Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 1.938
Filtrer
1.
Curr Protoc ; 4(9): e70000, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39228292

RÉSUMÉ

The sexually transmitted pathogen, Neisseria gonorrhoeae, undergoes natural transformation at high frequency. This property has led to the rapid dissemination of antibiotic resistance markers and the panmictic structure of the gonococcal population. However, high-frequency transformation also makes N. gonorrhoeae one of the easiest bacterial species to manipulate genetically in the laboratory. Techniques have been developed that result in transformation frequencies >50%, allowing the identification of mutants by screening and without selection. Constructs have been created to take advantage of this high-frequency transformation, facilitating genetic mutation, complementation, and heterologous gene expression. Similar methods have been developed for N. meningitidis and nonpathogenic Neisseria including N. mucosa and N. musculi. Techniques are described for genetic manipulation of N. gonorrhoeae and commensal Neisseria species, as well as for growth of these fastidious organisms. © 2024 The Author(s). Current Protocols published by Wiley Periodicals LLC. Basic Protocol 1: Spot transformation of Neisseria gonorrhoeae on agar plates Basic Protocol 2: Spot transformation of commensal Neisseria on agar plates Basic Protocol 3: Transformation of Neisseria gonorrhoeae in liquid culture Basic Protocol 4: Electroporation of Neisseria gonorrhoeae Basic Protocol 5: Creation of unmarked mutations using a positive and negative selection cassette Basic Protocol 6: In vitro mutagenesis of Neisseria gonorrhoeae chromosomal DNA using EZ-Tn5 Basic Protocol 7: Chemical mutagenesis Basic Protocol 8: Complementation on the Neisseria gonorrhoeae chromosome Alternate Protocol 1: Complementation with replicating plasmids Alternate Protocol 2: Complementation on the Neisseria musculi or Neisseria mucosa chromosome Basic Protocol 9: Preparation of chromosomal DNA from Neisseria gonorrhoeae grown on solid medium Alternate Protocol 3: Preparation of chromosomal DNA from Neisseria gonorrhoeae grown in broth Support Protocol: Preparing PCR templates from Neisseria gonorrhoeae colonies.


Sujet(s)
Neisseria gonorrhoeae , Neisseria , Transformation bactérienne , Neisseria gonorrhoeae/génétique , Neisseria gonorrhoeae/effets des médicaments et des substances chimiques , Neisseria/génétique , Neisseria/effets des médicaments et des substances chimiques , Électroporation , Gonorrhée/microbiologie , Gonorrhée/traitement médicamenteux , Humains
2.
J Med Microbiol ; 73(8)2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39212029

RÉSUMÉ

Introduction. Commensal Neisseria spp. are highly prevalent in the oropharynx as part of the healthy microbiome. N. meningitidis can colonise the oropharynx too from where it can cause invasive meningococcal disease. To identify N. meningitidis, clinical microbiology laboratories often rely on Matrix Assisted Laser Desorption/Ionisation Time of Flight Mass Spectrometry (MALDI-TOF MS).Hypothesis/Gap statement. N. meningitidis may be misidentified by MALDI-TOF MS.Aim. To conduct genomic surveillance of oropharyngeal Neisseria spp. in order to: (i) verify MALDI-TOF MS species identification, and (ii) characterize commensal Neisseria spp. genomes.Methodology. We analysed whole genome sequence (WGS) data from 119 Neisseria spp. isolates from a surveillance programme for oropharyngeal Neisseria spp. in Belgium. Different species identification methods were compared: (i) MALDI-TOF MS, (ii) Ribosomal Multilocus Sequence Typing (rMLST) and (iii) rplF gene species identification. WGS data were used to further characterize Neisseria species found with supplementary analyses of Neisseria cinerea genomes.Results. Based on genomic species identification, isolates from the oropharyngeal Neisseria surveilence study were composed of the following species: N. meningitidis (n=23), N. subflava (n=61), N. mucosa (n=15), N. oralis (n=8), N. cinerea (n=5), N. elongata (n=3), N. lactamica (n=2), N. bacilliformis (n=1) and N. polysaccharea (n=1). Of these 119 isolates, four isolates identified as N. meningitidis (n=3) and N. subflava (n=1) by MALDI-TOF MS, were determined to be N. polysaccharea (n=1), N. cinerea (n=2) and N. mucosa (n=1) by rMLST. Phylogenetic analyses revealed that N. cinerea isolates from the general population (n=3, cluster one) were distinct from those obtained from men who have sex with men (MSM, n=2, cluster two). The latter contained genomes misidentified as N. meningitidis using MALDI-TOF MS. These two N. cinerea clusters persisted after the inclusion of published N. cinerea WGS (n=42). Both N. cinerea clusters were further defined through pangenome and Average Nucleotide Identity (ANI) analyses.Conclusion. This study provides insights into the importance of genomic genus-wide Neisseria surveillance studies to improve the characterization and identification of the Neisseria genus.


Sujet(s)
Génome bactérien , Typage par séquençage multilocus , Partie orale du pharynx , Spectrométrie de masse MALDI , Séquençage du génome entier , Spectrométrie de masse MALDI/méthodes , Partie orale du pharynx/microbiologie , Humains , Typage par séquençage multilocus/méthodes , Neisseria cinerea/génétique , Phylogenèse , Neisseria/classification , Neisseria/génétique , Neisseria/isolement et purification , Belgique , Neisseria meningitidis/génétique , Neisseria meningitidis/classification , Neisseria meningitidis/isolement et purification , Infections à Neisseriaceae/microbiologie , Infections à Neisseriaceae/diagnostic
3.
FEMS Microbiol Lett ; 3712024 Jan 09.
Article de Anglais | MEDLINE | ID: mdl-39210455

RÉSUMÉ

BACKGROUND: commensal Neisseria species are part of the oropharyngeal microbiome and play an important role in nitrate reduction and protecting against colonization by pathogenic bacteria. They do, however, also serve as a reservoir of antimicrobial resistance. Little is known about the prevalence of these species in the general population, how this varies by age and how antimicrobial susceptibility varies between species. METHODS: we assessed the prevalence and antimicrobial susceptibility of commensal Neisseria species in the parents (n = 38) and children (n = 50) of 35 families in Belgium. RESULTS: various commensal Neisseria (n = 5) could be isolated from the participants. Most abundant were N. subflava and N. mucosa. Neisseria subflava was detected in 77 of 88 (87.5%) individuals and N. mucosa in 64 of 88 (72.7%). Neisseria mucosa was more prevalent in children [41/50 (82%)] than parents [23/38 (60.5%); P < .05], while N. bacilliformis was more prevalent in parents [7/36 (19.4%)] than children [2/50 (4%); P < .05]. Neisseria bacilliformis had high ceftriaxone minimum inhibitory concentrations (MICs; median MIC 0.5 mg/l; IQR 0.38-0.75). The ceftriaxone MICs of all Neisseria isolates were higher in the parents than in the children. This could be explained by a higher prevalence of N. bacilliformis in the parents. INTERPRETATION: the N. bacilliformis isolates had uniformly high ceftriaxone MICs which warrant further investigation.


Sujet(s)
Antibactériens , Tests de sensibilité microbienne , Neisseria , Parents , Humains , Belgique/épidémiologie , Neisseria/effets des médicaments et des substances chimiques , Neisseria/isolement et purification , Neisseria/génétique , Études transversales , Enfant , Antibactériens/pharmacologie , Femelle , Enfant d'âge préscolaire , Mâle , Adulte , Adulte d'âge moyen , Adolescent , Résistance bactérienne aux médicaments , Nourrisson , Partie orale du pharynx/microbiologie , Prévalence , Jeune adulte
4.
Nucleic Acids Res ; 52(17): 10563-10574, 2024 Sep 23.
Article de Anglais | MEDLINE | ID: mdl-39180405

RÉSUMÉ

CRISPR-Cas systems function as adaptive immune mechanisms in bacteria and archaea and offer protection against phages and other mobile genetic elements. Among many types of CRISPR-Cas systems, Type I CRISPR-Cas systems are most abundant, with target interference depending on a multi-subunit, RNA-guided complex known as Cascade that recruits a transacting helicase nuclease, Cas3, to degrade the target. While structural studies on several other types of Cas3 have been conducted long ago, it was only recently that the structural study of Type I-C Cas3 in complex with Cascade was revealed, shedding light on how Cas3 achieve its activity in the Cascade complex. In the present study, we elucidated the first structure of standalone Type I-C Cas3 from Neisseria lactamica (NlaCas3). Structural analysis revealed that the histidine-aspartate (HD) nuclease active site of NlaCas3 was bound to two Fe2+ ions that inhibited its activity. Moreover, NlaCas3 could cleave both single-stranded and double-stranded DNA in the presence of Ni2+ or Co2+, showing the highest activity in the presence of both Ni2+ and Mg2+ ions. By comparing the structural studies of various Cas3 proteins, we determined that our NlaCas3 stays in an inactive conformation, allowing us to understand the structural changes associated with its activation and their implication.


Sujet(s)
Protéines associées aux CRISPR , Systèmes CRISPR-Cas , Protéines associées aux CRISPR/métabolisme , Protéines associées aux CRISPR/composition chimique , Protéines associées aux CRISPR/génétique , Domaine catalytique , Modèles moléculaires , Neisseria/génétique , Neisseria/enzymologie , Protéines bactériennes/métabolisme , Protéines bactériennes/composition chimique , Protéines bactériennes/génétique , Cristallographie aux rayons X , ADN/métabolisme , ADN/composition chimique , ADN/génétique , Liaison aux protéines
5.
Biol Reprod ; 111(3): 613-624, 2024 Sep 14.
Article de Anglais | MEDLINE | ID: mdl-38972067

RÉSUMÉ

With ~78 million cases yearly, the sexually transmitted bacterium Neisseria gonorrhoeae is an urgent threat to global public health due to continued emergence of antimicrobial resistance. In the male reproductive tract, untreated infections may cause permanent damage, poor sperm quality, and subsequently subfertility. Currently, few animal models exist for N. gonorrhoeae infection, which has strict human tropism, and available models have limited translatability to human disease. The absence of appropriate models inhibits the development of vital new diagnostics and treatments. However, the discovery of Neisseria musculi, a mouse oral cavity bacterium, offers much promise. This bacterium has already been used to develop an oral Neisseria infection model, but the feasibility of establishing urogenital gonococcal models is unexplored. We inoculated mice via the intrapenile route with N. musculi. We assessed bacterial burden throughout the male reproductive tract, the systemic and tissue-specific immune response 2-weeks postinfection, and the effect of infection on sperm health. Neisseria musculi was found in penis (2/5) and vas deferens (3/5) tissues. Infection altered immune cell counts: CD19+ (spleen, lymph node, penis), F4/80+ (spleen, lymph node, epididymus), and Gr1+ (penis) compared with noninfected mice. This culminated in sperm from infected mice having poor viability, motility, and morphology. We hypothesize that in the absence of testis infection, infection and inflammation in other reproductive is sufficient to damage sperm quality. Many results herein are consistent with outcomes of gonorrhoea infection, indicating the potential of this model as a tool for enhancing the understanding of Neisseria infections of the human male reproductive tract.


Sujet(s)
Modèles animaux de maladie humaine , Neisseria , Mâle , Animaux , Souris , Neisseria/isolement et purification , Gonorrhée/microbiologie , Souris de lignée C57BL , Système génital de l'homme/microbiologie , Infections à Neisseriaceae/microbiologie
6.
Article de Anglais | MEDLINE | ID: mdl-39023135

RÉSUMÉ

A taxogenomic study of three strains (3986T, 51.81, and JF 2415) isolated from rabbits between 1972 and 2000 led to the description of a new Neisseria species. The highest sequence similarity of the 16S rRNA gene was found to Neisseria animalis NCTC 10212T (96.7 %). The 16S rRNA gene similarity above 99 % and average nucleotide identity (ANI) values above 96 % among the strains, indicated that they belong to the same species. At the same time, the strains shared ANI values below 81 % and dDDH values below 24 % with all described Neisseria species. In the bac120 gene phylogenetic tree, the three strains clustered near Neisseria elongata and Neisseria bacilliformis in the Neisseria clade. However, the Neisseria clade is not monophyletic, and includes the type strains of Morococcus cerebrosus, Bergeriella denitrificans, Kingella potus, Uruburuella suis, and Uruburuella testudinis. Neisseria shayeganii clustered outside the clade with members of the genus Eikenella. Amino acid identity (AAI) values were calculated, and a threshold of 71 % was used to circumscribe the genus Neisseria. According to this proposed AAI threshold, strains 3986T, 51.81, and JF 2415 were placed within the genus Neisseria. The cells of the three strains were Gram-stain-negative diplococcobacilli and non-motile. Optimal growth on trypticase soy agar occurred at 37 °C and pH 8.5 in aerobic conditions. Notably, all strains exhibited indole production in the API-NH test, which is atypical for Neisseria and the family Neisseriaceae. The strains exhibited a common set of 68 peaks in their MALDI-TOF MS profiles, facilitating the swift and accurate identification of this species. Based on genotypic and phenotypic data, it is proposed that strains 3986T, 51.81, and JF 2415 represent a novel species within the genus Neisseria, for which the name Neisseria leonii sp. nov. is proposed (type strain 3986T=R726T=CIP 109994T=LMG 32907T).


Sujet(s)
Techniques de typage bactérien , ADN bactérien , Foie , Poumon , Neisseria , Phylogenèse , ARN ribosomique 16S , Analyse de séquence d'ADN , Animaux , Lapins , ARN ribosomique 16S/génétique , Neisseria/isolement et purification , Neisseria/classification , Neisseria/génétique , ADN bactérien/génétique , Foie/microbiologie , Poumon/microbiologie , Acides gras/analyse , Composition en bases nucléiques
7.
Front Cell Infect Microbiol ; 14: 1356907, 2024.
Article de Anglais | MEDLINE | ID: mdl-38863832

RÉSUMÉ

Introduction: Microbial community composition is closely associated with host disease onset and progression, underscoring the importance of understanding host-microbiota dynamics in various health contexts. Methods: In this study, we utilized full-length 16S rRNA gene sequencing to conduct species-level identification of the microorganisms in the oral cavity of a giant panda (Ailuropoda melanoleuca) with oral malignant fibroma. Results: We observed a significant difference between the microbial community of the tumor side and non-tumor side of the oral cavity of the giant panda, with the latter exhibiting higher microbial diversity. The tumor side was dominated by specific microorganisms, such as Fusobacterium simiae, Porphyromonas sp. feline oral taxon 110, Campylobacter sp. feline oral taxon 100, and Neisseria sp. feline oral taxon 078, that have been reported to be associated with tumorigenic processes and periodontal diseases in other organisms. According to the linear discriminant analysis effect size analysis, more than 9 distinct biomarkers were obtained between the tumor side and non-tumor side samples. Furthermore, the Kyoto Encyclopedia of Genes and Genomes analysis revealed that the oral microbiota of the giant panda was significantly associated with genetic information processing and metabolism, particularly cofactor and vitamin, amino acid, and carbohydrate metabolism. Furthermore, a significant bacterial invasion of epithelial cells was predicted in the tumor side. Discussion: This study provides crucial insights into the association between oral microbiota and oral tumors in giant pandas and offers potential biomarkers that may guide future health assessments and preventive strategies for captive and aging giant pandas.


Sujet(s)
Campylobacter , Fusobacterium , Microbiote , Bouche , Porphyromonas , ARN ribosomique 16S , Ursidae , Ursidae/microbiologie , Animaux , ARN ribosomique 16S/génétique , Porphyromonas/génétique , Porphyromonas/isolement et purification , Porphyromonas/classification , Campylobacter/génétique , Campylobacter/isolement et purification , Campylobacter/classification , Bouche/microbiologie , Fusobacterium/génétique , Fusobacterium/isolement et purification , Fibrome/microbiologie , Fibrome/médecine vétérinaire , Neisseria/isolement et purification , Neisseria/génétique , Neisseria/classification , Tumeurs de la bouche/microbiologie , Tumeurs de la bouche/médecine vétérinaire , Tumeurs de la bouche/anatomopathologie , Phylogenèse , Analyse de séquence d'ADN
8.
J Oral Biosci ; 66(2): 300-303, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38703995

RÉSUMÉ

BACKGROUND: The oral microbiota has recently attracted attention owing to its association with oral and systemic diseases. Accordingly, gaining an understanding of oral microbiota development and the factors influencing it can contribute to preventing the establishment of dysbiotic oral microbiota and, eventually, oral microbiota-related diseases. HIGHLIGHT: In this review, we highlight the results of a longitudinal project focusing on oral microbiota development during early life. At 4 months of age, the oral microbiota of infants was found to differ considerably from the maternal oral microbiota, even though infants acquire oral bacteria from their mothers. At 18 months, although the infant microbiota is still not completely comparable with that of adults, from 4 to 18 months, there is a rapid phase of development, during which the microbial composition undergoes considerable change to a profile more similar to that in adults. During this development, the infant oral microbiota converges into two different profiles with adult-like traits, namely, Streptococcus salivarius- and Neisseria-dominant profiles. This divergence is strongly influenced by dietary habits, with a frequent intake of sweetened beverages being associated with an S. salivarius-dominant profile, which is suspected to be implicated in oral and systemic diseases. CONCLUSION: The foundation of the adult oral microbiota may be established by 18 months of age, and the developmental period from 4 to 18 months may be an appropriate period during which to modify the microbial balance to obtain a desirable healthy state. In particular, dietary habits during this period warrant close attention.


Sujet(s)
Microbiote , Bouche , Humains , Bouche/microbiologie , Nourrisson , Microbiote/physiologie , Streptococcus salivarius , Neisseria/isolement et purification
9.
Infect Immun ; 92(7): e0004824, 2024 Jul 11.
Article de Anglais | MEDLINE | ID: mdl-38814083

RÉSUMÉ

Commensal bacteria are crucial in maintaining host physiological homeostasis, immune system development, and protection against pathogens. Despite their significance, the factors influencing persistent bacterial colonization and their impact on the host still need to be fully understood. Animal models have served as valuable tools to investigate these interactions, but most have limitations. The bacterial genus Neisseria, which includes both commensal and pathogenic species, has been studied from a pathogenicity to humans perspective but lacks models that study immune responses in the context of long-term persistence. Neisseria musculi, a recently described natural commensal of mice, offers a unique opportunity to study long-term host-commensal interactions. In this study, for the first time, we have used this model to study the transcriptional, phenotypic, and functional dynamics of immune cell signatures in the mucosal and systemic tissue of mice in response to N. musculi colonization. We found key genes and pathways vital for immune homeostasis in palate tissue, validated by flow cytometry of immune cells from the lung, blood, and spleen. This study offers a novel avenue for advancing our understanding of host-bacteria dynamics and may provide a platform for developing efficacious interventions against mucosal persistence by pathogenic Neisseria.


Sujet(s)
Neisseria , Animaux , Souris , Neisseria/immunologie , Interactions hôte-pathogène/immunologie , Femelle , Souris de lignée C57BL , Modèles animaux de maladie humaine , Bouche/microbiologie , Bouche/immunologie
10.
Infect Immun ; 92(5): e0000424, 2024 May 07.
Article de Anglais | MEDLINE | ID: mdl-38563734

RÉSUMÉ

Neisseria gonorrhoeae, a human restricted pathogen, releases inflammatory peptidoglycan (PG) fragments that contribute to the pathophysiology of pelvic inflammatory disease. The genus Neisseria is also home to multiple species of human- or animal-associated Neisseria that form part of the normal microbiota. Here we characterized PG release from the human-associated nonpathogenic species Neisseria lactamica and Neisseria mucosa and animal-associated Neisseria from macaques and wild mice. An N. mucosa strain and an N. lactamica strain were found to release limited amounts of the proinflammatory monomeric PG fragments. However, a single amino acid difference in the PG fragment permease AmpG resulted in increased PG fragment release in a second N. lactamica strain examined. Neisseria isolated from macaques also showed substantial release of PG monomers. The mouse colonizer Neisseria musculi exhibited PG fragment release similar to that seen in N. gonorrhoeae with PG monomers being the predominant fragments released. All the human-associated species were able to stimulate NOD1 and NOD2 responses. N. musculi was a poor inducer of mouse NOD1, but ldcA mutation increased this response. The ability to genetically manipulate N. musculi and examine effects of different PG fragments or differing amounts of PG fragments during mouse colonization will lead to a better understanding of the roles of PG in Neisseria infections. Overall, we found that only some nonpathogenic Neisseria have diminished release of proinflammatory PG fragments, and there are differences even within a species as to types and amounts of PG fragments released.


Sujet(s)
Neisseria , Protéine adaptatrice de signalisation NOD1 , Protéine adaptatrice de signalisation NOD2 , Peptidoglycane , Animaux , Humains , Souris , Protéines bactériennes/métabolisme , Protéines bactériennes/génétique , Protéines de transport membranaire , Neisseria/génétique , Neisseria gonorrhoeae/immunologie , Neisseria gonorrhoeae/génétique , Neisseria gonorrhoeae/métabolisme , Protéine adaptatrice de signalisation NOD1/métabolisme , Protéine adaptatrice de signalisation NOD1/génétique , Protéine adaptatrice de signalisation NOD2/métabolisme , Protéine adaptatrice de signalisation NOD2/génétique , Peptidoglycane/métabolisme
11.
Vet Pathol ; 61(5): 783-791, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-38440886

RÉSUMÉ

Three cats, aged 2 to 11 years, presented to the University of Minnesota Veterinary Diagnostic Laboratory over a 3-year period following euthanasia or death due to respiratory distress. Thoracic radiographs revealed nodular, soft tissue opacities throughout the lung fields in all cases. On postmortem examination, approximately 60% to 80% of the lung parenchyma were expanded by multifocal to coalescing, well-demarcated, beige, semi-firm nodules. Histologically, large numbers of neutrophils, fewer macrophages, fibrin, and cellular and karyorrhectic debris effaced the pulmonary parenchyma. The inflammatory foci contained aggregates of gram-negative cocci. 16s rRNA Sanger sequencing and whole-genome sequencing identified the bacteria isolated from the lung of all cats under aerobic conditions as a novel Neisseria spp. Based on whole-genome sequence analysis, all 3 sequences shared 92.71% and 92.67% average nucleotide identity with closely related Neisseria animaloris NZ LR134440T and Neisseria animaloris GCA 002108605T, respectively. The in silico DNA-DNA hybridization identity compared to our isolates was 46.6% and 33.8% with strain DSM Neisseria zoodegmatis 21642 and strain DSM 21643, respectively. All 3 sequences have less than 95% average nucleotide identity and less than 70% DNA-DNA hybridization identity, suggesting that the 3 isolates are a novel species of the genus Neisseria. Infection with Neisseria spp. induces an embolic pneumonia in cats that radiographically and pathologically resembles a metastatic neoplastic process and should be considered among the etiologic differential diagnoses in cases of infectious pulmonary disease with a disseminated, nodular lung pattern.


Sujet(s)
Maladies des chats , Poumon , Neisseria , Animaux , Chats , Maladies des chats/microbiologie , Maladies des chats/anatomopathologie , Neisseria/isolement et purification , Poumon/anatomopathologie , Poumon/microbiologie , Mâle , Femelle , ARN ribosomique 16S/génétique , Pneumopathie bactérienne/médecine vétérinaire , Pneumopathie bactérienne/anatomopathologie , Pneumopathie bactérienne/microbiologie , Séquençage du génome entier , Phylogenèse
12.
Microb Genom ; 10(2)2024 Feb.
Article de Anglais | MEDLINE | ID: mdl-38381035

RÉSUMÉ

Mosaic penA alleles formed through horizontal gene transfer (HGT) have been instrumental to the rising incidence of ceftriaxone-resistant gonococcal infections. Although interspecies HGT of regions of the penA gene between Neisseria gonorrhoeae and commensal Neisseria species has been described, knowledge concerning which species are the most common contributors to mosaic penA alleles is limited, with most studies examining only a small number of alleles. Here, we investigated the origins of recombinant penA alleles through in silico analyses that incorporated 1700 penA alleles from 35 513 Neisseria isolates, comprising 15 different Neisseria species. We identified Neisseria subflava and Neisseria cinerea as the most common source of recombinant sequences in N. gonorrhoeae penA. This contrasted with Neisseria meningitidis penA, for which the primary source of recombinant DNA was other meningococci, followed by Neisseria lactamica. Additionally, we described the distribution of polymorphisms implicated in antimicrobial resistance in penA, and found that these are present across the genus. These results provide insight into resistance-related changes in the penA gene across human-associated Neisseria species, illustrating the importance of genomic surveillance of not only the pathogenic Neisseria, but also of the oral niche-associated commensals from which these pathogens are sourcing key genetic variation.


Sujet(s)
Gonorrhée , Neisseria meningitidis , Humains , Mosaïcisme , Neisseria/génétique , Neisseria gonorrhoeae/génétique
13.
Article de Anglais | MEDLINE | ID: mdl-38253396

RÉSUMÉ

Amylosucrase (EC 2.4.1.4) is a versatile enzyme with significant potential in biotechnology and food production. To facilitate its efficient preparation, a novel expression strategy was implemented in Bacillus licheniformis for the secretory expression of Neisseria polysaccharea amylosucrase (NpAS). The host strain B. licheniformis CBBD302 underwent genetic modification through the deletion of sacB, a gene responsible for encoding levansucrase that synthesizes extracellular levan from sucrose, resulting in a levan-deficient strain, B. licheniformis CBBD302B. Neisseria polysaccharea amylosucrase was successfully expressed in B. licheniformis CBBD302B using the highly efficient Sec-type signal peptide SamyL, but its extracellular translocation was unsuccessful. Consequently, the expression of NpAS via the twin-arginine translocation (TAT) pathway was investigated using the signal peptide SglmU. The study revealed that NpAS could be effectively translocated extracellularly through the TAT pathway, with the signal peptide SglmU facilitating the process. Remarkably, 62.81% of the total expressed activity was detected in the medium. This study marks the first successful secretory expression of NpAS in Bacillus species host cells, establishing a foundation for its future efficient production. ONE-SENTENCE SUMMARY: Amylosucrase was secreted in Bacillus licheniformis via the twin-arginine translocation pathway.


Sujet(s)
Bacillus licheniformis , Glucosyltransferases , Neisseria , Bacillus licheniformis/métabolisme , Signaux de triage des protéines/génétique , Fructanes , Arginine , Protéines bactériennes/génétique
14.
Sci Rep ; 14(1): 1179, 2024 01 12.
Article de Anglais | MEDLINE | ID: mdl-38216602

RÉSUMÉ

One of the most promising new treatments for gonorrhoea currently in phase 3 clinical trials is zoliflodacin. Studies have found very little resistance to zoliflodacin in currently circulating N. gonorrhoeae strains, and in-vitro experiments demonstrated that it is difficult to induce resistance. However, zoliflodacin resistance may emerge in commensal Neisseria spp., which could then be transferred to N. gonorrhoeae via transformation. In this study, we investigated this commensal-resistance-pathway hypothesis for zoliflodacin. To induce zoliflodacin resistance, ten wild-type susceptible isolates belonging to 5 Neisseria species were serially passaged for up to 48 h on gonococcal agar plates containing increasing zoliflodacin concentrations. Within 7 to 10 days, all strains except N. lactamica, exhibited MICs of ≥ 4 µg/mL, resulting in MIC increase ranging from 8- to 64-fold. The last passaged strains and their baseline were sequenced. We detected mutations previously reported to cause zoliflodacin resistance in GyrB (D429N and S467N), novel mutations in the quinolone resistance determining region (QRDR) (M464R and T472P) and mutations outside the QRDR at amino acid positions 28 and 29 associated with low level resistance (MIC 2 µg/mL). Genomic DNA from the laboratory evolved zoliflodacin-resistant strains was transformed into the respective baseline wild-type strain, resulting in MICs of ≥ 8 µg/mL in most cases. WGS of transformants with decreased zoliflodacin susceptibility revealed presence of the same zoliflodacin resistance determinants as observed in the donor strains. Two inter-species transformation experiments were conducted to investigate whether zoliflodacin resistance determinants of commensal Neisseria spp. could be acquired by N. gonorrhoeae. N. gonorrhoeae strain WHO P was exposed to (i) pooled genomic DNA from the two resistant N. mucosa strains and (ii) a gyrB amplicon of the resistant N. subflava strain 45/1_8. Transformants of both experiments exhibited an MIC of 2 µg/mL and whole genome analysis revealed uptake of the mutations detected in the donor strains. This is the first in-vitro study to report that zoliflodacin resistance can be induced in commensal Neisseria spp. and subsequently transformed into N. gonorrhoeae.


Sujet(s)
Barbituriques , Gonorrhée , Isoxazoles , Morpholines , Oxazolidinones , Quinolinone , Spiranes , Humains , Neisseria/génétique , Neisseria gonorrhoeae , Quinolinone/pharmacologie , Tests de sensibilité microbienne , ADN , Antibactériens/pharmacologie
15.
Microbiol Spectr ; 12(2): e0350723, 2024 Feb 06.
Article de Anglais | MEDLINE | ID: mdl-38179941

RÉSUMÉ

Species within the genus Neisseria are adept at sharing adaptive allelic variation, with commensal species repeatedly transferring resistance to their pathogenic relative Neisseria gonorrhoeae. However, resistance in commensals is infrequently characterized, limiting our ability to predict novel and potentially transferable resistance mechanisms that ultimately may become important clinically. Unique evolutionary starting places of each Neisseria species will have distinct genomic backgrounds, which may ultimately control the fate of evolving populations in response to selection as epistatic and additive interactions coerce lineages along divergent evolutionary trajectories. Alternatively, similar genetic content present across species due to shared ancestry may constrain existing adaptive solutions. Thus, identifying the paths to resistance across commensals may aid in characterizing the Neisseria resistome-or the reservoir of alleles within the genus as well as its depth. Here, we use in vitro evolution of four commensal species to investigate the potential and repeatability of resistance evolution to two antimicrobials, the macrolide azithromycin and the ß-lactam penicillin. After 20 days of selection, commensals evolved resistance to penicillin and azithromycin in 11/16 and 12/16 cases, respectively. Almost all cases of resistance emergence converged on mutations within ribosomal components or the mtrRCDE efflux pump for azithromycin-based selection and mtrRCDE, penA, and rpoB for penicillin selection, thus supporting constrained adaptive solutions despite divergent evolutionary starting points across the genus for these particular drugs. Though drug-selected loci were limited, we do identify novel resistance-imparting mutations. Continuing to explore paths to resistance across different experimental conditions and genomic backgrounds, which could shunt evolution down alternative evolutionary trajectories, will ultimately flesh out the full Neisseria resistome.IMPORTANCENeisseria gonorrhoeae is a global threat to public health due to its rapid acquisition of antibiotic resistance to all first-line treatments. Recent work has documented that alleles acquired from close commensal relatives have played a large role in the emergence of resistance to macrolides and beta-lactams within gonococcal populations. However, commensals have been relatively underexplored for the resistance genotypes they may harbor. This leaves a gap in our understanding of resistance that could be rapidly acquired by the gonococcus through a known highway of horizontal gene exchange. Here, we characterize resistance mechanisms that can emerge in commensal Neisseria populations via in vitro selection to multiple antimicrobials and begin to define the number of paths to resistance. This study, and other similar works, may ultimately aid both surveillance efforts and clinical diagnostic development by nominating novel and conserved resistance mechanisms that may be at risk of rapid dissemination to pathogen populations.


Sujet(s)
Anti-infectieux , Gonorrhée , Humains , Neisseria , Azithromycine/pharmacologie , Résistance bactérienne aux médicaments/génétique , Antibactériens/pharmacologie , Neisseria gonorrhoeae/génétique , Gonorrhée/traitement médicamenteux , Anti-infectieux/pharmacologie , bêta-Lactames/pharmacologie , Tests de sensibilité microbienne , Pénicillines
16.
Int J Pediatr Otorhinolaryngol ; 176: 111788, 2024 Jan.
Article de Anglais | MEDLINE | ID: mdl-38039804

RÉSUMÉ

OBJECTIVE: This study aimed to compare the tonsillar microbiota between post tonsillectomy patients with bleeding and without bleeding, and to investigate the potential role of tonsillar microbiota in the development of post-tonsillectomy hemorrhage (PTH). METHODS: Nineteen tonsillar tissues from PTH patients and 21 tissues from control patients were collected. Metagenomic sequencing was used to compare the microbiota in PTH and control groups. Alpha diversity indices were used to compare the richness and evenness of the microbiota between the two groups. PCoA and NMDS analyses were used to evaluate beta diversity. LDA analysis was conducted to identify significantly abundant genera. RESULTS: No significant difference in alpha diversity indices was found between PTH and control patients. The dominant bacteria in the tonsillar microbiota were Haemophilus, Streptococcus, and Fusobacterium. PCoA and NMDS analyses showed significant differences in beta diversity between PTH and control patients. PTH patients had a significantly higher relative abundance of Neisseria, Capnocytophaga, and Veillonella. Capnocytophaga was also identified as a significantly abundant genus by LDA analysis. CONCLUSION: This study demonstrates that there is a difference in the tonsillar microbiota between PTH and control patients. The results suggest that Neisseria, Capnocytophaga, and Veillonella may be associated with the development of PTH. These findings provide new insights into the potential role of the tonsillar microbiota in the development of PTH, and may help to develop new strategies for preventing and treating this potentially life-threatening complication.


Sujet(s)
Microbiote , Amygdalectomie , Enfant , Humains , Tonsille palatine/chirurgie , Tonsille palatine/microbiologie , Amygdalectomie/effets indésirables , Hémorragie , Hypertrophie , Neisseria
17.
mBio ; 15(1): e0279223, 2024 Jan 16.
Article de Anglais | MEDLINE | ID: mdl-38084997

RÉSUMÉ

IMPORTANCE: We describe the importance of Type IV pilus retraction to colonization and persistence by a mouse commensal Neisseria, N. musculi, in its native host. Our findings have implications for the role of Tfp retraction in mediating interactions of human-adapted pathogenic and commensal Neisseria with their human host due to the relatedness of these species.


Sujet(s)
Protéines de fimbriae , Fimbriae bactériens , Souris , Animaux , Humains , Neisseria/génétique , Symbiose , Neisseria gonorrhoeae , Protéines bactériennes
18.
Front Cell Infect Microbiol ; 13: 1308550, 2023.
Article de Anglais | MEDLINE | ID: mdl-38076458

RÉSUMÉ

Commensal Neisseria species of the oropharynx represent a significant reservoir of antimicrobial resistance determinants that can be transferred to Neisseria gonorrhoeae. This aspect is particularly crucial in 'men having sex with men' (MSM), a key population in which pharyngeal co-colonization by N. gonorrhoeae and non-pathogenic Neisseria species is frequent and associated with the emergence of antimicrobial resistance. Here, we explored the antimicrobial susceptibility of a large panel of non-pathogenic Neisseria species isolated from the oropharynx of two populations: a group of MSM attending a 'sexually transmitted infection' clinic in Bologna (Italy) (n=108) and a group of males representing a 'general population' (n=119). We collected 246 strains, mainly belonging to N. subflava (60%) and N. flavescens (28%) species. Their antimicrobial susceptibility was evaluated assessing the minimum inhibitory concentrations (MICs) for azithromycin, ciprofloxacin, cefotaxime, and ceftriaxone using E-test strips. Overall, commensal Neisseria spp. showed high rates of resistance to azithromycin (90%; median MICs: 4.0 mg/L), and ciprofloxacin (58%; median MICs: 0.12 mg/L), whereas resistance to cephalosporins was far less common (<15%). Neisseria strains from MSM were found to have significantly higher MICs for azithromycin (p=0.0001) and ciprofloxacin (p<0.0001) compared to those from the general population. However, there was no significant difference in cephalosporin MICs between the two groups. The surveillance of the antimicrobial resistance of non-pathogenic Neisseria spp. could be instrumental in predicting the risk of the spread of multi-drug resistant gonorrhea. This information could be an early predictor of an excessive use of antimicrobials, paving the way to innovative screening and prevention policies.


Sujet(s)
Antibactériens , Anti-infectieux , Humains , Mâle , Antibactériens/pharmacologie , Antibactériens/usage thérapeutique , Azithromycine/pharmacologie , Études transversales , Neisseria , Résistance bactérienne aux médicaments , Ciprofloxacine/pharmacologie , Neisseria gonorrhoeae , Céphalosporines/pharmacologie , Anti-infectieux/pharmacologie , Tests de sensibilité microbienne , Partie orale du pharynx
19.
Med Sci (Basel) ; 11(4)2023 12 01.
Article de Anglais | MEDLINE | ID: mdl-38132917

RÉSUMÉ

Neisseria meningitidis (N. meningitidis) serogroup B (MenB) is the leading cause of invasive meningococcal disease worldwide. The pathogen has a wide range of virulence factors, which are potential vaccine components. Studying the genetic variability of antigens within a population, especially their long-term persistence, is necessary to develop new vaccines and predict the effectiveness of existing ones. The multicomponent 4CMenB vaccine (Bexsero), used since 2014, contains three major genome-derived recombinant proteins: factor H-binding protein (fHbp), Neisserial Heparin-Binding Antigen (NHBA) and Neisserial adhesin A (NadA). Here, we assessed the prevalence and sequence variations of these vaccine antigens in a panel of 5667 meningococcal isolates collected worldwide over the past 10 years and deposited in the PubMLST database. Using multiple amino acid sequence alignments and Random Forest Classifier machine learning methods, we estimated the potential strain coverage of fHbp and NHBA vaccine variants (51 and about 25%, respectively); the NadA antigen sequence was found in only 18% of MenB genomes analyzed, but cross-reactive variants were present in less than 1% of isolates. Based on our findings, we proposed various strategies to improve the 4CMenB vaccine and broaden the coverage of N. meningitidis strains.


Sujet(s)
Infections à méningocoques , Vaccins antiméningococciques , Neisseria meningitidis sérogroupe B , Neisseria meningitidis , Humains , Antigènes bactériens/génétique , Infections à méningocoques/prévention et contrôle , Vaccins antiméningococciques/génétique , 59641 , Neisseria meningitidis sérogroupe B/génétique , Adhésines bactériennes/génétique , Neisseria meningitidis/génétique , Neisseria , Biologie informatique , Pronostic
20.
Nat Commun ; 14(1): 7706, 2023 Nov 24.
Article de Anglais | MEDLINE | ID: mdl-38001084

RÉSUMÉ

Core genome multilocus sequence typing (cgMLST) is commonly used to classify bacterial strains into different types, for taxonomical and epidemiological applications. However, cgMLST schemes require central databases for the nomenclature of new alleles and sequence types, which must be synchronized worldwide and involve increasingly intensive calculation and storage demands. Here, we describe a distributed cgMLST (dcgMLST) scheme that does not require a central database of allelic sequences and apply it to study evolutionary patterns of epidemic and endemic strains of the genus Neisseria. We classify 69,994 worldwide Neisseria strains into multi-level clusters that assign species, lineages, and local disease outbreaks. We divide Neisseria meningitidis into 168 endemic lineages and three epidemic lineages responsible for at least 9 epidemics in the past century. According to our analyses, the epidemic and endemic lineages experienced very different population dynamics in the past 100 years. Epidemic lineages repetitively emerged from endemic lineages, disseminated worldwide, and apparently disappeared rapidly afterward. We propose a stepwise model for the evolutionary trajectory of epidemic lineages in Neisseria, and expect that the development of similar dcgMLST schemes will facilitate epidemiological studies of other bacterial pathogens.


Sujet(s)
Neisseria meningitidis , Neisseria meningitidis/génétique , Neisseria/génétique , Génome bactérien/génétique , Génotype , Typage par séquençage multilocus , Analyse de regroupements
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE