Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Bacteriol ; 201(4)2019 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-30455284

RESUMEN

Rhodobacter sphaeroides is able to use 3-hydroxypropionate as the sole carbon source through the reductive conversion of 3-hydroxypropionate to propionyl coenzyme A (propionyl-CoA). The ethylmalonyl-CoA pathway is not required in this process because a crotonyl-CoA carboxylase/reductase (Ccr)-negative mutant still grew with 3-hydroxypropionate. Much to our surprise, a mutant defective for another specific enzyme of the ethylmalonyl-CoA pathway, mesaconyl-CoA hydratase (Mch), lost its ability for 3-hydroxypropionate-dependent growth. Interestingly, the Mch-deficient mutant was rescued either by introducing an additional ccr in-frame deletion that resulted in the blockage of an earlier step in the pathway or by heterologously expressing a gene encoding a thioesterase (YciA) that can act on several CoA intermediates of the ethylmalonyl-CoA pathway. The mch mutant expressing yciA metabolized only less than half of the 3-hydroxypropionate supplied, and over 50% of that carbon was recovered in the spent medium as free acids of the key intermediates mesaconyl-CoA and methylsuccinyl-CoA. A gradual increase in growth inhibition due to the blockage of consecutive steps of the ethylmalonyl-CoA pathway by gene deletions suggests that the growth defects were due to the titration of free CoA and depletion of the CoA pool in the cell rather than to detrimental effects arising from the accumulation of a specific metabolite. Recovery of carbon in mesaconate for the wild-type strain expressing yciA demonstrated that carbon flux through the ethylmalonyl-CoA pathway occurs during 3-hydroxypropionate-dependent growth. A possible role of the ethylmalonyl-CoA pathway is proposed that functions outside its known role in providing tricarboxylic acid intermediates during acetyl-CoA assimilation.IMPORTANCE Mutant analysis is an important tool utilized in metabolic studies to understand which role a particular pathway might have under certain growth conditions for a given organism. The importance of the enzyme and of the pathway in which it participates is discretely linked to the resulting phenotype observed after mutation of the corresponding gene. This work highlights the possibility of incorrectly interpreting mutant growth results that are based on studying a single unit (gene and encoded enzyme) of a metabolic pathway rather than the pathway in its entirety. This work also hints at the possibility of using an enzyme as a drug target although the enzyme may participate in a nonessential pathway and still be detrimental to the cell when inhibited.


Asunto(s)
Acilcoenzima A/metabolismo , Ácido Láctico/análogos & derivados , Redes y Vías Metabólicas/genética , Rhodobacter sphaeroides/crecimiento & desarrollo , Rhodobacter sphaeroides/metabolismo , Acilcoenzima A/deficiencia , Carbono/metabolismo , Eliminación de Gen , Ácido Láctico/metabolismo , Análisis de Flujos Metabólicos
2.
J Bacteriol ; 197(19): 3048-56, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26170412

RESUMEN

UNLABELLED: Propionyl coenzyme A (propionyl-CoA) assimilation by Rhodobacter sphaeroides proceeds via the methylmalonyl-CoA pathway. The activity of the key enzyme of the pathway, propionyl-CoA carboxylase (PCC), was upregulated 20-fold during growth with propionate compared to growth with succinate. Because propionyl-CoA is an intermediate in acetyl-CoA assimilation via the ethylmalonyl-CoA pathway, acetate growth also requires the methylmalonyl-CoA pathway. PCC activities were upregulated 8-fold in extracts of acetate-grown cells compared to extracts of succinate-grown cells. The upregulation of PCC activities during growth with propionate or acetate corresponded to increased expression of the pccB gene, which encodes a subunit of PCC. PccR (RSP_2186) was identified to be a transcriptional regulator required for the upregulation of pccB transcript levels and, consequently, PCC activity: growth substrate-dependent regulation was lost when pccR was inactivated by an in-frame deletion. In the pccR mutant, lacZ expression from a 215-bp plasmid-borne pccB upstream fragment including 27 bp of the pccB coding region was also deregulated. A loss of regulation as a result of mutations in the conserved motifs TTTGCAAA-X4-TTTGCAAA in the presence of PccR allowed the prediction of a possible operator site. PccR, together with homologs from other organisms, formed a distinct clade within the family of short-chain fatty acyl coenzyme A regulators (ScfRs) defined here. Some members from other clades within the ScfR family have previously been shown to be involved in regulating acetyl-CoA assimilation by the glyoxylate bypass (RamB) or propionyl-CoA assimilation by the methylcitrate cycle (MccR). IMPORTANCE: Short-chain acyl-CoAs are intermediates in essential biosynthetic and degradative pathways. The regulation of their accumulation is crucial for appropriate cellular function. This work identifies a regulator (PccR) that prevents the accumulation of propionyl-CoA by controlling expression of the gene encoding propionyl-CoA carboxylase, which is responsible for propionyl-CoA consumption by Rhodobacter sphaeroides. Many other Proteobacteria and Actinomycetales contain one or several PccR homologs that group into distinct clades on the basis of the pathway of acyl-CoA metabolism that they control. Furthermore, an upstream analysis of genes encoding PccR homologs allows the prediction of conserved binding motifs for these regulators. Overall, this study evaluates a single regulator of propionyl-CoA assimilation while expanding the knowledge of the regulation of short-chain acyl-CoAs in many bacterial species.


Asunto(s)
Proteínas Bacterianas/metabolismo , Regulación Bacteriana de la Expresión Génica/fisiología , Regulación Enzimológica de la Expresión Génica/fisiología , Rhodobacter sphaeroides/enzimología , Proteínas Bacterianas/genética , Eliminación de Gen , Familia de Multigenes , Filogenia , Subunidades de Proteína , ARN Bacteriano , Rhodobacter sphaeroides/genética , Rhodobacter sphaeroides/metabolismo , Transcripción Genética
3.
J Bacteriol ; 195(20): 4716-25, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23955006

RESUMEN

The anoxygenic phototroph Rhodobacter sphaeroides uses 3-hydroxypropionate as a sole carbon source for growth. Previously, we showed that the gene (RSP_1434) known as acuI, which encodes a protein of the medium-chain dehydrogenase/reductase (MDR) superfamily, was involved in 3-hydroxypropionate assimilation via the reductive conversion to propionyl-coenzyme A (CoA). Based on these results, we speculated that acuI encoded acrylyl-CoA reductase. In this work, we characterize the in vitro enzyme activity of purified, recombinant AcuI using a coupled spectrophotometric assay. AcuI from R. sphaeroides catalyzes the NADPH-dependent acrylyl-CoA reduction to produce propionyl-CoA. Two other members of the MDR012 family within the MDR superfamily, the products of SPO_1914 from Ruegeria pomeroyi and yhdH from Escherichia coli, were shown to also be part of this new class of NADPH-dependent acrylyl-CoA reductases. The activities of the three enzymes were characterized by an extremely low Km for acrylyl-CoA (<3 µM) and turnover numbers of 45 to 80 s(-1). These homodimeric enzymes were highly specific for NADPH (Km = 18 to 33 µM), with catalytic efficiencies of more than 10-fold higher for NADPH than for NADH. The introduction of codon-optimized SPO_1914 or yhdH into a ΔacuI::kan mutant of R. sphaeroides on a plasmid complemented 3-hydroxypropionate-dependent growth. However, in their native hosts, SPO_1914 and yhdH are believed to function in the metabolism of substrates other than 3-hydroxypropionate, where acrylyl-CoA is an intermediate. Complementation of the ΔacuI::kan mutant phenotype by crotonyl-CoA carboxylase/reductase from R. sphaeroides was attributed to the fact that the enzyme also uses acrylyl-CoA as a substrate.


Asunto(s)
Proteínas Bacterianas/metabolismo , Ácido Láctico/análogos & derivados , Oxidorreductasas/metabolismo , Rhodobacter sphaeroides/enzimología , Proteínas Bacterianas/genética , Regulación Bacteriana de la Expresión Génica/fisiología , Regulación Enzimológica de la Expresión Génica/fisiología , Histidina , Ácido Láctico/metabolismo , Oxidorreductasas/química , Oxidorreductasas/genética , Rhodobacter sphaeroides/metabolismo
4.
J Bacteriol ; 194(2): 225-32, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22056933

RESUMEN

3-Hydroxypropionate is a product or intermediate of the carbon metabolism of organisms from all three domains of life. However, little is known about how carbon derived from 3-hydroxypropionate is assimilated by organisms that can utilize this C(3) compound as a carbon source. This work uses the model bacterium Rhodobacter sphaeroides to begin to elucidate how 3-hydroxypropionate can be incorporated into cell constituents. To this end, a quantitative assay for 3-hydroxypropionate was developed by using recombinant propionyl coenzyme A (propionyl-CoA) synthase from Chloroflexus aurantiacus. Using this assay, we demonstrate that R. sphaeroides can utilize 3-hydroxypropionate as the sole carbon source and energy source. We establish that acetyl-CoA is not the exclusive entry point for 3-hydroxypropionate into the central carbon metabolism and that the reductive conversion of 3-hydroxypropionate to propionyl-CoA is a necessary route for the assimilation of this molecule by R. sphaeroides. Our conclusion is based on the following findings: (i) crotonyl-CoA carboxylase/reductase, a key enzyme of the ethylmalonyl-CoA pathway for acetyl-CoA assimilation, was not essential for growth with 3-hydroxypropionate, as demonstrated by mutant analyses and enzyme activity measurements; (ii) the reductive conversion of 3-hydroxypropionate or acrylate to propionyl-CoA was detected in cell extracts of R. sphaeroides grown with 3-hydroxypropionate, and both activities were upregulated compared to the activities of succinate-grown cells; and (iii) the inactivation of acuI, encoding a candidate acrylyl-CoA reductase, resulted in a 3-hydroxypropionate-negative growth phenotype.


Asunto(s)
Acilcoenzima A/metabolismo , Proteínas Bacterianas/metabolismo , Ácido Láctico/análogos & derivados , Rhodobacter sphaeroides/metabolismo , Acil-CoA Deshidrogenasas/metabolismo , Secuencia de Aminoácidos , Proteínas Bacterianas/genética , Metabolismo Energético , Eliminación de Gen , Regulación Bacteriana de la Expresión Génica/fisiología , Regulación Enzimológica de la Expresión Génica/fisiología , Ácido Láctico/metabolismo , Datos de Secuencia Molecular , Oxidación-Reducción
5.
Appl Microbiol Biotechnol ; 89(1): 17-25, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20882276

RESUMEN

The ethylmalonyl-CoA pathway is central to the carbon metabolism of many α-proteobacteria, like Rhodobacter sphaeroides and Methylobacterium extorquens as well as actinomycetes, like Streptomyces spp. Its function is to convert acetyl-CoA, a central carbon intermediate, to other precursor metabolites for cell carbon biosynthesis. In contrast to the glyoxylate cycle--another widely distributed acetyl-CoA assimilation strategy--the ethylmalonyl-CoA pathway contains many unique CoA-ester intermediates, such as (2R)- and (2S)-ethylmalonyl-CoA, (2S)-methylsuccinyl-CoA, mesaconyl-(C1)-CoA, and (2R, 3S)-methylmalyl-CoA. With this come novel catalysts that interconvert these compounds. Among these unique enzymes is a novel carboxylase that reductively carboxylates crotonyl-CoA, crotonyl-CoA carboxylase/reductase, and (3S)-malyl-CoA thioesterase. The latter represents the first example of a non-Claisen condensation enzyme of the malate synthase superfamily and defines a new class of thioesterases apart from the hotdog-fold and α/ß-fold thioesterases. The biotechnological implications of the ethylmalonyl-CoA pathway are tremendous as one looks to tap into the potential of using these new intermediates and catalysts to produce value-added products.


Asunto(s)
Acilcoenzima A/metabolismo , Bacterias/metabolismo , Vías Biosintéticas , Microbiología Industrial , Bacterias/enzimología , Bacterias/genética , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Carbono/metabolismo
6.
Arch Microbiol ; 193(2): 151-4, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21104179

RESUMEN

Rhodobacter sphaeroides ribulose-1,5-bisphosphate carboxylase/oxygenase (RubisCO)-deletion strain 16 was capable of photoheterotrophic growth with acetate, while Rhodopseudomonas palustris RubisCO-deletion strain 2040 could not grow under these conditions. The reason for this difference lies in the fact that Rba. sphaeroides and Rps. palustris use different pathways for acetate assimilation, the ethylmalonyl-CoA pathway, and glyoxylate-bypass cycle, respectively. The ethylmalonyl-CoA pathway is distinct from the glyoxylate cycle as one molecule of CO(2) and one molecule of HCO(3) (-) per three molecules of acetyl-CoA are co-assimilated to form two malate molecules. The glyoxylate cycle directly converts two acetyl-CoA molecules to malate. Each pathway, therefore, also dictates at what point, CO(2) and reductant are consumed, thereby determining the requirement for the Calvin-Benson-Bassham reductive pentose phosphate cycle.


Asunto(s)
Acetatos/metabolismo , Fotosíntesis , Rhodobacter sphaeroides/metabolismo , Rhodopseudomonas/metabolismo , Acetilcoenzima A/metabolismo , Acilcoenzima A/metabolismo , Eliminación de Gen , Pentosafosfatos/metabolismo , Rhodobacter sphaeroides/genética , Rhodobacter sphaeroides/crecimiento & desarrollo , Rhodopseudomonas/genética , Rhodopseudomonas/crecimiento & desarrollo , Ribulosa-Bifosfato Carboxilasa/genética
7.
Nat Rev Microbiol ; 8(6): 447-60, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20453874

RESUMEN

The acquisition of cellular carbon from inorganic carbon is a prerequisite for life and marked the transition from the inorganic to the organic world. Recent theories of the origins of life assume that chemo-evolution took place in a hot volcanic flow setting through a transition metal-catalysed, autocatalytic carbon fixation cycle. Many archaea live in volcanic habitats under such constraints, in high temperatures with only inorganic substances and often under anoxic conditions. In this Review, we describe the diverse carbon fixation mechanisms that are found in archaea. These reactions differ fundamentally from those of the well-known Calvin cycle, and their distribution mirrors the phylogenetic positions of the archaeal lineages and the needs of the ecological niches that they occupy.


Asunto(s)
Archaea/metabolismo , Procesos Autotróficos , Carbono/metabolismo , Acetilcoenzima A/metabolismo , Archaea/clasificación , Transportadores de Ácidos Dicarboxílicos/metabolismo , Ecosistema , Glucosa/metabolismo , Hidroxibutiratos/metabolismo , Redes y Vías Metabólicas , Filogenia
8.
J Bacteriol ; 192(5): 1249-58, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20047909

RESUMEN

Assimilation of acetyl coenzyme A (acetyl-CoA) is an essential process in many bacteria that proceeds via the glyoxylate cycle or the ethylmalonyl-CoA pathway. In both assimilation strategies, one of the final products is malate that is formed by the condensation of acetyl-CoA with glyoxylate. In the glyoxylate cycle this reaction is catalyzed by malate synthase, whereas in the ethylmalonyl-CoA pathway the reaction is separated into two proteins: malyl-CoA lyase, a well-known enzyme catalyzing the Claisen condensation of acetyl-CoA with glyoxylate and yielding malyl-CoA, and an unidentified malyl-CoA thioesterase that hydrolyzes malyl-CoA into malate and CoA. In this study the roles of Mcl1 and Mcl2, two malyl-CoA lyase homologs in Rhodobacter sphaeroides, were investigated by gene inactivation and biochemical studies. Mcl1 is a true (3S)-malyl-CoA lyase operating in the ethylmalonyl-CoA pathway. Notably, Mcl1 is a promiscuous enzyme and catalyzes not only the condensation of acetyl-CoA and glyoxylate but also the cleavage of beta-methylmalyl-CoA into glyoxylate and propionyl-CoA during acetyl-CoA assimilation. In contrast, Mcl2 was shown to be the sought (3S)-malyl-CoA thioesterase in the ethylmalonyl-CoA pathway, which specifically hydrolyzes (3S)-malyl-CoA but does not use beta-methylmalyl-CoA or catalyze a lyase or condensation reaction. The identification of Mcl2 as thioesterase extends the enzyme functions of malyl-CoA lyase homologs that have been known only as "Claisen condensation" enzymes so far. Mcl1 and Mcl2 are both related to malate synthase, an enzyme which catalyzes both a Claisen condensation and thioester hydrolysis reaction.


Asunto(s)
Acilcoenzima A/metabolismo , Proteínas Bacterianas/metabolismo , Malato Sintasa/metabolismo , Oxo-Ácido-Liasas/metabolismo , Rhodobacter sphaeroides/metabolismo , Tioléster Hidrolasas/metabolismo , Acetilcoenzima A/metabolismo , Acilcoenzima A/genética , Proteínas Bacterianas/genética , ADN Bacteriano/química , ADN Bacteriano/genética , Técnicas de Inactivación de Genes , Glioxilatos/metabolismo , Redes y Vías Metabólicas/genética , Datos de Secuencia Molecular , Oxo-Ácido-Liasas/genética , Análisis de Secuencia de ADN , Tioléster Hidrolasas/genética
9.
Mol Microbiol ; 73(6): 992-1008, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19703103

RESUMEN

Many organic substrates are metabolized via acetyl-coenzyme A (CoA) and enter central carbon metabolism at the level of this compound. We recently described the outlines of the ethylmalonyl-CoA pathway, a new acetyl-CoA assimilation strategy that operates in a number of bacteria such as Rhodobacter sphaeroides, Methylobacterium extorquens and streptomycetes and replaces the glyoxylate cycle. This new pathway involves a unique central reaction sequence catalysed by characteristic enzymes. Here, we identified and characterized (2S)-methylsuccinyl-CoA dehydrogenase from R. sphaeroides, a flavin adenine dinucleotide-containing enzyme that catalyses the last unknown step in the central part of the ethylmalonyl-CoA pathway, the oxidation of (2S)-methylsuccinyl-CoA to mesaconyl-(C1)-CoA. This enzyme is highly specific for its substrate and forms a distinct subgroup within the superfamily of flavin-dependent acyl-CoA dehydrogenases. Homology modelling and comparative sequence analyses with well-studied members of this superfamily identified amino acids that may contribute to the narrow substrate specificity of (2S)-methylsuccinyl-CoA dehydrogenase. The central part of the ethylmalonyl-CoA pathway was reconstituted in vitro using four recombinant enzymes. By this work, the ethylmalonyl-CoA pathway and its stereochemical course have been completely solved. This allowed defining the minimum set of enzymes necessary for its operation and to screen for further organisms following this acetyl-CoA assimilation strategy.


Asunto(s)
Acetilcoenzima A/metabolismo , Acilcoenzima A/metabolismo , Redes y Vías Metabólicas , Oxidorreductasas/metabolismo , Rhodobacter sphaeroides/enzimología , Secuencia de Aminoácidos , Coenzimas/farmacología , Flavina-Adenina Dinucleótido/farmacología , Datos de Secuencia Molecular , Homología de Secuencia de Aminoácido , Especificidad por Sustrato
10.
Proc Natl Acad Sci U S A ; 106(22): 8871-6, 2009 Jun 02.
Artículo en Inglés | MEDLINE | ID: mdl-19458256

RESUMEN

Chemo- and stereoselective reductions are important reactions in chemistry and biology, and reductases from biological sources are increasingly applied in organic synthesis. In contrast, carboxylases are used only sporadically. We recently described crotonyl-CoA carboxylase/reductase, which catalyzes the reduction of (E)-crotonyl-CoA to butyryl-CoA but also the reductive carboxylation of (E)-crotonyl-CoA to ethylmalonyl-CoA. In this study, the complete stereochemical course of both reactions was investigated in detail. The pro-(4R) hydrogen of NADPH is transferred in both reactions to the re face of the C3 position of crotonyl-CoA. In the course of the carboxylation reaction, carbon dioxide is incorporated in anti fashion at the C2 atom of crotonyl-CoA. For the reduction reaction that yields butyryl-CoA, a solvent proton is added in anti fashion instead of the CO(2). Amino acid sequence analysis showed that crotonyl-CoA carboxylase/reductase is a member of the medium-chain dehydrogenase/reductase superfamily and shares the same phylogenetic origin. The stereospecificity of the hydride transfer from NAD(P)H within this superfamily is highly conserved, although the substrates and reduction reactions catalyzed by its individual representatives differ quite considerably. Our findings led to a reassessment of the stereospecificity of enoyl(-thioester) reductases and related enzymes with respect to their amino acid sequence, revealing a general pattern of stereospecificity that allows the prediction of the stereochemistry of the hydride transfer for enoyl reductases of unknown specificity. Further considerations on the reaction mechanism indicated that crotonyl-CoA carboxylase/reductase may have evolved from enoyl-CoA reductases. This may be useful for protein engineering of enoyl reductases and their application in biocatalysis.


Asunto(s)
Acilcoenzima A/química , Acil-CoA Deshidrogenasas/química , NADH NADPH Oxidorreductasas/química , Catálisis , Oxidorreductasas actuantes sobre Donantes de Grupo CH-CH , Estereoisomerismo
11.
J Bacteriol ; 191(14): 4572-81, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19429610

RESUMEN

A 3-hydroxypropionate/4-hydroxybutyrate cycle operates in autotrophic CO(2) fixation in various Crenarchaea, as studied in some detail in Metallosphaera sedula. This cycle and the autotrophic 3-hydroxypropionate cycle in Chloroflexus aurantiacus have in common the conversion of acetyl-coenzyme A (CoA) and two bicarbonates via 3-hydroxypropionate to succinyl-CoA. Both cycles require the reductive conversion of 3-hydroxypropionate to propionyl-CoA. In M. sedula the reaction sequence is catalyzed by three enzymes. The first enzyme, 3-hydroxypropionyl-CoA synthetase, catalyzes the CoA- and MgATP-dependent formation of 3-hydroxypropionyl-CoA. The next two enzymes were purified from M. sedula or Sulfolobus tokodaii and studied. 3-Hydroxypropionyl-CoA dehydratase, a member of the enoyl-CoA hydratase family, eliminates water from 3-hydroxypropionyl-CoA to form acryloyl-CoA. Acryloyl-CoA reductase, a member of the zinc-containing alcohol dehydrogenase family, reduces acryloyl-CoA with NADPH to propionyl-CoA. Genes highly similar to the Metallosphaera CoA synthetase, dehydratase, and reductase genes were found in autotrophic members of the Sulfolobales. The encoded enzymes are only distantly related to the respective three enzyme domains of propionyl-CoA synthase from C. aurantiacus, where this trifunctional enzyme catalyzes all three reactions. This indicates that the autotrophic carbon fixation cycles in Chloroflexus and in the Sulfolobales evolved independently and that different genes/enzymes have been recruited in the two lineages that catalyze the same kinds of reactions.


Asunto(s)
Acilcoenzima A/metabolismo , Proteínas Arqueales/metabolismo , Enoil-CoA Hidratasa/metabolismo , Hidroxibutiratos/metabolismo , Oxidorreductasas/metabolismo , Propionatos/metabolismo , Sulfolobales/enzimología , Proteínas Arqueales/aislamiento & purificación , Enoil-CoA Hidratasa/genética , Enoil-CoA Hidratasa/aislamiento & purificación , Genes Arqueales , Redes y Vías Metabólicas , Modelos Biológicos , NADP/metabolismo , Oxidorreductasas/genética , Oxidorreductasas/aislamiento & purificación , Homología de Secuencia de Aminoácido , Sulfolobales/genética
12.
J Biol Chem ; 283(47): 32283-93, 2008 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-18819910

RESUMEN

Coenzyme B(12)-dependent mutases are radical enzymes that catalyze reversible carbon skeleton rearrangement reactions. Here we describe Rhodobacter sphaeroides ethylmalonyl-CoA mutase (Ecm), a novel member of the family of coenzyme B(12)-dependent acyl-CoA mutases, that operates in the recently discovered ethylmalonyl-CoA pathway for acetate assimilation. Ecm is involved in the central reaction sequence of this novel pathway and catalyzes the transformation of ethylmalonyl-CoA to methylsuccinyl-CoA in combination with a second enzyme that was further identified as promiscuous ethylmalonyl-CoA/methylmalonyl-CoA epimerase. In contrast to the epimerase, Ecm is highly specific for its substrate, ethylmalonyl-CoA, and accepts methylmalonyl-CoA only at 0.2% relative activity. Sequence analysis revealed that Ecm is distinct from (2R)-methylmalonyl-CoA mutase as well as isobutyryl-CoA mutase and defines a new subfamily of coenzyme B(12)-dependent acyl-CoA mutases. In combination with molecular modeling, two signature sequences were identified that presumably contribute to the substrate specificity of these enzymes.


Asunto(s)
Acilcoenzima A/química , Cobamidas/química , Transferasas Intramoleculares/química , Isomerasas/química , Rhodobacter sphaeroides/metabolismo , Clonación Molecular , Escherichia coli/genética , Transferasas Intramoleculares/fisiología , Espectroscopía de Resonancia Magnética , Modelos Biológicos , Modelos Químicos , Fenotipo , Filogenia , Proteínas Recombinantes/química , Programas Informáticos , Especificidad por Sustrato , Factores de Tiempo
13.
J Bacteriol ; 190(4): 1383-9, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18165310

RESUMEN

A modified 3-hydroxypropionate cycle has been proposed as the autotrophic CO2 fixation pathway for the thermoacidophilic crenarchaeon Metallosphaera sedula. The cycle requires the reductive conversion of 3-hydroxypropionate to propionyl-coenzyme A (propionyl-CoA). The specific activity of the 3-hydroxypropionate-, CoA-, and MgATP-dependent oxidation of NADPH in autotrophically grown cells was 0.023 micromol min(-1) mg protein(-1). The reaction sequence is catalyzed by at least two enzymes. The first enzyme, 3-hydroxypropionyl-CoA synthetase, catalyzes the following reaction: 3-hydroxypropionate + ATP + CoA --> 3-hydroxypropionyl-CoA + AMP + PP(i). The enzyme was purified 95-fold to a specific activity of 18 micromol min(-1) mg protein(-1) from autotrophically grown M. sedula cells. An internal peptide sequence was determined and a gene encoding a homologous protein identified in the genome of Sulfolobus tokodaii; similar genes were found in S. solfataricus and S. acidocaldarius. The gene was heterologously expressed in Escherichia coli, and the His-tagged protein was purified. Both the native enzyme from M. sedula and the recombinant enzyme from S. tokodaii not only activated 3-hydroxypropionate to its CoA ester but also activated propionate, acrylate, acetate, and butyrate; however, with the exception of propionate, the affinities for these substrates were reduced. 3-Hydroxypropionyl-CoA synthetase is up-regulated eightfold in autotrophically versus heterotrophically grown M. sedula, supporting its proposed role during CO2 fixation in this archaeon and possibly other members of the Sulfolobaceae family.


Asunto(s)
Proteínas Arqueales/metabolismo , Dióxido de Carbono/metabolismo , Coenzima A Ligasas/metabolismo , Sulfolobaceae/enzimología , Acilcoenzima A/química , Acilcoenzima A/metabolismo , Proteínas Arqueales/química , Proteínas Arqueales/genética , Coenzima A/química , Coenzima A/metabolismo , Coenzima A Ligasas/química , Coenzima A Ligasas/genética , Electroforesis en Gel de Poliacrilamida , Ácido Láctico/análogos & derivados , Ácido Láctico/química , Ácido Láctico/metabolismo , Malato Deshidrogenasa/metabolismo , Modelos Biológicos , Estructura Molecular , Especificidad por Sustrato , Sulfolobaceae/genética , Sulfolobaceae/metabolismo
14.
J Bacteriol ; 190(4): 1366-74, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18065535

RESUMEN

The coenzyme A (CoA)-activated C5-dicarboxylic acids mesaconyl-CoA and beta-methylmalyl-CoA play roles in two as yet not completely resolved central carbon metabolic pathways in bacteria. First, these compounds are intermediates in the 3-hydroxypropionate cycle for autotrophic CO2 fixation in Chloroflexus aurantiacus, a phototrophic green nonsulfur bacterium. Second, mesaconyl-CoA and beta-methylmalyl-CoA are intermediates in the ethylmalonyl-CoA pathway for acetate assimilation in various bacteria, e.g., in Rhodobacter sphaeroides, Methylobacterium extorquens, and Streptomyces species. In both cases, mesaconyl-CoA hydratase was postulated to catalyze the interconversion of mesaconyl-CoA and beta-methylmalyl-CoA. The putative genes coding for this enzyme in C. aurantiacus and R. sphaeroides were cloned and heterologously expressed in Escherichia coli, and the proteins were purified and studied. The recombinant homodimeric 80-kDa proteins catalyzed the reversible dehydration of erythro-beta-methylmalyl-CoA to mesaconyl-CoA with rates of 1,300 micromol min(-1) mg protein(-1). Genes coding for similar enzymes with two (R)-enoyl-CoA hydratase domains are present in the genomes of Roseiflexus, Methylobacterium, Hyphomonas, Rhodospirillum, Xanthobacter, Caulobacter, Magnetospirillum, Jannaschia, Sagittula, Parvibaculum, Stappia, Oceanicola, Loktanella, Silicibacter, Roseobacter, Roseovarius, Dinoroseobacter, Sulfitobacter, Paracoccus, and Ralstonia species. A similar yet distinct class of enzymes containing only one hydratase domain was found in various other bacteria, such as Streptomyces species. The role of this widely distributed new enzyme is discussed.


Asunto(s)
Proteínas Bacterianas/metabolismo , Hidroliasas/metabolismo , Acilcoenzima A/química , Acilcoenzima A/metabolismo , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Chloroflexus/metabolismo , Cromatografía Líquida de Alta Presión , Cromatografía en Capa Delgada , Clonación Molecular , Coenzima A/química , Coenzima A/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Hidroliasas/química , Hidroliasas/genética , Espectroscopía de Resonancia Magnética , Methylobacterium extorquens/genética , Methylobacterium extorquens/metabolismo , Modelos Biológicos , Estructura Molecular , Rhodobacter sphaeroides/genética , Rhodobacter sphaeroides/metabolismo , Espectrometría de Masa por Ionización de Electrospray , Espectrofotometría Ultravioleta , Streptomyces/genética , Streptomyces/metabolismo , Especificidad por Sustrato
15.
Proc Natl Acad Sci U S A ; 104(25): 10631-6, 2007 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-17548827

RESUMEN

Fifty years ago, Kornberg and Krebs established the glyoxylate cycle as the pathway for the synthesis of cell constituents from C2-units. However, since then, many bacteria have been described that do not contain isocitrate lyase, the key enzyme of this pathway. Here, a pathway termed the ethylmalonyl-CoA pathway operating in such organisms is described. Isotopically labeled acetate and bicarbonate were transformed to ethylmalonyl-CoA by cell extracts of acetate-grown, isocitrate lyase-negative Rhodobacter sphaeroides as determined by NMR spectroscopy. Crotonyl-CoA carboxylase/reductase, catalyzing crotonyl-CoA + CO2 + NADPH --> ethylmalonyl-CoA- + NADP+ was identified as the key enzyme of the ethylmalonyl-CoA pathway. The reductive carboxylation of an enoyl-thioester is a unique biochemical reaction, unprecedented in biology. The enzyme from R. sphaeroides was heterologously produced in Escherichia coli and characterized. Crotonyl-CoA carboxylase/reductase (or its gene) can be used as a marker for the presence of the ethylmalonyl-CoA pathway, which functions not only in acetyl-CoA assimilation. In Streptomyces sp., it may also supply precursors (ethylmalonyl-CoA) for antibiotic biosynthesis. For methylotrophic bacteria such as Methylobacterium extorquens, extension of the serine cycle with reactions of the ethylmalonyl-CoA pathway leads to a simplified scheme for isocitrate lyase-independent C1 assimilation.


Asunto(s)
Acilcoenzima A/metabolismo , Carboxiliasas/metabolismo , Ácidos Dicarboxílicos/metabolismo , Oxidorreductasas/metabolismo , Antibacterianos/biosíntesis , Cromatografía de Afinidad , Cromatografía DEAE-Celulosa , Cromatografía Líquida de Alta Presión , Ácidos Dicarboxílicos/química , Electroforesis en Gel de Poliacrilamida , Methylobacterium extorquens/enzimología , Modelos Biológicos , Resonancia Magnética Nuclear Biomolecular , Rhodobacter sphaeroides/enzimología , Streptomyces/metabolismo
16.
J Bacteriol ; 189(7): 2906-14, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17259315

RESUMEN

The autotrophic CO(2) fixation pathway (3-hydroxypropionate cycle) in Chloroflexus aurantiacus results in the fixation of two molecules of bicarbonate into one molecule of glyoxylate. Glyoxylate conversion to the CO(2) acceptor molecule acetyl-coenzyme A (CoA) requires condensation with propionyl-CoA (derived from one molecule of acetyl-CoA and one molecule of CO(2)) to beta-methylmalyl-CoA, which is converted to citramalyl-CoA. Extracts of autotrophically grown cells contained both S- and R-citramalyl-CoA lyase activities, which formed acetyl-CoA and pyruvate. Pyruvate is taken out of the cycle and used for cellular carbon biosynthesis. Both the S- and R-citramalyl-CoA lyases were up-regulated severalfold during autotrophic growth. S-Citramalyl-CoA lyase activity was found to be due to l-malyl-CoA lyase/beta-methylmalyl-CoA lyase. This promiscuous enzyme is involved in the CO(2) fixation pathway, forms acetyl-CoA and glyoxylate from l-malyl-CoA, and condenses glyoxylate with propionyl-CoA to beta-methylmalyl-CoA. R-Citramalyl-CoA lyase was further studied. Its putative gene was expressed and the recombinant protein was purified. This new enzyme belongs to the 3-hydroxy-3-methylglutaryl-CoA lyase family and is a homodimer with 34-kDa subunits that was 10-fold stimulated by adding Mg(2) or Mn(2+) ions and dithioerythritol. The up-regulation under autotrophic conditions suggests that the enzyme functions in the ultimate step of the acetyl-CoA regeneration route in C. aurantiacus. Genes similar to those involved in CO(2) fixation in C. aurantiacus, including an R-citramalyl-CoA lyase gene, were found in Roseiflexus sp., suggesting the operation of the 3-hydroxypropionate cycle in this bacterium. Incomplete sets of genes were found in aerobic phototrophic bacteria and in the gamma-proteobacterium Congregibacter litoralis. This may indicate that part of the reactions may be involved in a different metabolic process.


Asunto(s)
Liasas de Carbono-Carbono/metabolismo , Chloroflexus/enzimología , Ácido Láctico/análogos & derivados , Secuencia de Aminoácidos , Dióxido de Carbono/metabolismo , Liasas de Carbono-Carbono/genética , Gammaproteobacteria/enzimología , Glioxilatos/metabolismo , Cinética , Ácido Láctico/metabolismo , Datos de Secuencia Molecular , Filogenia , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/metabolismo , Alineación de Secuencia , Homología de Secuencia de Aminoácido
17.
J Bacteriol ; 188(18): 6460-8, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16952935

RESUMEN

The phototrophic bacterium Chloroflexus aurantiacus uses the 3-hydroxypropionate cycle for autotrophic CO(2) fixation. This cycle starts with acetyl-coenzyme A (CoA) and produces glyoxylate. Glyoxylate is an unconventional cell carbon precursor that needs special enzymes for assimilation. Glyoxylate is combined with propionyl-CoA to beta-methylmalyl-CoA, which is converted to citramalate. Cell extracts catalyzed the succinyl-CoA-dependent conversion of citramalate to acetyl-CoA and pyruvate, the central cell carbon precursor. This reaction is due to the combined action of enzymes that were upregulated during autotrophic growth, a coenzyme A transferase with the use of succinyl-CoA as the CoA donor and a lyase cleaving citramalyl-CoA to acetyl-CoA and pyruvate. Genomic analysis identified a gene coding for a putative coenzyme A transferase. The gene was heterologously expressed in Escherichia coli and shown to code for succinyl-CoA:d-citramalate coenzyme A transferase. This enzyme, which catalyzes the reaction d-citramalate + succinyl-CoA --> d-citramalyl-CoA + succinate, was purified and studied. It belongs to class III of the coenzyme A transferase enzyme family, with an aspartate residue in the active site. The homodimeric enzyme composed of 44-kDa subunits was specific for succinyl-CoA as a CoA donor but also accepted d-malate and itaconate instead of d-citramalate. The CoA transferase gene is part of a cluster of genes which are cotranscribed, including the gene for d-citramalyl-CoA lyase. It is proposed that the CoA transferase and the lyase catalyze the last two steps in the glyoxylate assimilation route.


Asunto(s)
Acilcoenzima A/metabolismo , Chloroflexus/enzimología , Coenzima A Transferasas/metabolismo , Ácido Láctico/análogos & derivados , Malatos/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/fisiología , Sitios de Unión/genética , Chloroflexus/genética , Chloroflexus/metabolismo , Clonación Molecular , Coenzima A Transferasas/genética , Coenzima A Transferasas/aislamiento & purificación , Dimerización , Escherichia coli/genética , Expresión Génica , Genes Bacterianos , Ácido Láctico/metabolismo , Peso Molecular , Familia de Multigenes , Subunidades de Proteína , ARN Bacteriano/análisis , ARN Mensajero/análisis , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/aislamiento & purificación , Especificidad por Sustrato , Succinatos/metabolismo , Transcripción Genética
18.
Mol Microbiol ; 61(2): 297-309, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16856937

RESUMEN

Organisms, which grow on organic substrates that are metabolized via acetyl-CoA, are faced with the problem to form all cell constituents from this C(2)-unit. The problem was solved by the seminal work of Kornberg and is known as the glyoxylate cycle. However, many bacteria are known to not contain isocitrate lyase, the key enzyme of this pathway. This problem was addressed in acetate-grown Rhodobacter sphaeroides. An acetate-minus mutant identified by transposon mutagenesis was affected in the gene for beta-ketothiolase forming acetoacetyl-CoA from two molecules of acetyl-CoA. This enzyme activity was missing in this mutant, which grew on acetoacetate and on acetate plus glyoxylate. A second acetate/acetoacetate-minus mutant was affected in the gene for a putative mesaconyl-CoA hydratase, an enzyme which catalyses the hydration of mesaconyl-CoA to beta-methylmalyl-CoA. Beta-methylmalyl-CoA is further cleaved into glyoxylate and propionyl-CoA. These results as well as identification of acetate-upregulated proteins by two-dimensional gel electrophoresis lead to the proposal of a new pathway for acetate assimilation. In a first part, affected by the mutations, two molecules of acetyl-CoA and one molecule CO(2) are converted via acetoacetyl-CoA and mesaconyl-CoA to glyoxylate and propionyl-CoA. In a second part glyoxylate and propionyl-CoA are converted with another molecule of acetyl-CoA and CO(2) to l-malyl-CoA and succinyl-CoA.


Asunto(s)
Acetatos/metabolismo , Glioxilatos/metabolismo , Rhodobacter sphaeroides/metabolismo , Acetilcoenzima A/metabolismo , Acetil-CoA C-Aciltransferasa/genética , Acetil-CoA C-Aciltransferasa/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Dióxido de Carbono/metabolismo , Elementos Transponibles de ADN , Enzimas/genética , Enzimas/metabolismo , Orden Génico , Genoma Bacteriano , Familia de Multigenes , Mutagénesis Sitio-Dirigida/métodos , Mutación , Rhodobacter sphaeroides/genética
19.
J Bacteriol ; 188(7): 2646-55, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16547052

RESUMEN

The 3-hydroxypropionate cycle has been proposed to operate as the autotrophic CO2 fixation pathway in the phototrophic bacterium Chloroflexus aurantiacus. In this pathway, acetyl coenzyme A (acetyl-CoA) and two bicarbonate molecules are converted to malate. Acetyl-CoA is regenerated from malyl-CoA by L-malyl-CoA lyase. The enzyme forming malyl-CoA, succinyl-CoA:L-malate coenzyme A transferase, was purified. Based on the N-terminal amino acid sequence of its two subunits, the corresponding genes were identified on a gene cluster which also contains the gene for L-malyl-CoA lyase, the subsequent enzyme in the pathway. Both enzymes were severalfold up-regulated under autotrophic conditions, which is in line with their proposed function in CO2 fixation. The two CoA transferase genes were cloned and heterologously expressed in Escherichia coli, and the recombinant enzyme was purified and studied. Succinyl-CoA:L-malate CoA transferase forms a large (alphabeta)n complex consisting of 46- and 44-kDa subunits and catalyzes the reversible reaction succinyl-CoA + L-malate --> succinate + L-malyl-CoA. It is specific for succinyl-CoA as the CoA donor but accepts L-citramalate instead of L-malate as the CoA acceptor; the corresponding d-stereoisomers are not accepted. The enzyme is a member of the class III of the CoA transferase family. The demonstration of the missing CoA transferase closes the last gap in the proposed 3-hydroxypropionate cycle.


Asunto(s)
Acilcoenzima A/metabolismo , Chloroflexus/enzimología , Coenzima A Transferasas/metabolismo , Ácido Láctico/análogos & derivados , Transferasas/metabolismo , Catálisis , Chloroflexus/citología , Chloroflexus/genética , Chloroflexus/metabolismo , Clonación Molecular , Ácido Láctico/metabolismo , Subunidades de Proteína , Transferasas/genética , Transferasas/aislamiento & purificación
20.
J Bacteriol ; 187(4): 1415-25, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15687206

RESUMEN

Cell extracts of Rhodobacter capsulatus grown on acetate contained an apparent malate synthase activity but lacked isocitrate lyase activity. Therefore, R. capsulatus cannot use the glyoxylate cycle for acetate assimilation, and a different pathway must exist. It is shown that the apparent malate synthase activity is due to the combination of a malyl-coenzyme A (CoA) lyase and a malyl-CoA-hydrolyzing enzyme. Malyl-CoA lyase activity was 20-fold up-regulated in acetate-grown cells versus glucose-grown cells. Malyl-CoA lyase was purified 250-fold with a recovery of 6%. The enzyme catalyzed not only the reversible condensation of glyoxylate and acetyl-CoA to L-malyl-CoA but also the reversible condensation of glyoxylate and propionyl-CoA to beta-methylmalyl-CoA. Enzyme activity was stimulated by divalent ions with preference for Mn(2+) and was inhibited by EDTA. The N-terminal amino acid sequence was determined, and a corresponding gene coding for a 34.2-kDa protein was identified and designated mcl1. The native molecular mass of the purified protein was 195 +/- 20 kDa, indicating a homohexameric composition. A homologous mcl1 gene was found in the genomes of the isocitrate lyase-negative bacteria Rhodobacter sphaeroides and Rhodospirillum rubrum in similar genomic environments. For Streptomyces coelicolor and Methylobacterium extorquens, mcl1 homologs are located within gene clusters implicated in acetate metabolism. We therefore propose that L-malyl-CoA/beta-methylmalyl-CoA lyase encoded by mcl1 is involved in acetate assimilation by R. capsulatus and possibly other glyoxylate cycle-negative bacteria.


Asunto(s)
Ácido Acético/metabolismo , Oxo-Ácido-Liasas/metabolismo , Rhodobacter capsulatus/enzimología , Acetilcoenzima A/metabolismo , Acilcoenzima A/metabolismo , Cationes Bivalentes/farmacología , Coenzimas/farmacología , ADN Bacteriano/química , Ácido Edético/farmacología , Inhibidores Enzimáticos/farmacología , Genes Bacterianos , Glioxilatos/metabolismo , Isocitratoliasa/metabolismo , Methylobacterium extorquens/genética , Datos de Secuencia Molecular , Peso Molecular , Sistemas de Lectura Abierta , Oxo-Ácido-Liasas/aislamiento & purificación , Subunidades de Proteína/química , Rhodobacter capsulatus/metabolismo , Rhodobacter sphaeroides/genética , Rhodospirillum rubrum/genética , Análisis de Secuencia de ADN , Análisis de Secuencia de Proteína , Homología de Secuencia , Streptomyces coelicolor/genética , Especificidad por Sustrato
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA