Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
J Ind Microbiol Biotechnol ; 46(7): 899-909, 2019 Jul.
Article in English | MEDLINE | ID: mdl-30963328

ABSTRACT

Lactoyl-CoA is critical for the biosynthesis of biodegradable and biocompatible lactate-based copolymers, which have wide applications. However, reports on acetyl-CoA: lactate CoA-transferases (ALCTs) are rare. To exploit novel ALCTs, amino acid sequence similarity searches based on the CoA-transferases from Clostridium propionicum and Megasphaera elsdenii were conducted. Two known and three novel enzymes were expressed, purified and characterized. Three novel ALCTs were identified, one each from Megasphaera sp. DISK 18, Clostridium lactatifermentans An75 and Firmicutes bacterium CAG: 466. ME-PCT from Megasphaera elsdenii had the highest catalytic efficiency for both acetyl-CoA (264.22 s-1 mM-1) and D-lactate (84.18 s-1 mM-1) with a broad temperature range for activity and good stability. This study, therefore, offers novel and efficient enzymes for lactoyl-CoA generation. To our best knowledge, this is the first report on the systematic mining of ALCTs, which offers valuable new tools for the engineering of pathways that rely on these enzymes.


Subject(s)
Acyl Coenzyme A/metabolism , Coenzyme A-Transferases/metabolism , Acetyl Coenzyme A/metabolism , Clostridium/metabolism , Coenzyme A-Transferases/genetics , Coenzyme A-Transferases/isolation & purification , Lactic Acid/metabolism
2.
J Biosci Bioeng ; 122(6): 660-665, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27342638

ABSTRACT

Thermostable enzymes are required for the rapid and sustainable production of polyhydroxyalkanoate (PHA) in vitro. The in vitro synthesis of PHA using the engineered thermostable synthase PhaC1SG(STQK) has been reported; however, the non-thermostable enzymes acetyl-CoA synthetase (ACS) and CoA transferase (CT) from mesophilic strains were used as monomer-supplying enzymes in this system. In the present study, acs and ct were cloned from the thermophilic bacteria Pelotomaculum thermopropionicum JCM10971 and Thermus thermophilus JCM10941 to construct an in vitro PHA synthesis system using only thermostable enzymes. ACS from P. thermopropionicum (ACSPt) and CT from T. thermophilus (CTTt) were confirmed to have high thermostability, and their optimal temperatures were around 60°C and 75°C, respectively. The in vitro PHA synthesis was successfully performed by ACSPt, CTTt, PhaC1SG(STQK), and poly(3-hydroxybutyrate) [P(3HB)] was synthesized at 45°C. Furthermore, the yields of P(3HB) and P(lactate-co-3HB) at 37°C were 1.4-fold higher than those of the in vitro synthesis system with non-thermostable ACS and CT from mesophilic strains. Overall, the thermostable ACS and CT were demonstrated to be useful for the efficient in vitro PHA synthesis at relatively high temperatures.


Subject(s)
Acetate-CoA Ligase/metabolism , Acyltransferases/metabolism , Coenzyme A-Transferases/metabolism , Peptococcaceae/enzymology , Polyhydroxyalkanoates/biosynthesis , Thermus thermophilus/enzymology , 3-Hydroxybutyric Acid/metabolism , Acetate-CoA Ligase/isolation & purification , Acetyl Coenzyme A/metabolism , Acyltransferases/isolation & purification , Coenzyme A-Transferases/isolation & purification , Enzyme Stability , Hydroxybutyrates/metabolism , Lactic Acid/metabolism , Peptococcaceae/metabolism , Polyesters/metabolism , Temperature , Thermus thermophilus/metabolism
3.
PLoS One ; 8(7): e67901, 2013.
Article in English | MEDLINE | ID: mdl-23935849

ABSTRACT

Many food plants accumulate oxalate, which humans absorb but do not metabolize, leading to the formation of urinary stones. The commensal bacterium Oxalobacter formigenes consumes oxalate by converting it to oxalyl-CoA, which is decarboxylated by oxalyl-CoA decarboxylase (OXC). OXC and the class III CoA-transferase formyl-CoA:oxalate CoA-transferase (FCOCT) are widespread among bacteria, including many that have no apparent ability to degrade or to resist external oxalate. The EvgA acid response regulator activates transcription of the Escherichia coli yfdXWUVE operon encoding YfdW (FCOCT), YfdU (OXC), and YfdE, a class III CoA-transferase that is ~30% identical to YfdW. YfdW and YfdU are necessary and sufficient for oxalate-induced protection against a subsequent acid challenge; neither of the other genes has a known function. We report the purification, in vitro characterization, 2.1-Å crystal structure, and functional assignment of YfdE. YfdE and UctC, an orthologue from the obligate aerobe Acetobacter aceti, perform the reversible conversion of acetyl-CoA and oxalate to oxalyl-CoA and acetate. The annotation of YfdE as acetyl-CoA:oxalate CoA-transferase (ACOCT) expands the scope of metabolic pathways linked to oxalate catabolism and the oxalate-induced acid tolerance response. FCOCT and ACOCT active sites contain distinctive, conserved active site loops (the glycine-rich loop and the GNxH loop, respectively) that appear to encode substrate specificity.


Subject(s)
Coenzyme A-Transferases/chemistry , Coenzyme A-Transferases/metabolism , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Escherichia coli/chemistry , Amino Acid Sequence , Catalytic Domain , Chromatography, High Pressure Liquid , Coenzyme A-Transferases/isolation & purification , Crystallography, X-Ray , Escherichia coli/genetics , Escherichia coli Proteins/isolation & purification , Genes, Bacterial/genetics , Kinetics , Models, Molecular , Molecular Sequence Data , Oxalates/chemistry , Oxalates/metabolism , Protein Multimerization , Substrate Specificity
4.
J Lipid Res ; 53(1): 66-76, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22021638

ABSTRACT

The human bile acid pool composition is composed of both primary bile acids (cholic acid and chenodeoxycholic acid) and secondary bile acids (deoxycholic acid and lithocholic acid). Secondary bile acids are formed by the 7α-dehydroxylation of primary bile acids carried out by intestinal anaerobic bacteria. We have previously described a multistep biochemical pathway in Clostridium scindens that is responsible for bile acid 7α-dehydroxylation. We have identified a large (12 kb) bile acid inducible (bai) operon in this bacterium that encodes eight genes involved in bile acid 7α-dehydroxylation. However, the function of the baiF gene product in this operon has not been elucidated. In the current study, we cloned and expressed the baiF gene in E. coli and discovered it has bile acid CoA transferase activity. In addition, we discovered a second bai operon encoding three genes. The baiK gene in this operon was expressed in E. coli and found to encode a second bile acid CoA transferase. Both bile acid CoA transferases were determined to be members of the type III family by amino acid sequence comparisons. Both bile acid CoA transferases had broad substrate specificity, except the baiK gene product, which failed to use lithocholyl-CoA as a CoA donor. Primary bile acids are ligated to CoA via an ATP-dependent mechanism during the initial steps of 7α-dehydroxylation. The bile acid CoA transferases conserve the thioester bond energy, saving the cell ATP molecules during bile acid 7α-dehydroxylation. ATP-dependent CoA ligation is likely quickly supplanted by ATP-independent CoA transfer.


Subject(s)
Clostridium/enzymology , Coenzyme A-Transferases/metabolism , Amino Acid Sequence , Bile Acids and Salts/metabolism , Cloning, Molecular , Coenzyme A-Transferases/chemistry , Coenzyme A-Transferases/isolation & purification , Escherichia coli/enzymology , Hydroxysteroid Dehydrogenases/metabolism , Intestines/microbiology , Molecular Sequence Data , Multigene Family , Substrate Specificity
5.
Mol Microbiol ; 68(3): 642-56, 2008 May.
Article in English | MEDLINE | ID: mdl-18331473

ABSTRACT

Filamentous fungi metabolize toxic propionyl-CoA via the methylcitrate cycle. Disruption of the methylcitrate synthase gene leads to an accumulation of propionyl-CoA and attenuates virulence of Aspergillus fumigatus. However, addition of acetate, but not ethanol, to propionate-containing medium strongly reduces the accumulation of propionyl-CoA and restores growth of the methylcitrate synthase mutant. Therefore, the existence of a CoA-transferase was postulated, which transfers the CoASH moiety from propionyl-CoA to acetate and, thereby, detoxifying the cell. In this study, we purified the responsible protein from Aspergillus nidulans and characterized its biochemical properties. The enzyme used succinyl-, propionyl- and acetyl-CoA as CoASH donors and the corresponding acids as acceptor molecules. Although the protein displayed high sequence similarity to acetyl-CoA hydrolases this activity was hardly detectable. We additionally identified and deleted the coding DNA sequence of the CoA-transferase. The mutant displayed weak phenotypes in the presence of propionate and behaved like the wild type when no propionate was present. However, when a double-deletion mutant defective in both methylcitrate synthase and CoA-transferase was constructed, the resulting strain was unable to grow on media containing acetate and propionate as sole carbon sources, which confirmed the in vivo activity of the CoA-transferase.


Subject(s)
Acyl Coenzyme A/metabolism , Aspergillus nidulans/enzymology , Coenzyme A-Transferases/genetics , Coenzyme A-Transferases/metabolism , Aspergillus nidulans/growth & development , Carbon/metabolism , Citrate (si)-Synthase/genetics , Citrate (si)-Synthase/metabolism , Cloning, Molecular , Coenzyme A-Transferases/chemistry , Coenzyme A-Transferases/isolation & purification , DNA, Complementary/genetics , Enzyme Stability , Fungal Proteins/analysis , Fungal Proteins/genetics , Fungal Proteins/isolation & purification , Fungal Proteins/metabolism , Genetic Complementation Test , Kinetics , Phenotype , Sequence Deletion , Substrate Specificity
6.
J Biol Chem ; 283(3): 1411-1418, 2008 Jan 18.
Article in English | MEDLINE | ID: mdl-18024431

ABSTRACT

Acetate:succinate CoA-transferases (ASCT) are acetate-producing enzymes in hydrogenosomes, anaerobically functioning mitochondria and in the aerobically functioning mitochondria of trypanosomatids. Although acetate is produced in the hydrogenosomes of a number of anaerobic microbial eukaryotes such as Trichomonas vaginalis, no acetate producing enzyme has ever been identified in these organelles. Acetate production is the last unidentified enzymatic reaction of hydrogenosomal carbohydrate metabolism. We identified a gene encoding an enzyme for acetate production in the genome of the hydrogenosome-containing protozoan parasite T. vaginalis. This gene shows high similarity to Saccharomyces cerevisiae acetyl-CoA hydrolase and Clostridium kluyveri succinyl-CoA:CoA-transferase. Here we demonstrate that this protein is expressed and is present in the hydrogenosomes where it functions as the T. vaginalis acetate:succinate CoA-transferase (TvASCT). Heterologous expression of TvASCT in CHO cells resulted in the expression of an active ASCT. Furthermore, homologous overexpression of the TvASCT gene in T. vaginalis resulted in an equivalent increase in ASCT activity. It was shown that the CoA transferase activity is succinate-dependent. These results demonstrate that this acetyl-CoA hydrolase/transferase homolog functions as the hydrogenosomal ASCT of T. vaginalis. This is the first hydrogenosomal acetate-producing enzyme to be identified. Interestingly, TvASCT does not share any similarity with the mitochondrial ASCT from Trypanosoma brucei, the only other eukaryotic succinate-dependent acetyl-CoA-transferase identified so far. The trichomonad enzyme clearly belongs to a distinct class of acetate:succinate CoA-transferases. Apparently, two completely different enzymes for succinate-dependent acetate production have evolved independently in ATP-generating organelles.


Subject(s)
Coenzyme A-Transferases/metabolism , Organelles/enzymology , Trichomonas vaginalis/enzymology , Acetates/metabolism , Acetyl Coenzyme A/metabolism , Amino Acid Sequence , Animals , Antibodies, Protozoan/immunology , Antibody Specificity , CHO Cells , Chromatography, Ion Exchange , Coenzyme A-Transferases/chemistry , Coenzyme A-Transferases/isolation & purification , Cricetinae , Cricetulus , Genes, Protozoan , Kinetics , Molecular Sequence Data , Peptides/chemistry , Protein Transport , Recombinant Proteins/metabolism , Sequence Alignment , Subcellular Fractions/enzymology , Succinic Acid/metabolism , Trichomonas vaginalis/genetics
7.
Biochemistry ; 46(38): 10852-63, 2007 Sep 25.
Article in English | MEDLINE | ID: mdl-17718512

ABSTRACT

Succinyl-CoA:3-ketoacid CoA transferase (SCOT) transfers CoA from succinyl-CoA to acetoacetate via a thioester intermediate with its active site glutamate residue, Glu 305. When CoA is linked to the enzyme, a cysteine residue can now be rapidly modified by 5,5'-dithiobis(2-nitrobenzoic acid), reflecting a conformational change of SCOT upon formation of the thioester. Since either Cys 28 or Cys 196 could be the target, each was mutated to Ser to distinguish between them. Like wild-type SCOT, the C196S mutant protein was modified rapidly in the presence of acyl-CoA substrates. In contrast, the C28S mutant protein was modified much more slowly under identical conditions, indicating that Cys 28 is the residue exposed on binding CoA. The specific activity of the C28S mutant protein was unexpectedly lower than that of wild-type SCOT. X-ray crystallography revealed that Ser adopts a different conformation than the native Cys. A chloride ion is bound to one of four active sites in the crystal structure of the C28S mutant protein, mimicking substrate, interacting with Lys 329, Asn 51, and Asn 52. On the basis of these results and the studies of the structurally similar CoA transferase from Escherichia coli, YdiF, bound to CoA, the conformational change in SCOT was deduced to be a domain rotation of 17 degrees coupled with movement of two loops: residues 321-329 that bury Cys 28 and interact with succinate or acetoacetate and residues 374-386 that interact with CoA. Modeling this conformational change has led to the proposal of a new mechanism for catalysis by SCOT.


Subject(s)
Amino Acid Substitution , Coenzyme A-Transferases/chemistry , Coenzyme A/metabolism , Cysteine/chemistry , Myocardium/enzymology , Amino Acid Sequence , Animals , Binding Sites , Coenzyme A/chemistry , Coenzyme A-Transferases/isolation & purification , Coenzyme A-Transferases/metabolism , Crystallography, X-Ray/methods , Escherichia coli/enzymology , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/isolation & purification , Escherichia coli Proteins/metabolism , Glutamic Acid/chemistry , Models, Chemical , Models, Molecular , Protein Binding , Protein Conformation , Protein Structure, Tertiary/genetics , Sequence Homology, Amino Acid , Serine/chemistry , Swine
8.
Biochemistry ; 46(25): 7365-73, 2007 Jun 26.
Article in English | MEDLINE | ID: mdl-17536835

ABSTRACT

Mature amyloid fibrils are believed to be formed by the lateral association of discrete structural units designated as protofibrils, but this lateral association of protofibrils has never been directly observed. We have recently characterized a thioesterase from Alcaligenes faecalis, which was shown to exist as homomeric oligomers with an average diameter of 21.6 nm consisting of 22 kDa subunits in predominantly beta-sheet structure. In this study, we have shown that upon incubation in a 75% ethanol solution, the oligomeric particles of protein were transformed into amyloid-like fibrils. TEM pictures obtained at various stages during fibril growth helped us to understand to a certain extent the early events in the fibrillization process. When incubated in 75% ethanol, oligomeric particles of protein grew to approximately 35-40 nm in diameter before fusion. Fusion of two oligomers of 35-40 nm resulted in the formation of a fibril. Fibril formation was accompanied by a reduction in the diameter of the particle to approximately 20-25 nm along with concomitant elongation to approximately 110 nm, indicating reorganization and strengthening of the structure. The elongation process continued by sequential addition of oligomeric units to give fibers 500-1000 nm in length with a further reduction in diameter to 17-20 nm. Further elongation resulted in the formation of fibers that were more than 4000 nm in length; the diameter, however, remained constant at 17-20 nm. These data clearly show that the mature fibrils have assembled via longitudinal growth of oligomers and not via lateral association of protofibrils.


Subject(s)
Amyloid/biosynthesis , Amyloid/chemistry , Bacterial Proteins/chemistry , Alcaligenes faecalis/enzymology , Amyloid/ultrastructure , Bacterial Proteins/isolation & purification , Birefringence , Buffers , Circular Dichroism , Coenzyme A-Transferases/chemistry , Coenzyme A-Transferases/isolation & purification , Ethanol/chemistry , Hydrogen-Ion Concentration , Molecular Weight , Phosphates/chemistry , Protein Structure, Secondary , Solutions/chemistry , Temperature , Time Factors
9.
Protein Expr Purif ; 53(2): 396-403, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17363272

ABSTRACT

The Bacillus subtilis genes scoA and scoB encode subunits of the heteromeric enzyme ScoAB, a putative succinyl-CoA:acetoacetate coenzyme A transferase. High-throughput, ligation-independent cloning (LIC) vectors used extensively for production and purification of single proteins were modified to allow simultaneous expression of interacting proteins and selective purification of functional complexes. Transfer of the LIC region of vector pMCSG7 (L. Stols, M. Gu, L. Dieckman, R. Raffen, F.R. Collart, M.I. Donnelly. A new vector for high-throughput, ligation-independent cloning encoding a tobacco etch virus protease cleavage site. Protein Expr. Purif. (2002) 25, 8-15) into commercial vectors with alternative, compatible origins of replication allowed introduction of standard LIC PCR products into the vectors by uniform protocols. Replacement of the His-tag encoding region of pMCSG7 with a sequence encoding the S-tag enabled selective purification of interacting proteins based on the His-tag associated with one member of the complex. When expressed separately and mixed, the ScoAB subunits failed to interact productively; no transferase activity was detected, and S-tagged ScoB failed to co-purify with His-tagged ScoA. Co-expression, in contrast, generated active transferase that catalyzed the predicted reaction. The ScoAB complex was purified by standard high-throughput metal-ion affinity chromatography procedures, crystallized robotically, and its structure was determined by molecular replacement.


Subject(s)
Bacillus subtilis/enzymology , Bacillus subtilis/genetics , Bacterial Proteins/genetics , Coenzyme A-Transferases/genetics , Genetic Vectors , Bacterial Proteins/isolation & purification , Bacterial Proteins/metabolism , Base Sequence , Cloning, Molecular , Coenzyme A-Transferases/isolation & purification , Coenzyme A-Transferases/metabolism , Crystallization , Crystallography, X-Ray , DNA, Bacterial/genetics , Gene Expression , Genes, Bacterial , Models, Molecular , Protein Subunits , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism
10.
J Bacteriol ; 188(18): 6460-8, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16952935

ABSTRACT

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.


Subject(s)
Acyl Coenzyme A/metabolism , Chloroflexus/enzymology , Coenzyme A-Transferases/metabolism , Lactic Acid/analogs & derivatives , Malates/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/physiology , Binding Sites/genetics , Chloroflexus/genetics , Chloroflexus/metabolism , Cloning, Molecular , Coenzyme A-Transferases/genetics , Coenzyme A-Transferases/isolation & purification , Dimerization , Escherichia coli/genetics , Gene Expression , Genes, Bacterial , Lactic Acid/metabolism , Molecular Weight , Multigene Family , Protein Subunits , RNA, Bacterial/analysis , RNA, Messenger/analysis , Recombinant Proteins/biosynthesis , Recombinant Proteins/isolation & purification , Substrate Specificity , Succinates/metabolism , Transcription, Genetic
11.
Microbiology (Reading) ; 150(Pt 11): 3821-3829, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15528667

ABSTRACT

Data are presented from two-dimensional (2-D) PAGE analysis of Mycobacterium tuberculosis strain Harlingen grown during aerobic and anaerobic culture, according to a modified Wayne dormancy model. M. tuberculosis cultures were grown to the transition point between exponential growth and stationary phase in the presence of oxygen (7 days) and then part of the cultures was shifted to anaerobic conditions for 16 days. Growth declined similarly during aerobic and anaerobic conditions, whereas the ATP consumption rapidly decreased in the anaerobic cultures. 2-D PAGE revealed 50 protein spots that were either unique to, or more abundant during, anaerobic conditions and 16 of these were identified by MALDI-TOF. These proteins were the alpha-crystalline homologue (HspX), elongation factor Tu (Tuf), GroEL2, succinyl-CoA : 3-oxoacid-CoA transferase (ScoB), mycolic acid synthase (CmaA2), thioredoxin (TrxB2), beta-ketoacyl-ACP synthase (KasB), l-alanine dehydrogenase (Ald), Rv2005c, Rv2629, Rv0560c, Rv2185c and Rv3866. Some protein spots were found to be proteolytic fragments, e.g. HspX and GroEL2. These data suggest that M. tuberculosis induces expression of about 1 % of its genes in response to dormancy.


Subject(s)
Mycobacterium tuberculosis/chemistry , Mycobacterium tuberculosis/growth & development , Proteome/analysis , 3-Oxoacyl-(Acyl-Carrier-Protein) Synthase/analysis , 3-Oxoacyl-(Acyl-Carrier-Protein) Synthase/isolation & purification , Adaptation, Physiological , Adenosine Triphosphate/metabolism , Aerobiosis , Alanine Dehydrogenase , Amino Acid Oxidoreductases/analysis , Amino Acid Oxidoreductases/isolation & purification , Anaerobiosis , Antigens, Bacterial/analysis , Antigens, Bacterial/isolation & purification , Bacterial Proteins/analysis , Bacterial Proteins/isolation & purification , Chaperonin 60/analysis , Chaperonin 60/isolation & purification , Coenzyme A-Transferases/analysis , Coenzyme A-Transferases/isolation & purification , Electrophoresis, Gel, Two-Dimensional , Fatty Acid Synthases/analysis , Fatty Acid Synthases/isolation & purification , Gene Expression Regulation, Bacterial , Mycobacterium tuberculosis/metabolism , Peptide Elongation Factor Tu/analysis , Peptide Elongation Factor Tu/isolation & purification , Proteome/isolation & purification , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Thioredoxins/analysis , Thioredoxins/isolation & purification
12.
Acta Crystallogr D Biol Crystallogr ; 59(Pt 7): 1276-7, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12832784

ABSTRACT

Formyl-CoA transferase from Oxalobacter formigenes has been expressed as a recombinant protein in Escherichia coli and purified to homogeneity. Crystals of formyl-CoA transferase were grown at 293 K using polyethylene glycol 4000 as a precipitant. The diffraction pattern of flash-frozen crystals at 100 K extends to 2.2 A resolution with synchrotron radiation (lambda = 0.933 nm). The crystals are tetragonal and belong to space group I4, with unit-cell parameters a = b = 151.44, c = 99.49 A. The asymmetric unit contains one dimer and the solvent content is 53%. Formyl-CoA transferase was crystallized both as the apoenzyme and as its complex with coenzyme A.


Subject(s)
Coenzyme A-Transferases/chemistry , Oxalobacter formigenes/enzymology , Apoenzymes/chemistry , Coenzyme A/chemistry , Coenzyme A-Transferases/genetics , Coenzyme A-Transferases/isolation & purification , Crystallization , Crystallography, X-Ray , Polyethylene Glycols , Recombinant Proteins
13.
Biochemistry ; 41(49): 14455-62, 2002 Dec 10.
Article in English | MEDLINE | ID: mdl-12463743

ABSTRACT

Ketoacidosis affects patients who are deficient in the enzyme activity of succinyl-CoA:3-ketoacid CoA transferase (SCOT), since SCOT catalyses the activation of acetoacetate in the metabolism of ketone bodies. Thus far, structure/function analysis of the mammalian enzyme has been predicted based on the three-dimensional structure of a CoA transferase determined from an anaerobic bacterium that utilizes its enzyme for glutamate fermentation. To better interpret clinical data, we have determined the structure of a mammalian CoA transferase from pig heart by X-ray crystallography to 2.5 A resolution. Instrumental to the structure determination were selenomethionine substitution and the use of argon during purification and crystallization. Although pig heart SCOT adopts an alpha/beta protein fold, resembling the overall fold of the bacterial CoA transferase, several loops near the active site of pig heart SCOT follow different paths than the corresponding loops in the bacterial enzyme, accounting for differences in substrate specificities. Two missense mutations found associated with SCOT of ketoacidosis patients were mapped to a location in the structure that might disrupt the stabilization of the amino-terminal strand and thereby interfere with the proper folding of the protein into a functional enzyme.


Subject(s)
Coenzyme A-Transferases/chemistry , Myocardium/enzymology , Amino Acid Sequence , Argon , Binding Sites , Coenzyme A-Transferases/isolation & purification , Crystallization , Crystallography, X-Ray/methods , Models, Molecular , Molecular Sequence Data , Selenium/chemistry , Sequence Homology, Amino Acid
14.
Mol Hum Reprod ; 8(1): 16-23, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11756565

ABSTRACT

Succinyl CoA: 3-oxo acid CoA transferase (scot; EC 2.8.3.5) is a key enzyme for metabolism of ketone bodies. Previously, we have cloned a testis-specific succinyl CoA: 3-oxo acid CoA transferase (scot-t) cDNA from a subtracted cDNA library of mouse testis and demonstrated its expression to be specific to late haploid male germ cells. In this study, the human orthologue of mouse scot-t was cloned and characterized. The entire coding region of the mRNA and the deduced amino acid sequence of human Scot-t (h-Scot-t) showed 75.4 and 75.8% identity with mouse scot-t respectively. The mRNA was exclusively expressed in the testis, and the protein was localized to the midpiece of ejaculated spermatozoa where mitochondria exist. We showed that the h-Scot-t gene was intronless by using a polymerase chain reaction technique and that a non-functional pseudogene, 98% similar to h-Scot-t, was also located on the human genome (1p33-34.3). Furthermore, the genomic structure of the actual h-Scot-t transcription unit was found to be located in an intron (1p34.1-35.3) of the bone morphogenetic protein 8 (BMP8) gene. In conclusion, we have demonstrated that h-Scot-t is a single intronless gene specifically expressed in the testis.


Subject(s)
Coenzyme A-Transferases/genetics , Coenzyme A-Transferases/metabolism , Testis/enzymology , Amino Acid Sequence , Animals , Base Sequence , Cloning, Molecular , Coenzyme A-Transferases/chemistry , Coenzyme A-Transferases/isolation & purification , Humans , Male , Mice , Molecular Sequence Data , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Alignment , Spermatozoa/cytology , Spermatozoa/enzymology , Tissue Distribution
15.
Eur J Biochem ; 269(1): 372-80, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11784332

ABSTRACT

Propionate CoA-transferase from Clostridium propionicum has been purified and the gene encoding the enzyme has been cloned and sequenced. The enzyme was rapidly and irreversibly inactivated by sodium borohydride or hydroxylamine in the presence of propionyl-CoA. The reduction of the thiol ester between a catalytic site glutamate and CoA with borohydride and the cleavage by hydroxylamine were used to introduce a site-specific label, which was followed by MALDI-TOF-MS. This allowed the identification of glutamate 324 at the active site. Propionate CoA-transferase and similar proteins deduced from the genomes of Escherichia coli, Staphylococcus aureus, Bacillus halodurans and Aeropyrum pernix are proposed to form a novel subclass of CoA-transferases. Secondary structure element predictions were generated and compared to known crystal structures in the databases. A high degree of structural similarity was observed between the arrangement of secondary structure elements in these proteins and glutaconate CoA-transferase from Acidaminococcus fermentans.


Subject(s)
Clostridium/enzymology , Coenzyme A-Transferases/chemistry , Amino Acid Sequence , Binding Sites , Cloning, Molecular , Coenzyme A-Transferases/genetics , Coenzyme A-Transferases/isolation & purification , Glutamic Acid , Molecular Sequence Data
16.
J Bacteriol ; 184(1): 207-15, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11741862

ABSTRACT

The degradation of 3-oxoadipate in Pseudomonas sp. strain B13 was investigated and was shown to proceed through 3-oxoadipyl-coenzyme A (CoA) to give acetyl-CoA and succinyl-CoA. 3-Oxoadipate:succinyl-CoA transferase of strain B13 was purified by heat treatment and chromatography on phenyl-Sepharose, Mono-Q, and Superose 6 gels. Estimation of the native molecular mass gave a value of 115,000 +/- 5,000 Da with a Superose 12 column. Polyacrylamide gel electrophoresis under denaturing conditions resulted in two distinct bands of equal intensities. The subunit A and B values were 32,900 and 27,000 Da. Therefore it can be assumed that the enzyme is a heterotetramer of the type A2B2 with a molecular mass of 120,000 Da. The N-terminal amino acid sequences of both subunits are as follows: subunit A, AELLTLREAVERFVNDGTVALEGFTHLIPT; subunit B, SAYSTNEMMTVAAARRLKNGAVVFV. The pH optimum was 8.4. Km values were 0.4 and 0.2 mM for 3-oxoadipate and succinyl-CoA, respectively. Reversibility of the reaction with succinate was shown. The transferase of strain B13 failed to convert 2-chloro- and 2-methyl-3-oxoadipate. Some activity was observed with 4-methyl-3-oxoadipate. Even 2-oxoadipate and 3-oxoglutarate were shown to function as poor substrates of the transferase. 3-oxoadipyl-CoA thiolase was purified by chromatography on DEAE-Sepharose, blue 3GA, and reactive brown-agarose. Estimation of the native molecular mass gave 162,000 +/- 5,000 Da with a Superose 6 column. The molecular mass of the subunit of the denatured protein, as determined by sodium dodecyl sulfate-polyacrylamide gel electrophoresis, was 42 kDa. On the basis of these results, 3-oxoadipyl-CoA thiolase should be a tetramer of the type A4. The N-terminal amino acid sequence of 3-oxoadipyl-CoA thiolase was determined to be SREVYI-DAVRTPIGRFG. The pH optimum was 7.8. Km values were 0.15 and 0.01 mM for 3-oxoadipyl-CoA and CoA, respectively. Sequence analysis of the thiolase terminus revealed high percentages of identity (70 to 85%) with thiolases of different functions. The N termini of the transferase subunits showed about 30 to 35% identical amino acids with the glutaconate-CoA transferase of an anaerobic bacterium but only an identity of 25% with the respective transferases of aromatic compound-degrading organisms was found.


Subject(s)
Acetyl-CoA C-Acyltransferase/metabolism , Adipates/metabolism , Benzoates/metabolism , Chlorobenzenes/metabolism , Coenzyme A-Transferases/metabolism , Pseudomonas/enzymology , Acetyl-CoA C-Acyltransferase/isolation & purification , Amino Acid Sequence , Biodegradation, Environmental , Coenzyme A-Transferases/isolation & purification , Molecular Sequence Data , Molecular Weight , Sequence Analysis, Protein , Substrate Specificity
17.
Proc Natl Acad Sci U S A ; 98(13): 7146-51, 2001 Jun 19.
Article in English | MEDLINE | ID: mdl-11416199

ABSTRACT

The tyrosine nitration of proteins has been observed in diverse inflammatory conditions and has been linked to the presence of reactive nitrogen species. From many in vitro experiments, it is apparent that tyrosine nitration may alter the function of proteins. A limited number of experiments under in vivo conditions also demonstrate that protein nitration is associated with altered cellular processes. To understand the association of protein nitration with the pathogenic mechanism of the disease, it is essential to identify specific protein targets of nitration with in vivo or intact tissue models. Using anti-nitrotyrosine antibodies, we demonstrated the accumulation of nitrotyrosine in a 52-kDa protein in rat kidney after lipopolysaccharide treatment. The 52-kDa protein was purified and identified with partial sequence as succinyl-CoA:3-oxoacid CoA-transferase (SCOT; EC ). Western blot analysis revealed that the nitration of this mitochondrial enzyme increased in the kidneys and hearts of lipopolysaccharide-treated rats, whereas its catalytic activity decreased. These data suggest that tyrosine nitration may be a mechanism for the inhibition of SCOT activity in inflammatory conditions. SCOT is a key enzyme for ketone body utilization. Thus, tyrosine nitration of the enzyme with sepsis or inflammation may explain the altered metabolism of ketone bodies present in these disorders.


Subject(s)
Coenzyme A-Transferases/metabolism , Endotoxins/pharmacology , Kidney/enzymology , Mitochondria, Heart/enzymology , Mitochondria/enzymology , Tyrosine/analogs & derivatives , Tyrosine/metabolism , Amino Acid Sequence , Animals , Brain/enzymology , Brain/metabolism , Coenzyme A-Transferases/chemistry , Coenzyme A-Transferases/isolation & purification , Escherichia coli , Lipopolysaccharides/pharmacology , Male , Rats , Rats, Wistar , Thiobarbituric Acid Reactive Substances/analysis
18.
J Bacteriol ; 183(14): 4288-95, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11418570

ABSTRACT

Anaerobic microbial toluene catabolism is initiated by addition of fumarate to the methyl group of toluene, yielding (R)-benzylsuccinate as first intermediate, which is further metabolized via beta-oxidation to benzoyl-coenzyme A (CoA) and succinyl-CoA. A specific succinyl-CoA:(R)-benzylsuccinate CoA-transferase activating (R)-benzylsuccinate to the CoA-thioester was purified and characterized from Thauera aromatica. The enzyme is fully reversible and forms exclusively the 2-(R)-benzylsuccinyl-CoA isomer. Only some close chemical analogs of the substrates are accepted by the enzyme: succinate was partially replaced by maleate or methylsuccinate, and (R)-benzylsuccinate was replaced by methylsuccinate, benzylmalonate, or phenylsuccinate. In contrast to all other known CoA-transferases, the enzyme consists of two subunits of similar amino acid sequences and similar sizes (44 and 45 kDa) in an alpha(2)beta(2) conformation. Identity of the subunits with the products of the previously identified toluene-induced bbsEF genes was confirmed by determination of the exact masses via electrospray-mass spectrometry. The deduced amino acid sequences resemble those of only two other characterized CoA-transferases, oxalyl-CoA:formate CoA-transferase and (E)-cinnamoyl-CoA:(R)-phenyllactate CoA-transferase, which represent a new family of CoA-transferases. As suggested by kinetic analysis, the reaction mechanism of enzymes of this family apparently involves formation of a ternary complex between the enzyme and the two substrates.


Subject(s)
Acyl Coenzyme A/metabolism , Coenzyme A-Transferases/metabolism , Succinates/metabolism , Thauera/enzymology , Toluene/metabolism , Anaerobiosis , Catalysis , Coenzyme A-Transferases/antagonists & inhibitors , Coenzyme A-Transferases/isolation & purification , Oxygen/metabolism
19.
Biochemistry ; 40(8): 2455-67, 2001 Feb 27.
Article in English | MEDLINE | ID: mdl-11327867

ABSTRACT

Pig heart succinate-coenzyme A transferase (succinyl-coenzyme A: 3-oxoacid coenzyme A transferase; E. C. 2.8.3.5.), a dimeric enzyme purified by affinity chromatography on Procion Blue MX-2G Sepharose, reacts with acetoacetyl-coenzyme A to form a covalent enzyme-coenzyme A thiolester intermediate in which the active site glutamate (E344) of both subunits each forms thiolester links with coenzyme A. Reaction of this dimeric enzyme-coenzyme A species with sodium borohydride leads to inactivation of the enzyme and reduction of the thiolester on both subunits to the corresponding enzyme alcohol, as judged by electrospray mass spectrometry. Reaction of the dimeric enzyme-coenzyme A intermediate with either succinate or acetoacetate, however, results in only one-half of the coenzyme A being transferred to the acceptor carboxylate to form either succinyl-coenzyme A or acetoacetyl-coenzyme A. Reaction of this latter enzyme species with borohydride caused no loss of enzyme activity despite the reduction of the remaining half of the enzyme-coenzyme A thiolester to the enzyme alcohol. That this catalytic asymmetry existed between subunits within the same enzyme dimer was demonstrated by showing that the enzyme species, created by successive reaction with acetoacetyl-coenzyme A and succinate, bound to Blue MX-2G Sepharose through the remaining available active site and could be eluted as a single chromatographic species by succinyl-coenzyme A. It is concluded that while both of the subunits of the succinate-coenzyme A transferase dimer are able to form enzyme-coenzyme A thiolester intermediates, only one subunit is competent to transfer the coenzyme A moiety to a carboxylic acid acceptor to form the new acyl-coenzyme A product. The possible structural basis for this catalytic asymmetry and its mechanistic implications are discussed.


Subject(s)
Coenzyme A-Transferases/metabolism , Myocardium/enzymology , Peptide Fragments/metabolism , Acyl Coenzyme A/chemistry , Acyl Coenzyme A/metabolism , Animals , Borohydrides , Carboxylic Acids/metabolism , Catalysis , Chromatography, Agarose , Coenzyme A-Transferases/antagonists & inhibitors , Coenzyme A-Transferases/isolation & purification , Dimerization , Esters , Mitochondria, Heart/enzymology , Sepharose/analogs & derivatives , Spectrometry, Mass, Electrospray Ionization/methods , Substrate Specificity , Sulfhydryl Compounds/metabolism , Swine
20.
Biochemistry ; 39(37): 11291-302, 2000 Sep 19.
Article in English | MEDLINE | ID: mdl-10985774

ABSTRACT

Pig heart CoA transferase (EC 2.8.3.5) has been shown previously to adopt a homodimeric structure, in which each subunit has a molecular weight of 52 197 and consists of N- and C-domains linked by a hydrophilic linker or "hinge". Here we identify and characterize a second oligomeric form constituent in purified enzyme preparations, albeit at low concentrations. Both species catalyze the transfer of CoA with similar values for k(cat) and K(M). This second form sediments more rapidly than the homodimer under the conditions of conventional sedimentation velocity and active enzyme centrifugation. Apparent molecular weight values determined by sedimentation equilibrium and gel filtration chromatography are 4-fold greater than the subunit molecular weight, confirming that this form is a homotetramer. The subunits of both oligomeric forms are indistinguishable with respect to molecular mass, far-UV CD, intrinsic tryptophan fluorescence, and equilibrium unfolding. Dissociation of the homotetramer to the homodimer occurs very slowly in benign solutions containing high salt concentrations (0.25-2.0 M KCl). The homotetramer is fully converted to homodimer during refolding from denaturant at low protein concentrations. Disruption of the hydrophilic linker between the N- and C-domains by mutagenesis or mild proteolysis causes a decrease in the relative amount of the larger conformer. The homotetramer is stabilized by interactions involving the helical hinge region, and a substantial kinetic barrier hinders interconversion of the two oligomeric species under nondenaturing conditions.


Subject(s)
Coenzyme A-Transferases/chemistry , Myocardium/enzymology , Animals , Catalysis , Coenzyme A-Transferases/genetics , Coenzyme A-Transferases/isolation & purification , Coenzyme A-Transferases/metabolism , Dimerization , Enzyme Activation , Enzyme Stability/genetics , Evolution, Molecular , Isoenzymes/chemistry , Isoenzymes/genetics , Isoenzymes/isolation & purification , Isoenzymes/metabolism , Kinetics , Molecular Weight , Mutagenesis, Site-Directed , Protein Conformation , Protein Folding , Protein Structure, Tertiary/genetics , Swine , Ultracentrifugation
SELECTION OF CITATIONS
SEARCH DETAIL