Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 231
Filtrar
1.
Science ; 366(6461): 100-104, 2019 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-31604309

RESUMEN

Cytochrome bd-type quinol oxidases catalyze the reduction of molecular oxygen to water in the respiratory chain of many human-pathogenic bacteria. They are structurally unrelated to mitochondrial cytochrome c oxidases and are therefore a prime target for the development of antimicrobial drugs. We determined the structure of the Escherichia coli cytochrome bd-I oxidase by single-particle cryo-electron microscopy to a resolution of 2.7 angstroms. Our structure contains a previously unknown accessory subunit CydH, the L-subfamily-specific Q-loop domain, a structural ubiquinone-8 cofactor, an active-site density interpreted as dioxygen, distinct water-filled proton channels, and an oxygen-conducting pathway. Comparison with another cytochrome bd oxidase reveals structural divergence in the family, including rearrangement of high-spin hemes and conformational adaption of a transmembrane helix to generate a distinct oxygen-binding site.


Asunto(s)
Grupo Citocromo b/química , Proteínas del Complejo de Cadena de Transporte de Electrón/química , Proteínas de Escherichia coli/química , Escherichia coli/enzimología , Oxidorreductasas/química , Dominio Catalítico , Microscopía por Crioelectrón , Hemo/química , Modelos Moleculares , Oxidación-Reducción , Oxígeno/química , Estructura Cuaternaria de Proteína , Subunidades de Proteína/química , Protones , Ubiquinona/química
2.
Biochim Biophys Acta Bioenerg ; 1859(2): 110-118, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29107655

RESUMEN

Microcin J25 has two targets in sensitive bacteria, the RNA polymerase, and the respiratory chain through inhibition of cellular respiration. In this work, the effect of microcin J25 in E. coli mutants that lack the terminal oxidases cytochrome bd-I and cytochrome bo3 was analyzed. The mutant strains lacking cytochrome bo3 or cytochrome bd-I were less sensitive to the peptide. In membranes obtained from the strain that only expresses cytochrome bd-I a great ROS overproduction was observed in the presence of microcin J25. Nevertheless, the oxygen consumption was less inhibited in this strain, probably because the oxygen is partially reduced to superoxide. There was no overproduction of ROS in membranes isolated from the mutant strain that only express cytochrome bo3 and the inhibition of the cellular respiration was similar to the wild type. It is concluded that both cytochromes bd-I and bo3 are affected by the peptide. The results establish for the first time a relationship between the terminal oxygen reductases and the mechanism of action of microcin J25.


Asunto(s)
Bacteriocinas/farmacología , Citocromos/biosíntesis , Proteínas del Complejo de Cadena de Transporte de Electrón/biosíntesis , Proteínas de Escherichia coli/biosíntesis , Escherichia coli/enzimología , Regulación Bacteriana de la Expresión Génica/efectos de los fármacos , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Oxidorreductasas/biosíntesis , Grupo Citocromo b , Citocromos/genética , Proteínas del Complejo de Cadena de Transporte de Electrón/genética , Escherichia coli/genética , Proteínas de Escherichia coli/genética , Oxidorreductasas/genética , Especies Reactivas de Oxígeno/metabolismo
3.
Appl Environ Microbiol ; 80(16): 4871-8, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24907324

RESUMEN

Reduction of NO to N2O by denitrifiying bacteria is catalyzed either by a monomeric quinol-nitric oxide reductase (qNor) or by a heterodimeric cytochrome c-dependent nitric oxide reductase (cNor). In ancient thermophilic bacteria belonging to the Thermales and Aquificales phylogenetic groups, the cluster encoding the cNor includes a small third gene (norH), in addition to those encoding homologues to the subunits of a typical cNor (norC and norB). We show in Thermus thermophilus that the three genes are cotranscribed in a single mRNA from an inducible promoter. The isolation of individual nor mutants and the production in vivo of His-tagged NorH protein followed by immobilized-metal affinity chromatography (IMAC) allowed us to conclude that NorH constitutes a third subunit of the cNor from T. thermophilus, which is involved in denitrification in vivo, likely allowing more efficient electron transport to cNor.


Asunto(s)
Proteínas Bacterianas/metabolismo , Citocromos c/metabolismo , Oxidorreductasas/metabolismo , Subunidades de Proteína/metabolismo , Thermus thermophilus/enzimología , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Óxido Nítrico/metabolismo , Operón , Oxidorreductasas/química , Oxidorreductasas/genética , Regiones Promotoras Genéticas , Subunidades de Proteína/química , Subunidades de Proteína/genética , Homología de Secuencia de Aminoácido , Thermus thermophilus/química , Thermus thermophilus/genética , Thermus thermophilus/metabolismo
4.
Biochemistry ; 40(32): 9695-708, 2001 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-11583170

RESUMEN

The reaction of cytochrome c oxidase (COX) from Rhodobacter sphaeroides with hydrogen peroxide has been studied at alkaline (pH 8.5) and acidic (pH 6.5) conditions with the aid of a stopped-flow apparatus. Absorption changes in the entire 350-800 nm spectral range were monitored and analyzed by a global fitting procedure. The reaction can be described by the sequential formation of two intermediates analogous to compounds I and II of peroxidases: oxidized COX + H2O2 --> intermediate I --> intermediate II. At pH as high as 8.5, intermediate I appears to be a mixture of at least two species characterized by absorption bands at approximately 607 nm (P607) and approximately 580 nm (F-I580) that rise synchronously. At acidic pH (6.5), intermediate I is represented mainly by a component with an alpha-peak around 575 nm (F-I575) that is probably equivalent to the so-called F* species observed with the bovine COX. The data are consistent with a pH-dependent reaction branching at the step of intermediate I formation. To get further insight into the mechanism of the pH-dependence, the peroxide reaction was studied using two mutants of the R. sphaeroides oxidase, K362M and D132N, that block, respectively, the proton-conducting K- and D-channels. The D132N mutation does not affect significantly the Ox --> intermediate I step of the peroxide reaction. In contrast, K362M replacement exerts a dramatic effect, eliminating the pH-dependence of intermediate I formation. The data obtained allow us to propose that formation of the acidic form of intermediate I (F-I575, F*) requires protonation of some group at/near the binuclear site that follows or is concerted with peroxide binding. The protonation involves specifically the K-channel. Presumably, a proton vacancy can be generated in the site as a consequence of the proton-assisted heterolytic scission of the O-O bond of the bound peroxide. The results are consistent with a proposal [Vygodina, T. V., Pecoraro, C., Mitchell, D., Gennis, R., and Konstantinov, A. A. (1998) Biochemistry 37, 3053-3061] that the K-channel may be involved in the delivery of the first four protons in the catalytic cycle (starting from reduction of the oxidized form) including proton uptake coupled to reduction of the binuclear site and transfer of protons driven by cleavage of the dioxygen O-O bond in the binculear site. Once peroxide intermediate I has been formed, generation of a strong oxene ligand at the heme a3 iron triggers a transition of the enzyme to the "peroxidase conformation" in which the K-channel is closed and the binuclear site becomes protonically disconnected from the bulk aqueous phase.


Asunto(s)
Complejo IV de Transporte de Electrones/metabolismo , Peróxido de Hidrógeno/metabolismo , Canales de Potasio/metabolismo , Animales , Complejo IV de Transporte de Electrones/genética , Concentración de Iones de Hidrógeno , Cinética , Modelos Químicos , Mutación , Oxidantes/metabolismo , Rhodobacter sphaeroides/enzimología , Análisis Espectral
5.
FEBS Lett ; 505(1): 63-7, 2001 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-11557043

RESUMEN

Attenuated total reflection (ATR) spectroscopy brings an added dimension to studies of structural changes of cytochrome c oxidase (CcO) because it enables the recording of reaction-induced infrared difference spectra under a wide variety of controlled conditions (e.g. pH and chemical composition), without relying on light or potentiometric changes to trigger the reaction. We have used the ATR method to record vibrational difference spectra of CcO with reduction induced by flow-exchange of the aqueous buffer. Films of CcO prepared from Rhodobacter sphaeroides and beef heart mitochondria by reconstitution with lipid were adhered to the internal reflection element of the ATR device and retained their full functionality as evidenced by visible spectroscopy and time-resolved vibrational spectroscopy. These results demonstrate that the technique of perfusion-induced Fourier-transform infrared difference spectroscopy can be successfully applied to a large, complex enzyme, such as CcO, with sufficient signal/noise to probe vibrational changes in individual residues of the enzyme under various conditions.


Asunto(s)
Complejo IV de Transporte de Electrones/química , Complejo IV de Transporte de Electrones/metabolismo , Espectroscopía Infrarroja por Transformada de Fourier/métodos , Animales , Bovinos , Enzimas Inmovilizadas/química , Enzimas Inmovilizadas/metabolismo , Mitocondrias Cardíacas/enzimología , Oxidación-Reducción , Perfusión , Fosfolípidos/química , Rhodobacter sphaeroides/enzimología , Vibración
6.
J Biol Chem ; 276(36): 33616-20, 2001 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-11443122

RESUMEN

The final step in the catalytic cycle of cytochrome oxidase, the reduction of oxyferryl heme a(3) in compound F, was investigated using a binuclear polypyridine ruthenium complex ([Ru(bipyridine)(2)](2)(1,4-bis[2-(4'-methyl-2, 2'-bipyrid-4-yl)ethenyl]benzene)(PF(6))(4)) as a photoactive reducing agent. In the untreated dimeric enzyme, the rate constant for reduction of compound F decreased from 700 s(-1) to 200 s(-1) as the pH was increased from 7.5 to 9.5. Incubation of dimeric enzyme at pH 10 led to an increase in the rate constant to 1650 s(-1), which was independent of pH between pH 7.4 and 10. This treatment resulted in a decrease in the sedimentation coefficient consistent with the irreversible conversion of the enzyme to a monomeric form. Similar results were obtained when the enzyme was incubated with Triton X-100 at pH 8.0. These treatments, which have traditionally been used to convert dimeric enzyme to monomeric form, have no effect on the steady-state activity. The data indicate that either the conversion of the bovine oxidase to a monomeric form or some structural change coincident with this conversion strongly influences the rate constant of this step in the catalytic cycle, perhaps by influencing the proton access to the heme-copper binuclear center.


Asunto(s)
Detergentes/farmacología , Complejo IV de Transporte de Electrones/química , Complejo IV de Transporte de Electrones/metabolismo , Octoxinol/farmacología , Animales , Catálisis , Bovinos , Dimerización , Transporte de Electrón , Concentración de Iones de Hidrógeno , Cinética , Luz , Modelos Químicos , Fotólisis , Protones , Factores de Tiempo , Ultracentrifugación
7.
Biochemistry ; 40(29): 8548-56, 2001 Jul 24.
Artículo en Inglés | MEDLINE | ID: mdl-11456494

RESUMEN

Cytochrome bd is one of the two quinol oxidases in the respiratory chain of Escherichia coli. The enzyme contains three heme prosthetic groups. The dioxygen binding site is heme d, which is thought to be part of the heme-heme binuclear center along with heme b(595), which is a high-spin heme whose function is not known. Protein sequence alignments [Osborne, J. P., and Gennis, R. B. (1999) Biochim. Biophys Acta 1410, 32--50] of cytochrome bd quinol oxidase sequences from different microorganisms have revealed a highly conserved sequence (GWXXXEXGRQPW; bold letters indicate strictly conserved residues) predicted to be on the periplasmic side of the membrane between transmembrane helices 8 and 9 in subunit I. The functional importance of this region is investigated in the current work by site-directed mutagenesis. Several mutations in this region (W441A, E445A/Q, R448A, Q449A, and W451A) resulted in a catalytically inactive enzyme with abnormal UV--vis spectra. E445A was selected for detailed analysis because of the absence of the absorption bands from heme b(595). Detailed spectroscopic and chemical analyses, indeed, show that one of the three heme prosthetic groups in the enzyme, heme b(595), is specifically perturbed and mostly missing from this mutant. Surprisingly, heme d, while known to interact with heme b(595), appears relatively unperturbed, whereas the low-spin heme b(558) shows some modification. This is the first report of a mutation that specifically affects the binding site of heme b(595).


Asunto(s)
Citocromos/genética , Proteínas del Complejo de Cadena de Transporte de Electrón , Proteínas de Escherichia coli , Escherichia coli/enzimología , Hemo/análogos & derivados , Hemo/química , Mutagénesis Sitio-Dirigida , Oxidorreductasas/genética , Alanina/genética , Secuencia de Aminoácidos , Monóxido de Carbono/química , Secuencia Conservada/genética , Cianuros/química , Grupo Citocromo b , Citocromos/química , Electroquímica , Espectroscopía de Resonancia por Spin del Electrón , Escherichia coli/genética , Ácido Glutámico/genética , Datos de Secuencia Molecular , Oxidación-Reducción , Oxidorreductasas/química , Oxidorreductasas N-Desmetilantes/química , Unión Proteica/genética , Estructura Terciaria de Proteína/genética , Quinona Reductasas/química , Espectrofotometría Ultravioleta , Espectroscopía Infrarroja por Transformada de Fourier , Espectrometría Raman
8.
Mol Genet Genomics ; 265(3): 394-404, 2001 May.
Artículo en Inglés | MEDLINE | ID: mdl-11405622

RESUMEN

Heme molecules play important roles in electron transfer by redox proteins such as cytochromes. In addition, a structural role for heme in protein folding and the assembly of enzymes has been suggested. Previous results obtained using Escherichia coli hemA mutants, which are unable to synthesize 5-aminolevulinic acid, a precursor of porphyrins and hemes, have demonstrated a requirement for heme biosynthesis in the assembly of a functional succinate-ubiquinone reductase (SQR or complex II), which is a component of the aerobic respiratory chain. In the present study, in order to investigate the role of the heme in the assembly of E. coli SQR, we used a hemH (encodes ferrochelatase) mutant that lacks the ability to insert iron into the porphyrin ring. The hemH mutant failed to insert functional SQR into the cytoplasmic membrane, and the catalytic portion of SQR [the flavoprotein subunit (Fp) and the iron-sulfur protein subunit (Ip)] was localized in the cytoplasm of the cell. It is of interest to note that protoporphyrin IX accumulated in the mutant cells and inactivated the cytoplasmic succinate dehydrogenase (SDH) activity associated with the catalytic Fp-Ip complex. In contrast, SQR was assembled into the membrane of a heme-permeable hemH double mutant when hemin was present in the culture. Only a low level of SQR activity was found in the membrane when hemin was replaced by non-iron metalloporphyrins: Mn-, Co-, Ni-, Zn- and Cu-protoporphyrin IX, or protoporphyrin IX These results indicate that heme iron is indispensable for the functional assembly of SQR in the cytoplasmic membrane of E. coli, and provide a new insight into the biological role of heme in the molecular assembly of the multi-subunit enzyme complex.


Asunto(s)
Escherichia coli/enzimología , Ferroquelatasa/genética , Complejos Multienzimáticos/química , Complejos Multienzimáticos/genética , Mutación , Oxidorreductasas/química , Oxidorreductasas/genética , Succinato Deshidrogenasa/química , Succinato Deshidrogenasa/genética , Membrana Celular/metabolismo , Grupo Citocromo b/metabolismo , Citoplasma/metabolismo , Relación Dosis-Respuesta a Droga , Complejo II de Transporte de Electrones , Hemo/química , Hemina/metabolismo , Modelos Biológicos , Porfirinas/metabolismo , Factores de Tiempo
9.
Biophys J ; 80(5): 2039-45, 2001 May.
Artículo en Inglés | MEDLINE | ID: mdl-11325707

RESUMEN

The resonance Raman spectra of the aa3 cytochrome c oxidase from Rhodobacter sphaeroides reveal pH-dependent structural changes in the binuclear site at room temperature. The binuclear site, which is the catalytic center of the enzyme, possesses two conformations at neutral pH, assessed from their distinctly different Fe-CO stretching modes in the resonance Raman spectra of the CO complex of the fully reduced enzyme. The two conformations (alpha and beta) interconvert reversibly in the pH 6-9 range with a pKa of 7.4, consistent with Fourier transform infrared spectroscopy measurements done at cryogenic temperatures (D.M. Mitchell, J.P. Sapleigh, A.M.Archer, J.O. Alben, and R.B.Gennis, 1996, Biochemistry 35:9446-9450). It is postulated that the different structures result from a change in the position of the Cu(B) atom with respect to the CO due to the presence of one or more ionizable groups in the vicinity of the binuclear center. The conserved tyrosine residue (Tyr-288 in R. sphaeroides, Tyr-244 in the bovine enzyme) that is adjacent to the oxygen-binding pocket or one of the histidines that coordinate Cu(B) are possible candidates. The existence of an equilibrium between the two conformers at physiological pH and room temperature suggests that the conformers may be functionally involved in enzymatic activity.


Asunto(s)
Cobre/química , Complejo IV de Transporte de Electrones/química , Hemo/química , Animales , Sitios de Unión , Dominio Catalítico , Bovinos , Complejo IV de Transporte de Electrones/aislamiento & purificación , Concentración de Iones de Hidrógeno , Modelos Moleculares , Conformación Proteica , Rhodobacter sphaeroides/enzimología , Espectroscopía Infrarroja por Transformada de Fourier , Espectrometría Raman , Temperatura , Tirosina/química
10.
Proc Natl Acad Sci U S A ; 98(9): 5013-8, 2001 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-11296255

RESUMEN

Cytochrome c oxidase is a membrane-bound enzyme that catalyzes the four-electron reduction of oxygen to water. This highly exergonic reaction drives proton pumping across the membrane. One of the key questions associated with the function of cytochrome c oxidase is how the transfer of electrons and protons is coupled and how proton transfer is controlled by the enzyme. In this study we focus on the function of one of the proton transfer pathways of the R. sphaeroides enzyme, the so-called K-proton transfer pathway (containing a highly conserved Lys(I-362) residue), leading from the protein surface to the catalytic site. We have investigated the kinetics of the reaction of the reduced enzyme with oxygen in mutants of the enzyme in which a residue [Ser(I-299)] near the entry point of the pathway was modified with the use of site-directed mutagenesis. The results show that during the initial steps of oxygen reduction, electron transfer to the catalytic site (to form the "peroxy" state, P(r)) requires charge compensation through the proton pathway, but no proton uptake from the bulk solution. The charge compensation is proposed to involve a movement of the K(I-362) side chain toward the binuclear center. Thus, in contrast to what has been assumed previously, the results indicate that the K-pathway is used during oxygen reduction and that K(I-362) is charged at pH approximately 7.5. The movement of the Lys is proposed to regulate proton transfer by "shutting off" the protonic connectivity through the K-pathway after initiation of the O(2) reduction chemistry. This "shutoff" prevents a short-circuit of the proton-pumping machinery of the enzyme during the subsequent reaction steps.


Asunto(s)
Complejo IV de Transporte de Electrones/metabolismo , Rhodobacter sphaeroides/enzimología , Sustitución de Aminoácidos/genética , Sitios de Unión , Transporte Biológico , Transporte de Electrón , Complejo IV de Transporte de Electrones/química , Complejo IV de Transporte de Electrones/genética , Electrones , Hemo/análogos & derivados , Hemo/metabolismo , Enlace de Hidrógeno , Lisina/metabolismo , Modelos Moleculares , Mutación/genética , Oxígeno/química , Oxígeno/metabolismo , Fotólisis , Conformación Proteica , Protones , Rhodobacter sphaeroides/genética , Análisis Espectral , Electricidad Estática
11.
FEBS Lett ; 492(1-2): 45-9, 2001 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-11248234

RESUMEN

The Na(+)-translocating NADH:quinone oxidoreductase (Na(+)-NQR) is present in the membranes of a number of marine bacteria and pathogenic bacteria. Two of the six subunits of the Na(+)-NQR, NqrB and NqrC, have been previously shown to contain covalently bound flavin adenine mononucleotide (FMN). In the current work, the cloning of nqrC from Vibrio cholerae is reported. The gene has been expressed in V. cholerae and shown to contain one equivalent of covalently bound FMN. In contrast, no covalent flavin was detected when threonine-225 was replaced by leucine. The data show that the FMN attachment does not require assembly of the enzyme and are consistent with the unusual threonine attachment site.


Asunto(s)
Mononucleótido de Flavina/metabolismo , Quinona Reductasas/genética , Vibrio cholerae/genética , Secuencia de Aminoácidos , Proteínas Bacterianas/genética , Proteínas Bacterianas/aislamiento & purificación , Proteínas Bacterianas/metabolismo , Expresión Génica , Histidina/genética , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Quinona Reductasas/biosíntesis , Quinona Reductasas/metabolismo , Treonina/genética , Vibrio cholerae/metabolismo
12.
Biochemistry ; 40(4): 1077-82, 2001 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-11170431

RESUMEN

Aspartate-75 (D75) was recently suggested to participate in a ubiquinone-binding site in subunit I of cytochrome bo(3) from Escherichia coli on the basis of a structural model [Abramson, J., Riistama, S., Larsson, G., Jasaitis, A., Svensson-Ek, M., Laakkonen, L., Puustinen, A., Iwata, S., and Wikström, M. (2000) Nat. Struct. Biol. 7 (10), 910-917]. We studied the protonation state of D75 for the reduced and oxidized forms of the enzyme, using a combined site-directed mutagenesis, electrochemical, and FTIR spectroscopic approach. The D75H mutant is catalytically inactive, whereas the more conservative D75E substitution has quinol oxidase activity equal to that of the wild-type enzyme. Electrochemically induced FTIR difference spectra of the inactive D75H mutant enzyme show a clear decrease in the spectroscopic region characteristic of protonated aspartates and glutamates. Strong variations in the amide I region of the FTIR difference spectrum, however, reflect a more general perturbation due to this mutation of both the protein and the bound quinone. Electrochemically induced FTIR difference spectra on the highly conservative D75E mutant enzyme show a shift from 1734 to 1750 cm(-1) in direct comparison to wild type. After H/D exchange, the mode at 1750 cm(-1) shifts to 1735 cm(-1). These modes, concomitant with the reduced state of the enzyme, can be assigned to the nu(C=O) vibrational mode of protonated D75 and E75, respectively. In the spectroscopic region where signals for deprotonated acidic groups are expected, band shifts for the nu(COO(-))(s/as) modes from 1563 to 1554-1539 cm(-1) and from 1315 to 1336 cm(-1), respectively, are found for the oxidized enzyme. These signals indicate that D75 (or E75 in the mutant) is deprotonated in the oxidized form of cytochrome bo(3) and is protonated upon full reduction of the enzyme. It is suggested that upon reduction of the bound ubiquinone at the high affinity site, D75 takes up a proton, possibly sharing it with ubiquinol.


Asunto(s)
Ácido Aspártico/metabolismo , Citocromos/metabolismo , Escherichia coli/enzimología , Protones , Ubiquinona/análogos & derivados , Ubiquinona/metabolismo , Amidas , Ácido Aspártico/genética , Sitios de Unión/genética , Grupo Citocromo b , Citocromos/genética , Electroquímica , Complejo IV de Transporte de Electrones/genética , Complejo IV de Transporte de Electrones/metabolismo , Escherichia coli/genética , Proteínas de Escherichia coli , Ácido Glutámico/genética , Ácido Glutámico/metabolismo , Histidina/genética , Mutagénesis Sitio-Dirigida , Oxidación-Reducción , Oxidorreductasas/genética , Oxidorreductasas/metabolismo , Espectrofotometría Ultravioleta , Espectroscopía Infrarroja por Transformada de Fourier/métodos
13.
Biochim Biophys Acta ; 1459(2-3): 533-9, 2000 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-11004473

RESUMEN

We have investigated the electron-proton coupling during the peroxy (P(R)) to oxo-ferryl (F) and F to oxidised (O) transitions in cytochrome c oxidase from Rhodobacter sphaeroides. The kinetics of these reactions were investigated in two different mutant enzymes: (1) ED(I-286), in which one of the key residues in the D-pathway, E(I-286), was replaced by an aspartate which has a shorter side chain than that of the glutamate and, (2) ML(II-263), in which the redox potential of Cu(A) is increased by approximately 100 mV, which slows electron transfer to the binuclear centre during the F-->O transition by a factor of approximately 200. In ED(I-286) proton uptake during P(R)-->F was slowed by a factor of approximately 5, which indicates that E(I-286) is the proton donor to P(R). In addition, in the mutant enzyme the F-->O transition rate displayed a deuterium isotope effect of approximately 2.5 as compared with approximately 7 in the wild-type enzyme. Since the entire deuterium isotope effect was shown to be associated with a single proton-transfer reaction in which the proton donor and acceptor must approach each other (M. Karpefors, P. Adelroth, P. Brzezinski, Biochemistry 39 (2000) 6850), the smaller deuterium isotope effect in ED(I-286) indicates that proton transfer from E(I-286) determines the rate also of the F-->O transition. In ML(II-263) the electron-transfer to the binuclear centre is slower than the intrinsic proton-transfer rate through the D-pathway. Nevertheless, both electron and proton transfer to the binuclear centre displayed a deuterium isotope effect of approximately 8, i.e., about the same as in the wild-type enzyme, which shows that these reactions are intimately coupled.


Asunto(s)
Complejo IV de Transporte de Electrones/química , Ácido Glutámico/química , Oxígeno/química , Protones , Deuterio , Transporte de Electrón , Complejo IV de Transporte de Electrones/genética , Mutación , Oxidación-Reducción , Fotólisis , Rhodobacter sphaeroides
14.
Biochemistry ; 39(33): 10154-62, 2000 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-10956004

RESUMEN

Changes in the FTIR difference spectra upon photoconversion of the M intermediate to its photoproduct(s) M' were studied in wild-type bacteriorhodopsin and several mutants at low temperatures. The studies aimed at examining whether internally bound water molecules interact with the chromophore and the key residues Asp85 and Asp96 in M, and whether these water molecules participate in the reprotonation of the Schiff base. We have found that three water molecules are perturbed by the isomerization of the chromophore in the M --> M' transition at 80 K. The perturbation of one water molecule, detected as a bilobe at 3567(+)/3550(-) cm(-)(1), relaxed in parallel with the relaxation of an Asp85 perturbation upon increasing temperature from 80 to 100 and 133 K (before the reprotonation of the Schiff base). Two water bands of M at 3588 and 3570 cm(-)(1) shift to 3640 cm(-)(1) upon photoconversion at 173 K. These bands were attributed to water molecules which are located in the vicinity of the Schiff base and Asp85 (Wat85). In the M to M' transition at 80 and 100 K, where the Schiff base remained unprotonated, the Wat85 pair stayed in similar states to those in M. The reprotonation of the Schiff base at 133 K occurred without the restoration of the Wat85 band around 3640 cm(-)(1). This band was restored at higher temperatures. Two water molecules in the region surrounded by Thr46, Asp96, and Phe219 (Wat219) were perturbed in the M to M' transition at 80 K and relaxed in parallel with the relaxation of the perturbation of Asp96 upon increasing the temperature. Mutant studies show that upon photoisomerization of the chromophore at 80 K one of the Wat219 water molecules moves closer to Val49 (located near the lysine side chain attached to retinal, and close to the Schiff base). These data along with our previous results indicate that the water molecules in the cytoplasmic domain participate in the connection of Asp96 with the Schiff base and undergo displacement during photoconversions, presumably shuttling between the Schiff base and a site close to Asp96 in the L to M to N transitions.


Asunto(s)
Bacteriorodopsinas/efectos de la radiación , Bombas de Protones/efectos de la radiación , Retinaldehído/efectos de la radiación , Agua , Ácido Aspártico , Bacteriorodopsinas/química , Bacteriorodopsinas/genética , Polaridad Celular , Ácido Glutámico , Halobacterium , Mutación , Bombas de Protones/química , Bombas de Protones/genética , Retinaldehído/química , Bases de Schiff , Espectroscopía Infrarroja por Transformada de Fourier , Treonina , Valina
15.
Biochim Biophys Acta ; 1458(1): 164-79, 2000 May 12.
Artículo en Inglés | MEDLINE | ID: mdl-10812031

RESUMEN

The current status of our knowledge about the mechanism of proton pumping by cytochrome oxidase is discussed. Significant progress has resulted from the study of site-directed mutants within the proton-conducting pathways of the bacterial oxidases. There appear to be two channels to facilitate proton translocation within the enzyme and they are important at different parts of the catalytic cycle. The use of hydrogen peroxide as an alternative substrate provides a very useful experimental tool to explore the enzymology of this system, and insights gained from this approach are described. Proton transfer is coupled to and appears to regulate the rate of electron transfer steps during turnover. It is proposed that the initial step in the reaction involves a proton transfer to the active site that is important to convert metal-ligated hydroxide to water, which can more rapidly dissociate from the metals and allow the reaction with dioxygen which, we propose, can bind the one-electron reduced heme-copper center. Coordinated movement of protons and electrons over both short and long distances within the enzyme appear to be important at different parts of the catalytic cycle. During the initial reduction of dioxygen, direct hydrogen transfer to form a tyrosyl radical at the active site seems likely. Subsequent steps can be effectively blocked by mutation of a residue at the surface of the protein, apparently preventing the entry of protons.


Asunto(s)
Complejo IV de Transporte de Electrones/química , Bombas de Protones/química , Animales , Catálisis , Cobre/química , Complejo IV de Transporte de Electrones/genética , Potenciales de la Membrana , Modelos Químicos , Mutación , Oxidación-Reducción , Oxígeno/química , Fuerza Protón-Motriz , Rhodobacter , Agua/química
16.
Biochemistry ; 39(11): 2989-96, 2000 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-10715119

RESUMEN

As the final electron acceptor in the respiratory chain of eukaryotic and many prokaryotic organisms, cytochrome c oxidase catalyzes the reduction of oxygen to water, concomitantly generating a proton gradient. X-ray structures of two cytochrome c oxidases have been reported, and in each structure three possible pathways for proton translocation are indicated: the D-, K-, and H-channels. The putative H-channel is most clearly delineated in the bovine heart oxidase and has been proposed to be functionally important for the translocation of pumped protons in the mammalian oxidase [Yoshikawa et al. (1998) Science 280, 1723-1729]. In the present work, the functional importance of residues lining the putative H-channel in the oxidase from Rhodobacter sphaeroides are examined by site-directed mutagenesis. Mutants were generated in eight different sites and the enzymes have been purified and characterized. The results suggest that the H-channel is not functionally important in the prokaryotic oxidase, in agreement with the conclusion from previous work with the oxidase from Paracoccus denitrificans [Pfitzner et al. (1998) J. Biomembr. Bioenerg. 30, 89-93]. Each of the mutants in R. sphaeroides, with an exception at only one position, is enzymatically active and pumps protons in reconstituted proteoliposomes. This includes H456A, where in the P. denitrificans oxidase a leucine residue substituted for the corresponding residue resulted in inactive enzyme. The only mutations that result in completely inactive enzyme in the set examined in the R. sphaeroides oxidase are in R52, a residue that, along with Q471, appears to be hydrogen-bonded to the formyl group of heme a in the X-ray structures. To characterize the interactions between this residue and the heme group, resonance Raman spectra of the R52 mutants were obtained. The frequency of the heme a formyl stretching mode in the R52A mutant is characteristic of that seen in non-hydrogen-bonded model heme a complexes. Thus the data confirm the presence of hydrogen bonding between the heme a formyl group and the R52 side chain, as suggested from crystallographic data. In the R52K mutant, this hydrogen bonding is maintained by the lysine residue, and this mutant enzyme retains near wild-type activity. The heme a formyl frequency is also affected by mutation of Q471, confirming the X-ray models that show this residue also has hydrogen-bonding interactions with the formyl group. Unlike R52, however, Q471 does not appear to be critical for the enzyme function.


Asunto(s)
Complejo IV de Transporte de Electrones/metabolismo , Hemo/análogos & derivados , Histidina/genética , Mutagénesis Sitio-Dirigida , Bombas de Protones/genética , Rhodobacter sphaeroides/enzimología , Alanina/genética , Alanina/metabolismo , Secuencia de Aminoácidos , Sustitución de Aminoácidos/genética , Animales , Arginina/genética , Arginina/metabolismo , Bovinos , Complejo IV de Transporte de Electrones/química , Complejo IV de Transporte de Electrones/genética , Activación Enzimática/genética , Glutamina/genética , Glutamina/metabolismo , Hemo/química , Hemo/metabolismo , Histidina/metabolismo , Lisina/genética , Lisina/metabolismo , Datos de Secuencia Molecular , Bombas de Protones/metabolismo , Rhodobacter sphaeroides/genética , Espectrometría Raman , Tirosina/genética , Tirosina/metabolismo
17.
Biochemistry ; 39(11): 3169-75, 2000 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-10715139

RESUMEN

Electron nuclear double resonance (ENDOR) was performed on the protein-bound, stabilized, high-affinity ubisemiquinone radical, QH*-, of bo3 quinol oxidase to determine its electronic spin distribution and to probe its interaction with its surroundings. Until this present work, such ENDOR studies of protein-stabilized ubisemiquinone centers have only been done on photosynthetic reaction centers whose function is to reduce a ubiquinol pool. In contrast, QH*- serves to oxidize a ubiquinol pool in the course of electron transfer from the ubiquinol pool to the oxygen-consuming center of terminal bo3 oxidase. As documented by large hyperfine couplings (>10 MHz) to nonexchangeable protons on the QH*- ubisemiquinone ring, we provide evidence for an electronic distribution on QH*- that is different from that of the semiquinones of reaction centers. Since the ubisemiquinone itself is physically nearly identical in both QH*- and the bacterial photosynthetic reaction centers, this electronic difference is evidently a function of the local protein environment. Interaction of QH*- with this local protein environment was explicitly shown by exchangeable deuteron ENDOR that implied hydrogen bonding to the quinone and by weak proton hyperfine couplings to the local protein matrix.


Asunto(s)
Citocromos/química , Escherichia coli/enzimología , Ubiquinona/análogos & derivados , Ubiquinona/química , Benzoquinonas/química , Coenzimas , Grupo Citocromo b , Deuterio , Espectroscopía de Resonancia por Spin del Electrón/métodos , Proteínas de Escherichia coli , Protones
18.
Biochemistry ; 38(49): 16246-52, 1999 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-10587447

RESUMEN

The Na(+)-translocating NADH: ubiquinone oxidoreductase (Na(+)-NQR) generates an electrochemical Na(+) potential driven by aerobic respiration. Previous studies on the enzyme from Vibrio alginolyticus have shown that the Na(+)-NQR has six subunits, and it is known to contain FAD and an FeS center as redox cofactors. In the current work, the enzyme from the marine bacterium Vibrio harveyi has been purified and characterized. In addition to FAD, a second flavin, tentatively identified as FMN, was discovered to be covalently attached to the NqrC subunit. The purified V. harveyi Na(+)-NQR was reconstituted into proteoliposomes. The generation of a transmembrane electric potential by the enzyme upon NADH:Q(1) oxidoreduction was strictly dependent on Na(+), resistant to the protonophore CCCP, and sensitive to the sodium ionophore ETH-157, showing that the enzyme operates as a primary electrogenic sodium pump. Interior alkalinization of the inside-out proteoliposomes due to the operation of the Na(+)-NQR was accelerated by CCCP, inhibited by valinomycin, and completely arrested by ETH-157. Hence, the protons required for ubiquinol formation must be taken up from the outside of the liposomes, which corresponds to the bacterial cytoplasm. The Na(+)-NQR operon from this bacterium was sequenced, and the sequence shows strong homology to the previously reported Na(+)-NQR operons from V. alginolyticus and Haemophilus influenzae. Homology studies show that a number of other bacteria, including a number of pathogenic species, also have an Na(+)-NQR operon.


Asunto(s)
NADH NADPH Oxidorreductasas/química , Vibrio/enzimología , Secuencia de Aminoácidos , Catálisis , Complejo I de Transporte de Electrón , Metabolismo Energético , Flavina-Adenina Dinucleótido/aislamiento & purificación , Ligandos , Datos de Secuencia Molecular , NADH NADPH Oxidorreductasas/genética , NADH NADPH Oxidorreductasas/aislamiento & purificación , NADH NADPH Oxidorreductasas/metabolismo , Operón , Análisis de Secuencia de Proteína , Homología de Secuencia de Aminoácido , Sodio/metabolismo
19.
Biochemistry ; 38(44): 14683-9, 1999 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-10545194

RESUMEN

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).


Asunto(s)
Citocromos/química , Ubiquinona/química , Sitios de Unión , Isótopos de Carbono , Grupo Citocromo b , Electroquímica , Escherichia coli/química , Proteínas de Escherichia coli , Enlace de Hidrógeno , Oxidación-Reducción , Conformación Proteica , Espectroscopía Infrarroja por Transformada de Fourier , Vibración
20.
Biochemistry ; 38(46): 15150-6, 1999 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-10563797

RESUMEN

Recent electrostatics calculations on the cytochrome c oxidase from Paracoccus denitrificans revealed an unexpected coupling between the redox state of the heme-copper center and the state of protonation of a glutamic acid (E78II) that is 25 A away in subunit II of the oxidase. Examination of more than 300 sequences of the homologous subunit in other heme-copper oxidases shows that this residue is virtually totally conserved and is in a cluster of very highly conserved residues at the "negative" end (bacterial cytoplasm or mitochondrial matrix) of the second transmembrane helix. The functional importance of several residues in this cluster (E89II, W93II, T94II, and P96II) was examined by site-directed mutagenesis of the corresponding region of the cytochrome bo(3) quinol oxidase from Escherichia coli (where E89II is the equivalent of residue E78II of the P. denitrificans oxidase). Substitution of E89II with either alanine or glutamine resulted in reducing the rate of turnover to about 43 or 10% of the wild-type value, respectively, whereas E89D has only about 60% of the activity of the control oxidase. The quinol oxidase activity of the W93V mutant is also reduced to about 30% of that of the wild-type oxidase. Spectroscopic studies with the purified E89A and E89Q mutants indicate no perturbation of the heme-copper center. The data suggest that E89II (E. coli numbering) is critical for the function of the heme copper oxidases. The proximity to K362 suggests that this glutamic acid residue may regulate proton entry or transit through the K-channel. This hypothesis is supported by the finding that the degree of oxidation of the low-spin heme b is greater in the steady state using hydrogen peroxide as an oxidant in place of dioxygen for the E89Q mutant. Thus, it appears that the inhibition resulting from the E89II mutation is due to a block in the reduction of the heme-copper binuclear center, expected for K-channel mutants.


Asunto(s)
Cobre/química , Citocromos/química , Escherichia coli/enzimología , Ácido Glutámico/química , Hemo/química , Alanina/genética , Secuencia Conservada , Grupo Citocromo b , Citocromos/genética , Complejo IV de Transporte de Electrones/química , Complejo IV de Transporte de Electrones/metabolismo , Activación Enzimática/genética , Escherichia coli/genética , Proteínas de Escherichia coli , Ácido Glutámico/genética , Concentración de Iones de Hidrógeno , Proteínas de la Membrana/química , Oxidación-Reducción , Bombas de Protones/química , Quinona Reductasas/química , Quinona Reductasas/genética , Espectrofotometría Ultravioleta , Electricidad Estática
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA