Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Arch Biochem Biophys ; 691: 108507, 2020 09 30.
Article in English | MEDLINE | ID: mdl-32710884

ABSTRACT

Mammalian carnitine acetyltransferase (CrAT) is a mitochondrial enzyme that catalyzes the reversible transfer of an acetyl group from acetyl-CoA to carnitine. CrAT knockout studies have shown that this enzyme is critical to sustain metabolic flexibility, or the ability to switch between different fuel types, an underlying theme of the metabolic syndrome. These recent physiological findings imply that CrAT dysfunction, or its catalytic impairment, may lead to disease. To gain insight into the CrAT kinetic mechanism, we conducted stopped-flow experiments in various enzyme substrate/product conditions and analyzed full progress curves by global fitting. Simultaneous mixing of both substrates with CrAT produced relatively fast kinetics that follows an ordered bi bi mechanism. A great preference for ordered binding is supported by stopped-flow double mixing experiments such that premixed CrAT with acetyl-CoA or CoA demonstrated a biphasic decrease in initial rate that produces about a 100-fold attenuation in catalysis. Double mixing experiments also revealed that the CrAT initial rate is inhibited by 50% in approximately 8 s by either acetyl-CoA or CoA premixing. Analysis of available CrAT structures support a substrate conformational change between acetyl-CoA/CoA binary versus ternary complexes. Additional viscosity-based kinetic experiments yielded strong evidence that product release is the rate limiting step in the CrAT-catalyzed reaction.


Subject(s)
Carnitine O-Acetyltransferase/chemistry , Acetyl Coenzyme A/chemistry , Acetyl Coenzyme A/metabolism , Animals , Carnitine/chemistry , Carnitine/metabolism , Carnitine O-Acetyltransferase/metabolism , Catalysis , Catalytic Domain , Coenzyme A/chemistry , Coenzyme A/metabolism , Columbidae , Crystallography, X-Ray , Kinetics , Mice , Protein Binding
2.
Hum Mutat ; 41(1): 110-114, 2020 01.
Article in English | MEDLINE | ID: mdl-31448845

ABSTRACT

Leigh syndrome, or subacute necrotizing encephalomyelopathy, is one of the most severe pediatric disorders of the mitochondrial energy metabolism. By performing whole-exome sequencing in a girl affected by Leigh syndrome and her parents, we identified two heterozygous missense variants (p.Tyr110Cys and p.Val569Met) in the carnitine acetyltransferase (CRAT) gene, encoding an enzyme involved in the control of mitochondrial short-chain acyl-CoA concentrations. Biochemical assays revealed carnitine acetyltransferase deficiency in the proband-derived fibroblasts. Functional analyses of recombinant-purified CRAT proteins demonstrated that both missense variants, located in the acyl-group binding site of the enzyme, severely impair its catalytic function toward acetyl-CoA, and the p.Val569Met variant also toward propionyl-CoA and octanoyl-CoA. Although a single recessive variant in CRAT has been recently associated with neurodegeneration with brain iron accumulation (NBIA), this study reports the first kinetic analysis of naturally occurring CRAT variants and demonstrates the genetic basis of carnitine acetyltransferase deficiency in a case of mitochondrial encephalopathy.


Subject(s)
Carnitine O-Acetyltransferase/genetics , Carnitine O-Acetyltransferase/metabolism , Leigh Disease/genetics , Leigh Disease/metabolism , Mutation, Missense , Age of Onset , Binding Sites , Carnitine O-Acetyltransferase/chemistry , DNA Mutational Analysis , Enzyme Activation , Humans , Leigh Disease/diagnosis , Models, Molecular , Protein Binding , Protein Conformation , Structure-Activity Relationship
3.
J Org Chem ; 79(10): 4644-9, 2014 May 16.
Article in English | MEDLINE | ID: mdl-24742295

ABSTRACT

N-Bromosuccinimide-induced electrophilic multicomponent reaction has been applied to the synthesis of Reboxetine intermediate, a highly potent selective norepinephrine reuptake inhibitor. By simply changing the olefinic partner, the synthesis of a carnitine acetyltransferase inhibitor, which contains a 2,6,6-trisubstituted morpholine system, can be accomplished.


Subject(s)
Adrenergic Uptake Inhibitors/chemistry , Adrenergic Uptake Inhibitors/pharmacology , Bromosuccinimide/chemistry , Carnitine O-Acetyltransferase/antagonists & inhibitors , Enzyme Inhibitors/chemical synthesis , Morpholines/chemical synthesis , Norepinephrine/chemistry , Norepinephrine/pharmacology , Carnitine O-Acetyltransferase/chemistry , Enzyme Inhibitors/chemistry , Morpholines/chemistry , Reboxetine
4.
Biochim Biophys Acta ; 1832(6): 773-9, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23485643

ABSTRACT

Carnitine acyltransferases catalyze the reversible conversion of acyl-CoAs into acylcarnitine esters. This family includes the mitochondrial enzymes carnitine palmitoyltransferase 2 (CPT2) and carnitine acetyltransferase (CrAT). CPT2 is part of the carnitine shuttle that is necessary to import fatty acids into mitochondria and catalyzes the conversion of acylcarnitines into acyl-CoAs. In addition, when mitochondrial fatty acid ß-oxidation is impaired, CPT2 is able to catalyze the reverse reaction and converts accumulating long- and medium-chain acyl-CoAs into acylcarnitines for export from the matrix to the cytosol. However, CPT2 is inactive with short-chain acyl-CoAs and intermediates of the branched-chain amino acid oxidation pathway (BCAAO). In order to explore the origin of short-chain and branched-chain acylcarnitines that may accumulate in various organic acidemias, we performed substrate specificity studies using purified recombinant human CrAT. Various saturated, unsaturated and branched-chain acyl-CoA esters were tested and the synthesized acylcarnitines were quantified by ESI-MS/MS. We show that CrAT converts short- and medium-chain acyl-CoAs (C2 to C10-CoA), whereas no activity was observed with long-chain species. Trans-2-enoyl-CoA intermediates were found to be poor substrates for this enzyme. Furthermore, CrAT turned out to be active towards some but not all the BCAAO intermediates tested and no activity was found with dicarboxylic acyl-CoA esters. This suggests the existence of another enzyme able to handle the acyl-CoAs that are not substrates for CrAT and CPT2, but for which the corresponding acylcarnitines are well recognized as diagnostic markers in inborn errors of metabolism.


Subject(s)
Amino Acids, Branched-Chain/chemistry , Amino Acids, Branched-Chain/metabolism , Carnitine O-Acetyltransferase/chemistry , Carnitine O-Acetyltransferase/metabolism , Fatty Acids/chemistry , Fatty Acids/metabolism , Acyl Coenzyme A/chemistry , Acyl Coenzyme A/genetics , Acyl Coenzyme A/metabolism , Amino Acids, Branched-Chain/genetics , Carnitine O-Acetyltransferase/genetics , Carnitine O-Palmitoyltransferase/chemistry , Carnitine O-Palmitoyltransferase/genetics , Carnitine O-Palmitoyltransferase/metabolism , Fatty Acids/genetics , Humans , Substrate Specificity/physiology
5.
J Biol Chem ; 285(32): 24335-46, 2010 Aug 06.
Article in English | MEDLINE | ID: mdl-20522553

ABSTRACT

Transport of acetyl-CoA between intracellular compartments is mediated by carnitine acetyltransferases (Cats) that reversibly link acetyl units to the carrier molecule carnitine. The genome of the opportunistic pathogenic yeast Candida albicans encodes several (putative) Cats: the peroxisomal and mitochondrial Cat2 isoenzymes encoded by a single gene and the carnitine acetyltransferase homologs Yat1 and Yat2. To determine the contributions of the individual Cats, various carnitine acetyltransferase mutant strains were constructed and subjected to phenotypic and biochemical analyses on different carbon sources. We show that mitochondrial Cat2 is required for the intramitochondrial conversion of acetylcarnitine to acetyl-CoA, which is essential for a functional tricarboxylic acid cycle during growth on oleate, acetate, ethanol, and citrate. Yat1 is cytosolic and contributes to acetyl-CoA transport from the cytosol during growth on ethanol or acetate, but its activity is not required for growth on oleate. Yat2 is also cytosolic, but we were unable to attribute any function to this enzyme. Surprisingly, peroxisomal Cat2 is essential neither for export of acetyl units during growth on oleate nor for the import of acetyl units during growth on acetate or ethanol. Oxidation of fatty acids still takes place in the absence of peroxisomal Cat2, but biomass formation is absent, and the strain displays a growth delay on acetate and ethanol that can be partially rescued by the addition of carnitine. Based on our results, we present a model for the intracellular flow of acetyl units under various growth conditions and the roles of each of the Cats in this process.


Subject(s)
Candida albicans/enzymology , Carnitine O-Acetyltransferase/metabolism , Biological Transport , Carbon/chemistry , Carnitine O-Acetyltransferase/chemistry , Cell Membrane/metabolism , Fatty Acids/chemistry , Mass Spectrometry/methods , Membrane Proteins/metabolism , Mitochondria/metabolism , Models, Biological , Mutation , Oxygen/chemistry , Peroxisomes/chemistry , Peroxisomes/metabolism , Phenotype , Saccharomyces cerevisiae Proteins/metabolism , Two-Hybrid System Techniques
6.
J Enzyme Inhib Med Chem ; 24(6): 1269-75, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19912061

ABSTRACT

Carnitine acetyltransferase (CrAT; EC 2.3.1.7) catalyzes the reversible transfer of acetyl groups between acetyl-coenzyme A (acetyl-CoA) and L-carnitine; it also regulates the cellular pool of CoA and the availability of activated acetyl groups. In this study, biochemical measurements, saturation transfer difference (STD) nuclear magnetic resonance (NMR) spectroscopy, and molecular docking were applied to give insights into the CrAT binding of a synthetic inhibitor, the cardioprotective drug mildronate (3-(2,2,2-trimethylhydrazinium)-propionate). The obtained results show that mildronate inhibits CrAT in a competitive manner through binding to the carnitine binding site, not the acetyl-CoA binding site. The bound conformation of mildronate closely resembles that of carnitine except for the orientation of the trimethylammonium group, which in the mildronate molecule is exposed to the solvent. The dissociation constant of the mildronate CrAT complex is approximately 0.1 mM, and the K(i) is 1.6 mM. The results suggest that the cardioprotective effect of mildronate might be partially mediated by CrAT inhibition and concomitant regulation of cellular energy metabolism pathways.


Subject(s)
Cardiovascular Agents/pharmacology , Carnitine O-Acetyltransferase/antagonists & inhibitors , Energy Metabolism/drug effects , Enzyme Inhibitors/pharmacology , Methylhydrazines/pharmacology , Animals , Binding Sites , Biocatalysis , Cardiovascular Agents/chemistry , Cardiovascular Agents/metabolism , Carnitine O-Acetyltransferase/chemistry , Carnitine O-Acetyltransferase/metabolism , Columbidae , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Magnetic Resonance Spectroscopy , Methylhydrazines/chemistry , Methylhydrazines/metabolism , Molecular Dynamics Simulation , Structure-Activity Relationship , Substrate Specificity
7.
Cell Mol Biol Lett ; 14(1): 90-9, 2009.
Article in English | MEDLINE | ID: mdl-18839069

ABSTRACT

Carnitine acetyltransferase (CRAT) is an important enzyme for energy homeostasis and fat metabolism. We characterized the predicted full length cDNA sequence of the porcine CRAT gene. Its structure is very similar to that in humans with respect to the size and organization of the 14 exons. We demonstrated the existence of a porcine alternative transcript resulting from a partial intron-retention at the 5' end of exon 2. To perform a comparison of the 5' end variants of the mammalian CRAT gene, we analyzed the Genbank data, and here we propose a new 5' variant for dog, rat and mouse. In contrast to other mammals where this variant encodes a shorter protein (-21 aa in human, mouse and rat, and -14 aa in dog), the pig variant encodes for a longer protein (+18 aa). In all mammalian species, variant 1 has a high probability of a preferential mitochondrial sub-cellular localization. Nevertheless, it is not evident, in particular in porcine and dog species, that the second variant is associated with a different sub-cellular specificity.


Subject(s)
5' Untranslated Regions/genetics , Carnitine O-Acetyltransferase/genetics , Mammals/genetics , Swine/genetics , Amino Acid Sequence , Animals , Carnitine O-Acetyltransferase/chemistry , Carnitine O-Acetyltransferase/metabolism , Gene Expression Regulation, Enzymologic , Humans , Molecular Sequence Data , Protein Transport , RNA, Messenger/genetics , RNA, Messenger/metabolism , Subcellular Fractions/enzymology , Transcription, Genetic
8.
FEMS Microbiol Lett ; 267(2): 159-66, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17156120

ABSTRACT

The wine yeast Saccharomyces cerevisiae is central in the production of aroma compounds during fermentation. Some of the most important yeast-derived aroma compounds produced are esters. The esters ethyl acetate and isoamyl acetate are formed from alcohols and acetyl-CoA in a reaction catalysed by alcohol acetyltransferases. The pool of acetyl-CoA available in yeast cells could play a key role in the development of ester aromas. Carnitine acetyltransferases catalyse the reversible reaction between carnitine and acetyl-CoA to form acetylcarnitine and free CoA. This reaction is important in transferring activated acetyl groups to the mitochondria and in regulating the acetyl-CoA/CoA pools within the cell. We investigated the effect of overexpressing CAT2, which encodes the major mitochondrial and peroxisomal carnitine acetyltransferase, on the formation of esters and other flavour compounds during fermentation. We also overexpressed a modified CAT2 that results in a protein that localizes to the cytosol. In general, the overexpression of both forms of CAT2 resulted in a reduction in ester concentrations, especially in ethyl acetate and isoamyl acetate. We hypothesize that overproduction of Cat2p favours the formation of acetylcarnitine and CoA and therefore limits the precursor for ester production. Carnitine acetyltransferase expression could potentially to be used successfully in order to modulate wine flavour.


Subject(s)
Carnitine O-Acetyltransferase/metabolism , Odorants , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/enzymology , Wine , Acetates/metabolism , Carnitine O-Acetyltransferase/chemistry , Carnitine O-Acetyltransferase/genetics , Esters/metabolism , Fermentation , Pentanols/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics
9.
J Biol Chem ; 281(38): 28480-7, 2006 Sep 22.
Article in English | MEDLINE | ID: mdl-16870616

ABSTRACT

Carnitine acyltransferases catalyze the reversible exchange of acyl groups between coenzyme A (CoA) and carnitine. They have important roles in many cellular processes, especially the oxidation of long-chain fatty acids in the mitochondria for energy production, and are attractive targets for drug discovery against diabetes and obesity. To help define in molecular detail the catalytic mechanism of these enzymes, we report here the high resolution crystal structure of wild-type murine carnitine acetyltransferase (CrAT) in a ternary complex with its substrates acetyl-CoA and carnitine, and the structure of the S554A/M564G double mutant in a ternary complex with the substrates CoA and hexanoylcarnitine. Detailed analyses suggest that these structures may be good mimics for the Michaelis complexes for the forward and reverse reactions of the enzyme, representing the first time that such complexes of CrAT have been studied in molecular detail. The structural information provides significant new insights into the catalytic mechanism of CrAT and possibly carnitine acyltransferases in general.


Subject(s)
Acetyl Coenzyme A/chemistry , Carnitine O-Acetyltransferase/chemistry , Carnitine/chemistry , Animals , Catalysis , Crystallization , Mice , X-Ray Diffraction
10.
Biochemistry ; 45(19): 6133-41, 2006 May 16.
Article in English | MEDLINE | ID: mdl-16681386

ABSTRACT

Carnitine acyltransferases catalyze the exchange of acyl groups between carnitine and CoA. The members of the family can be classified on the basis of their acyl-CoA selectivity. Carnitine acetyltransferases (CrATs) are very active toward short-chain acyl-CoAs but not toward medium- or long-chain acyl-CoAs. Previously, we identified an amino acid residue (Met(564) in rat CrAT) that was critical to fatty acyl-chain-length specificity. M564G-mutated CrAT behaved as if its natural substrates were medium-chain acyl-CoAs, similar to that of carnitine octanoyltransferase (COT). To extend the specificity of rat CrAT to other substrates, we have performed new mutations. Using in silico molecular modeling procedures, we have now identified a second putative amino acid involved in acyl-CoA specificity (Asp(356) in rat CrAT). The double CrAT mutant D356A/M564G showed 6-fold higher activity toward palmitoyl-CoA than that of the single CrAT mutant M564G and a new activity toward stearoyl-CoA. We show that by performing two amino acid replacements a CrAT can be converted into a pseudo carnitine palmitoyltransferase (CPT) in terms of substrate specificity. To change CrAT specificity from carnitine to choline, we also prepared a mutant CrAT that incorporates four amino acid substitutions (A106M/T465V/T467N/R518N). The quadruple mutant shifted the catalytic discrimination between l-carnitine and choline in favor of the latter substrate and showed a 9-fold increase in catalytic efficiency toward choline compared with that of the wild-type. Molecular in silico docking supports kinetic data for the positioning of substrates in the catalytic site of CrAT mutants.


Subject(s)
Amino Acids/chemistry , Carnitine O-Acetyltransferase/chemistry , Carnitine O-Palmitoyltransferase/chemistry , Amino Acid Sequence , Amino Acids/genetics , Animals , Carnitine O-Acetyltransferase/genetics , Carnitine O-Palmitoyltransferase/genetics , Fluorometry , Kinetics , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Radiometry , Rats , Sequence Homology, Amino Acid , Substrate Specificity
11.
J Biol Chem ; 279(29): 30634-42, 2004 Jul 16.
Article in English | MEDLINE | ID: mdl-15123643

ABSTRACT

Polyketide-associated protein A5 (PapA5) is an acyltransferase that is involved in production of phthiocerol and phthiodiolone dimycocerosate esters, a class of virulence-enhancing lipids produced by Mycobacterium tuberculosis. Structural analysis of PapA5 at 2.75-A resolution reveals a two-domain structure that shares unexpected similarity to structures of chloramphenicol acetyltransferase, dihydrolipoyl transacetylase, carnitine acetyltransferase, and VibH, a non-ribosomal peptide synthesis condensation enzyme. The PapA5 active site includes conserved histidine and aspartic acid residues that are critical to PapA5 acyltransferase activity. PapA5 catalyzes acyl transfer reactions on model substrates that contain long aliphatic carbon chains, and two hydrophobic channels were observed linking the PapA5 surface to the active site with properties consistent with these biochemical activities and substrate preferences. An additional alpha helix not observed in other acyltransferase structures blocks the putative entrance into the PapA5 active site, indicating that conformational changes may be associated with PapA5 activity. PapA5 represents the first structure solved for a protein involved in polyketide synthesis in Mycobacteria.


Subject(s)
Acyltransferases/chemistry , Lipid Metabolism , Mycobacterium tuberculosis/metabolism , Acetyltransferases/chemistry , Amino Acid Sequence , Binding Sites , Carnitine O-Acetyltransferase/chemistry , Chloramphenicol O-Acetyltransferase/chemistry , Chloramphenicol O-Acetyltransferase/metabolism , Crystallography, X-Ray , Dihydrolipoyllysine-Residue Acetyltransferase , Escherichia coli/metabolism , Models, Chemical , Models, Molecular , Molecular Sequence Data , Protein Conformation , Protein Structure, Secondary , Protein Structure, Tertiary , Pyruvate Dehydrogenase Complex/chemistry , Recombinant Proteins/chemistry , Sequence Homology, Amino Acid , Structure-Activity Relationship
12.
J Biol Chem ; 279(30): 31584-9, 2004 Jul 23.
Article in English | MEDLINE | ID: mdl-15155726

ABSTRACT

Carnitine acyltransferases catalyze the exchange of acyl groups between coenzyme A (CoA) and carnitine. They have important roles in many cellular processes, especially the oxidation of long-chain fatty acids, and are attractive targets for drug discovery against diabetes and obesity. These enzymes are classified based on their substrate selectivity for short-chain, medium-chain, or long-chain fatty acids. Structural information on carnitine acetyltransferase suggests that residues Met-564 and Phe-565 may be important determinants of substrate selectivity with the side chain of Met-564 located in the putative binding pocket for acyl groups. Both residues are replaced by glycine in carnitine palmitoyltransferases. To assess the functional relevance of this structural observation, we have replaced these two residues with small amino acids by mutagenesis, characterized the substrate preference of the mutants, and determined the crystal structures of two of these mutants. Kinetic studies confirm that the M564G or M564A mutation is sufficient to increase the activity of the enzyme toward medium-chain substrates with hexanoyl-CoA being the preferred substrate for the M564G mutant. The crystal structures of the M564G mutant, both alone and in complex with carnitine, reveal a deep binding pocket that can accommodate the larger acyl group. We have determined the crystal structure of the F565A mutant in a ternary complex with both the carnitine and CoA substrates at a 1.8-A resolution. The F565A mutation has minor effects on the structure or the substrate preference of the enzyme.


Subject(s)
Carnitine O-Acetyltransferase/chemistry , Carnitine O-Acetyltransferase/metabolism , Acyl Coenzyme A/metabolism , Amino Acid Sequence , Amino Acid Substitution , Animals , Carnitine O-Acetyltransferase/genetics , Catalytic Domain/genetics , Crystallography, X-Ray , In Vitro Techniques , Kinetics , Mice , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Protein Conformation , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid , Static Electricity , Substrate Specificity
13.
J Biol Chem ; 279(32): 33899-908, 2004 Aug 06.
Article in English | MEDLINE | ID: mdl-15155769

ABSTRACT

In eukaryotes, L-carnitine is involved in energy metabolism by facilitating beta-oxidation of fatty acids. Carnitine acetyltransferases (CrAT) catalyze the reversible conversion of acetyl-CoA and carnitine to acetylcarnitine and free CoA. To redesign the specificity of rat CrAT toward its substrates, we mutated Met564. The M564G mutated CrAT showed higher activity toward longer chain acyl-CoAs: activity toward myristoyl-CoA was 1250-fold higher than that of the wild-type CrAT, and lower activity toward its natural substrate, acetyl-CoA. Kinetic constants of the mutant CrAT showed modification in favor of longer acyl-CoAs as substrates. In the reverse case, mutation of the orthologous glycine (Gly553) to methionine in carnitine octanoyltransferase (COT) decreased activity toward its natural substrates, medium- and long-chain acyl-CoAs, and increased activity toward short-chain acyl-CoAs. Another CrAT mutant, M564A, was prepared and tested in the same way, with similar results. We conclude that Met564 blocks the entry of medium- and long-chain acyl-CoAs to the catalytic site of CrAT. Three-dimensional models of wild-type and mutated CrAT and COT support this hypothesis. We show for the first time that a single amino acid is able to determine the substrate specificity of CrAT and COT.


Subject(s)
Carnitine O-Acetyltransferase/genetics , Carnitine O-Acetyltransferase/metabolism , Protein Engineering , Acetyl Coenzyme A/analysis , Amino Acid Sequence , Animals , Binding Sites , Carnitine Acyltransferases/chemistry , Carnitine Acyltransferases/genetics , Carnitine Acyltransferases/metabolism , Carnitine O-Acetyltransferase/chemistry , Cloning, Molecular , Crystallization , Gene Expression , Glycine , Humans , Male , Mice , Models, Molecular , Molecular Structure , Mutagenesis, Site-Directed , RNA, Messenger/genetics , Rats , Reverse Transcriptase Polymerase Chain Reaction , Saccharomyces cerevisiae/genetics , Sequence Alignment , Structure-Activity Relationship , Substrate Specificity , Testis/chemistry
14.
J Struct Biol ; 146(3): 416-24, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15099582

ABSTRACT

We report the crystal structure of a binary complex of human peroxisomal carnitine acetyltransferase and the substrate l-carnitine, refined to a resolution of 1.8 Angstrom with an R(factor) value of 18.9% (R(free)=22.3%). L-carnitine binds to a preformed pocket in the active site tunnel of carnitine acetyltransferase aligned with His(322). The quaternary nitrogen of carnitine forms a pi-cation interaction with Phe(545), while Arg(497) forms an electrostatic interaction with the negatively charged carboxylate group. An extensive hydrogen bond network also occurs between the carboxylate group and Tyr(431), Thr(444), and a bound water molecule. Site-directed mutagenesis and kinetic characterization reveals that Tyr(431), Thr(444), Arg(497), and Phe(545) are essential for high affinity binding of L-carnitine.


Subject(s)
Carnitine O-Acetyltransferase/chemistry , Carnitine/chemistry , Mutation , Binding Sites , Carnitine/metabolism , Carnitine O-Acetyltransferase/metabolism , Crystallization , Humans , Hydrogen Bonding , Models, Molecular , Molecular Structure , Mutagenesis, Site-Directed , Protein Binding/genetics , Protein Conformation , Static Electricity
15.
Biochem J ; 379(Pt 3): 777-84, 2004 May 01.
Article in English | MEDLINE | ID: mdl-14711372

ABSTRACT

CPT I (carnitine palmitoyltransferase I) catalyses the conversion of palmitoyl-CoA into palmitoylcarnitine in the presence of L-carnitine, facilitating the entry of fatty acids into mitochondria. We propose a 3-D (three-dimensional) structural model for L-CPT I (liver CPT I), based on the similarity of this enzyme to the recently crystallized mouse carnitine acetyltransferase. The model includes 607 of the 773 amino acids of L-CPT I, and the positions of carnitine, CoA and the palmitoyl group were assigned by superposition and docking analysis. Functional analysis of this 3-D model included the mutagenesis of several amino acids in order to identify putative catalytic residues. Mutants D477A, D567A and E590D showed reduced L-CPT I activity. In addition, individual mutation of amino acids forming the conserved Ser685-Thr686-Ser687 motif abolished enzyme activity in mutants T686A and S687A and altered K(m) and the catalytic efficiency for carnitine in mutant S685A. We conclude that the catalytic residues are His473 and Asp477, while Ser687 probably stabilizes the transition state. Several conserved lysines, i.e. Lys455, Lys505, Lys560 and Lys561, were also mutated. Only mutants K455A and K560A showed decreases in activity of 50%. The model rationalizes the finding of nine natural mutations in patients with hereditary L-CPT I deficiencies.


Subject(s)
Carnitine O-Acetyltransferase/chemistry , Carnitine O-Palmitoyltransferase/chemistry , Amino Acid Sequence , Amino Acids/metabolism , Animals , Binding Sites , Blotting, Western , Carnitine/metabolism , Carnitine O-Palmitoyltransferase/deficiency , Carnitine O-Palmitoyltransferase/genetics , Carnitine O-Palmitoyltransferase/metabolism , Coenzyme A/metabolism , Crystallization , Crystallography, X-Ray , Humans , Kinetics , Mice , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Mutation, Missense/genetics , Palmitoyl Coenzyme A/metabolism , Protein Conformation , Rats , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism
17.
J Biol Chem ; 278(15): 13159-65, 2003 Apr 11.
Article in English | MEDLINE | ID: mdl-12562770

ABSTRACT

Carnitine acyltransferases are a family of ubiquitous enzymes that play a pivotal role in cellular energy metabolism. We report here the x-ray structure of human carnitine acetyltransferase to a 1.6-A resolution. This structure reveals a monomeric protein of two equally sized alpha/beta domains. Each domain is shown to have a partially similar fold to other known but oligomeric enzymes that are also involved in group-transfer reactions. The unique monomeric arrangement of the two domains constitutes a central narrow active site tunnel, indicating a likely universal feature for all members of the carnitine acyltransferase family. Superimposition of the substrate complex of a related protein, dihydrolipoyl trans-acetylase, reveals that both substrates localize to the active site tunnel of human carnitine acetyltransferase, suggesting the location of the ligand binding sites for carnitine and coenzyme A. Most significantly, this structure provides critical insights into the molecular basis for fatty acyl chain transfer and a possible common mechanism among a wide range of acyltransferases utilizing a catalytic dyad.


Subject(s)
Carnitine O-Acetyltransferase/chemistry , Carnitine O-Acetyltransferase/metabolism , Amino Acid Sequence , Crystallography, X-Ray , Fatty Acids/metabolism , Humans , Models, Molecular , Molecular Sequence Data , Protein Conformation , Protein Structure, Secondary , Sensitivity and Specificity , Sequence Alignment , Sequence Homology, Amino Acid , Software
18.
Cell ; 112(1): 113-22, 2003 Jan 10.
Article in English | MEDLINE | ID: mdl-12526798

ABSTRACT

Carnitine acyltransferases have crucial roles in the transport of fatty acids for beta-oxidation. Dysregulation of these enzymes can lead to serious diseases in humans, and they are targets for therapeutic development against diabetes. We report the crystal structures of murine carnitine acetyltransferase (CRAT), alone and in complex with its substrate carnitine or CoA. The structure contains two domains. Surprisingly, these two domains share the same backbone fold, which is also similar to that of chloramphenicol acetyltransferase and dihydrolipoyl transacetylase. The active site is located at the interface between the two domains. Carnitine and CoA are bound in deep channels in the enzyme, on opposite sides of the catalytic His343 residue. The structural information provides a molecular basis for understanding the catalysis by carnitine acyltransferases and for designing their inhibitors. Specifically, our structural information suggests that the substrate carnitine may assist the catalysis by stabilizing the oxyanion in the reaction intermediate.


Subject(s)
Carnitine O-Acetyltransferase/chemistry , Fatty Acids/metabolism , Acyltransferases/chemistry , Amino Acid Sequence , Animals , Binding Sites , Biological Transport, Active , Carnitine O-Acetyltransferase/isolation & purification , Carnitine O-Palmitoyltransferase/chemistry , Catalysis , Chloramphenicol O-Acetyltransferase/chemistry , Conserved Sequence , Crystallography, X-Ray , Escherichia coli , Humans , Mice , Models, Molecular , Molecular Sequence Data , Protein Folding , Protein Structure, Secondary , Protein Structure, Tertiary , Sequence Alignment , Substrate Specificity
19.
Acta Crystallogr D Biol Crystallogr ; 58(Pt 7): 1193-4, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12077440

ABSTRACT

In this paper, the purification, crystallization and preliminary X-ray crystallographic studies of human carnitine acetyltransferase are reported. Recombinant human carnitine acetyltransferase crystals were grown by the hanging-drop vapor-diffusion method and belong to the orthorhombic space group P2(1)2(1)2(1), with unit-cell parameters a = 137.65, b = 84.76, c = 57.65 A and one molecule per asymmetric unit. The intensity data were collected from a cryocooled crystal to 1.6 A resolution using a conventional X-ray source.


Subject(s)
Carnitine O-Acetyltransferase/chemistry , Crystallography, X-Ray/methods , Recombinant Proteins/chemistry , Diffusion , Humans
20.
Bioorg Med Chem ; 8(10): 2451-60, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11058040

ABSTRACT

An analogue 2 of coenzyme A (CoA) has been prepared in which the geminal methyl groups are replaced with hydrogens. An NMR titration study was conducted and shifts in frequency of protons in the pantetheine portion of the molecule upon titration of the adenine base were observed as has been previously reported with CoA. These studies indicate that the geminal dimethyl groups are not essential for adoption of a partially folded conformation in solution. Based on 1H-1H coupling constants, the distribution of conformations about the carbon-carbon bonds in the region of the methyl deletion were estimated. The results suggest that the conformer distribution is similar to that of CoA, but with small increases in population of the anti conformers. A simple model compound containing the didemethyl pantoamide moiety was prepared and subjected to similar conformational analysis. The coupling constants and predicted conformer distribution were almost identical to that of the CoA analogue, indicating that the conformer distribution is controlled by local interactions and not influenced by interactions between distant parts of the CoA molecule. The acetyl derivative of 2 was a fairly good substrate for the acetyl-CoA utilizing enzymes carnitine acetyltransferase, chloramphenicol acetyltransferase, and citrate synthase, with 1.3- to 10-fold increased Km values and 2.5- to 11-fold decreases in Vmax. The combined results indicate that the geminal dimethyl groups of CoA have modest effects on function and minimal effects on conformation.


Subject(s)
Coenzyme A/chemistry , Carnitine O-Acetyltransferase/chemistry , Carnitine O-Acetyltransferase/metabolism , Chloramphenicol O-Acetyltransferase/chemistry , Chloramphenicol O-Acetyltransferase/metabolism , Magnetic Resonance Spectroscopy , Molecular Structure , Protein Conformation
SELECTION OF CITATIONS
SEARCH DETAIL
...