Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 121(16): e2309211121, 2024 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-38593081

RESUMEN

Vesicular release of neurotransmitters and hormones relies on the dynamic assembly of the exocytosis/trans-SNARE complex through sequential interactions of synaptobrevins, syntaxins, and SNAP-25. Despite SNARE-mediated release being fundamental for intercellular communication in all excitable tissues, the role of auxiliary proteins modulating the import of reserve vesicles to the active zone, and thus, scaling repetitive exocytosis remains less explored. Secretagogin is a Ca2+-sensor protein with SNAP-25 being its only known interacting partner. SNAP-25 anchors readily releasable vesicles within the active zone, thus being instrumental for 1st phase release. However, genetic deletion of secretagogin impedes 2nd phase release instead, calling for the existence of alternative protein-protein interactions. Here, we screened the secretagogin interactome in the brain and pancreas, and found syntaxin-4 grossly overrepresented. Ca2+-loaded secretagogin interacted with syntaxin-4 at nanomolar affinity and 1:1 stoichiometry. Crystal structures of the protein complexes revealed a hydrophobic groove in secretagogin for the binding of syntaxin-4. This groove was also used to bind SNAP-25. In mixtures of equimolar recombinant proteins, SNAP-25 was sequestered by secretagogin in competition with syntaxin-4. Kd differences suggested that secretagogin could shape unidirectional vesicle movement by sequential interactions, a hypothesis supported by in vitro biological data. This mechanism could facilitate the movement of transport vesicles toward release sites, particularly in the endocrine pancreas where secretagogin, SNAP-25, and syntaxin-4 coexist in both α- and ß-cells. Thus, secretagogin could modulate the pace and fidelity of vesicular hormone release by differential protein interactions.


Asunto(s)
Fusión de Membrana , Secretagoginas , Proteínas Qa-SNARE/genética , Proteínas Qa-SNARE/metabolismo , Secretagoginas/metabolismo , Membrana Celular/metabolismo , Proteína 25 Asociada a Sinaptosomas/metabolismo , Exocitosis , Comunicación Celular , Sintaxina 1/metabolismo , Unión Proteica
2.
ACS Chem Biol ; 18(4): 794-802, 2023 04 21.
Artículo en Inglés | MEDLINE | ID: mdl-37005433

RESUMEN

Pseudouridimycin is a microbial C-nucleoside natural product that specifically inhibits bacterial RNA polymerases by binding to the active site and competing with uridine triphosphate for the nucleoside triphosphate (NTP) addition site. Pseudouridimycin consists of 5'-aminopseudouridine and formamidinylated, N-hydroxylated Gly-Gln dipeptide moieties to allow Watson-Crick base pairing and to mimic protein-ligand interactions of the triphosphates of NTP, respectively. The metabolic pathway of pseudouridimycin has been studied in Streptomyces species, but no biosynthetic steps have been characterized biochemically. Here, we show that the flavin-dependent oxidase SapB functions as a gate-keeper enzyme selecting pseudouridine (KM = 34 µM) over uridine (KM = 901 µM) in the formation of pseudouridine aldehyde. The pyridoxal phosphate (PLP)-dependent SapH catalyzes transamination, resulting in 5'-aminopseudouridine with a preference for arginine, methionine, or phenylalanine as cosubstrates as amino group donors. The binary structure of SapH in complex with pyridoxamine-5'-phosphate and site-directed mutagenesis identified Lys289 and Trp32 as key residues for catalysis and substrate binding, respectively. The related C-nucleoside oxazinomycin was accepted as a substrate by SapB with moderate affinity (KM = 181 µM) and was further converted by SapH, which opens possibilities for metabolic engineering to generate hybrid C-nucleoside pseudouridimycin analogues in Streptomyces.


Asunto(s)
Nucleósidos , Seudouridina , Vías Biosintéticas , ARN Polimerasas Dirigidas por ADN/metabolismo , Nucleósidos/metabolismo , Seudouridina/biosíntesis , Seudouridina/metabolismo , Fosfato de Piridoxal/química , Streptomyces/química , Streptomyces/metabolismo
3.
Eur J Med Chem ; 228: 113976, 2022 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-34815129

RESUMEN

Our previous studies on FabG have identified two compounds 5-bromo-2-(thiophene-2-carboxamido) benzoic acid (A) and ethyl 6-bromo-2-((dimethylamino)methyl)-5-hydroxy-1-phenyl-1H-indole-3-carboxylate(B) as best hits with allosteric mode of inhibition. FabG is an integral part of bacterial fatty acid biosynthetic system FAS II shown to be an essential gene in most ESKAPE Pathogens. The current work is focussed on lead expansion of these two hit molecules which ended up with forty-three analogues (twenty-nine analogues from lead compound A and fourteen compounds from lead compound B). The enzyme inhibition studies revealed that compound 15 (effective against EcFabG, AbFabG, StFabG, MtFabG1) and 19 (inhibiting EcFabG and StFabG) had potency of broad-spectrum inhibition on FabG panel.


Asunto(s)
Antibacterianos/farmacología , Inhibidores Enzimáticos/farmacología , Oxidorreductasas/antagonistas & inhibidores , Pseudomonas/efectos de los fármacos , Antibacterianos/química , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/química , Pruebas de Sensibilidad Microbiana , Estructura Molecular , Oxidorreductasas/metabolismo , Pseudomonas/enzimología , Relación Estructura-Actividad
4.
Cell Chem Biol ; 28(9): 1321-1332.e5, 2021 09 16.
Artículo en Inglés | MEDLINE | ID: mdl-33826941

RESUMEN

Effective treatment of tuberculosis is frequently hindered by the emerging antimicrobial resistance of Mycobacterium tuberculosis. The present study evaluates monocyclic ß-lactam compounds targeting the mycobacterial cell wall remodeling. Novel N-thio-ß-lactams were designed, synthesized, and characterized on the L,D-transpeptidase-2, a validated target in M. tuberculosis. The candidates were evaluated in biochemical assays identifying five compounds presenting target-specific kinetic constants equal or superior to meropenem, a carbapenem currently considered for tuberculosis therapy. Mass spectrometry in line with the crystal structures of five target-ligand complexes revealed that the N-thio-ß-lactams act via an unconventional mode of adduct formation, transferring the thio-residues from the lactam ring to the active-site cysteine of LdtMt2. The resulting stable adducts lead to a long-term inactivation of the target protein. Finally, the candidates were evaluated in vitro against a drug-susceptible and multidrug-resistant clinical isolates of M. tuberculosis, confirming the antimycobacterial effect of these novel compounds.


Asunto(s)
Antibacterianos/farmacología , Farmacorresistencia Bacteriana/efectos de los fármacos , Mycobacterium tuberculosis/efectos de los fármacos , Peptidil Transferasas/antagonistas & inhibidores , beta-Lactamas/farmacología , Antibacterianos/síntesis química , Antibacterianos/química , Pruebas de Sensibilidad Microbiana , Estructura Molecular , Mycobacterium tuberculosis/enzimología , Peptidil Transferasas/metabolismo , beta-Lactamas/síntesis química , beta-Lactamas/química
5.
J Am Chem Soc ; 143(10): 3794-3807, 2021 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-33496585

RESUMEN

Structural information is crucial for understanding catalytic mechanisms and to guide enzyme engineering efforts of biocatalysts, such as terpene cyclases. However, low sequence similarity can impede homology modeling, and inherent protein instability presents challenges for structural studies. We hypothesized that X-ray crystallography of engineered thermostable ancestral enzymes can enable access to reliable homology models of extant biocatalysts. We have applied this concept in concert with molecular modeling and enzymatic assays to understand the structure activity relationship of spiroviolene synthase, a class I terpene cyclase, aiming to engineer its specificity. Engineering a surface patch in the reconstructed ancestor afforded a template structure for generation of a high-confidence homology model of the extant enzyme. On the basis of structural considerations, we designed and crystallized ancestral variants with single residue exchanges that exhibited tailored substrate specificity and preserved thermostability. We show how the two single amino acid alterations identified in the ancestral scaffold can be transferred to the extant enzyme, conferring a specificity switch that impacts the extant enzyme's specificity for formation of the diterpene spiroviolene over formation of sesquiterpenes hedycaryol and farnesol by up to 25-fold. This study emphasizes the value of ancestral sequence reconstruction combined with enzyme engineering as a versatile tool in chemical biology.


Asunto(s)
Transferasas Alquil y Aril/metabolismo , Ingeniería de Proteínas , Transferasas Alquil y Aril/química , Transferasas Alquil y Aril/genética , Biocatálisis , Cristalografía por Rayos X , Ciclización , Diterpenos/química , Diterpenos/metabolismo , Mutagénesis Sitio-Dirigida , Conformación Proteica , Sesquiterpenos/química , Sesquiterpenos/metabolismo , Especificidad por Sustrato
6.
Bioorg Med Chem ; 30: 115898, 2021 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-33388594

RESUMEN

The spread of antibiotic resistance within the ESKAPE group of human pathogenic bacteria poses severe challenges in the treatment of infections and maintenance of safe hospital environments. This motivates efforts to validate novel target proteins within these species that could be pursued as potential targets for antibiotic development. Genetic data suggest that the enzyme FabG, which is part of the bacterial fatty acid biosynthetic system FAS-II, is essential in several ESKAPE pathogens. FabG catalyzes the NADPH dependent reduction of 3-keto-acyl-ACP during fatty acid elongation, thus enabling lipid supply for production and maintenance of the cell envelope. Here we report on small-molecule screening on the FabG enzymes from A. baumannii and S. typhimurium to identify a set of µM inhibitors, with the most potent representative (1) demonstrating activity against six FabG-orthologues. A co-crystal structure with FabG from A. baumannii (PDB:6T65) confirms inhibitor binding at an allosteric site located in the subunit interface, as previously demonstrated for other sub-µM inhibitors of FabG from P. aeruginosa. We show that inhibitor binding distorts the oligomerization interface in the FabG tetramer and displaces crucial residues involved in the interaction with the co-substrate NADPH. These observations suggest a conserved allosteric site across the FabG family, which can be potentially targeted for interference with fatty acid biosynthesis in clinically relevant ESKAPE pathogens.


Asunto(s)
Acinetobacter baumannii/enzimología , Oxidorreductasas de Alcohol/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , Pseudomonas aeruginosa/enzimología , Salmonella typhimurium/enzimología , Oxidorreductasas de Alcohol/metabolismo , Sitios de Unión/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , Estructura Molecular , Relación Estructura-Actividad
7.
J Mol Biol ; 430(18 Pt B): 3170-3189, 2018 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-30017920

RESUMEN

Many bacteria secrete cellulose, which forms the structural basis for bacterial multicellular aggregates, termed biofilms. The cellulose synthase complex of Salmonella typhimurium consists of the catalytic subunits BcsA and BcsB and several auxiliary subunits that are encoded by two divergently transcribed operons, bcsRQABZC and bcsEFG. Expression of the bcsEFG operon is required for full-scale cellulose production, but the functions of its products are not fully understood. This work aimed to characterize the BcsG subunit of the cellulose synthase, which consists of an N-terminal transmembrane fragment and a C-terminal domain in the periplasm. Deletion of the bcsG gene substantially decreased the total amount of BcsA and cellulose production. BcsA levels were partially restored by the expression of the transmembrane segment, whereas restoration of cellulose production required the presence of the C-terminal periplasmic domain and its characteristic metal-binding residues. The high-resolution crystal structure of the periplasmic domain characterized BcsG as a member of the alkaline phosphatase/sulfatase superfamily of metalloenzymes, containing a conserved Zn2+-binding site. Sequence and structural comparisons showed that BcsG belongs to a specific family within alkaline phosphatase-like enzymes, which includes bacterial Zn2+-dependent lipopolysaccharide phosphoethanolamine transferases such as MCR-1 (colistin resistance protein), EptA, and EptC and the Mn2+-dependent lipoteichoic acid synthase (phosphoglycerol transferase) LtaS. These enzymes use the phospholipids phosphatidylethanolamine and phosphatidylglycerol, respectively, as substrates. These data are consistent with the recently discovered phosphoethanolamine modification of cellulose by BcsG and show that its membrane-bound and periplasmic parts play distinct roles in the assembly of the functional cellulose synthase and cellulose production.


Asunto(s)
Glucosiltransferasas/química , Glucosiltransferasas/metabolismo , Subunidades de Proteína , Salmonella typhimurium/metabolismo , Fosfatasa Alcalina/química , Fosfatasa Alcalina/metabolismo , Orden Génico , Glucosiltransferasas/genética , Modelos Moleculares , Unión Proteica , Conformación Proteica , Salmonella typhimurium/genética , Relación Estructura-Actividad
8.
Mol Metab ; 14: 108-120, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29910119

RESUMEN

OBJECTIVE: Specification of endocrine cell lineages in the developing pancreas relies on extrinsic signals from non-pancreatic tissues, which initiate a cell-autonomous sequence of transcription factor activation and repression switches. The steps in this pathway share reliance on activity-dependent Ca2+ signals. However, the mechanisms by which phasic Ca2+ surges become converted into a dynamic, cell-state-specific and physiologically meaningful code made up by transcription factors constellations remain essentially unknown. METHODS: We used high-resolution histochemistry to explore the coincident expression of secretagogin and transcription factors driving ß cell differentiation. Secretagogin promoter activity was tested in response to genetically manipulating Pax6 and Pax4 expression. Secretagogin null mice were produced with their pancreatic islets morphologically and functionally characterized during fetal development. A proteomic approach was utilized to identify the Ca2+-dependent interaction of secretagogin with subunits of the 26S proteasome and verified in vitro by focusing on Pdx1 retention. RESULTS: Here, we show that secretagogin, a Ca2+ sensor protein that controls α and ß cell turnover in adult, is in fact expressed in endocrine pancreas from the inception of lineage segregation in a Pax4-and Pax6-dependent fashion. By genetically and pharmacologically manipulating secretagogin expression and interactome engagement in vitro, we find secretagogin to gate excitation-driven Ca2+ signals for ß cell differentiation and insulin production. Accordingly, secretagogin-/- fetuses retain a non-committed pool of endocrine progenitors that co-express both insulin and glucagon. We identify the Ca2+-dependent interaction of secretagogin with subunits of the 26S proteasome complex to prevent Pdx1 degradation through proteasome inactivation. This coincides with retained Nkx6.1, Pax4 and insulin transcription in prospective ß cells. CONCLUSIONS: In sum, secretagogin scales the temporal availability of a Ca2+-dependent transcription factor network to define ß cell identity.


Asunto(s)
Proteínas de Homeodominio/metabolismo , Células Secretoras de Insulina/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteolisis , Secretagoginas/metabolismo , Transactivadores/metabolismo , Animales , Calcio/metabolismo , Células Cultivadas , Células HEK293 , Proteínas de Homeodominio/genética , Humanos , Insulina/metabolismo , Secreción de Insulina , Ratones Endogámicos C57BL , Factor de Transcripción PAX6/genética , Factor de Transcripción PAX6/metabolismo , Factores de Transcripción Paired Box/genética , Factores de Transcripción Paired Box/metabolismo , Secretagoginas/genética
9.
Proteins ; 86(9): 912-923, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29722065

RESUMEN

RipA plays a vital role during cell division of Mycobacterium tuberculosis by degrading the cell wall peptidoglycan at the septum, allowing daughter cell separation. The peptidoglycan degrading activity relies on the NlpC/P60 domain, and as it is potentially harmful when deregulated, spatial and temporal control is necessary in this process. The N-terminal domain of RipA has been proposed to play an inhibitory role blocking the C-terminal NlpC/P60 domain. Accessibility of the active site cysteine residue is however not limited by the presence of the N-terminal domain, but by the lid-module of the inter-domain linker, which is situated in the peptide binding groove of the crystal structures of the catalytic domain. The 2.2 Å resolution structure of the N-terminal domain, determined by Se-SAD phasing, reveals an all-α-fold with 2 long α-helices, and shows similarity to bacterial periplasmic protein domains with scaffold-building role. Size exclusion chromatography and SAXS experiments are consistent with dimer formation of this domain in solution. The SAXS data from the periplasmic two-domain RipA construct suggest a rigid baton-like structure of the N-terminal module, with the catalytic domain connected by a 24 residue long flexible linker. This flexible linker allows for a catalytic zone, which is part of the spatiotemporal control of peptidoglycan degradation.


Asunto(s)
Proteínas Bacterianas/metabolismo , Pared Celular/enzimología , Hidrolasas/metabolismo , Proteínas Bacterianas/química , Biocatálisis , Dominio Catalítico , Hidrolasas/química , Mycobacterium tuberculosis/metabolismo , Peptidoglicano/metabolismo , Conformación Proteica , Multimerización de Proteína
10.
Bioorg Med Chem Lett ; 27(19): 4582-4587, 2017 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-28882483

RESUMEN

CysK1 and CysK2 are two members of the cysteine/S-sulfocysteine synthase family in Mycobacterium tuberculosis, responsible for the de novo biosynthesis of l-cysteine, which is subsequently used as a building block for mycothiol. This metabolite is the first line defense of this pathogen against reactive oxygen and nitrogen species released by host macrophages after phagocytosis. In a previous medicinal chemistry campaign we had developed urea-based inhibitors of the cysteine synthase CysM with bactericidal activity against dormant M. tuberculosis. In this study we extended these efforts by examination of the in vitro activities of a library consisting of 71 urea compounds against CysK1 and CysK2. Binding was established by fluorescence spectroscopy and inhibition by enzyme assays. Several of the compounds inhibited these two cysteine synthases, with the most potent inhibitor displaying an IC50 value of 2.5µM for CysK1 and 6.6µM for CysK2, respectively. Four of the identified molecules targeting CysK1 and CysK2 were also among the top ten inhibitors of CysM, suggesting that potent compounds could be developed with activity against all three enzymes.


Asunto(s)
Cisteína Sintasa/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , Mycobacterium tuberculosis/enzimología , Urea/farmacología , Cisteína Sintasa/metabolismo , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/química , Modelos Moleculares , Estructura Molecular , Relación Estructura-Actividad , Urea/análogos & derivados , Urea/química
11.
EMBO J ; 36(14): 2107-2125, 2017 07 14.
Artículo en Inglés | MEDLINE | ID: mdl-28637794

RESUMEN

Ca2+-sensor proteins are generally implicated in insulin release through SNARE interactions. Here, secretagogin, whose expression in human pancreatic islets correlates with their insulin content and the incidence of type 2 diabetes, is shown to orchestrate an unexpectedly distinct mechanism. Single-cell RNA-seq reveals retained expression of the TRP family members in ß-cells from diabetic donors. Amongst these, pharmacological probing identifies Ca2+-permeable transient receptor potential vanilloid type 1 channels (TRPV1) as potent inducers of secretagogin expression through recruitment of Sp1 transcription factors. Accordingly, agonist stimulation of TRPV1s fails to rescue insulin release from pancreatic islets of glucose intolerant secretagogin knock-out(-/-) mice. However, instead of merely impinging on the SNARE machinery, reduced insulin availability in secretagogin-/- mice is due to ß-cell loss, which is underpinned by the collapse of protein folding and deregulation of secretagogin-dependent USP9X deubiquitinase activity. Therefore, and considering the desensitization of TRPV1s in diabetic pancreata, a TRPV1-to-secretagogin regulatory axis seems critical to maintain the structural integrity and signal competence of ß-cells.


Asunto(s)
Regulación de la Expresión Génica , Células Secretoras de Insulina/fisiología , Proteínas/metabolismo , Secretagoginas/metabolismo , Canales Catiónicos TRPV/metabolismo , Animales , Supervivencia Celular , Perfilación de la Expresión Génica , Humanos , Ratones , Ratones Noqueados , Secretagoginas/deficiencia , Análisis de la Célula Individual
12.
FEBS J ; 284(5): 725-741, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28075068

RESUMEN

ß-lactam antibiotics represent a novel direction in the chemotherapy of tuberculosis that brings the peptidoglycan layer of the complex mycobacterial cell wall in focus as a therapeutic target. Peptidoglycan stability in Mycobacterium tuberculosis, especially during infection, relies on the nonconventional peptide cross-links formed by l,d-transpeptidases. These enzymes are known to be inhibited by ß-lactams, primarily carbapenems, leading to a stable covalent modification at the enzyme active site. A panel of 16 ß-lactam antibiotics was characterized by inhibition kinetics, mass spectrometry, and x-ray crystallography to identify efficient compounds and study their action on the essential transpeptidase, LdtMt2 . Members of the carbapenem class displayed fast binding kinetics, but faropenem, a penem type compound showed a three to four time higher rate in the adduct formation. In three cases, mass spectrometry indicated that carbapenems may undergo decarboxylation, while faropenem decomposition following the acylation step results in a small 87 Da ß-OH-butyryl adduct bound at the catalytic cysteine residue. The crystal structure of LdtMt2 at 1.54 Å resolution with this fragment bound revealed that the protein adopts a closed conformation that shields the thioester bond from the solvent, which is in line with the high stability of this dead-end complex observed also in biochemical assays. DATABASE: Structural data are available in Protein Data Bank under the accession numbers 5LB1 and 5LBG.


Asunto(s)
Mycobacterium tuberculosis/enzimología , Peptidil Transferasas/química , Tuberculosis/tratamiento farmacológico , beta-Lactamas/química , Carbapenémicos/química , Carbapenémicos/uso terapéutico , Cristalografía por Rayos X , Cinética , Mycobacterium tuberculosis/patogenicidad , Peptidoglicano/química , Peptidil Transferasas/metabolismo , Conformación Proteica , Tuberculosis/microbiología , beta-Lactamas/uso terapéutico
13.
J Med Chem ; 59(14): 6848-59, 2016 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-27379713

RESUMEN

Cysteine is an important amino acid in the redox defense of Mycobacterium tuberculosis, primarily as a building block of mycothiol. Genetic studies have implicated de novo cysteine biosynthesis in pathogen survival in infected macrophages, in particular for persistent M. tuberculosis. Here, we report on the identification and characterization of potent inhibitors of CysM, a critical enzyme in cysteine biosynthesis during dormancy. A screening campaign of 17 312 compounds identified ligands that bind to the active site with micromolar affinity. These were characterized in terms of their inhibitory potencies and structure-activity relationships through hit expansion guided by three-dimensional structures of enzyme-inhibitor complexes. The top compound binds to CysM with 300 nM affinity and displays selectivity over the mycobacterial homologues CysK1 and CysK2. Notably, two inhibitors show significant potency in a nutrient-starvation model of dormancy of Mycobacterium tuberculosis, with little or no cytotoxicity toward mammalian cells.


Asunto(s)
Antibacterianos/farmacología , Cisteína Sintasa/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , Mycobacterium tuberculosis/efectos de los fármacos , Animales , Antibacterianos/síntesis química , Antibacterianos/química , Línea Celular , Cisteína Sintasa/metabolismo , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , Humanos , Ratones , Pruebas de Sensibilidad Microbiana , Modelos Moleculares , Estructura Molecular , Mycobacterium tuberculosis/enzimología , Relación Estructura-Actividad
14.
Acta Crystallogr F Struct Biol Commun ; 72(Pt 2): 105-11, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26841760

RESUMEN

The locus PA4991 in Pseudomonas aeruginosa encodes an open reading frame that has been identified as essential for the virulence and/or survival of this pathogenic organism in the infected host. Here, it is shown that this gene encodes a monomeric FAD-binding protein of molecular mass 42.2 kDa. The structure of PA4991 was determined by a combination of molecular replacement using a search model generated with Rosetta and phase improvement by a low-occupancy heavy-metal derivative. PA4991 belongs to the GR2 family of FAD-dependent oxidoreductases, comprising an FAD-binding domain typical of the glutathione reductase family and a second domain dominated by an eight-stranded mixed ß-sheet. Most of the protein-FAD interactions are via the FAD-binding domain, but the isoalloxazine ring is located at the domain interface and interacts with residues from both domains. A comparison with the structurally related glycine oxidase and glycerol-3-phosphate dehydrogenase shows that in spite of very low amino-acid sequence identity (<18%) several active-site residues involved in substrate binding in these enzymes are conserved in PA4991. However, enzymatic assays show that PA4991 does not display amino-acid oxidase or glycerol-3-phosphate dehydrogenase activities, suggesting that it requires different substrates for activity.


Asunto(s)
Cristalización/métodos , Cristalografía por Rayos X/métodos , Flavoproteínas/química , Pseudomonas aeruginosa/enzimología , Modelos Moleculares , Conformación Proteica
15.
Acta Crystallogr F Struct Biol Commun ; 71(Pt 8): 1020-6, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26249693

RESUMEN

Bacterial infections remain a serious health concern, in particular causing life-threatening infections of hospitalized and immunocompromised patients. The situation is exacerbated by the rise in antibacterial drug resistance, and new treatments are urgently sought. In this endeavour, accurate structures of molecular targets can support early-stage drug discovery. Here, crystal structures, in three distinct forms, of recombinant Pseudomonas aeruginosa ß-ketoacyl-(acyl-carrier-protein) synthase II (FabF) are presented. This enzyme, which is involved in fatty-acid biosynthesis, has been validated by genetic and chemical means as an antibiotic target in Gram-positive bacteria and represents a potential target in Gram-negative bacteria. The structures of apo FabF, of a C164Q mutant in which the binding site is altered to resemble the substrate-bound state and of a complex with 3-(benzoylamino)-2-hydroxybenzoic acid are reported. This compound mimics aspects of a known natural product inhibitor, platensimycin, and surprisingly was observed binding outside the active site, interacting with a symmetry-related molecule. An unusual feature is a completely buried potassium-binding site that was identified in all three structures. Comparisons suggest that this may represent a conserved structural feature of FabF relevant to fold stability. The new structures provide templates for structure-based ligand design and, together with the protocols and reagents, may underpin a target-based drug-discovery project for urgently needed antibacterials.


Asunto(s)
Antibacterianos/química , Proteínas Bacterianas/química , Acido Graso Sintasa Tipo II/química , Potasio/química , Pseudomonas aeruginosa/química , Proteínas Recombinantes de Fusión/química , Adamantano/química , Secuencias de Aminoácidos , Aminobenzoatos/química , Anilidas/química , Proteínas Bacterianas/antagonistas & inhibidores , Proteínas Bacterianas/genética , Benzamidas/química , Dominio Catalítico , Clonación Molecular , Cristalización , Cristalografía por Rayos X , Descubrimiento de Drogas , Escherichia coli/genética , Escherichia coli/metabolismo , Acido Graso Sintasa Tipo II/antagonistas & inhibidores , Acido Graso Sintasa Tipo II/genética , Expresión Génica , Modelos Moleculares , Datos de Secuencia Molecular , Mutación , Unión Proteica , Estructura Secundaria de Proteína , Subunidades de Proteína/antagonistas & inhibidores , Subunidades de Proteína/química , Subunidades de Proteína/genética , Pseudomonas aeruginosa/enzimología , Proteínas Recombinantes de Fusión/genética , Salicilatos/química
16.
PLoS One ; 10(3): e0121494, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25807013

RESUMEN

In Mycobacterium tuberculosis the sulfate activating complex provides a key branching point in sulfate assimilation. The complex consists of two polypeptide chains, CysD and CysN. CysD is an ATP sulfurylase that, with the energy provided by the GTPase activity of CysN, forms adenosine-5'-phosphosulfate (APS) which can then enter the reductive branch of sulfate assimilation leading to the biosynthesis of cysteine. The CysN polypeptide chain also contains an APS kinase domain (CysC) that phosphorylates APS leading to 3'-phosphoadenosine-5'-phosphosulfate, the sulfate donor in the synthesis of sulfolipids. We have determined the crystal structures of CysC from M. tuberculosis as a binary complex with ADP, and as ternary complexes with ADP and APS and the ATP mimic AMP-PNP and APS, respectively, to resolutions of 1.5 Å, 2.1 Å and 1.7 Å, respectively. CysC shows the typical APS kinase fold, and the structures provide comprehensive views of the catalytic machinery, conserved in this enzyme family. Comparison to the structure of the human homolog show highly conserved APS and ATP binding sites, questioning the feasibility of the design of specific inhibitors of mycobacterial CysC. Residue Cys556 is part of the flexible lid region that closes off the active site upon substrate binding. Mutational analysis revealed this residue as one of the determinants controlling lid closure and hence binding of the nucleotide substrate.


Asunto(s)
Mycobacterium tuberculosis/química , Mycobacterium tuberculosis/metabolismo , Péptidos/química , Fosfotransferasas (Aceptor de Grupo Alcohol)/química , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Sulfatos/metabolismo , Adenosina Fosfosulfato/metabolismo , Adenosina Trifosfato/metabolismo , Secuencia de Aminoácidos , Dominio Catalítico , Datos de Secuencia Molecular , Nucleótidos/metabolismo , Péptidos/metabolismo , Fosfoadenosina Fosfosulfato/metabolismo , Estructura Terciaria de Proteína , Alineación de Secuencia , Sulfato Adenililtransferasa/química , Sulfato Adenililtransferasa/metabolismo
17.
Acta Crystallogr D Biol Crystallogr ; 71(Pt 3): 721-31, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25760619

RESUMEN

Locus PA4043 in the genome of Pseudomonas aeruginosa PAO1 has been annotated as coding for a farnesyl pyrophosphate synthase (FPPS). This open reading frame was cloned and expressed recombinantly in Escherichia coli. The dimeric enzyme shows farnesyl pyrophosphate synthase activity and is strongly inhibited by ibandronate and zoledronate, drugs that are presently in clinical use. The structures of the unliganded enzyme and complexes with the substrate geranyl diphosphate (GPP), the inhibitor ibandronate and two compounds obtained from a differential scanning fluorimetry-based screen of a fragment library were determined by X-ray crystallography to resolutions of better than 2.0 Å. The enzyme shows the typical α-helical fold of farnesyl pyrophosphate synthases. The substrate GPP binds in the S1 substrate site in an open conformation of the enzyme. In the enzyme-ibandronate complex three inhibitor molecules are bound in the active site of the enzyme. One inhibitor molecule occupies the allylic substrate site (S1) of each subunit, as observed in complexes of nitrogen-containing bisphosphonate inhibitors of farnesyl synthases from other species. Two (in subunit A) and one (in subunit B) additional ibandronate molecules are bound in the active site. The structures of the fragment complexes show two molecules bound in a hydrophobic pocket adjacent to the active site. This allosteric pocket, which has previously only been described for FPPS from eukaryotic organisms, is thus also present in enzymes from pathogenic prokaryotes and might be utilized for the design of inhibitors of bacterial FPPS with a different chemical scaffold to the highly charged bisphosphonates, which are less likely to pass bacterial membranes.


Asunto(s)
Proteínas Bacterianas/química , Difosfonatos/química , Inhibidores Enzimáticos/química , Geraniltranstransferasa/química , Pseudomonas aeruginosa/enzimología , Proteínas Bacterianas/antagonistas & inhibidores , Cristalografía por Rayos X , Geraniltranstransferasa/antagonistas & inhibidores , Ácido Ibandrónico , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína
18.
EMBO J ; 34(1): 36-54, 2015 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-25430741

RESUMEN

A hierarchical hormonal cascade along the hypothalamic-pituitary-adrenal axis orchestrates bodily responses to stress. Although corticotropin-releasing hormone (CRH), produced by parvocellular neurons of the hypothalamic paraventricular nucleus (PVN) and released into the portal circulation at the median eminence, is known to prime downstream hormone release, the molecular mechanism regulating phasic CRH release remains poorly understood. Here, we find a cohort of parvocellular cells interspersed with magnocellular PVN neurons expressing secretagogin. Single-cell transcriptome analysis combined with protein interactome profiling identifies secretagogin neurons as a distinct CRH-releasing neuron population reliant on secretagogin's Ca(2+) sensor properties and protein interactions with the vesicular traffic and exocytosis release machineries to liberate this key hypothalamic releasing hormone. Pharmacological tools combined with RNA interference demonstrate that secretagogin's loss of function occludes adrenocorticotropic hormone release from the pituitary and lowers peripheral corticosterone levels in response to acute stress. Cumulatively, these data define a novel secretagogin neuronal locus and molecular axis underpinning stress responsiveness.


Asunto(s)
Corticosterona/metabolismo , Hormona Liberadora de Corticotropina/metabolismo , Neuronas/metabolismo , Núcleo Hipotalámico Paraventricular/metabolismo , Secretagoginas/metabolismo , Estrés Fisiológico/fisiología , Animales , Corticosterona/genética , Hormona Liberadora de Corticotropina/genética , Masculino , Ratones , Neuronas/citología , Núcleo Hipotalámico Paraventricular/citología , Hipófisis/citología , Hipófisis/metabolismo , Interferencia de ARN , Secretagoginas/genética , Transcriptoma/fisiología
19.
Biochim Biophys Acta ; 1854(9): 1175-83, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25484279

RESUMEN

The alarming increase of drug resistance in Mycobacterium tuberculosis strains poses a severe threat to human health. Chemotherapy is particularly challenging because M. tuberculosis can persist in the lungs of infected individuals; estimates of the WHO indicate that about 1/3 of the world population is infected with latent tuberculosis providing a large reservoir for relapse and subsequent spread of the disease. Persistent M. tuberculosis shows considerable tolerance towards conventional antibiotics making treatment particularly difficult. In this phase the bacilli are exposed to oxygen and nitrogen radicals generated as part of the host response and redox-defense mechanisms are thus vital for the survival of the pathogen. Sulfur metabolism and de novo cysteine biosynthesis have been shown to be important for the redox homeostasis in persistent M. tuberculosis and these pathways could provide promising targets for novel antibiotics for the treatment of the latent form of the disease. Recent research has provided evidence for three de novo metabolic routes of cysteine biosynthesis in M. tuberculosis, each with a specific PLP dependent cysteine synthase with distinct substrate specificities. In this review we summarize our present understanding of these pathways, with a focus on the advances on functional and mechanistic characterization of mycobacterial PLP dependent cysteine synthases, their role in the various pathways to cysteine, and first attempts to develop specific inhibitors of mycobacterial cysteine biosynthesis. This article is part of a Special Issue entitled: Cofactor-dependent proteins: evolution, chemical diversity and bio-applications.


Asunto(s)
Cisteína Sintasa/química , Mycobacterium tuberculosis/enzimología , Fosfato de Piridoxal/fisiología , Antibacterianos/farmacología , Cisteína/biosíntesis , Cisteína Sintasa/antagonistas & inhibidores , Cisteína Sintasa/metabolismo , Humanos , Mycobacterium tuberculosis/efectos de los fármacos
20.
J Bacteriol ; 196(19): 3410-20, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25022854

RESUMEN

Mycobacterium tuberculosis is dependent on cysteine biosynthesis, and reduced sulfur compounds such as mycothiol synthesized from cysteine serve in first-line defense mechanisms against oxidative stress imposed by macrophages. Two biosynthetic routes to l-cysteine, each with its own specific cysteine synthase (CysK1 and CysM), have been described in M. tuberculosis, but the function of a third putative sulfhydrylase in this pathogen, CysK2, has remained elusive. We present biochemical and biophysical evidence that CysK2 is an S-sulfocysteine synthase, utilizing O-phosphoserine (OPS) and thiosulfate as substrates. The enzyme uses a mechanism via a central aminoacrylate intermediate that is similar to that of other members of this pyridoxal phosphate-dependent enzyme family. The apparent second-order rate of the first half-reaction with OPS was determined as kmax/Ks = (3.97 × 10(3)) ± 619 M(-1) s(-1), which compares well to the OPS-specific mycobacterial cysteine synthase CysM with a kmax/Ks of (1.34 × 10(3)) ± 48.2. Notably, CysK2 does not utilize thiocarboxylated CysO as a sulfur donor but accepts thiosulfate and sulfide as donor substrates. The specificity constant kcat/Km for thiosulfate is 40-fold higher than for sulfide, suggesting an annotation as S-sulfocysteine synthase. Mycobacterial CysK2 thus provides a third metabolic route to cysteine, either directly using sulfide as donor or indirectly via S-sulfocysteine. Hypothetically, S-sulfocysteine could also act as a signaling molecule triggering additional responses in redox defense in the pathogen upon exposure to reactive oxygen species during dormancy.


Asunto(s)
Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Liasas/química , Liasas/metabolismo , Mycobacterium tuberculosis/enzimología , Serina/metabolismo , Proteínas Bacterianas/genética , Cinética , Liasas/genética , Mycobacterium tuberculosis/química , Mycobacterium tuberculosis/genética , Serina/análogos & derivados , Especificidad por Sustrato
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...