Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 20
1.
Protein Pept Lett ; 29(3): 199-207, 2022.
Article En | MEDLINE | ID: mdl-35049426

BACKGROUND: Pseudomonas citronellolis SJTE-3 can efficiently degrade 17ß-estradiol (E2) and other estrogenic chemicals. However, the enzyme responsible for E2 metabolism within strain SJTE-3 has remained unidentified. OBJECTIVE: Here, a novel 3-oxoacyl-(acyl-carrier protein) (ACP) reductase, HSD-X1 (WP_ 009617962.1), was identified in SJTE-3 and its enzymatic characteristics for the transformation of E2 were investigated. METHODS: Multiple sequence alignment and homology modelling were used to predict the protein structure of HSD-X1. The concentrations of different steroids in the culture of recombinant strains expressing HSD-X1 were determined by high performance liquid chromatography. Additionally, the transcription of hsd-x1 gene was investigated using reverse transcription and quantitative PCR analysis. Heterologous expression and affinity purification were used to obtain recombinant HSD- X1. RESULTS: The transcription of hsd-x1 gene in P. citronellolis SJTE-3 was induced by E2. Multiple sequence alignment (MSA) indicated that HSD-X1 contained the two consensus regions and conserved residues of short-chain dehydrogenase/reductases (SDRs) and 17ß-hydroxysteroid dehydrogenases (17ß-HSDs). Over-expression of hsd-x1 gene allowed the recombinant strain to degrade E2. Recombinant HSD-X1 was purified with a yield of 22.15 mg/L and used NAD+ as its cofactor to catalyze the oxidization of E2 into estrone (E1) while exhibiting a Km value of 0.025 ± 0.044 mM and a Vmax value of 4.92 ± 0.31 mM/min/mg. HSD-X1 could tolerate a wide range of temperature and pH, while the presence of divalent ions exerted little influence on its activity. Further, the transformation efficiency of E2 into E1 was over 98.03% across 15 min. CONCLUSION: Protein HSD-X1 efficiently catalyzed the oxidization of E2 and participated in estrogen degradation by P. citronellolis SJTE-3.


Acyl Carrier Protein , Estrone , 3-Oxoacyl-(Acyl-Carrier-Protein) Reductase/metabolism , Estradiol/metabolism , Estrone/metabolism , Oxidoreductases/genetics , Oxidoreductases/metabolism , Pseudomonas
2.
Mol Biol Rep ; 49(2): 1643-1647, 2022 Feb.
Article En | MEDLINE | ID: mdl-35028856

BACKGROUND: Fatty acid elongases (FAEs), which catalyse elongation reactions of a carbon chain of very-long-chain fatty acids, play an important role in shoot development in rice. The elongation reactions consist of four sequential reactions catalysed by distinct enzymes, which are assumed to form an elongation complex. However, no interacting proteins of ONION1 (ONI1) and ONI2, which are ketoacyl CoA synthase catalyzing the first step and are required for shoot development in rice, are reported. METHODS AND RESULTS: In this study ketoacyl CoA reductase (KCR) that interacts with ONI1 and ONI2 was searched. A database search identified 10 KCR genes in the rice genome. Among the genes, the expression pattern of KCR1 was similar to that of ONI2. Yeast two-hybrid analysis showed interaction of ONI2 with KCR1, which was confirmed by GST pull-down assay. No interacting partner of ONI1 was identified. CONCLUSIONS: Our results suggest that ONI2 and KCR1 form an FAE complex that may play a role in biosynthesizing VLCFAs during shoot development.


3-Oxoacyl-(Acyl-Carrier-Protein) Reductase/metabolism , Fatty Acid Elongases/metabolism , Oryza/metabolism , 3-Oxoacyl-(Acyl-Carrier-Protein) Reductase/physiology , Acetyltransferases/genetics , Amino Acid Sequence/genetics , Cloning, Molecular/methods , Coenzyme A/genetics , Coenzyme A/metabolism , Fatty Acid Elongases/genetics , Fatty Acids/metabolism , Gene Expression/genetics , Gene Expression Regulation, Plant/genetics , Oryza/genetics , Oxidoreductases/genetics , Plant Proteins/genetics
3.
J Biomol Struct Dyn ; 40(5): 1952-1969, 2022 03.
Article En | MEDLINE | ID: mdl-33063633

Toxoplasma gondii is an opportunistic obligate parasite, ubiquitous around the globe with seropositivity rates that range from 10% to 90% and infection by the parasite of pregnant women causes pre-natal death of the foetus in most cases and severe neurodegenerative syndromes in some. No vaccine is currently available, and since drug-resistance is common among T. gondii strains, discovering lead compounds for drug design using diverse tactics is necessary. In this study, the sole constituent isoform of an enzymatic 3-oxoacyl-[acyl-carrier-protein] reduction step in an apicoplast-located fatty acid biosynthesis pathway was chosen as a possible drug target. FASII is prokaryotic therefore, targeting it would pose fewer side-effects to human hosts. After a homology 3D modelling of TgFabG, a high-throughput virtual screening of 9867 compounds, the elimination of ligands was carried out by a flexible ligand molecular docking and 200 ns molecular dynamics simulations, with additional DCCM and PC plot analyses. Molecular Dynamics and related post-MD analyses of the top 3 TgFabG binders selected for optimal free binding energies, showed that L2 maintained strong H-bonds with TgFabG and facilitated structural reorientation expected of FabGs, namely an expansion of the Rossmann Fold and a flexible lid capping. The most flexible TgFabG sites were the α7 helix (the flexible lid region) and the ß4-α4 and ß5-α6 loops. For TgFabG-L2, the movements of these regions toward the active site enabled greater ligand stability. Thus, L2 ("Skimmine"; PubChem ID: 320361), was ultimately selected as the optimal candidate for the discovery of lead compounds for rational drug design.Communicated by Ramaswamy H. Sarma.


3-Oxoacyl-(Acyl-Carrier-Protein) Reductase , Protozoan Proteins , Toxoplasma , 3-Oxoacyl-(Acyl-Carrier-Protein) Reductase/genetics , 3-Oxoacyl-(Acyl-Carrier-Protein) Reductase/metabolism , Female , Humans , Molecular Docking Simulation , Molecular Dynamics Simulation , Pregnancy , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Toxoplasma/enzymology , Toxoplasma/genetics
4.
Sci Rep ; 11(1): 7945, 2021 04 12.
Article En | MEDLINE | ID: mdl-33846444

Some Gram-negative bacteria harbor lipids with aryl polyene (APE) moieties. Biosynthesis gene clusters (BGCs) for APE biosynthesis exhibit striking similarities with fatty acid synthase (FAS) genes. Despite their broad distribution among pathogenic and symbiotic bacteria, the detailed roles of the metabolic products of APE gene clusters are unclear. Here, we determined the crystal structures of the ß-ketoacyl-acyl carrier protein (ACP) reductase ApeQ produced by an APE gene cluster from clinically isolated virulent Acinetobacter baumannii in two states (bound and unbound to NADPH). An in vitro visible absorption spectrum assay of the APE polyene moiety revealed that the ß-ketoacyl-ACP reductase FabG from the A. baumannii FAS gene cluster cannot be substituted for ApeQ in APE biosynthesis. Comparison with the FabG structure exhibited distinct surface electrostatic potential profiles for ApeQ, suggesting a positively charged arginine patch as the cognate ACP-binding site. Binding modeling for the aryl group predicted that Leu185 (Phe183 in FabG) in ApeQ is responsible for 4-benzoyl moiety recognition. Isothermal titration and arginine patch mutagenesis experiments corroborated these results. These structure-function insights of a unique reductase in the APE BGC in comparison with FAS provide new directions for elucidating host-pathogen interaction mechanisms and novel antibiotics discovery.


3-Oxoacyl-(Acyl-Carrier-Protein) Reductase/chemistry , 3-Oxoacyl-(Acyl-Carrier-Protein) Reductase/metabolism , Acinetobacter baumannii/enzymology , Fatty Acids/metabolism , Polyenes/metabolism , Amino Acid Sequence , Arginine/metabolism , Biosynthetic Pathways , Crystallography, X-Ray , Leucine/metabolism , Models, Molecular , NADP/metabolism , Protein Conformation , Static Electricity , Structural Homology, Protein , Substrate Specificity
5.
J Biol Chem ; 296: 100365, 2021.
Article En | MEDLINE | ID: mdl-33545175

The FabG 3-ketoacyl-acyl carrier protein (ACP) reductase of Escherichia coli has long been thought to be a classical member of the short-chain alcohol dehydrogenase/reductase (SDR) family. FabG catalyzes the essential 3-ketoacyl-ACP reduction step in the FAS II fatty acid synthesis pathway. Site-directed mutagenesis studies of several other SDR enzymes has identified three highly conserved amino acid residues, Ser, Tyr, and Lys, as the catalytic triad. Structural analyses of E. coli FabG suggested the triad S138-Y151-K155 to form a catalytically competent active site. To test this hypothesis, we constructed a series of E. coli FabG mutants and tested their 3-ketoacyl-ACP reductase activities both in vivo and in vitro. Our data show that plasmid-borne FabG mutants, including the double and triple mutants, restored growth of E. coli and Salmonella enterica fabG temperature-sensitive mutant strains under nonpermissive conditions. In vitro assays demonstrated that all of the purified FabG mutant proteins maintained fatty acid synthetic ability, although the activities of the single mutant proteins were 20% to 50% lower than that of wildtype FabG. The S138A, Y151F, and K155A residue substitutions were confirmed by tandem mass spectral sequencing of peptides that spanned all three residues. We conclude that FabG is not a classical short-chain alcohol dehydrogenase/reductase, suggesting that an alternative mode of 3-ketoacyl-ACP reduction awaits discovery.


3-Oxoacyl-(Acyl-Carrier-Protein) Reductase/metabolism , Alcohol Oxidoreductases/metabolism , 3-Oxoacyl-(Acyl-Carrier-Protein) Reductase/physiology , Alcohol Oxidoreductases/physiology , Amino Acid Sequence/genetics , Binding Sites/physiology , Catalytic Domain/physiology , Crystallography, X-Ray/methods , Escherichia coli/metabolism , Escherichia coli Proteins/metabolism , Fatty Acids/metabolism , Genetic Complementation Test/methods , Models, Molecular , Oxidoreductases/metabolism , Protein Binding/genetics
6.
Plant Physiol ; 183(2): 517-529, 2020 06.
Article En | MEDLINE | ID: mdl-32245791

Plant fatty acid biosynthesis occurs in both plastids and mitochondria. Here, we report the identification and characterization of Arabidopsis (Arabidopsis thaliana) genes encoding three enzymes shared between the mitochondria- and plastid-localized type II fatty acid synthase systems (mtFAS and ptFAS, respectively). Two of these enzymes, ß-ketoacyl-acyl carrier protein (ACP) reductase and enoyl-ACP reductase, catalyze two of the reactions that constitute the core four-reaction cycle of the FAS system, which iteratively elongates the acyl chain by two carbon atoms per cycle. The third enzyme, malonyl-coenzyme A:ACP transacylase, catalyzes the reaction that loads the mtFAS system with substrate by malonylating the phosphopantetheinyl cofactor of ACP. GFP fusion experiments revealed that the these enzymes localize to both chloroplasts and mitochondria. This localization was validated by characterization of mutant alleles, which were rescued by transgenes expressing enzyme variants that were retargeted only to plastids or only to mitochondria. The singular retargeting of these proteins to plastids rescued the embryo lethality associated with disruption of the essential ptFAS system, but these rescued plants displayed phenotypes typical of the lack of mtFAS function, including reduced lipoylation of the H subunit of the glycine decarboxylase complex, hyperaccumulation of glycine, and reduced growth. However, these latter traits were reversible in an elevated-CO2 atmosphere, which suppresses mtFAS-associated photorespiration-dependent chemotypes. Sharing enzymatic components between mtFAS and ptFAS systems constrains the evolution of these nonredundant fatty acid biosynthetic machineries.


Arabidopsis/metabolism , Fatty Acid Synthases/metabolism , Mitochondria/metabolism , 3-Oxoacyl-(Acyl-Carrier-Protein) Reductase/genetics , 3-Oxoacyl-(Acyl-Carrier-Protein) Reductase/metabolism , Arabidopsis/enzymology , Arabidopsis Proteins/genetics , Arabidopsis Proteins/metabolism , Enoyl-(Acyl-Carrier-Protein) Reductase (NADH)/genetics , Enoyl-(Acyl-Carrier-Protein) Reductase (NADH)/metabolism , Glycine/metabolism , Multienzyme Complexes/genetics , Multienzyme Complexes/metabolism , Plastids/metabolism
7.
Mol Plant Pathol ; 20(12): 1696-1709, 2019 12.
Article En | MEDLINE | ID: mdl-31560825

Xanthomonas campestris pv. campestris (Xcc), the causal agent of black rot in crucifers, produces a membrane-bound yellow pigment called xanthomonadin to protect against photobiological and peroxidative damage, and uses a quorum-sensing mechanism mediated by the diffusible signal factor (DSF) family signals to regulate virulence factors production. The Xcc gene XCC4003, annotated as Xcc fabG3, is located in the pig cluster, which may be responsible for xanthomonadin synthesis. We report that fabG3 expression restored the growth of the Escherichia coli fabG temperature-sensitive mutant CL104 under non-permissive conditions. In vitro assays demonstrated that FabG3 catalyses the reduction of 3-oxoacyl-acyl carrier protein (ACP) intermediates in fatty acid synthetic reactions, although FabG3 had a lower activity than FabG1. Moreover, the fabG3 deletion did not affect growth or fatty acid composition. These results indicate that Xcc fabG3 encodes a 3-oxoacyl-ACP reductase, but is not essential for growth or fatty acid synthesis. However, the Xcc fabG3 knock-out mutant abolished xanthomonadin production, which could be only restored by wild-type fabG3, but not by other 3-oxoacyl-ACP reductase-encoding genes, indicating that Xcc FabG3 is specifically involved in xanthomonadin biosynthesis. Additionally, our study also shows that the Xcc fabG3-disrupted mutant affects Xcc virulence in host plants.


3-Oxoacyl-(Acyl-Carrier-Protein) Reductase/metabolism , Pigments, Biological/biosynthesis , Xanthomonas campestris/metabolism , 3-Oxoacyl-(Acyl-Carrier-Protein) Reductase/genetics , Fatty Acids/biosynthesis , Gene Knockout Techniques , Genetic Complementation Test , Pigments, Biological/genetics , Quorum Sensing , Virulence/genetics , Xanthomonas campestris/enzymology , Xanthomonas campestris/genetics , Xanthomonas campestris/pathogenicity
8.
J Microbiol Biotechnol ; 29(11): 1769-1776, 2019 Nov 28.
Article En | MEDLINE | ID: mdl-30111070

Ethyl (S)-3-hydroxy-3-(2-thienyl)propanoate((S)-HEES)acts as a key chiral intermediate for the blockbuster antidepressant drug duloxetine, which canbe achieved viathe stereoselective bioreduction ofethyl 3-oxo-3-(2-thienyl) propanoate (KEES) that containsa 3-oxoacyl structure.The sequences of the short-chain dehydrogenase/reductases from Chryseobacterium sp. CA49 were analyzed, and the putative3-oxoacyl-acyl-carrier-protein reductase, ChKRED12, was able to stereoselectivelycatalyze theNADPH-dependent reduction to produce (S)-HEES.The reductase activity of ChKRED12 towardsothersubstrates with 3-oxoacyl structure were confirmed with excellent stereoselectivity (>99% enantiomeric excess) in most cases. When coupled with a cofactor recycling system using glucose dehydrogenase, the ChKRED12 was able to catalyze the complete conversion of 100 g/l KEES within 12h, yielding the enantiopure product with >99% ee, showing a remarkable potential to produce (S)-HEES.


3-Oxoacyl-(Acyl-Carrier-Protein) Reductase/metabolism , Bacterial Proteins/metabolism , Propionates/metabolism , Short Chain Dehydrogenase-Reductases/metabolism , 3-Oxoacyl-(Acyl-Carrier-Protein) Reductase/chemistry , 3-Oxoacyl-(Acyl-Carrier-Protein) Reductase/genetics , Amino Acid Sequence , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Catalysis , Chryseobacterium/enzymology , Chryseobacterium/genetics , Escherichia coli/genetics , Escherichia coli/metabolism , Glucose 1-Dehydrogenase/metabolism , Kinetics , Oxidation-Reduction , Propionates/chemistry , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Short Chain Dehydrogenase-Reductases/chemistry , Short Chain Dehydrogenase-Reductases/genetics , Stereoisomerism , Substrate Specificity
9.
Acta Crystallogr D Struct Biol ; 74(Pt 5): 383-393, 2018 05 01.
Article En | MEDLINE | ID: mdl-29717709

In mycobacteria, the ketoacyl-acyl carrier protein (ACP) reductase MabA (designated FabG in other bacteria) catalyzes the NADPH-dependent reduction of ß-ketoacyl-ACP substrates to ß-hydroxyacyl-ACP products. This first reductive step in the fatty-acid biosynthesis elongation cycle is essential for bacteria, which makes MabA/FabG an interesting drug target. To date, however, very few molecules targeting FabG have been discovered and MabA remains the only enzyme of the mycobacterial type II fatty-acid synthase that lacks specific inhibitors. Despite the existence of several MabA/FabG crystal structures, the structural rearrangement that occurs upon cofactor binding is still not fully understood. Therefore, unlocking this knowledge gap could help in the design of new inhibitors. Here, high-resolution crystal structures of MabA from Mycobacterium smegmatis in its apo, NADP+-bound and NADPH-bound forms are reported. Comparison of these crystal structures reveals the structural reorganization of the lid region covering the active site of the enzyme. The crystal structure of the apo form revealed numerous residues that trigger steric hindrance to the binding of NADPH and substrate. Upon NADPH binding, these residues are pushed away from the active site, allowing the enzyme to adopt an open conformation. The transition from an NADPH-bound to an NADP+-bound form is likely to facilitate release of the product. These results may be useful for subsequent rational drug design and/or for in silico drug-screening approaches targeting MabA/FabG.


3-Oxoacyl-(Acyl-Carrier-Protein) Reductase/chemistry , Mycobacterium smegmatis/chemistry , NADP/chemistry , 3-Oxoacyl-(Acyl-Carrier-Protein) Reductase/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Catalytic Domain , Crystallization , Crystallography, X-Ray , Fatty Acid Synthase, Type II , Mycobacterium smegmatis/enzymology , NADP/metabolism , Protein Binding , Protein Conformation
10.
Phys Chem Chem Phys ; 19(1): 347-355, 2016 Dec 21.
Article En | MEDLINE | ID: mdl-27905606

Human fatty acid synthase (hFAS) is a multifunctional enzyme involved in a wide diversity of biological functions. For instance, it is a precursor of phospholipids and other complex processes such as the de novo synthesis of long chain fatty acid. Human FAS is also a component of biological membranes and it is implicated in the overexpression of several types of cancers. In this work, we describe the catalytic mechanism of ß-ketoreductase (KR), which is a catalytic domain of the hFAS enzyme that catalyzes the reduction of ß-ketoacyl to ß-hydroxyacyl with the concomitant oxidation of the NADPH cofactor. The catalysis by KR is an intermediate step in the cycle of reactions that elongate the substrate's carbon chain until the final product is obtained. We study and propose the catalytic mechanism of the KR domain determined using the hybrid QM/MM methodology, at the ONIOM(B3LYP/6-311+G(2d,2p):AMBER) level of theory. The results indicate that the reaction mechanism occurs in two stages: (i) nucleophilic attack by a NADPH hydride to the ß-carbon of the substrate, together with an asynchronous deprotonation of the Tyr2034 by the oxygen of the ß-alkoxide to hold the final alcohol product; and (ii) an asynchronous deprotonation of the hydroxyl in the NADP+'s ribose by Tyr2034, and of the Lys1995 by the resulting alkoxide in the former ribose to restore the protonation state of Tyr2034. The reduction step occurs with a Gibbs energy barrier of 11.7 kcal mol-1 and a Gibbs reaction energy of -10.6 kcal mol-1. These results have provided an understanding of the catalytic mechanism of the KR hFAS domain, a piece of the heavy hFAS biosynthetic machinery.


3-Oxoacyl-(Acyl-Carrier-Protein) Reductase/chemistry , 3-Oxoacyl-(Acyl-Carrier-Protein) Reductase/metabolism , Alcohols/chemistry , NADP/chemistry , Catalysis , Catalytic Domain , Humans , Oxidation-Reduction , Quantum Theory
11.
Mol Plant Microbe Interact ; 29(6): 458-67, 2016 Jun.
Article En | MEDLINE | ID: mdl-26975437

In Sinorhizobium meliloti, the nodG gene is located in the nodFEG operon of the symbiotic plasmid. Although strong sequence similarity (53% amino acid identities) between S. meliloti NodG and Escherichia coli FabG was reported in 1992, it has not been determined whether S. meliloti NodG plays a role in fatty acid synthesis. We report that expression of S. meliloti NodG restores the growth of the E. coli fabG temperature-sensitive mutant CL104 under nonpermissive conditions. Using in vitro assays, we demonstrated that NodG is able to catalyze the reduction of the 3-oxoacyl-ACP intermediates in E. coli fatty acid synthetic reaction. Moreover, although deletion of the S. meliloti nodG gene does not cause any growth defects, upon overexpression of nodG from a plasmid, the S. meliloti fabG gene encoding the canonical 3-oxoacyl-ACP reductase (OAR) can be disrupted without any effects on growth or fatty acid composition. This indicates that S. meliloti nodG encodes an OAR and can play a role in fatty acid synthesis when expressed at sufficiently high levels. Thus, a bacterium can simultaneously possess two or more OARs that can play a role in fatty acid synthesis. Our data also showed that, although SmnodG increases alfalfa nodulation efficiency, it is not essential for alfalfa nodulation.


3-Oxoacyl-(Acyl-Carrier-Protein) Reductase/metabolism , Alcohol Oxidoreductases/metabolism , Bacterial Proteins/metabolism , Escherichia coli/metabolism , Fatty Acids/biosynthesis , Sinorhizobium meliloti/metabolism , 3-Oxoacyl-(Acyl-Carrier-Protein) Reductase/genetics , Alcohol Oxidoreductases/genetics , Bacterial Proteins/genetics , Escherichia coli/genetics , Escherichia coli/growth & development , Fatty Acids/genetics , Gene Expression Regulation, Bacterial , Medicago sativa/microbiology , Mutation , Root Nodules, Plant/microbiology , Sinorhizobium meliloti/genetics , Sinorhizobium meliloti/growth & development , Temperature
12.
J Bacteriol ; 198(3): 463-76, 2016 02 01.
Article En | MEDLINE | ID: mdl-26553852

UNLABELLED: ß-Ketoacyl-(acyl carrier protein) reductase (FabG) catalyzes the key reductive reaction in the elongation cycle of fatty acid synthesis (FAS), which is a vital metabolic pathway in bacteria and a promising target for new antibiotic development. The activation of the enzyme is usually linked to the formation of a catalytic triad and cofactor binding, and crystal structures of FabG from different organisms have been captured in either the active or inactive conformation. However, the structural elements which enable activation of FabG require further exploration. Here we report the findings of structural, enzymatic, and binding studies of the FabG protein found in the causative agent of cholera, Vibrio cholerae (vcFabG). vcFabG exists predominantly as a dimer in solution and is able to self-associate to form tetramers, which is the state seen in the crystal structure. The formation of the tetramer may be promoted by the presence of the cofactor NADP(H). The transition between the dimeric and tetrameric states of vcFabG is related to changes in the conformations of the α4/α5 helices on the dimer-dimer interface. Two glycine residues adjacent to the dimer interface (G92 and G141) are identified to be the hinge for the conformational changes, while the catalytic tyrosine (Y155) and a glutamine residue that forms hydrogen bonds to both loop ß4-α4 and loop ß5-α5 (Q152) stabilize the active conformation. The functions of the aforementioned residues were confirmed by binding and enzymatic assays for the corresponding mutants. IMPORTANCE: This paper describes the results of structural, enzymatic, and binding studies of FabG from Vibrio cholerae (vcFabG). In this work, we dissected the structural elements responsible for the activation of vcFabG. The structural information provided here is essential for the development of antibiotics specifically targeting bacterial FabG, especially for the multidrug-resistant strains of V. cholerae.


3-Oxoacyl-(Acyl-Carrier-Protein) Reductase/metabolism , Enzyme Activation/physiology , Gene Expression Regulation, Bacterial/physiology , Gene Expression Regulation, Enzymologic/physiology , Vibrio cholerae/enzymology , 3-Oxoacyl-(Acyl-Carrier-Protein) Reductase/genetics , Cloning, Molecular , Models, Molecular , Mutagenesis , Mutation , NADP/genetics , NADP/metabolism , Protein Binding , Protein Conformation , Tyrosine/chemistry , Vibrio cholerae/genetics , Vibrio cholerae/metabolism
13.
PLoS One ; 10(11): e0141543, 2015.
Article En | MEDLINE | ID: mdl-26539719

Ketoacyl-acyl carrier protein reductases (FabG) are ubiquitously expressed enzymes that catalyse the reduction of acyl carrier protein (ACP) linked thioesters within the bacterial type II fatty acid synthesis (FASII) pathway. The products of these enzymes, saturated and unsaturated fatty acids, are essential components of the bacterial cell envelope. The FASII reductase enoyl-ACP reductase (FabI) has been the focus of numerous drug discovery efforts, some of which have led to clinical trials, yet few studies have focused on FabG. Like FabI, FabG appears to be essential for survival in many bacteria, similarly indicating the potential of this enzyme as a drug target. FabG enzymes are members of the short-chain alcohol dehydrogenase/reductase (SDR) family, and like other SDRs, exhibit highly conserved secondary and tertiary structures, and contain a number of conserved sequence motifs. Here we describe the crystal structures of FabG from Yersinia pestis (YpFabG), the causative agent of bubonic, pneumonic, and septicaemic plague, and three human pandemics. Y. pestis remains endemic in many parts of North America, South America, Southeast Asia, and Africa, and a threat to human health. YpFabG shares a high degree of structural similarity with bacterial homologues, and the ketoreductase domain of the mammalian fatty acid synthase from both Homo sapiens and Sus scrofa. Structural characterisation of YpFabG, and comparison with other bacterial FabGs and the mammalian fatty acid synthase, provides a strong platform for virtual screening of potential inhibitors, rational drug design, and the development of new antimicrobial agents to combat Y. pestis infections.


3-Oxoacyl-(Acyl-Carrier-Protein) Reductase/chemistry , Fatty Acids/biosynthesis , Yersinia pestis/metabolism , 3-Oxoacyl-(Acyl-Carrier-Protein) Reductase/isolation & purification , 3-Oxoacyl-(Acyl-Carrier-Protein) Reductase/metabolism , Cloning, Molecular , Crystallization , Crystallography, X-Ray , Hydrogen Bonding , Molecular Structure , Protein Structure, Quaternary , Protein Structure, Tertiary , Structural Homology, Protein , Yersinia pestis/enzymology
14.
BMC Microbiol ; 15: 223, 2015 Oct 22.
Article En | MEDLINE | ID: mdl-26490537

BACKGROUND: FabG is the only known enzyme that catalyzes reduction of the 3-ketoacyl-ACP intermediates of bacterial fatty acid synthetic pathways. However, there are two Ralstonia solanacearum genes, RSc1052 (fabG1) and RSp0359 (fabG2), annotated as encoding putative 3-ketoacyl-ACP reductases. Both FabG homologues possess the conserved catalytic triad and the N-terminal cofactor binding sequence of the short chain dehydrogenase/reductase (SDR) family. Thus, it seems reasonable to hypothesize that RsfabG1 and RsfabG2 both encode functional 3-ketoacyl-ACP reductases and play important roles in R. solanacearum fatty acid synthesis and growth. METHODS: Complementation of Escherichia coli fabG temperature-sensitive mutant with R. solanacearum fabGs encoded plasmids was carried out to test the function of RsfabGs in fatty acid biosynthesis. RsFabGs proteins were purified by nickel chelate chromatography and fatty acid biosynthetic reaction was reconstituted to investigate the 3-ketoacyl-ACP reductase activity of RsFabGs in vitro. Disruption of both RsfabG genes was done via DNA homologous recombination to test the function of both RsfabG in vivo. And more we also carried out pathogenicity tests on tomato plants using RsfabG mutant strains.  RESULTS: We report that expression of either of the two proteins (RsFabG1 and RsFabG2) restores growth of the E. coli fabG temperature-sensitive mutant CL104 under non-permissive conditions. In vitro assays demonstrate that both proteins restore fatty acid synthetic ability to extracts of the E. coli strain. The RsfabG1 gene carried on the R. solanacearum chromosome is essential for growth of the bacterium, as is the case for fabG in E. coli. In contrast, the null mutant strain with the megaplasmid-encoded RsfabG2 gene is viable but has a fatty acid composition that differs significantly from that of the wild type strain. Our study also shows that RsFabG2 plays a role in adaptation to high salt concentration and low pH, and in pathogenesis of disease in tomato plants. CONCLUSION: R. solanacearum encodes two 3-ketoacyl-ACP reductases that both have functions in fatty acid synthesis. We supply the first evidence that, like other enzymes in the bacterial fatty acid biosynthetic pathway, one bacterium may simultaneously possess two or more 3-oxoacyl-ACP reductase isozymes.


3-Oxoacyl-(Acyl-Carrier-Protein) Reductase/genetics , 3-Oxoacyl-(Acyl-Carrier-Protein) Reductase/metabolism , Fatty Acids/analysis , Ralstonia solanacearum/chemistry , Ralstonia solanacearum/enzymology , Replicon , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Deletion , Genetic Complementation Test , Solanum lycopersicum/microbiology , Plant Diseases/microbiology , Ralstonia solanacearum/genetics , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Virulence
15.
FEBS Lett ; 589(20 Pt B): 3052-7, 2015 Oct 07.
Article En | MEDLINE | ID: mdl-26358291

PhaB (acetoacetyl-CoA reductase) catalyzes the reduction of acetoacetyl-CoA to (R)-3-hydroxybutyryl-CoA in polyhydroxybutyrate (PHB) synthesis and FabG (3-ketoacyl-acyl-carrier-protein reductase) catalyzes the ß-ketoacyl-ACP to yield (R)-3-hydroxyacyl-ACP in fatty acid biosynthesis. Both of them have been classified into the same group EC 1.1.1. PhaB is limited with substrate specificities, while FabG was considered as a potential PhaB due to broad substrate selectivity despite of low activity. Here, X-ray crystal structures of FabG and PhaB from the photosynthetic microorganism Synechocystis sp. PCC 6803 were resolved. Based on them, a high-performance FabG on acyl-CoA directed by structural evolution was constructed that may serve as a critical enzyme to partition carbon flow from fatty acid synthesis to PHA.


3-Oxoacyl-(Acyl-Carrier-Protein) Reductase/metabolism , Alcohol Oxidoreductases/metabolism , Bacterial Proteins/metabolism , Synechocystis/enzymology , 3-Oxoacyl-(Acyl-Carrier-Protein) Reductase/chemistry , 3-Oxoacyl-(Acyl-Carrier-Protein) Reductase/genetics , Acyl Carrier Protein/chemistry , Acyl Carrier Protein/metabolism , Acyl Coenzyme A/chemistry , Acyl Coenzyme A/metabolism , Alcohol Oxidoreductases/chemistry , Alcohol Oxidoreductases/genetics , Amino Acid Sequence , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Crystallography, X-Ray , Models, Molecular , Molecular Sequence Data , Molecular Structure , Mutation , Protein Conformation , Sequence Homology, Amino Acid , Substrate Specificity , Synechocystis/genetics , Synechocystis/metabolism
16.
Chembiochem ; 16(4): 631-40, 2015 Mar 02.
Article En | MEDLINE | ID: mdl-25662938

Streptomyces coelicolor produces fatty acids for both primary metabolism and for biosynthesis of the secondary metabolite undecylprodiginine. The first and last reductive steps during the chain elongation cycle of fatty acid biosynthesis are catalyzed by FabG and FabI. The S. coelicolor genome sequence has one fabI gene (SCO1814) and three likely fabG genes (SCO1815, SCO1345, and SCO1846). We report the expression, purification, and characterization of the corresponding gene products. Kinetic analyses revealed that all three FabGs and FabI are capable of utilizing both straight and branched-chain ß-ketoacyl-NAC and enoyl-NAC substrates, respectively. Furthermore, only SCO1345 differentiates between ACPs from both biosynthetic pathways. The data presented provide the first experimental evidence that SCO1815, SCO1346, and SCO1814 have the catalytic capability to process intermediates in both fatty acid and undecylprodiginine biosynthesis.


3-Oxoacyl-(Acyl-Carrier-Protein) Reductase/metabolism , Enoyl-(Acyl-Carrier-Protein) Reductase (NADH)/metabolism , Fatty Acid Synthases/metabolism , Fatty Acids/metabolism , Streptomyces coelicolor/enzymology , 3-Oxoacyl-(Acyl-Carrier-Protein) Reductase/genetics , Enoyl-(Acyl-Carrier-Protein) Reductase (NADH)/genetics , Genes, Bacterial , Genes, Fungal , Streptomyces coelicolor/genetics , Streptomyces coelicolor/metabolism
17.
Nat Commun ; 5: 4805, 2014 Sep 09.
Article En | MEDLINE | ID: mdl-25203508

Mitochondrial fatty acid synthesis (mtFAS) is essential for respiratory growth in yeast and mammalian embryonic survival. The human 3-ketoacyl-acyl carrier protein (ACP) reductase (KAR) of mtFAS is a heterotetrameric α2ß2-assembly composed of 17ß-hydroxysteroid dehydrogenase type-8 (HSD17B8, α-subunit) and carbonyl reductase type-4 (CBR4, ß-subunit). Here we provide a structural explanation for the stability of the heterotetramer from the crystal structure with NAD(+) and NADP(+) bound to the HSD17B8 and CBR4 subunits, respectively, and show that the catalytic activity of the NADPH- and ACP-dependent CBR4 subunit is crucial for a functional HsKAR. Therefore, mtFAS is NADPH- and ACP dependent, employing the 3R-hydroxyacyl-ACP intermediate. HSD17B8 assists in the formation of the competent HsKAR assembly. The intrinsic NAD(+)- and CoA-dependent activity of the HSD17B8 subunit on the 3R-hydroxyacyl-CoA intermediates may indicate a role for this subunit in routing 3R-hydroxyacyl-CoA esters, potentially arising from the metabolism of unsaturated fatty acids, into the mitochondrial ß-oxidation pathway.


Escherichia coli Proteins/metabolism , Fatty Acids/biosynthesis , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , NAD(P)H Dehydrogenase (Quinone)/metabolism , Oxidoreductases/metabolism , Protein Structure, Quaternary , Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , 3-Hydroxyacyl CoA Dehydrogenases/metabolism , 3-Oxoacyl-(Acyl-Carrier-Protein) Reductase/metabolism , Bacterial Proteins/metabolism , Crystallography, X-Ray , Enzyme Assays , Humans , Mycobacterium tuberculosis/metabolism , Saccharomyces cerevisiae/metabolism
18.
Nat Chem Biol ; 10(9): 774-9, 2014 Sep.
Article En | MEDLINE | ID: mdl-25086508

Human fatty acid synthase (hFAS) is a complex, multifunctional enzyme that is solely responsible for the de novo synthesis of long chain fatty acids. hFAS is highly expressed in a number of cancers, with low expression observed in most normal tissues. Although normal tissues tend to obtain fatty acids from the diet, tumor tissues rely on de novo fatty acid synthesis, making hFAS an attractive metabolic target for the treatment of cancer. We describe here the identification of GSK2194069, a potent and specific inhibitor of the ß-ketoacyl reductase (KR) activity of hFAS; the characterization of its enzymatic and cellular mechanism of action; and its inhibition of human tumor cell growth. We also present the design of a new protein construct suitable for crystallography, which resulted in what is to our knowledge the first co-crystal structure of the human KR domain and includes a bound inhibitor.


3-Oxoacyl-(Acyl-Carrier-Protein) Reductase/metabolism , Enzyme Inhibitors/metabolism , Fatty Acid Synthases/antagonists & inhibitors , Pyrrolidines/metabolism , Pyrrolidines/pharmacology , Triazoles/metabolism , Triazoles/pharmacology , 3-Oxoacyl-(Acyl-Carrier-Protein) Reductase/chemistry , Catalytic Domain , Cell Line, Tumor , Fatty Acid Synthases/chemistry , Humans , Models, Molecular , Protein Conformation , X-Ray Diffraction
19.
Org Biomol Chem ; 12(1): 73-85, 2014 Jan 07.
Article En | MEDLINE | ID: mdl-24129589

We report the design and synthesis of triazole-polyphenol hybrid compounds 1 and 2 as inhibitors of the FabG4 (Rv0242c) enzyme of Mycobacterium tuberculosis for the first time. A major advance in this field occurred only a couple of years ago with the X-ray crystal structure of FabG4, which has helped us to design these inhibitors by the computational fragment-based drug design (FBDD) approach. Compound 1 has shown competitive inhibition with an inhibition constant (Ki) value of 3.97 ± 0.02 µM. On the other hand, compound 2 has been found to be a mixed type inhibitor with a Ki value of 0.88 ± 0.01 µM. Thermodynamic analysis using isothermal titration calorimetry (ITC) reveals that both inhibitors bind at the NADH co-factor binding domain. Their MIC values, as determined by resazurin assay against M. smegmatis, indicated their good anti-mycobacterial properties. A preliminary structure-activity relationship (SAR) study supports the design of these inhibitors. These compounds may be possible candidates as lead compounds for alternate anti-tubercular drugs. All of the reductase enzymes of the Mycobacterium family have a similar ketoacyl reductase (KAR) domain. Hence, this work may be extrapolated to find structure-based inhibitors of other reductase enzymes.


3-Oxoacyl-(Acyl-Carrier-Protein) Reductase/antagonists & inhibitors , Antitubercular Agents/pharmacology , Drug Design , Enzyme Inhibitors/pharmacology , Mycobacterium tuberculosis/drug effects , Polyphenols/chemistry , Triazoles/chemistry , 3-Oxoacyl-(Acyl-Carrier-Protein) Reductase/metabolism , Antitubercular Agents/chemical synthesis , Antitubercular Agents/chemistry , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Microbial Sensitivity Tests , Models, Molecular , Molecular Structure , Mycobacterium tuberculosis/enzymology , Structure-Activity Relationship
20.
Genes Cells ; 18(9): 733-52, 2013 Sep.
Article En | MEDLINE | ID: mdl-23786411

Three types of mitosis, which are open, closed or semi-open mitosis, function in eukaryotic cells, respectively. The open mitosis involves breakage of the nuclear envelope before nuclear division, whereas the closed mitosis proceeds with an intact nuclear envelope. To understand the mechanism and significance of three types of mitotic division in eukaryotes, we investigated the process of semi-open mitosis, in which the nuclear envelope is only partially broken, in the fission yeast Schizosaccharomyces japonicus. In anaphase-promoting complex/cyclosome (APC/C) mutants of Sz. japonicus, the nuclear envelope remained relatively intact during anaphase, resulting in impaired semi-open mitosis. As a suppressor of apc2 mutant, a mutation of Oar2, which was a 3-oxoacyl-[acyl carrier protein] reductase, was obtained. The level of the Oar2, which had two destruction-box motifs recognized by APC/C, was increased in APC/C mutants. Furthermore, the defective semi-open mitosis observed in an apc2 mutant was restored by mutated oar2+. Based on these findings, we propose that APC/C regulates the dynamics of the nuclear envelope through degradation of Oar2 dependent on APC/C during the metaphase-to-anaphase transition of semi-open mitosis in Sz. japonicus.


Apc2 Subunit, Anaphase-Promoting Complex-Cyclosome/metabolism , Fungal Proteins/metabolism , Mitosis , Nuclear Envelope/metabolism , Schizosaccharomyces/metabolism , 3-Oxoacyl-(Acyl-Carrier-Protein) Reductase/chemistry , 3-Oxoacyl-(Acyl-Carrier-Protein) Reductase/genetics , 3-Oxoacyl-(Acyl-Carrier-Protein) Reductase/metabolism , Amino Acid Motifs , Amino Acid Sequence , Anaphase , Apc2 Subunit, Anaphase-Promoting Complex-Cyclosome/genetics , Fungal Proteins/genetics , Metaphase , Molecular Sequence Data , Mutation , Proteolysis , Schizosaccharomyces/cytology , Schizosaccharomyces/genetics
...