Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 74
Filter
Add more filters










Publication year range
1.
Biochem Biophys Res Commun ; 693: 149377, 2024 Jan 22.
Article in English | MEDLINE | ID: mdl-38101000

ABSTRACT

In most of the eukaryotes and archaea, isopentenyl pyrophosphate (IPP) and dimethyl allyl pyrophosphate (DMAPP) essential building blocks of all isoprenoids synthesized in the mevalonate pathway. Here, the first enzyme of this pathway, acetoacetyl CoA thiolase (PFC_04095) from an archaea Pyrococcus furiosus is structurally characterized. The crystal structure of PFC_04095 is determined at 2.7 Å resolution, and the crystal structure reveals the absence of catalytic acid/base cysteine in its active site, which is uncommon in thiolases. In place of cysteine, His285 of HDAF motif performs both protonation and abstraction of proton during the reaction. The crystal structure shows that the distance between Cys83 and His335 is 5.4 Å. So, His335 could not abstract a proton from nucleophilic cysteine (Cys83), resulting in the loss of enzymatic activity of PFC_04095. MD simulations of the docked PFC_04095-acetyl CoA complex show substrate binding instability to the active site pocket. Here, we have reported that the stable binding of acetyl CoA to the PFC_04095 pocket requires the involvement of three protein complexes, i.e., thiolase (PFC_04095), DUF35 (PFC_04100), and HMGCS (PFC_04090).


Subject(s)
Acetyl-CoA C-Acetyltransferase , Pyrococcus furiosus , Acetyl-CoA C-Acetyltransferase/chemistry , Acetyl Coenzyme A/metabolism , Pyrococcus furiosus/metabolism , Cysteine/metabolism , Protons , Models, Molecular
2.
Biochem Biophys Res Commun ; 688: 149151, 2023 12 25.
Article in English | MEDLINE | ID: mdl-37951156

ABSTRACT

Fusobacterium nucleatum (F. nucleatum) is an anaerobic gram-negative bacterium that was previously thought to be related to the progression of colorectal cancer. In F. nucleatum, thiolase participates in fatty acid metabolism, and it can catalyse the transfer of an acetyl group from acetyl-CoA to another molecule, typically a fatty acid or another molecule in the synthesis of lipids. To gain deeper insight into the molecular mechanism governing the function of thiolase in F. nucleatum (Fn0495), we herein report the structure of Fn0495. The monomer of Fn0495 consists of three subdomains, namely, the N-terminal domain (residues 1-117 and 252-270), the C-terminal domain (residues 273-393), and the loop domain (residues 118-251). Fn0495 shows a unique difference in the charge and structure of the substrate binding pocket compared with homologous proteins. This research found three conserved residues (Cys88, His357, and Cys387) in Fn0495 arranged near a potential substrate binding pocket. In this study, the conformational changes between the covering loop, catalytic cysteine loop, regulatory determinant region, and homologous protein were compared. These results will enhance our understanding of the molecular characteristics and roles of the thiolase family.


Subject(s)
Acetyl-CoA C-Acetyltransferase , Fusobacterium nucleatum , Fusobacterium nucleatum/metabolism , Acetyl-CoA C-Acetyltransferase/chemistry , Acetyl Coenzyme A , Cysteine/metabolism , Fatty Acids
3.
Annu Rev Biochem ; 92: 351-384, 2023 06 20.
Article in English | MEDLINE | ID: mdl-37068769

ABSTRACT

Thiolases are CoA-dependent enzymes that catalyze the thiolytic cleavage of 3-ketoacyl-CoA, as well as its reverse reaction, which is the thioester-dependent Claisen condensation reaction. Thiolases are dimers or tetramers (dimers of dimers). All thiolases have two reactive cysteines: (a) a nucleophilic cysteine, which forms a covalent intermediate, and (b) an acid/base cysteine. The best characterized thiolase is the Zoogloea ramigera thiolase, which is a bacterial biosynthetic thiolase belonging to the CT-thiolase subfamily. The thiolase active site is also characterized by two oxyanion holes, two active site waters, and four catalytic loops with characteristic amino acid sequence fingerprints. Three thiolase subfamilies can be identified, each characterized by a unique sequence fingerprint for one of their catalytic loops, which causes unique active site properties. Recent insights concerning the thiolase reaction mechanism, as obtained from recent structural studies, as well as from classical and recent enzymological studies, are addressed, and open questions are discussed.


Subject(s)
Coenzyme A , Cysteine , Coenzyme A/chemistry , Coenzyme A/metabolism , Cysteine/metabolism , Models, Molecular , Acetyl-CoA C-Acetyltransferase/chemistry , Acetyl-CoA C-Acetyltransferase/metabolism , Catalytic Domain
4.
FEBS J ; 290(16): 3997-4022, 2023 08.
Article in English | MEDLINE | ID: mdl-37026388

ABSTRACT

Tuberculosis (TB) is one of the leading causes of human death caused by Mycobacterium tuberculosis (Mtb). Mtb can enter into a long-lasting persistence where it can utilize fatty acids as the carbon source. Hence, fatty acid metabolism pathway enzymes are considered promising and pertinent mycobacterial drug targets. FadA2 (thiolase) is one of the enzymes involved in Mtb's fatty acid metabolism pathway. FadA2 deletion construct (ΔL136-S150) was designed to produce soluble protein. The crystal structure of FadA2 (ΔL136-S150) at 2.9 Å resolution was solved and analysed for membrane-anchoring region. The four catalytic residues of FadA2 are Cys99, His341, His390 and Cys427, and they belong to four loops with characteristic sequence motifs, i.e., CxT, HEAF, GHP and CxA. FadA2 is the only thiolase of Mtb which belongs to the CHH category containing the HEAF motif. Analysing the substrate-binding channel, it has been suggested that FadA2 is involved in the ß-oxidation pathway, i.e., the degradative pathway, as the long-chain fatty acid can be accommodated in the channel. The catalysed reaction is favoured by the presence of two oxyanion holes, i.e., OAH1 and OAH2. OAH1 formation is unique in FadA2, formed by the NE2 of His390 present in the GHP motif and NE2 of His341 present in the HEAF motif, whereas OAH2 formation is similar to CNH category thiolase. Sequence and structural comparison with the human trifunctional enzyme (HsTFE-ß) suggests the membrane-anchoring region in FadA2. Molecular dynamics simulations of FadA2 with a membrane containing POPE lipid were conducted to understand the role of a long insertion sequence of FadA2 in membrane anchoring.


Subject(s)
Mycobacterium tuberculosis , Humans , Mycobacterium tuberculosis/metabolism , Substrate Specificity , Acetyl-CoA C-Acetyltransferase/chemistry , Acetyl-CoA C-Acetyltransferase/metabolism
5.
Biochem J ; 478(15): 3047-3062, 2021 08 13.
Article in English | MEDLINE | ID: mdl-34338286

ABSTRACT

Activation of enzymes by monovalent cations (M+) is a widespread phenomenon in biology. Despite this, there are few structure-based studies describing the underlying molecular details. Thiolases are a ubiquitous and highly conserved family of enzymes containing both K+-activated and K+-independent members. Guided by structures of naturally occurring K+-activated thiolases, we have used a structure-based approach to engineer K+-activation into a K+-independent thiolase. To our knowledge, this is the first demonstration of engineering K+-activation into an enzyme, showing the malleability of proteins to accommodate M+ ions as allosteric regulators. We show that a few protein structural features encode K+-activation in this class of enzyme. Specifically, two residues near the substrate-binding site are sufficient for K+-activation: A tyrosine residue is required to complete the K+ coordination sphere, and a glutamate residue provides a compensating charge for the bound K+ ion. Further to these, a distal residue is important for positioning a K+-coordinating water molecule that forms a direct hydrogen bond to the substrate. The stability of a cation-π interaction between a positively charged residue and the substrate is determined by the conformation of the loop surrounding the substrate-binding site. Our results suggest that this cation-π interaction effectively overrides K+-activation, and is, therefore, destabilised in K+-activated thiolases. Evolutionary conservation of these amino acids provides a promising signature sequence for predicting K+-activation in thiolases. Together, our structural, biochemical and bioinformatic work provide important mechanistic insights into how enzymes can be allosterically activated by M+ ions.


Subject(s)
Acetyl-CoA C-Acetyltransferase/metabolism , Bacterial Proteins/metabolism , Cations, Monovalent/metabolism , Enzyme Activation , Potassium/metabolism , Zoogloea/isolation & purification , Acetyl Coenzyme A/chemistry , Acetyl Coenzyme A/metabolism , Acetyl-CoA C-Acetyltransferase/chemistry , Acetyl-CoA C-Acetyltransferase/genetics , Acyl Coenzyme A/chemistry , Acyl Coenzyme A/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Biocatalysis , Cations, Monovalent/chemistry , Crystallography, X-Ray , Kinetics , Models, Molecular , Mutation , Potassium/chemistry , Protein Binding , Protein Conformation , Protein Engineering , Protein Multimerization , Substrate Specificity , Zoogloea/enzymology , Zoogloea/genetics
6.
Biochem Biophys Res Commun ; 533(3): 442-448, 2020 12 10.
Article in English | MEDLINE | ID: mdl-32972748

ABSTRACT

Bacillus cereus ATCC 14579 is a known polyhydroxybutyrate (PHB)-producing microorganism that possesses genes associated with PHB synthesis such as PhaA, PhaB, and PHA synthases. PhaA (i.e., thiolase) is the first enzyme in the PHA biosynthetic pathway, which catalyze the condensation of two acetyl-CoA molecules to acetoacetyl-CoA. Our study elucidated the crystal structure of PhaA in Bacillus cereus ATCC 14579 (BcTHL) in its apo- and CoA-bound forms. BcTHL adopts a type II biosynthetic thiolase structure by forming a tetramer. The crystal structure of CoA-complexed BcTHL revealed that the substrate binding site of BcTHL is constituted by different residues compared with other known thiolases. Our study also revealed that Arg221, a residue involved in ADP binding, undergoes a positional conformational change upon the binding of the CoA molecule.


Subject(s)
Acetyl-CoA C-Acetyltransferase/chemistry , Bacillus cereus/enzymology , Bacterial Proteins/chemistry , Catalytic Domain , Coenzyme A/chemistry , Crystallography, X-Ray , Hydroxybutyrates/metabolism , Models, Molecular
7.
Protein Expr Purif ; 170: 105592, 2020 06.
Article in English | MEDLINE | ID: mdl-32032770

ABSTRACT

Acetyl-CoA C-acetyltransferase synthase gene (AACT) cDNA, DNA and promoter were cloned from Sanghuangporus baumii. The gene ORF (1260 bp) encoded 419 amino acids. The AACT DNA includes five exons (1-84 bp, 140-513 bp, 570-1027 bp, 1090-1282 bp, 1344-1494 bp) and four introns (85-139 bp, 514-569 bp, 1028-1089 bp, 1283-1343 bp). The molecular weight of AACT protein is 43.40 kDa, it is hydrophilic with a theoretical isoelectric point of 8.96. Furthermore, The region of the transcription start site is 1997-2047 bp of AACT promoter, and it contained promoter elements (TATA Boxs, CAAT Boxs, CAAT-box, ABRE, G-Boxs, Sp1, MSA-like, LTR). AACT recombinant protein (43.40 KDa + Tag protein 22.68 KDa) was subjected in SDS-PAGE. AACT the transcription levels of in different development stages were investigated. The expression of AACT in primordia (2.4-fold) and 15 d mycelia (2.3- fold) were significantly higher than 9 d mycelia (contral). The expression level of the AACT downstream genes and triterpenoids content were determined at different developmental stages. Triterpenoid content reached its peak on day 15(7.21 mg/g).


Subject(s)
Acetyl Coenzyme A/chemistry , Acetyl-CoA C-Acetyltransferase/chemistry , Basidiomycota/enzymology , Fruiting Bodies, Fungal/enzymology , Fungal Proteins/chemistry , Mycelium/enzymology , Acetyl Coenzyme A/metabolism , Acetyl-CoA C-Acetyltransferase/genetics , Acetyl-CoA C-Acetyltransferase/metabolism , Basidiomycota/chemistry , Cloning, Molecular , Escherichia coli/genetics , Escherichia coli/metabolism , Exons , Fruiting Bodies, Fungal/chemistry , Fungal Proteins/genetics , Fungal Proteins/metabolism , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Hydrophobic and Hydrophilic Interactions , Introns , Isoelectric Point , Models, Molecular , Molecular Weight , Mycelium/chemistry , Open Reading Frames , Phylogeny , Promoter Regions, Genetic , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Triterpenes/isolation & purification , Triterpenes/metabolism
8.
Hum Mutat ; 40(10): 1641-1663, 2019 10.
Article in English | MEDLINE | ID: mdl-31268215

ABSTRACT

Mitochondrial acetoacetyl-CoA thiolase (T2, encoded by the ACAT1 gene) deficiency is an inherited disorder of ketone body and isoleucine metabolism. It typically manifests with episodic ketoacidosis. The presence of isoleucine-derived metabolites is the key marker for biochemical diagnosis. To date, 105 ACAT1 variants have been reported in 149 T2-deficient patients. The 56 disease-associated missense ACAT1 variants have been mapped onto the crystal structure of T2. Almost all these missense variants concern residues that are completely or partially buried in the T2 structure. Such variants are expected to cause T2 deficiency by having lower in vivo T2 activity because of lower folding efficiency and/or stability. Expression and activity data of 30 disease-associated missense ACAT1 variants have been measured by expressing them in human SV40-transformed fibroblasts. Only two variants (p.Cys126Ser and p.Tyr219His) appear to have equal stability as wild-type. For these variants, which are inactive, the side chains point into the active site. In patients with T2 deficiency, the genotype does not correlate with the clinical phenotype but exerts a considerable effect on the biochemical phenotype. This could be related to variable remaining residual T2 activity in vivo and has important clinical implications concerning disease management and newborn screening.


Subject(s)
Acetyl-CoA C-Acetyltransferase/genetics , Acetyl-CoA C-Acyltransferase/deficiency , Amino Acid Metabolism, Inborn Errors/genetics , Genetic Predisposition to Disease , Mutation , Acetyl-CoA C-Acetyltransferase/chemistry , Acetyl-CoA C-Acetyltransferase/metabolism , Acetyl-CoA C-Acyltransferase/genetics , Acetyl-CoA C-Acyltransferase/metabolism , Amino Acid Metabolism, Inborn Errors/diagnosis , Amino Acid Metabolism, Inborn Errors/metabolism , Animals , Gene Expression Regulation, Enzymologic , Genetic Association Studies , Genetic Variation , Humans , Metabolic Networks and Pathways , Models, Molecular , Phenotype , Protein Binding , Protein Conformation , Protein Interaction Domains and Motifs , Protein Multimerization , Structure-Activity Relationship
9.
Proc Natl Acad Sci U S A ; 115(13): 3380-3385, 2018 03 27.
Article in English | MEDLINE | ID: mdl-29531083

ABSTRACT

Many reactions within a cell are thermodynamically unfavorable. To efficiently run some of those endergonic reactions, nature evolved intermediate-channeling enzyme complexes, in which the products of the first endergonic reactions are immediately consumed by the second exergonic reactions. Based on this concept, we studied how archaea overcome the unfavorable first reaction of isoprenoid biosynthesis-the condensation of two molecules of acetyl-CoA to acetoacetyl-CoA catalyzed by acetoacetyl-CoA thiolases (thiolases). We natively isolated an enzyme complex comprising the thiolase and 3-hydroxy-3-methylglutaryl (HMG)-CoA synthase (HMGCS) from a fast-growing methanogenic archaeon, Methanothermococcus thermolithotrophicus HMGCS catalyzes the second reaction in the mevalonate pathway-the exergonic condensation of acetoacetyl-CoA and acetyl-CoA to HMG-CoA. The 380-kDa crystal structure revealed that both enzymes are held together by a third protein (DUF35) with so-far-unknown function. The active-site clefts of thiolase and HMGCS form a fused CoA-binding site, which allows for efficient coupling of the endergonic thiolase reaction with the exergonic HMGCS reaction. The tripartite complex is found in almost all archaeal genomes and in some bacterial ones. In addition, the DUF35 proteins are also important for polyhydroxyalkanoate (PHA) biosynthesis, most probably by functioning as a scaffold protein that connects thiolase with 3-ketoacyl-CoA reductase. This natural and highly conserved enzyme complex offers great potential to improve isoprenoid and PHA biosynthesis in biotechnologically relevant organisms.


Subject(s)
Acetyl Coenzyme A/metabolism , Acetyl-CoA C-Acetyltransferase/chemistry , Acetyl-CoA C-Acetyltransferase/metabolism , Acyl Coenzyme A/metabolism , Archaea/enzymology , Hydroxymethylglutaryl-CoA Synthase/chemistry , Hydroxymethylglutaryl-CoA Synthase/metabolism , Binding Sites , Catalysis , Catalytic Domain , Crystallography, X-Ray , Protein Conformation
10.
Proc Natl Acad Sci U S A ; 115(15): E3378-E3387, 2018 04 10.
Article in English | MEDLINE | ID: mdl-29581275

ABSTRACT

Mycobacterium tuberculosis (Mtb) grows on host-derived cholesterol during infection. IpdAB, found in all steroid-degrading bacteria and a determinant of pathogenicity, has been implicated in the hydrolysis of the last steroid ring. Phylogenetic analyses revealed that IpdAB orthologs form a clade of CoA transferases (CoTs). In a coupled assay with a thiolase, IpdAB transformed the cholesterol catabolite (R)-2-(2-carboxyethyl)-3-methyl-6-oxocyclohex-1-ene-1-carboxyl-CoA (COCHEA-CoA) and CoASH to 4-methyl-5-oxo-octanedioyl-CoA (MOODA-CoA) and acetyl-CoA with high specificity (kcat/Km = 5.8 ± 0.8 × 104 M-1⋅s-1). The structure of MOODA-CoA was consistent with IpdAB hydrolyzing COCHEA-CoA to a ß-keto-thioester, a thiolase substrate. Contrary to characterized CoTs, IpdAB exhibited no activity toward small CoA thioesters. Further, IpdAB lacks the catalytic glutamate residue that is conserved in the ß-subunit of characterized CoTs and a glutamyl-CoA intermediate was not trapped during turnover. By contrast, Glu105A, conserved in the α-subunit of IpdAB, was essential for catalysis. A crystal structure of the IpdAB·COCHEA-CoA complex, solved to 1.4 Å, revealed that Glu105A is positioned to act as a catalytic base. Upon titration with COCHEA-CoA, the E105AA variant accumulated a yellow-colored species (λmax = 310 nm; Kd = 0.4 ± 0.2 µM) typical of ß-keto enolates. In the presence of D2O, IpdAB catalyzed the deuteration of COCHEA-CoA adjacent to the hydroxylation site at rates consistent with kcat Based on these data and additional IpdAB variants, we propose a retro-Claisen condensation-like mechanism for the IpdAB-mediated hydrolysis of COCHEA-CoA. This study expands the range of known reactions catalyzed by the CoT superfamily and provides mechanistic insight into an important determinant of Mtb pathogenesis.


Subject(s)
Bacterial Proteins/metabolism , Cholesterol/metabolism , Hydrolases/metabolism , Mycobacterium tuberculosis/enzymology , Tuberculosis/microbiology , Virulence Factors/metabolism , Acetyl-CoA C-Acetyltransferase/chemistry , Acetyl-CoA C-Acetyltransferase/genetics , Acetyl-CoA C-Acetyltransferase/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Cholesterol/chemistry , Crystallography, X-Ray , Humans , Hydrolases/chemistry , Hydrolases/genetics , Kinetics , Models, Molecular , Mycobacterium tuberculosis/chemistry , Mycobacterium tuberculosis/classification , Mycobacterium tuberculosis/genetics , Phylogeny , Tuberculosis/metabolism , Virulence Factors/chemistry , Virulence Factors/genetics
11.
Biochemistry ; 57(8): 1338-1348, 2018 02 27.
Article in English | MEDLINE | ID: mdl-29360348

ABSTRACT

Thiolases catalyze the condensation of acyl-CoA thioesters through the Claisen condensation reaction. The best described enzymes usually yield linear condensation products. Using a combined computational/experimental approach, and guided by structural information, we have studied the potential of thiolases to synthesize branched compounds. We have identified a bulky residue located at the active site that blocks proper accommodation of substrates longer than acetyl-CoA. Amino acid replacements at such a position exert effects on the activity and product selectivity of the enzymes that are highly dependent on a protein scaffold. Among the set of five thiolases studied, Erg10 thiolase from Saccharomyces cerevisiae showed no acetyl-CoA/butyryl-CoA branched condensation activity, but variants at position F293 resulted the most active and selective biocatalysts for this reaction. This is the first time that a thiolase has been engineered to synthesize branched compounds. These novel enzymes enrich the toolbox of combinatorial (bio)chemistry, paving the way for manufacturing a variety of α-substituted synthons. As a proof of concept, we have engineered Clostridium's 1-butanol pathway to obtain 2-ethyl-1-butanol, an alcohol that is interesting as a branched model compound.


Subject(s)
Acetyl-CoA C-Acetyltransferase/metabolism , Acyl Coenzyme A/metabolism , Hexanols/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Acetyl-CoA C-Acetyltransferase/chemistry , Acetyl-CoA C-Acetyltransferase/genetics , Catalytic Domain , Metabolic Networks and Pathways , Models, Molecular , Protein Engineering/methods , Saccharomyces cerevisiae/chemistry , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics
12.
Acta Crystallogr F Struct Biol Commun ; 74(Pt 1): 6-13, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29372902

ABSTRACT

Thiolases are vital enzymes which participate in both degradative and biosynthetic pathways. Biosynthetic thiolases catalyze carbon-carbon bond formation by a Claisen condensation reaction. The cytoplasmic acetoacetyl-CoA thiolase from Saccharomyces cerevisiae, ERG10, catalyses carbon-carbon bond formation in the mevalonate pathway. The structure of a S. cerevisiae biosynthetic thiolase has not previously been reported. Here, crystal structures of apo ERG10 and its Cys91Ala variant were solved at resolutions of 2.2 and 1.95 Å, respectively. The structure determined shows that ERG10 shares the characteristic thiolase superfamily fold, with a similar active-site architecture to those of type II thiolases and a similar binding pocket, apart from Ala159 at the entrance to the pantetheine-binding cavity, which appears to be a determinant of the poor binding ability of the substrate. Moreover, comparative binding-pocket analysis of molecule B in the asymmetric unit of the apo structure with that of the CoA-bound complex of human mitochondrial acetoacetyl-CoA thiolase indicates the canonical binding mode of CoA. Furthermore, the steric hindrance revealed in a structural comparison of molecule A with the CoA-bound form raise the possibility of conformational changes that are associated with substrate binding.


Subject(s)
Acetyl-CoA C-Acetyltransferase/chemistry , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae/enzymology , Acetyl-CoA C-Acetyltransferase/genetics , Amino Acid Sequence , Amino Acid Substitution , Catalytic Domain , Crystallography, X-Ray , Cytoplasm/enzymology , Genes, Fungal , Humans , Models, Molecular , Mutagenesis, Site-Directed , Protein Structure, Quaternary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Sequence Homology, Amino Acid , Species Specificity , Static Electricity , Structural Homology, Protein
13.
ACS Infect Dis ; 3(9): 666-675, 2017 09 08.
Article in English | MEDLINE | ID: mdl-28786661

ABSTRACT

Mycobacterium tuberculosis (Mtb), the causative agent of tuberculosis (TB), is a highly successful human pathogen and has infected approximately one-third of the world's population. Multiple drug resistant (MDR) and extensively drug resistant (XDR) TB strains and coinfection with HIV have increased the challenges of successfully treating this disease pandemic. The metabolism of host cholesterol by Mtb is an important factor for both its virulence and pathogenesis. In Mtb, the cholesterol side chain is degraded through multiple cycles of ß-oxidation and FadA5 (Rv3546) catalyzes side chain thiolysis in the first two cycles. Moreover, FadA5 is important during the chronic stage of infection in a mouse model of Mtb infection. Here, we report the redox control of FadA5 catalytic activity that results from reversible disulfide bond formation between Cys59-Cys91 and Cys93-Cys377. Cys93 is the thiolytic nucleophile, and Cys377 is the general acid catalyst for cleavage of the ß-keto-acyl-CoA substrate. The disulfide bond formed between the two catalytic residues Cys93 and Cys377 blocks catalysis. The formation of the disulfide bonds is accompanied by a large domain swap at the FadA5 dimer interface that serves to bring Cys93 and Cys377 in close proximity for disulfide bond formation. The catalytic activity of FadA5 has a midpoint potential of -220 mV, which is close to the Mtb mycothiol potential in the activated macrophage. The redox profile of FadA5 suggests that FadA5 is fully active when Mtb resides in the unactivated macrophage to maximize flux into cholesterol catabolism. Upon activation of the macrophage, the oxidative shift in the mycothiol potential will decrease the thiolytic activity by 50%. Thus, the FadA5 midpoint potential is poised to rapidly restrict cholesterol side chain degradation in response to oxidative stress from the host macrophage environment.


Subject(s)
Acetyl-CoA C-Acetyltransferase/chemistry , Acetyl-CoA C-Acetyltransferase/metabolism , Bacterial Proteins/chemistry , Cholesterol/metabolism , Mycobacterium tuberculosis/pathogenicity , Sulfhydryl Compounds/metabolism , Bacterial Proteins/metabolism , Catalytic Domain , Circular Dichroism , Cysteine/metabolism , Humans , Macrophage Activation , Models, Molecular , Oxidation-Reduction , Protein Conformation
14.
Mol Genet Metab ; 121(2): 111-118, 2017 06.
Article in English | MEDLINE | ID: mdl-28396157

ABSTRACT

BACKGROUND: 3-Hydroxy-3-Methylglutaryl-Coenzyme A (HMG-CoA) lyase deficiency is a rare inborn error of leucine metabolism and ketogenesis. Despite recurrent hypoglycemia and metabolic decompensations, most patients have a good clinical and neurological outcome contrasting with abnormal brain magnetic resonance imaging (MRI) signals and consistent abnormal brain proton magnetic resonance spectroscopy (1H-MRS) metabolite peaks. Identifying these metabolites could provide surrogate markers of the disease and improve understanding of MRI-clinical discrepancy and follow-up of affected patients. METHODS: Urine samples, brain MRI and 1H-MRS in 5 patients with HMG-CoA lyase deficiency (4 boys and 1 girl aged from 25days to 10years) were, for each patient, obtained on the same day. Brain and urine spectroscopy were performed at the same pH by studying urine at pH 7.4. Due to pH-induced modifications in chemical shifts and because reference 1H NMR spectra are obtained at pH 2.5, spectroscopy of normal urine added with the suspected metabolite was further performed at this pH to validate the correct identification of compounds. RESULTS: Mild to extended abnormal white matter MRI signals were observed in all cases. Brain spectroscopy abnormal peaks at 0.8-1.1ppm, 1.2-1.4ppm and 2.4ppm were also detected by urine spectroscopy at pH 7.4. Taking into account pH-induced changes in chemical shifts, brain abnormal peaks in patients were formally identified to be those of 3-hydroxyisovaleric, 3-methylglutaconic, 3-methylglutaric and 3-hydroxy-3-methylglutaric acids. CONCLUSION: 3-Methylglutaric, 3-hydroxyisovaleric and 3-hydroxy-3-methylglutaric acids identified on urine 1H-NMR spectra of 5 patients with HMG-CoA lyase deficiency are responsible for the cerebral spectroscopy signature seen in these patients, validating their local involvement in brain and putative contribution to brain neuropathology.


Subject(s)
Acetyl-CoA C-Acetyltransferase/deficiency , Amino Acid Metabolism, Inborn Errors/metabolism , Amino Acid Metabolism, Inborn Errors/urine , Brain Chemistry , Brain/diagnostic imaging , Brain/metabolism , Meglutol/urine , Metabolomics/methods , Acetyl-CoA C-Acetyltransferase/chemistry , Acetyl-CoA C-Acetyltransferase/metabolism , Acetyl-CoA C-Acetyltransferase/urine , Amino Acid Metabolism, Inborn Errors/diagnostic imaging , Cerebellum/metabolism , Child , Child, Preschool , Female , Humans , Hydrogen-Ion Concentration , Infant , Infant, Newborn , Magnetic Resonance Imaging , Male , Meglutol/analogs & derivatives , Meglutol/metabolism , Proton Magnetic Resonance Spectroscopy , Urine/chemistry , Valerates/metabolism , White Matter/metabolism
15.
Protein Eng Des Sel ; 30(3): 225-233, 2017 03 01.
Article in English | MEDLINE | ID: mdl-28062645

ABSTRACT

C: Structures of the C123A variant of the dimeric Leishmania mexicana SCP2-thiolase (type-2) (Lm-thiolase), complexed with acetyl-CoA and acetoacetyl-CoA, respectively, are reported. The catalytic site of thiolase contains two oxyanion holes, OAH1 and OAH2, which are important for catalysis. The two structures reveal for the first time the hydrogen bond interactions of the CoA-thioester oxygen atom of the substrate with the hydrogen bond donors of OAH1 of a CHH-thiolase. The amino acid sequence fingerprints ( xS, EAF, G P) of three catalytic loops identify the active site geometry of the well-studied CNH-thiolases, whereas SCP2-thiolases (type-1, type-2) are classified as CHH-thiolases, having as corresponding fingerprints xS, DCF and G P. In all thiolases, OAH2 is formed by the main chain NH groups of two catalytic loops. In the well-studied CNH-thiolases, OAH1 is formed by a water (of the Wat-Asn(NEAF) dyad) and NE2 (of the GHP-histidine). In the two described liganded Lm-thiolase structures, it is seen that in this CHH-thiolase, OAH1 is formed by NE2 of His338 (HDCF) and His388 (GHP). Analysis of the OAH1 hydrogen bond networks suggests that the GHP-histidine is doubly protonated and positively charged in these complexes, whereas the HDCF histidine is neutral and singly protonated.


Subject(s)
Acetyl-CoA C-Acetyltransferase/chemistry , Leishmania mexicana/enzymology , Protozoan Proteins/chemistry , Catalytic Domain , Crystallography, X-Ray , Protein Structure, Secondary
16.
J Biol Chem ; 291(52): 26698-26706, 2016 Dec 23.
Article in English | MEDLINE | ID: mdl-27815501

ABSTRACT

Phylogenetically diverse microbes that produce long chain, olefinic hydrocarbons have received much attention as possible sources of renewable energy biocatalysts. One enzyme that is critical for this process is OleA, a thiolase superfamily enzyme that condenses two fatty acyl-CoA substrates to produce a ß-ketoacid product and initiates the biosynthesis of long chain olefins in bacteria. Thiolases typically utilize a ping-pong mechanism centered on an active site cysteine residue. Reaction with the first substrate produces a covalent cysteine-thioester tethered acyl group that is transferred to the second substrate through formation of a carbon-carbon bond. Although the basics of thiolase chemistry are precedented, the mechanism by which OleA accommodates two substrates with extended carbon chains and a coenzyme moiety-unusual for a thiolase-are unknown. Gaining insights into this process could enable manipulation of the system for large scale olefin production with hydrocarbon chains lengths equivalent to those of fossil fuels. In this study, mutagenesis of the active site cysteine in Xanthomonas campestris OleA (Cys143) enabled trapping of two catalytically relevant species in crystals. In the resulting structures, long chain alkyl groups (C12 and C14) and phosphopantetheinate define three substrate channels in a T-shaped configuration, explaining how OleA coordinates its two substrates and product. The C143A OleA co-crystal structure possesses a single bound acyl-CoA representing the Michaelis complex with the first substrate, whereas the C143S co-crystal structure contains both acyl-CoA and fatty acid, defining how a second substrate binds to the acyl-enzyme intermediate. An active site glutamate (Gluß117) is positioned to deprotonate bound acyl-CoA and initiate carbon-carbon bond formation.


Subject(s)
Acetyl-CoA C-Acetyltransferase/metabolism , Acyl Coenzyme A/metabolism , Alkenes/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Coenzyme A/metabolism , Xanthomonas campestris/enzymology , Acetyl-CoA C-Acetyltransferase/chemistry , Acetyl-CoA C-Acetyltransferase/genetics , Bacterial Proteins/genetics , Binding Sites , Catalysis , Catalytic Domain , Crystallization , Crystallography, X-Ray , Cysteine/chemistry , Cysteine/metabolism , Models, Molecular , Mutant Proteins/chemistry , Mutant Proteins/genetics , Mutant Proteins/metabolism , Mutation/genetics , Protein Conformation , Substrate Specificity , Xanthomonas campestris/genetics
17.
Acta Crystallogr F Struct Biol Commun ; 72(Pt 7): 534-44, 2016 07.
Article in English | MEDLINE | ID: mdl-27380370

ABSTRACT

Thiolases catalyze the Claisen condensation of two acetyl-CoA molecules to give acetoacetyl-CoA, as well as the reverse degradative reaction. Four genes coding for thiolases or thiolase-like proteins are found in the Escherichia coli genome. In this communication, the successful cloning, purification, crystallization and structure determination at 1.8 Šresolution of a homotetrameric E. coli thiolase are reported. The structure of E. coli thiolase co-crystallized with acetyl-CoA at 1.9 Šresolution is also reported. As observed in other tetrameric thiolases, the present E. coli thiolase is a dimer of two tight dimers and probably functions as a biodegradative enzyme. Comparison of the structure and biochemical properties of the E. coli enzyme with those of other well studied thiolases reveals certain novel features of this enzyme, such as the modification of a lysine in the dimeric interface, the possible oxidation of the catalytic Cys88 in the structure of the enzyme obtained in the presence of CoA and active-site hydration. The tetrameric enzyme also displays an interesting departure from exact 222 symmetry, which is probably related to the deformation of the tetramerization domain that stabilizes the oligomeric structure of the protein. The current study allows the identification of substrate-binding amino-acid residues and water networks at the active site and provides the structural framework required for understanding the biochemical properties as well as the physiological function of this E. coli thiolase.


Subject(s)
Acetyl Coenzyme A/chemistry , Acetyl-CoA C-Acetyltransferase/chemistry , Escherichia coli Proteins/chemistry , Escherichia coli/enzymology , Water/chemistry , Acetyl Coenzyme A/metabolism , Acetyl-CoA C-Acetyltransferase/genetics , Acetyl-CoA C-Acetyltransferase/metabolism , Amino Acid Motifs , Catalytic Domain , Cloning, Molecular , Crystallography, X-Ray , Cysteine/chemistry , Cysteine/metabolism , Escherichia coli/genetics , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Gene Expression , Kinetics , Lysine/chemistry , Lysine/metabolism , Models, Molecular , Plasmids/chemistry , Plasmids/metabolism , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Protein Multimerization , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Substrate Specificity , Water/metabolism
18.
Poult Sci ; 95(6): 1406-10, 2016 Jun 01.
Article in English | MEDLINE | ID: mdl-26944984

ABSTRACT

Acetoacetyl-CoA thiolase (EC 2.3.1.9) catalyzes the cleavage of acetoacetyl-CoA into acetyl-CoA and its reverse reaction, the synthesis of acetoacetyl-CoA. Cytosolic acetoacetyl-CoA thiolase ( CT: ) is a key enzyme in the initial step of the cholesterol synthesis pathway. In the present study, we characterized the amino acid sequence of chicken CT and the tissue distribution of its mRNA and protein, together with their developmental changes in the liver. The amino acid sequence encoded by the nucleotide sequence of chicken CT cDNA showed a higher overall identity with those of human (74.3%) and rat (74.6%) CTs. Amino acid residues known to participate in enzymatic activity in human CT are conserved in chicken CT. Real-time PCR analysis revealed the expression of CT mRNA in the liver, kidney, adrenal gland, jejunum and ovary of adult hens, with higher levels in the liver, kidney, adrenal gland and ovary. Western blot analysis detected an immunoreactive protein of 41 kDa from cytoplasmic fraction but not particulate fractions of adult chicken liver. The immunoreactive protein was detected in all the tissues. The mRNA levels in the liver rapidly increased after hatching, with a maximum on d 5 post-hatching, after which they gradually decreased to adult levels. A similar change was observed in the protein levels. The increase in transcription and protein synthesis of CT suggests that the synthetic pathway of cholesterol from acetyl-CoA produced by CT replaces the hydrolysis of accumulated cholesteryl ester in the liver, in response to a change in the nutrient source from the lipid-rich yolk to a lower-lipid diet during the early post-hatching period.


Subject(s)
Acetyl-CoA C-Acetyltransferase/genetics , Avian Proteins/genetics , Chickens/genetics , Acetyl-CoA C-Acetyltransferase/chemistry , Acetyl-CoA C-Acetyltransferase/metabolism , Amino Acid Sequence , Animals , Avian Proteins/chemistry , Avian Proteins/metabolism , Base Sequence , Chickens/growth & development , Chickens/metabolism , Cytosol/metabolism , Liver/enzymology , Liver/metabolism , Organ Specificity , RNA, Messenger/genetics , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction/veterinary , Sequence Alignment/veterinary
19.
Zhongguo Zhong Yao Za Zhi ; 40(5): 847-52, 2015 Mar.
Article in Chinese | MEDLINE | ID: mdl-26087544

ABSTRACT

In this study, based on the transcriptome data, we cloned the full-length cDNAs of TwAACT gene from Tripterygium wilfordii suspension cells, and then analyzed the bioinformation of the sequence, detected the genetic differential expression after being induced by methyl jasmonate (MeJA) by RT-PCR. The full-length cDNA of the TwAACT was 1 704 bp containing a 1 218 bp open reading frame (ORF) encoding a polypeptide of 405 amino acids (GeneBank accession No. KP297934). The deduced isoelectric point (pI) was 6.10, a calculated molecular weight was about 41.20 kDa, and online prediction showed that TwAACT had two catalytic active sites. After the induction of MeJA, the relative expression level of TwAACT increased rapidly. The expression level of TwAACT was highest at 24 h. TwAACT was cloned firstly, that laid the foundation for identifying thegene and illustrating thebiosynthesis mechanism and its synthetic biology.


Subject(s)
Acetyl-CoA C-Acetyltransferase/genetics , Cloning, Molecular , Plant Proteins/genetics , Tripterygium/enzymology , Acetyl-CoA C-Acetyltransferase/chemistry , Acetyl-CoA C-Acetyltransferase/metabolism , Amino Acid Sequence , Gene Expression Regulation, Plant , Models, Molecular , Molecular Sequence Data , Phylogeny , Plant Proteins/chemistry , Plant Proteins/metabolism , Sequence Alignment , Tripterygium/chemistry , Tripterygium/classification , Tripterygium/genetics
20.
Chembiochem ; 16(7): 1047-51, 2015 May 04.
Article in English | MEDLINE | ID: mdl-25801592

ABSTRACT

Buff-tailed bumblebees, Bombus terrestris, use a male sex pheromone for premating communication. Its main component is a sesquiterpene, 2,3-dihydrofarnesol. This paper reports the isolation of a thiolase (acetyl-CoA thiolase, AACT_BT), the first enzyme involved in the biosynthetic pathway leading to formation of isoprenoids in the B. terrestris male sex pheromone. Characterisation of AACT_BT might contribute to a better understanding of pheromonogenesis in the labial gland of B. terrestris males. The protein was purified to apparent homogeneity by column chromatography with subsequent stepwise treatment. AACT_BT showed optimum acetyltransferase activity at pH 7.1 and was strongly inhibited by iodoacetamide. The enzyme migrated as a band with an apparent mass of 42.9 kDa on SDS-PAGE. MS analysis of an AACT_BT tryptic digest revealed high homology to representatives of the thiolase family. AACT_BT has 96 % amino acid sequence identity with the previously reported Bombus impatiens thiolase.


Subject(s)
Acetyl-CoA C-Acetyltransferase/metabolism , Bees/metabolism , Salivary Glands, Minor/enzymology , Sex Attractants/biosynthesis , Terpenes/metabolism , Acetyl-CoA C-Acetyltransferase/antagonists & inhibitors , Acetyl-CoA C-Acetyltransferase/chemistry , Amino Acid Sequence , Animals , Enzyme Inhibitors/pharmacology , Hydrogen-Ion Concentration , Male , Molecular Sequence Data , Molecular Weight , Sequence Analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...