Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
1.
Nat Chem Biol ; 14(7): 696-705, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29867142

RESUMEN

Colocation of the genes encoding ABC, TRAP, and TCT transport systems and catabolic pathways for the transported ligand provides a strategy for discovering novel microbial enzymes and pathways. We screened solute-binding proteins (SBPs) for ABC transport systems and identified three that bind D-apiose, a branched pentose in the cell walls of higher plants. Guided by sequence similarity networks (SSNs) and genome neighborhood networks (GNNs), the identities of the SBPs enabled the discovery of four catabolic pathways for D-apiose with eleven previously unknown reactions. The new enzymes include D-apionate oxidoisomerase, which catalyzes hydroxymethyl group migration, as well as 3-oxo-isoapionate-4-phosphate decarboxylase and 3-oxo-isoapionate-4-phosphate transcarboxylase/hydrolase, which are RuBisCO-like proteins (RLPs). The web tools for generating SSNs and GNNs are publicly accessible ( http://efi.igb.illinois.edu/efi-est/ ), so similar 'genomic enzymology' strategies for discovering novel pathways can be used by the community.


Asunto(s)
Pentosas/metabolismo , Biocatálisis , Humanos , Isomerasas/genética , Isomerasas/metabolismo , Modelos Moleculares , Pentosas/química
2.
J Bacteriol ; 201(2)2019 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-30249705

RESUMEN

We used comparative genomics to reconstruct d-galacturonic and d-glucuronic acid catabolic pathways and associated transcriptional regulons involving the tripartite ATP-independent periplasmic (TRAP) family transporters that bind hexuronates in proteobacteria. The reconstructed catabolic network involves novel transcription factors, catabolic enzymes, and transporters for utilization of both hexuronates and aldarates (d-glucarate and meso-galactarate). The reconstructed regulons for a novel GntR family transcription factor, GguR, include the majority of hexuronate/aldarate utilization genes in 47 species from the Burkholderiaceae, Comamonadaceae, Halomonadaceae, and Pseudomonadaceae families. GudR, GulR, and UdhR are additional local regulators of some hexuronate/aldarate utilization genes in some of the above-mentioned organisms. The predicted DNA binding motifs of GguR and GudR regulators from Ralstonia pickettii and Polaromonas were validated by in vitro binding assays. Genes from the GulR- and GguR-controlled loci were differentially expressed in R. pickettii grown on hexuronates and aldarates. By a combination of bioinformatics and experimental techniques we identified a novel variant of the oxidative pathway for hexuronate utilization, including two previously uncharacterized subfamilies of lactone hydrolases (UxuL and UxuF). The genomic context of respective genes and reconstruction of associated pathways suggest that both enzymes catalyze the conversion of d-galactaro- and d-glucaro-1,5-lactones to the ring-opened aldarates. The activities of the purified recombinant enzymes, UxuL and UxuF, from four proteobacterial species were directly confirmed and kinetically characterized. The inferred novel aldarate-specific transporter from the tripartite tricarboxylate transporter (TTT) family transporter TctC was confirmed to bind d-glucarate in vitro This study expands our knowledge of bacterial carbohydrate catabolic pathways by identifying novel families of catabolic enzymes, transcriptional regulators, and transporters.IMPORTANCE Hexuronate catabolic pathways and their transcriptional networks are highly variable among different bacteria. We identified novel transcriptional regulators that control the hexuronate and aldarate utilization genes in four families of proteobacteria. By regulon reconstruction and genome context analysis we identified several novel components of the common hexuronate/aldarate utilization pathways, including novel uptake transporters and catabolic enzymes. Two novel families of lactonases involved in the oxidative pathway of hexuronate catabolism were characterized. Novel transcriptional regulons were validated via in vitro binding assays and gene expression studies with Polaromonas and Ralstonia species. The reconstructed catabolic pathways are interconnected with each other metabolically and coregulated via the GguR regulons in proteobacteria.


Asunto(s)
Biología Computacional/métodos , Ácidos Hexurónicos/metabolismo , Redes y Vías Metabólicas/genética , Proteobacteria/genética , Proteobacteria/metabolismo , Regulación Bacteriana de la Expresión Génica , Genómica , Regulón , Transcripción Genética
3.
Nature ; 502(7473): 698-702, 2013 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-24056934

RESUMEN

Assigning valid functions to proteins identified in genome projects is challenging: overprediction and database annotation errors are the principal concerns. We and others are developing computation-guided strategies for functional discovery with 'metabolite docking' to experimentally derived or homology-based three-dimensional structures. Bacterial metabolic pathways often are encoded by 'genome neighbourhoods' (gene clusters and/or operons), which can provide important clues for functional assignment. We recently demonstrated the synergy of docking and pathway context by 'predicting' the intermediates in the glycolytic pathway in Escherichia coli. Metabolite docking to multiple binding proteins and enzymes in the same pathway increases the reliability of in silico predictions of substrate specificities because the pathway intermediates are structurally similar. Here we report that structure-guided approaches for predicting the substrate specificities of several enzymes encoded by a bacterial gene cluster allowed the correct prediction of the in vitro activity of a structurally characterized enzyme of unknown function (PDB 2PMQ), 2-epimerization of trans-4-hydroxy-L-proline betaine (tHyp-B) and cis-4-hydroxy-D-proline betaine (cHyp-B), and also the correct identification of the catabolic pathway in which Hyp-B 2-epimerase participates. The substrate-liganded pose predicted by virtual library screening (docking) was confirmed experimentally. The enzymatic activities in the predicted pathway were confirmed by in vitro assays and genetic analyses; the intermediates were identified by metabolomics; and repression of the genes encoding the pathway by high salt concentrations was established by transcriptomics, confirming the osmolyte role of tHyp-B. This study establishes the utility of structure-guided functional predictions to enable the discovery of new metabolic pathways.


Asunto(s)
Bacterias , Enzimas/química , Enzimas/genética , Genoma Bacteriano/genética , Redes y Vías Metabólicas/genética , Anotación de Secuencia Molecular/métodos , Homología Estructural de Proteína , Bacterias/enzimología , Bacterias/genética , Bacterias/metabolismo , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Enzimas/metabolismo , Perfilación de la Expresión Génica , Genes Bacterianos/genética , Glucólisis , Cinética , Metabolismo , Metabolómica , Modelos Moleculares , Familia de Multigenes/genética , Operón , Especificidad por Sustrato
4.
Nucleic Acids Res ; 45(7): 3785-3799, 2017 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-28073944

RESUMEN

Riboflavin (vitamin B2) is the precursor of flavin mononucleotide (FMN) and flavin adenine dinucleotide, which are essential coenzymes in all free-living organisms. Riboflavin biosynthesis in many Bacteria but not in Archaea is controlled by FMN-responsive riboswitches. We identified a novel bifunctional riboflavin kinase/regulator (RbkR), which controls riboflavin biosynthesis and transport genes in major lineages of Crenarchaeota, Euryarchaeota and Thaumarchaeota. RbkR proteins are composed of the riboflavin kinase domain and a DNA-binding winged helix-turn-helix-like domain. Using comparative genomics, we predicted RbkR operator sites and reconstructed RbkR regulons in 94 archaeal genomes. While the identified RbkR operators showed significant variability between archaeal lineages, the conserved core of RbkR regulons includes riboflavin biosynthesis genes, known/predicted vitamin uptake transporters and the rbkR gene. The DNA motifs and CTP-dependent riboflavin kinase activity of two RbkR proteins were experimentally validated in vitro. The DNA binding activity of RbkR was stimulated by CTP and suppressed by FMN, a product of riboflavin kinase. The crystallographic structure of RbkR from Thermoplasma acidophilum was determined in complex with CTP and its DNA operator revealing key residues for operator and ligand recognition. Overall, this study contributes to our understanding of metabolic and regulatory networks for vitamin homeostasis in Archaea.


Asunto(s)
Archaea/genética , Proteínas Arqueales/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Riboflavina/metabolismo , Factores de Transcripción/metabolismo , Archaea/enzimología , Archaea/metabolismo , Proteínas Arqueales/química , ADN de Archaea/química , ADN de Archaea/metabolismo , Evolución Molecular , Genoma Arqueal , Regiones Operadoras Genéticas , Fosfotransferasas (Aceptor de Grupo Alcohol)/química , Dominios Proteicos , Regulón , Factores de Transcripción/química
5.
Proc Natl Acad Sci U S A ; 113(29): E4161-9, 2016 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-27402745

RESUMEN

Using a large-scale "genomic enzymology" approach, we (i) assigned novel ATP-dependent four-carbon acid sugar kinase functions to members of the DUF1537 protein family (domain of unknown function; Pfam families PF07005 and PF17042) and (ii) discovered novel catabolic pathways for d-threonate, l-threonate, and d-erythronate. The experimentally determined ligand specificities of several solute binding proteins (SBPs) for TRAP (tripartite ATP-independent permease) transporters for four-carbon acids, including d-erythronate and l-erythronate, were used to constrain the substrates for the catabolic pathways that degrade the SBP ligands to intermediates in central carbon metabolism. Sequence similarity networks and genome neighborhood networks were used to identify the enzyme components of the pathways. Conserved genome neighborhoods encoded SBPs as well as permease components of the TRAP transporters, members of the DUF1537 family, and a member of the 4-hydroxy-l-threonine 4-phosphate dehydrogenase (PdxA) oxidative decarboxylase, class II aldolase, or ribulose 1,5-bisphosphate carboxylase/oxygenase, large subunit (RuBisCO) superfamily. Because the characterized substrates of members of the PdxA, class II aldolase, and RuBisCO superfamilies are phosphorylated, we postulated that the members of the DUF1537 family are novel ATP-dependent kinases that participate in catabolic pathways for four-carbon acid sugars. We determined that (i) the DUF1537/PdxA pair participates in a pathway for the conversion of d-threonate to dihydroxyacetone phosphate and CO2 and (ii) the DUF1537/class II aldolase pair participates in pathways for the conversion of d-erythronate and l-threonate (epimers at carbon-3) to dihydroxyacetone phosphate and CO2 The physiological importance of these pathways was demonstrated in vivo by phenotypic and genetic analyses.


Asunto(s)
Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Proteínas Quinasas/química , Proteínas Quinasas/metabolismo , Bacterias/enzimología , Bacterias/aislamiento & purificación , Butiratos/metabolismo , Fructosa-Bifosfato Aldolasa/metabolismo , Oxidorreductasas/metabolismo , Fosfatos/metabolismo , Dominios Proteicos
6.
PLoS Biol ; 12(4): e1001843, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24756107

RESUMEN

The cytosolic glutathione transferase (cytGST) superfamily comprises more than 13,000 nonredundant sequences found throughout the biosphere. Their key roles in metabolism and defense against oxidative damage have led to thousands of studies over several decades. Despite this attention, little is known about the physiological reactions they catalyze and most of the substrates used to assay cytGSTs are synthetic compounds. A deeper understanding of relationships across the superfamily could provide new clues about their functions. To establish a foundation for expanded classification of cytGSTs, we generated similarity-based subgroupings for the entire superfamily. Using the resulting sequence similarity networks, we chose targets that broadly covered unknown functions and report here experimental results confirming GST-like activity for 82 of them, along with 37 new 3D structures determined for 27 targets. These new data, along with experimentally known GST reactions and structures reported in the literature, were painted onto the networks to generate a global view of their sequence-structure-function relationships. The results show how proteins of both known and unknown function relate to each other across the entire superfamily and reveal that the great majority of cytGSTs have not been experimentally characterized or annotated by canonical class. A mapping of taxonomic classes across the superfamily indicates that many taxa are represented in each subgroup and highlights challenges for classification of superfamily sequences into functionally relevant classes. Experimental determination of disulfide bond reductase activity in many diverse subgroups illustrate a theme common for many reaction types. Finally, sequence comparison between an enzyme that catalyzes a reductive dechlorination reaction relevant to bioremediation efforts with some of its closest homologs reveals differences among them likely to be associated with evolution of this unusual reaction. Interactive versions of the networks, associated with functional and other types of information, can be downloaded from the Structure-Function Linkage Database (SFLD; http://sfld.rbvi.ucsf.edu).


Asunto(s)
Glutatión Transferasa/genética , Glutatión Transferasa/ultraestructura , Modelos Moleculares , Secuencia de Aminoácidos , Secuencia de Bases , Sitios de Unión , Biología Computacional , Bases de Datos de Proteínas , Glutatión/química , Estructura Terciaria de Proteína , Alineación de Secuencia , Relación Estructura-Actividad
7.
J Biol Chem ; 290(48): 28963-76, 2015 Nov 27.
Artículo en Inglés | MEDLINE | ID: mdl-26472925

RESUMEN

Innovations in the discovery of the functions of uncharacterized proteins/enzymes have become increasingly important as advances in sequencing technology flood protein databases with an exponentially growing number of open reading frames. This study documents one such innovation developed by the Enzyme Function Initiative (EFI; U54GM093342), the use of solute-binding proteins for transport systems to identify novel metabolic pathways. In a previous study, this strategy was applied to the tripartite ATP-independent periplasmic transporters. Here, we apply this strategy to the ATP-binding cassette transporters and report the discovery of novel catabolic pathways for d-altritol and galactitol in Agrobacterium tumefaciens C58. These efforts resulted in the description of three novel enzymatic reactions as follows: 1) oxidation of d-altritol to d-tagatose via a dehydrogenase in Pfam family PF00107, a previously unknown reaction; 2) phosphorylation of d-tagatose to d-tagatose 6-phosphate via a kinase in Pfam family PF00294, a previously orphan EC number; and 3) epimerization of d-tagatose 6-phosphate C-4 to d-fructose 6-phosphate via a member of Pfam family PF08013, another previously unknown reaction. The epimerization reaction catalyzed by a member of PF08013 is especially noteworthy, because the functions of members of PF08013 have been unknown. These discoveries were assisted by the following two synergistic bioinformatics web tools made available by the Enzyme Function Initiative: the EFI-Enzyme Similarity Tool and the EFI-Genome Neighborhood Tool.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/metabolismo , Agrobacterium tumefaciens/metabolismo , Proteínas Bacterianas/metabolismo , Galactitol/metabolismo , Alcoholes del Azúcar/metabolismo , Transportadoras de Casetes de Unión a ATP/genética , Agrobacterium tumefaciens/genética , Proteínas Bacterianas/genética , Galactitol/genética
8.
Biochemistry ; 54(3): 909-31, 2015 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-25540822

RESUMEN

The rate at which genome sequencing data is accruing demands enhanced methods for functional annotation and metabolism discovery. Solute binding proteins (SBPs) facilitate the transport of the first reactant in a metabolic pathway, thereby constraining the regions of chemical space and the chemistries that must be considered for pathway reconstruction. We describe high-throughput protein production and differential scanning fluorimetry platforms, which enabled the screening of 158 SBPs against a 189 component library specifically tailored for this class of proteins. Like all screening efforts, this approach is limited by the practical constraints imposed by construction of the library, i.e., we can study only those metabolites that are known to exist and which can be made in sufficient quantities for experimentation. To move beyond these inherent limitations, we illustrate the promise of crystallographic- and mass spectrometric-based approaches for the unbiased use of entire metabolomes as screening libraries. Together, our approaches identified 40 new SBP ligands, generated experiment-based annotations for 2084 SBPs in 71 isofunctional clusters, and defined numerous metabolic pathways, including novel catabolic pathways for the utilization of ethanolamine as sole nitrogen source and the use of d-Ala-d-Ala as sole carbon source. These efforts begin to define an integrated strategy for realizing the full value of amassing genome sequence data.


Asunto(s)
Proteínas Portadoras/metabolismo , Redes y Vías Metabólicas , Metaboloma , Metabolómica/métodos , Anotación de Secuencia Molecular , Bacillus/metabolismo , Carbohidratos/química , Clonación Molecular , Cristalografía por Rayos X , Fluorometría , Cinética , Ligandos , Reproducibilidad de los Resultados , Homología de Secuencia de Aminoácido
9.
J Am Chem Soc ; 137(4): 1388-91, 2015 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-25608448

RESUMEN

The genome of Labrenzia aggregata IAM 12614 encodes an uncharacterized member of the muconate lactonizing enzyme (MLE) subgroup of the enolase superfamily (UniProt ID A0NXQ8 ). The gene encoding A0NXQ8 is located between genes that encode members of the proline racemase superfamily, 4R-hydroxyproline 2-epimerase (UniProt ID A0NXQ7 ; 4HypE) and trans-3-hydroxy-l-proline dehydratase (UniProt ID A0NXQ9 ; t3LHypD). A0NXQ8 was screened with a library of proline analogues; two reactions were observed with cis-3-hydroxy-l-proline (c3LHyp), competing 2-epimerization to trans-3-hydroxy-d-proline (1,1-proton transfer) and dehydration to Δ(1)-pyrroline-2-carboxylate (ß-elimination; c3LHyp dehydratase), with eventual total dehydration. The genome context encoding A0NXQ8 both (1) confirms its novel c3LHyp dehydratase function and (2) provides evidence for metabolic pathways that allow L. aggregata to utilize several isomeric 3- and 4-hydroxyprolines as sole carbon sources.


Asunto(s)
Hidroliasas/metabolismo , Hidroxiprolina/metabolismo , Rhodobacteraceae/enzimología , Isomerasas de Aminoácido/genética , Isomerasas de Aminoácido/metabolismo , Genoma Bacteriano , Hidroliasas/genética , Hidroxiprolina/química , Modelos Moleculares , Rhodobacteraceae/genética , Rhodobacteraceae/metabolismo , Especificidad por Sustrato
10.
J Am Chem Soc ; 137(46): 14570-3, 2015 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-26560079

RESUMEN

We describe a general integrated bioinformatic and experimental strategy to discover the in vitro enzymatic activities and in vivo functions (metabolic pathways) of uncharacterized enzymes discovered in microbial genome projects using the ligand specificities of the solute binding proteins (SBPs) for ABC transporters. Using differential scanning fluorimetry, we determined that the SBP for an ABC transporter encoded by the genome of Mycobacterium smegmatis is stabilized by d-threitol. Using sequence similarity networks and genome neighborhood networks to guide selection of target proteins for pathway enzymes, we applied both in vitro and in vivo experimental approaches to discover novel pathways for catabolism of d-threitol, l-threitol, and erythritol.


Asunto(s)
Eritritol/metabolismo , Mycobacterium smegmatis/metabolismo , Alcoholes del Azúcar/metabolismo , Estereoisomerismo
11.
Proc Natl Acad Sci U S A ; 109(11): 4122-7, 2012 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-22392983

RESUMEN

The rapid advance in genome sequencing presents substantial challenges for protein functional assignment, with half or more of new protein sequences inferred from these genomes having uncertain assignments. The assignment of enzyme function in functionally diverse superfamilies represents a particular challenge, which we address through a combination of computational predictions, enzymology, and structural biology. Here we describe the results of a focused investigation of a group of enzymes in the enolase superfamily that are involved in epimerizing dipeptides. The first members of this group to be functionally characterized were Ala-Glu epimerases in Eschericiha coli and Bacillus subtilis, based on the operon context and enzymological studies; these enzymes are presumed to be involved in peptidoglycan recycling. We have subsequently studied more than 65 related enzymes by computational methods, including homology modeling and metabolite docking, which suggested that many would have divergent specificities;, i.e., they are likely to have different (unknown) biological roles. In addition to the Ala-Phe epimerase specificity reported previously, we describe the prediction and experimental verification of: (i) a new group of presumed Ala-Glu epimerases; (ii) several enzymes with specificity for hydrophobic dipeptides, including one from Cytophaga hutchinsonii that epimerizes D-Ala-D-Ala; and (iii) a small group of enzymes that epimerize cationic dipeptides. Crystal structures for certain of these enzymes further elucidate the structural basis of the specificities. The results highlight the potential of computational methods to guide experimental characterization of enzymes in an automated, large-scale fashion.


Asunto(s)
Dipéptidos/metabolismo , Familia de Multigenes , Fosfopiruvato Hidratasa/metabolismo , Racemasas y Epimerasas/metabolismo , Homología de Secuencia de Aminoácido , Dominio Catalítico , Cationes , Análisis por Conglomerados , Biología Computacional , Cristalografía por Rayos X , Interacciones Hidrofóbicas e Hidrofílicas , Cinética , Modelos Moleculares , Racemasas y Epimerasas/química , Especificidad por Sustrato
12.
Biochemistry ; 53(25): 4192-203, 2014 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-24926996

RESUMEN

The genome of Agrobacterium tumefaciens C58 encodes 12 members of the enolase superfamily (ENS), eight of which are members of the mandelate racemase (MR) subgroup and, therefore, likely to be acid sugar dehydratases. Using a library of 77 acid sugars for high-throughput screening, one protein (UniProt entry A9CG74; locus tag Atu4196) showed activity with both m-galactarate and d-galacturonate. Two families of galactarate dehydratases had been discovered previously in the ENS, GalrD/TalrD [Yew, W. S., et al. (2007) Biochemistry 46, 9564-9577] and GalrD-II [Rakus, J. F., et al. (2009) Biochemistry 48, 11546-11558]; these have different active site acid/base catalysis and have no activity with d-galacturonate. A9CG74 dehydrates m-galactarate to form 2-keto-3-deoxy-galactarate but does not dehydrate d-galacturonate as expected. Instead, when A9CG74 is incubated with d-galacturonate, 3-deoxy-d-xylo-hexarate or 3-deoxy-d-lyxo-hexarate is formed. In this reaction, instead of abstracting the C5 proton α to the carboxylate group, the expected reaction for a member of the ENS, the enzyme apparently abstracts the proton α to the aldehyde group to form 3-deoxy-d-threo-hexulosuronate that undergoes a 1,2-hydride shift similar to the benzylic acid rearrangement to form the observed product. A. tumefaciens C58 does not utilize m-galactarate as a carbon source under the conditions tested in this study, although it does utilize d-galacturonate, which is a likely precursor to m-galactarate. The gene encoding A9CG74 and several genome proximal genes were upregulated with d-galacturonate as the carbon source. One of these, a member of the dihydrodipicolinate synthase superfamily, catalyzes the dehydration and subsequent decarboxylation of 2-keto-3-deoxy-d-galactarate to α-ketoglutarate semialdehyde, thereby providing a pathway for the conversion of m-galactarate to α-ketoglutarate semialdehyde.


Asunto(s)
Agrobacterium tumefaciens/enzimología , Proteínas Bacterianas/metabolismo , Hidroliasas/metabolismo , Azúcares Ácidos/metabolismo , Agrobacterium tumefaciens/genética , Secuencia de Aminoácidos , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Cristalografía por Rayos X , Genoma Bacteriano , Ácidos Hexurónicos/metabolismo , Hidroliasas/química , Hidroliasas/genética , Simulación del Acoplamiento Molecular , Datos de Secuencia Molecular , Mutación , Estereoisomerismo
13.
Biochemistry ; 53(16): 2722-31, 2014 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-24697546

RESUMEN

The continued increase in the size of the protein sequence databases as a result of advances in genome sequencing technology is overwhelming the ability to perform experimental characterization of function. Consequently, functions are assigned to the vast majority of proteins via automated, homology-based methods, with the result that as many as 50% are incorrectly annotated or unannotated ( Schnoes et al. PLoS Comput. Biol. 2009 , 5 ( 12 ), e1000605 ). This manuscript describes a study of the D-mannonate dehydratase (ManD) subgroup of the enolase superfamily (ENS) to investigate how function diverges as sequence diverges. Previously, one member of the subgroup had been experimentally characterized as ManD [dehydration of D-mannonate to 2-keto-3-deoxy-D-mannonate (equivalently, 2-keto-3-deoxy-D-gluconate)]. In this study, 42 additional members were characterized to sample sequence-function space in the ManD subgroup. These were found to differ in both catalytic efficiency and substrate specificity: (1) high efficiency (kcat/KM = 10(3) to 10(4) M(-1) s(-1)) for dehydration of D-mannonate, (2) low efficiency (kcat/KM = 10(1) to 10(2) M(-1) s(-1)) for dehydration of d-mannonate and/or D-gluconate, and 3) no-activity with either D-mannonate or D-gluconate (or any other acid sugar tested). Thus, the ManD subgroup is not isofunctional and includes D-gluconate dehydratases (GlcDs) that are divergent from the GlcDs that have been characterized in the mandelate racemase subgroup of the ENS (Lamble et al. FEBS Lett. 2004 , 576 , 133 - 136 ) (Ahmed et al. Biochem. J. 2005 , 390 , 529 - 540 ). These observations signal caution for functional assignment based on sequence homology and lay the foundation for the studies of the physiological functions of the GlcDs and the promiscuous ManDs/GlcDs.


Asunto(s)
Hidroliasas/química , Hidroliasas/metabolismo , Fosfopiruvato Hidratasa/metabolismo , Dominio Catalítico , Cristalografía por Rayos X , Gluconatos/metabolismo , Hidroliasas/genética , Cinética , Datos de Secuencia Molecular , Mutación , Fosfopiruvato Hidratasa/química , Conformación Proteica , Relación Estructura-Actividad , Especificidad por Sustrato , Azúcares Ácidos/metabolismo
14.
J Biol Chem ; 286(28): 25265-73, 2011 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-21597116

RESUMEN

QnrB1 is a plasmid-encoded pentapeptide repeat protein (PRP) that confers a moderate degree of resistance to fluoroquinolones. Its gene was cloned into an expression vector with an N-terminal polyhistidine tag, and the protein was purified by nickel affinity chromatography. The structure of QnrB1 was determined by a combination of trypsinolysis, surface mutagenesis, and single anomalous dispersion phasing. QnrB1 folds as a right-handed quadrilateral ß-helix with a highly asymmetric dimeric structure typical of PRP-topoisomerase poison resistance factors. The threading of pentapeptides into the ß-helical fold is interrupted by two noncanonical PRP sequences that produce outward projecting loops that interrupt the regularity of the PRP surface. Deletion of the larger upper loop eliminated the protective effect of QnrB1 on DNA gyrase toward inhibition by quinolones, whereas deletion of the smaller lower loop drastically reduced the protective effect. These loops are conserved among all plasmid-based Qnr variants (QnrA, QnrC, QnrD, and QnrS) and some chromosomally encoded Qnr varieties. A mechanism in which PRP-topoisomerase poison resistance factors bind to and disrupt the quinolone-DNA-gyrase interaction is proposed.


Asunto(s)
Proteínas Bacterianas/química , Farmacorresistencia Bacteriana/fisiología , Enterococcus faecalis/química , Fluoroquinolonas/química , Plásmidos , Secuencia de Aminoácidos , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Girasa de ADN/química , Girasa de ADN/genética , Girasa de ADN/metabolismo , Enterococcus faecalis/genética , Enterococcus faecalis/metabolismo , Fluoroquinolonas/farmacología , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Eliminación de Secuencia , Relación Estructura-Actividad
15.
Nat Chem Biol ; 6(11): 797-9, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20852636

RESUMEN

The Mycobacterium tuberculosis enzyme Rv2275 catalyzes the formation of cyclo(L-Tyr-L-Tyr) using two molecules of Tyr-tRNA(Tyr) as substrates. The three-dimensional (3D) structure of Rv2275 was determined to 2.0-Å resolution, revealing that Rv2275 is structurally related to the class Ic aminoacyl-tRNA synthetase family of enzymes. Mutagenesis and radioactive labeling suggests a covalent intermediate in which L-tyrosine is transferred from Tyr-tRNA(Tyr) to an active site serine (Ser88) by transesterification with Glu233 serving as a critical base, catalyzing dipeptide bond formation.


Asunto(s)
Dipéptidos/biosíntesis , Mycobacterium tuberculosis/enzimología , Péptido Sintasas/química , Péptido Sintasas/metabolismo , Péptidos Cíclicos/biosíntesis , Tirosina-ARNt Ligasa/química , Biocatálisis , Dominio Catalítico , Cristalografía por Rayos X , Ciclización , Esterificación , Ácido Glutámico/química , Ácido Glutámico/metabolismo , Modelos Moleculares , Conformación Proteica , ARN de Transferencia de Tirosina/química , ARN de Transferencia de Tirosina/metabolismo , Serina/genética , Serina/metabolismo , Tirosina/química , Tirosina/metabolismo
17.
Antimicrob Agents Chemother ; 55(1): 110-7, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20937785

RESUMEN

The chromosomally encoded Qnr homolog protein from Enterococcus faecalis (EfsQnr), when expressed, confers to its host a decreased susceptibility to quinolones and consists mainly of tandem repeats, which is consistent with belonging to the pentapeptide repeat family of proteins (PRPs). EfsQnr was cloned with an N-terminal 6× His tag and purified to homogeneity. EfsQnr partially protected DNA gyrase from fluoroquinolone inhibition at concentrations as low as 20 nM. EfsQnr inhibited the ATP-dependent supercoiling activity of DNA gyrase with a 50% inhibitory concentration (IC(50)) of 1.2 µM, while no significant inhibition of ATP-independent relaxation activity was observed. EfsQnr was cytotoxic when overexpressed in Escherichia coli, resulting in the clumping of cells and a loss of viability. The X-ray crystal structure of EfsQnr was determined to 1.6-Å resolution. EfsQnr exhibits the right-handed quadrilateral beta-helical fold typical of PRPs, with features more analogous to MfpA (mycobacterium fluoroquinolone resistance pentapeptide) than to the PRPs commonly found in cyanobacteria.


Asunto(s)
Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/farmacología , Enterococcus faecalis/metabolismo , Inhibidores de Topoisomerasa II , Secuencia de Aminoácidos , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Enterococcus faecalis/genética , Escherichia coli/citología , Escherichia coli/genética , Escherichia coli/metabolismo , Escherichia coli/ultraestructura , Viabilidad Microbiana , Microscopía Electrónica de Rastreo , Datos de Secuencia Molecular , Multimerización de Proteína , Estructura Secundaria de Proteína , Homología de Secuencia de Aminoácido
18.
Nat Struct Mol Biol ; 13(5): 408-13, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16648861

RESUMEN

Isoniazid is a key drug used in the treatment of tuberculosis. Isoniazid is a pro-drug, which, after activation by the katG-encoded catalase peroxidase, reacts nonenzymatically with NAD(+) and NADP(+) to generate several isonicotinoyl adducts of these pyridine nucleotides. One of these, the acyclic 4S isomer of isoniazid-NAD, targets the inhA-encoded enoyl-ACP reductase, an enzyme essential for mycolic acid biosynthesis in Mycobacterium tuberculosis. Here we show that the acyclic 4R isomer of isoniazid-NADP inhibits the M. tuberculosis dihydrofolate reductase (DHFR), an enzyme essential for nucleic acid synthesis. This biologically relevant form of the isoniazid adduct is a subnanomolar bisubstrate inhibitor of M. tuberculosis DHFR. Expression of M. tuberculosis DHFR in Mycobacterium smegmatis mc(2)155 protects cells against growth inhibition by isoniazid by sequestering the drug. Thus, M. tuberculosis DHFR is the first new target for isoniazid identified in the last decade.


Asunto(s)
Antagonistas del Ácido Fólico/farmacología , Isoniazida/farmacología , Mycobacterium tuberculosis/efectos de los fármacos , Mycobacterium tuberculosis/enzimología , Tetrahidrofolato Deshidrogenasa/metabolismo , Proliferación Celular , Clonación Molecular , Cristalografía por Rayos X , Antagonistas del Ácido Fólico/química , Antagonistas del Ácido Fólico/metabolismo , Expresión Génica , Isoniazida/química , Isoniazida/metabolismo , Modelos Moleculares , Mycobacterium tuberculosis/citología , Mycobacterium tuberculosis/genética , NADP/química , NADP/metabolismo , Profármacos/química , Profármacos/metabolismo , Profármacos/farmacología , Unión Proteica , Estructura Terciaria de Proteína , Tetrahidrofolato Deshidrogenasa/genética , Tetrahidrofolato Deshidrogenasa/aislamiento & purificación
19.
Acta Crystallogr Sect F Struct Biol Cryst Commun ; 67(Pt 3): 296-302, 2011 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-21393830

RESUMEN

The protein AlbG is a self-resistance factor against albicidin, a nonribosomally encoded hybrid polyketide-peptide with antibiotic and phytotoxic properties produced by Xanthomonas albilineans. Primary-sequence analysis indicates that AlbG is a member of the pentapeptide-repeat family of proteins (PRP). The structure of AlbG from X. albilineans was determined at 2.0 Šresolution by SAD phasing using data collected from a single trimethyllead acetate derivative on a home source. AlbG folds into a right-handed quadrilateral ß-helix composed of approximately eight semi-regular coils. The regularity of the ß-helix is blemished by a large loop/deviation in the ß-helix between coils 4 and 5. The C-terminus of the ß-helix is capped by a dimerization module, yielding a dimer with a 110 Šsemi-collinear ß-helical axis. This method of dimer formation appears to be common to all PRP proteins that confer resistance to topoisomerase poisons and contrasts with most PRP proteins, which are typically monomeric.


Asunto(s)
Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Oligopéptidos/química , Oligopéptidos/genética , Estructura Secundaria de Proteína , Factores R/química , Factores R/genética , Secuencia de Aminoácidos , Proteínas Bacterianas/metabolismo , Modelos Moleculares , Datos de Secuencia Molecular , Oligopéptidos/metabolismo , Compuestos Orgánicos/química , Compuestos Orgánicos/metabolismo , Multimerización de Proteína , Factores R/metabolismo , Xanthomonas/química , Xanthomonas/genética , Xanthomonas/metabolismo
20.
Biochemistry ; 48(27): 6461-8, 2009 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-19485344

RESUMEN

The galacto-, homoserine-, mevalonate-, phosphomevalonate-kinase (GHMP) superfamily encompases a wide-range of protein function. Three members of the family (mevalonate kinase, phosphomevalonate kinase, and diphosphomevalonate decarboxylase) comprise the mevalonate pathway found in S. pneumoniae and other organisms. We have determined the 1.9 A crystal structure of phosphomevalonate kinase (PMK) from S. pneumoniae in complex with phosphomevalonate and AMPPNP.Mg(2+). Comparison of the apo and ternary PMK structures suggests that ligand binding reverses the side-chain orientations of two antiparallel lysines residues (100 and 101) with the result that Lys101 is switched into a position in which its ammonium ion is in direct contact with the beta,gamma-bridging atom of the nucleotide, where it is expected to stabilize both the ground and transition states of the reaction. Analysis of all available GHMP kinase ternary complex structures reveals that while their C(alpha)-scaffolds are highly conserved, their substrates bind in one of two conformations, which appear to be either reactive or nonreactive. The active site of PMK seems spacious enough to accommodate interconversion of the reactive and nonreactive conformers. A substantial fraction of the PMK active site is occupied by ordered water, which clusters near the charged regions of the substrate. Notably, a water pentamer that interacts extensively with the reactive groups of both substrates was discovered at the active site.


Asunto(s)
Fosfotransferasas (Aceptor del Grupo Fosfato)/química , Secuencia de Aminoácidos , Dominio Catalítico , Cristalografía por Rayos X , Ligandos , Modelos Moleculares , Datos de Secuencia Molecular , Fosfotransferasas (Aceptor del Grupo Fosfato)/metabolismo , Conformación Proteica , Homología de Secuencia de Aminoácido , Solventes , Streptococcus pneumoniae/enzimología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA