Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
Antimicrob Agents Chemother ; 66(6): e0238321, 2022 06 21.
Artigo em Inglês | MEDLINE | ID: mdl-35652645

RESUMO

Invasive meningococcal disease (IMD) due to serogroup Y Neisseria meningitidis (NmY) is rare in China; recently, an invasive NmY isolate, Nm512, was discovered in Shanghai with decreased susceptibility to penicillin (PenNS). Here, we investigated the epidemiology of NmY isolates in Shanghai and explored the potential commensal Neisseria lactamica donor of the PenNS NmY isolate. A total of 491 N. meningitidis and 724 commensal Neisseria spp. isolates were collected. Eleven NmY isolates were discovered from IMD (n = 1) and carriers (n = 10), including two PenNS isolates with five-key-mutation-harboring (F504L-A510V-I515V-H541N-I566V) penA genes. Five of the eight ST-175 complex (CC175) isolates had a genotype [Y:P1.5-1,2-2:F5-8:ST-175(CC175)] identical to that of the predominant invasive clone found in South Africa. Only one invasive NmY CC23 isolate (Nm512) was discovered; this isolate carried a novel PenNSpenA832 allele, which was identified in commensal N. lactamica isolates locally. Recombination analysis and transformation of the penA allele highlighted that N. meningitidis Nm512 may acquire resistance from its commensal donor; this was supported by the similar distribution of transformation-required DNA uptake sequence variants and the highly cognate receptor ComP between N. meningitidis and N. lactamica. In 2,309 NmY CC23 genomes from the PubMLST database, isolates with key-mutation-harboring penA genes comprised 12% and have been increasing since the 1990s, accompanied by recruitment of the blaROB-1 and/or quinolone resistance allele. Moreover, penA22 was predominant among genomes without key mutations in penA. These results strongly suggest that Nm512 is a descendant of the penA22-harboring CC23 isolate from Europe and acquired its penicillin resistance locally from commensal N. lactamica species by natural transformation.


Assuntos
Infecções Meningocócicas , Neisseria lactamica , Neisseria meningitidis , China/epidemiologia , Humanos , Neisseria lactamica/genética , Neisseria meningitidis/genética , Neisseria meningitidis Sorogrupo Y , Resistência às Penicilinas/genética , Sorogrupo
2.
BMJ Open ; 12(5): e056081, 2022 05 18.
Artigo em Inglês | MEDLINE | ID: mdl-35584870

RESUMO

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.


Assuntos
Microbiota , Neisseria lactamica , Neisseria meningitidis , Adulto , Criança , Pré-Escolar , Feminino , Humanos , Lactente , Recém-Nascido , Microbiota/genética , Mães , Neisseria lactamica/genética , Faringe , Projetos Piloto , Gravidez , RNA Ribossômico 16S
3.
Mol Cell ; 82(4): 852-867.e5, 2022 02 17.
Artigo em Inglês | MEDLINE | ID: mdl-35051351

RESUMO

Leading CRISPR-Cas technologies employ Cas9 and Cas12 enzymes that generate RNA-guided dsDNA breaks. Yet, the most abundant microbial adaptive immune systems, Type I CRISPRs, are under-exploited for eukaryotic applications. Here, we report the adoption of a minimal CRISPR-Cas3 from Neisseria lactamica (Nla) type I-C system to create targeted large deletions in the human genome. RNP delivery of its processive Cas3 nuclease and target recognition complex Cascade can confer ∼95% editing efficiency. Unexpectedly, NlaCascade assembly in bacteria requires internal translation of a hidden component Cas11 from within the cas8 gene. Furthermore, expressing a separately encoded NlaCas11 is the key to enable plasmid- and mRNA-based editing in human cells. Finally, we demonstrate that supplying cas11 is a universal strategy to systematically implement divergent I-C, I-D, and I-B CRISPR-Cas3 editors with compact sizes, distinct PAM preferences, and guide orthogonality. These findings greatly expand our ability to engineer long-range genome edits.


Assuntos
Proteínas de Bactérias/genética , Proteínas Associadas a CRISPR/genética , Sistemas CRISPR-Cas , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , Deleção de Genes , Edição de Genes , Genoma Humano , Neisseria lactamica/genética , Proteínas de Bactérias/metabolismo , Proteínas Associadas a CRISPR/metabolismo , Células HEK293 , Células HeLa , Humanos , Neisseria lactamica/enzimologia , Ribonucleoproteínas/genética , Ribonucleoproteínas/metabolismo
4.
J Infect Chemother ; 27(1): 65-69, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-32873462

RESUMO

INTRODUCTION: Neisseria lactamica is a commensal bacterium of the upper respiratory tract in humans and is closely related to Neisseria meningitidis. N. lactamica colonization may contribute to preventing N. meningitidis colonization and invasive meningococcal disease. However, the transference of antimicrobial resistance genes from N. lactamica to N. meningitidis has been reported. METHODS: In this study, we aimed to identify N. lactamica using matrix-assisted laser desorption ionization-time of flight mass spectrometry (MALDI-TOF MS) and performed multilocus sequence typing of seven N. lactamica strains isolated from Japanese children. We also analyzed the antimicrobial susceptibility of these strains and the mutations in their antimicrobial resistance genes (penA, gyrA, and parC). RESULTS: All the N. lactamica strains could be identified using MALDI-TOF MS. All strains were of different sequence types (STs), including five new STs. Five strains had intermediate susceptibility, two were resistant to ampicillin, and all had five out of the five known PBP2 mutations. Six strains were resistant to levofloxacin. Among the quinolone-resistant strains, three had GyrA mutations, and three had both ParC and GyrA mutations. CONCLUSIONS: N. lactamica STs may vary in Japanese children, and penicillin- and quinolone-resistant strains may be prevalent. We should pay attention not only to the drug resistance of N. meningitidis but also to the drug susceptibility of N. lactamica whose drug-resistance genes may transfer to N. meningitidis.


Assuntos
Infecções Meningocócicas , Neisseria lactamica , Neisseria meningitidis , Criança , Humanos , Japão/epidemiologia , Neisseria lactamica/genética , Neisseria meningitidis/genética , Sistema Respiratório
5.
BMJ Open ; 9(4): e026544, 2019 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-31048443

RESUMO

INTRODUCTION: Neisseria lactamica is a commensal organism found in the human nasopharynx and is closely related to the pathogen N. meningitidis (meningococcus). Carriage of N. lactamica is associated with reduced meningococcal carriage and disease. We summarise an ethically approved protocol for an experimental human challenge study using a genetically modified strain of N. lactamica that expresses the meningococcal antigen NadA. We aim to develop a model to study the role of specific bacterial antigens in nasopharyngeal carriage and immunity, to evaluate vaccines for their efficacy in preventing colonisation and to provide a proof of principle for the development of bacterial medicines. METHODS AND ANALYSIS: Healthy adult volunteers aged 18-45 years will receive an intranasal inoculation of either the NadA containing strain of N. lactamica or a genetically modified, but wild-type equivalent control strain. These challenge volunteers will be admitted for 4.5 days observation following inoculation and will then be discharged with strict infection control rules. Bedroom contacts of the challenge volunteers will also be enrolled as contact volunteers. Safety, colonisation, shedding, transmission and immunogenicity will be assessed over 90 days after which carriage will be terminated with antibiotic eradication therapy. ETHICS AND DISSEMINATION: This study has been approved by the Department for Environment, Food and Rural Affairs and South Central Oxford A Research Ethics Committee (reference: 18/SC/0133). Findings will be published in peer-reviewed open-access journals as soon as possible. TRIAL REGISTRATION NUMBER: NCT03630250; Pre-results.


Assuntos
Adesinas Bacterianas/biossíntese , Adesinas Bacterianas/imunologia , Antígenos/imunologia , Vacinas Meningocócicas/imunologia , Microrganismos Geneticamente Modificados , Neisseria lactamica/genética , Neisseria lactamica/metabolismo , Neisseria meningitidis/imunologia , Projetos de Pesquisa , Adolescente , Adulto , Pesquisa Biomédica , Humanos , Pessoa de Meia-Idade , Estudos Prospectivos , Adulto Jovem
6.
J Mol Biol ; 431(11): 2082-2094, 2019 05 17.
Artigo em Inglês | MEDLINE | ID: mdl-30995450

RESUMO

Specificity engineering is challenging and particularly difficult for enzymes that have the catalytic machinery and specificity determinants in close proximity. Restriction endonucleases have been used as a paradigm for protein engineering, but successful cases are rare. Here, we present the results of a directed evolution approach to the engineering of a dimeric, blunt end cutting restriction enzyme NlaIV (GGN/NCC). Based on the remote similarity to EcoRV endonuclease, regions for random mutagenesis and in vitro evolution were chosen. The obtained variants cleaved target sites with an up to 100-fold kcat/KM preference for AT or TA (GGW/WCC) over GC or CG (GGS/SCC) in the central dinucleotide step, compared to the only ~17-fold preference of the wild-type enzyme. To understand the basis of the increased specificity, we determined the crystal structure of NlaIV. Despite the presence of DNA in the crystallization mix, the enzyme crystallized in the free form. We therefore constructed a computational model of the NlaIV-DNA complex. According to the model, the mutagenesis of the regions that were in the proximity of DNA did not lead to the desired specificity change, which was instead conveyed in an indirect manner by substitutions in the more distant regions.


Assuntos
Proteínas de Bactérias/química , Desoxirribonucleases de Sítio Específico do Tipo II/química , Neisseria lactamica/química , Proteínas de Bactérias/genética , Cristalografia por Raios X , Desoxirribonucleases de Sítio Específico do Tipo II/genética , Humanos , Modelos Moleculares , Mutagênese Sítio-Dirigida , Neisseria lactamica/genética , Infecções por Neisseriaceae/microbiologia , Conformação Proteica , Especificidade por Substrato
7.
Nat Commun ; 9(1): 4753, 2018 11 12.
Artigo em Inglês | MEDLINE | ID: mdl-30420631

RESUMO

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.


Assuntos
Nasofaringe/microbiologia , Neisseria lactamica/crescimento & desenvolvimento , Infecções por Neisseriaceae/microbiologia , Portador Sadio , Contagem de Colônia Microbiana , Genes Bacterianos , Humanos , Mutação/genética , Taxa de Mutação , Neisseria lactamica/genética , Neisseria lactamica/isolamento & purificação , Recombinação Genética/genética
8.
Int J Med Microbiol ; 308(4): 454-458, 2018 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-29605532

RESUMO

Neisseria lactamica is a nonpathogenic commensal bacterium that is potentially associated with the development of natural immunity against N. meningitidis. However, the genetic variation present in natural populations of N. lactamica has not been fully investigated. To better understand its epidemiology and genetic variation, we studied N. lactamica carriage in 1200 students aged 11-19 years old in Salvador, Brazil. The carriage prevalence was 4.5% (54/1200), with no statistical difference among sex and age, although we observed a trend towards higher carriage prevalence among 11-year-old individuals. Whole genome sequence analysis revealed a high genetic diversity among the isolates, with the presence of 32 different STs, 28 (87.5%) of which were new. A total of 21/50 (42%) isolates belonged to three different clonal complexes. While none of the isolates contained nadA or fHpb alleles, we detected 21 FetA variants, 20 NhbA variants and two variants of PorB. The data provide detailed information on circulating N. lactamica isolates in adolescents in Brazil and are complementary to studies in other countries.


Assuntos
Portador Sadio/epidemiologia , Portador Sadio/microbiologia , Infecções Meningocócicas/epidemiologia , Infecções Meningocócicas/microbiologia , Neisseria lactamica/genética , Adolescente , Alelos , Proteínas da Membrana Bacteriana Externa/genética , Brasil/epidemiologia , El Salvador/epidemiologia , Feminino , Genótipo , Humanos , Masculino , Epidemiologia Molecular , Neisseria lactamica/isolamento & purificação , Neisseria meningitidis/genética , Polimorfismo de Nucleotídeo Único , Porinas/genética , Estudantes , Sequenciamento Completo do Genoma , Adulto Jovem
9.
J Clin Microbiol ; 54(12): 2891-2899, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27629899

RESUMO

A carriage study was undertaken (n = 112) to ascertain the prevalence of Neisseria spp. following the eighth case of invasive meningococcal disease in young children (5 to 46 months) and members of a large extended indigenous ethnic minority Traveller family (n = 123), typically associated with high-occupancy living conditions. Nested multilocus sequence typing (MLST) was employed for case specimen extracts. Isolates were genome sequenced and then were assembled de novo and deposited into the Bacterial Isolate Genome Sequencing Database (BIGSdb). This facilitated an expanded MLST approach utilizing large numbers of loci for isolate characterization and discrimination. A rare sequence type, ST-6697, predominated in disease specimens and isolates that were carried (n = 8/14), persisting for at least 44 months, likely driven by the high population density of houses (n = 67/112) and trailers (n = 45/112). Carriage for Neisseria meningitidis (P < 0.05) and Neisseria lactamica (P < 0.002) (2-sided Fisher's exact test) was more likely in the smaller, more densely populated trailers. Meningococcal carriage was highest in 24- to 39-year-olds (45%, n = 9/20). Evidence of horizontal gene transfer (HGT) was observed in four individuals cocolonized by Neisseria lactamica and Neisseria meningitidis One HGT event resulted in the acquisition of 26 consecutive N. lactamica alleles. This study demonstrates how housing density can drive meningococcal transmission and carriage, which likely facilitated the persistence of ST-6697 and prolonged the outbreak. Whole-genome MLST effectively distinguished between highly similar outbreak strain isolates, including those isolated from person-to-person transmission, and also highlighted how a few HGT events can distort the true phylogenetic relationship between highly similar clonal isolates.


Assuntos
Surtos de Doenças , Infecções Meningocócicas/epidemiologia , Infecções Meningocócicas/transmissão , Neisseria lactamica/isolamento & purificação , Neisseria meningitidis Sorogrupo B/isolamento & purificação , Densidade Demográfica , Adolescente , Adulto , Criança , Pré-Escolar , Transferência Genética Horizontal/genética , Genoma Bacteriano/genética , Humanos , Lactente , Infecções Meningocócicas/microbiologia , Tipagem de Sequências Multilocus , Neisseria lactamica/genética , Neisseria meningitidis Sorogrupo B/genética , Adulto Jovem
10.
Int J Med Microbiol ; 305(7): 729-35, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26321007

RESUMO

In meningococci, reduced penicillin susceptibility is associated with five specific mutations in the transpeptidase region of penicillin binding protein 2 (PBP2). We showed that the same set of mutations was present in 64 of 123 Neisseria lactamica strains obtained from a carriage study (MIC range: 0.125-2.0mg/L). The PBP2 encoding penA alleles in these strains were genetically similar to those found in intermediate resistant meningococci suggesting frequent interspecies genetic exchange. Fifty-six N. lactamica isolates with mostly lower penicillin MICs (range: 0.064-0.38mg/L) exhibited only three of the five mutations. The corresponding penA alleles were unique to N. lactamica and formed a distinct genetic clade. PenA alleles with no mutations on the other hand were unique to meningococci. Under penicillin selective pressure, genetic transformation of N. lactamica penA alleles in meningococci was only possible for alleles encoding five mutations, but not for those encoding three mutations; the transfer resulted in MICs comparable to those of meningococci harboring penA alleles that encoded PBP2 with five mutations, but considerably lower than those of the corresponding N. lactamica donor strains. Due to a transformation barrier the complete N. lactamica penA could not be transformed into N. meningitidis. In summary, penicillin MICs in N. lactamica were associated with the number of mutations in the transpeptidase region of PBP2. Evidence for interspecific genetic transfer was only observed for penA alleles associated with higher MICs, suggesting that alleles encoding only three mutations in the transpeptidase region are biologically not effective in N. meningitidis. Factors other than PBP2 seem to be responsible for the high levels of penicillin resistance in N. lactamica. A reduction of penicillin susceptibility in N. meningitidis by horizontal gene transfer from N. lactamica is unlikely to happen.


Assuntos
Neisseria lactamica/efeitos dos fármacos , Neisseria meningitidis/efeitos dos fármacos , Resistência às Penicilinas , Proteínas de Ligação às Penicilinas/genética , Penicilinas/farmacologia , Polimorfismo Genético , Substituição de Aminoácidos , Transferência Genética Horizontal , Genótipo , Alemanha , Humanos , Testes de Sensibilidade Microbiana , Mutação de Sentido Incorreto , Neisseria lactamica/genética , Neisseria lactamica/isolamento & purificação , Neisseria meningitidis/genética , Neisseria meningitidis/isolamento & purificação , Infecções por Neisseriaceae/microbiologia , Transformação Bacteriana
11.
Mol Microbiol ; 93(2): 346-55, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24910087

RESUMO

Neisseria meningitidis is an important human pathogen that is capable of killing within hours of infection. Its normal habitat is the nasopharynx of adult humans. Here we identify a genomic island (the prp gene cluster) in N. meningitidis that enables this species to utilize propionic acid as a supplementary carbon source during growth, particularly under nutrient poor growth conditions. The prp gene cluster encodes enzymes for a methylcitrate cycle. Novel aspects of the methylcitrate cycle in N. meningitidis include a propionate kinase which was purified and characterized, and a putative propionate transporter. This genomic island is absent from the close relative of N. meningitidis, the commensal Neisseria lactamica, which chiefly colonizes infants not adults. We reason that the possession of the prp genes provides a metabolic advantage to N. meningitidis in the adult oral cavity, which is rich in propionic acid-generating bacteria. Data from classical microbiological and sequence-based microbiome studies provide several lines of supporting evidence that N. meningitidis colonization is correlated with propionic acid generating bacteria, with a strong correlation between prp-containing Neisseria and propionic acid generating bacteria from the genus Porphyromonas, and that this may explain adolescent/adult colonization by N. meningitidis.


Assuntos
Regulação Bacteriana da Expressão Gênica , Ilhas Genômicas , Nasofaringe/microbiologia , Neisseria meningitidis/genética , Neisseria meningitidis/metabolismo , Propionatos/metabolismo , Adolescente , Adulto , Carbono/metabolismo , Feminino , Genoma Bacteriano , Humanos , Masculino , Microbiota , Família Multigênica , Neisseria lactamica/genética , Neisseria meningitidis/crescimento & desenvolvimento , Neisseria meningitidis/isolamento & purificação , Porphyromonas/metabolismo
12.
Clin Vaccine Immunol ; 20(9): 1360-9, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23803905

RESUMO

The poor immunogenicity of the meningococcal serogroup B (MenB) capsule has led to the development of vaccines targeting subcapsular antigens, in particular the immunodominant and diverse outer membrane porin, PorA. These vaccines are largely strain specific; however, they offer limited protection against the diverse MenB-associated diseases observed in many industrialized nations. To broaden the scope of its protection, the multicomponent vaccine (4CMenB) incorporates a PorA-containing outer membrane vesicle (OMV) alongside relatively conserved recombinant protein components, including factor H-binding protein (fHbp), Neisseria adhesin A (NadA), and neisserial heparin-binding antigen (NHBA). The expression of PorA is unique to meningococci (Neisseria meningitidis); however, many subcapsular antigens are shared with nonpathogenic members of the genus Neisseria that also inhabit the nasopharynx. These organisms may elicit cross-protective immunity against meningococci and/or occupy a niche that might otherwise accommodate pathogens. The potential for 4CMenB responses to impact such species (and vice versa) was investigated by determining the genetic distribution of the primary 4CMenB antigens among diverse members of the common childhood commensal, Neisseria lactamica. All the isolates possessed nhba but were devoid of fhbp and nadA. The nhba alleles were mainly distinct from but closely related to those observed among a representative panel of invasive MenB isolates from the same broad geographic region. We made similar findings for the immunogenic typing antigen, FetA, which constitutes a major part of the 4CMenB OMV. Thus, 4CMenB vaccine responses may impact or be impacted by nasopharyngeal carriage of commensal neisseriae. This highlights an area for further research and surveillance should the vaccine be routinely implemented.


Assuntos
Antígenos de Bactérias/imunologia , Vacinas Meningocócicas/imunologia , Neisseria lactamica/imunologia , Neisseria meningitidis Sorogrupo B/imunologia , Adolescente , Adulto , Antígenos de Bactérias/genética , Portador Sadio/imunologia , Portador Sadio/microbiologia , Criança , Pré-Escolar , Proteção Cruzada , Reações Cruzadas , Feminino , Humanos , Lactente , Masculino , Vacinas Meningocócicas/genética , Pessoa de Meia-Idade , Nasofaringe/microbiologia , Neisseria lactamica/genética , Neisseria meningitidis Sorogrupo B/genética , Adulto Jovem
13.
J Clin Microbiol ; 50(12): 4020-7, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23035186

RESUMO

Neisseria lactamica is a true commensal bacterium occupying the same ecological niche as the pathogenic Neisseria meningitidis, which is responsible for outbreaks and large epidemics, especially in sub-Saharan Africa. To better understand the epidemiology of N. lactamica in Africa and its relationship to N. meningitidis, we studied N. lactamica carriage in 1- to 29-year-old people living in three districts of Burkina Faso from 2009 to 2011. N. lactamica was detected in 18.2% of 45,847 oropharyngeal samples. Carriage prevalence was highest among the 2-year-olds (40.1%) and decreased with age. Overall prevalence was higher for males (19.1%) than females (17.5%) (odds ratio [OR], 1.11; 95% confidence interval [CI], 1.04 to 1.18), while among the 18- to 29-year-olds, carriage prevalence was significantly higher in women (9.1%) than in men (3.9%) (OR, 2.49; 95% CI, 1.94 to 3.19). Carriage prevalence of N. lactamica was remarkably homogeneous in the three districts of Burkina Faso and stable over time, in comparison with carriage of N. meningitidis (P. A. Kristiansen et al., Clin. Vaccine Immunol. 18:435-443, 2011). There was no significant seasonal variation of N. lactamica carriage and no significant change in carriage prevalence after introduction of the serogroup A meningococcal conjugate vaccine, MenAfriVac. Multilocus sequence typing was performed on a selection of 142 isolates. The genetic diversity was high, as we identified 62 different genotypes, of which 56 were new. The epidemiology of N. lactamica carriage and the molecular characteristics of carried isolates were similar to those reported from industrialized countries, in contrast to the particularities of N. meningitidis carriage and disease epidemiology in Burkina Faso.


Assuntos
Portador Sadio/epidemiologia , Neisseria lactamica/isolamento & purificação , Infecções por Neisseriaceae/epidemiologia , Adolescente , Adulto , Burkina Faso/epidemiologia , Portador Sadio/microbiologia , Criança , Pré-Escolar , Estudos Transversais , Feminino , Variação Genética , Humanos , Lactente , Masculino , Epidemiologia Molecular , Tipagem de Sequências Multilocus , Neisseria lactamica/classificação , Neisseria lactamica/genética , Infecções por Neisseriaceae/microbiologia , Orofaringe/microbiologia , Prevalência , Adulto Jovem
14.
Infect Immun ; 80(10): 3417-28, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22825445

RESUMO

Toll-like receptors (TLRs) play a major role in host mucosal and systemic defense mechanisms by recognizing a diverse array of conserved pathogen-associated molecular patterns (PAMPs). TLR2, with TLR1 and TLR6, recognizes structurally diverse bacterial products such as lipidated factors (lipoproteins and peptidoglycans) and nonlipidated proteins, i.e., bacterial porins. PorB is a pan-neisserial porin expressed regardless of organisms' pathogenicity. However, commensal Neisseria lactamica organisms and purified N. lactamica PorB (published elsewhere as Nlac PorB) induce TLR2-dependent proinflammatory responses of lower magnitude than N. meningitidis organisms and N. meningitidis PorB (published elsewhere as Nme PorB). Both PorB types bind to TLR2 in vitro but with different apparent specificities. The structural and molecular details of PorB-TLR2 interaction are only beginning to be unraveled and may be due to electrostatic attraction. PorB molecules have significant strain-specific sequence variability within surface-exposed regions (loops) putatively involved in TLR2 interaction. By constructing chimeric recombinant PorB loop mutants in which surface-exposed loop residues have been switched between N. lactamica PorB and N. meningitidis PorB, we identified residues in loop 5 and loop 7 that influence TLR2-dependent cell activation using HEK cells and BEAS-2B cells. These loops are not uniquely responsible for PorB interaction with TLR2, but NF-κB and MAP kinases signaling downstream of TLR2 recognition are likely influenced by a hypothetical "TLR2-binding signature" within the sequence of PorB surface-exposed loops. Consistent with the effect of purified PorB in vitro, a chimeric N. meningitidis strain expressing N. lactamica PorB induces lower levels of interleukin 8 (IL-8) secretion than wild-type N. meningitidis, suggesting a role for PorB in induction of host cell activation by whole bacteria.


Assuntos
Neisseria lactamica/metabolismo , Porinas/metabolismo , Receptor 2 Toll-Like/metabolismo , Sequência de Aminoácidos , Quimiocina CCL5/genética , Quimiocina CCL5/metabolismo , Células HEK293 , Humanos , Interleucina-8/genética , Interleucina-8/metabolismo , Modelos Moleculares , Mutação , Neisseria lactamica/genética , Porinas/química , Porinas/genética , Ligação Proteica , Conformação Proteica , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais
15.
Hum Vaccin ; 7(4): 398-401, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21795847

RESUMO

From pertussis to meningococcal disease and back represents nearly 30 years of research at Porton, first at the Centre for Applied Microbiology and Research and latterly as part of the Health Protection Agency. I joined the group lead by Andy Robinson developing an acellular pertussis vaccine and was part of an exciting period that encompassed basic antigen characterisation and pathogenesis studies with the development of an acellular vaccine containing fimbriae. Research then changed to focus on serogroup B meningococcal disease, studying the vaccine potential of iron-regulated proteins and then Neisseria lactamica. The resurgence of pertussis seen in some countries alerted me to the lack of understanding of protective immune responses to Bordetella pertussis infection and disease and this is now an active area of research.


Assuntos
Meningite Meningocócica/epidemiologia , Meningite Meningocócica/prevenção & controle , Vacinas Meningocócicas/imunologia , Vacina contra Coqueluche/imunologia , Coqueluche/epidemiologia , Coqueluche/prevenção & controle , Bordetella pertussis/genética , Bordetella pertussis/imunologia , Bordetella pertussis/patogenicidade , História do Século XX , História do Século XXI , Humanos , Meningite Meningocócica/imunologia , Neisseria lactamica/genética , Neisseria lactamica/imunologia , Neisseria lactamica/patogenicidade , Coqueluche/imunologia
16.
BMC Genomics ; 11: 652, 2010 Nov 23.
Artigo em Inglês | MEDLINE | ID: mdl-21092259

RESUMO

BACKGROUND: The genus Neisseria contains two important yet very different pathogens, N. meningitidis and N. gonorrhoeae, in addition to non-pathogenic species, of which N. lactamica is the best characterized. Genomic comparisons of these three bacteria will provide insights into the mechanisms and evolution of pathogenesis in this group of organisms, which are applicable to understanding these processes more generally. RESULTS: Non-pathogenic N. lactamica exhibits very similar population structure and levels of diversity to the meningococcus, whilst gonococci are essentially recent descendents of a single clone. All three species share a common core gene set estimated to comprise around 1190 CDSs, corresponding to about 60% of the genome. However, some of the nucleotide sequence diversity within this core genome is particular to each group, indicating that cross-species recombination is rare in this shared core gene set. Other than the meningococcal cps region, which encodes the polysaccharide capsule, relatively few members of the large accessory gene pool are exclusive to one species group, and cross-species recombination within this accessory genome is frequent. CONCLUSION: The three Neisseria species groups represent coherent biological and genetic groupings which appear to be maintained by low rates of inter-species horizontal genetic exchange within the core genome. There is extensive evidence for exchange among positively selected genes and the accessory genome and some evidence of hitch-hiking of housekeeping genes with other loci. It is not possible to define a 'pathogenome' for this group of organisms and the disease causing phenotypes are therefore likely to be complex, polygenic, and different among the various disease-associated phenotypes observed.


Assuntos
Evolução Molecular , Genes Bacterianos/genética , Neisseria lactamica/genética , Neisseria lactamica/isolamento & purificação , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Composição de Bases/genética , Sequência de Bases , Ordem dos Genes/genética , Dados de Sequência Molecular , Neisseria lactamica/patogenicidade , Filogenia , Homologia de Sequência do Ácido Nucleico , Virulência/genética
17.
PLoS One ; 5(7): e11835, 2010 Jul 28.
Artigo em Inglês | MEDLINE | ID: mdl-20676376

RESUMO

Commensal bacteria comprise a large part of the microbial world, playing important roles in human development, health and disease. However, little is known about the genomic content of commensals or how related they are to their pathogenic counterparts. The genus Neisseria, containing both commensal and pathogenic species, provides an excellent opportunity to study these issues. We undertook a comprehensive sequencing and analysis of human commensal and pathogenic Neisseria genomes. Commensals have an extensive repertoire of virulence alleles, a large fraction of which has been exchanged among Neisseria species. Commensals also have the genetic capacity to donate DNA to, and take up DNA from, other Neisseria. Our findings strongly suggest that commensal Neisseria serve as reservoirs of virulence alleles, and that they engage extensively in genetic exchange.


Assuntos
Transferência Genética Horizontal/genética , Genoma Bacteriano/genética , Neisseria/genética , Virulência/genética , Humanos , Neisseria/patogenicidade , Neisseria gonorrhoeae/genética , Neisseria gonorrhoeae/patogenicidade , Neisseria lactamica/genética , Neisseria lactamica/patogenicidade , Neisseria meningitidis/genética , Neisseria meningitidis/patogenicidade
19.
PLoS Comput Biol ; 5(8): e1000455, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19662158

RESUMO

The evolution of bacterial populations has recently become considerably better understood due to large-scale sequencing of population samples. It has become clear that DNA sequences from a multitude of genes, as well as a broad sample coverage of a target population, are needed to obtain a relatively unbiased view of its genetic structure and the patterns of ancestry connected to the strains. However, the traditional statistical methods for evolutionary inference, such as phylogenetic analysis, are associated with several difficulties under such an extensive sampling scenario, in particular when a considerable amount of recombination is anticipated to have taken place. To meet the needs of large-scale analyses of population structure for bacteria, we introduce here several statistical tools for the detection and representation of recombination between populations. Also, we introduce a model-based description of the shape of a population in sequence space, in terms of its molecular variability and affinity towards other populations. Extensive real data from the genus Neisseria are utilized to demonstrate the potential of an approach where these population genetic tools are combined with an phylogenetic analysis. The statistical tools introduced here are freely available in BAPS 5.2 software, which can be downloaded from http://web.abo.fi/fak/mnf/mate/jc/software/baps.html.


Assuntos
Bactérias/genética , Biologia Computacional/métodos , Pool Gênico , Genes Bacterianos , Modelos Genéticos , Algoritmos , Teorema de Bayes , Simulação por Computador , Fluxo Gênico , Neisseria lactamica/genética , Neisseria meningitidis/genética , Análise de Sequência de DNA/métodos
20.
J Infect Dis ; 200(1): 94-8, 2009 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-19476432

RESUMO

The distribution of the hemoglobin receptor gene (hmbR) was investigated among disease and carriage Neisseria meningitidis isolates, revealing that the gene was detected at a significantly higher frequency among disease isolates than among carriage isolates. In isolates without hmbR, the locus was occupied by the cassettes exl2 or exl3 or by a "pseudo hmbR" gene, designated exl4. The hmbR locus exhibited characteristics of a pathogenicity island in published genomes of N. meningitidis, Neisseria gonorrhoeae, and Neisseria lactamica sequence type-640. These data are consistent with a role for the hmbR gene in meningococcal disease.


Assuntos
Proteínas de Bactérias/genética , Infecções Meningocócicas/genética , Neisseria gonorrhoeae/genética , Neisseria meningitidis/genética , Receptores de Superfície Celular/genética , Proteínas de Bactérias/fisiologia , Primers do DNA , Humanos , Infecções Meningocócicas/imunologia , Neisseria lactamica/genética , Neisseria lactamica/patogenicidade , Neisseria meningitidis/patogenicidade , Receptores de Superfície Celular/fisiologia , Virulência/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA