Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
J Biol Chem ; 299(2): 102789, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36509144

RESUMO

α-Isopropylmalate synthase (IPMS) catalyzes the first step in leucine (Leu) biosynthesis and is allosterically regulated by the pathway end product, Leu. IPMS is a dimeric enzyme with each chain consisting of catalytic, accessory, and regulatory domains, with the accessory and regulatory domains of each chain sitting adjacent to the catalytic domain of the other chain. The IPMS crystal structure shows significant asymmetry because of different relative domain conformations in each chain. Owing to the challenges posed by the dynamic and asymmetric structures of IPMS enzymes, the molecular details of their catalytic and allosteric mechanisms are not fully understood. In this study, we have investigated the allosteric feedback mechanism of the IPMS enzyme from the bacterium that causes meningitis, Neisseria meningitidis (NmeIPMS). By combining molecular dynamics simulations with small-angle X-ray scattering, mutagenesis, and heterodimer generation, we demonstrate that Leu-bound NmeIPMS is in a rigid conformational state stabilized by asymmetric interdomain polar interactions. Furthermore, we found removing these polar interactions by mutagenesis impaired the allosteric response without compromising Leu binding. Our results suggest that the allosteric inhibition of NmeIPMS is achieved by restricting the flexibility of the accessory and regulatory domains, demonstrating that significant conformational flexibility is required for catalysis.


Assuntos
2-Isopropilmalato Sintase , Biocatálise , Leucina , Neisseria meningitidis , Domínios Proteicos , 2-Isopropilmalato Sintase/química , 2-Isopropilmalato Sintase/genética , 2-Isopropilmalato Sintase/metabolismo , Regulação Alostérica , Domínio Catalítico , Leucina/biossíntese , Leucina/química , Leucina/metabolismo , Neisseria meningitidis/enzimologia , Neisseria meningitidis/metabolismo , Simulação de Dinâmica Molecular , Espalhamento a Baixo Ângulo , Difração de Raios X , Multimerização Proteica , Mutagênese , Maleabilidade
2.
New Phytol ; 235(3): 1129-1145, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35485988

RESUMO

We show here that the side chain of pogostone, one of the major components of patchouli oil obtained from Pogostemon cablin and possessing a variety of pharmacological activities, is derived from 4-methylvaleric acid. We also show that 4-methylvaleric acid is produced through the one-carbon α-ketoacid elongation pathway with the involvement of the key enzyme 2-isobutylmalate synthase (IBMS), a newly identified enzyme related to isopropylmalate synthase (IPMS) of leucine (Leu) biosynthesis. Site-directed mutagenesis identified Met132 in the N-terminal catalytic region as affecting the substrate specificity of PcIBMS1. Even though PcIBMS1 possesses the C-terminal domain that in IPMS serves to mediate Leu inhibition, it is insensitive to Leu. The observation of the evolution of IBMS from IPMS, as well as previously reported examples of IPMS-related genes involved in making glucosinolates in Brassicaceae, acylsugars in Solanaceae, and flavour compounds in apple, indicate that IPMS genes represent an important pool for the independent evolution of genes for specialised metabolism.


Assuntos
2-Isopropilmalato Sintase , Óleos Voláteis , 2-Isopropilmalato Sintase/química , 2-Isopropilmalato Sintase/genética , 2-Isopropilmalato Sintase/metabolismo , Cinética , Leucina/metabolismo
3.
FEMS Yeast Res ; 22(1)2022 04 08.
Artigo em Inglês | MEDLINE | ID: mdl-35266531

RESUMO

The first committed step in the leucine biosynthetic pathway is catalyzed by α-isopropylmalate synthase (α-IPMS, EC 2.3.3.13), which in the Saccaromycotina subphylum of Ascomycete yeasts is frequently encoded by duplicated genes. Following a gene duplication event, the two copies may be preserved presumably because the encoded proteins diverge in either functional properties and/or cellular localization. The genome of the petite-negative budding yeast Lachancea kluyveri includes two SAKL0E10472 (LkLEU4) and SAKL0F05170 g (LkLEU4BIS) paralogous genes, which are homologous to other yeast α-IPMS sequences. Here, we investigate whether these paralogous genes encode functional α-IPMS isozymes and whether their functions have diverged. Molecular phylogeny suggested that the LkLeu4 isozyme is located in the mitochondria and LkLeu4BIS in the cytosol. Comparison of growth rates, leucine intracellular pools and mRNA levels, indicate that the LkLeu4 isozyme is the predominant α-IPMS enzyme during growth on glucose as carbon source. Determination of the kinetic parameters indicates that the isozymes have similar affinities for the substrates and for the feedback inhibitor leucine. Thus, the diversification of the physiological roles of the genes LkLEU4 and LkLEU4BIS involves preferential transcription of the LkLEU4 gene during growth on glucose and different subcellular localization, although ligand interactions have not diverged.


Assuntos
2-Isopropilmalato Sintase , Saccharomycetales , 2-Isopropilmalato Sintase/química , 2-Isopropilmalato Sintase/genética , 2-Isopropilmalato Sintase/metabolismo , Glucose/metabolismo , Isoenzimas/genética , Isoenzimas/metabolismo , Leucina/metabolismo , Saccharomyces cerevisiae/metabolismo , Saccharomycetales/metabolismo
4.
Extremophiles ; 23(4): 377-388, 2019 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-30919057

RESUMO

Protein lysine Nε-acetylation is one of the important factors regulating cellular metabolism. We performed a proteomic analysis to identify acetylated proteins in the extremely thermophilic bacterium, Thermus thermophilus HB27. A total of 335 unique acetylated lysine residues, including many metabolic enzymes and ribosomal proteins, were identified in 208 proteins. Enzymes involved in amino acid metabolism were the most abundant among acetylated metabolic proteins. 2-Isopropylmalate synthase (IPMS), which catalyzes the first step in leucine biosynthesis, was acetylated at four lysine residues. Acetylation-mimicking mutations at Lys332 markedly decreased IPMS activity in vitro, suggesting that Lys332, which is located in subdomain II, plays a regulatory role in IPMS activity. We also investigated the acetylation-deacetylation mechanism of IPMS and revealed that it was acetylated non-enzymatically by acetyl-CoA and deacetylated enzymatically by TT_C0104. The present results suggest that leucine biosynthesis is regulated by post-translational protein modifications, in addition to feedback inhibition/repression, and that metabolic enzymes are regulated by protein acetylation in T. thermophilus.


Assuntos
2-Isopropilmalato Sintase/metabolismo , Proteínas de Bactérias/metabolismo , Processamento de Proteína Pós-Traducional , Thermus thermophilus/enzimologia , 2-Isopropilmalato Sintase/química , 2-Isopropilmalato Sintase/genética , Acetilação , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Leucina/biossíntese , Thermus thermophilus/metabolismo
5.
Biochem Biophys Res Commun ; 501(2): 465-470, 2018 06 22.
Artigo em Inglês | MEDLINE | ID: mdl-29738774

RESUMO

2-Isopropylmalate synthase (IPMS) catalyzes the first step of leucine biosynthesis and is regulated via feedback inhibition by leucine. The thermophilic bacterium, Thermus thermophilus HB27, has two IPMS homologous genes: TTC0847 and TTC0849, both of which are in the branched-chain amino acid biosynthetic gene cluster. Since enzymes involved in the leucine biosynthetic pathway are evolutionarily related to those in isoleucine biosynthesis, TTC0847 and TTC0849 are expected to function as IPMS or citramalate synthase, which is the first enzyme in the isoleucine biosynthetic pathway from pyruvate. We characterized these proteins in vitro and in vivo, and revealed that TTC0849 plays a key role in the biosynthesis of leucine and isoleucine, whereas TTC0847 is only involved in that of isoleucine.


Assuntos
2-Isopropilmalato Sintase/metabolismo , Thermus thermophilus/enzimologia , 2-Isopropilmalato Sintase/química , 2-Isopropilmalato Sintase/genética , Sequência de Aminoácidos , Vias Biossintéticas , Domínio Catalítico , Deleção de Genes , Isoleucina/metabolismo , Leucina/metabolismo , Modelos Moleculares , Família Multigênica , Alinhamento de Sequência , Thermus thermophilus/química , Thermus thermophilus/genética , Thermus thermophilus/metabolismo
6.
Biochemistry ; 55(12): 1863-72, 2016 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-26935545

RESUMO

Within the DRE-TIM metallolyase superfamily, members of the Claisen-like condensation (CC-like) subgroup catalyze C-C bond-forming reactions between various α-ketoacids and acetyl-coenzyme A. These reactions are important in the metabolic pathways of many bacterial pathogens and serve as engineering scaffolds for the production of long-chain alcohol biofuels. To improve functional annotation and identify sequences that might use novel substrates in the CC-like subgroup, a combination of structural modeling and multiple-sequence alignments identified active site residues on the third, fourth, and fifth ß-strands of the TIM-barrel catalytic domain that are differentially conserved within the substrate-diverse enzyme families. Using α-isopropylmalate synthase and citramalate synthase from Methanococcus jannaschii (MjIPMS and MjCMS), site-directed mutagenesis was used to test the role of each identified position in substrate selectivity. Kinetic data suggest that residues at the ß3-5 and ß4-7 positions play a significant role in the selection of α-ketoisovalerate over pyruvate in MjIPMS. However, complementary substitutions in MjCMS fail to alter substrate specificity, suggesting residues in these positions do not contribute to substrate selectivity in this enzyme. Analysis of the kinetic data with respect to a protein similarity network for the CC-like subgroup suggests that evolutionarily distinct forms of IPMS utilize residues at the ß3-5 and ß4-7 positions to affect substrate selectivity while the different versions of CMS use unique architectures. Importantly, mapping the identities of residues at the ß3-5 and ß4-7 positions onto the protein similarity network allows for rapid annotation of probable IPMS enzymes as well as several outlier sequences that may represent novel functions in the subgroup.


Assuntos
2-Isopropilmalato Sintase/química , 2-Isopropilmalato Sintase/fisiologia , Methanocaldococcus/enzimologia , Mapeamento de Peptídeos/métodos , Domínio Catalítico/fisiologia , Estrutura Secundária de Proteína
7.
J Biol Chem ; 289(40): 27966-78, 2014 Oct 03.
Artigo em Inglês | MEDLINE | ID: mdl-25128527

RESUMO

The committed step of leucine biosynthesis, converting acetyl-CoA and α-ketoisovalerate into α-isopropylmalate, is catalyzed by α-isopropylmalate synthase (IPMS), an allosteric enzyme subjected to feedback inhibition by the end product L-leucine. We characterized the short form IPMS from Leptospira biflexa (LbIPMS2), which exhibits a catalytic activity comparable with that of the long form IPMS (LbIPMS1) and has a similar N-terminal domain followed by subdomain I and subdomain II but lacks the whole C-terminal regulatory domain. We found that partial deletion of the regulatory domain of LbIPMS1 resulted in a loss of about 50% of the catalytic activity; however, when the regulatory domain was deleted up to Arg-385, producing a protein that is almost equivalent to the intact LbIPMS2, about 90% of the activity was maintained. Moreover, in LbIPMS2 or LbIPMS1, further deletion of several residues from the C terminus of subdomain II significantly impaired or completely abolished the catalytic activity, respectively. These results define a complete and independently functional catalytic module of IPMS consisting of both the N-terminal domain and the two subdomains. Structural comparison of LbIPMS2 and the Mycobacterium tuberculosis IPMS revealed two different conformations of subdomain II that likely represent two substrate-binding states related to cooperative catalysis. The biochemical and structural analyses together with the previously published hydrogen-deuterium exchange data led us to propose a conformation transition mechanism for feedback inhibition mediated by subdomains I and II that might associated with alteration of the binding affinity toward acetyl-CoA.


Assuntos
2-Isopropilmalato Sintase/química , 2-Isopropilmalato Sintase/metabolismo , Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Retroalimentação Fisiológica , Leptospira/enzimologia , 2-Isopropilmalato Sintase/genética , Acetilcoenzima A/química , Acetilcoenzima A/metabolismo , Sequência de Aminoácidos , Proteínas de Bactérias/genética , Sítios de Ligação , Cristalografia por Raios X , Cinética , Leptospira/química , Leptospira/genética , Leucina/química , Leucina/metabolismo , Modelos Moleculares , Dados de Sequência Molecular , Estrutura Terciária de Proteína , Alinhamento de Sequência , Especificidade por Substrato
8.
Biochemistry ; 53(29): 4847-56, 2014 Jul 29.
Artigo em Inglês | MEDLINE | ID: mdl-24991690

RESUMO

Understanding the evolution of allostery in multidomain enzymes remains an important step in improving our ability to identify and exploit structure-function relationships in allosteric mechanisms. A recent protein similarity network for the DRE-TIM metallolyase superfamily indicated there are two evolutionarily distinct forms of the enzyme α-isopropylmalate synthase (IPMS) sharing approximately 20% sequence identity. IPMS from Mycobacterium tuberculosis has been extensively characterized with respect to catalysis and the mechanism of feedback regulation by l-leucine. Here, IPMS from Methanococcus jannaschii (MjIPMS) is used as a representative of the second form of the enzyme, and its catalytic and regulatory mechanism is compared with that of MtIPMS to identify any functional differences between the two forms. MjIPMS exhibits kinetic parameters similar to those of other reported IPMS enzymes and is partially inhibited by l-leucine in a V-type manner. Identical values of (D2O)kcat (3.1) were determined in the presence and absence of l-leucine, indicating the hydrolytic step is rate-determining in the absence of l-leucine and remains so in the inhibited form of the enzyme. This mechanism is identical to the mechanism identified for MtIPMS ((D2O)kcat = 3.3 ± 0.3 in the presence of l-leucine) despite product release being rate-determining in the uninhibited MtIPMS enzyme. The identification of identical regulatory mechanisms in enzymes with low sequence identity raises important evolutionary questions concerning the acquisition and divergence of multidomain allosteric enzymes and highlights the need for caution when comparing regulatory mechanisms for homologous enzymes.


Assuntos
2-Isopropilmalato Sintase/química , Proteínas de Bactérias/química , 2-Isopropilmalato Sintase/antagonistas & inibidores , Regulação Alostérica , Proteínas de Bactérias/antagonistas & inibidores , Biocatálise , Evolução Molecular , Concentração de Íons de Hidrogênio , Cinética , Leucina/química , Methanocaldococcus/enzimologia , Modelos Moleculares , Mycobacterium tuberculosis/enzimologia , Estrutura Quaternária de Proteína , Estrutura Terciária de Proteína , Especificidade da Espécie
9.
Biochemistry ; 53(18): 2915-25, 2014 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-24720347

RESUMO

The characterization of functionally diverse enzyme superfamilies provides the opportunity to identify evolutionarily conserved catalytic strategies, as well as amino acid substitutions responsible for the evolution of new functions or specificities. Isopropylmalate synthase (IPMS) belongs to the DRE-TIM metallolyase superfamily. Members of this superfamily share common active site elements, including a conserved active site helix and an HXH divalent metal binding motif, associated with stabilization of a common enolate anion intermediate. These common elements are overlaid by variations in active site architecture resulting in the evolution of a diverse set of reactions that include condensation, lyase/aldolase, and carboxyl transfer activities. Here, using IPMS, an integrated biochemical and bioinformatics approach has been utilized to investigate the catalytic role of residues on an active site helix that is conserved across the superfamily. The construction of a sequence similarity network for the DRE-TIM metallolyase superfamily allows for the biochemical results obtained with IPMS variants to be compared across superfamily members and within other condensation-catalyzing enzymes related to IPMS. A comparison of our results with previous biochemical data indicates an active site arginine residue (R80 in IPMS) is strictly required for activity across the superfamily, suggesting that it plays a key role in catalysis, most likely through enolate stabilization. In contrast, differential results obtained from substitution of the C-terminal residue of the helix (Q84 in IPMS) suggest that this residue plays a role in reaction specificity within the superfamily.


Assuntos
2-Isopropilmalato Sintase/química , Mycobacterium tuberculosis/enzimologia , 2-Isopropilmalato Sintase/antagonistas & inibidores , 2-Isopropilmalato Sintase/genética , 2-Isopropilmalato Sintase/metabolismo , Regulação Alostérica , Sequência de Aminoácidos , Substituição de Aminoácidos , Arginina/química , Sítios de Ligação/genética , Catálise , Domínio Catalítico/genética , Biologia Computacional , Cinética , Leucina/farmacologia , Modelos Moleculares , Estrutura Secundária de Proteína , Alinhamento de Sequência
10.
FEBS Lett ; 588(9): 1603-7, 2014 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-24613923

RESUMO

α-Isopropylmalate synthase (IPMS) catalyses the reaction between α-ketoisovalerate and acetyl coenzyme A (AcCoA) in the first step of leucine biosynthesis. IPMS is closely related to homocitrate synthase, which catalyses the reaction between AcCoA and the unbranched α-ketoacid α-ketoglutarate. Analysis of these enzymes suggests that several differently conserved key residues are responsible for the different substrate selectivity. These residues were systematically substituted in the Mycobacterium tuberculosis IPMS, resulting in changes in substrate specificity. A variant of IPMS was constructed with a preference for the unbranched α-ketoacids α-ketobutyrate and pyruvate over the natural branched substrate α-ketoisovalerate.


Assuntos
2-Isopropilmalato Sintase/genética , Proteínas de Bactérias/genética , Mycobacterium tuberculosis/enzimologia , 2-Isopropilmalato Sintase/química , Sequência de Aminoácidos , Substituição de Aminoácidos , Proteínas de Bactérias/química , Domínio Catalítico , Cetoácidos/química , Cinética , Modelos Moleculares , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Especificidade por Substrato
11.
Biochem Biophys Res Commun ; 433(2): 249-54, 2013 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-23500460

RESUMO

α-Isopropylmalate synthase (α-IPMS) is a multi-domain protein catalysing the condensation of α-ketoisovalerate (α-KIV) and acetyl coenzyme A (AcCoA) to form α-isopropylmalate. This reaction is the first committed step in the leucine biosynthetic pathway in bacteria and plants, and α-IPMS is allosterically regulated by this amino acid. Existing crystal structures of α-IPMS from Mycobacterium tuberculosis (MtuIPMS) indicate that this enzyme has a strikingly different domain arrangement in each monomer of the homodimeric protein. This asymmetry results in two distinct interfaces between the N-terminal catalytic domains and the C-terminal regulatory domains in the dimer. In this study, residues Arg97 and Asp444 across one of these unequal domain interfaces were substituted to evaluate the importance of protein asymmetry and salt bridge formation between this pair of residues. Analysis of solution-phase structures of wild-type and variant MtuIPMS indicates that substitutions of these residues have little effect on overall protein conformation, a result also observed for addition of the feedback inhibitor leucine to the wild-type enzyme. All variants had increased catalytic efficiency relative to wild-type MtuIPMS, and those with an Asp444 substitution displayed increased affinity for the substrate AcCoA. All variants also showed reduced sensitivity to leucine and altered biphasic reaction kinetics when compared with those of the wild-type enzyme. It is proposed that substituting residues at the asymmetric domain interface increases flexibility in the protein, particularly affecting the AcCoA binding site and the response to leucine, without penalty on catalysis.


Assuntos
2-Isopropilmalato Sintase/antagonistas & inibidores , 2-Isopropilmalato Sintase/química , 2-Isopropilmalato Sintase/metabolismo , Leucina/metabolismo , Mycobacterium tuberculosis/enzimologia , 2-Isopropilmalato Sintase/genética , Substituição de Aminoácidos , Arginina/metabolismo , Sítios de Ligação , Cinética , Leucina/química , Modelos Moleculares , Conformação Proteica , Estrutura Terciária de Proteína , Espalhamento a Baixo Ângulo , Difração de Raios X
12.
Biochemistry ; 51(24): 4773-5, 2012 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-22662746

RESUMO

The identification of structure-function relationships in allosteric enzymes is essential to describing a molecular mechanism for allosteric processes. The enzyme α-isopropylmalate synthase from Mycobacterium tuberculosis (MtIPMS) is subject to slow-onset, allosteric inhibition by l-leucine. Here we report that alternate amino acids act as rapid equilibrium noncompetitive inhibitors of MtIPMS failing to display biphasic inhibition kinetics. Amino acid substitutions on a flexible loop covering the regulatory binding pocket generate enzyme variants that have significant affinity for l-leucine but lack biphasic inhibition kinetics. Taken together, these results are consistent with the flexible loop mediating the slow-onset step of allosteric inhibition.


Assuntos
2-Isopropilmalato Sintase/antagonistas & inibidores , 2-Isopropilmalato Sintase/química , Mycobacterium tuberculosis/enzimologia , 2-Isopropilmalato Sintase/genética , 2-Isopropilmalato Sintase/metabolismo , Regulação Alostérica , Domínio Catalítico , Cinética , Modelos Moleculares , Mutagênese Sítio-Dirigida , Mutação
13.
Nat Chem Biol ; 8(6): 518-26, 2012 May 17.
Artigo em Inglês | MEDLINE | ID: mdl-22596203

RESUMO

Recursive pathways are broadly defined as those that catalyze a series of reactions such that the key, bond-forming functional group of the substrate is always regenerated in each cycle, allowing for a new cycle of reactions to begin. Recursive carbon-chain elongation pathways in nature produce fatty acids, polyketides, isoprenoids and α-keto acids (αKAs), which all use modular or iterative approaches for chain elongation. Recently, an artificial pathway for αKA elongation has been built that uses an engineered isopropylmalate synthase to recursively condense acetyl-CoA with αKAs. This synthetic approach expands the possibilities for recursive pathways beyond the modular or iterative synthesis of natural products and serves as a case study for understanding the challenges of building recursive pathways from nonrecursive enzymes. There exists the potential to design synthetic recursive pathways far beyond what nature has evolved.


Assuntos
2-Isopropilmalato Sintase/química , Acetilcoenzima A/química , Cetoácidos/química , Bibliotecas de Moléculas Pequenas/síntese química , 2-Isopropilmalato Sintase/genética , Sítios de Ligação , Ciclo do Carbono , Catálise , Modelos Moleculares , Estrutura Molecular , Engenharia de Proteínas/métodos , Bibliotecas de Moléculas Pequenas/química , Especificidade por Substrato , Biologia Sintética/métodos
14.
Biochemistry ; 51(11): 2289-97, 2012 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-22352945

RESUMO

α-Isopropylmalate synthase (α-IPMS) catalyzes the metal-dependent aldol reaction between α-ketoisovalerate (α-KIV) and acetyl-coenzyme A (AcCoA) to give α-isopropylmalate (α-IPM). This reaction is the first committed step in the biosynthesis of leucine in bacteria. α-IPMS is homodimeric, with monomers consisting of (ß/α)(8) barrel catalytic domains fused to a C-terminal regulatory domain, responsible for binding leucine and providing feedback regulation for leucine biosynthesis. In these studies, we demonstrate that removal of the regulatory domain from the α-IPMS enzymes of both Neisseria meningitidis (NmeIPMS) and Mycobacterium tuberculosis (MtuIPMS) results in enzymes that are unable to catalyze the formation of α-IPM, although truncated NmeIPMS was still able to slowly hydrolyze AcCoA. The lack of catalytic activity of these truncation variants was confirmed by complementation studies with Escherichia coli cells lacking the α-IPMS gene, where transformation with the plasmids encoding the truncated α-IPMS enzymes was not able to rescue α-IPMS activity. X-ray crystal structures of both truncation variants reveal that both proteins are dimeric and that the catalytic sites of the proteins are intact, although the divalent metal ion that is thought to be responsible for activating substrate α-KIV is displaced slightly relative to its position in the substrate-bound, wild-type structure. Isothermal titration calorimetry and WaterLOGSY nuclear magnetic resonance experiments demonstrate that although these truncation variants are not able to catalyze the reaction between α-KIV and AcCoA, they are still able to bind the substrate α-KIV. It is proposed that the regulatory domain is crucial for ensuring protein dynamics necessary for competent catalysis.


Assuntos
2-Isopropilmalato Sintase/química , 2-Isopropilmalato Sintase/metabolismo , Acetilcoenzima A/química , Acetilcoenzima A/metabolismo , Sítios de Ligação , Catálise , Domínio Catalítico , Cristalografia por Raios X , Hemiterpenos , Cetoácidos/química , Cetoácidos/metabolismo , Cinética , Mycobacterium tuberculosis/enzimologia , Mycobacterium tuberculosis/metabolismo , Neisseria meningitidis/enzimologia , Neisseria meningitidis/metabolismo , Especificidade por Substrato
15.
Arch Biochem Biophys ; 519(2): 202-9, 2012 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-22033339

RESUMO

The manipulation of modular regulatory domains from allosteric enzymes represents a possible mechanism to engineer allostery into non-allosteric systems. Currently, there is insufficient understanding of the structure/function relationships in modular regulatory domains to rationally implement this methodology. The LeuA dimer regulatory domain represents a well-conserved, novel fold responsible for the regulation of two enzymes involved in branched chain amino acid biosynthesis, α-isopropylmalate synthase and citramalate synthase. The LeuA dimer regulatory domain is responsible for the feedback inhibition of these enzymes by their respective downstream products. Both enzymes display multidomain architecture with a conserved N-terminal TIM barrel catalytic domain and a C-terminal (ßßßα)2 LeuA dimer domain joined by a flexible linker region. Due to the similarity of three-dimensional structure and catalytic mechanism combined with low sequence similarity, we propose these enzymes can be classified as members of the LeuA dimer superfamily. Despite their similarity, members of the LeuA dimer superfamily display diversity in their allosteric mechanisms. In this review, structural aspects of the LeuA dimer superfamily are discussed followed by three examples highlighting the diversity of allosteric mechanisms in the LeuA dimer superfamily.


Assuntos
2-Isopropilmalato Sintase/química , 2-Isopropilmalato Sintase/metabolismo , Acetiltransferases/química , Acetiltransferases/metabolismo , Multimerização Proteica , Regulação Alostérica , Bactérias/enzimologia , Estrutura Terciária de Proteína
16.
Biochem Biophys Res Commun ; 393(1): 168-73, 2010 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-20117081

RESUMO

alpha-Isopropylmalate synthase (alpha-IPMS) catalyses the first committed step in leucine biosynthesis in many pathogenic bacteria, including Neisseria meningitidis. This enzyme (NmeIPMS) has been purified, characterised, and compared to alpha-IPMS proteins from other bacteria. NmeIPMS is a homodimer which catalyses the condensation of alpha-ketoisovalerate (alpha-KIV) and acetyl coenzyme A (AcCoA), and is inhibited by leucine. NmeIPMS can use alternate alpha-ketoacids as substrates and, in contrast to alpha-IPMS from other sources, is activated by a range of metal ions including Cd(2+) and Zn(2+) that have previously been reported as inhibitory, since they suppress the dithiodipyridone assay system rather than the enzyme itself. Previous studies indicate that alpha-IPMS is a TIM barrel enzyme with an allosteric leucine-binding domain. To assess the importance of this domain, a truncated form of NmeIPMS was generated and characterised. Loss of the regulatory domain resulted in a loss of the ability to catalyse the aldol reaction, although the enzyme was still able to slowly hydrolyse AcCoA independently of alpha-KIV at a rate similar to that of the WT enzyme. This implies that the regulatory domain is not only required for control of enzymatic activity but may assist in the positioning of key residues in the catalytic TIM barrel. The importance of this domain to catalytic function may offer new strategies for inhibitor design.


Assuntos
2-Isopropilmalato Sintase/química , Neisseria meningitidis/enzimologia , 2-Isopropilmalato Sintase/antagonistas & inibidores , 2-Isopropilmalato Sintase/genética , Regulação Alostérica , Cádmio/química , Catálise , Clonagem Molecular , Desenho de Fármacos , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Estabilidade Enzimática , Multimerização Proteica , Estrutura Terciária de Proteína , Especificidade por Substrato , Zinco/química
17.
Biochemistry ; 48(31): 7457-64, 2009 Aug 11.
Artigo em Inglês | MEDLINE | ID: mdl-19606873

RESUMO

As it is becoming accepted that allosteric regulation can occur through a change in local conformational equilibria as opposed to a change in overall static structure, a thorough description of the structural aspects of these types of mechanisms will be essential to understanding this fundamental biological process. Here we report the experimental identification of key regions of conformational perturbation in the allosteric network of a large (144 kDa), multidomain enzyme by use of solution-phase hydrogen/deuterium exchange. Large perturbations in the regulatory domain induced by effector molecule binding are linked to a very specific, targeted perturbation in the active site, some 50 A away. Binding of L-leucine to an enzyme variant (Y410F) that is kinetically insensitive to effector binding was shown to elicit similar changes in the regulatory domain, but perturbs an alternate region of the catalytic domain, consistent with the proposed allosteric mechanism. These results comprise one of the first reports of an experimentally mapped allosteric mechanism in a protein of this size and provide necessary information to be used toward the development of allostery-based drugs or enzymes with engineered regulatory properties.


Assuntos
2-Isopropilmalato Sintase/antagonistas & inibidores , 2-Isopropilmalato Sintase/química , Medição da Troca de Deutério , Regulação para Baixo , Leucina/química , Mycobacterium tuberculosis/enzimologia , 2-Isopropilmalato Sintase/metabolismo , Regulação Alostérica , Retroalimentação Fisiológica , Transição de Fase , Ligação Proteica , Estrutura Terciária de Proteína , Soluções , Espectroscopia de Infravermelho com Transformada de Fourier
18.
Biochemistry ; 48(9): 1996-2004, 2009 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-19166329

RESUMO

The enzyme alpha-isopropylmalate synthase from Mycobacterium tuberculosis (MtIPMS) has been identified as a possible target for the design of new antitubercular therapeutics. Recently, it was shown that MtIPMS is subject to slow-onset, feedback inhibition by l-leucine, the first instance of an allosteric regulator utilizing this mechanism. Structural studies are inconsistent with canonical allosteric mechanisms, including changes to the quaternary structure or large, rigid-body conformational changes to the enzyme upon l-leucine binding. Thus, the allosteric regulation may result from a discrete inhibitory signal transmitted to the active site upon l-leucine binding in the regulatory domain, a distance of more than 50 A. To test this mechanism, site-directed mutagenesis was employed to construct enzymes with substitutions at phylogenetically conserved active site residues near the interface of the catalytic and linker domains. The substitutions had wide-ranging effects on the kinetics of l-leucine inhibition, with some modest effects on the kinetic parameters of catalysis. The most dramatic result was the finding that the Y410F mutant form of MtIPMS is insensitive to l-leucine inhibition, suggesting that this residue has completely uncoupled the inhibitory signal to the active site. Overall, the data are consistent with a mechanism of allosteric regulation described by the interdomain communication of the inhibitory signal from the regulatory to catalytic domain and implicate the interactions between the linker and catalytic domains as critical determinants of inhibitory signal transmission.


Assuntos
2-Isopropilmalato Sintase/química , Proteínas de Bactérias/química , Mycobacterium tuberculosis/enzimologia , 2-Isopropilmalato Sintase/genética , 2-Isopropilmalato Sintase/metabolismo , Regulação Alostérica , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Sítios de Ligação/genética , Calorimetria/métodos , Catálise/efeitos dos fármacos , Cinética , Leucina/metabolismo , Leucina/farmacologia , Espectroscopia de Ressonância Magnética , Malatos/química , Malatos/metabolismo , Modelos Moleculares , Estrutura Molecular , Mutagênese Sítio-Dirigida , Mutação de Sentido Incorreto , Mycobacterium tuberculosis/genética , Conformação Proteica , Estrutura Terciária de Proteína , Titulometria
19.
FEMS Microbiol Lett ; 286(2): 166-70, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18647358

RESUMO

The 57-bp tandem repeats located in the Mycobacterium tuberculosis leuA gene code for the alpha-isopropylmalate synthase (alpha-IPMS). It is unique to this pathogen. It was previously demonstrated that the leuA-coding sequence Rv3710, containing the tandem repeats, can be translated to an active alpha-IPMS. The objective of the present study was to investigate the significance and effect of the two 57-bp tandem repeats upon gene expression and the general properties of alpha-IPMS. The putative M. tuberculosis H37Rv leuA gene with and without the tandem repeats was cloned by PCR and expressed in an Escherichia coli host. The enzyme product was studied for general properties, comparing that from a native leuA gene containing two repeats and that from the 57-bp tandem repeats deletion mutant. Upon deletion of the two 57-bp tandem repeats, the expression level of leuA from M. tuberculosis H37Rv was comparable with that of the native form. The general properties of the two types of enzymes were similar. They were both functional with the same range of optimal temperature and optimal pH for activity and with similar enzyme stability. Deletion of the repeats had no detectable effect on leuA expression level or the general properties of the enzyme product.


Assuntos
2-Isopropilmalato Sintase/metabolismo , Proteínas de Bactérias/metabolismo , Mycobacterium tuberculosis/enzimologia , Sequências de Repetição em Tandem , 2-Isopropilmalato Sintase/química , 2-Isopropilmalato Sintase/genética , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Clonagem Molecular , Estabilidade Enzimática , Escherichia coli/genética , Expressão Gênica , Concentração de Íons de Hidrogênio , Mycobacterium tuberculosis/genética , Deleção de Sequência , Temperatura
20.
BMC Struct Biol ; 7: 39, 2007 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-17577419

RESUMO

BACKGROUND: Alpha-isopropylmalate synthase (MtalphaIPMS), an enzyme that catalyzes the first committed step of the leucine biosynthetic pathway of Mycobacterium tuberculosis is a potential drug target for the anti-tuberculosis drugs. Cations induce differential effect of activation and inhibition of MtalphaIPMS. To date no concrete mechanism for such an opposite effect of similarly charged cations on the functional activity of enzyme has been presented. RESULTS: Effect of cations on the structure and function of the MtalphaIPMS has been studied in detail. The studies for the first time demonstrate that different cations interact specifically at different sites in the enzyme and modulate the enzyme structure differentially. The inhibitors Zn2+ and Cd2+ ions interact directly with the catalytic domain of the enzyme and induce unfolding/denaturation of the domain. The activator K+ also interacts with the catalytic TIM barrel domain however, it does not induce any significant effect on the enzyme structure. Studies with isolated catalytic TIM barrel domain showed that it can carry out the catalytic function on its own but probably requires the non-catalytic C-terminal domain for optimum functioning. An important observation was that divalent cations induce significant interaction between the regulatory and the catalytic domain of MtalphaIPMS thus inducing structural cooperativity in the enzyme. This divalent cation induced structural cooperativity might result in modulation of activity of the catalytic domain by regulatory domain. CONCLUSION: The studies for the first time demonstrate that different cations bind at different sites in the enzyme leading to their differential effects on the structure and functional activity of the enzyme.


Assuntos
2-Isopropilmalato Sintase/química , 2-Isopropilmalato Sintase/metabolismo , Mycobacterium tuberculosis/enzimologia , 2-Isopropilmalato Sintase/genética , 2-Isopropilmalato Sintase/isolamento & purificação , Cátions/química , Cátions/farmacologia , Dimerização , Estabilidade Enzimática/efeitos dos fármacos , Expressão Gênica , Metais/química , Metais/metabolismo , Modelos Moleculares , Mycobacterium tuberculosis/genética , Desnaturação Proteica , Dobramento de Proteína , Estrutura Quaternária de Proteína/efeitos dos fármacos , Estrutura Secundária de Proteína/efeitos dos fármacos , Eletricidade Estática , Temperatura
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA