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










Base de datos
Intervalo de año de publicación
1.
Philos Trans R Soc Lond B Biol Sci ; 378(1871): 20220035, 2023 02 27.
Artículo en Inglés | MEDLINE | ID: mdl-36633276

RESUMEN

Menaquinones (MKs) are electron carriers in bacterial respiratory chains. In Staphylococcus aureus (Sau), MKs are essential for aerobic and anaerobic respiration. As MKs are redox-active, their biosynthesis likely requires tight regulation to prevent disruption of cellular redox balance. We recently found that the Mycobacterium tuberculosis MenD, the first committed enzyme of the MK biosynthesis pathway, is allosterically inhibited by the downstream metabolite 1,4-dihydroxy-2-naphthoic acid (DHNA). To understand if this is a conserved mechanism in phylogenetically distant genera that also use MK, we investigated whether the Sau-MenD is allosterically inhibited by DHNA. Our results show that DHNA binds to and inhibits the SEPHCHC synthase activity of Sau-MenD enzymes. We identified residues in the DHNA binding pocket that are important for catalysis (Arg98, Lys283, Lys309) and inhibition (Arg98, Lys283). Furthermore, we showed that exogenous DHNA inhibits the growth of Sau, an effect that can be rescued by supplementing the growth medium with MK-4. Our results demonstrate that, despite a lack of strict conservation of the DHNA binding pocket between Mtb-MenD and Sau-MenD, feedback inhibition by DHNA is a conserved mechanism in Sau-MenD and hence the Sau MK biosynthesis pathway. These findings may have implications for the development of anti-staphylococcal agents targeting MK biosynthesis. This article is part of the theme issue 'Reactivity and mechanism in chemical and synthetic biology'.


Asunto(s)
Naftalenos , Staphylococcus aureus , Vitamina K 2/farmacología , Vitamina K 2/metabolismo , Staphylococcus aureus/metabolismo , Retroalimentación , Naftalenos/farmacología
2.
Acta Crystallogr F Struct Biol Commun ; 78(Pt 12): 416-422, 2022 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-36458621

RESUMEN

The 1.72 Šresolution structure of purine nucleoside phosphorylase from Geobacillus stearothermophilus, a thermostable protein of potential interest for the biocatalytic synthesis of antiviral nucleoside compounds, is reported. The structure of the N-terminally His-tagged enzyme is a hexamer, as is typical of bacterial homologues, with a trimer-of-dimers arrangement. Unexpectedly, several residues of the recombinant tobacco etch virus protease (rTEV) cleavage site from the N-terminal tag are located in the active site of the neighbouring subunit in the dimer. Key to this interaction is a tyrosine residue, which sits where the nucleoside ring of the substrate would normally be located. Tag binding appears to be driven by a combination of enthalpic, entropic and proximity effects, which convey a particularly high affinity in the crystallized form. Attempts to cleave the tag in solution yielded only a small fraction of untagged protein, suggesting that the enzyme predominantly exists in the tag-bound form in solution, preventing rTEV from accessing the cleavage site. However, the tagged protein retained some activity in solution, suggesting that the tag does not completely block the active site, but may act as a competitive inhibitor. This serves as a warning that it is prudent to establish how affinity tags may affect protein structure and function, especially for industrial biocatalytic applications that rely on the efficiency and convenience of one-pot purifications and in cases where tag removal is difficult.


Asunto(s)
Geobacillus stearothermophilus , Purina-Nucleósido Fosforilasa , Purina-Nucleósido Fosforilasa/genética , Nucleósidos , Cristalografía por Rayos X , Biocatálisis
3.
Biochem Biophys Res Commun ; 549: 194-199, 2021 04 16.
Artículo en Inglés | MEDLINE | ID: mdl-33721671

RESUMEN

Chemotaxis is the process of sensing chemical gradients and navigating towards favourable conditions. Bacterial chemotaxis is mediated by arrays of trans-membrane chemoreceptor proteins. The most common class of chemoreceptors have periplasmic ligand-binding domains (LBDs) that detect extracellular chemical signs and transduce these signals to the downstream chemotaxis machinery. The repertoire of chemoreceptor proteins in a bacterium determines the range of environmental signals to which it can respond. Pseudomonas syringae pv. actinidiae (Psa) is a plant pathogen which causes bacterial canker of kiwifruit (Actinidia sp.). Compared to many other bacteria, Psa has a large number of chemoreceptors encoded in its genome (43) and most of these remain uncharacterized. A previous study identified PscC as a potential chemoreceptor for l-proline and other amino acid ligands. Here, we have characterized the interaction of PscC-LBD with l-proline using a combination of isothermal titration calorimetry (ITC) and X-ray crystallography. ITC confirmed direct binding of l-proline to PscC-LBD with KD value of 5.0 µM. We determined the structure of PscC-LBD in complex with l-proline. Our structural analysis showed that PscC-LBD adopts similar double-CACHE fold to several other amino acid chemoreceptors. A comparison of the PscC-LDB to other dCACHE structures highlights residues in the binding cavity which contribute to its ligand specificity.


Asunto(s)
Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Células Quimiorreceptoras/metabolismo , Prolina/metabolismo , Pseudomonas syringae/metabolismo , Sitios de Unión , Calorimetría , Cristalografía por Rayos X , Ligandos , Modelos Moleculares , Dominios Proteicos
4.
Curr Opin Struct Biol ; 65: 33-41, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32634692

RESUMEN

Menaquinones (vitamin K2) are a family of redox-active small molecules with critical functions across all domains of life, including energy generation in bacteria and bone health in humans. The enzymes involved in menaquinone biosynthesis also have bioengineering applications and are potential antimicrobial drug targets. New insights into the essential roles of menaquinones, and their potential to cause redox-related toxicity, have highlighted the need for this pathway to be tightly controlled. Here, we provide an overview of our current understanding of the classical menaquinone biosynthesis pathway in bacteria. We also review recent discoveries on protein-level allostery and sublocalisation of membrane-bound enzymes that have provided insight into the regulation of flux through this biosynthetic pathway.


Asunto(s)
Bacterias/metabolismo , Vitamina K 2/metabolismo , Regulación Alostérica
5.
J Biol Chem ; 295(12): 3759-3770, 2020 03 20.
Artículo en Inglés | MEDLINE | ID: mdl-32029475

RESUMEN

Menaquinone (vitamin K2) plays a vital role in energy generation and environmental adaptation in many bacteria, including the human pathogen Mycobacterium tuberculosis (Mtb). Although menaquinone levels are known to be tightly linked to the cellular redox/energy status of the cell, the regulatory mechanisms underpinning this phenomenon are unclear. The first committed step in menaquinone biosynthesis is catalyzed by MenD, a thiamine diphosphate-dependent enzyme comprising three domains. Domains I and III form the MenD active site, but no function has yet been ascribed to domain II. Here, we show that the last cytosolic metabolite in the menaquinone biosynthesis pathway, 1,4-dihydroxy-2-naphthoic acid (DHNA), binds to domain II of Mtb-MenD and inhibits its activity. Using X-ray crystallography of four apo- and cofactor-bound Mtb-MenD structures, along with several spectroscopy assays, we identified three arginine residues (Arg-97, Arg-277, and Arg-303) that are important for both enzyme activity and the feedback inhibition by DHNA. Among these residues, Arg-277 appeared to be particularly important for signal propagation from the allosteric site to the active site. This is the first evidence of feedback regulation of the menaquinone biosynthesis pathway in bacteria, identifying a protein-level regulatory mechanism that controls menaquinone levels within the cell and may therefore represent a good target for disrupting menaquinone biosynthesis in M. tuberculosis.


Asunto(s)
Proteínas Bacterianas/metabolismo , Mycobacterium tuberculosis/metabolismo , Vitamina K 2/metabolismo , Regulación Alostérica/efectos de los fármacos , Sitio Alostérico , Secuencia de Aminoácidos , Proteínas Bacterianas/antagonistas & inhibidores , Proteínas Bacterianas/genética , Sitios de Unión , Biocatálisis , Dominio Catalítico , Cristalografía por Rayos X , Humanos , Mutagénesis Sitio-Dirigida , Mycobacterium tuberculosis/enzimología , Naftoles/química , Naftoles/metabolismo , Naftoles/farmacología , Conformación Proteica , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/química , Proteínas Recombinantes/aislamiento & purificación , Alineación de Secuencia
6.
Structure ; 24(7): 1167-77, 2016 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-27291649

RESUMEN

Menaquinone (MQ) is an essential component of the respiratory chains of many pathogenic organisms, including Mycobacterium tuberculosis (Mtb). The first committed step in MQ biosynthesis is catalyzed by 2-succinyl-5-enolpyruvyl-6-hydroxy-3-cyclohexadiene-1-carboxylate synthase (MenD), a thiamin diphosphate (ThDP)-dependent enzyme. Catalysis proceeds through two covalent intermediates as the substrates 2-oxoglutarate and isochorismate are successively added to the cofactor before final cleavage of the product. We have determined a series of crystal structures of Mtb-MenD that map the binding of both substrates, visualizing each step in the MenD catalytic cycle, including both intermediates. ThDP binding induces a marked asymmetry between the coupled active sites of each dimer, and possible mechanisms of communication can be identified. The crystal structures also reveal conformational features of the two intermediates that facilitate reaction but prevent premature product release. These data fully map chemical space to inform early-stage drug discovery targeting MenD.


Asunto(s)
Proteínas Bacterianas/química , Mycobacterium tuberculosis/enzimología , Piruvato Oxidasa/química , Proteínas Bacterianas/metabolismo , Dominio Catalítico , Simulación de Dinámica Molecular , Unión Proteica , Multimerización de Proteína , Piruvato Oxidasa/metabolismo , Tiamina/metabolismo
7.
FEBS Open Bio ; 6(12): 1220-1226, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-28203522

RESUMEN

Phosphopantetheinyl transferases (PPTases) are key elements in the modular syntheses performed by multienzyme systems such as polyketide synthases. PPTases transfer phosphopantetheine derivatives from Coenzyme A to carrier proteins (CPs), thus orchestrating substrate supply. We describe an efficient mass spectrometry-based protocol for determining CP specificity for a particular PPTase in organisms possessing several candidate PPTases. We show that the CPs MbtL and PpsC, both involved in synthesis of essential metabolites in Mycobacterium tuberculosis, are exclusively activated by the type 2 PPTase PptT and not the type 1 AcpS. The assay also enables conclusive identification of the reactive serine on each CP.

8.
Acta Crystallogr D Biol Crystallogr ; 71(Pt 11): 2297-308, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26527146

RESUMEN

The tryptophan-biosynthesis pathway is essential for Mycobacterium tuberculosis (Mtb) to cause disease, but not all of the enzymes that catalyse this pathway in this organism have been identified. The structure and function of the enzyme complex that catalyses the first committed step in the pathway, the anthranilate synthase (AS) complex, have been analysed. It is shown that the open reading frames Rv1609 (trpE) and Rv0013 (trpG) encode the chorismate-utilizing (AS-I) and glutamine amidotransferase (AS-II) subunits of the AS complex, respectively. Biochemical assays show that when these subunits are co-expressed a bifunctional AS complex is obtained. Crystallization trials on Mtb-AS unexpectedly gave crystals containing only AS-I, presumably owing to its selective crystallization from solutions containing a mixture of the AS complex and free AS-I. The three-dimensional structure reveals that Mtb-AS-I dimerizes via an interface that has not previously been seen in AS complexes. As is the case in other bacteria, it is demonstrated that Mtb-AS shows cooperative allosteric inhibition by tryptophan, which can be rationalized based on interactions at this interface. Comparative inhibition studies on Mtb-AS-I and related enzymes highlight the potential for single inhibitory compounds to target multiple chorismate-utilizing enzymes for TB drug discovery.


Asunto(s)
Antranilato Sintasa/antagonistas & inhibidores , Antranilato Sintasa/química , Mycobacterium tuberculosis/enzimología , Triptófano/metabolismo , Tuberculosis/microbiología , Antranilato Sintasa/metabolismo , Vías Biosintéticas , Cristalografía por Rayos X , Diseño de Fármacos , Inhibidores Enzimáticos/farmacología , Humanos , Modelos Moleculares , Mycobacterium tuberculosis/metabolismo , Conformación Proteica , Multimerización de Proteína , Subunidades de Proteína/antagonistas & inhibidores , Subunidades de Proteína/química , Subunidades de Proteína/metabolismo
9.
Acta Crystallogr D Biol Crystallogr ; 71(Pt 4): 862-72, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25849397

RESUMEN

Mycobacterium tuberculosis (Mtb) is the causative agent of tuberculosis. Access to iron in host macrophages depends on iron-chelating siderophores called mycobactins and is strongly correlated with Mtb virulence. Here, the crystal structure of an Mtb enzyme involved in mycobactin biosynthesis, MbtN, in complex with its FAD cofactor is presented at 2.30 Šresolution. The polypeptide fold of MbtN conforms to that of the acyl-CoA dehydrogenase (ACAD) family, consistent with its predicted role of introducing a double bond into the acyl chain of mycobactin. Structural comparisons and the presence of an acyl carrier protein, MbtL, in the same gene locus suggest that MbtN acts on an acyl-(acyl carrier protein) rather than an acyl-CoA. A notable feature of the crystal structure is the tubular density projecting from N(5) of FAD. This was interpreted as a covalently bound polyethylene glycol (PEG) fragment and resides in a hydrophobic pocket where the substrate acyl group is likely to bind. The pocket could accommodate an acyl chain of 14-21 C atoms, consistent with the expected length of the mycobactin acyl chain. Supporting this, steady-state kinetics show that MbtN has ACAD activity, preferring acyl chains of at least 16 C atoms. The acyl-binding pocket adopts a different orientation (relative to the FAD) to other structurally characterized ACADs. This difference may be correlated with the apparent ability of MbtN to catalyse the formation of an unusual cis double bond in the mycobactin acyl chain.


Asunto(s)
Enoil-ACP Reductasa (NADPH Específica B)/química , Mycobacterium tuberculosis/enzimología , Tuberculosis/microbiología , Secuencia de Aminoácidos , Sitios de Unión , Cristalografía por Rayos X , Flavina-Adenina Dinucleótido/metabolismo , Humanos , Modelos Moleculares , Datos de Secuencia Molecular , Mycobacterium tuberculosis/química , Mycobacterium tuberculosis/metabolismo , Oxazoles/metabolismo , Conformación Proteica , Alineación de Secuencia
10.
J Struct Biol ; 188(3): 274-8, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25450595

RESUMEN

Phosphopantetheinyl transferases (PPTases) are key enzymes in the assembly-line production of complex molecules such as fatty acids, polyketides and polypeptides, where they activate acyl or peptidyl carrier proteins, transferring a 4'-phosphopantetheinyl moiety from coenzyme A (CoA) to a reactive serine residue on the carrier protein. The human pathogen Mycobacterium tuberculosis encodes two PPTases, both essential and therefore attractive drug targets. We report the structure of the type-II PPTase PptT, obtained from crystals of a fusion protein with maltose binding protein. The structure, at 1.75Å resolution (R=0.156, Rfree=0.191), reveals an α/ß fold broadly similar to other type-II PPTases, but with differences in peripheral structural elements. A bound CoA is clearly defined with its pantetheinyl arm tucked into a hydrophobic pocket. Interactions involving the CoA diphosphate, bound Mg(2+) and three active site acidic side chains suggest a plausible pathway for proton transfer during catalysis.


Asunto(s)
Proteínas Bacterianas/metabolismo , Proteínas de Unión a Maltosa/metabolismo , Mycobacterium tuberculosis/metabolismo , Transferasas (Grupos de Otros Fosfatos Sustitutos)/metabolismo , Secuencia de Aminoácidos , Cristalografía por Rayos X , Estructura Secundaria de Proteína
11.
Artículo en Inglés | MEDLINE | ID: mdl-24316828

RESUMEN

In Mycobacterium tuberculosis, the protein MbtN (Rv1346) catalyzes the formation of a double bond in the fatty-acyl moiety of the siderophore mycobactin, which is used by this organism to acquire essential iron. MbtN is homologous to acyl-CoA dehydrogenases, whose general role is to catalyze the α,ß-dehydrogenation of fatty-acyl-CoA conjugates. Mycobactins, however, contain a long unsaturated fatty-acid chain with an unusual cis double bond conjugated to the carbonyl group of the mycobactin core. To characterize the role of MbtN in the dehydrogenation of this fatty-acyl moiety, the enzyme has been expressed, purified and crystallized. The crystals diffracted to 2.3 Å resolution at a synchrotron source and were found to belong to the hexagonal space group H32, with unit-cell parameters a = b = 139.10, c = 253.09 Å, α = ß = 90, γ = 120°.


Asunto(s)
Acil-CoA Deshidrogenasas/química , Proteínas Bacterianas/química , Mycobacterium tuberculosis/química , Acil-CoA Deshidrogenasas/genética , Acil-CoA Deshidrogenasas/aislamiento & purificación , Proteínas Bacterianas/genética , Proteínas Bacterianas/aislamiento & purificación , Cristalización , Cristalografía por Rayos X , Escherichia coli/genética , Escherichia coli/metabolismo , Expresión Génica , Mycobacterium tuberculosis/genética , Oxazoles/química , Oxazoles/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/aislamiento & purificación , Sideróforos/química , Sideróforos/metabolismo
12.
PLoS One ; 8(4): e61325, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23637813

RESUMEN

Recent revision of the biosynthetic pathway for menaquinone has led to the discovery of a previously unrecognized enzyme 2-succinyl-6-hydroxy-2,4-cyclohexadiene-1-carboxylate synthase, also known as MenH. This enzyme has an α/ß hydrolase fold with a catalytic triad comprising Ser86, His232, and Asp210. Mutational studies identified a number of conserved residues of importance to activity, and modeling further implicated the side chains of Tyr85 and Trp147 in formation of a non-standard oxyanion hole. We have solved the structure of E. coli MenH (EcMenH) at 2.75 Å resolution, together with the structures of the active site mutant proteins Tyr85Phe and Arg124Ala, both at 2.5 Å resolution. EcMenH has the predicted α/ß hydrolase fold with its core α/ß domain capped by a helical lid. The active site, a long groove beneath the cap, contains a number of conserved basic residues and is found to bind exogeneous anions, modeled as sulfate and chloride, in all three crystal structures. Docking studies with the MenH substrate and a transition state model indicate that the bound anions mark the binding sites for anionic groups on the substrate. The docking studies, and careful consideration of the active site geometry, further suggest that the oxyanion hole is of a conventional nature, involving peptide NH groups, rather than the proposed site involving Tyr85 and Trp147. This is in accord with conclusions from the structure of S. aureus MenH. Comparisons with the latter do, however, indicate differences in the periphery of the active site that could be of relevance to selective inhibition of MenH enzymes.


Asunto(s)
Oxo-Ácido-Liasas/química , Secuencia de Aminoácidos , Sitios de Unión , Dominio Catalítico/genética , Cristalografía por Rayos X , Escherichia coli/enzimología , Modelos Moleculares , Simulación del Acoplamiento Molecular , Oxo-Ácido-Liasas/genética , Oxo-Ácido-Liasas/metabolismo , Alineación de Secuencia
13.
Biochemistry ; 51(24): 4868-79, 2012 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-22607697

RESUMEN

MbtI is the salicylate synthase that catalyzes the first committed step in the synthesis of the iron chelating compound mycobactin in Mycobacterium tuberculosis. We previously developed a series of aromatic inhibitors against MbtI based on the reaction intermediate for this enzyme, isochorismate. The most potent of these inhibitors had hydrophobic substituents, ranging in size from a methyl to a phenyl group, appended to the terminal alkene of the enolpyruvyl group. These compounds exhibited low micromolar inhibition constants against MbtI and were at least an order of magnitude more potent than the parental compound for the series, which carries a native enolpyruvyl group. In this study, we sought to understand how the substituted enolpyruvyl group confers greater potency, by determining cocrystal structures of MbtI with six inhibitors from the series. A switch in binding mode at the MbtI active site is observed for inhibitors carrying a substituted enolpyruvyl group, relative to the parental compound. Computational studies suggest that the change in binding mode, and higher potency, is due to the effect of the substituents on the conformational landscape of the core inhibitor structure. The crystal structures and fluorescence-based thermal shift assays indicate that substituents larger than a methyl group are accommodated in the MbtI active site through significant but localized flexibility in the peptide backbone. These findings have implications for the design of improved inhibitors of MbtI, as well as other chorismate-utilizing enzymes from this family.


Asunto(s)
Dominio Catalítico , Inhibidores Enzimáticos/metabolismo , Inhibidores Enzimáticos/farmacología , Liasas/antagonistas & inhibidores , Liasas/metabolismo , Mycobacterium tuberculosis/enzimología , Ácido Corísmico/metabolismo , Cristalografía por Rayos X , Inhibidores Enzimáticos/química , Liasas/química , Modelos Moleculares , Unión Proteica , Piruvatos/química , Piruvatos/metabolismo , Piruvatos/farmacología , Soluciones , Temperatura
14.
Tuberculosis (Edinb) ; 91(2): 155-72, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21247804

RESUMEN

The TB Structural Genomics Consortium is a worldwide organization of collaborators whose mission is the comprehensive structural determination and analyses of Mycobacterium tuberculosis proteins to ultimately aid in tuberculosis diagnosis and treatment. Congruent to the overall vision, Consortium members have additionally established an integrated facilities core to streamline M. tuberculosis structural biology and developed bioinformatics resources for data mining. This review aims to share the latest Consortium developments with the TB community, including recent structures of proteins that play significant roles within M. tuberculosis. Atomic resolution details may unravel mechanistic insights and reveal unique and novel protein features, as well as important protein-protein and protein-ligand interactions, which ultimately lead to a better understanding of M. tuberculosis biology and may be exploited for rational, structure-based therapeutics design.


Asunto(s)
Genómica/métodos , Cooperación Internacional , Mycobacterium tuberculosis/genética , Proteínas Bacterianas/química , Cristalografía por Rayos X , Bases de Datos de Proteínas , Diseño de Fármacos , Genoma Bacteriano , Genómica/tendencias , Humanos , Modelos Moleculares , Mycobacterium tuberculosis/efectos de los fármacos , Mycobacterium tuberculosis/metabolismo
15.
Acta Crystallogr D Biol Crystallogr ; 66(Pt 8): 909-17, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20693690

RESUMEN

Mycobacterium tuberculosis, the cause of tuberculosis, is a devastating human pathogen against which new drugs are urgently needed. Enzymes from the biosynthetic pathway for menaquinone are considered to be valid drug targets. The protein encoded by the open reading frame Rv0554 has been expressed, purified and subjected to structural and functional analysis to test for a putative role in menaquinone biosynthesis. The crystal structure of Rv0554 has been solved and refined in two different space groups at 2.35 and 1.9 A resolution. The protein is dimeric, with an alpha/beta-hydrolase monomer fold. In each monomer, a large cavity adjacent to the catalytic triad is enclosed by a helical lid. Dimerization is mediated by the lid regions. Small-molecule additives used in crystallization bind in the active site, but no binding of ligands related to menaquinone biosynthesis could be detected and functional assays failed to support possible roles in menaquinone biosynthesis.


Asunto(s)
Mycobacterium tuberculosis/enzimología , Peroxidasa/química , Vitamina K 2/química , Cristalografía por Rayos X , Ligandos , Modelos Moleculares , Estructura Cuaternaria de Proteína , Estructura Terciaria de Proteína , Vitamina K 2/metabolismo
16.
J Biol Chem ; 281(31): 22131-22141, 2006 Aug 04.
Artículo en Inglés | MEDLINE | ID: mdl-16737961

RESUMEN

Phosphorylation of both small molecules and proteins plays a central role in many biological processes. In proteins, phosphorylation most commonly targets the oxygen atoms of Ser, Thr, and Tyr. In contrast, stably phosphorylated His residues are rarely found, due to the lability of the N-P bond, and histidine phosphorylation features most often in transient processes. Here we present the crystal structure of a protein of previously unknown function, which proves to contain a stably phosphorylated histidine residue. The protein is the product of open reading frame PAE2307, from the hyperthermophilic archaeon Pyrobaculum aerophilum, and is representative of a highly conserved protein family found in archaea and bacteria. The crystal structure of PAE2307, solved at 1.45-A resolution (R = 0.208, R(free) = 0.227), forms a remarkably tightly associated hexamer. The phosphorylated histidine at the proposed active site, pHis85, occupies a cavity that is at the interface between two subunits and contains a number of fully conserved residues. Stable phosphorylation is attributed to favorable hydrogen bonding of the phosphoryl group and a salt bridge with pHis85 that provides electronic stabilization. In silico modeling suggested that the protein may function as an adenosine kinase, a conclusion that is supported by in vitro assays of adenosine binding, using fluorescence spectroscopy, and crystallographic visualization of an adenosine complex of PAE2307 at 2.25-A resolution.


Asunto(s)
Adenosina Quinasa/química , Proteínas Arqueales/química , Secuencia Conservada , Histidina/metabolismo , Fosforilación , Adenosina/química , Adenosina/metabolismo , Adenosina Quinasa/genética , Adenosina Quinasa/metabolismo , Proteínas Arqueales/genética , Proteínas Arqueales/metabolismo , Sitios de Unión , Cristalografía por Rayos X , Enlace de Hidrógeno , Familia de Multigenes , Pyrobaculum/enzimología , Electricidad Estática
17.
Drug Discov Today ; 11(1-2): 28-34, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16478688

RESUMEN

Mycobacterium tuberculosis, the causative agent of tuberculosis (TB) in humans, is a devastating infectious organism that kills approximately two million people annually. The current suite of antibiotics used to treat TB faces two main difficulties: (i) the emergence of multidrug-resistant (MDR) strains of M. tuberculosis, and (ii) the persistent state of the bacterium, which is less susceptible to antibiotics and causes very long antibiotic treatment regimes. The complete genome sequences of a laboratory strain (H37Rv) and a clinical strain (CDC1551) of M. tuberculosis and the concurrent identification of all the open reading frames that encode proteins within this organism, present structural biologists with a wide array of protein targets for structure determination. Comparative genomics of the species that make up the M. tuberculosis complex has also added an array of genomic information to our understanding of these organisms. In response to this, structural genomics consortia have been established for targeting proteins from M. tuberculosis. This review looks at the progress of these major initiatives and the potential impact of large scale structure determination efforts on the development of inhibitors to many proteins. Increasing sophistication in structure-based drug design approaches, in combination with increasing numbers of protein structures and inhibitors for TB proteins, will have a significant impact on the downstream development of TB antibiotics.


Asunto(s)
Antituberculosos , Diseño de Fármacos , Mycobacterium tuberculosis/genética , Animales , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Genómica , Humanos , Conformación Proteica , Tuberculosis/tratamiento farmacológico
18.
Acta Crystallogr D Biol Crystallogr ; 61(Pt 9): 1199-206, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16131752

RESUMEN

Mycobacterium tuberculosis, the cause of tuberculosis, is one of the most devastating human pathogens. New drugs for its control are urgently needed. Menaquinone, also known as vitamin K, is an essential cofactor that is required for electron transfer and the enzymes that synthesize it are therefore potential drug targets. The enzyme naphthoate synthase (MenB) from M. tuberculosis has been expressed in Escherichia coli, purified and crystallized both as the native enzyme and in complex with naphthoyl-CoA. Both structures have been determined by X-ray crystallography: native MenB at 2.15 A resolution (R = 0.203, R(free) = 0.231) and its napthoyl-CoA complex at 2.30 A resolution (R = 0.197, R(free) = 0.225). The protein structure, which has a fold characteristic of the crotonase family of enzymes, is notable for the presence of several highly flexible regions around the active site. The bound naphthoyl-CoA is only visible for one of the three molecules in the asymmetric unit and only partly rigidifies the structure. The C-terminal region of the protein is seen to play a critical role both in completion of the binding pocket and in stabilization of the hexamer, suggesting a link between oligomerization and catalytic activity.


Asunto(s)
Hidroliasas/química , Mycobacterium tuberculosis/enzimología , Sitios de Unión , Clonación Molecular , Cristalografía por Rayos X , Dimerización , Escherichia coli/genética , Modelos Moleculares , Estructura Molecular , Unión Proteica , Conformación Proteica
19.
J Bacteriol ; 185(14): 4057-65, 2003 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12837779

RESUMEN

Bioinformatic analyses of whole genome sequences highlight the problem of identifying the biochemical and cellular functions of many gene products that are at present uncharacterized. The open reading frame Rv3853 from Mycobacterium tuberculosis has been annotated as menG and assumed to encode an S-adenosylmethionine (SAM)-dependent methyltransferase that catalyzes the final step in menaquinone biosynthesis. The Rv3853 gene product has been expressed, refolded, purified, and crystallized in the context of a structural genomics program. Its crystal structure has been determined by isomorphous replacement and refined at 1.9 A resolution to an R factor of 19.0% and R(free) of 22.0%. The structure strongly suggests that this protein is not a SAM-dependent methyltransferase and that the gene has been misannotated in this and other genomes that contain homologs. The protein forms a tightly associated, disk-like trimer. The monomer fold is unlike that of any known SAM-dependent methyltransferase, most closely resembling the phosphohistidine domains of several phosphotransfer systems. Attempts to bind cofactor and substrate molecules have been unsuccessful, but two adventitiously bound small-molecule ligands, modeled as tartrate and glyoxalate, are present on each monomer. These may point to biologically relevant binding sites but do not suggest a function. In silico screening indicates a range of ligands that could occupy these and other sites. The nature of these ligands, coupled with the location of binding sites on the trimer, suggests that proteins of the Rv3853 family, which are distributed throughout microbial and plant species, may be part of a larger assembly binding to nucleic acids or proteins.


Asunto(s)
Genoma Bacteriano , Metiltransferasas/química , Metiltransferasas/metabolismo , Mycobacterium tuberculosis/enzimología , Secuencia de Aminoácidos , Sitios de Unión , Biología Computacional , Secuencia Conservada , Cristalización , Cristalografía por Rayos X , Ligandos , Metiltransferasas/genética , Modelos Moleculares , Datos de Secuencia Molecular , Mycobacterium tuberculosis/genética , Pliegue de Proteína , Estructura Cuaternaria de Proteína , Alineación de Secuencia
20.
Curr Drug Targets Infect Disord ; 2(2): 121-41, 2002 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12462144

RESUMEN

Structural genomics, the large-scale determination of protein structures, promises to provide a broad structural foundation for drug discovery. The tuberculosis (TB) Structural Genomics Consortium is devoted to encouraging, coordinating, and facilitating the determination of structures of proteins from Mycobacterium tuberculosis and hopes to determine 400 TB protein structures over 5 years. The Consortium has determined structures of 28 proteins from TB to date. These protein structures are already providing a basis for drug discovery efforts.


Asunto(s)
Aciltransferasas , Antibacterianos/farmacología , Antígenos Bacterianos , Proteínas Bacterianas/química , Diseño de Fármacos , Genómica , Mycobacterium tuberculosis/química , Mycobacterium tuberculosis/efectos de los fármacos , Aldehído-Liasas/química , Sistema Enzimático del Citocromo P-450/química , Glutamato-Amoníaco Ligasa/química , Metiltransferasas/química , Mio-Inositol-1-Fosfato Sintasa/química , Oxidorreductasas/química , Proteína Disulfuro Isomerasas/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA