Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
Proc Natl Acad Sci U S A ; 106(44): 18527-32, 2009 Nov 03.
Artigo em Inglês | MEDLINE | ID: mdl-19846791

RESUMO

Cobalamin-dependent methionine synthase (MetH) is a modular protein that catalyzes the transfer of a methyl group from methyltetrahydrofolate to homocysteine to produce methionine and tetrahydrofolate. The cobalamin cofactor, which serves as both acceptor and donor of the methyl group, is oxidized once every approximately 2,000 catalytic cycles and must be reactivated by the uptake of an electron from reduced flavodoxin and a methyl group from S-adenosyl-L-methionine (AdoMet). Previous structures of a C-terminal fragment of MetH (MetH(CT)) revealed a reactivation conformation that juxtaposes the cobalamin- and AdoMet-binding domains. Here we describe 2 structures of a disulfide stabilized MetH(CT) ((s-s)MetH(CT)) that offer further insight into the reactivation of MetH. The structure of (s-s)MetH(CT) with cob(II)alamin and S-adenosyl-L-homocysteine represents the enzyme in the reactivation step preceding electron transfer from flavodoxin. The structure supports earlier suggestions that the enzyme acts to lower the reduction potential of the Co(II)/Co(I) couple by elongating the bond between the cobalt and its upper axial water ligand, effectively making the cobalt 4-coordinate, and illuminates the role of Tyr-1139 in the stabilization of this 4-coordinate state. The structure of (s-s)MetH(CT) with aquocobalamin may represent a transient state at the end of reactivation as the newly remethylated 5-coordinate methylcobalamin returns to the 6-coordinate state, triggering the rearrangement to a catalytic conformation.


Assuntos
5-Metiltetra-Hidrofolato-Homocisteína S-Metiltransferase/metabolismo , Escherichia coli/enzimologia , Vitamina B 12/metabolismo , 5-Metiltetra-Hidrofolato-Homocisteína S-Metiltransferase/química , Substituição de Aminoácidos , Domínio Catalítico , Cobalto/metabolismo , Ativação Enzimática , Modelos Biológicos , Modelos Moleculares , Mutação/genética , Oxirredução , Estrutura Secundária de Proteína , S-Adenosil-Homocisteína/metabolismo , Análise Espectral , Vitamina B 12/análogos & derivados
2.
Proc Natl Acad Sci U S A ; 105(11): 4115-20, 2008 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-18332423

RESUMO

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.


Assuntos
5-Metiltetra-Hidrofolato-Homocisteína S-Metiltransferase/química , 5-Metiltetra-Hidrofolato-Homocisteína S-Metiltransferase/metabolismo , Dissulfetos/química , Dissulfetos/metabolismo , Histidina/metabolismo , Vitamina B 12/química , Vitamina B 12/metabolismo , 5-Metiltetra-Hidrofolato-Homocisteína S-Metiltransferase/genética , Sítios de Ligação , Catálise , Cristalografia por Raios X , Ativação Enzimática , Escherichia coli/enzimologia , Escherichia coli/genética , Expressão Gênica , Histidina/genética , Ligantes , Modelos Moleculares , Fotoquímica , Estrutura Quaternária de Proteína , Estrutura Terciária de Proteína , Homologia Estrutural de Proteína , Compostos de Sulfidrila/química , Compostos de Sulfidrila/metabolismo , Temperatura , Titulometria
3.
Proc Natl Acad Sci U S A ; 105(9): 3286-91, 2008 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-18296644

RESUMO

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.


Assuntos
5-Metiltetra-Hidrofolato-Homocisteína S-Metiltransferase/química , Homocisteína/metabolismo , Metais , 5-Metiltetra-Hidrofolato-Homocisteína S-Metiltransferase/metabolismo , Sítios de Ligação , Catálise , Cristalografia por Raios X , Elasticidade , Metilação , Metiltransferases , Conformação Proteica , Thermotoga maritima/enzimologia , Vitamina B 12 , Zinco
4.
J Bacteriol ; 191(10): 3407-10, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19286805

RESUMO

Cobalamin-independent methionine synthase (MetE) catalyzes the final step in Escherichia coli methionine biosynthesis but is inactivated under oxidative conditions, triggering a methionine deficiency. This study demonstrates that the mutation of MetE cysteine 645 to alanine completely eliminates the methionine auxotrophy imposed by diamide treatment, suggesting that modulation of MetE activity via cysteine 645 oxidation has significant physiological consequences for oxidatively stressed cells.


Assuntos
Cisteína/fisiologia , Escherichia coli/metabolismo , Metionina/metabolismo , Metiltransferases/metabolismo , Alanina/genética , Alanina/fisiologia , Cisteína/genética , Diamida/farmacologia , Escherichia coli/efeitos dos fármacos , Escherichia coli/genética , Metiltransferases/efeitos dos fármacos , Metiltransferases/genética , Mutação , Oxirredução/efeitos dos fármacos , Relação Estrutura-Atividade
5.
J Phys Chem B ; 113(15): 5245-54, 2009 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-19298066

RESUMO

The one-electron-reduced form of vitamin B(12), cob(II)alamin (Co(2+)Cbl), is found in several essential human enzymes, including the cobalamin-dependent methionine synthase (MetH). In this work, experimentally validated electronic structure descriptions for two "base-off" Co(2+)Cbl species have been generated using a combined spectroscopic and computational approach, so as to obtain definitive clues as to how these and related enzymes catalyze the thermodynamically challenging reduction of Co(2+)Cbl to cob(I)alamin (Co(1+)Cbl). Specifically, electron paramagnetic resonance (EPR), electronic absorption (Abs), and magnetic circular dichroism (MCD) spectroscopic techniques have been employed as complementary tools to characterize the two distinct forms of base-off Co(2+)Cbl that can be trapped in the H759G variant of MetH, one containing a five-coordinate and the other containing a four-coordinate, square-planar Co(2+) center. Accurate spin Hamiltonian parameters for these low-spin Co(2+) centers have been determined by collecting EPR data using both X- and Q-band microwave frequencies, and Abs and MCD spectroscopic techniques have been employed to probe the corrin-centered pi --> pi* and Co-based d --> d excitations, respectively. By using these spectroscopic data to evaluate electronic structure calculations, we found that density functional theory provides a reasonable electronic structure description for the five-coordinate form of base-off Co(2+)Cbl. However, it was necessary to resort to a multireference ab initio treatment to generate a more realistic description of the electronic structure of the four-coordinate form. Consistent with this finding, our computational data indicate that, in the five-coordinate Co(2+)Cbl species, the unpaired spin density is primarily localized in the Co 3d(z(2))-based molecular orbital, as expected, whereas in the four-coordinate form, extensive Co 3d orbital mixing, configuration interaction, and spin-orbit coupling cause the unpaired electron to delocalize over several Co 3d orbitals. These results provide important clues to the mechanism of enzymatic Co(2+)Cbl --> Co(1+)Cbl reduction.


Assuntos
Simulação por Computador , Modelos Químicos , Vitamina B 12/análogos & derivados , Espectroscopia de Ressonância de Spin Eletrônica/métodos , Conformação Molecular , Vitamina B 12/química
6.
J Am Chem Soc ; 130(48): 16374-81, 2008 Dec 03.
Artigo em Inglês | MEDLINE | ID: mdl-19006389

RESUMO

The cobalamin-dependent methionine synthase (MetH) from Escherichia coli is a modular enzyme that catalyzes a methyl group transfer from methyltetrahydrofolate to homocysteine via a methylcob(III)alamin (MeCbl) intermediate, generating tetrahydrofolate and methionine (Met). Once every approximately 2000 turnovers, the cobalamin cofactor is converted to the inactive cob(II)alamin (Co(2+)Cbl) form, from which MeCbl has to be recovered for MetH to re-enter the catalytic cycle. A particularly puzzling aspect of this reactivation process is that it requires the reduction of the Co(2+)Cbl species to cob(I)alamin (Co(1+)Cbl) by flavodoxin, a reaction that would appear to be endergonic on the basis of the corresponding reduction potentials. To explore how MetH may overcome this apparent thermodynamic challenge, we have prepared the I690C/G743C variant of a C-terminal fragment of MetH (MetH(CT)) to lock the enzyme into the activation conformation without perturbing any of the residues in the vicinity of the active site. A detailed spectroscopic characterization of this species and the I690C/G743C/Y1139F MetH(CT) triple mutant reveals that the strategy employed by MetH to activate Co(2+)Cbl for Co(2+) --> Co(1+) reduction likely involves (i) an axial ligand switch to generate a five-coordinate species with an axially coordinated water molecule and (ii) a significant lengthening, or perhaps complete rupture, of the Co-OH(2) bond of the cofactor, thereby causing a large stabilization of the Co 3d(z(2))-based "redox-active" molecular orbital. The lengthening of the Co-OH(2) bond is mediated by the Y1139 active-site residue and becomes much more dramatic when the S-adenosylmethionine substrate is present in the enzyme active site. This substrate requirement provides MetH a means to suppress deleterious side reactions involving the transiently formed Co(1+)Cbl "supernucleophile".


Assuntos
5-Metiltetra-Hidrofolato-Homocisteína S-Metiltransferase/química , 5-Metiltetra-Hidrofolato-Homocisteína S-Metiltransferase/metabolismo , S-Adenosilmetionina/química , S-Adenosilmetionina/metabolismo , Vitamina B 12/química , Vitamina B 12/metabolismo , Dicroísmo Circular , Cobalto/química , Cristalografia por Raios X , Espectroscopia de Ressonância de Spin Eletrônica , Ativação Enzimática , Modelos Moleculares , Estrutura Terciária de Proteína , Tirosina/genética , Tirosina/metabolismo
7.
Mol Genet Metab ; 94(3): 336-42, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18413293

RESUMO

Low dietary folate and polymorphisms in genes of folate metabolism can influence risk for pregnancy complications and birth defects. Methionine synthase reductase (MTRR) is required for activation of methionine synthase, a folate- and vitamin B(12)-dependent enzyme. A polymorphism in MTRR (p.I22M), present in the homozygous state in 25% of many populations, may increase risk for neural tube defects. To examine the impact of MTRR deficiency on early development and congenital heart defects, we used mice harboring a gene-trapped (gt) allele in Mtrr. Female mice (Mtrr(+/+), Mtrr(+/gt), and Mtrr(gt/gt)) were mated with male Mtrr(+/g) mice. Reproductive outcomes and cardiac phenotype (presence of defects and myocardial thickness) were assessed at E14.5. Mtrr-deficient mothers had more resorptions and more delayed embryos per litter (resorptions per litter: 0.29+/-0.13; 1.21+/-0.41; 1.87+/-0.38 and delayed embryos per litter: 0.07+/-0.07; 0.14+/-0.14; 0.60+/-0.24 in Mtrr(+/+), Mtrr(+/gt), and Mtrr(gt/gt) mothers respectively). Placentae of Mtrr(gt/gt) mothers were smaller and their embryos were smaller, with myocardial hypoplasia and a higher incidence of ventricular septal defects (VSD) per litter (0; 0.57+/-0.30; 1.57+/-0.67 in Mtrr(+/+), Mtrr(+/gt), and Mtrr(gt/gt) groups respectively). Embryonic Mtrr(gt/gt) genotype was associated with reduced embryonic length, reduced embryonic and placental weight, and higher incidence of VSD, but did not affect myocardial thickness or embryonic delay. We conclude that Mtrr deficiency adversely impacts reproductive outcomes and cardiac development in mice. These findings may have implications for nutritional prevention of heart defects, particularly in women with the common MTRR polymorphism.


Assuntos
Ferredoxina-NADP Redutase/genética , Cardiopatias Congênitas/genética , Resultado da Gravidez/genética , Reprodução/genética , Animais , Embrião de Mamíferos , Feminino , Ferredoxina-NADP Redutase/deficiência , Genótipo , Cardiopatias Congênitas/epidemiologia , Incidência , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Gravidez , Caracteres Sexuais
8.
Protein Sci ; 16(8): 1588-95, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17656578

RESUMO

The crystal structure of the Thermotoga maritima gene product TM0269, determined as part of genome-wide structural coverage of T. maritima by the Joint Center for Structural Genomics, revealed structural homology with the fourth module of the cobalamin-dependent methionine synthase (MetH) from Escherichia coli, despite the lack of significant sequence homology. The gene specifying TM0269 lies in close proximity to another gene, TM0268, which shows sequence homology with the first three modules of E. coli MetH. The fourth module of E. coli MetH is required for reductive remethylation of the cob(II)alamin form of the cofactor and binds the methyl donor for this reactivation, S-adenosylmethionine (AdoMet). Measurements of the rates of methionine formation in the presence and absence of TM0269 and AdoMet demonstrate that both TM0269 and AdoMet are required for reactivation of the inactive cob(II)alamin form of TM0268. These activity measurements confirm the structure-based assignment of the function of the TM0269 gene product. In the presence of TM0269, AdoMet, and reductants, the measured activity of T. maritima MetH is maximal near 80 degrees C, where the specific activity of the purified protein is approximately 15% of that of E. coli methionine synthase (MetH) at 37 degrees C. Comparisons of the structures and sequences of TM0269 and the reactivation domain of E. coli MetH suggest that AdoMet may be bound somewhat differently by the homologous proteins. However, the conformation of a hairpin that is critical for cobalamin binding in E. coli MetH, which constitutes an essential structural element, is retained in the T. maritima reactivation protein despite striking divergence of the sequences.


Assuntos
5-Metiltetra-Hidrofolato-Homocisteína S-Metiltransferase/química , Proteínas de Bactérias/química , Thermotoga maritima/enzimologia , 5-Metiltetra-Hidrofolato-Homocisteína S-Metiltransferase/genética , 5-Metiltetra-Hidrofolato-Homocisteína S-Metiltransferase/metabolismo , Sequência de Aminoácidos , Apoproteínas/química , Apoproteínas/genética , Apoproteínas/metabolismo , Proteínas de Bactérias/metabolismo , Ativação Enzimática , Metilação , Modelos Moleculares , Dados de Sequência Molecular , Oxirredução , Conformação Proteica , S-Adenosilmetionina/química , S-Adenosilmetionina/metabolismo , Temperatura , Thermotoga maritima/química , Thermotoga maritima/genética , Vitamina B 12/química , Vitamina B 12/metabolismo
9.
Antioxid Redox Signal ; 9(11): 1911-21, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17696766

RESUMO

Mouse models that perturb homocysteine metabolism, including genetic mouse models that result in deficiencies of methylenetetrahydrofolate reductase, methionine synthase, methionine synthase reductase, and cystathionine beta-synthase, and a pharmaceutically induced mouse model with a transient deficiency in betainehomocysteine methyl transferase, have now been characterized and can be compared. Although each of these enzyme deficiencies is associated with moderate to severe hyperhomocyst(e)inemia, the broader metabolic profiles are profoundly different. In particular, the various models differ in the degree to which tissue ratios of S-adenosylmethionine to S-adenosylhomocysteine are reduced in the face of elevated plasma homocyst(e)ine, and in the distribution of the tissue folate pools. These different metabolic profiles illustrate the potential complexities of hyperhomocyst(e)inemia in humans and suggest that comparison of the disease phenotypes of the various mouse models may be extremely useful in dissecting the underlying risk factors associated with human hyperhomocyst(e)inemia.


Assuntos
5-Metiltetra-Hidrofolato-Homocisteína S-Metiltransferase/metabolismo , Cistationina beta-Sintase/metabolismo , Ferredoxina-NADP Redutase/metabolismo , Hiper-Homocisteinemia/metabolismo , Metilenotetra-Hidrofolato Redutase (NADPH2)/metabolismo , 5-Metiltetra-Hidrofolato-Homocisteína S-Metiltransferase/sangue , 5-Metiltetra-Hidrofolato-Homocisteína S-Metiltransferase/química , Animais , Cistationina beta-Sintase/sangue , Cistationina beta-Sintase/química , Modelos Animais de Doenças , Ferredoxina-NADP Redutase/sangue , Ferredoxina-NADP Redutase/química , Hiper-Homocisteinemia/enzimologia , Metilenotetra-Hidrofolato Redutase (NADPH2)/sangue , Metilenotetra-Hidrofolato Redutase (NADPH2)/química , Camundongos , Camundongos Transgênicos , Modelos Biológicos , Estrutura Molecular , S-Adenosil-Homocisteína/metabolismo , S-Adenosilmetionina/metabolismo
10.
BMC Microbiol ; 7: 2, 2007 Jan 18.
Artigo em Inglês | MEDLINE | ID: mdl-17233899

RESUMO

BACKGROUND: The glutamate synthase operon (gltBDF) contributes to one of the two main pathways of ammonia assimilation in Escherichia coli. Of the seven most-global regulators, together affecting expression of about half of all E. coli genes, two were previously shown to exert direct, positive control on gltBDF transcription: Lrp and IHF. The involvement of Lrp is unusual in two respects: first, it is insensitive to the usual coregulator leucine, and second, Lrp binds more than 150 bp upstream of the transcription starting point. There was indirect evidence for involvement of a third global regulator, Crp. Given the physiological importance of gltBDF, and the potential opportunity to learn about integration of global regulatory signals, a combination of in vivo and in vitro approaches was used to investigate the involvement of additional regulatory proteins, and to determine their relative binding positions and potential interactions with one another and with RNA polymerase (RNAP). RESULTS: Crp and a more local regulator, ArgR, directly control gltBDF transcription, both acting negatively. Crp-cAMP binds a sequence centered at -65.5 relative to the transcript start. Mutation of conserved nucleotides in the Crp binding site abolishes the Crp-dependent repression. ArgR also binds to the gltBDF promoter region, upstream of the Lrp binding sites, and decreases transcription. RNAP only yields a defined DNAse I footprint under two tested conditions: in the presence of both Lrp and IHF, or in the presence of Crp-cAMP. The DNAse I footprint of RNAP in the presence of Lrp and IHF is altered by ArgR. CONCLUSION: The involvement of nearly half of E. coli's most-global regulatory proteins in the control of gltBDF transcription is striking, but seems consistent with the central metabolic role of this operon. Determining the mechanisms of activation and repression for gltBDF was beyond the scope of this study. However the results are consistent with a model in which IHF bends the DNA to allow stabilizing contacts between Lrp and RNAP, ArgR interferes with such contacts, and Crp introduces an interfering bend in the DNA and/or stabilizes RNAP in a poised but inactive state.


Assuntos
Proteínas de Escherichia coli/metabolismo , Escherichia coli/metabolismo , Glutamato Sintase/metabolismo , Óperon , Sequência de Bases , Sítios de Ligação , Proteína Receptora de AMP Cíclico/genética , Proteína Receptora de AMP Cíclico/metabolismo , RNA Polimerases Dirigidas por DNA/metabolismo , Desoxirribonuclease I/metabolismo , Ensaio de Desvio de Mobilidade Eletroforética , Escherichia coli/enzimologia , Escherichia coli/genética , Proteínas de Escherichia coli/genética , Regulação Bacteriana da Expressão Gênica , Glutamato Sintase/genética , Fatores Hospedeiros de Integração/genética , Fatores Hospedeiros de Integração/metabolismo , Proteína Reguladora de Resposta a Leucina/genética , Proteína Reguladora de Resposta a Leucina/metabolismo , Redes e Vias Metabólicas , Dados de Sequência Molecular , Regiões Promotoras Genéticas , Ligação Proteica , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo
11.
PLoS Biol ; 2(11): e336, 2004 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-15502870

RESUMO

In nature, Escherichia coli are exposed to harsh and non-ideal growth environments-nutrients may be limiting, and cells are often challenged by oxidative stress. For E. coli cells confronting these realities, there appears to be a link between oxidative stress, methionine availability, and the enzyme that catalyzes the final step of methionine biosynthesis, cobalamin-independent methionine synthase (MetE). We found that E. coli cells subjected to transient oxidative stress during growth in minimal medium develop a methionine auxotrophy, which can be traced to an effect on MetE. Further experiments demonstrated that the purified enzyme is inactivated by oxidized glutathione (GSSG) at a rate that correlates with protein oxidation. The unique site of oxidation was identified by selectively cleaving N-terminally to each reduced cysteine and analyzing the results by liquid chromatography mass spectrometry. Stoichiometric glutathionylation of MetE by GSSG occurs at cysteine 645, which is strategically located at the entrance to the active site. Direct evidence of MetE oxidation in vivo was obtained from thiol-trapping experiments in two different E. coli strains that contain highly oxidizing cytoplasmic environments. Moreover, MetE is completely oxidized in wild-type E. coli treated with the thiol-oxidizing agent diamide; reduced enzyme reappears just prior to the cells resuming normal growth. We argue that for E. coli experiencing oxidizing conditions in minimal medium, MetE is readily inactivated, resulting in cellular methionine limitation. Glutathionylation of the protein provides a strategy to modulate in vivo activity of the enzyme while protecting the active site from further damage, in an easily reversible manner. While glutathionylation of proteins is a fairly common mode of redox regulation in eukaryotes, very few proteins in E. coli are known to be modified in this manner. Our results are complementary to the independent findings of Leichert and Jakob presented in the accompanying paper (Leichert and Jakob 2004), which provide evidence that MetE is one of the proteins in E. coli most susceptible to oxidation. In eukaryotes, glutathionylation of key proteins involved in protein synthesis leads to inhibition of translation. Our studies suggest a simpler mechanism is employed by E. coli to achieve the same effect.


Assuntos
Proteínas de Escherichia coli/fisiologia , Escherichia coli/metabolismo , Metiltransferases/fisiologia , Estresse Oxidativo , Vitamina B 12/química , Dicroísmo Circular , Cisteína/química , Dissulfetos/química , Proteínas de Escherichia coli/metabolismo , Glutationa/química , Dissulfeto de Glutationa/química , Peróxido de Hidrogênio/farmacologia , Focalização Isoelétrica , Espectrometria de Massas , Metionina/química , Metiltransferases/metabolismo , Modelos Biológicos , Modelos Químicos , Dados de Sequência Molecular , Oxigênio/química , Conformação Proteica , Temperatura , Tripsina/química , Zinco/química
13.
Biochem J ; 376(Pt 2): 517-24, 2003 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-12954077

RESUMO

Roles played by homocysteine and choline in the regulation of MS (methionine synthase) have been examined in fungi. The Aspergillus nidulans metH gene encoding MS was cloned and characterized. Its transcription was not regulated by methionine, but was enhanced by homocysteine and repressed by choline and betaine. MS activity levels were regulated in a similar way. The repression by betaine was due to its metabolic conversion to choline, which was found to be very efficient in A. nidulans. Betaine and choline supplementation stimulated growth of leaky metH mutants apparently by decreasing the demand for methyl groups and thus saving methionine and S -adenosylmethionine. We have also found that homocysteine stimulates transcription of MS-encoding genes in Saccharomyces cerevisiae and Schizosaccharomyces pombe.


Assuntos
5-Metiltetra-Hidrofolato-Homocisteína S-Metiltransferase/genética , Aspergillus nidulans/enzimologia , Aspergillus nidulans/genética , Colina/farmacologia , Regulação Fúngica da Expressão Gênica , Homocisteína/farmacologia , 5-Metiltetra-Hidrofolato-Homocisteína S-Metiltransferase/biossíntese , 5-Metiltetra-Hidrofolato-Homocisteína S-Metiltransferase/metabolismo , Aspergillus nidulans/efeitos dos fármacos , Sequência de Bases , Betaína/farmacologia , Clonagem Molecular , Ativação Enzimática , Regulação Enzimológica da Expressão Gênica , Metionina/metabolismo , Modelos Biológicos , Dados de Sequência Molecular , RNA Mensageiro/biossíntese , Transcrição Gênica
16.
Met Ions Life Sci ; 6: 53-114, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-20877792

RESUMO

This chapter reviews the literature on cobalamin- and corrinoid-containing enzymes. These enzymes fall into two broad classes, those using methylcobalamin or related methylcorrinoids as prosthetic groups and catalyzing methyl transfer reactions, and those using adenosylcobalamin as the prosthetic group and catalyzing the generation of substrate radicals that in turn undergo rearrangements and/or eliminations.

17.
Curr Opin Struct Biol ; 18(6): 658-66, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19059104

RESUMO

Methyltransferases that employ cobalamin cofactors, or their analogs the cobamides, as intermediates in catalysis of methyl transfer play vital roles in energy generation in anaerobic unicellular organisms. In a broader range of organisms they are involved in the conversion of homocysteine to methionine. Although the individual methyl transfer reactions catalyzed are simple S(N)2 displacements, the required change in coordination at the cobalt of the cobalamin or cobamide cofactors and the lability of the reduced Co(+1) intermediates introduces the necessity for complex conformational changes during the catalytic cycle. Recent spectroscopic and structural studies on several of these methyltransferases have helped to reveal the strategies by which these conformational changes are facilitated and controlled.


Assuntos
Cobamidas/química , Metiltransferases/química , Vitamina B 12/química , 5-Metiltetra-Hidrofolato-Homocisteína S-Metiltransferase/química , 5-Metiltetra-Hidrofolato-Homocisteína S-Metiltransferase/metabolismo , Acil Coenzima A/metabolismo , Animais , Cobamidas/metabolismo , Corrinoides/química , Corrinoides/metabolismo , Histidina/química , Histidina/metabolismo , Ferro/química , Ligantes , Metiltransferases/metabolismo , Complexos Multienzimáticos/química , Enxofre/química , Vitamina B 12/metabolismo
18.
Clin Chem Lab Med ; 45(12): 1700-3, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17937607

RESUMO

There are now four genetic mouse models that induce hyperhomocyst(e)inemia by decreasing the activity of an enzyme involved in homocysteine metabolism: cystathionine beta-synthase, methylenetetrahydrofolate reductase, methionine synthase and methionine synthase reductase. While each enzyme deficiency leads to murine hyperhomocyst(e)inemia, the accompanying metabolic profiles are significantly and often unexpectedly, different. Deficiencies in cystathionine beta-synthase lead to elevated plasma methionine, while deficiencies of the remaining three enzymes lead to hypomethioninemia. The liver [S-adenosylmethionine]/[S-adenosylhomocysteine] ratio is decreased in mice lacking methylenetetrahydrofolate reductase or cystathionine beta-synthase, but unexpectedly increased in mice with deficiencies in methionine synthase or methionine synthase reductase. Folate pool imbalances are observed in complete methylenetetrahydrofolate reductase deficiency, where methyltetra-hydrofolate is a minor component, and in methionine synthase reductase deficiency, where methyltetrahydrofolate is increased relative to wild-type mice. These differences illustrate the potential diversity among human patients with hyperhomocyst(e)inemia, and strengthen the argument that the pathologies associated with the dissimilar forms of the condition will require different treatments.


Assuntos
Homocisteína/sangue , Hiper-Homocisteinemia/sangue , Animais , Humanos , Camundongos , Modelos Animais
19.
EcoSal Plus ; 2(2)2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26443588

RESUMO

Many microorganisms and plants possess the ability to synthesize folic acid derivatives de novo, initially forming dihydrofolate. All the folic acid derivatives that serve as recipients and donors of one-carbon units are derivatives of tetrahydrofolate, which is formed from dihydrofolate by an NADPH-dependent reduction catalyzed by dihydrofolate reductase (FolA). This review discusses the biosynthesis of dihydrofolate monoglutamate, its reduction to tetrahydrofolate monoglutamate, and the addition of glutamyl residues to form folylpolyglutamates. Escherichia coli and Salmonella, like many microorganisms that can synthesize folate de novo, appear to lack the ability to transport folate into the cell and are thus highly susceptible to inhibitors of folate biosynthesis. The review includes a brief discussion of the inhibition of folate biosynthesis by sulfa drugs. The folate biosynthetic pathway can be divided into two sections. First, the aromatic precursor chorismate is converted to paminobenzoic acid (PABA) by the action of three proteins. Second, the pteridine portion of folate is made from GTP and coupled to PABA to generate dihydropteroate, and the bifunctional protein specified by folC, dihydrofolate synthetase, or folylpolyglutamate synthetase, adds the initial glutamate molecule to form dihydrofolate (H2PteGlu1, or dihydropteroylmonoglutamate). Bacteriophage T4 infection of E. coli has been shown to cause alterations in the metabolism of folate derivatives. Infection is associated with an increase in the chain lengths in folylpolyglutamates and particularly the accumulation of hexaglutamate derivatives.

20.
Biochemistry ; 46(43): 12382-92, 2007 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-17924667

RESUMO

Cobalamin-dependent methionine synthase (MetH) of Escherichia coli is a large, modular enzyme that uses a cobalamin prosthetic group as a donor or acceptor in three separate methyl transfer reactions. The prosthetic group alternates between methylcobalamin and cob(I)alamin during catalysis as homocysteine is converted to methionine using a methyl group derived from methyltetrahydrofolate. Occasional oxidation of cob(I)alamin to cob(II)alamin inactivates the enzyme. Reductive methylation with flavodoxin and adenosylmethionine returns the enzyme to an active methylcobalamin state. At different points during the reaction cycle, the coordination state of the cobalt of the cobalamin changes. The imidazole side chain of His759 coordinates to cobalamin in a "His-on" state and dissociates to produce a "His-off" state. The His-off state has been associated with a conformation of MetH that is poised for reactivation of cobalamin by reductive methylation rather than catalysis. Our studies on cob(III)alamins bound to MetH, specifically aqua-, methyl-, and n-propylcobalamin, show a correlation between the accessibility of the reactivation conformation and the order of the established ligand trans influence. The trans influence also controls the affinity of MetH in the cob(III)alamin form for flavodoxin. Flavodoxin, which acts to shift the conformational equilibrium toward the reactivation conformation, binds less tightly to MetH when the cob(III)alamin has a strong trans ligand and therefore has less positive charge on cobalt. These results are compared to those for cob(II)alamin MetH, illustrating that access to the reactivation conformation is governed by the net charge on the cobalt as well as the trans influence in cob(III)alamins.


Assuntos
5-Metiltetra-Hidrofolato-Homocisteína S-Metiltransferase/química , Vitamina B 12/metabolismo , 5-Metiltetra-Hidrofolato-Homocisteína S-Metiltransferase/metabolismo , Ligantes , Conformação Proteica , Espectrofotometria/métodos
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa