Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Biochemistry ; 39(20): 6250-7, 2000 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-10821701

RESUMO

The hydrate of glycolaldehyde is a substrate analogue that induces the formation of cob(II)alamin and 5'-deoxyadenosine from adenosylcobalamin at the active site of dioldehydrase, and the resulting complex is inactive. The carbon atoms of glycolaldehyde hydrate remain bound to this complex, and it has been postulated that the first step or steps of the catalytic process on glycolaldehyde hydrate generate an intermediate that undergoes a destructive side reaction leading to inactivation of the enzyme [Wagner, O. W., Lee, H. A., Jr., Frey, P. A., and Abeles, R. H. (1966) J. Biol. Chem. 249, 1751-1762]. All evidence suggests that dioldehydrase reaction proceeds by a radical mechanism, and the glycolaldehyde hydrate is expected to be converted initially into a radical. Electron paramagnetic resonance (EPR) spectroscopic analysis of the inactivated complex shows that glycolaldehyde is transformed into a cis-ethanesemidione radical that is weakly spin-coupled to the cob(II)alamin in the active site of the enzyme. This radical has been identified by analysis of EPR spectra obtained from samples with (13)C- and (2)H-labeled forms of glycolaldehyde. The analysis shows that the stable radical associated with the inactive complex is symmetrical and that it contains a single solvent-exchangeable proton, consistent with a cis-ethanesemidione. Glycolaldehyde also inactivates ethanolamine ammonia-lyase (EAL). EPR studies of ethanolamine ammonia-lyase reveal that treatment with glycolaldehyde also results in formation of an ethanesemidione radical bound in the active site. The suicide inactivation in both enzymatic reactions is postulated to result from formation of this stable radical, which cannot react further to abstract a hydrogen atom from 5'-deoxyadenosine. Analysis of the electron spin-spin coupling between the semidione radicals and cob(II)alamin in both enzymes indicates that the distance between the radical and Co(2+) is approximately 11 A in each case.


Assuntos
Acetaldeído/análogos & derivados , Inibidores Enzimáticos/química , Etanolamina Amônia-Liase/química , Propanodiol Desidratase/química , Acetaldeído/química , Cobalto/química , Espectroscopia de Ressonância de Spin Eletrônica , Ativação Enzimática , Radicais Livres/química , Modelos Químicos , Ressonância Magnética Nuclear Biomolecular , Propanodiol Desidratase/antagonistas & inibidores , Propilenoglicol/química , Salmonella typhimurium/enzimologia , Solventes , Marcadores de Spin , Especificidade por Substrato
2.
Arch Biochem Biophys ; 370(1): 138-41, 1999 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-10496987

RESUMO

Ethanolamine ammonia-lyase (EAL, EC 4.3.1.7) catalyzes a coenzyme B(12)-dependent deamination of vicinal amino alcohols. The mode of binding of coenzyme B(12) to EAL has been investigated by electron paramagnetic resonance spectroscopy (EPR) using [(15)N]-dimethylbenzimidazole-coenzyme B(12). EAL was incubated with either unlabeled or (15)N-enriched coenzyme B(12) and then either exposed to light or treated with ethanol to generate the cleaved form of the cofactor, cob(II)alamin (B(12r)) bound in the active site. The reaction mixtures were examined by EPR spectroscopy at 77 K. (15)N superhyperfine splitting in the EPR signals of the low-spin Co(2+) of B(12r), bound in the active site of EAL, indicates that the dimethylbenzimidazole moiety of the cofactor contributes the lower axial ligand consistent with "base-on" binding of coenzyme B(12) to EAL.


Assuntos
Cobamidas/metabolismo , Etanolamina Amônia-Liase/química , Etanolamina Amônia-Liase/metabolismo , Benzimidazóis/química , Benzimidazóis/farmacocinética , Sítios de Ligação , Cobalto , Espectroscopia de Ressonância de Spin Eletrônica , Cinética , Isótopos de Nitrogênio , Especificidade por Substrato
3.
Biochemistry ; 38(12): 3668-76, 1999 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-10090754

RESUMO

Fhit is the protein product of FHIT, a candidate human tumor suppressor gene. Fhit catalyzes the hydrolysis of diadenosine triphosphate (Ap3A) to AMP and ADP. Fhit is here shown to catalyze the hydrolysis in H218O with production of adenosine 5'-[18O]phosphate and ADP, proving that the substitution of water is at Palpha and not at Pbeta. The chain fold of Fhit is similar to that of galactose-1-phosphate uridylyltransferase, which functions by a double-displacement mechanism through the formation of a covalent nucleotidyl-enzyme intermediate and overall retention of configuration at Palpha. The active site of Fhit contains a histidine motif that is reminiscent of the HPH motif in galactose-1-phosphate uridylyltransferases, in which the first histidine residue serves as the nucleophilic catalyst to which the nucleotidyl group is bonded covalently in the covalent intermediate. In this work, the Fhit-catalyzed cleavage of (RP)- and (SP)-gamma-(m-nitrobenzyl) adenosine 5'-O-1-thiotriphosphate (mNBATPalphaS) in H218O to adenosine 5'-[18O]thiophosphate is shown to proceed with overall retention of configuration at phosphorus. gamma-(m-Nitrobenzyl) adenosine 5'-O-triphosphate (mNBATP) is approximately as good a substrate for Fhit as Ap3A, and both (RP)- and (SP)-mNBATPalphaS are substrates that react at about 0.5% of the rate of Ap3A. The stereochemical evidence indicates that hydrolysis by Fhit proceeds by a double-displacement mechanism, presumably through a covalent AMP-enzyme intermediate.


Assuntos
Hidrolases Anidrido Ácido , Proteínas de Neoplasias , Proteínas/química , Trifosfato de Adenosina/análogos & derivados , Trifosfato de Adenosina/metabolismo , Catálise , Fosfatos de Dinucleosídeos/metabolismo , Genes Supressores de Tumor , Humanos , Hidrólise , Fósforo/química , Fósforo/metabolismo , Conformação Proteica , Proteínas/genética , Proteínas/metabolismo , Estereoisomerismo , Especificidade por Substrato
4.
Eur J Biochem ; 249(1): 180-6, 1997 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-9363770

RESUMO

Novel analogues of methylmalonyl-CoA and succinyl-CoA have been prepared and used for mechanistic investigations on the coenzyme-B12-dependent methylmalonyl-CoA mutase. 1-Carboxyethyl-CoA (1) and 2-carboxyethyl-CoA (2) as well as their sulphoxides (3 and 4) were moderately good inhibitors with Ki values 4-20 times higher than the Km for succinyl-CoA. 2-Carboxyethyl-CoA (2) and its sulphoxide 4 induced EPR signals when bound to the enzyme-coenzyme-B12 complex. The EPR spectrum of 2 and its sulphoxide 4 differed very much from those induced by the other substrates. In the case of 2 the EPR spectrum of the holoenzyme/inhibitor complex showed the presence of an organic radical coupled to cobal(II)amin. The same experiment with 4 leads to the formation of enzyme-bound cobal(II)amin with no detectable organic radical. The analogues 1 and 3 exhibited higher Ki values and did not induce EPR signals binding to the enzyme-coenzyme-B12 complex. Formyl-CoA and acrylate inhibited the enzyme synergistically but were unable to induce EPR signals and to form the product. Ethylmalonyl-CoA, known as a poor substrate, induced a similar but less intense EPR signal than the natural substrate methylmalonyl-CoA. The results are discussed in terms of the mechanism of the methylmalonyl-CoA mutase reaction.


Assuntos
Metilmalonil-CoA Mutase/química , Metilmalonil-CoA Mutase/metabolismo , Acil Coenzima A/química , Acil Coenzima A/farmacologia , Cobamidas/metabolismo , Espectroscopia de Ressonância de Spin Eletrônica , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Cinética , Metilmalonil-CoA Mutase/antagonistas & inibidores , Propionibacterium/enzimologia , Especificidade por Substrato
7.
Eur J Biochem ; 225(3): 891-6, 1994 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-7957226

RESUMO

The substrate-dependent homolysis of the cobalt-carbon bond and generation of organic radicals in the coenzyme-B12-methylmalonyl-CoA-mutase complex have been demonstrated by EPR measurements. Both the natural substrate methylmalonyl-CoA, its 13C-substituted analogue and the non-hydrolysable synthetic substrates succinyl-dethia(carba)-CoA, succinyl-dethia(dicarba)-CoA and 4-carboxy-2-oxo-butyl-CoA induced similar but not identical EPR signals. 3-Carboxypropyl-CoA, a novel competitive inhibitor, has been synthesised. Its Ki value of 89 +/- 6 microM was in the same range as the Km of succinyl-CoA. Using [5'-3H]adenosylcobalamin, an enzyme-dependent tritium transfer to the inhibitor has been shown. The enzyme-coenzyme-inhibitor complex also exhibited EPR signals that were less structured and less intensive than the corresponding signals with active substrates. These results prove that the inhibitor also induces cobalt-carbon bond homolysis and undergoes reversible hydrogen transfer but not rearrangement.


Assuntos
Metilmalonil-CoA Mutase/metabolismo , Ligação Competitiva , Carbono/química , Cobalto/química , Cobamidas/metabolismo , Coenzima A/farmacologia , Espectroscopia de Ressonância de Spin Eletrônica , Radicais Livres , Hidrogênio/química , Cinética , Metilmalonil-CoA Mutase/antagonistas & inibidores , Propionibacterium/enzimologia , Especificidade por Substrato
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...