Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Lancet Microbe ; 3(12): e931-e943, 2022 12.
Article in English | MEDLINE | ID: mdl-36462524

ABSTRACT

BACKGROUND: Pharyngeal colonisation by the commensal bacterium Neisseria lactamica inhibits colonisation by Neisseria meningitidis and has an inverse epidemiological association with meningococcal disease. The mechanisms that underpin this relationship are unclear, but could involve the induction of cross-reactive immunity. In this study, we aimed to evaluate whether colonisation with N lactamica induces N lactamica-specific B-cell responses that are cross-reactive with N meningitidis. METHODS: In this randomised, placebo-controlled, human infection trial at University Hospital Southampton Clinical Research Facility (Southampton, UK), healthy adults aged 18-45 years were randomly assigned (2:1) to receive intranasal inoculation with either 105 colony-forming units of N lactamica in 1 mL phosphate-buffered saline (PBS) or 1 mL PBS alone. Participants and researchers conducting participant sampling and immunological assays were masked to allocation. The primary endpoint was the frequency of circulating N lactamica-specific plasma cells and memory B cells after N lactamica inoculation (day 7-28) compared with baseline values (day 0), measured using enzyme-linked immunospot assays. The secondary endpoint was to measure the frequency of N meningitidis-specific B cells. In a second study, we measured the effect of duration of N lactamica colonisation on seroconversion by terminating carriage at either 4 days or 14 days with single-dose oral ciprofloxacin. The studies are now closed to participants. The trials are registered with ClinicalTrials.gov, NCT03633474 and NCT03549325. FINDINGS: Of 50 participants assessed for eligibility between Sept 5, 2018, and March 3, 2019, 31 were randomly assigned (n=20 N lactamica, n=11 PBS). Among the 17 participants who were colonised with N lactamica, the median baselines compared with peak post-colonisation N lactamica-specific plasma-cell frequencies (per 105 peripheral blood mononuclear cells) were 0·0 (IQR 0·0-0·0) versus 5·0 (1·5-10·5) for IgA-secreting plasma cells (p<0·0001), and 0·0 (0·0-0·0) versus 3·0 (1·5-9·5) for IgG-secreting plasma cells (p<0·0001). Median N lactamica-specific IgG memory-B-cell frequencies (percentage of total IgG memory B cells) increased from 0·0024% (0·0000-0·0097) at baseline to 0·0384% (0·0275-0·0649) at day 28 (p<0·0001). The frequency of N meningitidis-specific IgA-secreting and IgG-secreting plasma cells and memory B cells also increased signficantly in participants who were colonised with N lactamica. Upper respiratory tract symptoms were reported in ten (50%) of 20 participants who were inoculated with N lactamica and six (55%) of 11 participants who were inoculated with PBS (p>0·99). Three additional adverse events (two in the N lactamica group and one in the PBS group) and no serious adverse events were reported. In the second study, anti-N lactamica and anti-N meningitidis serum IgG titres increased only in participants who were colonised with N lactamica for 14 days. INTERPRETATION: Natural immunity to N meningitidis after colonisation with N lactamica might be due to cross-reactive adaptive responses. Exploitation of this microbial mechanism with a genetically modified live vector could protect against N meningitidis colonisation and disease. FUNDING: Wellcome Trust, Medical Research Council, and NIHR Southampton Biomedical Research Centre.


Subject(s)
Neisseria lactamica , Neisseria meningitidis , Adult , Humans , Leukocytes, Mononuclear , Immunoglobulin A, Secretory , Phosphates , Saline Solution , Immunoglobulin G
2.
BMJ Open ; 12(5): e056081, 2022 05 18.
Article in English | MEDLINE | ID: mdl-35584870

ABSTRACT

INTRODUCTION: Infant upper respiratory microbiota are derived partly from the maternal respiratory tract, and certain microbiota are associated with altered risk of infections and respiratory disease. Neisseria lactamica is a common pharyngeal commensal in young children and is associated with reduced carriage and invasive disease by Neisseria meningitidis. Nasal inoculation with N. lactamica safely and reproducibly reduces N. meningitidis colonisation in healthy adults. We propose nasal inoculation of pregnant women with N. lactamica, to establish if neonatal pharyngeal colonisation occurs after birth, and to characterise microbiome evolution in mother-infant pairs over 1 month post partum. METHODS AND ANALYSIS: 20 healthy pregnant women will receive nasal inoculation with N. lactamica (wild type strain Y92-1009) at 36-38 weeks gestation. Upper respiratory samples, as well as optional breastmilk, umbilical cord blood and infant venous blood samples, will be collected from mother-infant pairs over 1 month post partum. We will assess safety, N. lactamica colonisation (by targeted PCR) and longitudinal microevolution (by whole genome sequencing), and microbiome evolution (by 16S rRNA gene sequencing). ETHICS AND DISSEMINATION: This study has been approved by the London Central Research Ethics Committee (21/PR/0373). Findings will be published in peer-reviewed open-access journals as soon as possible. TRIAL REGISTRATION NUMBER: NCT04784845.


Subject(s)
Microbiota , Neisseria lactamica , Neisseria meningitidis , Adult , Child , Child, Preschool , Female , Humans , Infant , Infant, Newborn , Microbiota/genetics , Mothers , Neisseria lactamica/genetics , Pharynx , Pilot Projects , Pregnancy , RNA, Ribosomal, 16S
3.
Methods Mol Biol ; 2414: 387-404, 2022.
Article in English | MEDLINE | ID: mdl-34784048

ABSTRACT

Neisseria lactamica is a nonpathogenic commensal of the human upper respiratory tract that has been associated with protection against N. meningitidis colonization and disease. We have previously utilized the N. lactamica controlled human infection model to investigate the protective effect of N. lactamica colonization on N. meningitidis colonization, the nature of cross-reactive immune responses mounted toward N. meningitidis following N. lactamica colonization, and the microevolution of N. lactamica over a 5-month colonization period. More recently, we have assessed the possibility of utilizing genetically modified strains of N. lactamica to enable use of the commensal as a vehicle for prolonged exposure of the nasopharynx of humans to antigens of interest, expressed in carried organisms. A controlled infection with N. lactamica expressing the meningococcal antigen NadA has been executed and the results demonstrate that this strategy is effective at generating immune responses to the target antigen. Throughout this chapter, we outline in a step-by-step manner the methodologies utilized when performing controlled human infection with N. lactamica including procedures relating to: (1) the dilution of N. lactamica stock vials to derive intranasal inocula, (2) the delivery of intranasal inocula to human volunteers, (3) the determination of N. lactamica colonization status following intranasal inoculation using oropharyngeal swabbing and nasal wash sampling, (4) the microbiological procedures utilized to identify N. lactamica colonization among study volunteers, and (5) the identification of N. lactamica colonies as strain Y92-1009 using polymerase chain reaction.


Subject(s)
Neisseria lactamica , Antigens , Cross Reactions , Humans , Nasopharynx , Neisseria meningitidis , Neisseriaceae Infections
4.
Sci Transl Med ; 13(601)2021 07 07.
Article in English | MEDLINE | ID: mdl-34233953

ABSTRACT

The human nasopharynx contains a stable microbial ecosystem of commensal and potentially pathogenic bacteria, which can elicit protective primary and secondary immune responses. Experimental intranasal infection of human adults with the commensal Neisseria lactamica produced safe, sustained pharyngeal colonization. This has potential utility as a vehicle for sustained release of antigen to the human mucosa, but commensals in general are thought to be immunologically tolerated. Here, we show that engineered N. lactamica, chromosomally transformed to express a heterologous vaccine antigen, safely induces systemic, antigen-specific immune responses during carriage in humans. When the N. lactamica expressing the meningococcal antigen Neisseria Adhesin A (NadA) was inoculated intranasally into human volunteers, all colonized participants carried the bacteria asymptomatically for at least 28 days, with most (86%) still carrying the bacteria at 90 days. Compared to an otherwise isogenic but phenotypically wild-type strain, colonization with NadA-expressing N. lactamica generated NadA-specific immunoglobulin G (IgG)- and IgA-secreting plasma cells within 14 days of colonization and NadA-specific IgG memory B cells within 28 days of colonization. NadA-specific IgG memory B cells were detected in peripheral blood of colonized participants for at least 90 days. Over the same period, there was seroconversion against NadA and generation of serum bactericidal antibody activity against a NadA-expressing meningococcus. The controlled infection was safe, and there was no transmission to adult bedroom sharers during the 90-day period. Genetically modified N. lactamica could therefore be used to generate beneficial immune responses to heterologous antigens during sustained pharyngeal carriage.


Subject(s)
Meningococcal Vaccines , Neisseria lactamica , Adult , Antibodies, Bacterial , Antigens, Heterophile , Ecosystem , Humans , Immunologic Memory
5.
J Infect ; 82(6): 247-252, 2021 06.
Article in English | MEDLINE | ID: mdl-33753151

ABSTRACT

BACKGROUND: The association between infant respiratory microbiota and disease (including respiratory tract infections and asthma) is increasingly recognised, although the mechanism remains unclear. Respiratory infections and asthma account for a large proportion of infant morbidity and mortality, so the possibility of preventing disease or modifying clinical outcomes by manipulating microbiome development warrants investigation. OBJECTIVES AND METHODS: We identified studies that investigated the efficacy of live bacteria (probiotics or human challenge) or their substrates to modify respiratory colonisation or clinical outcomes in infants. ELIGIBILITY CRITERIA: Interventional studies involving infants under one year of age, administration of live bacteria or their substrates, and outcome measures including bacterial colonisation, microbiome profile, or respiratory disease phenotypes. RESULTS AND LIMITATIONS: Some bacterial interventions can reduce infant respiratory infections, although none have been shown to reduce asthma incidence. The literature is heterogeneous in design and quality, precluding meaningful meta-analysis. CONCLUSIONS: Upper respiratory tract infant microbiome manipulation may alter outcomes in respiratory tract infection, but further well-conducted research is needed to confirm this. Improved regulation of proprietary bacterial products is essential for further progress.


Subject(s)
Asthma , Microbiota , Probiotics , Respiratory Tract Infections , Asthma/epidemiology , Bacteria , Humans , Infant , Respiratory Tract Infections/epidemiology , Respiratory Tract Infections/prevention & control
7.
J Infect ; 77(6): 534-543, 2018 12.
Article in English | MEDLINE | ID: mdl-30391630

ABSTRACT

OBJECTIVES: Escherichia coli is the leading cause of bacteraemia. In an era of emerging multi-drug-resistant strains, development of effective preventative strategies will be informed by knowledge of strain diversity associated with specific infective syndromes/patient groups. We hypothesised that the number of virulence factor (VF) genes amongst bacteraemia isolates from neutropaenic patients would be lower than isolates from immunocompetent patients. METHODS: Immunocompetent and neutropaenic adults with E. coli bacteraemia were recruited prospectively and the source of bacteraemia determined. VF gene profiles were established in silico following whole genome sequencing. RESULTS: Isolates from individual patients were monoclonal. Strains from immunocompetent patients with urinary tract infective foci (UTIF) harboured more VF genes (median number of VF genes 16, range 8-24) than isolates from both immunocompetent patients with non-UTIF (10, 2-22, p = 0.0058) and neutropaenic patients with unknown focus of infection (NPUFI) (8, 3-13, p < 0.0001). Number of VF genes (OR 1.21, 95% CIs 1.01-1.46, p = 0.039) and urinary catheter/recurrent urinary tract infection (OR 12.82, 95% CIs 1.24-132.65, p = 0.032) were independent predictors of bacteraemia secondary to UTIF vs. non-UTIF in immunocompetent patients. papA, papC, papE/F, papG, agn43, tia, iut, fyuA, kpsM and sat were significantly more prevalent amongst UTIF- vs non-UTIF-originating isolates amongst immunocompetent patients, while papC, papE/F, papG, agn43, tia, fyuA, hlyA, usp and clb were significantly more prevalent amongst UTIF- vs NPUFI-associated isolates. CONCLUSIONS: Bacteraemia-associated E. coli strains originating from UTIF have distinct VF gene profiles from strains associated with non-UTIF- and NPUFI. This diversity must be addressed in the design of future vaccines to ensure adequate coverage of strains responsible for site-specific disease.


Subject(s)
Escherichia coli Infections/urine , Escherichia coli/genetics , Genome, Bacterial , Urinary Tract Infections/microbiology , Virulence Factors/genetics , Adult , Aged , Aged, 80 and over , Bacteremia/microbiology , Escherichia coli/isolation & purification , Escherichia coli/pathogenicity , Escherichia coli Infections/blood , Female , Humans , Immunocompetence , Male , Middle Aged , Neutropenia/microbiology , Phylogeny , Prospective Studies , Sequence Analysis, DNA , United Kingdom , Virulence , Whole Genome Sequencing , Young Adult
8.
Nat Commun ; 9(1): 4753, 2018 11 12.
Article in English | MEDLINE | ID: mdl-30420631

ABSTRACT

Neisseria lactamica is a harmless coloniser of the infant respiratory tract, and has a mutually-excluding relationship with the pathogen Neisseria meningitidis. Here we report controlled human infection with genomically-defined N. lactamica and subsequent bacterial microevolution during 26 weeks of colonisation. We find that most mutations that occur during nasopharyngeal carriage are transient indels within repetitive tracts of putative phase-variable loci associated with host-microbe interactions (pgl and lgt) and iron acquisition (fetA promotor and hpuA). Recurrent polymorphisms occurred in genes associated with energy metabolism (nuoN, rssA) and the CRISPR-associated cas1. A gene encoding a large hypothetical protein was often mutated in 27% of the subjects. In volunteers who were naturally co-colonised with meningococci, recombination altered allelic identity in N. lactamica to resemble meningococcal alleles, including loci associated with metabolism, outer membrane proteins and immune response activators. Our results suggest that phase variable genes are often mutated during carriage-associated microevolution.


Subject(s)
Nasopharynx/microbiology , Neisseria lactamica/growth & development , Neisseriaceae Infections/microbiology , Carrier State , Colony Count, Microbial , Genes, Bacterial , Humans , Mutation/genetics , Mutation Rate , Neisseria lactamica/genetics , Neisseria lactamica/isolation & purification , Recombination, Genetic/genetics
9.
Stand Genomic Sci ; 13: 4, 2018.
Article in English | MEDLINE | ID: mdl-29467915

ABSTRACT

[This corrects the article DOI: 10.1186/s40793-017-0250-6.].

10.
Stand Genomic Sci ; 12: 41, 2017.
Article in English | MEDLINE | ID: mdl-28770026

ABSTRACT

We present the high quality, complete genome assembly of Neisseria lactamica Y92-1009 used to manufacture an outer membrane vesicle (OMV)-based vaccine, and a member of the Neisseria genus. The strain is available on request from the Public Health England Meningococcal Reference Unit. This Gram negative, dipplococcoid bacterium is an organism of worldwide clinical interest because human nasopharyngeal carriage is related inversely to the incidence of meningococcal disease, caused by Neisseria meningitidis. The organism sequenced was isolated during a school carriage survey in Northern Ireland in 1992 and has been the subject of a variety of laboratory and clinical studies. Four SMRT cells on a RSII machine by Pacific Biosystems were used to produce a complete, closed genome assembly. Sequence data were obtained for a total of 30,180,391 bases from 2621 reads and assembled using the HGAP algorithm. The assembly was corrected using short reads obtained from an Illumina HiSeq 2000instrument. This resulted in a 2,146,723 bp assembly with approximately 460 fold mean coverage depth and a GC ratio of 52.3%.

11.
Microbiology (Reading) ; 163(7): 1093-1104, 2017 07.
Article in English | MEDLINE | ID: mdl-28699879

ABSTRACT

Non-typeable Haemophilus influenzae (NTHi) is an opportunistic pathogen that plays a major role in a number of respiratory tract infections, including otitis media, cystic fibrosis and chronic obstructive pulmonary disease. Biofilm formation has been implicated in both NTHi colonization and disease, and is responsible for the increased tolerance of this pathogen towards antibiotic treatment. Targeting metabolic pathways that are important in NTHi biofilm formation represents a potential strategy to combat this antibiotic recalcitrance. A previous investigation demonstrated increased expression of a putative d-methionine uptake protein following exposure of NTHi biofilms to the ubiquitous signalling molecule, nitric oxide. We therefore hypothesized that treatment with exogenous d-methionine would impact on NTHi biofilm formation and increase antibiotic sensitivity. Treatment of NTHi during the process of biofilm formation resulted in a reduction in biofilm viability, increased biomass, changes in the overall biofilm architecture and the adoption of an amorphous cellular morphology. Quantitative proteomic analyses identified 124 proteins that were differentially expressed following d-methionine treatment, of which 51 (41 %) were involved in metabolic and transport processes. Nine proteins involved in peptidoglycan synthesis and cell division showed significantly increased expression. Furthermore, d-methionine treatment augmented the efficacy of azithromycin treatment and highlighted the potential of d-methionine as an adjunctive therapeutic approach for NTHi biofilm-associated infections.


Subject(s)
Biofilms , Haemophilus Infections/microbiology , Haemophilus influenzae/growth & development , Haemophilus influenzae/metabolism , Methionine/metabolism , Peptidoglycan/biosynthesis , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Gene Expression Regulation, Bacterial , Haemophilus influenzae/drug effects , Haemophilus influenzae/genetics , Humans
12.
Pathogens ; 5(3)2016 Aug 03.
Article in English | MEDLINE | ID: mdl-27527221

ABSTRACT

The bronchial epithelium provides protection against pathogens from the inhaled environment through the formation of a highly-regulated barrier. In order to understand the pulmonary diseases melioidosis and tularemia caused by Burkholderia thailandensis and Fransicella tularensis, respectively, the barrier function of the human bronchial epithelium were analysed. Polarised 16HBE14o- or differentiated primary human bronchial epithelial cells (BECs) were exposed to increasing multiplicities of infection (MOI) of B. thailandensis or F. tularensis Live Vaccine Strain and barrier responses monitored over 24-72 h. Challenge of polarized BECs with either bacterial species caused an MOI- and time-dependent increase in ionic permeability, disruption of tight junctions, and bacterial passage from the apical to the basolateral compartment. B. thailandensis was found to be more invasive than F. tularensis. Both bacterial species induced an MOI-dependent increase in TNF-α release. An increase in ionic permeability and TNF-α release was induced by B. thailandensis in differentiated BECs. Pretreatment of polarised BECs with the corticosteroid fluticasone propionate reduced bacterial-dependent increases in ionic permeability, bacterial passage, and TNF-α release. TNF blocking antibody Enbrel(®) reduced bacterial passage only. BEC barrier properties are disrupted during respiratory bacterial infections and targeting with corticosteroids or anti-TNF compounds may represent a therapeutic option.

13.
Vaccine ; 33(36): 4486-94, 2015 Aug 26.
Article in English | MEDLINE | ID: mdl-26207592

ABSTRACT

The nmb1612 (NEIS1533) gene encoding the ~27-kDa putative amino acid ATP-binding cassette (ABC) transporter, periplasmic substrate-binding protein from Neisseria meningitidis serogroup B (MenB) strain MC58 was cloned and expressed in Escherichia coli, and the purified recombinant (r)NMB1612 was used for animal immunization studies. Immunization of mice with rNMB1612 adsorbed to Al(OH)3 and in liposomes with and without MPLA, induced antiserum with bactericidal activity in an assay using baby rabbit complement, against the homologous strain MC58 (encoding protein representative of Allele 62) and killed heterologous strains encoding proteins of three other alleles (representative of Alleles 1, 64 and 68), with similar SBA titres. However, strain MC58 was not killed (titre <4) in a human serum bactericidal assay (hSBA) using anti-rNMB1612 sera, although another strain (MC168) expressing the same protein was killed (median titres of 16-64 in the hSBA). Analysis of the NMB1612 amino acid sequences from 4351 meningococcal strains in the pubmlst.org/Neisseria database and a collection of 13 isolates from colonized individuals and from patients, showed that antibodies raised against rNMB1612 could potentially kill at least 72% of the MenB strains in the complete sequence database. For MenB disease occurring specifically in the UK from 2013 to 2015, >91% of the isolates causing disease in this recent period expressed NMB1612 protein encoded by Allele 1 and could be potentially killed by sera raised to the recombinant antigen in the current study. The NMB1612 protein was surface-accessible and expressed by different meningococcal strains. In summary, the properties of (i) NMB1612 protein conservation and expression, (ii) limited amino acid sequence variation between proteins encoded by different alleles, and (iii) the ability of a recombinant protein to induce cross-strain bactericidal antibodies, would all suggest a promising antigen for consideration for inclusion in new meningococcal vaccines.


Subject(s)
ATP-Binding Cassette Transporters/immunology , Antibodies, Bacterial/blood , Bacterial Proteins/immunology , Blood Bactericidal Activity , Immunity, Heterologous , Meningococcal Vaccines/immunology , Neisseria meningitidis, Serogroup B/immunology , ATP-Binding Cassette Transporters/genetics , Adjuvants, Immunologic/administration & dosage , Aluminum Hydroxide/administration & dosage , Animals , Bacterial Proteins/genetics , Cloning, Molecular , Cross Reactions , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Humans , Liposomes/administration & dosage , Meningococcal Vaccines/administration & dosage , Meningococcal Vaccines/genetics , Mice, Inbred BALB C , Neisseria meningitidis, Serogroup B/genetics , Rabbits , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology
14.
Adv Microb Physiol ; 66: 323-55, 2015.
Article in English | MEDLINE | ID: mdl-26210107

ABSTRACT

The exclusive reservoir of the genus Neisseria is the human. Of the broad range of species that comprise the Neisseria, only two are frequently pathogenic, and only one of those is a resident of the nasopharynx. Although Neisseria meningitidis can cause severe disease if it invades the bloodstream, the vast majority of interactions between humans and Neisseria are benign, with the bacteria inhabiting its mucosal niche as a non-invasive commensal. Understandably, with the exception of Neisseria gonorrhoeae, which preferentially colonises the urogenital tract, the neisseriae are extremely well adapted to survival in the human nasopharynx, their sole biological niche. The purpose of this review is to provide an overview of the molecular mechanisms evolved by Neisseria to facilitate colonisation and survival within the nasopharynx, focussing on N. meningitidis. The organism has adapted to survive in aerosolised transmission and to attach to mucosal surfaces. It then has to replicate in a nutrition-poor environment and resist immune and competitive pressure within a polymicrobial complex. Temperature and relative gas concentrations (nitric oxide and oxygen) are likely to be potent initial signals of arrival within the nasopharyngeal environment, and this review will focus on how N. meningitidis responds to these to increase the likelihood of its survival.


Subject(s)
Adaptation, Biological , Nasopharynx/microbiology , Neisseria meningitidis/physiology , Bacteremia , Bacterial Adhesion , Carrier State , Host-Pathogen Interactions , Humans , Meningitis, Meningococcal , Meningococcal Infections/transmission , Neisseria meningitidis/genetics , Neisseria meningitidis/growth & development
15.
Antioxid Redox Signal ; 18(3): 309-22, 2013 Jan 20.
Article in English | MEDLINE | ID: mdl-22768799

ABSTRACT

SIGNIFICANCE: The formation and degradation of S-nitrosothiols (SNOs) are important mechanisms of post-translational protein modification and appear to be ubiquitous in biology. These processes play well-characterized roles in eukaryotic cells, including a variety of pathologies and in relation to chronic conditions. We know little of the roles of these processes in pathogenic and other bacteria. RECENT ADVANCES: It is clear, mostly from growth and transcriptional studies, that bacteria sense and respond to exogenous SNOs. These responses are phenotypically and mechanistically distinct from the responses of bacteria to nitric oxide (NO) and NO-releasing agents, as well as peroxynitrite. Small SNOs, such as S-nitrosoglutathione (GSNO), are accumulated by bacteria with the result that intracellular S-nitrosoproteins (the 'S-nitrosoproteome') are detectable. Recently, conditions for endogenous SNO formation in enterobacteria have been described. CRITICAL ISSUES: The propensity of intracellular proteins to form SNOs is presumably constrained by the same rules of selectivity that have been discovered in eukaryotic systems, but is also influenced by uniquely bacterial NO detoxification systems, exemplified by the flavohemoglobin Hmp in enterobacteria and NO reductase of meningococci. Furthermore, the bacterial expression of such proteins impacts upon the formation of SNOs in mammalian hosts. FUTURE DIRECTIONS: The impairment during bacterial infections of specific SNO events in the mammalian host is of considerable interest in the context of proteins involved in innate immunity and intracellular signalling. In bacteria, numerous mechanisms of S-nitrosothiol degradation have been reported (e.g., GSNO reductase); others are thought to operate, based on consideration of their mammalian counterparts. The nitrosothiols of bacteria and particularly of pathogens warrant more intensive investigation.


Subject(s)
Bacterial Proteins/metabolism , S-Nitrosothiols/metabolism , Animals , Bacterial Infections/metabolism , Bacterial Infections/microbiology , Cysteine/analogs & derivatives , Cysteine/metabolism , Gene Expression Regulation, Bacterial , Gram-Negative Bacteria/metabolism , Gram-Negative Bacteria/physiology , Gram-Positive Bacteria/metabolism , Gram-Positive Bacteria/physiology , Host-Pathogen Interactions , Humans , Nitrosation , Oxidative Stress , Protein Processing, Post-Translational , Reactive Nitrogen Species/metabolism , S-Nitrosoglutathione/metabolism
16.
FASEB J ; 24(1): 286-95, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19720623

ABSTRACT

S-nitrosylation is an important mediator of multiple nitric oxide-dependent biological processes, including eukaryotic cellular events such as macrophage apoptosis and proinflammatory signaling. Many pathogenic bacteria possess NO detoxification mechanisms, such as the nitric oxide reductase (NorB) of Neisseria meningitidis and the flavohemoglobins (Hmp) of Salmonella enterica and Escherichia coli, which serve to protect the microorganism from nitrosative stress within the intracellular environment. In this study, we demonstrate that expression of meningococcal NorB increases the rate at which low-molecular-weight S-nitrosothiol (SNO) decomposes in vitro. To determine whether this effect occurs in cells during infection by bacteria, we induced SNO formation in murine macrophages by activation with lipopolysaccharide and gamma-interferon and observed a reduced abundance of SNO during coincubation with N. meningitidis, S. enterica, or E. coli. In each case, this effect was shown to be dependent on bacterial NO detoxification genes, which act to prevent SNO formation through the removal of NO. This may represent a novel mechanism of host cell injury by bacteria.


Subject(s)
Bacteria/metabolism , Bacteria/pathogenicity , Nitric Oxide/metabolism , S-Nitrosothiols/metabolism , Animals , Cell Line , Escherichia coli K12/metabolism , Host-Pathogen Interactions/physiology , Inactivation, Metabolic , Interferon-gamma/pharmacology , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Macrophages/metabolism , Macrophages/microbiology , Mice , Neisseria meningitidis, Serogroup B/genetics , Neisseria meningitidis, Serogroup B/metabolism , Oxidoreductases/genetics , Oxidoreductases/metabolism , Recombinant Proteins , Salmonella typhimurium/metabolism
17.
Methods Enzymol ; 436: 113-27, 2008.
Article in English | MEDLINE | ID: mdl-18237630

ABSTRACT

Nitric oxide (NO) is a ubiquitous gas with potent biological effects, including vasodilation, neuronal signaling, and antimicrobial activity. NO is a free radical and can readily react with other molecules, in particular, iron centers and oxygen. At physiological concentrations in aqueous solutions, even in the presence of oxygen, NO is reasonably stable. Under these conditions, NO is oxidized almost exclusively to nitrite (NO2-). In cell lysates and tissue extracts with iron-containing proteins, however, NO is postulated to have a very short half-life, with the major oxidation product being nitrate (NO3-). In mammalian cells, NO is generated via the action of the NO synthases (NOS), of which there are three known isotypes. NO can also be generated from the chemical decomposition of S-nitrosothiols, and there is some indication that naturally occurring S-nitrosothiols, such as S-nitrosoalbumin, may be natural reservoirs of NO in vivo. Here we describe a methodology to measure variations in NO in liquid samples using chemiluminescence. The protocols described allow us to distinguish between various products of NO chemistry, thus providing a sensitive method of measurement of NO concentration within a sample. They also allow us to distinguish between the various products that may be generated when NO reacts with molecules in complex biological samples such as cell lysates and supernatants.


Subject(s)
Luminescent Measurements/methods , Nitric Oxide/analysis , Animals , Cell Line , Indicators and Reagents , Luminescent Measurements/standards , Macrophages/chemistry , Mice , Nitrates/analysis , Nitric Oxide/chemistry , Nitrites/analysis , Reactive Nitrogen Species/analysis , Reference Standards , S-Nitrosothiols/analysis
18.
Microbes Infect ; 9(8): 981-7, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17544805

ABSTRACT

Macrophages produce nitric oxide (NO) via the inducible nitric oxide synthase as part of a successful response to infection. The gene norB of Neisseria meningitidis encodes a NO reductase which enables utilization and consumption of NO during microaerobic respiration and confers resistance to nitrosative stress-related killing by human monocyte-derived macrophages (MDM). In this study we confirmed that NO regulates cytokine and chemokine release by resting MDM: accumulation of TNF-alpha, IL-12, IL-10, CCL5 (RANTES) and CXCL8 (IL-8) in MDM supernatants was significantly modified by the NO-donor S-nitroso-N-penicillamine (SNAP). Using a protein array, infection of MDM with N. meningitidis was shown to be associated with secretion of a wide range of cytokines and chemokines. To test whether NO metabolism by N. meningitidis modifies release of NO-regulated cytokines, we infected MDM with wild-type organisms and an isogenic norB strain. Resulting expression of the cytokines TNF-alpha and IL-12, and the chemokine CXCL8 was increased and production of the cytokine IL-10 and the chemokine CCL5 was decreased in norB-infected MDM, in comparison to wild-type. Addition of SNAP to cultures infected with wild-type mimicked the effect observed in cultures infected with the norB mutant. In conclusion, NorB-catalysed removal of NO modifies cellular release of NO-regulated cytokines and chemokines.


Subject(s)
Cytokines/metabolism , Gene Expression Regulation , Macrophages/microbiology , Monocytes/microbiology , Neisseria meningitidis/pathogenicity , Nitric Oxide/metabolism , Chemokines/genetics , Chemokines/metabolism , Cytokines/genetics , Humans , Macrophages/metabolism , Monocytes/metabolism , Neisseria meningitidis/enzymology , Neisseria meningitidis/genetics , Neisseria meningitidis/metabolism , Nitric Oxide/pharmacology , Nitric Oxide Donors/pharmacology , Nitrite Reductases/genetics , Nitrite Reductases/metabolism , Penicillamine/analogs & derivatives , Penicillamine/pharmacology , Phagocytosis
SELECTION OF CITATIONS
SEARCH DETAIL
...