Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 163
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 120(22): e2216304120, 2023 05 30.
Artículo en Inglés | MEDLINE | ID: mdl-37216558

RESUMEN

The oral microbiome is critical to human health and disease, yet the role that host salivary proteins play in maintaining oral health is unclear. A highly expressed gene in human salivary glands encodes the lectin zymogen granule protein 16 homolog B (ZG16B). Despite the abundance of this protein, its interaction partners in the oral microbiome are unknown. ZG16B possesses a lectin fold, but whether it binds carbohydrates is unclear. We postulated that ZG16B would bind microbial glycans to mediate recognition of oral microbes. To this end, we developed a microbial glycan analysis probe (mGAP) strategy based on conjugating the recombinant protein to fluorescent or biotin reporter functionality. Applying the ZG16B-mGAP to dental plaque isolates revealed that ZG16B predominantly binds to a limited set of oral microbes, including Streptococcus mitis, Gemella haemolysans, and, most prominently, Streptococcus vestibularis. S. vestibularis is a commensal bacterium widely distributed in healthy individuals. ZG16B binds to S. vestibularis through the cell wall polysaccharides attached to the peptidoglycan, indicating that the protein is a lectin. ZG16B slows the growth of S. vestibularis with no cytotoxicity, suggesting that it regulates S. vestibularis abundance. The mGAP probes also revealed that ZG16B interacts with the salivary mucin MUC7. Analysis of S. vestibularis and MUC7 with ZG16B using super-resolution microscopy supports ternary complex formation that can promote microbe clustering. Together, our data suggest that ZG16B influences the compositional balance of the oral microbiome by capturing commensal microbes and regulating their growth using a mucin-assisted clearance mechanism.


Asunto(s)
Interacciones Microbiota-Huesped , Péptidos y Proteínas de Señalización Intercelular , Lectinas , Humanos , Pared Celular/metabolismo , Lectinas/metabolismo , Mucinas/metabolismo , Polisacáridos/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo
2.
Biochemistry ; 63(1): 141-151, 2024 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-38110367

RESUMEN

The Campylobacter genus of Gram-negative bacteria is characterized by the expression of N-linked protein glycosylation (pgl) pathways. As Campylobacter concisus is an emerging human pathogen, a better understanding of the variation of the biosynthetic pathways across the genus is necessary to identify the relationships between protein glycosylation and disease. The pgl pathways of C. concisus strains have been reported to diverge from other Campylobacter in steps after the biosynthesis of N-acetylgalactosamine-α1,3-N,N'-diacetylbacillosamine-α-1-diphosphate undecaprenyl (GalNAc-diNAcBac-PP-Und), which is catalyzed by PglC and PglA, a phosphoglycosyltransferase (PGT) and a glycosyltransferase (GT), respectively. Here we characterize the PglJ GTs from two strains of C. concisus. Chemical synthesis was employed to access the stereochemically defined glycan donor substrates, uridine diphosphate N-acetyl-d-galactosaminuronic acid (UDP-GalNAcA) and uridine diphosphate N-acetyl-d-glucosaminuronic acid (UDP-GlcNAcA), to allow biochemical investigation of PglJ. Evidence for the PglJ substrate specificity structural determinants for the C6″ carboxylate-containing sugar was obtained through variant-based biochemical assays. Additionally, characterization of a UDP-sugar dehydrogenase encoded in the pgl operon, which is similar to the Pseudomonas aeruginosa WbpO responsible for the oxidization of a UDP-HexNAc to UDP-HexNAcA, supports the availability of a UDP-HexNAcA substrate for a GT that incorporates the modified sugar and provides evidence for the presence of a HexNAcA in the N-linked glycan. Utilizing sequence similarity network (SSN) analysis, we identified conserved sequence motifs among PglJ glycosyltransferases, shedding light on substrate preferences and offering predictive insights into enzyme functions across the Campylobacter genus. These studies now allow detailed characterization of the later steps in the pgl pathway in C. concisus strains and provide insights into enzyme substrate specificity determinants for glycan assembly enzymes.


Asunto(s)
Campylobacter , Glicosiltransferasas , Humanos , Glicosiltransferasas/genética , Glicosiltransferasas/metabolismo , Glicosilación , Polisacáridos , Campylobacter/genética , Campylobacter/metabolismo , Uridina Difosfato/metabolismo , Azúcares
3.
J Biol Chem ; 299(10): 105194, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37633332

RESUMEN

Complex glycans serve essential functions in all living systems. Many of these intricate and byzantine biomolecules are assembled employing biosynthetic pathways wherein the constituent enzymes are membrane-associated. A signature feature of the stepwise assembly processes is the essentiality of unusual linear long-chain polyprenol phosphate-linked substrates of specific isoprene unit geometry, such as undecaprenol phosphate (UndP) in bacteria. How these enzymes and substrates interact within a lipid bilayer needs further investigation. Here, we focus on a small enzyme, PglC from Campylobacter, structurally characterized for the first time in 2018 as a detergent-solubilized construct. PglC is a monotopic phosphoglycosyl transferase that embodies the functional core structure of the entire enzyme superfamily and catalyzes the first membrane-committed step in a glycoprotein assembly pathway. The size of the enzyme is significant as it enables high-level computation and relatively facile, for a membrane protein, experimental analysis. Our ensemble computational and experimental results provided a high-level view of the membrane-embedded PglC/UndP complex. The findings suggested that it is advantageous for the polyprenol phosphate to adopt a conformation in the same leaflet where the monotopic membrane protein resides as opposed to additionally disrupting the opposing leaflet of the bilayer. Further, the analysis showed that electrostatic steering acts as a major driving force contributing to the recognition and binding of both UndP and the soluble nucleotide sugar substrate. Iterative computational and experimental mutagenesis support a specific interaction of UndP with phosphoglycosyl transferase cationic residues and suggest a role for critical conformational transitions in substrate binding and specificity.


Asunto(s)
Membrana Celular , Poliprenoles , Transferasas , Ligandos , Proteínas de la Membrana , Fosfatos , Poliprenoles/metabolismo , Transferasas/química , Fosfatos de Poliisoprenilo/química , Membrana Celular/química , Bacterias/química , Bacterias/citología
4.
J Am Chem Soc ; 146(5): 3220-3229, 2024 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-38271668

RESUMEN

Complex bacterial glycoconjugates drive interactions between pathogens, symbionts, and their human hosts. Glycoconjugate biosynthesis is initiated at the membrane interface by phosphoglycosyl transferases (PGTs), which catalyze the transfer of a phosphosugar from a soluble uridine diphosphosugar (UDP-sugar) substrate to a membrane-bound polyprenol-phosphate (Pren-P). The two distinct superfamilies of PGT enzymes (polytopic and monotopic) show striking differences in their structure and mechanism. We designed and synthesized a series of uridine bisphosphonates (UBPs), wherein the diphosphate of the UDP and UDP-sugar is replaced by a substituted methylene bisphosphonate (CXY-BPs; X/Y = F/F, Cl/Cl, (S)-H/F, (R)-H/F, H/H, CH3/CH3). UBPs and UBPs incorporating an N-acetylglucosamine (GlcNAc) substituent at the ß-phosphonate were evaluated as inhibitors of a polytopic PGT (WecA from Thermotoga maritima) and a monotopic PGT (PglC from Campylobacter jejuni). Although CHF-BP most closely mimics diphosphate with respect to its acid/base properties, the less basic CF2-BP conjugate more strongly inhibited PglC, whereas the more basic CH2-BP analogue was the strongest inhibitor of WecA. These surprising differences indicate different modes of ligand binding for the different PGT superfamilies, implicating a modified P-O- interaction with the structural Mg2+. For the monoPGT enzyme, the two diastereomeric CHF-BP conjugates, which feature a chiral center at the Pα-CHF-Pß carbon, also exhibited strikingly different binding affinities and the inclusion of GlcNAc with the native α-anomer configuration significantly improved binding affinity. UBP-sugars are thus revealed as informative new mechanistic probes of PGTs that may aid development of novel antibiotic agents for the exclusively prokaryotic monoPGT superfamily.


Asunto(s)
Difosfatos , Transferasas , Humanos , Transferasas/química , Uridina , Glicoconjugados/química , Difosfonatos , Azúcares , Uridina Difosfato
5.
Proc Natl Acad Sci U S A ; 118(4)2021 01 26.
Artículo en Inglés | MEDLINE | ID: mdl-33472976

RESUMEN

The monotopic phosphoglycosyl transferase (monoPGT) superfamily comprises over 38,000 nonredundant sequences represented in bacterial and archaeal domains of life. Members of the superfamily catalyze the first membrane-committed step in en bloc oligosaccharide biosynthetic pathways, transferring a phosphosugar from a soluble nucleoside diphosphosugar to a membrane-resident polyprenol phosphate. The singularity of the monoPGT fold and its employment in the pivotal first membrane-committed step allows confident assignment of both protein and corresponding pathway. The diversity of the family is revealed by the generation and analysis of a sequence similarity network for the superfamily, with fusion of monoPGTs with other pathway members being the most frequent and extensive elaboration. Three common fusions were identified: sugar-modifying enzymes, glycosyl transferases, and regulatory domains. Additionally, unexpected fusions of the monoPGT with members of the polytopic PGT superfamily were discovered, implying a possible evolutionary link through the shared polyprenol phosphate substrate. Notably, a phylogenetic reconstruction of the monoPGT superfamily shows a radial burst of functionalization, with a minority of members comprising only the minimal PGT catalytic domain. The commonality and identity of the fusion partners in the monoPGT superfamily is consistent with advantageous colocalization of pathway members at membrane interfaces.


Asunto(s)
Proteínas Bacterianas/química , Glicoconjugados/química , Glicosiltransferasas/química , Bacterias Gramnegativas/enzimología , Bacterias Grampositivas/enzimología , Polisacáridos/química , Secuencia de Aminoácidos , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Sitios de Unión , Citoplasma/enzimología , Citoplasma/genética , Evolución Molecular , Expresión Génica , Redes Reguladoras de Genes , Glicoconjugados/metabolismo , Glicosiltransferasas/genética , Glicosiltransferasas/metabolismo , Bacterias Gramnegativas/clasificación , Bacterias Gramnegativas/genética , Bacterias Grampositivas/clasificación , Bacterias Grampositivas/genética , Redes y Vías Metabólicas/genética , Modelos Moleculares , Periplasma/enzimología , Periplasma/genética , Filogenia , Polisacáridos/metabolismo , Unión Proteica , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta , Dominios y Motivos de Interacción de Proteínas , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Especificidad por Sustrato
6.
Trends Biochem Sci ; 44(1): 7-20, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30337134

RESUMEN

Monotopic membrane proteins, classified by topology, are proteins that embed into a single face of the membrane. These proteins are generally underrepresented in the Protein Data Bank (PDB), but the past decade of research has revealed new examples that allow the description of generalizable features. This Opinion article summarizes shared characteristics including oligomerization states, modes of membrane association, mechanisms of interaction with hydrophobic or amphiphilic substrates, and homology to soluble folds. We also discuss how associations of monotopic enzymes in pathways can be used to promote substrate specificity and product composition. These examples highlight the challenges in structure determination specific to this class of proteins, but also the promise of new understanding from future study of these proteins that reside at the interface.


Asunto(s)
Proteínas de la Membrana/química , Animales , Bases de Datos de Proteínas , Humanos , Interacciones Hidrofóbicas e Hidrofílicas
7.
Protein Expr Purif ; 207: 106273, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37068720

RESUMEN

Phosphoglycosyl transferases (PGTs) are among the first membrane-bound enzymes involved in the biosynthesis of bacterial glycoconjugates. Robust expression and purification protocols for an abundant subfamily of PGTs remains lacking. Recent advancements in detergent-free methods for membrane protein solubilization open the door for purification of difficult membrane proteins directly from cell membranes into native-like liponanoparticles. By leveraging autoinduction, in vivo SUMO tag cleavage, styrene maleic acid co-polymer liponanoparticles (SMALPs), and Strep-Tag purification, we have established a robust workflow for expression and purification of previously unobtainable PGTs. The material generated from this workflow is extremely pure and can be directly visualized by Cryogenic Electron Microscopy (CryoEM). The methods presented here promise to be generalizable to additional membrane proteins recombinantly expressed in E. coli and should be of interest to the greater membrane proteomics community.


Asunto(s)
Escherichia coli , Transferasas , Transferasas/genética , Escherichia coli/genética , Membrana Celular/genética , Proteínas de la Membrana/genética
8.
Org Biomol Chem ; 21(28): 5873-5879, 2023 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-37417819

RESUMEN

Nucleoside analogs show useful bioactive properties. A versatile solid-phase synthesis that readily enables the diversification of thymine-containing nucleoside analogs is presented. The utility of the approach is demonstrated with the preparation of a library of compounds for analysis with SNM1A, a DNA damage repair enzyme that contributes to cytotoxicity. This exploration provided the most promising nucleoside-derived inhibitor of SNM1A to date with an IC50 of 12.3 µM.


Asunto(s)
Nucleósidos , Timina , Nucleósidos/farmacología , Timina/farmacología , Técnicas de Síntesis en Fase Sólida , Exodesoxirribonucleasas/metabolismo , Reparación del ADN
9.
Trends Biochem Sci ; 43(1): 10-17, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29183665

RESUMEN

In the three domains of life, lipid-linked glycans contribute to various cellular processes ranging from protein glycosylation to glycosylphosphatidylinositol anchor biosynthesis to peptidoglycan assembly. In generating many of these glycoconjugates, phosphorylated polyprenol-based lipids are charged with single sugars by polyprenol phosphate glycosyltransferases. The resultant substrates serve as glycosyltransferase donors, complementing the more common nucleoside diphosphate sugars. It had been accepted that these polyprenol phosphate glycosyltransferases acted similarly, given their considerable sequence homology. Recent findings, however, suggest that matters may not be so simple. In this Opinion we propose that the stereochemistry of sugar addition by polyprenol phosphate glycosyltransferases is not conserved across evolution, even though the GT-A fold that characterizes such enzymes is omnipresent.


Asunto(s)
Glicosiltransferasas/metabolismo , Pentanoles/química , Pentanoles/metabolismo , Fosfatos/metabolismo , Polímeros/química , Polímeros/metabolismo , Hemiterpenos , Humanos , Fosfatos/química , Estereoisomerismo
10.
Biochem Soc Trans ; 49(3): 1189-1203, 2021 06 30.
Artículo en Inglés | MEDLINE | ID: mdl-34100892

RESUMEN

Phosphoglycosyl transferases (PGTs) play a pivotal role at the inception of complex glycoconjugate biosynthesis pathways across all domains of life. PGTs promote the first membrane-committed step in the en bloc biosynthetic strategy by catalyzing the transfer of a phospho-sugar from a nucleoside diphospho-sugar to a membrane-resident polyprenol phosphate. Studies on the PGTs have been hampered because they are integral membrane proteins, and often prove to be recalcitrant to expression, purification and analysis. However, in recent years exciting new information has been derived on the structures and the mechanisms of PGTs, revealing the existence of two unique superfamilies of PGT enzymes that enact catalysis at the membrane interface. Genome neighborhood analysis shows that these superfamilies, the polytopic PGT (polyPGT) and monotopic PGT (monoPGT), may initiate different pathways within the same organism. Moreover, the same fundamental two-substrate reaction is enacted through two different chemical mechanisms with distinct modes of catalysis. This review highlights the structural and mechanistic divergence between the PGT enzyme superfamilies and how this is reflected in differences in regulation in their varied glycoconjugate biosynthesis pathways.


Asunto(s)
Proteínas Bacterianas/química , Dominio Catalítico , Glicoconjugados/química , Glicosiltransferasas/química , Proteínas de la Membrana/química , Proteínas Bacterianas/metabolismo , Biocatálisis , Conformación de Carbohidratos , Membrana Celular/enzimología , Membrana Celular/metabolismo , Glicoconjugados/biosíntesis , Glicosiltransferasas/metabolismo , Cinética , Proteínas de la Membrana/metabolismo , Modelos Químicos , Conformación Proteica , Especificidad por Sustrato
11.
Glycobiology ; 30(6): 374-381, 2020 05 19.
Artículo en Inglés | MEDLINE | ID: mdl-31965157

RESUMEN

An in vitro gut-immune co-culture model with apical and basal accessibility, designed to more closely resemble a human intestinal microenvironment, was employed to study the role of the N-linked protein glycosylation pathway in Campylobacter jejuni pathogenicity. The gut-immune co-culture (GIC) was developed to model important aspects of the human small intestine by the inclusion of mucin-producing goblet cells, human enterocytes and dendritic cells, bringing together a mucus-containing epithelial monolayer with elements of the innate immune system. The utility of the system was demonstrated by characterizing host-pathogen interactions facilitated by N-linked glycosylation, such as host epithelial barrier functions, bacterial invasion and immunogenicity. Changes in human intestinal barrier functions in the presence of 11168 C. jejuni (wildtype) strains were quantified using GICs. The glycosylation-impaired strain 11168 ΔpglE was 100-fold less capable of adhering to and invading this intestinal model in cell infectivity assays. Quantification of inflammatory signaling revealed that 11168ΔpglE differentially modulated inflammatory responses in different intestinal microenvironments, suppressive in some but activating in others. Virulence-associated outer membrane vesicles produced by wildtype and 11168ΔpglE C. jejuni were shown to have differential composition and function, with both leading to immune system activation when provided to the gut-immune co-culture model. This analysis of aspects of C. jejuni infectivity in the presence and absence of its N-linked glycome is enabled by application of the gut-immune model, and we anticipate that this system will be applicable to further studies of C. jejuni and other enteropathogens of interest.


Asunto(s)
Campylobacter jejuni/inmunología , Técnicas de Cocultivo , Microbioma Gastrointestinal/inmunología , Interacciones Huésped-Patógeno/inmunología , Polisacáridos/inmunología , Animales , Humanos , Polisacáridos/química
12.
J Am Chem Soc ; 142(5): 2145-2149, 2020 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-31923358

RESUMEN

We report the application of lanthanide-binding tags (LBTs) for two- and three-dimensional X-ray imaging of individual proteins in cells with a sub-15 nm beam. The method combines encoded LBTs, which are tags of minimal size (ca. 15-20 amino acids) affording high-affinity lanthanide ion binding, and X-ray fluorescence microscopy (XFM). This approach enables visualization of LBT-tagged proteins while simultaneously measuring the elemental distribution in cells at a spatial resolution necessary for visualizing cell membranes and eukaryotic subcellular organelles.


Asunto(s)
Imagenología Tridimensional/métodos , Elementos de la Serie de los Lantanoides/metabolismo , Proteínas/química , Espectrometría por Rayos X/métodos , Secuencia de Aminoácidos , Unión Proteica
13.
Chembiochem ; 21(1-2): 108-112, 2020 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-31709708

RESUMEN

High-throughput small-molecule screening in drug discovery processes commonly rely on fluorescence-based methods including fluorescent polarization and fluorescence/Förster resonance energy transfer. These techniques use highly accessible instrumentation; however, they can suffer from high false-negative rates and background signals, or might involve complex schemes for the introduction of fluorophore pairs. Herein we present the synthesis and application of fluorescent nucleoside analogues as the foundation for directed approaches for competitive binding analyses. The general approach describes selective fluorescent environment-sensitive (ES) nucleoside analogues that are adaptable to diverse enzymes that act on nucleoside-based substrates. We demonstrate screening a set of uridine analogues and development of an assay for fragment-based lead discovery with the TcdB glycosyltransferase (GT), an enzyme associated with virulence in Clostridium difficile. The uridine-based probe used for this high-throughput screen has a KD value of 7.2 µm with the TcdB GT and shows a >30-fold increase in fluorescence intensity upon binding. The ES-based probe assay is benchmarked against two other screening approaches.


Asunto(s)
Clostridioides difficile/enzimología , Inhibidores Enzimáticos/farmacología , Colorantes Fluorescentes/farmacología , Glicosiltransferasas/antagonistas & inhibidores , Secuenciación de Nucleótidos de Alto Rendimiento , Nucleósidos/farmacología , Evaluación Preclínica de Medicamentos , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , Colorantes Fluorescentes/síntesis química , Colorantes Fluorescentes/química , Glicosiltransferasas/metabolismo , Modelos Moleculares , Nucleósidos/síntesis química , Nucleósidos/química
14.
Nat Chem Biol ; 14(6): 538-541, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29769739

RESUMEN

Polyprenol phosphate phosphoglycosyl transferases (PGTs) catalyze the first membrane-committed step in assembly of essential glycoconjugates. Currently there is no structure-function information to describe how monotopic PGTs coordinate the reaction between membrane-embedded and soluble substrates. We describe the structure and mode of membrane association of PglC, a PGT from Campylobacter concisus. The structure reveals a unique architecture, provides mechanistic insight and identifies ligand-binding determinants for PglC and the monotopic PGT superfamily.


Asunto(s)
Campylobacter/enzimología , Membrana Celular/enzimología , Glicosiltransferasas/química , Fosfatos/química , Catálisis , Dominio Catalítico , Clonación Molecular , Cisteína/química , Glicoconjugados/química , Ligandos , Mutación , Fosforilación , Dominios Proteicos , Pliegue de Proteína , Estructura Secundaria de Proteína , Relación Estructura-Actividad , Especificidad por Sustrato
15.
Bioorg Med Chem ; 28(18): 115661, 2020 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-32828427

RESUMEN

Nucleoside derivatives, in particular those featuring uridine, are familiar components of the nucleoside family of bioactive natural products. The structural complexity and biological activities of these compounds have inspired research from organic chemistry and chemical biology communities seeking to develop novel approaches to assemble the challenging molecular targets, to gain inspiration for enzyme inhibitor development and to fuel antibiotic discovery efforts. This review will present recent case studies describing the total synthesis and biosynthesis of uridine natural products, and de novo synthetic efforts exploiting features of the natural products to produce simplified scaffolds. This research has culminated in the development of complementary strategies that can lead to effective uridine-based inhibitors and antibiotics. The strengths and challenges of the juxtaposing methods will be illustrated by examining select uridine natural products. Moreover, structure-activity relationships (SAR) for each natural product-inspired scaffold will be discussed, highlighting the impact on inhibitor development, with the aim of future uridine-based small molecule expansion.


Asunto(s)
Productos Biológicos/síntesis química , Inhibidores Enzimáticos/síntesis química , Uridina/síntesis química , Antibacterianos/farmacología , Productos Biológicos/metabolismo , Productos Biológicos/farmacología , Descubrimiento de Drogas , Inhibidores Enzimáticos/metabolismo , Inhibidores Enzimáticos/farmacología , Humanos , Estructura Molecular , Fosfatos/química , Poliprenoles/química , Relación Estructura-Actividad , Uridina/metabolismo , Uridina/farmacología
16.
Proc Natl Acad Sci U S A ; 114(27): 7019-7024, 2017 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-28630348

RESUMEN

Phosphoglycosyl transferases (PGTs) are integral membrane proteins with diverse architectures that catalyze the formation of polyprenol diphosphate-linked glycans via phosphosugar transfer from a nucleotide diphosphate-sugar to a polyprenol phosphate. There are two PGT superfamilies that differ significantly in overall structure and topology. The polytopic PGT superfamily, represented by MraY and WecA, has been the subject of many studies because of its roles in peptidoglycan and O-antigen biosynthesis. In contrast, less is known about a second, extensive superfamily of PGTs that reveals a core structure with dual domain architecture featuring a C-terminal soluble globular domain and a predicted N-terminal membrane-associated domain. Representative members of this superfamily are the Campylobacter PglCs, which initiate N-linked glycoprotein biosynthesis and are implicated in virulence and pathogenicity. Despite the prevalence of dual domain PGTs, their mechanism of action is unknown. Here, we present the mechanistic analysis of PglC, a prototypic dual domain PGT from Campylobacter concisus Using a luminescence-based assay, together with substrate labeling and kinetics-based approaches, complementary experiments were carried out that support a ping-pong mechanism involving a covalent phosphosugar intermediate for PglC. Significantly, mass spectrometry-based approaches identified Asp93, which is part of a highly conserved AspGlu dyad found in all dual domain PGTs, as the active-site nucleophile of the enzyme involved in the formation of the covalent adduct. The existence of a covalent phosphosugar intermediate provides strong support for a ping-pong mechanism of PglC, differing fundamentally from the ternary complex mechanisms of representative polytopic PGTs.


Asunto(s)
Antibacterianos/química , Proteínas Bacterianas/química , Campylobacter/enzimología , Transferasas/química , Ácido Aspártico/química , Dominio Catalítico , Ácido Glutámico/química , Cinética , Luminiscencia , Modelos Químicos , Peptidoglicano/metabolismo , Especificidad por Sustrato , Azúcares/química
17.
Arch Biochem Biophys ; 675: 108111, 2019 10 30.
Artículo en Inglés | MEDLINE | ID: mdl-31563509

RESUMEN

Long-chain polyprenol phosphates feature in membrane-associated glycoconjugate biosynthesis pathways across domains of life. These unique amphiphilic molecules are best known as substrates of polytopic membrane proteins, including polyprenol-phosphate phosphoglycosyl and glycosyl transferases, and as components of more complex substrates. The linear polyprenols are constrained by double bond geometry and lend themselves well to interactions with polytopic membrane proteins, in which multiple transmembrane helices form a rich landscape for interactions. Recently, a new superfamily of monotopic phosphoglycosyl transferase enzymes has been identified that interacts with polyprenol phosphate substrates via a single reentrant membrane helix. Intriguingly, despite the dramatic differences in their membrane-interaction domains, both polytopic and monotopic enzymes similarly favor a unique cis/trans geometry in their polyprenol phosphate substrates. Herein, we present a multipronged biochemical and biophysical study of PglC, a monotopic phosphoglycosyl transferase that catalyzes the first membrane-committed step in N-linked glycoprotein biosynthesis in Campylobacter jejuni. We probe the significance of polyprenol phosphate geometry both in mediating substrate binding to PglC and in modulating the local membrane environment. Geometry is found to be important for binding to PglC; a conserved proline residue in the reentrant membrane helix is determined to drive polyprenol phosphate recognition and specificity. Pyrene fluorescence studies show that polyprenol phosphates at physiologically-relevant levels increase the disorder of the local lipid bilayer; however, this effect is confined to polyprenol phosphates with specific isoprene geometries. The molecular insights from this study may shed new light on the interactions of polyprenol phosphates with diverse membrane-associated proteins in glycoconjugate biosynthesis.


Asunto(s)
Poliprenoles/metabolismo , Transferasas (Grupos de Otros Fosfatos Sustitutos)/metabolismo , Campylobacter jejuni/crecimiento & desarrollo , Campylobacter jejuni/metabolismo , Secuencia Conservada , Fluidez de la Membrana , Lípidos de la Membrana/metabolismo , Unión Proteica , Conformación Proteica , Especificidad por Sustrato , Transferasas (Grupos de Otros Fosfatos Sustitutos)/química
18.
Bioorg Med Chem ; 26(19): 5314-5321, 2018 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-29685683

RESUMEN

The human enteropathogen Campylobacter jejuni, like many bacteria, employs siderophores such as enterobactin for cellular uptake of ferric iron. This transport process has been shown to be essential for virulence and presents an attractive opportunity for further study of the permissiveness of this pathway to small-molecule intervention and as inspiration for the development of synthetic carriers that may effectively transport cargo into Gram-negative bacteria. In this work, we have developed a facile and robust microscale assay to measure growth recovery of C. jejuni NCTC 11168 in liquid culture as a result of ferric iron uptake. In parallel, we have established the solid-phase synthesis of catecholamide compounds modeled on enterobactin fragments. Applying these methodological developments, we show that small synthetic iron chelators of minimal dimensions provide ferric iron to C. jejuni with equal or greater efficiency than enterobactin.


Asunto(s)
Campylobacter jejuni/metabolismo , Enterobactina/análogos & derivados , Hierro/química , Campylobacter jejuni/crecimiento & desarrollo , Enterobactina/síntesis química , Humanos , Hierro/metabolismo , Sideróforos/síntesis química , Sideróforos/química , Técnicas de Síntesis en Fase Sólida
19.
J Biol Chem ; 291(42): 22001-22010, 2016 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-27573243

RESUMEN

Site selectivity of protein N-linked glycosylation is dependent on many factors, including accessibility of the modification site, amino acid composition of the glycosylation consensus sequence, and cellular localization of target proteins. Previous studies have shown that the bacterial oligosaccharyltransferase, PglB, of Campylobacter jejuni favors acceptor proteins with consensus sequences ((D/E)X1NX2(S/T), where X1,2 ≠ proline) in flexible, solvent-exposed motifs; however, several native glycoproteins are known to harbor consensus sequences within structured regions of the acceptor protein, suggesting that unfolding or partial unfolding is required for efficient N-linked glycosylation in the native environment. To derive insight into these observations, we generated structural homology models of the N-linked glycoproteome of C. jejuni This evaluation highlights the potential diversity of secondary structural conformations of previously identified N-linked glycosylation sequons. Detailed assessment of PglB activity with a structurally characterized acceptor protein, PEB3, demonstrated that this natively folded substrate protein is not efficiently glycosylated in vitro, whereas structural destabilization increases glycosylation efficiency. Furthermore, in vivo glycosylation studies in both glyco-competent Escherichia coli and the native system, C. jejuni, revealed that efficient glycosylation of glycoproteins, AcrA and PEB3, depends on translocation to the periplasmic space via the general secretory pathway. Our studies provide quantitative evidence that many acceptor proteins are likely to be N-linked-glycosylated before complete folding and suggest that PglB activity is coupled to general secretion-mediated translocation to the periplasm. This work extends our understanding of the molecular mechanisms underlying N-linked glycosylation in bacteria.


Asunto(s)
Campylobacter jejuni/metabolismo , Proteínas de Escherichia coli/metabolismo , Escherichia coli/metabolismo , Hexosiltransferasas/metabolismo , Lipoproteínas/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Sistemas de Secreción Bacterianos/genética , Sistemas de Secreción Bacterianos/metabolismo , Campylobacter jejuni/genética , Escherichia coli/genética , Proteínas de Escherichia coli/genética , Glicosilación , Hexosiltransferasas/genética , Lipoproteínas/genética , Proteínas de la Membrana/genética , Proteínas de Transporte de Membrana/genética , Periplasma/genética , Periplasma/metabolismo
20.
Glycobiology ; 27(9): 820-833, 2017 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-28810664

RESUMEN

Phosphoglycosyl transferases (PGTs) initiate the biosynthesis of both essential and virulence-associated bacterial glycoconjugates including lipopolysaccharide, peptidoglycan and glycoproteins. PGTs catalyze the transfer of a phosphosugar moiety from a nucleoside diphosphate sugar to a polyprenol phosphate, to form a membrane-bound polyprenol diphosphosugar product. PGTs are integral membrane proteins, which include between 1 and 11 predicted transmembrane domains. Despite this variation, common motifs have been identified in PGT families through bioinformatics and mutagenesis studies. Bacterial PGTs represent important antibacterial and virulence targets due to their significant role in initiating the biosynthesis of key bacterial glycoconjugates. Considerable effort has gone into mechanistic and inhibition studies for this class of enzymes, both of which depend on reliable, high-throughput assays for easy quantification of activity. This review summarizes recent advances made in the characterization of this challenging but important class of enzymes.


Asunto(s)
Membrana Celular/enzimología , Proteínas de Escherichia coli/metabolismo , Escherichia coli/enzimología , Regulación Bacteriana de la Expresión Génica , Glicoconjugados/biosíntesis , Hexosiltransferasas/metabolismo , Metabolismo de los Hidratos de Carbono , Membrana Celular/química , Secuencia Conservada , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Escherichia coli/genética , Proteínas de Escherichia coli/antagonistas & inhibidores , Proteínas de Escherichia coli/genética , Glicoconjugados/química , Glicoconjugados/genética , Hexosiltransferasas/antagonistas & inhibidores , Hexosiltransferasas/genética , Ensayos Analíticos de Alto Rendimiento , Cinética , Dominios Proteicos , Especificidad por Sustrato
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA