Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 151
Filter
Add more filters

Publication year range
1.
Biochemistry ; 60(41): 3098-3113, 2021 10 19.
Article in English | MEDLINE | ID: mdl-34609833

ABSTRACT

The MtrCDE system confers multidrug resistance to Neisseria gonorrhoeae, the causative agent of gonorrhea. Using free and directed molecular dynamics (MD) simulations, we analyzed the interactions between MtrD and azithromycin, a transport substrate of MtrD, and a last-resort clinical treatment for multidrug-resistant gonorrhea. We then simulated the interactions between MtrD and streptomycin, an apparent nonsubstrate of MtrD. Using known conformations of MtrD homologues, we simulated a potential dynamic transport cycle of MtrD using targeted MD techniques (TMD), and we noted that forces were not applied to ligands of interest. In these TMD simulations, we observed the transport of azithromycin and the rejection of streptomycin. In an unbiased, long-time scale simulation of AZY-bound MtrD, we observed the spontaneous diffusion of azithromycin through the periplasmic cleft. Our simulations show how the peristaltic motions of the periplasmic cleft facilitate the transport of substrates by MtrD. Our data also suggest that multiple transport pathways for macrolides may exist within the periplasmic cleft of MtrD.


Subject(s)
Bacterial Proteins/metabolism , Membrane Proteins/metabolism , Membrane Transport Proteins/metabolism , Neisseria gonorrhoeae/chemistry , Azithromycin/chemistry , Azithromycin/metabolism , Bacterial Proteins/chemistry , Binding Sites , Biological Transport , Hydrogen Bonding , Ligands , Membrane Proteins/chemistry , Membrane Transport Proteins/chemistry , Molecular Dynamics Simulation , Protein Binding , Streptomycin/chemistry , Streptomycin/metabolism
2.
J Bacteriol ; 202(8)2020 03 26.
Article in English | MEDLINE | ID: mdl-32041800

ABSTRACT

The Gram-negative pathogen Neisseria gonorrhoeae (gonococcus [Gc]) colonizes lysozyme-rich mucosal surfaces. Lysozyme hydrolyzes peptidoglycan, leading to bacterial lysis. Gc expresses two proteins, SliC and NgACP, that bind and inhibit the enzymatic activity of lysozyme. SliC is a surface-exposed lipoprotein, while NgACP is found in the periplasm and also released extracellularly. Purified SliC and NgACP similarly inhibit lysozyme. However, whereas mutation of ngACP increases Gc susceptibility to lysozyme, the sliC mutant is only susceptible to lysozyme when ngACP is inactivated. In this work, we examined how lipidation contributes to SliC expression, cellular localization, and resistance of Gc to killing by lysozyme. To do so, we mutated the conserved cysteine residue (C18) in the N-terminal lipobox motif of SliC, the site for lipid anchor attachment, to alanine. SliC(C18A) localized to soluble rather than membrane fractions in Gc and was not displayed on the bacterial surface. Less SliC(C18A) was detected in Gc lysates compared to the wild-type protein. This was due in part to some release of the C18A mutant, but not wild-type, protein into the extracellular space. Surprisingly, Gc expressing SliC(C18A) survived better than SliC (wild type)-expressing Gc after exposure to lysozyme. We conclude that lipidation is not required for the ability of SliC to inhibit lysozyme, even though the lipidated cysteine is 100% conserved in Gc SliC alleles. These findings shed light on how members of the growing family of lysozyme inhibitors with distinct subcellular localizations contribute to bacterial defense against lysozyme.IMPORTANCENeisseria gonorrhoeae is one of many bacterial species that express multiple lysozyme inhibitors. It is unclear how inhibitors that differ in their subcellular localization contribute to defense from lysozyme. We investigated how lipidation of SliC, an MliC (membrane-bound lysozyme inhibitor of c-type lysozyme)-type inhibitor, contributes to its localization and lysozyme inhibitory activity. We found that lipidation was required for surface exposure of SliC and yet was dispensable for protecting the gonococcus from killing by lysozyme. To our knowledge, this is the first time the role of lipid anchoring of a lysozyme inhibitor has been investigated. These results help us understand how different lysozyme inhibitors are localized in bacteria and how this impacts resistance to lysozyme.


Subject(s)
Bacterial Proteins/metabolism , Enzyme Inhibitors/metabolism , Gonorrhea/microbiology , Lipoproteins/metabolism , Muramidase/antagonists & inhibitors , Neisseria gonorrhoeae/metabolism , Amino Acid Motifs , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Enzyme Inhibitors/chemistry , Gonorrhea/enzymology , Host-Pathogen Interactions , Humans , Lipoproteins/chemistry , Lipoproteins/genetics , Muramidase/metabolism , Neisseria gonorrhoeae/chemistry , Neisseria gonorrhoeae/genetics , Periplasm/genetics , Periplasm/metabolism , Protein Transport
3.
Molecules ; 25(20)2020 Oct 16.
Article in English | MEDLINE | ID: mdl-33081246

ABSTRACT

Aminoacyl-tRNA synthetases (aaRSs) have become viable targets for the development of antimicrobial agents due to their crucial role in protein translation. A series of six amino acids were coupled to the purine-like 7-amino-5-hydroxymethylbenzimidazole nucleoside analogue following an optimized synthetic pathway. These compounds were designed as aaRS inhibitors and can be considered as 1,3-dideazaadenine analogues carrying a 2-hydroxymethyl substituent. Despite our intentions to obtain N1-glycosylated 4-aminobenzimidazole congeners, resembling the natural purine nucleosides glycosylated at the N9-position, we obtained the N3-glycosylated benzimidazole derivatives as the major products, resembling the respective purine N7-glycosylated nucleosides. A series of X-ray crystal structures of class I and II aaRSs in complex with newly synthesized compounds revealed interesting interactions of these "base-flipped" analogues with their targets. While the exocyclic amine of the flipped base mimics the reciprocal interaction of the N3-purine atom of aminoacyl-sulfamoyl adenosine (aaSA) congeners, the hydroxymethyl substituent of the flipped base apparently loses part of the standard interactions of the adenine N1 and the N6-amine as seen with aaSA analogues. Upon the evaluation of the inhibitory potency of the newly obtained analogues, nanomolar inhibitory activities were noted for the leucine and isoleucine analogues targeting class I aaRS enzymes, while rather weak inhibitory activity against the corresponding class II aaRSs was observed. This class bias could be further explained by detailed structural analysis.


Subject(s)
Amino Acyl-tRNA Synthetases/ultrastructure , Benzimidazoles/chemistry , Enzyme Inhibitors/chemical synthesis , Ribonucleosides/chemistry , Amino Acyl-tRNA Synthetases/antagonists & inhibitors , Amino Acyl-tRNA Synthetases/chemistry , Benzimidazoles/chemical synthesis , Benzimidazoles/pharmacology , Crystallography, X-Ray , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Humans , Neisseria gonorrhoeae/chemistry , Neisseria gonorrhoeae/enzymology , Neisseria gonorrhoeae/pathogenicity , Protein Conformation/drug effects , Structure-Activity Relationship
4.
Dokl Biochem Biophys ; 495(1): 334-337, 2020 Nov.
Article in English | MEDLINE | ID: mdl-33368046

ABSTRACT

Three-dimensional full-atom model of the enzyme complex with acetyl-CoA and substrate was constructed on the basis of the primary sequence of amino acid residues of N-acetyl glutamate synthase. Bioinformatics approaches of computer modeling were applied, including multiple sequence alignment, prediction of co-evolutionary contacts, and ab initio folding. On the basis of the results of calculations by classical molecular dynamics and combined quantum and molecular mechanics (QM/MM) methods, the structure of the active site and the reaction mechanism of N-acetylglutamate formation are described. Agreement of the structures of the enzyme-product complexes obtained in computer modeling and in the X-ray studies validates the reliability of modeling predictions.


Subject(s)
Amino-Acid N-Acetyltransferase/chemistry , Amino-Acid N-Acetyltransferase/metabolism , Neisseria gonorrhoeae/enzymology , Catalysis , Catalytic Domain , Computer Simulation , Crystallography, X-Ray , Models, Molecular , Neisseria gonorrhoeae/chemistry , Neisseria gonorrhoeae/metabolism , Structure-Activity Relationship , Substrate Specificity
5.
J Bacteriol ; 201(20)2019 10 15.
Article in English | MEDLINE | ID: mdl-31331979

ABSTRACT

Neisseria gonorrhoeae responds to host-derived antimicrobials by inducing the expression of the mtrCDE-encoded multidrug efflux pump, which expels microbicides, such as bile salts, fatty acids, and multiple extrinsically administered drugs, from the cell. In the absence of these cytotoxins, the TetR family member MtrR represses the mtrCDE genes. Although antimicrobial-dependent derepression of mtrCDE is clear, the physiological inducers of MtrR are unknown. Here, we report the crystal structure of an induced form of MtrR. In the binding pocket of MtrR, we observed electron density that we hypothesized was N-cyclohexyl-3-aminopropanesulfonic acid (CAPS), a component of the crystallization reagent. Using the MtrR-CAPS structure as an inducer-bound template, we hypothesized that bile salts, which bear significant chemical resemblance to CAPS, are physiologically relevant inducers. Indeed, characterization of MtrR-chenodeoxycholate and MtrR-taurodeoxycholate interactions, both in vitro and in vivo, revealed that these bile salts, but not glyocholate or taurocholate, bind MtrR tightly and can act as bona fide inducers. Furthermore, two residues, W136 and R176, were shown to be important in binding chenodeoxycholate but not taurodeoxycholate, suggesting different binding modes of the bile salts. These data provide insight into a crucial mechanism utilized by the pathogen to overcome innate human defenses.IMPORTANCENeisseria gonorrhoeae causes a significant disease burden worldwide, and a meteoric rise in its multidrug resistance has reduced the efficacy of antibiotics previously or currently approved for therapy of gonorrheal infections. The multidrug efflux pump MtrCDE transports multiple drugs and host-derived antimicrobials from the bacterial cell and confers survival advantage on the pathogen within the host. Transcription of the pump is repressed by MtrR but relieved by the cytosolic influx of antimicrobials. Here, we describe the structure of induced MtrR and use this structure to identify bile salts as physiological inducers of MtrR. These findings provide a mechanistic basis for antimicrobial sensing and gonococcal protection by MtrR through the derepression of mtrCDE expression after exposure to intrinsic and clinically applied antimicrobials.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Drug Resistance, Multiple, Bacterial , Neisseria gonorrhoeae/pathogenicity , Repressor Proteins/chemistry , Repressor Proteins/metabolism , Binding Sites , Chenodeoxycholic Acid/metabolism , Crystallography, X-Ray , Humans , Models, Molecular , Neisseria gonorrhoeae/chemistry , Neisseria gonorrhoeae/metabolism , Protein Binding , Taurodeoxycholic Acid/metabolism
6.
J Biol Chem ; 293(4): 1106-1119, 2018 01 26.
Article in English | MEDLINE | ID: mdl-29229778

ABSTRACT

The ß-barrel assembly machinery (BAM) is a conserved multicomponent protein complex responsible for the biogenesis of ß-barrel outer membrane proteins (OMPs) in Gram-negative bacteria. Given its role in the production of OMPs for survival and pathogenesis, BAM represents an attractive target for the development of therapeutic interventions, including drugs and vaccines against multidrug-resistant bacteria such as Neisseria gonorrhoeae The first structure of BamA, the central component of BAM, was from N. gonorrhoeae, the etiological agent of the sexually transmitted disease gonorrhea. To aid in pharmaceutical targeting of BAM, we expanded our studies to BamD and BamE within BAM of this clinically relevant human pathogen. We found that the presence of BamD, but not BamE, is essential for gonococcal viability. However, BamE, but not BamD, was cell-surface-displayed under native conditions; however, in the absence of BamE, BamD indeed becomes surface-exposed. Loss of BamE altered cell envelope composition, leading to slower growth and an increase in both antibiotic susceptibility and formation of membrane vesicles containing greater amounts of vaccine antigens. Both BamD and BamE are expressed in diverse gonococcal isolates, under host-relevant conditions, and throughout different phases of growth. The solved structures of Neisseria BamD and BamE share overall folds with Escherichia coli proteins but contain differences that may be important for function. Together, these studies highlight that, although BAM is conserved across Gram-negative bacteria, structural and functional differences do exist across species, which may be leveraged in the development of species-specific therapeutics in the effort to combat multidrug resistance.


Subject(s)
Bacterial Outer Membrane Proteins/chemistry , Neisseria gonorrhoeae/chemistry , Bacterial Outer Membrane Proteins/genetics , Humans , Microbial Viability , Neisseria gonorrhoeae/genetics , Protein Domains , Structure-Activity Relationship
7.
Nature ; 501(7467): 385-90, 2013 Sep 19.
Article in English | MEDLINE | ID: mdl-23995689

ABSTRACT

ß-barrel membrane proteins are essential for nutrient import, signalling, motility and survival. In Gram-negative bacteria, the ß-barrel assembly machinery (BAM) complex is responsible for the biogenesis of ß-barrel membrane proteins, with homologous complexes found in mitochondria and chloroplasts. Here we describe the structure of BamA, the central and essential component of the BAM complex, from two species of bacteria: Neisseria gonorrhoeae and Haemophilus ducreyi. BamA consists of a large periplasmic domain attached to a 16-strand transmembrane ß-barrel domain. Three structural features shed light on the mechanism by which BamA catalyses ß-barrel assembly. First, the interior cavity is accessible in one BamA structure and conformationally closed in the other. Second, an exterior rim of the ß-barrel has a distinctly narrowed hydrophobic surface, locally destabilizing the outer membrane. And third, the ß-barrel can undergo lateral opening, suggesting a route from the interior cavity in BamA into the outer membrane.


Subject(s)
Bacterial Outer Membrane Proteins/biosynthesis , Bacterial Outer Membrane Proteins/chemistry , Haemophilus/chemistry , Neisseria gonorrhoeae/chemistry , Bacterial Outer Membrane Proteins/genetics , Cell Membrane/chemistry , Cell Membrane/metabolism , Crystallography, X-Ray , Escherichia coli/chemistry , Escherichia coli/genetics , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Hydrophobic and Hydrophilic Interactions , Models, Molecular , Mutagenesis , Protein Conformation , Structural Homology, Protein
8.
Infect Immun ; 86(8)2018 08.
Article in English | MEDLINE | ID: mdl-29844237

ABSTRACT

Sialylation of lacto-N-neotetraose (LNnT) extending from heptose I (HepI) of gonococcal lipooligosaccharide (LOS) contributes to pathogenesis. Previously, gonococcal LOS sialyltransterase (Lst) was shown to sialylate LOS in Triton X-100 extracts of strain 15253, which expresses lactose from both HepI and HepII, the minimal structure required for monoclonal antibody (MAb) 2C7 binding. Ongoing work has shown that growth of 15253 in cytidine monophospho-N-acetylneuraminic acid (CMP-Neu5Ac)-containing medium enables binding to CD33/Siglec-3, a cell surface receptor that binds sialic acid, suggesting that lactose termini on LOSs of intact gonococci can be sialylated. Neu5Ac was detected on LOSs of strains 15253 and an MS11 mutant with lactose only from HepI and HepII by mass spectrometry; deleting HepII lactose rendered Neu5Ac undetectable. Resistance of HepII lactose Neu5Ac to desialylation by α2-3-specific neuraminidase suggested an α2-6 linkage. Although not associated with increased factor H binding, HepII lactose sialylation inhibited complement C3 deposition on gonococci. Strain 15253 mutants that lacked Lst or HepII lactose were significantly attenuated in mice, confirming the importance of HepII Neu5Ac in virulence. All 75 minimally passaged clinical isolates from Nanjing, China, expressed HepII lactose, evidenced by reactivity with MAb 2C7; MAb 2C7 was bactericidal against the first 62 (of 75) isolates that had been collected sequentially and were sialylated before testing. MAb 2C7 effectively attenuated 15253 vaginal colonization in mice. In conclusion, this novel sialylation site could explain the ubiquity of gonococcal HepII lactose in vivo Our findings reinforce the candidacy of the 2C7 epitope as a vaccine antigen and MAb 2C7 as an immunotherapeutic antibody.


Subject(s)
Gonorrhea/microbiology , Heptoses/metabolism , Lactose/metabolism , Lipopolysaccharides/metabolism , N-Acetylneuraminic Acid/metabolism , Neisseria gonorrhoeae/metabolism , Neisseria gonorrhoeae/pathogenicity , Adult , Animals , Antibodies, Bacterial/immunology , Antibodies, Bacterial/metabolism , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/metabolism , China , Disease Models, Animal , Female , Healthy Volunteers , Humans , Lipopolysaccharides/chemistry , Male , Mass Spectrometry , Mice , Microbial Viability/drug effects , N-Acetylneuraminic Acid/analysis , Neisseria gonorrhoeae/chemistry , Neisseria gonorrhoeae/isolation & purification
9.
Biochem J ; 474(6): 1017-1039, 2017 03 07.
Article in English | MEDLINE | ID: mdl-28126741

ABSTRACT

Guanosine 5'-triphosphate (GTP) cyclohydrolase-I (GCYH-I) catalyzes the first step in folic acid biosynthesis in bacteria and plants, biopterin biosynthesis in mammals, and the biosynthesis of 7-deazaguanosine-modified tRNA nucleosides in bacteria and archaea. The type IB GCYH (GCYH-IB) is a prokaryotic-specific enzyme found in many pathogens. GCYH-IB is structurally distinct from the canonical type IA GCYH involved in biopterin biosynthesis in humans and animals, and thus is of interest as a potential antibacterial drug target. We report kinetic and inhibition data of Neisseria gonorrhoeae GCYH-IB and two high-resolution crystal structures of the enzyme; one in complex with the reaction intermediate analog and competitive inhibitor 8-oxoguanosine 5'-triphosphate (8-oxo-GTP), and one with a tris(hydroxymethyl)aminomethane molecule bound in the active site and mimicking another reaction intermediate. Comparison with the type IA enzyme bound to 8-oxo-GTP (guanosine 5'-triphosphate) reveals an inverted mode of binding of the inhibitor ribosyl moiety and, together with site-directed mutagenesis data, shows that the two enzymes utilize different strategies for catalysis. Notably, the inhibitor interacts with a conserved active-site Cys149, and this residue is S-nitrosylated in the structures. This is the first structural characterization of a biologically S-nitrosylated bacterial protein. Mutagenesis and biochemical analyses demonstrate that Cys149 is essential for the cyclohydrolase reaction, and S-nitrosylation maintains enzyme activity, suggesting a potential role of the S-nitrosothiol in catalysis.


Subject(s)
Bacterial Proteins/chemistry , GTP Cyclohydrolase/chemistry , Guanosine Triphosphate/analogs & derivatives , Neisseria gonorrhoeae/chemistry , Tromethamine/chemistry , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Catalytic Domain , Cloning, Molecular , Crystallography, X-Ray , Enzyme Inhibitors/chemistry , Escherichia coli/genetics , Escherichia coli/metabolism , GTP Cyclohydrolase/antagonists & inhibitors , GTP Cyclohydrolase/genetics , GTP Cyclohydrolase/metabolism , Gene Expression , Guanosine Triphosphate/chemistry , Kinetics , Models, Molecular , Mutation , Neisseria gonorrhoeae/enzymology , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , S-Nitrosothiols/chemistry , Substrate Specificity
10.
J Biol Chem ; 291(18): 9818-26, 2016 Apr 29.
Article in English | MEDLINE | ID: mdl-26975373

ABSTRACT

Multidrug and toxic compound extrusion (MATE) transporters contribute to multidrug resistance by extruding different drugs across cell membranes. The MATE transporters alternate between their extracellular and intracellular facing conformations to propel drug export, but how these structural changes occur is unclear. Here we combine site-specific cross-linking and functional studies to probe the movement of transmembrane helices in NorM from Neiserria gonorrheae (NorM-NG), a MATE transporter with known extracellular facing structure. We generated an active, cysteine-less NorM-NG and conducted pairwise cysteine mutagenesis on this variant. We found that copper phenanthroline catalyzed disulfide bond formation within five cysteine pairs and increased the electrophoretic mobility of the corresponding mutants. Furthermore, copper phenanthroline abolished the activity of the five paired cysteine mutants, suggesting that these substituted amino acids come in spatial proximity during transport, and the proximity changes are functionally indispensable. Our data also implied that the substrate-binding transmembrane helices move up to 10 Å in NorM-NG during transport and afforded distance restraints for modeling the intracellular facing transporter, thereby casting new light on the underlying mechanism.


Subject(s)
Antiporters/chemistry , Bacterial Proteins/chemistry , Disulfides/chemistry , Neisseria gonorrhoeae/chemistry , Antiporters/genetics , Bacterial Proteins/genetics , Biological Transport, Active , Neisseria gonorrhoeae/genetics , Protein Structure, Secondary
11.
Biochim Biophys Acta ; 1857(2): 169-176, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26589091

ABSTRACT

Neisseria gonorrhoeae colonizes the genitourinary track, and in these environments, especially in the female host, the bacteria are subjected to low levels of oxygen, and reactive oxygen and nitrosyl species. Here, the biochemical characterization of N. gonorrhoeae Laz is presented, as well as, the solution structure of its soluble domain determined by NMR. N. gonorrhoeae Laz is a type 1 copper protein of the azurin-family based on its spectroscopic properties and structure, with a redox potential of 277±5 mV, at pH7.0, that behaves as a monomer in solution. The globular Laz soluble domain adopts the Greek-key motif, with the copper center located at one end of the ß-barrel coordinated by Gly48, His49, Cys113, His118 and Met122, in a distorted trigonal geometry. The edge of the His118 imidazole ring is water exposed, in a surface that is proposed to be involved in the interaction with its redox partners. The heterologously expressed Laz was shown to be a competent electron donor to N. gonorrhoeae cytochrome c peroxidase. This is an evidence for its involvement in the mechanism of protection against hydrogen peroxide generated by neighboring lactobacilli in the host environment.


Subject(s)
Azurin/chemistry , Copper/chemistry , Cytochrome-c Peroxidase/chemistry , Electrons , Hydrogen Peroxide/chemistry , Neisseria gonorrhoeae/chemistry , Amino Acid Sequence , Azurin/genetics , Azurin/metabolism , Cloning, Molecular , Copper/metabolism , Cytochrome-c Peroxidase/genetics , Cytochrome-c Peroxidase/metabolism , Electron Transport , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Models, Molecular , Molecular Sequence Data , Neisseria gonorrhoeae/enzymology , Oxidation-Reduction , Protein Folding , Protein Structure, Secondary , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Substrate Specificity
12.
Glycobiology ; 27(9): 888-899, 2017 09 01.
Article in English | MEDLINE | ID: mdl-28460017

ABSTRACT

O-acetylation is a common modification of bacterial glycoconjugates. By modifying oligosaccharide structure and chemistry, O-acetylation has important consequences for biotic and abiotic recognition events and thus bacterial fitness in general. Previous studies of the broad-spectrum O-linked protein glycosylation in pathogenic Neisseria species (including N. gonorrhoeae and N. meningitidis) have revealed O-acetylation of some of their diverse glycoforms and identified the committed acetylase, PglI. Herein, we extend these observations by using mass spectrometry to examine a complete set of all glycan variants identified to date. Regardless of composition, all glycoforms and all sugars in the oligosaccharide are subject to acetylation in a PglI-dependent fashion with the only exception of di-N-acetyl-bacillosamine. Moreover, multiple sugars in a single oligosaccharide could be simultaneously modified. Interestingly, O-acetylation status was found to be correlated with altered chain lengths of oligosaccharides expressed in otherwise isogenic backgrounds. Models for how this unprecedented phenomenon might arise are discussed with some having potentially important implications for the membrane topology of glycan O-acetylation. Together, the findings provide better insight into how O-acetylation can both directly and indirectly govern glycoform structure and diversity.


Subject(s)
Acetyltransferases/metabolism , Bacterial Proteins/metabolism , Fimbriae Proteins/metabolism , Glycosyltransferases/metabolism , Neisseria gonorrhoeae/metabolism , Polysaccharides/biosynthesis , Protein Processing, Post-Translational , Acetylation , Acetyltransferases/chemistry , Acetyltransferases/genetics , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Carbohydrate Sequence , Cell Membrane/chemistry , Cell Membrane/metabolism , Fimbriae Proteins/chemistry , Fimbriae Proteins/genetics , Glycoconjugates/chemistry , Glycoconjugates/metabolism , Glycosylation , Glycosyltransferases/chemistry , Glycosyltransferases/genetics , Neisseria gonorrhoeae/chemistry , Neisseria gonorrhoeae/genetics , Oligosaccharides/chemistry , Oligosaccharides/metabolism , Polysaccharides/chemistry , Spectrometry, Mass, Electrospray Ionization
13.
J Bacteriol ; 198(21): 3029-3040, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27551020

ABSTRACT

Neisseria gonorrhoeae (gonococci) and Neisseria meningitidis (meningococci) are human pathogens that cause gonorrhea and meningococcal meningitis, respectively. Both N. gonorrhoeae and N. meningitidis release a number of small peptidoglycan (PG) fragments, including proinflammatory PG monomers, although N. meningitidis releases fewer PG monomers. The PG fragments released by N. gonorrhoeae and N. meningitidis are generated in the periplasm during cell wall remodeling, and a majority of these fragments are transported into the cytoplasm by an inner membrane permease, AmpG; however, a portion of the PG fragments are released into the extracellular environment through unknown mechanisms. We previously reported that the expression of meningococcal ampG in N. gonorrhoeae reduced PG monomer release by gonococci. This finding suggested that the efficiency of AmpG-mediated PG fragment recycling regulates the amount of PG fragments released into the extracellular milieu. We determined that three AmpG residues near the C-terminal end of the protein modulate AmpG's efficiency. We also investigated the association between PG fragment recycling and release in two species of human-associated nonpathogenic Neisseria: N. sicca and N. mucosa Both N. sicca and N. mucosa release lower levels of PG fragments and are more efficient at recycling PG fragments than N. gonorrhoeae Our results suggest that N. gonorrhoeae has evolved to increase the amounts of toxic PG fragments released by reducing its PG recycling efficiency. IMPORTANCE: Neisseria gonorrhoeae and Neisseria meningitidis are human pathogens that cause highly inflammatory diseases, although N. meningitidis is also frequently found as a normal member of the nasopharyngeal microbiota. Nonpathogenic Neisseria, such as N. sicca and N. mucosa, also colonize the nasopharynx without causing disease. Although all four species release peptidoglycan fragments, N. gonorrhoeae is the least efficient at recycling and releases the largest amount of proinflammatory peptidoglycan monomers, partly due to differences in the recycling permease AmpG. Studying the interplay between bacterial physiology (peptidoglycan metabolism) and pathogenesis (release of toxic monomers) leads to an increased understanding of how different bacterial species maintain asymptomatic colonization or cause disease and may contribute to efforts to mitigate disease.


Subject(s)
Bacterial Proteins/metabolism , Membrane Transport Proteins/metabolism , Neisseria gonorrhoeae/enzymology , Neisseria meningitidis/enzymology , Neisseriaceae Infections/microbiology , Peptidoglycan/metabolism , Amino Acid Motifs , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Cell Wall/chemistry , Cell Wall/genetics , Cell Wall/metabolism , Membrane Transport Proteins/chemistry , Membrane Transport Proteins/genetics , Neisseria/classification , Neisseria/enzymology , Neisseria/growth & development , Neisseria/metabolism , Neisseria gonorrhoeae/chemistry , Neisseria gonorrhoeae/cytology , Neisseria gonorrhoeae/growth & development , Neisseria meningitidis/chemistry , Neisseria meningitidis/cytology , Neisseria meningitidis/growth & development , Peptidoglycan/chemistry , Peptidoglycan/toxicity
14.
Microbiology (Reading) ; 162(1): 177-190, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26475082

ABSTRACT

Initially, pilE transcription in Neisseria gonorrhoeae appeared to be complicated, yet it was eventually simplified into a model where integration host factor activates a single -35/ -10 promoter. However, with the advent of high-throughput RNA sequencing, numerous small pil-specific RNAs (sense as well as antisense) have been identified at the pilE locus as well as at various pilS loci. Using a combination of in vitro transcription, site-directed mutagenesis, Northern analysis and quantitative reverse transcriptase PCR (qRT-PCR) analysis, we have identified three additional non-canonical promoter elements within the pilE gene; two are located within the midgene region (one sense and one antisense), with the third, an antisense promoter, located immediately downstream of the pilE ORF. Using strand-specific qRT-PCR analysis, an inverse correlation exists between the level of antisense expression and the amount of sense message. By their nature, promoter sequences tend to be AT-rich. In Escherichia coli, the small DNA-binding protein H-NS binds to AT-rich sequences and inhibits intragenic transcription. In N. gonorrhoeae hns mutants, pilE antisense transcription was increased twofold, with a concomitant decrease in sense transcript levels. However, most noticeably in these mutants, the absence of H-NS protein caused pilE/pilS recombination to increase dramatically when compared with WT values. Consequently, H-NS protein suppresses pilE intragenic transcription as well as antigenic variation through the pilE/pilS recombination system.


Subject(s)
Antigenic Variation , Bacterial Proteins/genetics , DNA-Binding Proteins/genetics , Fimbriae Proteins/genetics , Fimbriae Proteins/immunology , Neisseria gonorrhoeae/genetics , Bacterial Proteins/immunology , Base Sequence , DNA-Binding Proteins/immunology , Gene Expression Regulation, Bacterial , Molecular Sequence Data , Neisseria gonorrhoeae/chemistry , Neisseria gonorrhoeae/immunology , Promoter Regions, Genetic , Recombination, Genetic , Sequence Alignment , Transcription, Genetic
15.
Microbiology (Reading) ; 162(11): 2005-2016, 2016 11.
Article in English | MEDLINE | ID: mdl-27590250

ABSTRACT

Regulation of the Neisseria gonorrhoeae pilE gene is ill-defined. In this study, post-transcriptional effects on expression were assessed. In silico analysis predicts the formation of three putative stable stem-loop structures with favourable free energies within the 5' untranslated region of the pilE message. Using quantitative reverse transcriptase PCR analyses, we show that each loop structure forms, with introduced destabilizing stem-loop mutations diminishing loop stability. Utilizing a series of pilE translational fusions, deletion of either loop 1 or loop 2 caused a significant reduction of pilE mRNA resulting in reduced expression of the reporter gene. Consequently, the formation of the loops apparently protects the pilE transcript from degradation. Putative loop 3 contains the pilE ribosomal binding site. Consequently, its formation may influence translation. Analysis of a small RNA transcriptome revealed an antisense RNA being produced upstream of the pilE promoter that is predicted to hybridize across the 5' untranslated region loops. Insertional mutants were created where the antisense RNA is not transcribed. In these mutants, pilE transcript levels are greatly diminished, with any residual message apparently not being translated. Complementation of these insertion mutants in trans with the antisense RNA gene facilitates pilE translation yielding a pilus + phenotype. Overall, this study demonstrates a complex relationship between loop-dependent transcript protection and antisense RNA in modulating pilE expression levels.


Subject(s)
5' Untranslated Regions , Fimbriae Proteins/genetics , Gene Expression Regulation, Bacterial , Neisseria gonorrhoeae/metabolism , RNA, Antisense/chemistry , RNA, Antisense/metabolism , RNA, Bacterial/chemistry , RNA, Bacterial/metabolism , Fimbriae Proteins/chemistry , Fimbriae Proteins/metabolism , Inverted Repeat Sequences , Neisseria gonorrhoeae/chemistry , Neisseria gonorrhoeae/genetics , Nucleic Acid Conformation , RNA, Antisense/genetics , RNA, Bacterial/genetics
16.
J Clin Microbiol ; 54(4): 1130-2, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26763972

ABSTRACT

Identification ofNeisseria gonorrhoeaeby the Bruker matrix-assisted laser desorption ionization-time of flight mass spectrometry (MALDI-TOF MS) system may be affected by "B consistency categorization." A supplementary database of 17N. gonorrhoeaemain spectra was constructed. Twelve of 64N. gonorrhoeaeidentifications were categorized with B consistency, which disappeared using the supplementary database. Database extension did not result in misidentification ofNeisseria meningitidis.


Subject(s)
Computational Biology/methods , Databases as Topic , Neisseria gonorrhoeae/isolation & purification , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Female , Humans , Male , Neisseria gonorrhoeae/chemistry
17.
Anal Biochem ; 510: 33-40, 2016 10 01.
Article in English | MEDLINE | ID: mdl-27325503

ABSTRACT

Nucleic acid-based detection of gonorrhea infections typically require a two-step process involving isolation of the nucleic acid, followed by detection of the genomic target often involving polymerase chain reaction (PCR)-based approaches. In an effort to improve on current detection approaches, we have developed a unique two-step microwave-accelerated approach for rapid extraction and detection of Neisseria gonorrhoeae (gonorrhea, GC) DNA. Our approach is based on the use of highly focused microwave radiation to rapidly lyse bacterial cells, release, and subsequently fragment microbial DNA. The DNA target is then detected by a process known as microwave-accelerated metal-enhanced fluorescence (MAMEF), an ultra-sensitive direct DNA detection analytical technique. In the current study, we show that highly focused microwaves at 2.45 GHz, using 12.3-mm gold film equilateral triangles, are able to rapidly lyse both bacteria cells and fragment DNA in a time- and microwave power-dependent manner. Detection of the extracted DNA can be performed by MAMEF, without the need for DNA amplification, in less than 10 min total time or by other PCR-based approaches. Collectively, the use of a microwave-accelerated method for the release and detection of DNA represents a significant step forward toward the development of a point-of-care (POC) platform for detection of gonorrhea infections.


Subject(s)
DNA, Bacterial , Microwaves , Neisseria gonorrhoeae/chemistry , DNA, Bacterial/analysis , DNA, Bacterial/chemistry , DNA, Bacterial/isolation & purification
18.
J Theor Biol ; 410: 36-43, 2016 12 07.
Article in English | MEDLINE | ID: mdl-27596531

ABSTRACT

OBJECTIVES: Resistance to the currently recommended extended-spectrum cephalosporins, which is used to treat Gonorrhea, is increasing continuously and leading to a threat of untreatable infection. It is, therefore, becoming extremely essential to search for new therapeutic strategies to control Gonorrhea. Vaccination may be considered as an effective control measure to control this disease, which is caused by Neisseria gonorrhoeae. METHODS: In-silico hierarchical approach was used to help identify candidate proteins of N. gonorrhoeae that might contribute significantly in vaccine research. In contrast to the conventional vaccine research which requires at least 10-12 years, the present approach would reduce the time period drastically and help to identify Potential Universal Vaccine Candidates (PUVCs). These proteins were further analyzed for the presence of T-cell and linear B-cell epitopes, by using HLAPred and ABCpred servers respectively, in order to facilitate the identification of Multi Epitope Peptide Vaccine Constructs. RESULTS: We have identified 23 non-host candidate proteins, using the proteomic information of four sequenced strains of N. gonorrhoeae namely FA 1090, TCDC_NG08107, NCCP11945 and MS11 and labeled them as PUVCs. Since all these identified 23 PUVCs contained both T cell and B cell epitopes, these have been further reiterated as PUVCs which could be used as promising leads for vaccine development. CONCLUSIONS: This hierarchical approach is the first comprehensive study to identify potential vaccine candidates which once utilized for vaccine development would surely serve as promising tools for effective control of Gonorrhea.


Subject(s)
Bacterial Vaccines/immunology , Computer Simulation , Epitopes, B-Lymphocyte/immunology , Epitopes, T-Lymphocyte/immunology , Neisseria gonorrhoeae/immunology , Bacterial Vaccines/chemistry , Bacterial Vaccines/therapeutic use , Epitopes, B-Lymphocyte/chemistry , Epitopes, T-Lymphocyte/chemistry , Gonorrhea/immunology , Gonorrhea/prevention & control , Humans , Neisseria gonorrhoeae/chemistry , Proteomics
19.
Infect Immun ; 83(11): 4438-49, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26351283

ABSTRACT

Neisseria gonorrhoeae, the causative agent of the sexually transmitted infection gonorrhea, is not preventable by vaccination and is rapidly developing resistance to antibiotics. However, the transferrin (Tf) receptor system, composed of TbpA and TbpB, is an ideal target for novel therapeutics and vaccine development. Using a three-dimensional structure of gonococcal TbpA, we investigated two hypotheses, i.e., that loop-derived antibodies can interrupt ligand-receptor interactions in the native bacterium and that the loop 3 helix is a critical functional domain. Preliminary loop-derived antibodies, as well as optimized second-generation antibodies, demonstrated similar modest ligand-blocking effects on the gonococcal surface but different effects in Escherichia coli. Mutagenesis of loop 3 helix residues was employed, generating 11 mutants. We separately analyzed the mutants' abilities to (i) bind Tf and (ii) internalize Tf-bound iron in the absence of the coreceptor TbpB. Single residue mutations resulted in up to 60% reductions in ligand binding and up to 85% reductions in iron utilization. All strains were capable of growing on Tf as the sole iron source. Interestingly, in the presence of TbpB, only a 30% reduction in Tf-iron utilization was observed, indicating that the coreceptor can compensate for TbpA impairment. Complete deletion of the loop 3 helix of TbpA eliminated the abilities to bind Tf, internalize iron, and grow with Tf as the sole iron source. Our studies demonstrate that while the loop 3 helix is a key functional domain, its function does not exclusively rely on any single residue.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Neisseria gonorrhoeae/metabolism , Transferrin-Binding Protein A/chemistry , Transferrin-Binding Protein A/metabolism , Bacterial Proteins/genetics , Bacterial Vaccines/chemistry , Bacterial Vaccines/genetics , Bacterial Vaccines/metabolism , Gonorrhea/genetics , Gonorrhea/metabolism , Gonorrhea/microbiology , Humans , Neisseria gonorrhoeae/chemistry , Neisseria gonorrhoeae/genetics , Protein Binding , Protein Structure, Secondary , Transferrin/genetics , Transferrin/metabolism , Transferrin-Binding Protein A/genetics
20.
Acta Crystallogr D Biol Crystallogr ; 71(Pt 11): 2287-96, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26527145

ABSTRACT

NorM from Neisseria gonorrhoeae (NorM-NG) belongs to the multidrug and toxic compound extrusion (MATE) family of membrane-transport proteins, which can extrude cytotoxic chemicals across cell membranes and confer multidrug resistance. Here, the structure determination of NorM-NG is described, which had been hampered by low resolution (∼ 4 Å), data anisotropy and pseudo-merohedral twinning. The crystal structure was solved using molecular replacement and was corroborated by conducting a difference Fourier analysis. The NorM-NG structure displays an extracellular-facing conformation, similar to that of NorM-NG bound to a crystallization chaperone. The approaches taken to determine the NorM-NG structure and the lessons learned from this study are discussed, which may be useful for analyzing X-ray diffraction data with similar shortcomings.


Subject(s)
Antiporters/chemistry , Bacterial Proteins/chemistry , Neisseria gonorrhoeae/chemistry , Anisotropy , Antiporters/metabolism , Bacterial Proteins/metabolism , Binding Sites , Crystallization , Crystallography, X-Ray , Fourier Analysis , Ligands , Models, Molecular , Neisseria gonorrhoeae/metabolism , Protein Conformation
SELECTION OF CITATIONS
SEARCH DETAIL