Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Life (Basel) ; 13(8)2023 Jul 28.
Artículo en Inglés | MEDLINE | ID: mdl-37629500

RESUMEN

Short- and medium-chain acyl-CoA synthetases catalyze similar two-step reactions in which acyl substrate and ATP bind to form an enzyme-bound acyl-adenylate, then CoA binds for formation of the acyl-CoA product. We investigated the roles of active site residues in CoA binding in acetyl-CoA synthetase (Acs) and a medium-chain acyl-CoA synthetase (Macs) that uses 2-methylbutyryl-CoA. Three highly conserved residues, Arg193, Arg528, and Arg586 of Methanothermobacter thermautotrophicus Acs (AcsMt), are predicted to form important interactions with the 5'- and 3'-phosphate groups of CoA. Kinetic characterization of AcsMt variants altered at each of these positions indicates these Arg residues play a critical role in CoA binding and catalysis. The predicted CoA binding site of Methanosarcina acetivorans Macs (MacsMa) is structurally more closely related to that of 4-chlorobenzoate:coenzyme A ligase (CBAL) than Acs. Alteration of MacsMa residues Tyr460, Arg490, Tyr525, and Tyr527, which correspond to CoA binding pocket residues in CBAL, strongly affected CoA binding and catalysis without substantially affecting acyl-adenylate formation. Both enzymes discriminate between 3'-dephospho-CoA and CoA, indicating interaction between the enzyme and the 3'-phosphate group is important. Alteration of MacsMa residues Lys461 and Lys519, located at positions equivalent to AcsMt Arg528 and Arg586, respectively, had only a moderate effect on CoA binding and catalysis. Overall, our results indicate the active site architecture in AcsMt and MacsMa differs even though these enzymes catalyze mechanistically similar reactions. The significance of this study is that we have delineated the active site architecture with respect to CoA binding and catalysis in this important enzyme superfamily.

2.
Life (Basel) ; 13(7)2023 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-37511904

RESUMEN

Entamoeba histolytica is a parasitic protozoan that causes diarrheal disease in approximately 100 million people worldwide every year. E. histolytica has two forms, the growing trophozoite and the infectious cyst. Trophozoites colonizing the large intestine form cysts that are released into the environment. The ingestion of the cysts in contaminated food and water continues the disease cycle. Here, we investigated the role of glycogen in trophozoite growth and encystation. Glycogen is thought to provide precursors for the synthesis of chitin, a major component of the protective cyst wall. We propose that glycogen also serves as an energy source during metabolic adaptation to different nutrient environments. We examined the role of glycogen in E. histolytica by analyzing the growth and encystation of RNAi strains with reduced expression of the single gene-encoding glycogen synthase (GYS) or two of three genes encoding glycogen phosphorylase (PYG). The GYS RNAi strain had a greatly reduced glycogen accumulation, and both the GYS and PYG RNAi strains exhibited reduced growth in the glucose-poor medium. Both RNAi strains also showed reduced cyst production. Our results suggest glycogen synthesis and degradation are vital to the growth and adaptation of E. histolytica to a low-glucose environment such as that encountered in the large intestine.

3.
Parasitologia ; 2(2): 147-159, 2022 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-36872919

RESUMEN

The human parasite Entamoeba histolytica, which causes approximately 100 million cases of amoebic dysentery each year, relies on glycolysis as the major source of ATP production from glucose as it lacks a citric acid cycle and oxidative phosphorylation. Ethanol and acetate, the two major glycolytic end products for E. histolytica, are produced at a ratio of 2:1 under anaerobic conditions, creating an imbalance between NADH production and utilization. In this study we investigated the role of acetate kinase (ACK) in acetate production during glycolysis in E. histolytica metabolism. Analysis of intracellular and extracellular metabolites demonstrated that acetate levels were unaffected in an ACK RNAi cell line, but acetyl-CoA levels and the NAD+/NADH ratio were significantly elevated. Moreover, we demonstrated that glyceraldehyde 3-phosphate dehydrogenase catalyzes the ACK-dependent conversion of acetaldehyde to acetyl phosphate in E. histolytica. We propose that ACK is not a major contributor to acetate production, but instead provides a mechanism for maintaining the NAD+/NADH balance during ethanol production in the extended glycolytic pathway.

4.
Microorganisms ; 9(4)2021 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-33919506

RESUMEN

Entamoeba histolytica is a parasitic protozoan that causes amoebic dysentery, which affects approximately 90 million people each year worldwide. E. histolytica is transmitted through ingestion of food and water contaminated with the cyst form, which undergoes excystation in the small intestine to the trophozoite form that colonizes the large intestine. The reptile pathogen Entamoeba invadens has served as a model for studying stage conversion between the trophozoite and cyst form due to lack of reproducible encystation of E. histolytica in the laboratory. Although much has been learned about encystation and excystation using E. invadens, the findings do not fully translate to E. histolytica due to the extensive genetic and host differences between these species. Here, we present the first reproducible encystation of E. histolytica in vitro. The cysts produced were viable and displayed the four characteristic hallmarks: round shape, chitinous cell wall, tetranucleation, and detergent resistance. Using flow cytometry analysis, glucose limitation and high cell density were key for encystation, as for E. invadens. Entry into encystation was enhanced by the short-chain fatty acids acetate and propionate, unlike for E. invadens. This new model will now allow the further study of E. histolytica stage conversion, transmission, and treatment.

5.
Sci Rep ; 7(1): 5912, 2017 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-28724909

RESUMEN

Acetate kinase (ACK; E.C. 2.7.2.1), which catalyzes the interconversion of acetate and acetyl phosphate, is nearly ubiquitous in bacteria but is present only in one genus of archaea and certain eukaryotic microbes. All ACKs utilize ATP/ADP as the phosphoryl donor/acceptor in the respective directions of the reaction (acetate + ATP [Formula: see text] acetyl phosphate + ADP), with the exception of the Entamoeba histolytica ACK (EhACK) which uses pyrophosphate (PPi)/inorganic phosphate (Pi) (acetyl phosphate + Pi [Formula: see text] acetate + PPi). Structural analysis and modeling of EhACK indicated steric hindrance by active site residues constricts entry to the adenosine pocket as compared to ATP-utilizing Methanosarcina thermophila ACK (MtACK). Reciprocal alterations were made to enlarge the adenosine pocket of EhACK and reduce that of MtACK. The EhACK variants showed a step-wise increase in ADP and ATP binding but were still unable to use these as substrates, and enzymatic activity with Pi/PPi was negatively impacted. Consistent with this, ATP utilization by MtACK variants was negatively affected but the alterations were not sufficient to convert this enzyme to Pi/PPi utilization. Our results suggest that controlling access to the adenosine pocket can contribute to substrate specificity but is not the sole determinant.


Asunto(s)
Acetato Quinasa/metabolismo , Adenosina Trifosfato/farmacología , Difosfatos/farmacología , Entamoeba histolytica/enzimología , Acetato Quinasa/química , Adenosina/metabolismo , Secuencia de Aminoácidos , Sitios de Unión , Entamoeba histolytica/efectos de los fármacos , Concentración 50 Inhibidora , Cinética , Methanosarcina/enzimología , Modelos Moleculares , Especificidad por Sustrato/efectos de los fármacos
6.
FEBS Lett ; 591(4): 603-612, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28129670

RESUMEN

ADP-forming acetyl-CoA synthetase (ACD) catalyzes the interconversion of acetyl-CoA and acetate. The related succinyl-CoA synthetase follows a three-step mechanism involving a single phosphoenzyme, but a novel four-step mechanism with two phosphoenzyme intermediates was proposed for Pyrococcus ACD. Characterization of enzyme variants of Entamoeba ACD in which the two proposed phosphorylated His residues were individually altered revealed that only His252 is essential for enzymatic activity. Analysis of variants altered at two residues proposed to interact with the phosphohistidine loop that swings between distinct parts of the active site are consistent with a mechanism involving a single phosphoenzyme intermediate. Our results suggest ACDs with different subunit structures may employ slightly different mechanisms to bridge the span between active sites I and II.


Asunto(s)
Adenosina Difosfato/metabolismo , Coenzima A Ligasas/metabolismo , Entamoeba histolytica/enzimología , Proteínas Protozoarias/metabolismo , Acetatos/metabolismo , Acetilcoenzima A/metabolismo , Secuencia de Aminoácidos , Biocatálisis , Dominio Catalítico , Coenzima A Ligasas/química , Coenzima A Ligasas/genética , Entamoeba histolytica/genética , Entamoeba histolytica/metabolismo , Histidina/análogos & derivados , Histidina/química , Histidina/genética , Histidina/metabolismo , Cinética , Modelos Moleculares , Mutación Missense , Fosforilación , Dominios Proteicos , Proteínas Protozoarias/química , Proteínas Protozoarias/genética , Especificidad por Sustrato
7.
Eukaryot Cell ; 14(7): 652-60, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25956919

RESUMEN

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.


Asunto(s)
Acetilcoenzima A/metabolismo , Fosfato Acetiltransferasa/metabolismo , Phytophthora/enzimología , Catálisis , Escherichia coli/enzimología , Escherichia coli/genética , Cinética , Mutagénesis Sitio-Dirigida , Mutación/genética , Fosfato Acetiltransferasa/clasificación , Fosfato Acetiltransferasa/genética , Filogenia , Especificidad por Sustrato
8.
Life (Basel) ; 5(1): 861-71, 2015 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-25775277

RESUMEN

Acetate kinase (ACK), which catalyzes the reversible phosphorylation of acetate by ATP, is a member of the acetate and sugar kinase/heat shock cognate/actin (ASKHA) superfamily. ASKHA family members share a common core fold that includes an ATPase domain with five structural motifs. The PHOSPHATE1 motif has previously been shown to be important for catalysis. We have investigated the role of two of these motifs in the Methanosarcina thermophila ACK (MtACK) and have shown that residues projecting into the ACK active site from the PHOSPHATE2 and ADENOSINE loops and a third highly conserved loop designated here as LOOP3 play key roles in nucleotide triphosphate (NTP) selection and utilization. Alteration of Asn211 of PHOSPHATE2, Gly239 of LOOP3, and Gly331 of ADENOSINE greatly reduced catalysis. In particular, Gly331, which is highly conserved throughout the ASKHA superfamily, has the greatest effect on substrate selection. Alteration at this site strongly skewed MtACK toward utilization of purines over pyrimidines, unlike the wild type enzyme that shows broad NTP utilization. Further investigation into differences between the ATPase domain in MtACK and other acetate kinases that show different substrate preferences will provide us with a better understanding of the diversity of phosphoryl donor selection in this enzyme family.

9.
Plant Cell ; 26(11): 4499-518, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25381350

RESUMEN

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.


Asunto(s)
Acetato Quinasa/metabolismo , Acetatos/metabolismo , Chlamydomonas reinhardtii/enzimología , Cloroplastos/metabolismo , Fosfato Acetiltransferasa/metabolismo , Acetato Quinasa/genética , Proteínas Algáceas/genética , Proteínas Algáceas/metabolismo , Chlamydomonas reinhardtii/genética , Fermentación , Mitocondrias/metabolismo , Mutagénesis Insercional , Fosfato Acetiltransferasa/genética
10.
Eukaryot Cell ; 13(12): 1530-7, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25303954

RESUMEN

Entamoeba histolytica, an amitochondriate protozoan parasite that relies on glycolysis as a key pathway for ATP generation, has developed a unique extended PPi-dependent glycolytic pathway in which ADP-forming acetyl-coenzyme A (CoA) synthetase (ACD; acetate:CoA ligase [ADP-forming]; EC 6.2.1.13) converts acetyl-CoA to acetate to produce additional ATP and recycle CoA. We characterized the recombinant E. histolytica ACD and found that the enzyme is bidirectional, allowing it to potentially play a role in ATP production or in utilization of acetate. In the acetate-forming direction, acetyl-CoA was the preferred substrate and propionyl-CoA was used with lower efficiency. In the acetyl-CoA-forming direction, acetate was the preferred substrate, with a lower efficiency observed with propionate. The enzyme can utilize both ADP/ATP and GDP/GTP in the respective directions of the reaction. ATP and PPi were found to inhibit the acetate-forming direction of the reaction, with 50% inhibitory concentrations of 0.81 ± 0.17 mM (mean ± standard deviation) and 0.75 ± 0.20 mM, respectively, which are both in the range of their physiological concentrations. ATP and PPi displayed mixed inhibition versus each of the three substrates, acetyl-CoA, ADP, and phosphate. This is the first example of regulation of ACD enzymatic activity, and possible roles for this regulation are discussed.


Asunto(s)
Acetato CoA Ligasa/química , Entamoeba histolytica/enzimología , Proteínas Protozoarias/química , Acetato CoA Ligasa/antagonistas & inhibidores , Acetilcoenzima A/química , Adenosina Difosfato/química , Adenosina Trifosfato/química , Difosfatos/química , Cinética , Magnesio/química , Proteínas Protozoarias/antagonistas & inhibidores , Proteínas Recombinantes/química , Especificidad por Sustrato
11.
Eukaryot Cell ; 13(5): 657-63, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24659577

RESUMEN

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 µM AMP is capable of increasing Xfp2 activity by 24.8% ± 1.0% (mean ± standard error of the mean), while 50 µM 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.


Asunto(s)
Aldehído-Liasas/química , Cryptococcus neoformans/enzimología , Proteínas Fúngicas/química , Adenosina Trifosfato/química , Adenosina Trifosfato/metabolismo , Aldehído-Liasas/genética , Aldehído-Liasas/metabolismo , Regulación Alostérica , Cryptococcus neoformans/química , Cryptococcus neoformans/genética , Activación Enzimática , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/metabolismo , Fructosafosfatos/metabolismo , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Cinética , Pentosafosfatos/metabolismo , Especificidad por Sustrato
12.
J Struct Biol ; 181(2): 185-9, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23159802

RESUMEN

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.


Asunto(s)
Acetato Quinasa/química , Cryptococcus neoformans/enzimología , Entamoeba histolytica/enzimología , Modelos Moleculares , Conformación Proteica , Acetato Quinasa/genética , Acetatos/metabolismo , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Dominio Catalítico/genética , Cristalografía por Rayos X , Datos de Secuencia Molecular , Fosfatos/metabolismo , Especificidad de la Especie , Especificidad por Sustrato/genética
13.
Archaea ; 2012: 509579, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22973162

RESUMEN

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.


Asunto(s)
Acetato CoA Ligasa/metabolismo , Proteínas Arqueales/metabolismo , Secuencia Conservada , Methanobacteriaceae/enzimología , Acetato CoA Ligasa/genética , Adenosina/metabolismo , Adenosina Trifosfato/metabolismo , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Proteínas Arqueales/genética , Dominio Catalítico , Activación Enzimática , Pruebas de Enzimas , Escherichia coli/genética , Escherichia coli/metabolismo , Cinética , Methanobacteriaceae/genética , Mutagénesis Sitio-Dirigida , Unión Proteica , Conformación Proteica , Mapeo de Interacción de Proteínas , Ribosa/metabolismo , Alineación de Secuencia , Tirosina/metabolismo
14.
Eukaryot Cell ; 11(10): 1249-56, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22903977

RESUMEN

Acetate kinase (ACK) catalyzes the reversible synthesis of acetyl phosphate by transfer of the γ-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.


Asunto(s)
Difosfatos/metabolismo , Entamoeba histolytica/enzimología , Fosfotransferasas (aceptor de Grupo Carboxilo)/metabolismo , Proteínas Protozoarias/metabolismo , Secuencia de Aminoácidos , Sitios de Unión , Dominio Catalítico , Entamoeba histolytica/genética , Datos de Secuencia Molecular , Organofosfatos/metabolismo , Fosfotransferasas (aceptor de Grupo Carboxilo)/química , Fosfotransferasas (aceptor de Grupo Carboxilo)/genética , Proteínas Protozoarias/química , Proteínas Protozoarias/genética
15.
J Bacteriol ; 193(14): 3668-9, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21571998

RESUMEN

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.


Asunto(s)
Genoma Arqueal , Metano/metabolismo , Methanosarcinaceae/genética , Methanosarcinaceae/metabolismo , Secuencia de Bases , ADN de Archaea/genética , Methanosarcinaceae/clasificación , Datos de Secuencia Molecular , Análisis de Secuencia de ADN
16.
J Vis Exp ; (58)2011 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-22214984

RESUMEN

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).


Asunto(s)
Acetato Quinasa/análisis , Espectrofotometría/métodos , Acetato Quinasa/metabolismo , Acetatos/análisis , Acetatos/metabolismo , Compuestos Férricos/análisis , Compuestos Férricos/química , Ácidos Hidroxámicos/análisis , Ácidos Hidroxámicos/química , Organofosfatos/análisis , Organofosfatos/metabolismo
17.
J Bacteriol ; 192(22): 5982-90, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20851904

RESUMEN

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.


Asunto(s)
Acilcoenzima A/metabolismo , Proteínas Arqueales/genética , Proteínas Arqueales/metabolismo , Coenzima A Ligasas/genética , Coenzima A Ligasas/metabolismo , Methanosarcina/enzimología , Ácido Acético/metabolismo , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Sitios de Unión , Butiratos/metabolismo , Clonación Molecular , ADN Bacteriano/química , ADN Bacteriano/genética , Difosfatos/metabolismo , Expresión Génica , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Ácidos Pentanoicos/metabolismo , Propionatos , Análisis de Secuencia de ADN , Especificidad por Sustrato
18.
Proteins ; 77(3): 685-98, 2009 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-19544569

RESUMEN

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.


Asunto(s)
Coenzima A Ligasas/química , Cristalografía por Rayos X/métodos , Methanosarcina/enzimología , Secuencia de Aminoácidos , Catálisis , Dominio Catalítico , Modelos Químicos , Modelos Moleculares , Conformación Molecular , Datos de Secuencia Molecular , Unión Proteica , Homología de Secuencia de Aminoácido , Especificidad por Sustrato
19.
Archaea ; 2(2): 95-107, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17350930

RESUMEN

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.


Asunto(s)
Acetato CoA Ligasa/química , Acetato CoA Ligasa/metabolismo , Archaea/enzimología , Acetato CoA Ligasa/genética , Adenosina Monofosfato/metabolismo , Secuencia de Aminoácidos , Archaea/clasificación , Archaea/metabolismo , Methanobacteriaceae/enzimología , Methanobacteriaceae/genética , Modelos Moleculares , Datos de Secuencia Molecular , Sistemas de Lectura Abierta , Filogenia , Alineación de Secuencia , Especificidad por Sustrato
20.
Trends Microbiol ; 15(4): 150-5, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17320399

RESUMEN

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.


Asunto(s)
Acetato CoA Ligasa/metabolismo , Metano/metabolismo , Methanosarcinaceae/metabolismo , Acetato CoA Ligasa/genética , Acetatos/metabolismo , Methanosarcinaceae/clasificación , Methanosarcinaceae/genética , Modelos Biológicos , Filogenia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...