Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 109
Filtrar
1.
Biochemistry ; 42(19): 5632-9, 2003 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-12741819

RESUMO

Ubiquinone-2 (UQ-2) selectively labeled with (13)C (I =(1)/(2)) at either the position 1- or the 4-carbonyl carbon is incorporated into the ubiquinol oxidase bo(3) from Escherichia coli in which the native quinone (UQ-8) has been previously removed. The resulting stabilized anion radical in the high-affinity quinone-binding site (Q(H)(*)(-)) is investigated using multifrequency (9, 34, and 94 GHz) electron paramagnetic resonance (EPR) spectroscopy. The corresponding spectra reveal dramatic differences in (13)C hyperfine couplings indicating a strongly asymmetric spin density distribution over the quinone headgroup. By comparison with previous results on labeled ubisemiquinones in proteins as well as in organic solvents, it is concluded that Q(H)(*)(-) is most probably bound to the protein via a one-sided hydrogen bond or a strongly asymmetric hydrogen-bonding network. This observation is discussed with regard to the function of Q(H) in the enzyme and contrasted with the information available on other protein-bound semiquinone radicals.


Assuntos
Oxirredutases/química , Oxirredutases/metabolismo , Ubiquinona/análogos & derivados , Ubiquinona/química , Ubiquinona/metabolismo , Sítios de Ligação , Coenzimas , Espectroscopia de Ressonância de Spin Eletrônica , Escherichia coli/enzimologia , Ligação de Hidrogênio , Cinética , Modelos Moleculares , Eletricidade Estática
2.
Biochem Biophys Res Commun ; 289(1): 181-90, 2001 Nov 23.
Artigo em Inglês | MEDLINE | ID: mdl-11708797

RESUMO

Previous studies have indicated that Ess1/Pin1, a gene in the parvulin family of peptidyl-prolyl isomerases (PPIases), plays an important role in regulating the G(2)/M transition of the cell cycle by binding cell-cycle-regulating proteins in eukaryotic cells. Although the ess1 gene has been considered to be essential in yeast, we have isolated viable ess1 deletion mutants and demonstrated, via analysis of yeast gene expression profiles using microarray techniques, a novel regulatory role for ESS1 in the G(1) phase. Although the overall expression profiles in the tested strains (C110-1, W303, S288c, and RAY-3AD) were similar, marked changes were detected for a number of genes involved in the molecular action of ESS1. Among these, the expression levels of a cyclophilin A gene, also a member of the PPIase family, increased in the ess1 null mutant derived from C110-1. Subsequent treatment with cyclosporin A significantly retarded growth, which suggests that ESS1 and cyclophilin A are functionally linked in yeast cells and play important roles at the G(1) phase of the cell cycle.


Assuntos
Ciclofilina A/genética , Ciclofilina A/metabolismo , Peptidilprolil Isomerase/genética , Peptidilprolil Isomerase/metabolismo , Saccharomyces cerevisiae/enzimologia , Saccharomyces cerevisiae/genética , Sequência de Bases , Ciclo Celular/fisiologia , DNA Fúngico/genética , Fase G1/fisiologia , Deleção de Genes , Expressão Gênica , Genes Fúngicos , Peptidilprolil Isomerase de Interação com NIMA , Análise de Sequência com Séries de Oligonucleotídeos , Fenótipo , RNA Fúngico/genética , RNA Fúngico/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Saccharomyces cerevisiae/citologia , Proteínas de Saccharomyces cerevisiae
3.
Biochemistry ; 39(50): 15620-5, 2000 Dec 19.
Artigo em Inglês | MEDLINE | ID: mdl-11112550

RESUMO

To elucidate a unique mechanism for the quinol oxidation in the Escherichia coli cytochrome bo, we applied pulse radiolysis technique to the wild-type enzyme with or without a single bound ubiquinone-8 at the high-affinity quinone binding site (Q(H)), using N-methylnicotinamide (NMA) as an electron mediator. With the ubiquinone bound enzyme, the reduction of the oxidase occurred in two phases as judged from kinetic difference spectra. In the faster phase, the transient species with an absorption maximum at 440 nm, a characteristic of the formation of ubisemiquinone anion radical, appeared within 10 micros after pulse radiolysis. In the slower phase, a decrease of absorption at 440 nm was accompanied by an increase of absorption at 428 and 561 nm, characteristic of the reduced form. In contrast, with the bound ubiquinone-8-free wild-type enzyme, NMA radicals directly reduced hemes b and o, though the reduction yield was low. These results indicate that a pathway for an intramolecular electron transfer from ubisemiquinone anion radical at the Q(H) site to heme b exists in cytochrome bo. The first-order rate constant of this process was calculated to be 1.5 x 10(3) s(-1) and is comparable to a turnover rate for ubiquinol-1. The rate constant for the intramolecular electron transfer decreased considerably with increasing pH, though the yields of the formation of ubisemiquinone anion radical and the subsequent reduction of the hemes were not affected. The pH profile was tightly linked to the stability of the bound ubisemiquinone in cytochrome bo [Ingledew, W. J., Ohnishi, T., and Salerno, J. C. (1995) Eur. J. Biochem. 227, 903-908], indicating that electron transfer from the bound ubisemiquinone at the Q(H) site to the hemes slows down at the alkaline pH where the bound ubisemiquinone can be stabilized. These findings are consistent with our previous proposal that the bound ubiquinone at the Q(H) site mediates electron transfer from the low-affinity quinol oxidation site in subunit II to low-spin heme b in subunit I.


Assuntos
Grupo dos Citocromos b , Citocromos/química , Proteínas de Escherichia coli , Hidroquinonas/química , Citocromos/metabolismo , Transporte de Elétrons , Escherichia coli , Radicais Livres , Hidroquinonas/metabolismo , Dados de Sequência Molecular , Oxirredução , Especificidade por Substrato
4.
Biochemistry ; 39(22): 6669-78, 2000 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-10828985

RESUMO

Cytochrome bo from Escherichia coli, a member of the heme-copper terminal oxidase superfamily, physiologically catalyzes reduction of O(2) by quinols and simultaneously translocates protons across the cytoplasmic membrane. The reaction of its ferric pulsed form with hydrogen peroxide was investigated with steady-state resonance Raman spectroscopy using a homemade microcirculating system. Three oxygen-isotope-sensitive Raman bands were observed at 805/X, 783/753, and (767)/730 cm(-)(1) for intermediates derived from H(2)(16)O(2)/H(2)(18)O(2). The experiments using H(2)(16)O(18)O yielded no new bands, indicating that all the bands arose from the Fe=O stretching (nu(Fe)(=)(O)) mode. Among them, the intensity of the 805/X cm(-)(1) pair increased at higher pH, and the species giving rise to this band seemed to correspond to the P intermediate of bovine cytochrome c oxidase (CcO) on the basis of the reported fact that the P intermediate of cytochrome bo appeared prior to the formation of the F species at higher pH. For this intermediate, a Raman band assignable to the C-O stretching mode of a tyrosyl radical was deduced at 1489 cm(-)(1) from difference spectra. This suggests that the P intermediate of cytochrome bo contains an Fe(IV)=O heme and a tyrosyl radical like compound I of prostaglandin H synthase. The 783/753 cm(-)(1) pair, which was dominant at neutral pH and close to the nu(Fe)(=)(O) frequency of the oxoferryl intermediate of CcO, presumably arises from the F intermediate. On the contrary, the (767)/730 cm(-)(1) species has no counterpart in CcO. Its presence may support the branched reaction scheme proposed previously for O(2) reduction by cytochrome bo.


Assuntos
Proteínas de Bactérias/química , Grupo dos Citocromos b , Citocromos/química , Proteínas de Escherichia coli , Peróxido de Hidrogênio/química , Monóxido de Carbono/química , Óxido de Deutério , Escherichia coli , Radicais Livres , Heme/química , Concentração de Íons de Hidrogênio , Ferro/química , Isótopos de Oxigênio , Análise Espectral Raman , Tirosina/química
5.
J Inorg Biochem ; 82(1-4): 19-25, 2000 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11132627

RESUMO

Cytochromes bo and bd are structurally unrelated terminal ubiquinol oxidases in the aerobic respiratory chain of Escherichia coli. The high-spin heme o-CuB binuclear center serves as the dioxygen reduction site for cytochrome bo, and the heme b595-heme d binuclear center for cytochrome bd. CuB coordinates three histidine ligands and serves as a transient ligand binding site en route to high-spin heme o one-electron donor to the oxy intermediate, and a binding site for bridging ligands like cyanide. In addition, it can protect the dioxygen reduction site through binding of a peroxide ion in the resting state, and connects directly or indirectly Tyr288 and Glu286 to carry out redox-driven proton pumping in the catalytic cycle. Contrary, heme b595 of cytochrome bd participate a similar role to CuB in ligand binding and dioxygen reduction but cannot perform such versatile roles because of its rigid structure.


Assuntos
Citocromos/química , Complexo de Proteínas da Cadeia de Transporte de Elétrons , Complexo IV da Cadeia de Transporte de Elétrons/química , Proteínas de Escherichia coli , Escherichia coli/enzimologia , Oxirredutases/química , Sítios de Ligação , Grupo dos Citocromos b , Citocromos/metabolismo , Espectroscopia de Ressonância de Spin Eletrônica , Complexo IV da Cadeia de Transporte de Elétrons/metabolismo , Ligantes , Oxirredução , Oxirredutases/metabolismo , Estrutura Quaternária de Proteína
6.
Biochemistry ; 38(44): 14683-9, 1999 Nov 02.
Artigo em Inglês | MEDLINE | ID: mdl-10545194

RESUMO

In this study we present the infrared spectroscopic characterization of the bound ubiquinone in cytochrome bo(3) from Escherichia coli. Electrochemically induced Fourier transform infrared (FTIR) difference spectra of DeltaUbiA (an oxidase devoid of bound ubiquinone) and DeltaUbiA reconstituted with ubiquinone 2 and with isotopically labeled ubiquinone 2, where (13)C was introduced either at the 1- or at the 4-position of the ring (C=O groups), have been obtained. The vibrational modes of the quinone bound to the discussed high-affinity binding site (Q(H)) are compared to those from the synthetic quinones in solution, leading to the assignment of the C=O modes to a split signal at 1658/1668 cm(-)(1), with both carbonyls similarly contributing. The FTIR spectra of DeltaUbiA reconstituted with the labeled quinones indicate an essentially symmetrical and weak hydrogen bonding of the two C=O groups from the neutral quinone with the protein and distinct conformations of the 2- and 3-methoxy groups. Perturbations of the vibrational modes of the 5-methyl side groups are discussed for a signal at 1452 cm(-)(1). Only negligible shifts of the aromatic ring modes can be reported for the reduced and the protonated form of the quinone. Alterations of the protein upon quinone binding are reflected in the electrochemically induced FTIR difference spectra. In particular, difference signals at 1640-1633 cm(-)(1) and 1700-1670 cm(-)(1) indicate variations of beta-sheet secondary structure elements and loops, bands at 1706 and 1678 cm(-)(1) are tentatively attributed to individual amino acids, and a difference signal a 1540 cm(-)(1) is discussed to reflect an influence on C=C modes of the porphyrin ring or on deprotonated propionate groups of the hemes. Further tentative assignments are presented and discussed. The (13)C labeling experiments allow the assignment of the vibrational modes of a bound ubiquinone 8 in the electrochemically induced FTIR difference spectra of wild-type bo(3).


Assuntos
Citocromos/química , Ubiquinona/química , Sítios de Ligação , Isótopos de Carbono , Grupo dos Citocromos b , Eletroquímica , Escherichia coli/química , Proteínas de Escherichia coli , Ligação de Hidrogênio , Oxirredução , Conformação Proteica , Espectroscopia de Infravermelho com Transformada de Fourier , Vibração
7.
J Biochem ; 126(5): 934-9, 1999 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-10544288

RESUMO

To probe the location of the quinol oxidation site and physical interactions for inter-subunit electron transfer, we constructed and characterized two chimeric oxidases in which subunit II (CyoA) of cytochrome bo-type ubiquinol oxidase from Escherichia coli was replaced with the counterpart (CaaA) of caa(3)-type cytochrome c oxidase from thermophilic Bacillus PS3. In pHNchi5, the C-terminal hydrophilic domain except a connecting region as to transmembrane helix II of CyoA was replaced with the counterpart of CaaA, which carries the Cu(A) site and cytochrome c domain. The resultant chimeric oxidase was detected immunochemically and spectroscopically, and the turnover numbers for Q(1)H(2) (ubiquinol-1) and TMPD (N,N, N',N'-tetramethyl-p-phenylenediamine) oxidation were 28 and 8.5 s(-1), respectively. In pHNchi6, the chimeric oxidase was designed to carry a minimal region of the cupredoxin fold containing all the Cu(A) ligands, and showed enzymatic activities of 65 and 5.1 s(-1), and an expression level better than that of pHNchi5. Kinetic analyses proved that the apparent lower turnover of the chimeric enzyme by pHNchi6 was due to the higher K(m) of the enzyme for Q(1)H(2) (220 microM) than that of cytochrome bo (48 microM), while in the enzyme by pHNchi5, both substrate-binding and internal electron transfer were perturbed. These results suggest that the connecting region and the C-terminal alpha(1)-alpha(2)-beta(11)-alpha(3) domain of CyoA are involved in the quinol oxidation and/or physical interactions for inter-subunit electron transfer, supporting our previous proposal [Sato-Watanabe, M., Mogi, T., Miyoshi, H., and Anraku, Y. (1998) Biochemistry 37, 12744-12752]. The close relationship of E. coli quinol oxidases to cytochrome c oxidase of Gram-positive bacteria like Bacillus was also indicated.


Assuntos
Complexo IV da Cadeia de Transporte de Elétrons/química , Sequência de Aminoácidos , Bacillus/enzimologia , Bacillus/genética , Sítios de Ligação , Transporte de Elétrons , Complexo IV da Cadeia de Transporte de Elétrons/genética , Complexo IV da Cadeia de Transporte de Elétrons/metabolismo , Escherichia coli/enzimologia , Escherichia coli/genética , Escherichia coli/crescimento & desenvolvimento , Dados de Sequência Molecular , Fenótipo , Estrutura Terciária de Proteína , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Espectrofotometria , Transformação Genética
8.
FEBS Lett ; 457(1): 61-4, 1999 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-10486564

RESUMO

To probe the functional role of a bound ubiquinone-8 in cytochrome bo-type ubiquinol oxidase from Escherichia coli, we examined reactions with ubiquinol-1 and dioxygen. Stopped-flow studies showed that anaerobic reduction of the wild-type and the bound ubiquinone-free (delta UbiA) enzymes with ubiquinol-1 immediately takes place with four kinetic phases. Replacement of the bound ubiquinone with 2,6-dibromo-4-cyanophenol (PC32) suppressed the anaerobic reduction of the hemes with ubiquinol-1 by eliminating the fast phase. Flow-flash studies in the reaction of the fully reduced enzyme with dioxygen showed that the heme b to heme o electron transfer occurs with a rate constant of approximately 10(4) s-1 in all three preparations. These results support our previous proposal that the bound ubiquinone is involved in facile oxidation of substrates in subunit II and subsequent intramolecular electron transfer to low-spin heme b in subunit I.


Assuntos
Grupo dos Citocromos b , Citocromos/metabolismo , Proteínas de Escherichia coli , Escherichia coli/metabolismo , Oxigênio/metabolismo , Ubiquinona/análogos & derivados , Ubiquinona/fisiologia , Catálise , Citocromos/isolamento & purificação , Heme/metabolismo , Oxirredução , Oxirredutases/metabolismo , Análise Espectral Raman , Fatores de Tempo , Ubiquinona/metabolismo
9.
J Biochem ; 126(3): 510-9, 1999 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10467166

RESUMO

Cytochrome bd-type ubiquinol oxidase contains two hemes b (b(558) and b(595)) and one heme d as the redox metal centers. To clarify the structure of the reaction center, we analyzed Escherichia coli cytochrome bd by visible absorption, EPR and FTIR spectroscopies using azide and cyanide as monitoring probes for the exogenous ligand binding site. Azide-binding caused the appearance of a new EPR low-spin signal characteristic of ferric iron-chlorin-azide species and a new visible absorption band at 647 nm. However, the bound azide ((14)N(3)) anti-symmetric stretching infrared band (2, 010.5 cm(-1)) showed anomalies upon (15)N-substitutions, indicating interactions with surrounding protein residues or heme b(595) in close proximity. The spectral changes upon cyanide-binding in the visible region were typical of those observed for ferric iron-chlorin species with diol substituents in macrocycles. However, we found no indication of a low-spin EPR signal corresponding to the ferric iron-chlorin-cyanide complexes. Instead, derivative-shaped signals at g = 3.19 and g = 7.15, which could arise from the heme d(Fe(3+))-CN-heme b(595)(Fe(3+)) moiety, were observed. Further, after the addition of cyanide, a part of ferric heme d showed the rhombic high-spin signal that coexisted with the g(z) = 2.85 signal ascribed to the minor heme b(595)-CN species. This indicates strong steric hindrance of cyanide-binding to ferric heme d with the bound cyanide at ferric heme b(595).


Assuntos
Azidas/metabolismo , Cianetos/metabolismo , Citocromos/metabolismo , Complexo de Proteínas da Cadeia de Transporte de Elétrons , Proteínas de Escherichia coli , Escherichia coli/enzimologia , Oxirredutases/metabolismo , Sítios de Ligação , Grupo dos Citocromos b , Citocromos/química , Espectroscopia de Ressonância de Spin Eletrônica , Oxirredutases/química , Ligação Proteica , Espectroscopia de Infravermelho com Transformada de Fourier
10.
FEBS Lett ; 457(2): 223-6, 1999 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-10471783

RESUMO

To probe the functional role of a bound ubiquinone-8 in cytochrome bo-type ubiquinol oxidase from Escherichia coli, we examined reactions with ubiquinol-1 and dioxygen. Stopped-flow studies showed that anaerobic reduction of the wild-type and the bound ubiquinone-free (DeltaUbiA) enzymes with ubiquinol-1 immediately takes place with four kinetic phases. Replacement of the bound ubiquinone with 2,6-dibromo-4-cyanophenol (PC32) suppressed the anaerobic reduction of the hemes with ubiquinol-1 by eliminating the fast phase. Flow-flash studies in the reaction of the fully reduced enzyme with dioxygen showed that the heme b-to-heme o electron transfer occurs with a rate constant of approximately 1x10(4) s(-1) in all three preparations. These results support our previous proposal that the bound ubiquinone is involved in facile oxidation of substrates in subunit II and subsequent intramolecular electron transfer to low-spin heme b in subunit I.


Assuntos
Grupo dos Citocromos b , Citocromos/metabolismo , Proteínas de Escherichia coli , Escherichia coli/metabolismo , Oxigênio/metabolismo , Ubiquinona/análogos & derivados , Ubiquinona/metabolismo , Transporte de Elétrons , Escherichia coli/enzimologia , Oxirredução
11.
J Biochem ; 126(1): 98-103, 1999 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10393326

RESUMO

Cytochrome bd-type ubiquinol oxidase in the aerobic respiratory chain of Escherichia coli contains two hemes b (b558 and b595) and one heme d as redox metal centers. To clarify the structure of the reaction center, we analyzed the fully oxidized enzyme by visible and EPR spectroscopies using fluoride ion as a monitoring probe. The visible spectral changes upon fluoride-binding were typical of ferric iron-chlorine species, indicating heme d as a primary binding site. The negative peak at 645 nm in the difference spectrum indicates that heme b595 also provides the low-affinity fluoride-binding site. Fluoride-binding caused a complete disappearance from the EPR spectra of the low-spin signals ascribable to heme d and spectral changes in both rhombic and axial high-spin signals. After fluoride-binding, each component of the rhombic high-spin signal showed superhyperfine splitting arising from the interaction of the unpaired spin of the heme d iron with the nuclear magnetic moment of 19F. The axial high-spin species was converted to a new rhombic high-spin species assignable to heme b595-fluoride. The g = 2 component of this new species also gave 19F-superhyperfine splitting. These results indicate that both heme d and heme b595 can coordinate with a fluoride ion with different affinities in the fully oxidized state.


Assuntos
Citocromos/química , Citocromos/metabolismo , Complexo de Proteínas da Cadeia de Transporte de Elétrons , Proteínas de Escherichia coli , Escherichia coli/enzimologia , Fluoretos/metabolismo , Oxirredutases/química , Oxirredutases/metabolismo , Sítios de Ligação , Domínio Catalítico , Grupo dos Citocromos b , Espectroscopia de Ressonância de Spin Eletrônica/métodos , Análise Espectral/métodos
12.
J Biochem ; 126(1): 194-9, 1999 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10393339

RESUMO

Cytochrome bo is the heme-copper terminal ubiquinol oxidase in the aerobic respiratory chain of Escherichia coli, and functions as a redox-coupled proton pump. As an extension to our mutagenesis and Fourier-transform infrared studies on ion pumps, we examined the effects of subunit I mutations on redox-linked protein structural changes in cytochrome bo. Upon photo-reduction in the presence of riboflavin, Y288F and H333A showed profound effects in their peptide backbone vibrations (amide-I and amide-II), probably due to the loss of CuB or replacement of high-spin heme o with heme B. In the frequency region of protonated carboxylic C=O stretching vibrations, negative 1,743 cm-1 and positive 1,720 cm-1 bands were observed in the wild-type; the former shifted to 1,741 cm-1 in E286D but not in other mutants including D135N. This suggests that Glu286 in the D-channel is protonated in the air-oxidized state and undergoes hydrogen bonding changes upon reduction of the redox metal centers. Two pairs of band shifts at 2,566 (+)/2,574 (-) and 2,546 (+)/2,556 (-) cm-1 in all mutants indicate that two cysteine residues not in the vicinity of the metal centers undergo redox-linked hydrogen bonding changes. Cyanide had no effect on the protein structural changes because of the rigid local protein structure around the binuclear center or the presence of a ligand(s) at the binuclear center, and was released from the binuclear center upon reduction. This study establishes that cytochrome bo undergoes unique redox-linked protein structural changes. Localization and time-resolved analysis of the structural changes during dioxygen reduction will facilitate understanding of the molecular mechanism of redox-coupled proton pumping at the atomic level.


Assuntos
Grupo dos Citocromos b , Citocromos/química , Citocromos/genética , Proteínas de Escherichia coli , Escherichia coli/enzimologia , Espectroscopia de Infravermelho com Transformada de Fourier/métodos , Sítios de Ligação , Cobre/metabolismo , Cianetos/metabolismo , Citocromos/metabolismo , Ácido Glutâmico , Heme , Mutação , Oxirredução , Conformação Proteica , Riboflavina/metabolismo , Análise Espectral/métodos
13.
J Biochem ; 125(6): 1131-6, 1999 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10348916

RESUMO

Cytochrome bd is a two-subunit ubiquinol oxidase in the aerobic respiratory chain of Escherichia coli that does not belong to the heme-copper terminal oxidase superfamily. To explore unique protein structural changes associated with the reduction of the redox metal centers, we carried out Fourier-transform infrared and visible spectroscopic studies on cytochrome bd. For infrared measurements of a partially dehydrated thin sample solution, the air-oxidized enzyme was fully reduced by the intermolecular electron transfer of photo-excited riboflavin in the absence and presence of KCN, and redox difference spectra were calculated. Upon reduction, the bound cyanide was released from the heme b595-heme d binuclear center but remained in a protein pocket as a deprotonated form. Reduction of heme b558, heme b595, and heme d resulted in large changes in amide-I and protonated carboxylic CO-stretching vibrations and also a small change in the cysteine SH-stretching vibration. The location of the redox metal centers and the effects of cyanide suggest that these protein structural changes occur at the heme-binding pockets near the protein surface. Systematic site-directed mutagenesis and time-resolved FTIR studies on cytochrome bd will facilitate an understanding of the unique molecular mechanisms for dioxygen reduction and delivery of chemical protons to the active center at the atomic level.


Assuntos
Citocromos/química , Complexo de Proteínas da Cadeia de Transporte de Elétrons , Proteínas de Escherichia coli , Escherichia coli/enzimologia , Oxirredutases/química , Sítios de Ligação , Grupo dos Citocromos b , Citocromos/efeitos da radiação , Heme/química , Metais/química , Oxirredução , Oxirredutases/efeitos da radiação , Fotoquímica , Conformação Proteica , Espectrofotometria , Espectroscopia de Infravermelho com Transformada de Fourier
14.
FEBS Lett ; 449(2-3): 191-5, 1999 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-10338130

RESUMO

Azide-binding to the heme-copper binuclear center of bo-type ubiquinol oxidase from Escherichia coli was investigated with Fourier-transform infrared spectroscopy. Deconvolution analyses of infrared spectra of the azide (14N3)-inhibited air-oxidized form showed a major infrared azide antisymmetric stretching band at 2041 cm(-1). An additional band developed at 2062.5 cm(-1) during a longer incubation. Isotope substitutions with terminally 15N-labelled azides did not show a splitting of the major band, indicating that the geometry of the bound azide is mainly in a bridging configuration between high-spin heme o and CuB. The band at 2062.5 cm(-1) showed clear splittings upon substitution with the terminally 15N-labelled azides, indicating the Cu(2+)B-N=N=N structure. Partial reduction of the oxidase with beta-NADH in the presence of azide caused an appearance of new infrared bands at 2038.5 (major) and 2009 (minor) cm(-1). The former band also showed clear splittings in the presence of the terminally 15N-labelled azides, indicating that reduction of low-spin heme b alters the structure of the binuclear center leading to the Fe(3+)o-N=N=N configuration.


Assuntos
Azidas/metabolismo , Complexo IV da Cadeia de Transporte de Elétrons/metabolismo , Escherichia coli/enzimologia , NAD/metabolismo , Espectroscopia de Infravermelho com Transformada de Fourier
15.
Biochemistry ; 38(18): 5913-7, 1999 May 04.
Artigo em Inglês | MEDLINE | ID: mdl-10231544

RESUMO

Cytochrome bd is a two-subunit ubiquinol oxidase in the aerobic respiratory chain of Escherichia coli and binds hemes b558, b595, and d as the redox metal centers. Taking advantage of spectroscopic properties of three hemes which exhibit distinct absorption peaks, we investigated electron transfer within the enzyme by the technique of pulse radiolysis. Reduction of the hemes in the air-oxidized, resting-state enzyme, where heme d exists in mainly an oxygenated form and partially an oxoferryl and a ferric low-spin forms, occurred in two phases. In the faster phase, radiolytically generated N-methylnicotinamide radicals simultaneously reduced the ferric hemes b558 and b595 with a second-order rate constant of 3 x 10(8) M-1 s-1, suggesting that a rapid equilibrium occurs for electron transfer between two b-type hemes long before 10 micros. In the slower phase, an intramolecular electron transfer from heme b to the oxoferryl and the ferric heme d occurred with the first-order rate constant of 4.2-5.6 x 10(2) s-1. In contrast, the oxygenated heme d did not exhibit significant spectral change. Reactions with the fully oxidized and hydrogen peroxide-treated forms demonstrated that the oxidation and/or ligation states of heme d do not affect the heme b reduction. The following intramolecular electron transfer transformed the ferric and oxoferryl forms of heme d to the ferrous and ferric forms, respectively, with the first-order rate constants of 3.4 x 10(3) and 5.9 x 10(2) s-1, respectively.


Assuntos
Citocromos/química , Citocromos/metabolismo , Complexo de Proteínas da Cadeia de Transporte de Elétrons , Proteínas de Escherichia coli , Escherichia coli/enzimologia , Oxirredutases/química , Oxirredutases/metabolismo , Grupo dos Citocromos b , Transporte de Elétrons , Heme/química , Peróxido de Hidrogênio/química , Oxirredução , Radiólise de Impulso/métodos , Fatores de Tempo
16.
J Biochem ; 125(2): 270-6, 1999 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-9990123

RESUMO

We constructed a time-resolved photovoltage measurement system and examined the photovoltage kinetics of wild-type bacteriorhodopsin, its D96N mutant, and halorhodopsins from Halobacterium salinarum and Natronobacterium pharaonis. Upon illumination with a laser flash, wild-type bacteriorhodopsin showed photovoltage generation with fast (10-100 micros range) and slow (ms range) components while D96N lacked the latter, as reported previously [Holz, M., Drachev, L.A., Mogi, T., Otto, H., Kaulen, A.D., Heyn, M.P., Skulachev, V.P., and Khorana, H.G. (1989) Proc. Natl. Acad. Sci. USA 86, 2167-2171]. In contrast, photovoltage generation in halorhodopsins from H. salinarum and N. pharaonis was significant only in the ms time range. On the basis of the photovoltage kinetics and photocycle, we conclude that major charge (chloride) movements within halorhodopsin occur during the formation and decay of the N intermediate in the ms range. These observations are discussed in terms of the "Energization-Relaxation Channel Model" [Muneyuki, E., Ikematsu, M., and Yoshida, M. (1996) J. Phys. Chem. 100, 19687-19691].


Assuntos
Bacteriorodopsinas/química , Fotoquímica/métodos , Halobacterium/química , Halorrodopsinas , Cinética , Filmes Cinematográficos , Natronobacterium/química , Polímeros/química , Fatores de Tempo
18.
J Biochem ; 125(1): 138-42, 1999 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-9880809

RESUMO

Natural aurachin C is the most potent inhibitor of oxidation of ubiquinols by cytochromes bo and bd from Escherichia coli. To probe the structural properties of the substrate oxidation site in the ubiquinol oxidases, we synthesized a systematic set of aurachin C analogues (N-hydroxy-4-quinolone derivatives) and examined how their structure affects their activity towards cytochromes bo and bd, which are structurally unrelated. We found that the presence of the 3-methyl group of the 2-n-decyl and 2-n-undecyl derivatives increased the inhibitory potency towards both enzymes, probably due to a local steric congestion that allows favorable interaction of the alkyl tail with the enzyme. Increase in the chain length of the 3-alkyl tail of the 2-n-undecyl derivatives decreased the inhibitory potency only in cytochrome bo, indicating that the binding site for the alkyl tails of cytochrome bo is smaller than that of cytochrome bd. Based on these findings, we discuss the differences in the molecular mechanism of substrate oxidation by these two terminal ubiquinol oxidases.


Assuntos
Grupo dos Citocromos b , Citocromos/metabolismo , Complexo de Proteínas da Cadeia de Transporte de Elétrons , Inibidores Enzimáticos/química , Proteínas de Escherichia coli , Escherichia coli/enzimologia , Oxirredutases/metabolismo , Sítios de Ligação , Citocromos/antagonistas & inibidores , Citocromos/genética , Inibidores Enzimáticos/farmacologia , Mutação , Oxirredução , Oxirredutases/antagonistas & inibidores , Quinolonas/química , Quinolonas/farmacologia , Relação Estrutura-Atividade , Ubiquinona/análogos & derivados , Ubiquinona/metabolismo
19.
Int Arch Occup Environ Health ; 71 Suppl: S33-6, 1998 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-9827877

RESUMO

OBJECTIVES: To summarize the results of two studies that attempted to clarify: (1) the health effects of hydrazine hydrate (HH) (N2H4 x H2O: CAS No. 7803-57-8); and (2) the influence of allelic polymorphism of N-acetyltransferase (NAT2) on the metabolism of HH. METHODS: A cross-sectional survey was carried out on 172 male HH-exposed workers and 125 male referent workers at five factories in Japan. The biological half-lives of HH after 1 h of exposure were determined in 12 workers, four workers in each of three NAT2 phenotypes. Clinical examinations were performed and acute and chronic subjective symptoms related to HH were examined by self-administered questionnaires. NAT2 phenotypes were assessed. RESULTS: No hydrazine was detected in either the breathing zones or the urine of the referent workers. The mean hydrazine concentration in the breathing zones, hydrazine and acetylhydrazine in urine, and the cumulative exposure level were 0.0109 ppm, 0.8660 micromol/g x Cr, and 2.80 ppm-years, respectively. There was no difference and no dose-dependent change in the health examination items between HH-exposed and referent workers after adjusting confounding factors, nor in terms of the differences of NAT2 phenotypes. Of 90 subjective symptoms, complaints of nightmares were significantly related to HH exposure. The half-life of urinary hydrazine and acetylhydrazine on rapid, intermediate, and slow phenotypes was 1.68, 3.01, and 4.46 h, respectively. CONCLUSION: This study suggested that current and cumulative exposure to HH did not affect the workers' health, and the half-life of the slow phenotype was longer than those of the rapid and intermediate phenotypes.


Assuntos
Hidrazinas , Adolescente , Adulto , Arilamina N-Acetiltransferase , Estudos Transversais , Humanos , Hidrazinas/metabolismo , Masculino , Pessoa de Meia-Idade , Fenótipo , Polimorfismo Genético
20.
Biochemistry ; 37(43): 15106-13, 1998 Oct 27.
Artigo em Inglês | MEDLINE | ID: mdl-9790673

RESUMO

The hydrophobic isoprene tail of ubiquinone-2 (Q2) exihibits binding specificity in redox reactions with bovine heart mitochondrial complex I (Ohshima, M., Miyoshi, H., Sakamoto, K., Takegami, K., Iwata, J., Kuwabara, K., Iwamura, H., and Yagi, T. (1998) Biochemistry 37, 6436-6445) and the Escherichia coli bo-type ubiquinol oxidase (Sakamoto, K., Miyoshi, H., Takegami, K., Mogi, T., Anraku, Y., and Iwamura, H. (1996) J. Biol. Chem. 271, 29897-29902). To identify the structural factor(s) of the diprenyl tail of Q2 governing the specific interaction with these enzymes, we synthesized a series of novel Q2 analogues in which only one of the structural factors of the diprenyl tail was systematically modified. In bovine complex I, the presence of the methyl branch and the pi-electron system in the first isoprene unit are responsible for high-affinity binding of Q2 to the ubiquinone reduction site, which results in a low Km and kcat values of Q2 reduction. The position of the methyl group in the tail is strictly recognized by the enzyme. In contrast to complex I, in bo-type ubiquinol oxidase, either of the two pi-electron systems in the tail is required for high-affinity binding of Q2H2 to the enzyme, while the presence of the methyl branch and the location of the pi-electron systems are not strictly recognized by the enzyme. We concluded that the role of the ubiquinone tail is not simply the enhancement of the hydrophobicity of the molecule and that molecular recognition of the tail by the quinone redox site differs among the respiratory enzymes.


Assuntos
Complexo IV da Cadeia de Transporte de Elétrons/metabolismo , Escherichia coli/enzimologia , Mitocôndrias Cardíacas/enzimologia , NAD(P)H Desidrogenase (Quinona)/metabolismo , Prenilação de Proteína , Ubiquinona/metabolismo , Animais , Sítios de Ligação , Bovinos , Transporte de Elétrons , Cinética , Oxirredução , Conformação Proteica , Ubiquinona/análogos & derivados , Ubiquinona/síntese química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...