Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Microorganisms ; 9(2)2021 Jan 26.
Artigo em Inglês | MEDLINE | ID: mdl-33530338

RESUMO

The genetic heterogeneity of Heyndrickxia sporothermodurans (formerly Bacillussporothermodurans) was evaluated using whole genome sequencing. The genomes of 29 previously identified Heyndrickxiasporothermodurans and two Heyndrickxia vini strains isolated from ultra-high-temperature (UHT)-treated milk were sequenced by short-read (Illumina) sequencing. After sequence analysis, the two H. vini strains could be reclassified as H. sporothermodurans. In addition, the genomes of the H.sporothermodurans type strain (DSM 10599T) and the closest phylogenetic neighbors Heyndrickxiaoleronia (DSM 9356T) and Heyndrickxia vini (JCM 19841T) were also sequenced using both long (MinION) and short-read (Illumina) sequencing. By hybrid sequence assembly, the genome of the H. sporothermodurans type strain was enlarged by 15% relative to the short-read assembly. This noticeable increase was probably due to numerous mobile elements in the genome that are presumptively related to spore heat tolerance. Phylogenetic studies based on 16S rDNA gene sequence, core genome, single-nucleotide polymorphisms and ANI/dDDH, showed that H. vini is highly related to H. sporothermodurans. When examining the genome sequences of all H.sporothermodurans strains from this study, together with 4 H. sporothermodurans genomes available in the GenBank database, the majority of the 36 strains examined occurred in a clonal lineage with less than 100 SNPs. These data substantiate previous reports on the existence and spread of a genetically highly homogenous and heat resistant spore clone, i.e., the HRS-clone.

2.
Infect Immun ; 84(12): 3484-3495, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27672084

RESUMO

Neisseria meningitidis, the meningococcus, bears the potential to cause life-threatening invasive diseases, but it usually colonizes the nasopharynx without causing any symptoms. Within the nasopharynx, Neisseria meningitidis must face temperature changes depending on the ambient air temperature. Indeed, the nasopharyngeal temperature can be substantially lower than 37°C, the temperature commonly used in experimental settings. Here, we compared the levels of meningococcal biofilm formation, autoaggregation, and cellular adherence at 32°C and 37°C and found a clear increase in all these phenotypes at 32°C suggestive of a stronger in vivo colonization capability at this temperature. A comparative proteome analysis approach revealed differential protein expression levels between 32°C and 37°C, predominantly affecting the bacterial envelope. A total of 375 proteins were detected. Use of database annotation or the PSORTb algorithm predicted 49 of those proteins to be localized in the outer membrane, 21 in either the inner or outer membrane, 35 in the periplasm, 56 in the inner membrane, and 208 in the cytosol; for 6 proteins, no annotation or prediction was available. Temperature-dependent regulation of protein expression was seen particularly in the periplasm as well as in the outer and inner membranes. Neisserial heparin binding antigen (NHBA), NMB1030, and adhesin complex protein (ACP) showed the strongest upregulation at 32°C and were partially responsible for the observed temperature-dependent phenotypes. Screening of different global regulators of Neisseria meningitidis suggested that the extracytoplasmic sigma factor σE might be involved in temperature-dependent biofilm formation. In conclusion, subtle temperature changes trigger adaptation events promoting mucosal colonization by meningococci.


Assuntos
Aderência Bacteriana/fisiologia , Proteínas de Bactérias/metabolismo , Regulação Bacteriana da Expressão Gênica/fisiologia , Neisseria meningitidis/fisiologia , Proteoma/metabolismo , Temperatura , Proteínas de Bactérias/genética , Biofilmes/crescimento & desenvolvimento , Fatores de Virulência/genética , Fatores de Virulência/metabolismo
3.
PLoS One ; 11(5): e0154047, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27167067

RESUMO

Meningococci spread via respiratory droplets, whereas the closely related gonococci are transmitted sexually. Several outbreaks of invasive meningococcal disease have been reported in Europe and the United States among men who have sex with men (MSM). We recently identified an outbreak of serogroup C meningococcal disease among MSM in Germany and France. In this study, genomic and proteomic techniques were used to analyze the outbreak isolates. In addition, genetically identical urethritis isolates were recovered from France and Germany and included in the analysis. Genome sequencing revealed that the isolates from the outbreak among MSM and from urethritis cases belonged to a clade within clonal complex 11. Proteome analysis showed they expressed nitrite reductase, enabling anaerobic growth as previously described for gonococci. Invasive isolates from MSM, but not urethritis isolates, further expressed functional human factor H binding protein associated with enhanced survival in a newly developed transgenic mouse model expressing human factor H, a complement regulatory protein. In conclusion, our data suggest that urethritis and outbreak isolates followed a joint adaptation route including adaption to the urogenital tract.


Assuntos
Surtos de Doenças , Evolução Molecular , Homossexualidade Masculina , Meningite Meningocócica/epidemiologia , Neisseria meningitidis Sorogrupo C/classificação , Neisseria meningitidis/classificação , Nitrito Redutases/genética , Uretrite/epidemiologia , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Fator H do Complemento/antagonistas & inibidores , Fator H do Complemento/genética , Fator H do Complemento/metabolismo , França/epidemiologia , Expressão Gênica , Alemanha/epidemiologia , Interações Hospedeiro-Patógeno , Humanos , Masculino , Meningite Meningocócica/diagnóstico , Meningite Meningocócica/microbiologia , Meningite Meningocócica/patologia , Camundongos , Camundongos Transgênicos , Neisseria meningitidis/genética , Neisseria meningitidis/isolamento & purificação , Neisseria meningitidis/patogenicidade , Neisseria meningitidis Sorogrupo C/genética , Neisseria meningitidis Sorogrupo C/isolamento & purificação , Neisseria meningitidis Sorogrupo C/patogenicidade , Nitrito Redutases/metabolismo , Filogenia , Proteoma/genética , Proteoma/metabolismo , Uretrite/diagnóstico , Uretrite/microbiologia , Uretrite/patologia
4.
J Bacteriol ; 195(19): 4425-35, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23893116

RESUMO

Outer membrane vesicles (OMVs) of Gram-negative bacteria receive increasing attention because of various biological functions and their use as vaccines. However, the mechanisms of OMV release and selective sorting of proteins into OMVs remain unclear. Comprehensive quantitative proteome comparisons between spontaneous OMVs (SOMVs) and the outer membrane (OM) have not been conducted so far. Here, we established a protocol for metabolic labeling of neisserial proteins with (15)N. SOMV and OM proteins labeled with (15)N were used as an internal standard for proteomic comparison of the SOMVs and OMs of two different strains. This labeling approach, coupled with high-sensitivity mass spectrometry, allowed us to comprehensively unravel the proteome of the SOMVs and OMs. We quantified the relative distribution of 155 proteins between SOMVs and the OM. Complement regulatory proteins, autotransporters, proteins involved in iron and zinc acquisition, and a two-partner secretion system were enriched in SOMVs. The highly abundant porins PorA and PorB and proteins connecting the OM with peptidoglycan or the inner membrane, such as RmpM, MtrE, and PilQ, were depleted in SOMVs. Furthermore, the three lytic transglycosylases MltA, MltB, and Slt were less abundant in SOMVs. In conclusion, SOMVs are likely to be released from surface areas with a low local abundance of membrane-anchoring proteins and lytic transglycosylases. The enrichment of complement regulatory proteins, autotransporters, and trace metal binding and transport proteins needs to be explored in the context of the pathogenesis of meningococcal disease.


Assuntos
Proteínas da Membrana Bacteriana Externa/metabolismo , Regulação Bacteriana da Expressão Gênica/fisiologia , Neisseria meningitidis/metabolismo , Proteoma , Proteínas da Membrana Bacteriana Externa/genética , Neisseria meningitidis/genética , Isótopos de Nitrogênio , Porinas , Transporte Proteico
5.
Mol Microbiol ; 89(3): 433-49, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23750848

RESUMO

Neisseria meningitidis (Nm) is a leading cause of septicemia in childhood. Nm septicemia is unique with respect to very quick disease progression, high in vivo bacterial replication rate and its considerable mortality. Nm circumvents major mechanisms of innate immunity such as complement system and phagocytosis. Neutrophil extracellular traps (NETs) are formed from neutrophils during systemic infection and are suggested to contain invading microorganisms. Here, we investigated the interaction of Nm with NETs. Both, meningococci and spontaneously released outer membrane vesicles (SOMVs) were potent NET inducers. NETs were unable to kill NET bound meningococci, but slowed down their proliferation rate. Using Nm as model organism we identified three novel mechanisms how bacteria can evade NET-mediated killing: (i) modification of lipid A of meningococcal LPS with phosphoethanolamine protected Nm from NET-bound cathepsin G; (ii) expression of the high-affinity zinc uptake receptor ZnuD allowed Nm to escape NET-mediated nutritional immunity; (iii) binding of SOMVs to NETs saved Nm from NET binding and the consequent bacteriostatic effect. Escape from NETs may contribute to the most rapid progression of meningococcal disease. The induction of NET formation by Nm in vivo might aggravate thrombosis in vessels ultimately directing to disseminated intravascular coagulation (DIC).


Assuntos
Evasão da Resposta Imune , Neisseria meningitidis/imunologia , Neutrófilos/imunologia , Aderência Bacteriana , Proteínas de Bactérias/metabolismo , Catepsina G/imunologia , Proteínas de Transporte de Cátions/metabolismo , Membrana Celular/metabolismo , Etanolaminas/química , Fímbrias Bacterianas/fisiologia , Técnicas de Inativação de Genes , Granulócitos/imunologia , Granulócitos/microbiologia , Humanos , Imunidade Inata , Lipídeo A/química , Infecções Meningocócicas/imunologia , Microscopia Eletrônica de Transmissão , Neisseria meningitidis/genética , Neisseria meningitidis/ultraestrutura , Neutrófilos/microbiologia , Zinco/metabolismo
6.
Proteomics ; 10(24): 4512-21, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21136603

RESUMO

Neisseria meningitidis is a commensal of the human nasopharynx occasionally causing invasive disease. In vitro biofilms have been employed to model meningococcal carriage. A proteomic analysis of meningococcal biofilms was conducted and metabolic changes related to oxygen and nutrient limitation and upregulation of proteins involved in ROS defense were observed. The upregulated MntC which protects against ROS was shown to be required for meningococcal biofilm formation, but not for planktonic growth. ROS-induced proteomic changes might train the biofilm to cope with immune effectors.


Assuntos
Proteínas de Bactérias/metabolismo , Biofilmes , Neisseria meningitidis/metabolismo , Proteoma/metabolismo , Proteínas de Bactérias/genética , Biofilmes/crescimento & desenvolvimento , Mutação , Neisseria meningitidis/crescimento & desenvolvimento , Espécies Reativas de Oxigênio/metabolismo
7.
Med Microbiol Immunol ; 199(3): 173-83, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20376486

RESUMO

The past decade has seen an increasing interest in biofilm formation by Neisseria meningitidis, a human facultative pathogen causing life-threatening childhood disease commencing from asymptomatic nasopharyngeal colonization. Studying the biology of in vitro biofilm formation improves the understanding of inter-bacterial processes in asymptomatic carriage, of bacterial aggregate formation on host cells, and of meningococcal population biology. This paper reviews publications referring to meningococcal biofilm formation with an emphasis on the role of motility and of extracellular DNA. The theory of sub-dividing the meningococcal population in settler and spreader lineages is discussed, which provides a mechanistic framework for the assumed balance of colonization efficacy and transmission frequency.


Assuntos
Biofilmes/crescimento & desenvolvimento , Neisseria meningitidis/fisiologia , DNA Bacteriano/metabolismo , Fímbrias Bacterianas/fisiologia , Locomoção , Modelos Biológicos , Neisseria meningitidis/crescimento & desenvolvimento
8.
Appl Environ Microbiol ; 76(7): 2271-9, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20139319

RESUMO

Listeria monocytogenes is a food-borne pathogen that is capable of living in harsh environments. It is believed to do this by forming biofilms, which are surface-associated multicellular structures encased in a self-produced matrix. In this paper we show that in L. monocytogenes extracellular DNA (eDNA) may be the only central component of the biofilm matrix and that it is necessary for both initial attachment and early biofilm formation for 41 L. monocytogenes strains that were tested. DNase I treatment resulted in dispersal of biofilms, not only in microtiter tray assays but also in flow cell biofilm assays. However, it was also demonstrated that in a culture without eDNA, neither Listeria genomic DNA nor salmon sperm DNA by itself could restore the capacity to adhere. A search for additional necessary components revealed that peptidoglycan (PG), specifically N-acetylglucosamine (NAG), interacted with the DNA in a manner which restored adhesion. If a short DNA fragment (less than approximately 500 bp long) was added to an eDNA-free culture prior to addition of genomic or salmon sperm DNA, adhesion was prevented, indicating that high-molecular-weight DNA is required for adhesion and that the number of attachment sites on the cell surface can be saturated.


Assuntos
Biofilmes/crescimento & desenvolvimento , DNA Bacteriano/metabolismo , Listeria monocytogenes/fisiologia , Acetilglucosamina/metabolismo , Animais , Aderência Bacteriana , DNA/metabolismo , Desoxirribonuclease I/metabolismo , Matriz Extracelular/metabolismo , Listeria monocytogenes/metabolismo , Peptidoglicano/metabolismo , Salmão
9.
Mol Microbiol ; 75(6): 1355-71, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20180907

RESUMO

Major pathogenic clonal complexes (cc) of Neisseria meningitidis differ substantially in their point prevalence among healthy carriers. We show that frequently carried pathogenic cc (e.g. sequence type ST-41/44 cc and ST-32 cc) depend on extracellular DNA (eDNA) to initiate in vitro biofilm formation, whereas biofilm formation of cc with low point prevalence (ST-8 cc and ST-11 cc) was eDNA-independent. For initial biofilm formation, a ST-32 cc type strain, but not a ST-11 type strain, utilized eDNA. The release of eDNA was mediated by lytic transglycosylase and cytoplasmic N-acetylmuramyl-L-alanine amidase genes. In late biofilms, outer membrane phospholipase A-dependent autolysis, which was observed in most cc, but not in ST-8 and ST-11 strains, was required for shear force resistance of microcolonies. Taken together, N. meningitidis evolved two different biofilm formation strategies, an eDNA-dependent one yielding shear force resistant microcolonies, and an eDNA-independent one. Based on the experimental findings and previous epidemiological observations, we hypothesize that most meningococcal cc display a settler phenotype, which is eDNA-dependent and results in a stable interaction with the host. On the contrary, spreaders (ST-11 and ST-8 cc) are unable to use eDNA for biofilm formation and might compensate for poor colonization properties by high transmission rates.


Assuntos
Biofilmes/crescimento & desenvolvimento , DNA Bacteriano/metabolismo , Neisseria meningitidis/fisiologia , Proteínas de Bactérias/metabolismo , Bacteriólise , Glicosiltransferases/metabolismo , N-Acetil-Muramil-L-Alanina Amidase/metabolismo , Neisseria meningitidis/citologia , Neisseria meningitidis/crescimento & desenvolvimento , Neisseria meningitidis/metabolismo , Fosfolipases/metabolismo
10.
Environ Microbiol ; 11(12): 3154-65, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19671077

RESUMO

Escherichia coli K1 causes disease in humans and birds. Its polysialic acid capsule can be O-acetylated via phase-variable expression of the acetyltransferase NeuO encoded by prophage CUS-3. The role of capsule O-acetylation in ecological adaptation or pathogenic invasion of E. coli K1 is largely unclear. A population genetics approach was performed to study the distribution of neuO among E. coli K1 isolates from human and avian sources. Multilocus sequence typing revealed 39 different sequence types (STs) among 183 E. coli K1 strains. The proportion of the ST95 complex (STC95) was 44%. NeuO was found in 98% of the STC95 strains, but only in 24% of other STs. Grouping of STs and prophage genotypes revealed a segregation of prophage types according to STs, suggesting coevolution of CUS-3 and the E. coli K1 host. Within the STC95, which is known to harbour both human and avian pathogenic isolates, CUS-3 genotypes were shared irrespective of the host species. Functional analysis of a variety of strain pairs revealed that NeuO-mediated K1 capsule O-acetylation enhanced desiccation resistance. In contrast, NeuO expression led to a reduced biofilm formation in biofilm positive E. coli K1 isolates. These findings suggest a delicate ecological balance of neuO'on'/'off' switching.


Assuntos
Acetiltransferases/genética , Adaptação Fisiológica/genética , Cápsulas Bacterianas/metabolismo , Proteínas de Escherichia coli/genética , Escherichia coli/genética , Genes Bacterianos , Acetiltransferases/classificação , Acetiltransferases/metabolismo , Animais , Sequência de Bases , Escherichia coli/classificação , Escherichia coli/isolamento & purificação , Proteínas de Escherichia coli/classificação , Proteínas de Escherichia coli/metabolismo , Fezes/microbiologia , Regulação Bacteriana da Expressão Gênica , Humanos , Dados de Sequência Molecular , Filogenia , Prófagos/genética , Análise de Sequência de DNA
11.
Mol Microbiol ; 62(5): 1292-309, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17121595

RESUMO

We show that in a standardized in vitro flow system unencapsulated variants of genetically diverse lineages of Neisseria meningitidis formed biofilms, that could be maintained for more than 96 h. Biofilm cells were resistant to penicillin, but not to rifampin or ciprofloxacin. For some strains, microcolony formation within biofilms was observed. Microcolony formation in strain MC58 depended on a functional copy of the pilE gene encoding the pilus subunit pilin, and was associated with twitching of cells. Nevertheless, unpiliated pilE mutants formed biofilms showing that attachment and accumulation of cells did not depend on pilus expression. Mutation and complementation analysis revealed that the type IV pilus-associated protein PilX, which was recently shown to mediate interbacterial aggregation, indirectly supported microcolony formation by contributing to pilus expression. A large number of PilX alleles was identified among genetically diverse meningococcal strains. PilX alleles differed in their propensity to support autoaggregation of cells in suspension, but not in their ability to support microcolony formation within biofilms in the continuous flow system.


Assuntos
Biofilmes/crescimento & desenvolvimento , Proteínas de Fímbrias/fisiologia , Neisseria meningitidis/fisiologia , Cápsulas Bacterianas , Regulação Bacteriana da Expressão Gênica , Testes de Sensibilidade Microbiana , Neisseria meningitidis/genética , Fenótipo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...