Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
2.
Appl Microbiol Biotechnol ; 58(6): 695-703, 2002 May.
Article in English | MEDLINE | ID: mdl-12021787

ABSTRACT

Fatty acid biosynthesis is an emerging target for the development of novel antibacterial chemotherapeutics. The dissociated bacterial system is substantially different from the large, multifunctional protein of mammals, and many possibilities exist for type-selective drugs. Several compounds, both synthetic and natural, target bacterial fatty acid synthesis. Three compounds target the FabI enoyl-ACP reductase step; isoniazid, a clinically used antituberculosis drug, triclosan, a widely used consumer antimicrobial, and diazaborines. In addition, cerulenin and thiolactomycin, two fungal products, inhibit the FabH, FabB and FabF condensation enzymes. Finally, the synthetic reaction intermediates BP1 and decynoyl- N-acetyl cysteamine inhibit the acetyl-CoA carboxylase and dehydratase isomerase steps, respectively. The mechanisms of action of these compounds, as well as the potential development of new drugs targeted against this pathway, are discussed.


Subject(s)
Anti-Infective Agents/pharmacology , Fatty Acids/antagonists & inhibitors , Enzymes/metabolism , Fatty Acids/biosynthesis
3.
Prog Lipid Res ; 40(6): 467-97, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11591436

ABSTRACT

Fatty acid biosynthesis, the first stage in membrane lipid biogenesis, is catalyzed in most bacteria by a series of small, soluble proteins that are each encoded by a discrete gene (Fig. 1; Table 1). This arrangement is termed the type II fatty acid synthase (FAS) system and contrasts sharply with the type I FAS of eukaryotes which is a dimer of a single large, multifunctional polypeptide. Thus, the bacterial pathway offers several unique sites for selective inhibition by chemotherapeutic agents. The site of action of isoniazid, used in the treatment of tuberculosis for 50 years, and the consumer antimicrobial agent triclosan were revealed recently to be the enoyl-ACP reductase of the type II FAS. The fungal metabolites, cerulenin and thiolactomycin, target the condensing enzymes of the bacterial pathway while the dehydratase/isomerase is inhibited by a synthetic acetylenic substrate analogue. Transfer of fatty acids to the membrane has also been inhibited via interference with the first acyltransferase step, while a new class of drugs targets lipid A synthesis. This review will summarize the data generated on these inhibitors to date, and examine where additional efforts will be required to develop new chemotherapeutics to help combat microbial infections.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacteria/metabolism , Enzyme Inhibitors/pharmacology , Fatty Acids/biosynthesis , Gene Expression Regulation/drug effects , Lipids/biosynthesis , Acetyl-CoA Carboxylase/metabolism , Bacteria/genetics , Bacterial Infections/drug therapy , Bacterial Proteins/genetics , Base Sequence , Desiccation , Enzyme Inhibitors/therapeutic use , Escherichia coli/genetics , Fatty Acid Synthases/metabolism , Fatty Acids/genetics , Humans , Lipid A/biosynthesis , Models, Biological , Molecular Sequence Data , Phospholipids/biosynthesis , Protein Conformation , Sequence Alignment , Streptococcus pneumoniae/genetics
4.
J Bacteriol ; 183(16): 4927-31, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11466299
6.
J Biol Chem ; 276(20): 17373-9, 2001 May 18.
Article in English | MEDLINE | ID: mdl-11279025

ABSTRACT

In Escherichia coli, the expression of fatty acid metabolic genes is controlled by the transcription factor, FadR. The affinity of FadR for DNA is controlled by long chain acyl-CoA molecules, which bind to the protein and modulate gene expression. The crystal structure of FadR reveals a two domain dimeric molecule where the N-terminal domains bind DNA, and the C-terminal domains bind acyl-CoA. The DNA binding domain has a winged-helix motif, and the C-terminal domain resembles the sensor domain of the Tet repressor. The FadR.DNA complex reveals how the protein interacts with DNA and specifically recognizes a palindromic sequence. Structural and functional similarities to the Tet repressor and the BmrR transcription factors suggest how the binding of the acyl-CoA effector molecule to the C-terminal domain may affect the DNA binding affinity of the N-terminal domain. We suggest that the binding of acyl-CoA disrupts a buried network of charged and polar residues in the C-terminal domain, and the resulting conformational change is transmitted to the N-terminal domain via a domain-spanning alpha-helix.


Subject(s)
Acyl Coenzyme A/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , DNA, Bacterial/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression Regulation, Bacterial , Repressor Proteins/chemistry , Repressor Proteins/metabolism , Bacterial Proteins/genetics , Binding Sites , Crystallography, X-Ray , DNA, Bacterial/chemistry , DNA, Bacterial/genetics , Models, Molecular , Nucleic Acid Conformation , Protein Structure, Secondary , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Repressor Proteins/genetics , Transcription Factors/metabolism
7.
J Biol Chem ; 276(9): 6551-9, 2001 Mar 02.
Article in English | MEDLINE | ID: mdl-11050088

ABSTRACT

The beta-ketoacyl-acyl carrier protein (ACP) synthases are key regulators of type II fatty acid synthesis and are the targets for two natural products, thiolactomycin (TLM) and cerulenin. The high resolution structures of the FabB-TLM and FabB-cerulenin binary complexes were determined. TLM mimics malonyl-ACP in the FabB active site. It forms strong hydrogen bond interactions with the two catalytic histidines, and the unsaturated alkyl side chain interaction with a small hydrophobic pocket is stabilized by pi stacking interactions. Cerulenin binding mimics the condensation transition state. The subtle differences between the FabB-cerulenin and FabF-cerulenin (Moche, M., Schneider, G., Edwards, P., Dehesh, K., and Lindqvist, Y. (1999) J. Biol. Chem. 244, 6031-6034) structures explain the differences in the sensitivity of the two enzymes to the antibiotic and may reflect the distinct substrate specificities that differentiate the two enzymes. The FabB[H333N] protein was prepared to convert the FabB His-His-Cys active site triad into the FabH His-Asn-Cys configuration to test the importance of the two His residues in TLM and cerulenin binding. FabB[H333N] was significantly more resistant to both antibiotics than FabB and had an affinity for TLM an order of magnitude less than the wild-type enzyme, illustrating that the two-histidine active site architecture is critical to protein-antibiotic interaction. These data provide a structural framework for understanding antibiotic sensitivity within this group of enzymes.


Subject(s)
3-Oxoacyl-(Acyl-Carrier-Protein) Synthase/antagonists & inhibitors , Cerulenin/pharmacology , Enzyme Inhibitors/pharmacology , Isoenzymes/antagonists & inhibitors , Thiophenes/pharmacology , 3-Oxoacyl-(Acyl-Carrier-Protein) Synthase/chemistry , Binding Sites , Cerulenin/chemistry , Isoenzymes/chemistry , Structure-Activity Relationship , Thiophenes/chemistry , Thiophenes/metabolism
8.
J Biol Chem ; 276(11): 8231-8, 2001 Mar 16.
Article in English | MEDLINE | ID: mdl-11078736

ABSTRACT

The molecular details that govern the specific interactions between acyl carrier protein (ACP) and the enzymes of fatty acid biosynthesis are unknown. We investigated the mechanism of ACP-protein interactions using a computational analysis to dock the NMR structure of ACP with the crystal structure of beta-ketoacyl-ACP synthase III (FabH) and experimentally tested the model by the biochemical analysis of FabH mutants. The activities of the mutants were assessed using both an ACP-dependent and an ACP-independent assay. The ACP interaction surface was defined by mutations that compromised FabH activity in the ACP-dependent assay but had no effect in the ACP-independent assay. ACP docked to a positively charged/hydrophobic patch adjacent to the active site tunnel on FabH, which included a conserved arginine (Arg-249) that was required for ACP docking. Kinetic analysis and direct binding studies between FabH and ACP confirmed the identification of Arg-249 as critical for FabH-ACP interaction. Our experiments reveal the significance of the positively charged/hydrophobic patch located adjacent to the active site cavities of the fatty acid biosynthesis enzymes and the high degree of sequence conservation in helix II of ACP across species.


Subject(s)
3-Oxoacyl-(Acyl-Carrier-Protein) Synthase/chemistry , Acyl Carrier Protein/metabolism , 3-Oxoacyl-(Acyl-Carrier-Protein) Synthase/metabolism , Amino Acid Sequence , Binding Sites , Magnetic Resonance Spectroscopy , Models, Molecular , Molecular Sequence Data , Protein Conformation , Protein Structure, Secondary
9.
J Biol Chem ; 275(51): 40128-33, 2000 Dec 22.
Article in English | MEDLINE | ID: mdl-11007778

ABSTRACT

Enoyl-[acyl-carrier-protein] (ACP) reductase is a key enzyme in type II fatty-acid synthases that catalyzes the last step in each elongation cycle. The FabI component of Bacillus subtilis (bsFabI) was identified in the genomic data base by homology to the Escherichia coli protein. bsFabI was cloned and purified and exhibited properties similar to those of E. coli FabI, including a marked preference for NADH over NADPH as a cofactor. Overexpression of the B. subtilis fabI gene complemented the temperature-sensitive growth phenotype of an E. coli fabI mutant. Triclosan was a slow-binding inhibitor of bsFabI and formed a stable bsFabI.NAD(+). triclosan ternary complex. Analysis of the B. subtilis genomic data base revealed a second open reading frame (ygaA) that was predicted to encode a protein with a relatively low overall similarity to FabI, but contained the Tyr-Xaa(6)-Lys enoyl-ACP reductase catalytic architecture. The purified YgaA protein catalyzed the NADPH-dependent reduction of trans-2-enoyl thioesters of both N-acetylcysteamine and ACP. YgaA was reversibly inhibited by triclosan, but did not form the stable ternary complex characteristic of the FabI proteins. Expression of YgaA complemented the fabI(ts) defect in E. coli and conferred complete triclosan resistance. Single knockouts of the ygaA or fabI gene in B. subtilis were viable, but double knockouts were not obtained. The fabI knockout was as sensitive as the wild-type strain to triclosan, whereas the ygaA knockout was 250-fold more sensitive to the drug. YgaA was renamed FabL to denote the discovery of a new family of proteins that carry out the enoyl-ACP reductase step in type II fatty-acid synthases.


Subject(s)
Bacillus subtilis/enzymology , Oxidoreductases/metabolism , Amino Acid Sequence , Base Sequence , DNA Primers , Enoyl-(Acyl-Carrier-Protein) Reductase (NADH) , Enzyme Inhibitors/pharmacology , Molecular Sequence Data , Oxidoreductases/antagonists & inhibitors , Oxidoreductases/chemistry , Oxidoreductases/genetics , Sequence Homology, Amino Acid , Triclosan/pharmacology
11.
J Biol Chem ; 275(7): 4654-9, 2000 Feb 18.
Article in English | MEDLINE | ID: mdl-10671494

ABSTRACT

Enoyl-acyl carrier protein reductase (FabI) plays a determinant role in completing cycles of elongation in type II fatty acid synthase systems and is an important target for antibacterial drugs. The FabI component of Staphylococcus aureus (saFabI) was identified, and its properties were compared with Escherichia coli FabI (ecFabI). ecFabI and saFabI had similar specific activities, and saFabI expression complemented the E. coli fabI(Ts) mutant, illustrating that the Gram-positive FabI was interchangeable with the Gram-negative FabI enzyme. However, ecFabI was specific for NADH, whereas saFabI exhibited specific and positive cooperative binding of NADPH. Triclosan and hexachlorophene inhibited both ecFabI and saFabI. The triclosan-resistant ecFabI(G93V) protein was also refractory to hexachlorophene inhibition, illustrating that both drugs bind at the FabI active site. Both the introduction of a plasmid expressing the safabI gene or a missense mutation in the chromosomal safabI gene led to triclosan resistance in S. aureus; however, these strains did not exhibit cross-resistance to hexachlorophene. The replacement of the ether linkage in triclosan by a carbon bridge in hexachlorophene prevented the formation of a stable FabI-NAD(P)(+)-drug ternary complex. Thus, the formation of this ternary complex is a key determinant of the antibacterial activity of FabI inhibitors.


Subject(s)
Enzyme Inhibitors/pharmacology , Hexachlorophene/pharmacology , Oxidoreductases/antagonists & inhibitors , Staphylococcus aureus/enzymology , Triclosan/pharmacology , Amino Acid Sequence , Base Sequence , Cloning, Molecular , DNA Primers , Enoyl-(Acyl-Carrier Protein) Reductase (NADPH, B-Specific) , Enzyme Inhibitors/chemistry , Escherichia coli/drug effects , Escherichia coli/genetics , Hexachlorophene/chemistry , Microbial Sensitivity Tests , Molecular Sequence Data , Molecular Structure , Sequence Homology, Amino Acid , Staphylococcus aureus/drug effects , Staphylococcus aureus/growth & development , Triclosan/chemistry
12.
Structure ; 8(2): 185-95, 2000 Feb 15.
Article in English | MEDLINE | ID: mdl-10673437

ABSTRACT

BACKGROUND: beta-Ketoacyl-acyl carrier protein synthase III (FabH) initiates elongation in type II fatty acid synthase systems found in bacteria and plants. FabH is a ubiquitous component of the type II system and is positioned ideally in the pathway to control the production of fatty acids. The elucidation of the structure of FabH is important for the understanding of its regulation by feedback inhibition and its interaction with drugs. Although the structures of two related condensing enzymes are known, the roles of the active-site residues have not been experimentally tested. RESULTS: The 1.8 A crystal structure of FabH was determined using a 12-site selenium multiwavelength anomalous dispersion experiment. The active site (Cys112, His244 and Asn274) is formed by the convergence of two alpha helices and is accessed via a narrow hydrophobic tunnel. Hydrogen-bonding networks that include two tightly bound water molecules fix the positions of His244 and Asn274, which are critical for the decarboxylation and condensation reactions. Surprisingly, the His244-->Ala mutation does not affect the transacylation reaction suggesting that His244 has only a minor influence on the nucleophilicity of Cys112. CONCLUSIONS: The histidine and asparagine active-site residues are both required for the decarboxylation step in the condensation reaction. The nucleophilicity of the active-site cysteine is enhanced by the alpha-helix dipole effect, and an oxyanion hole promotes the formation of the tetrahedral transition state.


Subject(s)
3-Oxoacyl-(Acyl-Carrier-Protein) Synthase/chemistry , Escherichia coli/enzymology , 3-Oxoacyl-(Acyl-Carrier-Protein) Synthase/genetics , 3-Oxoacyl-(Acyl-Carrier-Protein) Synthase/metabolism , Amino Acid Sequence , Base Sequence , Binding Sites , Catalysis , Crystallography, X-Ray , DNA Primers , Hydrogen Bonding , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Protein Conformation
13.
J Bacteriol ; 182(2): 365-70, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10629181

ABSTRACT

A universal set of genes encodes the components of the dissociated, type II, fatty acid synthase system that is responsible for producing the multitude of fatty acid structures found in bacterial membranes. We examined the biochemical basis for the production of branched-chain fatty acids by gram-positive bacteria. Two genes that were predicted to encode homologs of the beta-ketoacyl-acyl carrier protein synthase III of Escherichia coli (eFabH) were identified in the Bacillus subtilis genome. Their protein products were expressed, purified, and biochemically characterized. Both B. subtilis FabH homologs, bFabH1 and bFabH2, carried out the initial condensation reaction of fatty acid biosynthesis with acetyl-coenzyme A (acetyl-CoA) as a primer, although they possessed lower specific activities than eFabH. bFabH1 and bFabH2 also utilized iso- and anteiso-branched-chain acyl-CoA primers as substrates. eFabH was not able to accept these CoA thioesters. Reconstitution of a complete round of fatty acid synthesis in vitro with purified E. coli proteins showed that eFabH was the only E. coli enzyme incapable of using branched-chain substrates. Expression of either bFabH1 or bFabH2 in E. coli resulted in the appearance of a branched-chain 17-carbon fatty acid. Thus, the substrate specificity of FabH is an important determinant of branched-chain fatty acid production.


Subject(s)
3-Oxoacyl-(Acyl-Carrier-Protein) Synthase/metabolism , Fatty Acids/biosynthesis , Amino Acid Sequence , Escherichia coli/enzymology , Escherichia coli/metabolism , Molecular Sequence Data , Substrate Specificity
14.
J Biol Chem ; 274(16): 11110-4, 1999 Apr 16.
Article in English | MEDLINE | ID: mdl-10196195

ABSTRACT

Triclosan is a broad-spectrum antibacterial agent that inhibits bacterial fatty acid synthesis at the enoyl-acyl carrier protein reductase (FabI) step. Resistance to triclosan in Escherichia coli is acquired through a missense mutation in the fabI gene that leads to the expression of FabI[G93V]. The specific activity and substrate affinities of FabI[G93V] are similar to FabI. Two different binding assays establish that triclosan dramatically increases the affinity of FabI for NAD+. In contrast, triclosan does not increase the binding of NAD+ to FabI[G93V]. The x-ray crystal structure of the FabI-NAD+-triclosan complex confirms that hydrogen bonds and hydrophobic interactions between triclosan and both the protein and the NAD+ cofactor contribute to the formation of a stable ternary complex, with the drug binding at the enoyl substrate site. These data show that the formation of a noncovalent "bi-substrate" complex accounts for the effectiveness of triclosan as a FabI inhibitor and illustrates that mutations in the FabI active site that interfere with the formation of a stable FabI-NAD+-triclosan ternary complex acquire resistance to the drug.


Subject(s)
Fatty Acids/antagonists & inhibitors , Oxidoreductases/antagonists & inhibitors , Triclosan/pharmacology , Enoyl-(Acyl-Carrier-Protein) Reductase (NADH) , Fatty Acids/biosynthesis , Models, Molecular , Molecular Structure , Mutation, Missense , NAD/metabolism , Oxidoreductases/genetics , Oxidoreductases/metabolism , Protein Binding , Triclosan/chemistry , Triclosan/metabolism
15.
J Bacteriol ; 181(6): 1944-6, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10074094

ABSTRACT

The sn-glycerol-3-phosphate acyltransferase (plsB) catalyzes the first step in membrane phospholipid formation. A conditional Escherichia coli mutant (plsB26) has a single missense mutation (G1045A) predicting the expression of an acyltransferase with an Ala349Thr substitution. The PlsB26 protein had a significantly reduced glycerol-3-phosphate acyltransferase specific activity coupled with an elevated Km for glycerol-3-phosphate.


Subject(s)
Escherichia coli/enzymology , Escherichia coli/genetics , Glycerol-3-Phosphate O-Acyltransferase/genetics , Glycerol-3-Phosphate O-Acyltransferase/metabolism , Mutation, Missense , Alleles , Base Sequence , DNA Primers/genetics , DNA, Bacterial/genetics , Gene Expression , Genes, Bacterial , Kinetics , Molecular Sequence Data
16.
J Biol Chem ; 273(46): 30316-20, 1998 Nov 13.
Article in English | MEDLINE | ID: mdl-9804793

ABSTRACT

The broad spectrum antibacterial properties of 2-hydroxydiphenyl ethers have been appreciated for decades, and their use in consumer products is rapidly increasing. We identify the enoyl-acyl carrier protein reductase (fabI) component of the type II fatty acid synthase system as the specific cellular target for these antibacterials. Biologically active 2-hydroxydiphenyl ethers effectively inhibit fatty acid synthesis in vivo and FabI activity in vitro. Resistant mechanisms include up-regulation of fabI expression and spontaneously arising missense mutations in the fabI gene. These results contradict the view that these compounds directly disrupt membranes and suggest that their widespread use will select for resistant bacterial populations.


Subject(s)
Fatty Acid Synthases/metabolism , Oxidoreductases/antagonists & inhibitors , Animals , Anti-Infective Agents, Local/pharmacology , Cattle , Enoyl-(Acyl-Carrier-Protein) Reductase (NADH) , In Vitro Techniques , Models, Chemical , Mutagenesis, Site-Directed , Oxidoreductases/genetics , Phenols/pharmacology , Triclosan/pharmacology
17.
J Bacteriol ; 180(6): 1425-30, 1998 Mar.
Article in English | MEDLINE | ID: mdl-9515909

ABSTRACT

Sequence analysis of membrane-bound glycerolipid acyltransferases revealed that proteins from the bacterial, plant, and animal kingdoms share a highly conserved domain containing invariant histidine and aspartic acid residues separated by four less conserved residues in an HX4D configuration. We investigated the role of the invariant histidine residue in acyltransferase catalysis by site-directed mutagenesis of two representative members of this family, the sn-glycerol-3-phosphate acyltransferase (PlsB) and the bifunctional 2-acyl-glycerophosphoethanolamine acyltransferase/acyl-acyl carrier protein synthetase (Aas) of Escherichia coli. Both the PlsB[H306A] and Aas[H36A] mutants lacked acyltransferase activity. However, the Aas[H36A] mutant retained significant acyl-acyl carrier protein synthetase activity, illustrating that the lack of acyltransferase activity was specifically associated with the H36A substitution. The invariant aspartic acid residue in the HX4D pattern was also important. The substitution of aspartic acid 311 with glutamic acid in PlsB resulted in an enzyme with significantly reduced catalytic activity. Substitution of an alanine at this position eliminated acyltransferase activity; however, the PlsB[D311A] mutant protein did not assemble into the membrane, indicating that aspartic acid 311 is also important for the proper folding and membrane insertion of the acyltransferases. These data are consistent with a mechanism for glycerolipid acyltransferase catalysis where the invariant histidine functions as a general base to deprotonate the hydroxyl moiety of the acyl acceptor.


Subject(s)
Acyltransferases/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Glycerol-3-Phosphate O-Acyltransferase/metabolism , Histidine/genetics , Acyltransferases/genetics , Alanine/genetics , Amino Acid Sequence , Amino Acid Substitution , Aspartic Acid/genetics , Cell Membrane/metabolism , Cloning, Molecular , DNA, Bacterial/analysis , DNA, Bacterial/genetics , Gene Expression , Genetic Complementation Test , Glutamic Acid/genetics , Glycerol-3-Phosphate O-Acyltransferase/genetics , Molecular Sequence Data , Plasmids , Polymerase Chain Reaction , Protein Biosynthesis , Sequence Alignment , Sequence Analysis, DNA , Transcription, Genetic
18.
J Bacteriol ; 179(23): 7257-63, 1997 Dec.
Article in English | MEDLINE | ID: mdl-9393688

ABSTRACT

The sn-glycerol-3-phosphate acyltransferase (plsB) of Escherichia coli is a key regulatory enzyme that catalyzes the first committed step in phospholipid biosynthesis. We report the initial characterization of a novel gene (termed plsD) from Clostridium butyricum, cloned based on its ability to complement the sn-glycerol-3-phosphate auxotrophic phenotype of a plsB mutant strain of E. coli. Unlike the 83-kDa PlsB acyltransferase from E. coli, the predicted plsD open reading frame encoded a protein of 26.5 kDa. Two regions of strong homology to other lipid acyltransferases, including PlsB and PlsC analogs from mammals, plants, yeast, and bacteria, were identified. PlsD was most closely related to the 1-acyl-sn-glycerol-3-phosphate acyltransferase (plsC) gene family but did not complement the growth of plsC(Ts) mutants. An in vivo metabolic labeling experiment using a plsB plsX plsC(Ts) strain of E. coli confirmed that the plsD expression restored the ability of the cells to synthesize 1-acyl-glycerol-3-phosphate. However, glycerol-3-phosphate acyltransferase activity was not detected in vitro in assays using either acyl-acyl carrier protein or acyl coenzyme A as the substrate.


Subject(s)
Clostridium/genetics , Genes, Bacterial/genetics , Glycerol-3-Phosphate O-Acyltransferase/genetics , Amino Acid Sequence , Base Sequence , Cloning, Molecular , Clostridium/enzymology , Escherichia coli/enzymology , Escherichia coli/genetics , Genetic Complementation Test , Molecular Sequence Data , Sequence Analysis, DNA , Sequence Homology, Amino Acid , Species Specificity
19.
J Biol Chem ; 271(44): 27795-801, 1996 Nov 01.
Article in English | MEDLINE | ID: mdl-8910376

ABSTRACT

There are two genes, fabA and fabZ, encoding beta-hydroxyacyl-acyl carrier protein (ACP) dehydratases that function in the dissociated, type II fatty acid synthase system of Escherichia coli. We have investigated their roles in fatty acid synthesis by purifying the two proteins and reconstituting cycles of fatty acid synthesis in vitro using five other purified proteins. FabA and FabZ exhibited broad, overlapping chain length specificities. The FabZ dehydratase efficiently catalyzed the dehydration of short chain beta-hydroxyacyl-ACPs and long chain saturated and unsaturated beta-hydroxyacyl-ACPs. FabA was most active on intermediate chain length beta-hydroxyacyl-ACPs and also possessed significant activity toward both short and long chain saturated beta-hydroxyacyl-ACPs. Significantly, FabA was virtually inactive in the dehydration of long chain unsaturated beta-hydroxyacyl-ACP. The introduction of the double bond at the 10-carbon stage of fatty acid synthesis by FabA was only detected in the presence of beta-ketoacyl-ACP synthase I (FabB). A yeast two-hybrid analysis failed to detect an interaction between FabA and FabB, therefore the channeling of intermediates toward unsaturated fatty acid synthesis by FabB was attributed to the affinity of the condensing enzyme for cis-decenoyl-ACP. The broad substrate specificity of FabZ coupled with the inactivity of FabA toward a long chain unsaturated beta-hydroxyacyl-ACP provides a biochemical explanation for the phenotypes of cells with genetically altered levels of the two dehydratases.


Subject(s)
Escherichia coli/enzymology , Fatty Acids/biosynthesis , Hydro-Lyases/metabolism , Cloning, Molecular , Fatty Acid Synthase, Type II , Fatty Acids, Unsaturated/metabolism , Kinetics , Models, Biological , Recombinant Proteins/metabolism , Saccharomyces cerevisiae , Substrate Specificity
20.
J Biol Chem ; 271(18): 10996-1000, 1996 May 03.
Article in English | MEDLINE | ID: mdl-8631920

ABSTRACT

beta-Ketoacyl-acyl carrier protein (ACP) synthase III (the fabH gene product) condenses acetyl-CoA with malonyl-ACP to initiate fatty acid biosynthesis in the dissociated, type II fatty acid synthase systems typified by Escherichia coli. The accumulation of malonyl-acyl carrier protein (ACP) following the inhibition of a reconstituted fatty acid synthase system by acyl-ACP implicated synthase III (FabH) as a target for acyl-ACP regulation (Heath, R. J., and Rock, C. O. (1996) J. Biol. Chem. 271, 1833-1836); therefore, the FabH protein was purified and its biochemical and regulatory properties examined. FabH exhibited a Km of 40 microM for acetyl-CoA and 5 microM for malonyl-ACP. FabH also accepted other acyl-CoAs as primers with the rank order of activity being acetyl-CoA approximately propionyl-CoA >> butyryl-CoA. FabH utilized neither hexanoyl-CoA nor octanoyl-CoA. Acyl-ACPs suppressed Fabh activity, and their potency increased with increasing acyl chain length between 12 and 20 carbon atoms. Nonesterified ACP was not an inhibitor. Acyl-ACP inhibition kinetics were mixed with respect to acetyl-CoA, but were competitive with malonyl-ACP, indicating that acyl-ACPs decrease FabH activity by binding to either the free enzyme or the acyl-enzyme intermediate. These data support the concept that the inhibition of chain initiation at the beta-ketoacyl-ACP synthase III step contributes to the attenuation of fatty acid biosynthesis by acyl-ACP.


Subject(s)
3-Oxoacyl-(Acyl-Carrier-Protein) Synthase/antagonists & inhibitors , Acyl Carrier Protein/physiology , Escherichia coli/enzymology , 3-Oxoacyl-(Acyl-Carrier-Protein) Synthase/isolation & purification , 3-Oxoacyl-(Acyl-Carrier-Protein) Synthase/metabolism , Acetyl Coenzyme A/metabolism , Kinetics , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...