Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros











Intervalo de año de publicación
2.
J Biol Chem ; 296: 100740, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33957129

RESUMEN

The modern-day respiratory complex I shares a common ancestor with the membrane-bound hydrogenase (MBH) and membrane-bound sulfane sulfur reductase (MBS). MBH and MBS use protons and sulfur as their respective electron sinks, which helped to conserve energy during early life in the Proterozoic era when the Earth's atmosphere was low in oxygen. MBH and MBS likely evolved from an integration of an ancestral, membrane-embedded, multiple resistance and pH antiporter and a soluble redox-active module encompassing a [NiFe] hydrogenase. In this review, we discuss how the structures of MBH, MBS, multiple resistance and pH, photosynthetic NADH dehydrogenase-like complex type-1, and complex I, which have been determined recently, thanks to the advent of high-resolution cryo-EM, have significantly improved our understanding of the catalytic reaction mechanisms and the evolutionary relationships of the respiratory complexes.


Asunto(s)
Evolución Biológica , Complejo I de Transporte de Electrón/metabolismo , Adenosina Trifosfato/biosíntesis , Catálisis , Complejo I de Transporte de Electrón/genética , Transporte Iónico , Oxidación-Reducción , Protones , Sodio/metabolismo
3.
Nat Commun ; 11(1): 5953, 2020 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-33230146

RESUMEN

Modern day aerobic respiration in mitochondria involving complex I converts redox energy into chemical energy and likely evolved from a simple anaerobic system now represented by hydrogen gas-evolving hydrogenase (MBH) where protons are the terminal electron acceptor. Here we present the cryo-EM structure of an early ancestor in the evolution of complex I, the elemental sulfur (S0)-reducing reductase MBS. Three highly conserved protein loops linking cytoplasmic and membrane domains enable scalable energy conversion in all three complexes. MBS contains two proton pumps compared to one in MBH and likely conserves twice the energy. The structure also reveals evolutionary adaptations of MBH that enabled S0 reduction by MBS catalyzed by a site-differentiated iron-sulfur cluster without participation of protons or amino acid residues. This is the simplest mechanism proposed for reduction of inorganic or organic disulfides. It is of fundamental significance in the iron and sulfur-rich volcanic environments of early earth and possibly the origin of life. MBS provides a new perspective on the evolution of modern-day respiratory complexes and of catalysis by biological iron-sulfur clusters.


Asunto(s)
Proteínas Hierro-Azufre/química , Proteínas Hierro-Azufre/metabolismo , Oxidorreductasas/química , Oxidorreductasas/metabolismo , Azufre/metabolismo , Catálisis , Dominio Catalítico , Microscopía por Crioelectrón , Complejo I de Transporte de Electrón/química , Complejo I de Transporte de Electrón/metabolismo , Hidrogenasas/química , Hidrogenasas/metabolismo , Membranas Mitocondriales/enzimología , Membranas Mitocondriales/metabolismo , Modelos Moleculares , Origen de la Vida , Oxidación-Reducción , Bombas de Protones/química , Pyrococcus furiosus/química , Pyrococcus furiosus/enzimología , Intercambiadores de Sodio-Hidrógeno/química
4.
J Am Chem Soc ; 142(3): 1227-1235, 2020 01 22.
Artículo en Inglés | MEDLINE | ID: mdl-31816235

RESUMEN

Hydrogenases display a wide range of catalytic rates and biases in reversible hydrogen gas oxidation catalysis. The interactions of the iron-sulfur-containing catalytic site with the local protein environment are thought to contribute to differences in catalytic reactivity, but this has not been demonstrated. The microbe Clostridium pasteurianum produces three [FeFe]-hydrogenases that differ in "catalytic bias" by exerting a disproportionate rate acceleration in one direction or the other that spans a remarkable 6 orders of magnitude. The combination of high-resolution structural work, biochemical analyses, and computational modeling indicates that protein secondary interactions directly influence the relative stabilization/destabilization of different oxidation states of the active site metal cluster. This selective stabilization or destabilization of oxidation states can preferentially promote hydrogen oxidation or proton reduction and represents a simple yet elegant model by which a protein catalytic site can confer catalytic bias.


Asunto(s)
Hidrógeno/metabolismo , Hidrogenasas/metabolismo , Catálisis , Clostridium/enzimología , Oxidación-Reducción , Difracción de Rayos X
5.
J Biol Chem ; 293(43): 16687-16696, 2018 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-30181217

RESUMEN

Hyperthermophilic archaea contain a hydrogen gas-evolving,respiratory membrane-bound NiFe-hydrogenase (MBH) that is very closely related to the aerobic respiratory complex I. During growth on elemental sulfur (S°), these microorganisms also produce a homologous membrane-bound complex (MBX), which generates H2S. MBX evolutionarily links MBH to complex I, but its catalytic function is unknown. Herein, we show that MBX reduces the sulfane sulfur of polysulfides by using ferredoxin (Fd) as the electron donor, and we rename it membrane-bound sulfane reductase (MBS). Two forms of affinity-tagged MBS were purified from genetically engineered Pyrococcus furiosus (a hyperthermophilic archaea species): the 13-subunit holoenzyme (S-MBS) and a cytoplasmic 4-subunit catalytic subcomplex (C-MBS). S-MBS and C-MBS reduced dimethyl trisulfide (DMTS) with comparable Km (∼490 µm) and Vmax values (12 µmol/min/mg). The MBS catalytic subunit (MbsL), but not that of complex I (NuoD), retains two of four NiFe-coordinating cysteine residues of MBH. However, these cysteine residues were not involved in MBS catalysis because a mutant P. furiosus strain (MbsLC85A/C385A) grew normally with S°. The products of the DMTS reduction and properties of polysulfides indicated that in the physiological reaction, MBS uses Fd (Eo' = -480 mV) to reduce sulfane sulfur (Eo' -260 mV) and cleave organic (RS n R, n ≥ 3) and anionic polysulfides (S n2-, n ≥ 4) but that it does not produce H2S. Based on homology to MBH, MBS also creates an ion gradient for ATP synthesis. This work establishes the electrochemical reaction catalyzed by MBS that is intermediate in the evolution from proton- to quinone-reducing respiratory complexes.


Asunto(s)
Proteínas Arqueales/metabolismo , Membrana Celular/metabolismo , Complejo I de Transporte de Electrón/metabolismo , Proteínas de la Membrana/metabolismo , Oxidorreductasas/metabolismo , Pyrococcus furiosus/enzimología , Sulfuros/química , Proteínas Arqueales/genética , Dominio Catalítico , Complejo I de Transporte de Electrón/genética , Proteínas de la Membrana/genética , Oxidación-Reducción , Oxidorreductasas/genética , Pyrococcus furiosus/crecimiento & desarrollo
6.
J Biol Chem ; 289(5): 2873-9, 2014 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-24318960

RESUMEN

Biohydrogen gas has enormous potential as a source of reductant for the microbial production of biofuels, but its low solubility and poor gas mass transfer rates are limiting factors. These limitations could be circumvented by engineering biofuel production in microorganisms that are also capable of generating H2 from highly soluble chemicals such as formate, which can function as an electron donor. Herein, the model hyperthermophile, Pyrococcus furiosus, which grows optimally near 100 °C by fermenting sugars to produce H2, has been engineered to also efficiently convert formate to H2. Using a bacterial artificial chromosome vector, the 16.9-kb 18-gene cluster encoding the membrane-bound, respiratory formate hydrogen lyase complex of Thermococcus onnurineus was inserted into the P. furiosus chromosome and expressed as a functional unit. This enabled P. furiosus to utilize formate as well as sugars as an H2 source and to do so at both 80° and 95 °C, near the optimum growth temperature of the donor (T. onnurineus) and engineered host (P. furiosus), respectively. This accomplishment also demonstrates the versatility of P. furiosus for metabolic engineering applications.


Asunto(s)
Biocombustibles/microbiología , Formiatos/metabolismo , Hidrógeno/metabolismo , Pyrococcus furiosus/enzimología , Pyrococcus furiosus/genética , Thermococcus/enzimología , Gases/metabolismo , Genes Bacterianos/genética , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ingeniería Metabólica/métodos , Complejos Multienzimáticos/genética , Complejos Multienzimáticos/metabolismo , Operón/genética , Thermococcus/genética
7.
Mol Microbiol ; 77(5): 1111-22, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20598080

RESUMEN

We present structural and biochemical evidence for a redox switch in the archaeal transcriptional regulator SurR of Pyrococcus furiosus, a hyperthermophilic anaerobe. P. furiosus produces H(2) during fermentation, but undergoes a metabolic shift to produce H(2) S when elemental sulfur (S(0) ) becomes available. Changes in gene expression occur within minutes of S(0) addition, and the majority of these S(0) -responsive genes are regulatory targets of SurR, a key regulator involved in primary S(0) response. SurR was shown in vitro to have dual functionality, activating transcription of some of these genes, notably the hydrogenase operons, and repressing others, including a gene-encoding sulfur reductase. This work demonstrates via biochemical and structural evidence that the activity of SurR is modulated by cysteine residues in a CxxC motif that constitutes a redox switch. Oxidation of the switch with S(0) inhibits sequence-specific DNA binding by SurR, leading to deactivation of genes related to H(2) production and derepression of genes involved in S(0) metabolism.


Asunto(s)
Proteínas Arqueales/metabolismo , Regulación de la Expresión Génica Arqueal , Sulfuro de Hidrógeno/metabolismo , Hidrógeno/metabolismo , Pyrococcus furiosus/fisiología , Azufre/metabolismo , Factores de Transcripción/metabolismo , Secuencia de Aminoácidos , Modelos Moleculares , Datos de Secuencia Molecular , Oxidación-Reducción , Multimerización de Proteína , Estructura Cuaternaria de Proteína , Pyrococcus furiosus/metabolismo
8.
J Bacteriol ; 191(13): 4451-7, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19411328

RESUMEN

The hyperthermophilic and anaerobic bacterium Thermotoga maritima ferments a wide variety of carbohydrates, producing acetate, CO(2), and H(2). Glucose is degraded through a classical Embden-Meyerhof pathway, and both NADH and reduced ferredoxin are generated. The oxidation of these electron carriers must be coupled to H(2) production, but the mechanism by which this occurs is unknown. The trimeric [FeFe]-type hydrogenase that was previously purified from T. maritima does not use either reduced ferredoxin or NADH as a sole electron donor. This problem has now been resolved by the demonstration that this hydrogenase requires the presence of both electron carriers for catalysis of H(2) production. The enzyme oxidizes NADH and ferredoxin simultaneously in an approximately 1:1 ratio and in a synergistic fashion to produce H(2). It is proposed that the enzyme represents a new class of bifurcating [FeFe] hydrogenase in which the exergonic oxidation of ferredoxin (midpoint potential, -453 mV) is used to drive the unfavorable oxidation of NADH (E(0)' = -320 mV) to produce H(2) (E(0)' = -420 mV). From genome sequence analysis, it is now clear that there are two major types of [FeFe] hydrogenases: the trimeric bifurcating enzyme and the more well-studied monomeric ferredoxin-dependent [FeFe] hydrogenase. Almost one-third of the known H(2)-producing anaerobes appear to contain homologs of the trimeric bifurcating enzyme, although many of them also harbor one or more homologs of the simpler ferredoxin-dependent hydrogenase. The discovery of the bifurcating hydrogenase gives a new perspective on our understanding of the bioenergetics and mechanism of H(2) production and of anaerobic metabolism in general.


Asunto(s)
Ferredoxinas/metabolismo , Hidrógeno/metabolismo , Hidrogenasas/metabolismo , Proteínas Hierro-Azufre/metabolismo , NAD/metabolismo , Thermotoga maritima/enzimología , Thermotoga maritima/metabolismo , Anaerobiosis , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Genoma Bacteriano/genética , Hidrogenasas/genética , Proteínas Hierro-Azufre/genética , Modelos Biológicos , Thermotoga maritima/genética
9.
Appl Environ Microbiol ; 75(7): 1820-5, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19201977

RESUMEN

The iron-sulfur nitroso compound [Fe(4)S(3)(NO)(7)](-) is a broad-spectrum antimicrobial agent that has been used for more than 100 years to combat pathogenic anaerobes. Known as Roussin's black salt (RBS), it contains seven moles of nitric oxide, the release of which was always assumed to mediate its cytotoxicity. Using the hyperthermophilic archaeon Pyrococcus furiosus, it is demonstrated through growth studies, membrane analyses, and scanning electron microscopy that nitric oxide does not play a role in RBS toxicity; rather, the mechanism involves membrane disruption leading to cell lysis. Moreover, insoluble elemental sulfur (S(0)), which is reduced by P. furiosus to hydrogen sulfide, prevents cell lysis by RBS. It is proposed that S(0) also directly interacts with the membranes of P. furiosus during its transfer into the cell, ultimately for reduction by a cytosolic NADPH sulfur reductase. RBS is proposed to be a new class of inorganic antimicrobial agent that also has potential use as an inert cell-lysing agent.


Asunto(s)
Desinfectantes/farmacología , Compuestos de Hierro/farmacología , Compuestos Nitrosos/farmacología , Pyrococcus furiosus/efectos de los fármacos , Membrana Celular/efectos de los fármacos , Membrana Celular/ultraestructura , Sulfuro de Hidrógeno/metabolismo , Microscopía Electrónica de Rastreo , Pyrococcus furiosus/crecimiento & desarrollo , Pyrococcus furiosus/ultraestructura , Azufre/metabolismo
10.
Biochem J ; 375(Pt 2): 231-46, 2003 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-12921536

RESUMEN

An early divergence in evolution has resulted in two prokaryotic domains, the Bacteria and the Archaea. Whereas the central metabolic routes of bacteria and eukaryotes are generally well-conserved, variant pathways have developed in Archaea involving several novel enzymes with a distinct control. A spectacular example of convergent evolution concerns the glucose-degrading pathways of saccharolytic archaea. The identification, characterization and comparison of the glycolytic enzymes of a variety of phylogenetic lineages have revealed a mosaic of canonical and novel enzymes in the archaeal variants of the Embden-Meyerhof and the Entner-Doudoroff pathways. By means of integrating results from biochemical and genetic studies with recently obtained comparative and functional genomics data, the structure and function of the archaeal glycolytic routes, the participating enzymes and their regulation are re-evaluated.


Asunto(s)
Archaea/metabolismo , Glucosa/metabolismo , Glucólisis , Adenosina Difosfato/metabolismo , Archaea/enzimología , Archaea/genética , Evolución Molecular , Fosfotransferasas/genética , Fosfotransferasas/metabolismo , Filogenia , Especificidad de la Especie
11.
J Bacteriol ; 185(13): 3935-47, 2003 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12813088

RESUMEN

The first complete-genome DNA microarray was constructed for a hyperthermophile or a nonhalophilic archaeon by using the 2,065 open reading frames (ORFs) that have been annotated in the genome of Pyrococcus furiosus (optimal growth temperature, 100 degrees C). This was used to determine relative transcript levels in cells grown at 95 degrees C with either peptides or a carbohydrate (maltose) used as the primary carbon source. Approximately 20% (398 of 2065) of the ORFs did not appear to be significantly expressed under either growth condition. Of the remaining 1,667 ORFs, the expression of 125 of them (8%) differed by more than fivefold between the two cultures, and 82 of the 125 (65%) appear to be part of operons, indicating extensive coordinate regulation. Of the 27 operons that are regulated, 5 of them encode (conserved) hypothetical proteins. A total of 18 operons are up-regulated (greater than fivefold) in maltose-grown cells, including those responsible for maltose transport and for the biosynthesis of 12 amino acids, of ornithine, and of citric acid cycle intermediate products. A total of nine operons are up-regulated (greater than fivefold) in peptide-grown cells, including those encoding enzymes involved in the production of acyl and aryl acids and 2-ketoacids, which are used for energy conservation. Analyses of the spent growth media confirmed the production of branched-chain and aromatic acids during growth on peptides. In addition, six nonlinked enzymes in the pathways of sugar metabolism were regulated more than fivefold--three in maltose-grown cells that are unique to the unusual glycolytic pathway and three in peptide-grown cells that are unique to gluconeogenesis. The catalytic activities of 16 metabolic enzymes whose expression appeared to be highly regulated in the two cell types correlated very well with the microarray data. The degree of coordinate regulation revealed by the microarray data was unanticipated and shows that P. furiosus can readily adapt to a change in its primary carbon source.


Asunto(s)
Proteínas Arqueales/metabolismo , Perfilación de la Expresión Génica , Maltosa/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Péptidos/metabolismo , Pyrococcus furiosus/crecimiento & desarrollo , Proteínas Arqueales/genética , Medios de Cultivo , Regulación de la Expresión Génica Arqueal , Genoma Arqueal , Calor , Sistemas de Lectura Abierta , Pyrococcus furiosus/genética , Pyrococcus furiosus/metabolismo , Transcripción Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA