Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Arch Biochem Biophys ; 692: 108516, 2020 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-32745463

RESUMO

Ketol-acid reductoisomerase (KARI), the second enzyme in the branched-chain amino acid (BCAA) biosynthesis pathway, is an emerging target for the discovery of biocides. Here, we demonstrate that cyclopropane-1,1-dicarboxylate (CPD) inhibits KARIs from the pathogens Mycobacterium tuberculosis (Mt) and Campylobacter jejuni (Cj) reversibly with Ki values of 3.03 µM and 0.59 µM, respectively. Another reversible inhibitor of both KARIs, Hoe 704, is more potent than CPD with Ki values of 300 nM and 110 nM for MtKARI and CjKARI, respectively. The most potent inhibitor tested here is N-hydroxy-N-isopropyloxamate (IpOHA). It has a Ki of ~26 nM for MtKARI, but binds rather slowly (kon ~900 M-1s-1). In contrast, IpOHA binds more rapidly (kon ~7000 M-1s-1) to CjKARI and irreversibly.


Assuntos
Proteínas de Bactérias/antagonistas & inibidores , Campylobacter jejuni/enzimologia , Inibidores Enzimáticos/química , Cetol-Ácido Redutoisomerase/antagonistas & inibidores , Mycobacterium tuberculosis/enzimologia , Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Campylobacter jejuni/química , Ciclopropanos/química , Ácidos Dicarboxílicos/química , Ácidos Hidroxâmicos/química , Cetol-Ácido Redutoisomerase/química , Cetol-Ácido Redutoisomerase/metabolismo , Mycobacterium tuberculosis/química , Compostos Organofosforados/química
2.
Chemistry ; 26(41): 8958-8968, 2020 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-32198779

RESUMO

Ketol-acid reductoisomerase (KARI), the second enzyme in the branched-chain amino acid biosynthesis pathway, is a potential drug target for bacterial infections including Mycobacterium tuberculosis. Here, we have screened the Medicines for Malaria Venture Pathogen Box against purified M. tuberculosis (Mt) KARI and identified two compounds that have Ki values below 200 nm. In Mt cell susceptibility assays one of these compounds exhibited an IC50 value of 0.8 µm. Co-crystallization of this compound, 3-((methylsulfonyl)methyl)-2H-benzo[b][1,4]oxazin-2-one (MMV553002), in complex with Staphylococcus aureus KARI, which has 56 % identity with Mt KARI, NADPH and Mg2+ yielded a structure to 1.72 Šresolution. However, only a hydrolyzed product of the inhibitor (i.e. 3-(methylsulfonyl)-2-oxopropanic acid, missing the 2-aminophenol attachment) is observed in the active site. Surprisingly, Mt cell susceptibility assays showed that the 2-aminophenol product is largely responsible for the anti-TB activity of the parent compound. Thus, 3-(methylsulfonyl)-2-oxopropanic acid was identified as a potent KARI inhibitor that could be further explored as a potential biocidal agent and we have shown 2-aminophenol, as an anti-TB drug lead, especially given it has low toxicity against human cells. The study highlights that careful analysis of broad screening assays is required to correctly interpret cell-based activity data.


Assuntos
Cetol-Ácido Redutoisomerase/metabolismo , Magnésio/química , Mycobacterium tuberculosis/enzimologia , NADP/química , Staphylococcus aureus/metabolismo , Domínio Catalítico , Cristalização , Cristalografia por Raios X , Humanos , Cetol-Ácido Redutoisomerase/química , Mycobacterium tuberculosis/química , NADP/metabolismo , Staphylococcus aureus/química
3.
Chembiochem ; 21(3): 381-391, 2020 02 03.
Artigo em Inglês | MEDLINE | ID: mdl-31309701

RESUMO

Binuclear Mg ketol-acid reductoisomerase (KARI), which converts (S)-2-acetolactate into (R)-2,3-dihydroxyisovalerate, is responsible for the second step of the biosynthesis of branched-chain amino acids in plants and microorganisms and thus serves as a key inhibition target potentially without effects on mammals. Here, through the use of density functional calculations and a chemical model, the KARI-catalyzed reaction has been demonstrated to include the initial deprotonation of the substrate C2 hydroxy group, bridged by the two Mg ions, alkyl migration from the C2-alkoxide carbon atom to the C3-carbonyl carbon atom, and hydride transfer from a nicotinamide adenine dinucleotide phosphate [NAD(P)H] cofactor to C2. A dead-end mechanism with a hydride transferred to the C3 carbonyl group has been ruled out. The nucleophilicity (migratory aptitude) of the migrating carbon atom and the provision of additional negative charge to the di-Mg coordination sphere have significant effects on the steps of alkyl migration and hydride transfer, respectively. Other important mechanistic characteristics are also revealed. Inspired by the mechanism, an inhibitor (2-carboxylate-lactic acid) was designed and predicted by barrier analysis to be effective in inactivating KARI, hence probably enriching the antifungal and antibacterial library. Two types of slow substrate analogues (2-trihalomethyl acetolactic acids and 2-glutaryl lactic acid) were also found.


Assuntos
Aminoácidos de Cadeia Ramificada/antagonistas & inibidores , Ácidos Carboxílicos/farmacologia , Inibidores Enzimáticos/farmacologia , Cetol-Ácido Redutoisomerase/antagonistas & inibidores , Ácido Láctico/farmacologia , Magnésio/metabolismo , Aminoácidos de Cadeia Ramificada/biossíntese , Ácidos Carboxílicos/síntese química , Ácidos Carboxílicos/química , Cristalografia por Raios X , Teoria da Densidade Funcional , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/química , Cetol-Ácido Redutoisomerase/química , Cetol-Ácido Redutoisomerase/metabolismo , Ácido Láctico/síntese química , Ácido Láctico/química , Magnésio/química , Modelos Moleculares , Estrutura Molecular
4.
J Am Chem Soc ; 141(51): 19983-19987, 2019 12 26.
Artigo em Inglês | MEDLINE | ID: mdl-31829582

RESUMO

Protein functions are temperature-dependent, but protein structures are usually solved at a single (often low) temperature because of limitations on the conditions of crystal growth or protein vitrification. Here we demonstrate the feasibility of solving cryo-EM structures of proteins vitrified at high temperatures, solve 12 structures of an archaeal ketol-acid reductoisomerase (KARI) vitrified at 4-70 °C, and show that structures of both the Mg2+ form (KARI:2Mg2+) and its ternary complex (KARI:2Mg2+:NADH:inhibitor) are temperature-dependent in correlation with the temperature dependence of enzyme activity. Furthermore, structural analyses led to dissection of the induced-fit mechanism into ligand-induced and temperature-induced effects and to capture of temperature-resolved intermediates of the temperature-induced conformational change. The results also suggest that it is preferable to solve cryo-EM structures of protein complexes at functional temperatures. These studies should greatly expand the landscapes of protein structure-function relationships and enhance the mechanistic analysis of enzymatic functions.


Assuntos
Cetol-Ácido Redutoisomerase/metabolismo , Temperatura , Microscopia Crioeletrônica , Cristalografia por Raios X , Cetol-Ácido Redutoisomerase/química , Modelos Moleculares , Conformação Molecular , Sulfolobus solfataricus/enzimologia
5.
J Agric Food Chem ; 67(31): 8527-8535, 2019 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-31298526

RESUMO

l-Valine belongs to the branched-chain amino acids (BCAAs) and is an essential amino acid that is crucial for all living organisms. l-Valine is industrially produced by the nonpathogenic bacterium Corynebacterium glutamicum and is synthesized by the BCAA biosynthetic pathway. Ketol-acid reductoisomerase (KARI) is the second enzyme in the BCAA pathway and catalyzes the conversion of (S)-2-acetolactate into (R)-2,3-dihydroxy-isovalerate, or the conversion of (S)-2-aceto-2-hydroxybutyrate into (R)-2,3-dihydroxy-3-methylvalerate. To elucidate the enzymatic properties of KARI from C. glutamicum (CgKARI), we successfully produced CgKARI protein and determined its crystal structure in complex with NADP+ and two Mg2+ ions. Based on the complex structure, docking simulations, and site-directed mutagenesis experiments, we revealed that CgKARI belongs to Class I KARI and identified key residues involved in stabilization of the substrate, metal ions, and cofactor. Furthermore, we confirmed the difference in the binding of metal ions that depended on the conformational change.


Assuntos
Proteínas de Bactérias/química , Corynebacterium glutamicum/enzimologia , Cetol-Ácido Redutoisomerase/química , Sequência de Aminoácidos , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Sítios de Ligação , Domínio Catalítico , Corynebacterium glutamicum/química , Corynebacterium glutamicum/genética , Cristalografia por Raios X , Cetol-Ácido Redutoisomerase/genética , Cetol-Ácido Redutoisomerase/metabolismo , Metais/química , Metais/metabolismo , Simulação de Acoplamento Molecular , NADP/química , NADP/metabolismo
6.
J Am Chem Soc ; 141(15): 6136-6140, 2019 04 17.
Artigo em Inglês | MEDLINE | ID: mdl-30921515

RESUMO

While cryo-EM is revolutionizing structural biology, its impact on enzymology is yet to be fully demonstrated. The ketol-acid reductoisomerase (KARI) catalyzes conversion of (2 S)-acetolactate or (2 S)-aceto-2-hydroxybutyrate to 2,3-dihydroxy-3-alkylbutyrate. We found that KARI from archaea Sulfolobus solfataricus (Sso-KARI) is unusual in being a dodecamer, bispecific to NADH and NADPH, and losing activity above pH 7.8. While crystals were obtainable only at pH 8.5, cryo-EM structures were solved at pH 7.5 and 8.5 for Sso-KARI:2Mg2+. The results showed that the distances of the two catalytic Mg2+ ions are lengthened in both structures at pH 8.5. We next solved cryo-EM structures of two Sso-KARI complexes, with NADH+inhibitor and NADPH+inhibitor at pH 7.5, which indicate that the bispecificity can be attributed to a unique asparagine at the cofactor binding loop. Unexpectedly, Sso-KARI also differs from other KARI enzymes in lacking "induced-fit", reflecting structural rigidity. Thus, cryo-EM is powerful for structural and mechanistic enzymology.


Assuntos
Álcoois/metabolismo , Archaea/enzimologia , Cetol-Ácido Redutoisomerase/química , Cetonas/metabolismo , Álcoois/química , Cristalografia por Raios X , Concentração de Íons de Hidrogênio , Cetol-Ácido Redutoisomerase/metabolismo , Cetonas/química , Modelos Moleculares , Conformação Molecular
7.
J Am Chem Soc ; 141(9): 4108-4118, 2019 03 06.
Artigo em Inglês | MEDLINE | ID: mdl-30761897

RESUMO

Despite tremendous progress in understanding and engineering enzymes, knowledge of how enzyme structures and their dynamics induce observed catalytic properties is incomplete, and capabilities to engineer enzymes fall far short of industrial needs. Here, we investigate the structural and dynamic drivers of enzyme catalysis for the rate-limiting step of the industrially important enzyme ketol-acid reductoisomerase (KARI) and identify a region of the conformational space of the bound enzyme-substrate complex that, when populated, leads to large increases in reactivity. We apply computational statistical mechanical methods that implement transition interface sampling to simulate the kinetics of the reaction and combine this with machine learning techniques from artificial intelligence to select features relevant to reactivity and to build predictive models for reactive trajectories. We find that conformational descriptors alone, without the need for dynamic ones, are sufficient to predict reactivity with greater than 85% accuracy (90% AUC). Key descriptors distinguishing reactive from almost-reactive trajectories quantify substrate conformation, substrate bond polarization, and metal coordination geometry and suggest their role in promoting substrate reactivity. Moreover, trajectories constrained to visit a region of the reactant well, separated from the rest by a simple hyperplane defined by ten conformational parameters, show increases in computed reactivity by many orders of magnitude. This study provides evidence for the existence of reactivity promoting regions within the conformational space of the enzyme-substrate complex and develops methodology for identifying and validating these particularly reactive regions of phase space. We suggest that identification of reactivity promoting regions and re-engineering enzymes to preferentially populate them may lead to significant rate enhancements.


Assuntos
Cetol-Ácido Redutoisomerase/metabolismo , Aprendizado de Máquina , Simulação de Dinâmica Molecular , Biocatálise , Cetol-Ácido Redutoisomerase/química , Método de Monte Carlo , Conformação Proteica , Especificidade por Substrato
8.
Sci Rep ; 8(1): 7176, 2018 05 08.
Artigo em Inglês | MEDLINE | ID: mdl-29739976

RESUMO

Ketol-acid reductoisomerase (KARI) is a bifunctional enzyme in the second step of branched-chain amino acids biosynthetic pathway. Most KARIs prefer NADPH as a cofactor. However, KARI with a preference for NADH is desirable in industrial applications including anaerobic fermentation for the production of branched-chain amino acids or biofuels. Here, we characterize a thermoacidophilic archaeal Sac-KARI from Sulfolobus acidocaldarius and present its crystal structure at a 1.75-Å resolution. By comparison with other holo-KARI structures, one sulphate ion is observed in each binding site for the 2'-phosphate of NADPH, implicating its NADPH preference. Sac-KARI has very high affinity for NADPH and NADH, with K M values of 0.4 µM for NADPH and 6.0 µM for NADH, suggesting that both are good cofactors at low concentrations although NADPH is favoured over NADH. Furthermore, Sac-KARI can catalyze 2(S)-acetolactate (2S-AL) with either cofactor from 25 to 60 °C, but the enzyme has higher activity by using NADPH. In addition, the catalytic activity of Sac-KARI increases significantly with elevated temperatures and reaches an optimum at 60 °C. Bi-cofactor utilization and the thermoactivity of Sac-KARI make it a potential candidate for use in metabolic engineering or industrial applications under anaerobic or harsh conditions.


Assuntos
Aminoácidos/biossíntese , Vias Biossintéticas , Cetol-Ácido Redutoisomerase/química , Sulfolobus acidocaldarius/enzimologia , Sequência de Aminoácidos , Sítios de Ligação , Cristalografia por Raios X , Estabilidade Enzimática/genética , Fermentação , Cetol-Ácido Redutoisomerase/biossíntese , Cetol-Ácido Redutoisomerase/genética , Cetol-Ácido Redutoisomerase/metabolismo , NAD/química , NAD/metabolismo , NADP/química , NADP/metabolismo , Sulfolobus acidocaldarius/genética , Temperatura
9.
Chemistry ; 23(72): 18289-18295, 2017 Dec 22.
Artigo em Inglês | MEDLINE | ID: mdl-28975665

RESUMO

Ketol-acid reductoisomerase (KARI) is an NAD(P)H and Mg2+ -dependent enzyme of the branched-chain amino acid (BCAA) biosynthesis pathway. Here, the first crystal structures of Staphylococcus aureus (Sa) KARI in complex with two transition state analogues, cyclopropane-1,1-dicarboxylate (CPD) and N-isopropyloxalyl hydroxamate (IpOHA) are reported. These compounds bind competitively and in multi-dentate manner to KARI with Ki values of 2.73 µm and 7.9 nm, respectively; however, IpOHA binds slowly to the enzyme. Interestingly, intact IpOHA is present in only ≈25 % of binding sites, whereas its deoxygenated form is present in the remaining sites. This deoxy form of IpOHA binds rapidly to Sa KARI, but with much weaker affinity (Ki =21 µm). Thus, our data pinpoint the origin of the slow binding mechanism of IpOHA. Furthermore, we propose that CPD mimics the early stage of the catalytic reaction (preceding the reduction step), whereas IpOHA mimics the late stage (after the reduction took place). These structural insights will guide strategies to design potent and rapidly binding derivatives of these compounds for the development of novel biocides.


Assuntos
Proteínas de Bactérias/química , Ciclopropanos/química , Ácidos Dicarboxílicos/química , Ácidos Hidroxâmicos/química , Cetol-Ácido Redutoisomerase/química , Staphylococcus aureus/enzimologia , Proteínas de Bactérias/metabolismo , Sítios de Ligação , Domínio Catalítico , Cristalização , Cristalografia por Raios X/métodos , Cetol-Ácido Redutoisomerase/metabolismo , Modelos Moleculares , NAD/química , Oxirredução , Ligação Proteica , Conformação Proteica , Relação Estrutura-Atividade , Termodinâmica
10.
FEBS J ; 283(7): 1184-96, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26876563

RESUMO

UNLABELLED: The biosynthetic pathway for the branched-chain amino acids is present in plants, fungi and bacteria, but not in animals, making it an attractive target for herbicidal and antimicrobial drug discovery. Ketol-acid reductoisomerase (KARI; EC 1.1.1.86) is the second enzyme in this pathway, converting in a Mg(2+) - and NADPH-dependent reaction either 2-acetolactate or 2-aceto-2-hydroxybutyrate to their corresponding 2,3-dihydroxy-3-alkylbutyrate products. Here, we have determined the crystal structure of Mycobacterium tuberculosis (Mt) KARI, a class I KARI, with two magnesium ions bound in the active site. X-ray data were obtained to 1.0 Å resolution and the final model has an Rfree of 0.163. The structure shows that the active site is solvent-accessible with the two metal ions separated by 4.7 Å. A comparison of this structure with that of Mg(2+) -free Pseudomonas aeruginosa KARI suggests that upon magnesium binding no movement of the N domain relative to the C domain occurs. However, upon formation of the Michaelis complex, as illustrated in the structure of Slackia exigua KARI in complex with NADH.Mg(2+) . N-hydroxy-N-isopropyloxamate (IpOHA, a transition state analog), domain movements and reduction of the metal-metal distance to 3.5 Å are observed. This inherent flexibility therefore appears to be critical for initiation of the KARI-catalyzed reaction. This study provides new insights into the complex structural rearrangements required for activity of KARIs, particularly those belonging to class I, and provides the framework for the rational design of Mt KARI inhibitors that can be tested as novel antituberculosis agents. DATABASE: Coordinates and structure factors for the Mt KARI.Mg(2+) complex are available in the Protein Data Bank under accession number 4YPO.


Assuntos
Proteínas de Bactérias/química , Cetol-Ácido Redutoisomerase/química , Mycobacterium tuberculosis/enzimologia , Antituberculosos/uso terapêutico , Proteínas de Bactérias/antagonistas & inibidores , Proteínas de Bactérias/metabolismo , Sítios de Ligação , Biocatálise/efeitos dos fármacos , Domínio Catalítico , Cristalografia por Raios X , Descoberta de Drogas , Inibidores Enzimáticos/uso terapêutico , Humanos , Cetol-Ácido Redutoisomerase/antagonistas & inibidores , Cetol-Ácido Redutoisomerase/metabolismo , Magnésio/química , Magnésio/metabolismo , Modelos Moleculares , Terapia de Alvo Molecular , Mycobacterium tuberculosis/fisiologia , Multimerização Proteica , Estrutura Terciária de Proteína , Soluções , Tuberculose/microbiologia , Tuberculose/prevenção & controle
11.
Protein Sci ; 25(7): 1241-8, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-26644020

RESUMO

The duplication of protein structural domains has been proposed as a common mechanism for the generation of new protein folds. A particularly interesting case is the class II ketol-acid reductoisomerase (KARI), which putatively arose from an ancestral class I KARI by duplication of the C-terminal domain and corresponding loss of obligate dimerization. As a result, the class II enzymes acquired a deeply embedded figure-of-eight knot. To test this evolutionary hypothesis we constructed a novel class II KARI by duplicating the C-terminal domain of a hyperthermostable class I KARI. The new protein is monomeric, as confirmed by gel filtration and X-ray crystallography, and has the deeply knotted class II KARI fold. Surprisingly, its catalytic activity is nearly unchanged from the parent KARI. This provides strong evidence in support of domain duplication as the mechanism for the evolution of the class II KARI fold and demonstrates the ability of domain duplication to generate topological novelty in a function-neutral manner.


Assuntos
Archaea/enzimologia , Duplicação Gênica , Cetol-Ácido Redutoisomerase/química , Cetol-Ácido Redutoisomerase/genética , Sequência de Aminoácidos , Archaea/química , Domínio Catalítico , Cristalografia por Raios X , Modelos Moleculares , Dobramento de Proteína , Estrutura Secundária de Proteína
12.
Biochem J ; 468(3): 475-84, 2015 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-25849365

RESUMO

Although most sequenced members of the industrially important ketol-acid reductoisomerase (KARI) family are class I enzymes, structural studies to date have focused primarily on the class II KARIs, which arose through domain duplication. In the present study, we present five new crystal structures of class I KARIs. These include the first structure of a KARI with a six-residue ß2αB (cofactor specificity determining) loop and an NADPH phosphate-binding geometry distinct from that of the seven- and 12-residue loops. We also present the first structures of naturally occurring KARIs that utilize NADH as cofactor. These results show insertions in the specificity loops that confounded previous attempts to classify them according to loop length. Lastly, we explore the conformational changes that occur in class I KARIs upon binding of cofactor and metal ions. The class I KARI structures indicate that the active sites close upon binding NAD(P)H, similar to what is observed in the class II KARIs of rice and spinach and different from the opening of the active site observed in the class II KARI of Escherichia coli. This conformational change involves a decrease in the bending of the helix that runs between the domains and a rearrangement of the nicotinamide-binding site.


Assuntos
Alicyclobacillus/enzimologia , Azotobacter vinelandii/enzimologia , Proteínas de Bactérias/metabolismo , Coenzimas/metabolismo , Desulfurococcaceae/enzimologia , Cetol-Ácido Redutoisomerase/metabolismo , Modelos Moleculares , Adenosina Difosfato Ribose/análogos & derivados , Adenosina Difosfato Ribose/química , Adenosina Difosfato Ribose/metabolismo , Sequência de Aminoácidos , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Sítios de Ligação , Domínio Catalítico , Coenzimas/química , Cristalografia por Raios X , Cetol-Ácido Redutoisomerase/química , Cetol-Ácido Redutoisomerase/genética , Magnésio/química , Magnésio/metabolismo , Conformação Molecular , Dados de Sequência Molecular , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , NAD/química , NAD/metabolismo , NADP/química , NADP/metabolismo , Fosforilação , Dobramento de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Alinhamento de Sequência
13.
Biochimie ; 108: 76-84, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25446654

RESUMO

Due to its enhanced energy content and hydrophobicity, isobutanol is flagged as a next generation biofuel and chemical building block. For cellular and cell-free isobutanol production, NADH dependent (over NADPH dependent) enzyme systems are desired. To improve cell-free isobutanol processes, we characterized and catalytically optimized a NADH dependent, thermo- and solvent stable ketol-acid reductoisomerase (KARI) derived from the bacterium Meiothermus ruber (Mr). The wild type Mr-KARI has the most temperature tolerant KARI specific activity reported to date. The KARI screening procedure developed in this study allows accelerated molecular optimization. Thus, a KARI variant with a 350% improved activity and enhanced NADH cofactor specificity was identified. Other KARI variants gave insights into Mr-KARI structure-function relationships.


Assuntos
Butanóis/metabolismo , Cetol-Ácido Redutoisomerase/química , Cetol-Ácido Redutoisomerase/metabolismo , Mutagênese , Solventes/química , Temperatura , Sequência de Aminoácidos , Biocatálise , Deinococcus/enzimologia , Estabilidade Enzimática , Cetol-Ácido Redutoisomerase/genética , Cinética , Modelos Moleculares , Dados de Sequência Molecular , Conformação Proteica
14.
Methods Mol Biol ; 1216: 129-59, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25213414

RESUMO

Faced with a protein engineering challenge, a contemporary researcher can choose from myriad design strategies. Library-scale computational protein design (LCPD) is a hybrid method suitable for the engineering of improved protein variants with diverse sequences. This chapter discusses the background and merits of several practical LCPD techniques. First, LCPD methods suitable for delocalized protein design are presented in the context of example design calculations for cellobiohydrolase II. Second, localized design methods are discussed in the context of an example design calculation intended to shift the substrate specificity of a ketol-acid reductoisomerase Rossmann domain from NADPH to NADH.


Assuntos
Proteínas/química , Biologia Computacional/métodos , Cetol-Ácido Redutoisomerase/química , NAD/química , NADP/química , Biblioteca de Peptídeos , Engenharia de Proteínas/métodos , Estrutura Terciária de Proteína
15.
Metab Eng ; 26: 17-22, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25172159

RESUMO

All members of the ketol-acid reductoisomerase (KARI) enzyme family characterized to date have been shown to prefer the nicotinamide adenine dinucleotide phosphate hydride (NADPH) cofactor to nicotinamide adenine dinucleotide hydride (NADH). However, KARIs with the reversed cofactor preference are desirable for industrial applications, including anaerobic fermentation to produce branched-chain amino acids. By applying insights gained from structural and engineering studies of this enzyme family to a comprehensive multiple sequence alignment of KARIs, we identified putative NADH-utilizing KARIs and characterized eight whose catalytic efficiencies using NADH were equal to or greater than NADPH. These are the first naturally NADH-preferring KARIs reported and demonstrate that this property has evolved independently multiple times, using strategies unlike those used previously in the laboratory to engineer a KARI cofactor switch.


Assuntos
Cetol-Ácido Redutoisomerase/química , NAD/química , Alinhamento de Sequência/métodos , Análise de Sequência de Proteína/métodos , Sequência de Aminoácidos , Sítios de Ligação , Ativação Enzimática , Dados de Sequência Molecular , Ligação Proteica , Especificidade por Substrato
17.
Proc Natl Acad Sci U S A ; 110(27): 10946-51, 2013 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-23776225

RESUMO

To date, efforts to switch the cofactor specificity of oxidoreductases from nicotinamide adenine dinucleotide phosphate (NADPH) to nicotinamide adenine dinucleotide (NADH) have been made on a case-by-case basis with varying degrees of success. Here we present a straightforward recipe for altering the cofactor specificity of a class of NADPH-dependent oxidoreductases, the ketol-acid reductoisomerases (KARIs). Combining previous results for an engineered NADH-dependent variant of Escherichia coli KARI with available KARI crystal structures and a comprehensive KARI-sequence alignment, we identified key cofactor specificity determinants and used this information to construct five KARIs with reversed cofactor preference. Additional directed evolution generated two enzymes having NADH-dependent catalytic efficiencies that are greater than the wild-type enzymes with NADPH. High-resolution structures of a wild-type/variant pair reveal the molecular basis of the cofactor switch.


Assuntos
Proteínas de Escherichia coli/metabolismo , Cetol-Ácido Redutoisomerase/metabolismo , Sequência de Aminoácidos , Evolução Molecular Direcionada , Escherichia coli/enzimologia , Escherichia coli/genética , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/genética , Cetol-Ácido Redutoisomerase/química , Cetol-Ácido Redutoisomerase/genética , Modelos Moleculares , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , NAD/metabolismo , NADP/metabolismo , Estrutura Secundária de Proteína , Homologia de Sequência de Aminoácidos
18.
J Mol Biol ; 424(3-4): 168-79, 2012 Dec 07.
Artigo em Inglês | MEDLINE | ID: mdl-23036858

RESUMO

Ketol-acid reductoisomerase (KARI) is the second enzyme in the branched-chain amino acid biosynthesis pathway, which is found in plants, fungi and bacteria but not in animals. This difference in metabolism between animals and microorganisms makes KARI an attractive target for the development of antimicrobial agents. Herein we report the crystal structure of Escherichia coli KARI in complex with Mg(2+) and NADPH at 2.3Å resolution. Ultracentrifugation studies confirm that the enzyme exists as a tetramer in solution, and isothermal titration calorimetry shows that the binding of Mg(2+) increases structural disorder while the binding of NADPH increases the structural rigidity of the enzyme. Comparison of the structure of the E. coli KARI-Mg(2+)-NADPH complex with that of enzyme in the absence of cofactors shows that the binding of Mg(2+) and NADPH opens the interface between the N- and C-domains, thereby allowing access for the substrates to bind: the existence of only a small opening between the domains in the crystal structure of the unliganded enzyme signifies restricted access to the active site. This observation contrasts with that in the plant enzyme, where the N-domain can rotate freely with respect to the C-domain until the binding of Mg(2+) and/or NADPH stabilizes the relative positions of these domains. Support is thereby provided for the idea that plant and bacterial KARIs have evolved different mechanisms of induced fit to prepare the active site for catalysis.


Assuntos
Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/metabolismo , Escherichia coli/enzimologia , Cetol-Ácido Redutoisomerase/química , Cetol-Ácido Redutoisomerase/metabolismo , Coenzimas/metabolismo , Cristalografia por Raios X , Magnésio/metabolismo , Modelos Moleculares , NADP/metabolismo , Ligação Proteica , Conformação Proteica , Multimerização Proteica
19.
Biochemistry ; 48(36): 8731-7, 2009 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-19653643

RESUMO

The large, catalytic subunits (LSUs; ilvB, ilvG and ilvI, respectively) of enterobacterial acetohydroxyacid synthases isozymes (AHAS I, II and III) have molecular weights approximately 60 kDa and are paralogous with a family of other thiamin diphosphate dependent enzymes. The small, regulatory subunits (SSUs) of AHAS I and AHAS III (ilvN and ilvH) are required for valine inhibition, but ilvN and ilvH can only confer valine sensitivity on their own LSUs. AHAS II is valine resistant. The LSUs have only approximately 15, <<1 and approximately 3%, respectively, of the activity of their respective holoenzymes, but the holoenzymes can be reconstituted with complete recovery of activity. We have examined the activation of each of the LSUs by SSUs from different isozymes and ask to what extent such activation is specific; that is, is effective nonspecific interaction possible between LSUs and SSUs of different isozymes? To our surprise, the AHAS II SSU ilvM is able to activate the LSUs of all three of the isozymes, and the truncated AHAS III SSUs ilvH-Delta80, ilvH-Delta86 and ilvH-Delta89 are able to activate the LSUs of both AHAS I and AHAS III. However, none of the heterologously activated enzymes have any feedback sensitivity. Our results imply the existence of a common region in all three LSUs to which regulatory subunits may bind, as well as a similarity between the surfaces of ilvM and the other SSUs. This surface must be included within the N-terminal betaalphabetabetaalphabeta-domain of the SSUs, probably on the helical face of this domain. We suggest hypotheses for the mechanism of valine inhibition, and reject one involving induced dissociation of subunits.


Assuntos
Acetolactato Sintase/metabolismo , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/metabolismo , Escherichia coli/enzimologia , Cetol-Ácido Redutoisomerase/metabolismo , Subunidades Proteicas/metabolismo , Acetolactato Sintase/química , Acetolactato Sintase/genética , Acetolactato Sintase/fisiologia , Proteínas de Bactérias/química , Proteínas de Bactérias/fisiologia , Domínio Catalítico/genética , Escherichia coli/genética , Proteínas de Escherichia coli/antagonistas & inibidores , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/fisiologia , Isoenzimas/antagonistas & inibidores , Isoenzimas/química , Isoenzimas/genética , Isoenzimas/metabolismo , Cetol-Ácido Redutoisomerase/antagonistas & inibidores , Cetol-Ácido Redutoisomerase/química , Cetol-Ácido Redutoisomerase/genética , Peso Molecular , Subunidades Proteicas/química , Subunidades Proteicas/genética , Deleção de Sequência/genética , Valina/química , Valina/fisiologia
20.
J Mol Biol ; 389(1): 167-82, 2009 May 29.
Artigo em Inglês | MEDLINE | ID: mdl-19362563

RESUMO

Ketol-acid reductoisomerase (KARI; EC 1.1.1.86) is an enzyme in the branched-chain amino acid biosynthesis pathway where it catalyzes the conversion of 2-acetolactate into (2R)-2,3-dihydroxy-3-isovalerate or the conversion of 2-aceto-2-hydroxybutyrate into (2R,3R)-2,3-dihydroxy-3-methylvalerate. KARI catalyzes two reactions-alkyl migration and reduction-and requires Mg(2+) and NADPH for activity. To date, the only reported structures for a plant KARI are those of the spinach enzyme-Mn(2+)-(phospho)ADP ribose-(2R,3R)-2,3-dihydroxy-3-methylvalerate complex and the spinach KARI-Mg(2)(+)-NADPH-N-hydroxy-N-isopropyloxamate complex, where N-hydroxy-N-isopropyloxamate is a predicted transition-state analog. These studies demonstrated that the enzyme consists of two domains, N-domain and C-domain, with the active site at the interface of these domains. Here, we have determined the structures of the rice KARI-Mg(2+) and rice KARI-Mg(2)(+)-NADPH complexes to 1.55 A and 2.80 A resolutions, respectively. In comparing the structures of all the complexes, several differences are observed. Firstly, the N-domain is rotated up to 15 degrees relative to the C-domain, expanding the active site by up to 4 A. Secondly, an alpha-helix in the C-domain that includes residues V510-T519 and forms part of the active site moves by approximately 3.9 A upon binding of NADPH. Thirdly, the 15 C-terminal amino acid residues in the rice KARI-Mg(2+) complex are disordered. In the rice KARI-Mg(2)(+)-NADPH complex and the spinach KARI structures, many of the 15 residues bind to NADPH and the N-domain and cover the active site. Fourthly, the location of the metal ions within the active site can vary by up to 2.7 A. The new structures allow us to propose that an induced-fit mechanism operates to (i) allow substrate to enter the active site, (ii) close over the active site during catalysis, and (iii) open the active site to facilitate product release.


Assuntos
Cetol-Ácido Redutoisomerase/química , Magnésio/metabolismo , NADP/metabolismo , Oryza/enzimologia , Sequência de Aminoácidos , Biocatálise , Domínio Catalítico , Cristalografia por Raios X , Desenho de Fármacos , Escherichia coli/enzimologia , Herbicidas/química , Ligação de Hidrogênio , Ligantes , Modelos Moleculares , Dados de Sequência Molecular , Multimerização Proteica , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Subunidades Proteicas/química , Pseudomonas aeruginosa/enzimologia , Alinhamento de Sequência , Soluções
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...