Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Plant Cell ; 26(11): 4499-518, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25381350

ABSTRACT

Chlamydomonas reinhardtii insertion mutants disrupted for genes encoding acetate kinases (EC 2.7.2.1) (ACK1 and ACK2) and a phosphate acetyltransferase (EC 2.3.1.8) (PAT2, but not PAT1) were isolated to characterize fermentative acetate production. ACK1 and PAT2 were localized to chloroplasts, while ACK2 and PAT1 were shown to be in mitochondria. Characterization of the mutants showed that PAT2 and ACK1 activity in chloroplasts plays a dominant role (relative to ACK2 and PAT1 in mitochondria) in producing acetate under dark, anoxic conditions and, surprisingly, also suggested that Chlamydomonas has other pathways that generate acetate in the absence of ACK activity. We identified a number of proteins associated with alternative pathways for acetate production that are encoded on the Chlamydomonas genome. Furthermore, we observed that only modest alterations in the accumulation of fermentative products occurred in the ack1, ack2, and ack1 ack2 mutants, which contrasts with the substantial metabolite alterations described in strains devoid of other key fermentation enzymes.


Subject(s)
Acetate Kinase/metabolism , Acetates/metabolism , Chlamydomonas reinhardtii/enzymology , Chloroplasts/metabolism , Phosphate Acetyltransferase/metabolism , Acetate Kinase/genetics , Algal Proteins/genetics , Algal Proteins/metabolism , Chlamydomonas reinhardtii/genetics , Fermentation , Mitochondria/metabolism , Mutagenesis, Insertional , Phosphate Acetyltransferase/genetics
2.
Eukaryot Cell ; 14(7): 652-60, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25956919

ABSTRACT

Phosphotransacetylase (Pta), a key enzyme in bacterial metabolism, catalyzes the reversible transfer of an acetyl group from acetyl phosphate to coenzyme A (CoA) to produce acetyl-CoA and Pi. Two classes of Pta have been identified based on the absence (Pta(I)) or presence (Pta(II)) of an N-terminal regulatory domain. Pta(I) has been fairly well studied in bacteria and one genus of archaea; however, only the Escherichia coli and Salmonella enterica Pta(II) enzymes have been biochemically characterized, and they are allosterically regulated. Here, we describe the first biochemical and kinetic characterization of a eukaryotic Pta from the oomycete Phytophthora ramorum. The two Ptas from P. ramorum, designated PrPta(II)1 and PrPta(II)2, both belong to class II. PrPta(II)1 displayed positive cooperativity for both acetyl phosphate and CoA and is allosterically regulated. We compared the effects of different metabolites on PrPta(II)1 and the S. enterica Pta(II) and found that, although the N-terminal regulatory domains share only 19% identity, both enzymes are inhibited by ATP, NADP, NADH, phosphoenolpyruvate (PEP), and pyruvate in the acetyl-CoA/Pi-forming direction but are differentially regulated by AMP. Phylogenetic analysis of bacterial, archaeal, and eukaryotic sequences identified four subtypes of Pta(II) based on the presence or absence of the P-loop and DRTGG subdomains within the N-terminal regulatory domain. Although the E. coli, S. enterica, and P. ramorum enzymes all belong to the IIa subclass, our kinetic analysis has indicated that enzymes within a subclass can still display differences in their allosteric regulation.


Subject(s)
Acetyl Coenzyme A/metabolism , Phosphate Acetyltransferase/metabolism , Phytophthora/enzymology , Catalysis , Escherichia coli/enzymology , Escherichia coli/genetics , Kinetics , Mutagenesis, Site-Directed , Mutation/genetics , Phosphate Acetyltransferase/classification , Phosphate Acetyltransferase/genetics , Phylogeny , Substrate Specificity
3.
J Bacteriol ; 197(7): 1157-63, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25605308

ABSTRACT

UNLABELLED: Xylulose 5-phosphate/fructose 6-phosphate phosphoketolase (Xfp), which catalyzes the conversion of xylulose 5-phosphate (X5P) or fructose 6-phosphate (F6P) to acetyl phosphate, plays a key role in carbohydrate metabolism in a number of bacteria. Recently, we demonstrated that the fungal Cryptococcus neoformans Xfp2 exhibits both substrate cooperativity for all substrates (X5P, F6P, and Pi) and allosteric regulation in the forms of inhibition by phosphoenolpyruvate (PEP), oxaloacetic acid (OAA), and ATP and activation by AMP (K. Glenn, C. Ingram-Smith, and K. S. Smith. Eukaryot Cell 13: 657-663, 2014). Allosteric regulation has not been reported previously for the characterized bacterial Xfps. Here, we report the discovery of substrate cooperativity and allosteric regulation among bacterial Xfps, specifically the Lactobacillus plantarum Xfp. L. plantarum Xfp is an allosteric enzyme inhibited by PEP, OAA, and glyoxylate but unaffected by the presence of ATP or AMP. Glyoxylate is an additional inhibitor to those previously reported for C. neoformans Xfp2. As with C. neoformans Xfp2, PEP and OAA share the same or possess overlapping sites on L. plantarum Xfp. Glyoxylate, which had the lowest half-maximal inhibitory concentration of the three inhibitors, binds at a separate site. This study demonstrates that substrate cooperativity and allosteric regulation may be common properties among bacterial and eukaryotic Xfp enzymes, yet important differences exist between the enzymes in these two domains. IMPORTANCE: Xylulose 5-phosphate/fructose 6-phosphate phosphoketolase (Xfp) plays a key role in carbohydrate metabolism in a number of bacteria. Although we recently demonstrated that the fungal Cryptococcus Xfp is subject to substrate cooperativity and allosteric regulation, neither phenomenon has been reported for a bacterial Xfp. Here, we report that the Lactobacillus plantarum Xfp displays substrate cooperativity and is allosterically inhibited by phosphoenolpyruvate and oxaloacetate, as is the case for Cryptococcus Xfp. The bacterial enzyme is unaffected by the presence of AMP or ATP, which act as a potent activator and inhibitor of the fungal Xfp, respectively. Our results demonstrate that substrate cooperativity and allosteric regulation may be common properties among bacterial and eukaryotic Xfps, yet important differences exist between the enzymes in these two domains.


Subject(s)
Aldehyde-Lyases/metabolism , Gene Expression Regulation, Bacterial/physiology , Gene Expression Regulation, Enzymologic/physiology , Lactobacillus plantarum/enzymology , Pentosephosphates/metabolism , Adenosine Monophosphate/metabolism , Adenosine Triphosphate/metabolism , Aldehyde-Lyases/antagonists & inhibitors , Aldehyde-Lyases/genetics , Gene Expression Regulation, Bacterial/drug effects , Gene Expression Regulation, Enzymologic/drug effects , Glyoxylates/pharmacology , Hydrogen-Ion Concentration , Oxaloacetic Acid/pharmacology , Phosphoenolpyruvate/pharmacology
4.
Eukaryot Cell ; 13(5): 657-63, 2014 May.
Article in English | MEDLINE | ID: mdl-24659577

ABSTRACT

Xylulose 5-phosphate/fructose 6-phosphate phosphoketolase (Xfp), previously thought to be present only in bacteria but recently found in fungi, catalyzes the formation of acetyl phosphate from xylulose 5-phosphate or fructose 6-phosphate. Here, we describe the first biochemical and kinetic characterization of a eukaryotic Xfp, from the opportunistic fungal pathogen Cryptococcus neoformans, which has two XFP genes (designated XFP1 and XFP2). Our kinetic characterization of C. neoformans Xfp2 indicated the existence of both substrate cooperativity for all three substrates and allosteric regulation through the binding of effector molecules at sites separate from the active site. Prior to this study, Xfp enzymes from two bacterial genera had been characterized and were determined to follow Michaelis-Menten kinetics. C. neoformans Xfp2 is inhibited by ATP, phosphoenolpyruvate (PEP), and oxaloacetic acid (OAA) and activated by AMP. ATP is the strongest inhibitor, with a half-maximal inhibitory concentration (IC50) of 0.6 mM. PEP and OAA were found to share the same or have overlapping allosteric binding sites, while ATP binds at a separate site. AMP acts as a very potent activator; as little as 20 ĀµM AMP is capable of increasing Xfp2 activity by 24.8% Ā± 1.0% (mean Ā± standard error of the mean), while 50 ĀµM prevented inhibition caused by 0.6 mM ATP. AMP and PEP/OAA operated independently, with AMP activating Xfp2 and PEP/OAA inhibiting the activated enzyme. This study provides valuable insight into the metabolic role of Xfp within fungi, specifically the fungal pathogen Cryptococcus neoformans, and suggests that at least some Xfps display substrate cooperative binding and allosteric regulation.


Subject(s)
Aldehyde-Lyases/chemistry , Cryptococcus neoformans/enzymology , Fungal Proteins/chemistry , Adenosine Triphosphate/chemistry , Adenosine Triphosphate/metabolism , Aldehyde-Lyases/genetics , Aldehyde-Lyases/metabolism , Allosteric Regulation , Cryptococcus neoformans/chemistry , Cryptococcus neoformans/genetics , Enzyme Activation , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Fructosephosphates/metabolism , Fungal Proteins/genetics , Fungal Proteins/metabolism , Kinetics , Pentosephosphates/metabolism , Substrate Specificity
5.
J Struct Biol ; 181(2): 185-9, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23159802

ABSTRACT

Acetate kinases (ACKs) are members of the acetate and sugar kinase/hsp70/actin (ASKHA) superfamily and catalyze the reversible phosphorylation of acetate, with ADP/ATP the most common phosphoryl acceptor/donor. While prokaryotic ACKs have been the subject of extensive biochemical and structural characterization, there is a comparative paucity of information on eukaryotic ACKs, and prior to this report, no structure of an ACK of eukaryotic origin was available. We determined the structures of ACKs from the eukaryotic pathogens Entamoeba histolytica and Cryptococcus neoformans. Each active site is located at an interdomain interface, and the acetate and phosphate binding pockets display sequence and structural conservation with their prokaryotic counterparts. Interestingly, the E. histolytica ACK has previously been shown to be pyrophosphate (PP(i))-dependent, and is the first ACK demonstrated to have this property. Examination of its structure demonstrates how subtle amino acid substitutions within the active site have converted cosubstrate specificity from ATP to PP(i) while retaining a similar backbone conformation. Differences in the angle between domains surrounding the active site suggest that interdomain movement may accompany catalysis. Taken together, these structures are consistent with the eukaryotic ACKs following a similar reaction mechanism as is proposed for the prokaryotic homologs.


Subject(s)
Acetate Kinase/chemistry , Cryptococcus neoformans/enzymology , Entamoeba histolytica/enzymology , Models, Molecular , Protein Conformation , Acetate Kinase/genetics , Acetates/metabolism , Amino Acid Sequence , Amino Acid Substitution , Catalytic Domain/genetics , Crystallography, X-Ray , Molecular Sequence Data , Phosphates/metabolism , Species Specificity , Substrate Specificity/genetics
6.
Eukaryot Cell ; 11(10): 1249-56, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22903977

ABSTRACT

Acetate kinase (ACK) catalyzes the reversible synthesis of acetyl phosphate by transfer of the ƎĀ³-phosphate of ATP to acetate. Here we report the first biochemical and kinetic characterization of a eukaryotic ACK, that from the protist Entamoeba histolytica. Our characterization revealed that this protist ACK is the only known member of the ASKHA structural superfamily, which includes acetate kinase, hexokinase, and other sugar kinases, to utilize inorganic pyrophosphate (PP(i))/inorganic phosphate (P(i)) as the sole phosphoryl donor/acceptor. Detection of ACK activity in E. histolytica cell extracts in the direction of acetate/PP(i) formation but not in the direction of acetyl phosphate/P(i) formation suggests that the physiological direction of the reaction is toward acetate/PP(i) production. Kinetic parameters determined for each direction of the reaction are consistent with this observation. The E. histolytica PP(i)-forming ACK follows a sequential mechanism, supporting a direct in-line phosphoryl transfer mechanism as previously reported for the well-characterized Methanosarcina thermophila ATP-dependent ACK. Characterizations of enzyme variants altered in the putative acetate/acetyl phosphate binding pocket suggested that acetyl phosphate binding is not mediated solely through a hydrophobic interaction but also through the phosphoryl group, as for the M. thermophila ACK. However, there are key differences in the roles of certain active site residues between the two enzymes. The absence of known ACK partner enzymes raises the possibility that ACK is part of a novel pathway in Entamoeba.


Subject(s)
Diphosphates/metabolism , Entamoeba histolytica/enzymology , Phosphotransferases (Carboxyl Group Acceptor)/metabolism , Protozoan Proteins/metabolism , Amino Acid Sequence , Binding Sites , Catalytic Domain , Entamoeba histolytica/genetics , Molecular Sequence Data , Organophosphates/metabolism , Phosphotransferases (Carboxyl Group Acceptor)/chemistry , Phosphotransferases (Carboxyl Group Acceptor)/genetics , Protozoan Proteins/chemistry , Protozoan Proteins/genetics
7.
Archaea ; 2012: 509579, 2012.
Article in English | MEDLINE | ID: mdl-22973162

ABSTRACT

The acyl-adenylate-forming enzyme superfamily, consisting of acyl- and aryl-CoA synthetases, the adenylation domain of the nonribosomal peptide synthetases, and luciferase, has three signature motifs (I-III) and ten conserved core motifs (A1-A10), some of which overlap the signature motifs. The consensus sequence for signature motif III (core motif A7) in acetyl-CoA synthetase is Y-X-S/T/A-G-D, with an invariant fifth position, highly conserved first and fourth positions, and variable second and third positions. Kinetic studies of enzyme variants revealed that an alteration at any position resulted in a strong decrease in the catalytic rate, although the most deleterious effects were observed when the first or fifth positions were changed. Structural modeling suggests that the highly conserved Tyr in the first position plays a key role in active site architecture through interaction with a highly conserved active-site Gln, and the invariant Asp in the fifth position plays a critical role in ATP binding and catalysis through interaction with the 2'- and 3'-OH groups of the ribose moiety. Interactions between these Asp and ATP are observed in all structures available for members of the superfamily, consistent with a critical role in substrate binding and catalysis for this invariant residue.


Subject(s)
Acetate-CoA Ligase/metabolism , Archaeal Proteins/metabolism , Conserved Sequence , Methanobacteriaceae/enzymology , Acetate-CoA Ligase/genetics , Adenosine/metabolism , Adenosine Triphosphate/metabolism , Amino Acid Motifs , Amino Acid Sequence , Archaeal Proteins/genetics , Catalytic Domain , Enzyme Activation , Enzyme Assays , Escherichia coli/genetics , Escherichia coli/metabolism , Kinetics , Methanobacteriaceae/genetics , Mutagenesis, Site-Directed , Protein Binding , Protein Conformation , Protein Interaction Mapping , Ribose/metabolism , Sequence Alignment , Tyrosine/metabolism
8.
J Bacteriol ; 193(14): 3668-9, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21571998

ABSTRACT

The genome sequence of the aceticlastic methanoarchaeon Methanosaeta concilii GP6, comprised of a 3,008,626-bp chromosome and an 18,019-bp episome, has been determined and exhibits considerable differences in gene content from that of Methanosaeta thermophila.


Subject(s)
Genome, Archaeal , Methane/metabolism , Methanosarcinaceae/genetics , Methanosarcinaceae/metabolism , Base Sequence , DNA, Archaeal/genetics , Methanosarcinaceae/classification , Molecular Sequence Data , Sequence Analysis, DNA
9.
J Bacteriol ; 192(22): 5982-90, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20851904

ABSTRACT

Short- and medium-chain acyl coenzyme A (acyl-CoA) synthetases catalyze the formation of acyl-CoA from an acyl substrate, ATP, and CoA. These enzymes catalyze mechanistically similar two-step reactions that proceed through an enzyme-bound acyl-AMP intermediate. Here we describe the characterization of a member of this enzyme family from the methane-producing archaeon Methanosarcina acetivorans. This enzyme, a medium-chain acyl-CoA synthetase designated Macs(Ma), utilizes 2-methylbutyrate as its preferred substrate for acyl-CoA synthesis but cannot utilize acetate and thus cannot catalyze the first step of acetoclastic methanogenesis in M. acetivorans. When propionate or other less favorable acyl substrates, such as butyrate, 2-methylpropionate, or 2-methylvalerate, were utilized, the acyl-CoA was not produced or was produced at reduced levels. Instead, acyl-AMP and PP(i) were released in the absence of CoA, whereas in the presence of CoA, the intermediate was broken down into AMP and the acyl substrate, which were released along with PP(i). These results suggest that although acyl-CoA synthetases may have the ability to utilize a broad range of substrates for the acyl-adenylate-forming first step of the reaction, the intermediate may not be suitable for the thioester-forming second step. The Macs(Ma) structure has revealed the putative acyl substrate- and CoA-binding pockets. Six residues proposed to form the acyl substrate-binding pocket, Lys(256), Cys(298), Gly(351), Trp(259), Trp(237), and Trp(254), were targeted for alteration. Characterization of the enzyme variants indicates that these six residues are critical in acyl substrate binding and catalysis, and even conservative alterations significantly reduced the catalytic ability of the enzyme.


Subject(s)
Acyl Coenzyme A/metabolism , Archaeal Proteins/genetics , Archaeal Proteins/metabolism , Coenzyme A Ligases/genetics , Coenzyme A Ligases/metabolism , Methanosarcina/enzymology , Acetic Acid/metabolism , Amino Acid Sequence , Amino Acid Substitution , Binding Sites , Butyrates/metabolism , Cloning, Molecular , DNA, Bacterial/chemistry , DNA, Bacterial/genetics , Diphosphates/metabolism , Gene Expression , Molecular Sequence Data , Mutagenesis, Site-Directed , Pentanoic Acids/metabolism , Propionates , Sequence Analysis, DNA , Substrate Specificity
10.
Proteins ; 77(3): 685-98, 2009 Nov 15.
Article in English | MEDLINE | ID: mdl-19544569

ABSTRACT

The acyl-AMP forming family of adenylating enzymes catalyze two-step reactions to activate a carboxylate with the chemical energy derived from ATP hydrolysis. X-ray crystal structures have been determined for multiple members of this family and, together with biochemical studies, provide insights into the active site and catalytic mechanisms used by these enzymes. These studies have shown that the enzymes use a domain rotation of 140 degrees to reconfigure a single active site to catalyze the two partial reactions. We present here the crystal structure of a new medium chain acyl-CoA synthetase from Methanosarcina acetivorans. The binding pocket for the three substrates is analyzed, with many conserved residues present in the AMP binding pocket. The CoA binding pocket is compared to the pockets of both acetyl-CoA synthetase and 4-chlorobenzoate:CoA ligase. Most interestingly, the acyl-binding pocket of the new structure is compared with other acyl- and aryl-CoA synthetases. A comparison of the acyl-binding pocket of the acyl-CoA synthetase from M. acetivorans with other structures identifies a shallow pocket that is used to bind the medium chain carboxylates. These insights emphasize the high sequence and structural diversity among this family in the area of the acyl-binding pocket.


Subject(s)
Coenzyme A Ligases/chemistry , Crystallography, X-Ray/methods , Methanosarcina/enzymology , Amino Acid Sequence , Catalysis , Catalytic Domain , Models, Chemical , Models, Molecular , Molecular Conformation , Molecular Sequence Data , Protein Binding , Sequence Homology, Amino Acid , Substrate Specificity
11.
Trends Microbiol ; 15(4): 150-5, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17320399

ABSTRACT

Although the aceticlastic methanoarchaea Methanosarcina and Methanosaeta employ different enzymes to catalyze the first step of aceticlastic methanogenesis, it has long been assumed that the remainder of the pathway was the same. Analysis of the recently completed genome sequence of Methanosaeta thermophila confirms that the majority of core steps of the pathway are similar in both genera, but striking differences have been discovered in electron transfer and energy conservation. In addition, the presence of genes encoding enzymes for the CO(2) reduction pathway in the Msa. thermophila genome suggests the possibility that Methanosaeta might be more metabolically diverse than previously thought. Thus, genome analysis of Msa. thermophila presents new research avenues for this forgotten methanogen and reminds us of the questions that still remain unanswered about aceticlastic methanogenesis in both Methanosaeta and Methanosarcina.


Subject(s)
Acetate-CoA Ligase/metabolism , Methane/metabolism , Methanosarcinaceae/metabolism , Acetate-CoA Ligase/genetics , Acetates/metabolism , Methanosarcinaceae/classification , Methanosarcinaceae/genetics , Models, Biological , Phylogeny
12.
Biochem Biophys Res Commun ; 365(3): 420-5, 2008 Jan 18.
Article in English | MEDLINE | ID: mdl-17996732

ABSTRACT

Trypanosoma brucei harbors two hexokinases (TbHK1 and TbHK2) that are 98% identical at the amino acid level. We previously found that recombinant TbHK1 (rTbHK1) has hexokinase activity, while rTbHK2 has not, a finding attributed to differences in the C-termini of the proteins. Sequence analysis suggests that the C-termini of TbHKs are part of a newly identified conserved motif found in other eukaryotic hexokinases. Here, we have explored the role of tail residues in the differences in catalytic activity between TbHK1 and TbHK2. Our studies reveal that tail residues D454, F462, M466, and N469 are essential for HK activity while both I458 and V468 are required for catalysis and substrate specificity. To activate rTbHK2, all of the residues important for activity in rTbHK1 (D454, V458, F462, M466, V468, and N469) were required. These results indicate that the overall structure of the C-terminal tail influences the HK activity of rTbHK1.


Subject(s)
Fructose/chemistry , Glucose/chemistry , Hexokinase/chemistry , Trypanosoma brucei brucei/enzymology , Actins/chemistry , Adenosine Triphosphate/chemistry , Amino Acid Motifs , Animals , Catalysis , Hexokinase/genetics , Mutagenesis, Site-Directed , Protein Folding , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Substrate Specificity
13.
Trends Microbiol ; 14(6): 249-53, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16678422

ABSTRACT

The bacterial enzymes acetate kinase (AK) and phosphotransacetylase (PTA) form a key pathway for synthesis of the central metabolic intermediate acetyl coenzyme A (acetyl-CoA) from acetate or for generation of ATP from excess acetyl-CoA. Putative AK genes have now been identified in some eukaryotic microbes. In Chlamydomonas reinhardtii and Phytophthora species, AK forms a pathway with PTA. AK has also been identified in non-yeast fungi but these fungi do not have PTA. Instead, AK forms a pathway with D-xylulose 5-phosphate phosphoketolase (XFP), a pathway that was also previously found only in bacteria. In Entamoeba histolytica, neither PTA nor XFP was found as a partner for AK. Thus, eukaryotic microbes seem to have incorporated the 'bacterial' enzyme AK into at least three different metabolic pathways.


Subject(s)
Acetate Kinase/genetics , Eukaryotic Cells/enzymology , Sequence Homology, Amino Acid , Acetate Kinase/metabolism , Algal Proteins/genetics , Algal Proteins/metabolism , Amoeba/genetics , Amoeba/metabolism , Animals , Fungal Proteins/genetics , Fungal Proteins/metabolism , Phosphate Acetyltransferase/genetics , Phosphate Acetyltransferase/metabolism , Phylogeny
14.
J Vis Exp ; (58)2011 Dec 19.
Article in English | MEDLINE | ID: mdl-22214984

ABSTRACT

Acetate kinase, a member of the acetate and sugar kinase-Hsp70-actin (ASKHA) enzyme superfamily, is responsible for the reversible phosphorylation of acetate to acetyl phosphate utilizing ATP as a substrate. Acetate kinases are ubiquitous in the Bacteria, found in one genus of Archaea, and are also present in microbes of the Eukarya. The most well characterized acetate kinase is that from the methane-producing archaeon Methanosarcina thermophila. An acetate kinase which can only utilize PP(i) but not ATP in the acetyl phosphate-forming direction has been isolated from Entamoeba histolytica, the causative agent of amoebic dysentery, and has thus far only been found in this genus. In the direction of acetyl phosphate formation, acetate kinase activity is typically measured using the hydroxamate assay, first described by Lipmann, a coupled assay in which conversion of ATP to ADP is coupled to oxidation of NADH to NAD(+) by the enzymes pyruvate kinase and lactate dehydrogenase, or an assay measuring release of inorganic phosphate after reaction of the acetyl phosphate product with hydroxylamine. Activity in the opposite, acetate-forming direction is measured by coupling ATP formation from ADP to the reduction of NADP(+) to NADPH by the enzymes hexokinase and glucose 6-phosphate dehydrogenase. Here we describe a method for the detection of acetate kinase activity in the direction of acetate formation that does not require coupling enzymes, but is instead based on direct determination of acetyl phosphate consumption. After the enzymatic reaction, remaining acetyl phosphate is converted to a ferric hydroxamate complex that can be measured spectrophotometrically, as for the hydroxamate assay. Thus, unlike the standard coupled assay for this direction that is dependent on the production of ATP from ADP, this direct assay can be used for acetate kinases that produce ATP or PP(i).


Subject(s)
Acetate Kinase/analysis , Spectrophotometry/methods , Acetate Kinase/metabolism , Acetates/analysis , Acetates/metabolism , Ferric Compounds/analysis , Ferric Compounds/chemistry , Hydroxamic Acids/analysis , Hydroxamic Acids/chemistry , Organophosphates/analysis , Organophosphates/metabolism
15.
Archaea ; 2(2): 95-107, 2007 May.
Article in English | MEDLINE | ID: mdl-17350930

ABSTRACT

Adenosine monophosphate (AMP)-forming acetyl-CoA synthetase (ACS; acetate:CoA ligase (AMP-forming), EC 6.2.1.1) is a key enzyme for conversion of acetate to acetyl-CoA, an essential intermediate at the junction of anabolic and catabolic pathways. Phylogenetic analysis of putative short and medium chain acyl-CoA synthetase sequences indicates that the ACSs form a distinct clade from other acyl-CoA synthetases. Within this clade, the archaeal ACSs are not monophyletic and fall into three groups composed of both bacterial and archaeal sequences. Kinetic analysis of two archaeal enzymes, an ACS from Methanothermobacter thermautotrophicus (designated as MT-ACS1) and an ACS from Archaeoglobus fulgidus (designated as AF-ACS2), revealed that these enzymes have very different properties. MT-ACS1 has nearly 11-fold higher affinity and 14-fold higher catalytic efficiency with acetate than with propionate, a property shared by most ACSs. However, AF-ACS2 has only 2.3-fold higher affinity and catalytic efficiency with acetate than with propionate. This enzyme has an affinity for propionate that is almost identical to that of MT-ACS1 for acetate and nearly tenfold higher than the affinity of MT-ACS1 for propionate. Furthermore, MT-ACS1 is limited to acetate and propionate as acyl substrates, whereas AF-ACS2 can also utilize longer straight and branched chain acyl substrates. Phylogenetic analysis, sequence alignment and structural modeling suggest a molecular basis for the altered substrate preference and expanded substrate range of AF-ACS2 versus MT-ACS1.


Subject(s)
Acetate-CoA Ligase/chemistry , Acetate-CoA Ligase/metabolism , Archaea/enzymology , Acetate-CoA Ligase/genetics , Adenosine Monophosphate/metabolism , Amino Acid Sequence , Archaea/classification , Archaea/metabolism , Methanobacteriaceae/enzymology , Methanobacteriaceae/genetics , Models, Molecular , Molecular Sequence Data , Open Reading Frames , Phylogeny , Sequence Alignment , Substrate Specificity
16.
Biochemistry ; 45(38): 11482-90, 2006 Sep 26.
Article in English | MEDLINE | ID: mdl-16981708

ABSTRACT

AMP-forming acetyl-CoA synthetase [ACS; acetate:CoA ligase (AMP-forming), EC 6.2.1.1] catalyzes the activation of acetate to acetyl-CoA in a two-step reaction. This enzyme is a member of the adenylate-forming enzyme superfamily that includes firefly luciferase, nonribosomal peptide synthetases, and acyl- and aryl-CoA synthetases/ligases. Although the structures of several superfamily members demonstrate that these enzymes have a similar fold and domain structure, the low sequence conservation and diversity of the substrates utilized have limited the utility of these structures in understanding substrate binding in more distantly related enzymes in this superfamily. The crystal structures of the Salmonella enterica ACS and Saccharomyces cerevisiae ACS1 have allowed a directed approach to investigating substrate binding and catalysis in ACS. In the S. enterica ACS structure, the propyl group of adenosine 5'-propylphosphate, which mimics the acyl-adenylate intermediate, lies in a hydrophobic pocket. Modeling of the Methanothermobacter thermautotrophicus Z245 ACS (MT-ACS1) on the S. cerevisiae ACS structure showed similar active site architecture, and alignment of the amino acid sequences of proven ACSs indicates that the four residues that compose the putative acetate binding pocket are well conserved. These four residues, Ile312, Thr313, Val388, and Trp416 of MT-ACS1, were targeted for alteration, and our results support that they do indeed form the acetate binding pocket and that alterations at these positions significantly alter the enzyme's affinity for acetate as well as the range of acyl substrates that can be utilized. In particular, Trp416 appears to be the primary determinant for acyl chain length that can be accommodated in the binding site.


Subject(s)
Acetate-CoA Ligase/metabolism , Acetates/metabolism , Methanobacteriaceae/enzymology , Acetate-CoA Ligase/chemistry , Acetate-CoA Ligase/isolation & purification , Adenosine Triphosphate/metabolism , Amino Acid Sequence , Binding Sites , Coenzyme A/metabolism , Isoleucine/metabolism , Kinetics , Models, Molecular , Molecular Sequence Data , Mutant Proteins/isolation & purification , Mutant Proteins/metabolism , Saccharomyces cerevisiae/enzymology , Salmonella enterica/enzymology , Sequence Alignment , Substrate Specificity , Threonine/metabolism , Tryptophan/metabolism , Valine/metabolism
17.
Eukaryot Cell ; 5(12): 2014-23, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17028241

ABSTRACT

Trypanosoma brucei expresses two hexokinases that are 98% identical, namely, TbHK1 and TbHK2. Homozygous null TbHK2-/- procyclic-form parasites exhibit an increased doubling time, a change in cell morphology, and, surprisingly, a twofold increase in cellular hexokinase activity. Recombinant TbHK1 enzymatic activity is similar to that of other hexokinases, with apparent Km values for glucose and ATP of 0.09 +/- 0.02 mM and 0.28 +/- 0.1 mM, respectively. The k(cat) value for TbHK1 is 2.9 x 10(4) min(-1). TbHK1 can use mannose, fructose, 2-deoxyglucose, and glucosamine as substrates. In addition, TbHK1 is inhibited by fatty acids, with lauric, myristic, and palmitic acids being the most potent (with 50% inhibitory concentrations of 75.8, 78.4, and 62.4 microM, respectively). In contrast to TbHK1, recombinant TbHK2 lacks detectable enzymatic activity. Seven of the 10 amino acid differences between TbHK1 and TbHK2 lie within the C-terminal 18 amino acids of the polypeptides. Modeling of the proteins maps the C-terminal tails near the interdomain cleft of the enzyme that participates in the conformational change of the enzyme upon substrate binding. Replacing the last 18 amino acids of TbHK2 with the corresponding residues of TbHK1 yields an active recombinant protein with kinetic properties similar to those of TbHK1. Conversely, replacing the C-terminal tail of TbHK1 with the TbHK2 tail inactivates the enzyme. These findings suggest that the C-terminal tail of TbHK1 is important for hexokinase activity. The altered C-terminal tail of TbHK2, along with the phenotype of the knockout parasites, suggests a distinct function for the protein.


Subject(s)
Hexokinase/chemistry , Hexokinase/metabolism , Trypanosoma brucei brucei/enzymology , Amino Acid Sequence , Animals , Base Sequence , DNA, Protozoan/genetics , Gene Deletion , Genes, Protozoan , Hexokinase/genetics , Isoenzymes/chemistry , Isoenzymes/genetics , Isoenzymes/metabolism , Kinetics , Microbodies/enzymology , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid , Trypanosoma brucei brucei/genetics , Trypanosoma brucei brucei/growth & development
18.
J Bacteriol ; 184(15): 4240-5, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12107142

ABSTRACT

The roles of an aspartate and an arginine, which are completely conserved in the active sites of beta-class carbonic anhydrases, were investigated by steady-state kinetic analyses of replacement variants of the beta-class enzyme (Cab) from the archaeon Methanobacterium thermoautotrophicum. Previous kinetic analyses of wild-type Cab indicated a two-step zinc-hydroxide mechanism of catalysis in which the k(cat)/K(m) value depends only on the rate constants for the CO(2) hydration step, whereas k(cat) also depends on rate constants from the proton transfer step (K. S. Smith, N. J. Cosper, C. Stalhandske, R. A. Scott, and J. G. Ferry, J. Bacteriol. 182:6605-6613, 2000). The recently solved crystal structure of Cab shows the presence of a buffer molecule within hydrogen bonding distance of Asp-34, implying a role for this residue in the proton transport step (P. Strop, K. S. Smith, T. M. Iverson, J. G. Ferry, and D. C. Rees, J. Biol. Chem. 276:10299-10305, 2001). The k(cat)/K(m) values of Asp-34 variants were decreased relative to those of the wild type, although not to an extent which supports an essential role for this residue in the CO(2) hydration step. Parallel decreases in k(cat) and k(cat)/K(m) values for the variants precluded any conclusions regarding a role for Asp-34 in the proton transfer step; however, the k(cat) of the D34A variant was chemically rescued by replacement of 2-(N-morpholino)propanesulfonic acid buffer with imidazole at pH 7.2, supporting a role for the conserved aspartate in the proton transfer step. The crystal structure of Cab also shows Arg-36 with two hydrogen bonds to Asp-34. Arg-36 variants had both k(cat) and k(cat)/K(m) values that were decreased at least 250-fold relative to those of the wild type, establishing an essential function for this residue. Imidazole was unable to rescue the k(cat) of the R36A variant; however, partial rescue of the kinetic parameter was obtained with guanidine-HCl indicating that the guanido group of this residue is important.


Subject(s)
Arginine/metabolism , Aspartic Acid/metabolism , Carbonic Anhydrases/metabolism , Methanobacterium/enzymology , Arginine/genetics , Aspartic Acid/genetics , Binding Sites , Carbonic Anhydrases/chemistry , Carbonic Anhydrases/genetics , Kinetics , Models, Molecular
SELECTION OF CITATIONS
SEARCH DETAIL