Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
1.
Antimicrob Agents Chemother ; 68(10): e0079424, 2024 Oct 08.
Artigo em Inglês | MEDLINE | ID: mdl-39150247

RESUMO

Each year, Neisseria gonorrhoeae (Ngo) causes over 1.5 million new infections in the United States, and >87 million worldwide. The absence of a vaccine for preventing gonorrhea, the rapid emergence of multidrug-resistant and extremely drug-resistant Ngo strains, and the limited number of antibiotics available for treating gonorrhea underscore the importance of developing new modalities for addressing Ngo infection. Here, we describe DNA-based microbicides that kill Ngo but not commensals. Previously, we showed that Ngo is killed when it takes up differentially methylated DNA with homology to its genome. We exploited this Achilles heel to develop a new class of microbicides for preventing Ngo infection. These microbicides consist of DNA molecules with specific sequences and a methylation pattern different from Ngo DNA. These DNAs kill low-passage and antibiotic-resistant clinical isolates with high efficiency but leave commensals unharmed. Equally important, the DNAs are equally effective against Ngo whether they are in buffered media or personal lubricants. These findings illustrate the potential of this new class of practical, low-cost, self-administered DNA-based microbicides for preventing Ngo transmission during sexual intercourse.


Assuntos
Gonorreia , Neisseria gonorrhoeae , Neisseria gonorrhoeae/efeitos dos fármacos , Neisseria gonorrhoeae/genética , Gonorreia/prevenção & controle , Gonorreia/microbiologia , Gonorreia/tratamento farmacológico , Humanos , Antibacterianos/farmacologia , Feminino , Animais , Testes de Sensibilidade Microbiana , Metilação de DNA/efeitos dos fármacos , DNA Bacteriano/genética
2.
PLoS Pathog ; 18(5): e1010497, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35580146

RESUMO

The mechanisms used by human adapted commensal Neisseria to shape and maintain a niche in their host are poorly defined. These organisms are common members of the mucosal microbiota and share many putative host interaction factors with Neisseria meningitidis and Neisseria gonorrhoeae. Evaluating the role of these shared factors during host carriage may provide insight into bacterial mechanisms driving both commensalism and asymptomatic infection across the genus. We identified host interaction factors required for niche development and maintenance through in vivo screening of a transposon mutant library of Neisseria musculi, a commensal of wild-caught mice which persistently and asymptomatically colonizes the oral cavity and gut of CAST/EiJ and A/J mice. Approximately 500 candidate genes involved in long-term host interaction were identified. These included homologs of putative N. meningitidis and N. gonorrhoeae virulence factors which have been shown to modulate host interactions in vitro. Importantly, many candidate genes have no assigned function, illustrating how much remains to be learned about Neisseria persistence. Many genes of unknown function are conserved in human adapted Neisseria species; they are likely to provide a gateway for understanding the mechanisms allowing pathogenic and commensal Neisseria to establish and maintain a niche in their natural hosts. Validation of a subset of candidate genes confirmed a role for a polysaccharide capsule in N. musculi persistence but not colonization. Our findings highlight the potential utility of the Neisseria musculi-mouse model as a tool for studying the pathogenic Neisseria; our work represents a first step towards the identification of novel host interaction factors conserved across the genus.


Assuntos
Elementos de DNA Transponíveis , Interações entre Hospedeiro e Microrganismos , Neisseria , Animais , Portador Sadio/microbiologia , Portador Sadio/fisiopatologia , Elementos de DNA Transponíveis/genética , Biblioteca Gênica , Interações entre Hospedeiro e Microrganismos/genética , Interações entre Hospedeiro e Microrganismos/fisiologia , Camundongos , Microbiota/genética , Mucosa/microbiologia , Neisseria/genética , Neisseria/patogenicidade , Neisseria gonorrhoeae/genética , Neisseria gonorrhoeae/patogenicidade , Neisseria meningitidis/genética , Neisseria meningitidis/patogenicidade , Simbiose/genética , Simbiose/fisiologia , Fatores de Virulência/genética
3.
PLoS Pathog ; 15(2): e1007495, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30753248

RESUMO

The Gram-negative human pathogen N. gonorrhoeae (Ngo) quickly attaches to epithelial cells, and large numbers of the bacteria remain on the cell surface for prolonged periods. Ngo invades cells but few viable intracellular bacteria are recovered until later stages of infection, leading to the assumption that Ngo is a weak invader. On the cell surface, Ngo quickly recruits CD46-cyt1 to the epithelial cell cortex directly beneath the bacteria and causes its cleavage by metalloproteinases and Presenilin/γSecretease; how these interactions affect the Ngo lifecycle is unknown. Here, we show Ngo induces an autophagic response in the epithelial cell through CD46-cyt1/GOPC, and this response kills early invaders. Throughout infection, the pathogen slowly downregulates CD46-cyt1 and remodeling of lysosomes, another key autophagy component, and these activities ultimately promote intracellular survival. We present a model on the dynamics of Ngo infection and describe how this dual interference with the autophagic pathway allows late invaders to survive within the cell.


Assuntos
Gonorreia/metabolismo , Proteína Cofatora de Membrana/fisiologia , Neisseria gonorrhoeae/patogenicidade , Autofagia/fisiologia , Aderência Bacteriana , Linhagem Celular , Colo do Útero , Regulação para Baixo , Células Epiteliais , Feminino , Fímbrias Bacterianas , Gonorreia/fisiopatologia , Humanos , Lisossomos , Proteína Cofatora de Membrana/imunologia , Glicoproteínas de Membrana , Proteínas de Membrana/metabolismo , Neisseria gonorrhoeae/metabolismo , Cultura Primária de Células , Isoformas de Proteínas
4.
Mol Microbiol ; 110(5): 677-688, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-29719082

RESUMO

Post-translational acetylation is a common protein modification in bacteria. It was recently reported that Neisseria gonorrhoeae acetylates the Type IV pilus retraction motor, PilT. Here, we show recombinant PilT can be acetylated in vitro and acetylation does not affect PilT ultrastructure. To investigate the function of PilT acetylation, we mutated an acetylated lysine, K117, to mimic its acetylated or unacetylated forms. These mutations were not tolerated by wild-type N. gonorrhoeae, but they were tolerated by N. gonorrhoeae carrying an inducible pilE when grown without inducer. We identified additional mutations in pilT and pilU that suppress the lethality of K117 mutations. To investigate the link between PilE and PilT acetylation, we found the lack of PilE decreases PilT acetylation levels and increases the amount of PilT associated with the inner membrane. Finally, we found no difference between wild-type and mutant cells in transformation efficiency, suggesting neither mutation inhibits Type IV pilus retraction. Mutant cells, however, form microcolonies morphologically distinct from wt cells. We conclude that interfering with the acetylation status of PilTK117 greatly reduces N. gonorrhoeae viability, and mutations in pilT, pilU and pilE can overcome this lethality. We discuss the implications of these findings in the context of Type IV pilus retraction regulation.


Assuntos
Proteínas de Fímbrias , Proteínas Motores Moleculares , Neisseria gonorrhoeae/genética , Neisseria gonorrhoeae/metabolismo , Acetilação , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Proteínas de Fímbrias/genética , Proteínas de Fímbrias/metabolismo , Fímbrias Bacterianas/metabolismo , Proteínas Motores Moleculares/genética , Proteínas Motores Moleculares/metabolismo , Mutação , Processamento de Proteína Pós-Traducional
5.
Infect Immun ; 86(5)2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29440372

RESUMO

Commensals are important for the proper functioning of multicellular organisms. How a commensal establishes persistent colonization of its host is little understood. Studies of this aspect of microbe-host interactions are impeded by the absence of an animal model. We have developed a natural small animal model for identifying host and commensal determinants of colonization and of the elusive process of persistence. Our system couples a commensal bacterium of wild mice, Neisseria musculi, with the laboratory mouse. The pairing of a mouse commensal with its natural host circumvents issues of host restriction. Studies are performed in the absence of antibiotics, hormones, invasive procedures, or genetic manipulation of the host. A single dose of N. musculi, administered orally, leads to long-term colonization of the oral cavity and gut. All mice are healthy. Susceptibility to colonization is determined by host genetics and innate immunity. For N. musculi, colonization requires the type IV pilus. Reagents and powerful tools are readily available for manipulating the laboratory mouse, allowing easy dissection of host determinants controlling colonization resistance. N. musculi is genetically related to human-dwelling commensal and pathogenic Neisseria and encodes host interaction factors and vaccine antigens of pathogenic Neisseria Our system provides a natural approach for studying Neisseria-host interactions and is potentially useful for vaccine efficacy studies.


Assuntos
Infecções por Bactérias Gram-Negativas/imunologia , Infecções por Bactérias Gram-Negativas/transmissão , Interações Hospedeiro-Patógeno , Imunidade Inata , Camundongos/microbiologia , Neisseria/patogenicidade , Simbiose , Animais , Modelos Animais de Doenças
6.
Infect Immun ; 85(3)2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28052997

RESUMO

Colonization of the endometrium by pathogenic bacteria ascending from the lower female reproductive tract (FRT) is associated with many gynecologic and obstetric health complications. To study these host-microbe interactions in vitro, we developed a human three-dimensional (3-D) endometrial epithelial cell (EEC) model using the HEC-1A cell line and the rotating wall vessel (RWV) bioreactor technology. Our model, composed of 3-D EEC aggregates, recapitulates several functional/structural characteristics of human endometrial epithelial tissue, including cell differentiation, the presence of junctional complexes/desmosomes and microvilli, and the production of membrane-associated mucins and Toll-like receptors (TLRs). TLR function was evaluated by exposing the EEC aggregates to viral and bacterial products. Treatment with poly(I·C) and flagellin but not with synthetic lipoprotein (fibroblast-stimulating lipoprotein 1 [FSL-1]) or lipopolysaccharide (LPS) significantly induced proinflammatory mediators in a dose-dependent manner. To simulate ascending infection, we infected EEC aggregates with commensal and pathogenic bacteria: Lactobacillus crispatus, Gardnerella vaginalis, and Neisseria gonorrhoeae All vaginal microbiota and N. gonorrhoeae efficiently colonized the 3-D surface, localizing to crevices of the EEC model and interacting with multiple adjacent cells simultaneously. However, only infection with pathogenic N. gonorrhoeae and not infection with the other bacteria tested significantly induced proinflammatory mediators and significant ultrastructural changes to the host cells. The latter observation is consistent with clinical findings and illustrated the functional specificity of our system. Additionally, we highlighted the utility of the 3-D EEC model for the study of the pathogenesis of N. gonorrhoeae using a well-characterized ΔpilT mutant. Overall, this study demonstrates that the human 3-D EEC model is a robust tool for studying host-microbe interactions and bacterial pathogenesis in the upper FRT.


Assuntos
Gonorreia/microbiologia , Interações Hospedeiro-Patógeno , Mucosa/microbiologia , Neisseria gonorrhoeae/fisiologia , Vagina/microbiologia , Técnicas de Cultura de Células , Linhagem Celular , Quimiocinas/metabolismo , Citocinas/metabolismo , Feminino , Humanos , Técnicas In Vitro , Mucinas/metabolismo , Mucosa/ultraestrutura , Mutação
7.
Int J Syst Evol Microbiol ; 66(9): 3585-3593, 2016 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-27298306

RESUMO

Members of the genus Neisseria have been isolated from or detected in a wide range of animals, from non-human primates and felids to a rodent, the guinea pig. By means of selective culture, biochemical testing, Gram staining and PCR screening for the Neisseria-specific internal transcribed spacer region of the rRNA operon, we isolated four strains of the genus Neisseria from the oral cavity of the wild house mouse, Mus musculus subsp. domesticus. The isolates are highly related and form a separate clade in the genus, as judged by tree analyses using either multi-locus sequence typing of ribosomal genes or core genes. One isolate, provisionally named Neisseria musculi sp. nov. (type strain AP2031T=DSM 101846T=CCUG 68283T=LMG 29261T), was studied further. Strain AP2031T/N. musculi grew well in vitro. It was naturally competent, taking up DNA in a DNA uptake sequence and pilT-dependent manner, and was amenable to genetic manipulation. These and other genomic attributes of N. musculi sp. nov. make it an ideal candidate for use in developing a mouse model for studying Neisseria-host interactions.


Assuntos
Camundongos/microbiologia , Neisseria/classificação , Filogenia , Animais , Técnicas de Tipagem Bacteriana , Composição de Bases , DNA Bacteriano/genética , Genes Bacterianos , Boca/microbiologia , Tipagem de Sequências Multilocus , Neisseria/genética , Neisseria/isolamento & purificação , América do Norte , Hibridização de Ácido Nucleico , RNA Ribossômico 16S/genética , Análise de Sequência de DNA
8.
Proc Natl Acad Sci U S A ; 110(8): 3059-64, 2013 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-23382234

RESUMO

The strict tropism of many pathogens for man hampers the development of animal models that recapitulate important microbe-host interactions. We developed a rhesus macaque model for studying Neisseria-host interactions using Neisseria species indigenous to the animal. We report that Neisseria are common inhabitants of the rhesus macaque. Neisseria isolated from the rhesus macaque recolonize animals after laboratory passage, persist in the animals for at least 72 d, and are transmitted between animals. Neisseria are naturally competent and acquire genetic markers from each other in vivo, in the absence of selection, within 44 d after colonization. Neisseria macacae encodes orthologs of known or presumed virulence factors of human-adapted Neisseria, as well as current or candidate vaccine antigens. We conclude that the rhesus macaque model will allow studies of the molecular mechanisms of Neisseria colonization, transmission, persistence, and horizontal gene transfer. The model can potentially be developed further for preclinical testing of vaccine candidates.


Assuntos
Transferência Genética Horizontal , Infecções por Bactérias Gram-Negativas/microbiologia , Neisseria/patogenicidade , Animais , Marcadores Genéticos , Infecções por Bactérias Gram-Negativas/genética , Infecções por Bactérias Gram-Negativas/transmissão , Interações Hospedeiro-Patógeno , Macaca mulatta , Dados de Sequência Molecular , Neisseria/classificação , Neisseria/genética , Filogenia , Virulência
9.
Mol Microbiol ; 90(1): 103-13, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23899162

RESUMO

Human-adapted Neisseria includes two pathogens, Neisseria gonorrhoeae and Neisseria meningitidis, and at least 13 species of commensals that colonize many of the same niches as the pathogens. The Type IV pilus plays an important role in the biology of pathogenic Neisseria. In these species, Sigma factor RpoD (σ(70)), Integration Host Factor, and repressors RegF and CrgA regulate transcription of pilE, the gene encoding the pilus structural subunit. The Type IV pilus is also a strictly conserved trait in commensal Neisseria. We present evidence that a different mechanism regulates pilE transcription in commensals. Using Neisseria elongata as a model, we show that Sigma factor RpoN (σ(54)), Integration Host Factor, and an activator we name Npa regulate pilE transcription. Taken in context with previous reports, our findings indicate pilE regulation switched from an RpoN- to an RpoD-dependent mechanism as pathogenic Neisseria diverged from commensals during evolution. Our findings have implications for the timing of Tfp expression and Tfp-mediated host cell interactions in these two groups of bacteria.


Assuntos
Proteínas de Fímbrias/biossíntese , Regulação Bacteriana da Expressão Gênica , Neisseria elongata/genética , RNA Polimerase Sigma 54/metabolismo , Fatores Hospedeiros de Integração/metabolismo , Fatores de Transcrição/metabolismo , Transcrição Gênica
10.
Cell Microbiol ; 15(11): 1837-50, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23648135

RESUMO

Neisseria gonorrhoeae regulates the expression of epithelial cell genes, activates cytoprotective pathways in the infected cell and protects it from apoptosis. Many of these responses are enhanced by the Type IV pilus (Tfp). We tested the hypothesis that N. gonorrhoeae modulates the innate immune response by inducing expression of ATF3, a transcription factor that negatively regulates the expression of many cytokine genes. We further determined whether Tfp are involved in these events. We found that N. gonorrhoeae induces ATF3 expression in mucosal epithelial cells through activation of mitogen-activated protein kinases. Maximal ATF3 expression requires Tfp retraction. Knocking down endogenous levels of ATF3 results in higher levels of IL-6 transcript. Our findings strongly suggest that ATF3 is involved in suppressing cytokine expression during gonococcal infection. We propose a model for the role of ATF3 in the context of N. gonorrhoeae infection.


Assuntos
Fator 3 Ativador da Transcrição/metabolismo , Interações Hospedeiro-Patógeno , Evasão da Resposta Imune , Interleucina-6/antagonistas & inibidores , Neisseria gonorrhoeae/fisiologia , Linhagem Celular Tumoral , Células Epiteliais/imunologia , Células Epiteliais/microbiologia , Regulação da Expressão Gênica , Humanos
11.
mBio ; 15(1): e0279223, 2024 Jan 16.
Artigo em Inglês | MEDLINE | ID: mdl-38084997

RESUMO

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.


Assuntos
Proteínas de Fímbrias , Fímbrias Bacterianas , Camundongos , Animais , Humanos , Neisseria/genética , Simbiose , Neisseria gonorrhoeae , Proteínas de Bactérias
12.
Proc Natl Acad Sci U S A ; 107(25): 11358-63, 2010 Jun 22.
Artigo em Inglês | MEDLINE | ID: mdl-20534431

RESUMO

Through evolution, nature has produced exquisite nanometric structures, with features unrealized in the most advanced man-made devices. Type IV pili (Tfp) represent such a structure: 6-nm-wide retractable filamentous appendages found in many bacteria, including human pathogens. Whereas the structure of Neisseria gonorrhoeae Tfp has been defined by conventional structural techniques, it remains difficult to explain the wide spectrum of functions associated with Tfp. Here we uncover a previously undescribed force-induced quaternary structure of the N. gonorrhoeae Tfp. By using a combination of optical and magnetic tweezers, atomic force microscopy, and molecular combing to apply forces on purified Tfp, we demonstrate that Tfp subjected to approximately 100 pN of force will transition into a new conformation. The new structure is roughly 3 times longer and 40% narrower than the original structure. Upon release of the force, the Tfp fiber regains its original form, indicating a reversible transition. Equally important, we show that the force-induced conformation exposes hidden epitopes previously buried in the Tfp fiber. We postulate that this transition provides a means for N. gonorrhoeae to maintain attachment to its host while withstanding intermittent forces encountered in the environment. Our findings demonstrate the need to reassess our understanding of Tfp dynamics and functions. They could also explain the structural diversity of other helical polymers while presenting a unique mechanism for polymer elongation and exemplifying the extreme structural plasticity of biological polymers.


Assuntos
Proteínas de Fímbrias/metabolismo , Fímbrias Bacterianas/química , Fímbrias Bacterianas/metabolismo , Neisseria gonorrhoeae/metabolismo , Conformação Proteica , Bactérias/metabolismo , Biomimética , Epitopos , Magnetismo , Microscopia de Força Atômica/métodos , Modelos Biológicos , Pinças Ópticas , Polímeros/química , Estresse Mecânico
13.
J Immunol ; 184(2): 694-701, 2010 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-20018629

RESUMO

CD46 is a type I transmembrane protein with complement and T cell regulatory functions in human cells. CD46 has signaling and receptor properties in immune and nonimmune cells, many of which are dependent on the expression of cytoplasmic tail (cyt) isoforms cyt1 or cyt2. Little is known about how cyt1 and cyt2 mediate cellular responses. We show that CD46-cyt1 and CD46-cyt2 are substrates for presenilin/gamma-secretase (PS/gammaS), an endogenous protease complex that regulates many important signaling proteins through proteolytic processing. PS/gammaS processing of CD46 releases immunoprecipitable cyt1 and cyt2 tail peptides into the cell, is blocked by chemical inhibitors, and is prevented in dominant negative presenilin mutant cell lines. Two human pathogens, Neisseria gonorrhoeae and Neisseria meningitidis, stimulate PS/gammaS processing of CD46-cyt1 and CD46-cyt2. This stimulation requires type IV pili and PilT, the type IV pilus retraction motor, implying that mechanotransduction plays a role in this event. We present a model for PS/gammaS processing of CD46 that provides a mechanism by which signals are transduced via the cyt1 and cyt2 tails to regulate CD46-dependent cellular responses. Our findings have broad implications for understanding the full range of CD46 functions in infection and noninfection situations.


Assuntos
Secretases da Proteína Precursora do Amiloide/metabolismo , Gonorreia/metabolismo , Proteína Cofatora de Membrana/metabolismo , Infecções Meningocócicas/metabolismo , Presenilinas/metabolismo , Fímbrias Bacterianas , Humanos , Mecanotransdução Celular , Proteína Cofatora de Membrana/fisiologia , Neisseria gonorrhoeae , Neisseria meningitidis , Isoformas de Proteínas , Transdução de Sinais
14.
PLoS Biol ; 6(4): e87, 2008 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-18416602

RESUMO

The causative agent of gonorrhea, Neisseria gonorrhoeae, bears retractable filamentous appendages called type IV pili (Tfp). Tfp are used by many pathogenic and nonpathogenic bacteria to carry out a number of vital functions, including DNA uptake, twitching motility (crawling over surfaces), and attachment to host cells. In N. gonorrhoeae, Tfp binding to epithelial cells and the mechanical forces associated with this binding stimulate signaling cascades and gene expression that enhance infection. Retraction of a single Tfp filament generates forces of 50-100 piconewtons, but nothing is known, thus far, on the retraction force ability of multiple Tfp filaments, even though each bacterium expresses multiple Tfp and multiple bacteria interact during infection. We designed a micropillar assay system to measure Tfp retraction forces. This system consists of an array of force sensors made of elastic pillars that allow quantification of retraction forces from adherent N. gonorrhoeae bacteria. Electron microscopy and fluorescence microscopy were used in combination with this novel assay to assess the structures of Tfp. We show that Tfp can form bundles, which contain up to 8-10 Tfp filaments, that act as coordinated retractable units with forces up to 10 times greater than single filament retraction forces. Furthermore, single filament retraction forces are transient, whereas bundled filaments produce retraction forces that can be sustained. Alterations of noncovalent protein-protein interactions between Tfp can inhibit both bundle formation and high-amplitude retraction forces. Retraction forces build over time through the recruitment and bundling of multiple Tfp that pull cooperatively to generate forces in the nanonewton range. We propose that Tfp retraction can be synchronized through bundling, that Tfp bundle retraction can generate forces in the nanonewton range in vivo, and that such high forces could affect infection.


Assuntos
Fímbrias Bacterianas/fisiologia , Neisseria gonorrhoeae/patogenicidade , Aderência Bacteriana/fisiologia , Fímbrias Bacterianas/ultraestrutura , Microscopia Eletrônica
15.
Microbiology (Reading) ; 155(Pt 12): 4084-4092, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19762436

RESUMO

Early in infection, Neisseria gonorrhoeae can be observed to attach to the epithelial cell surface as microcolonies and induce dramatic changes to the host cell cortex. We tested the hypothesis that type IV pili (Tfp) retraction plays a role in the ultrastructure of both the host cell cortex and the bacterial microcolony. Using serial ultrathin sectioning, transmission electron microscopy and 3D reconstruction of serial 2D images, we have obtained what we believe to be the first 3D reconstructions of the N. gonorrhoeae-host cell interface, and determined the architecture of infected cell microvilli as well as the attached microcolony. Tfp connect both wild-type (wt) and Tfp retraction-deficient bacteria with each other, and with the host cell membrane. Tfp fibres and microvilli form a lattice in the wt microcolony and at its periphery. Wt microcolonies induce microvilli formation and increases of surface area, leading to an approximately ninefold increase in the surface area of the host cell membrane at the site of attachment. In contrast, Tfp retraction-deficient microcolonies do not affect these parameters. Wt microcolonies had a symmetrical, dome-shaped structure with a circular 'footprint', while Tfp retraction-deficient microcolonies were notably less symmetrical. These findings support a major role for Tfp retraction in microvilli and microcolony architecture. They are consistent with the biophysical attributes of Tfp and the effects of Tfp retraction on epithelial cell signalling.


Assuntos
Fímbrias Bacterianas/ultraestrutura , Neisseria gonorrhoeae/patogenicidade , Neisseria gonorrhoeae/ultraestrutura , Aderência Bacteriana/fisiologia , Linhagem Celular , Fímbrias Bacterianas/fisiologia , Humanos , Imageamento Tridimensional , Microscopia Eletrônica de Varredura , Microscopia Eletrônica de Transmissão , Neisseria gonorrhoeae/fisiologia , Virulência/fisiologia
16.
J Cell Biol ; 156(6): 951-7, 2002 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-11901164

RESUMO

The Neisseria type IV pilus promotes bacterial adhesion to host cells. The pilus binds CD46, a complement-regulatory glycoprotein present on nucleated human cells (Källström et al., 1997). CD46 mutants with truncated cytoplasmic tails fail to support bacterial adhesion (Källström et al., 2001), suggesting that this region of the molecule also plays an important role in infection. Here, we report that infection of human epithelial cells by piliated Neisseria gonorrhoeae (GC) leads to rapid tyrosine phosphorylation of CD46. Studies with wild-type and mutant tail fusion constructs demonstrate that Src kinase phosphorylates tyrosine 354 in the Cyt2 isoform of the CD46 cytoplasmic tail. Consistent with these findings, infection studies show that PP2, a specific Src family kinase inhibitor, but not PP3, an inactive variant of this drug, reduces the ability of epithelial cells to support bacterial adhesion. Several lines of evidence point to the role of c-Yes, a member of the Src family of nonreceptor tyrosine kinases, in CD46 phosphorylation. GC infection causes c-Yes to aggregate in the host cell cortex beneath adherent bacteria, increases binding of c-Yes to CD46, and stimulates c-Yes kinase activity. Finally, c-Yes immunoprecipitated from epithelial cells is able to phosphorylate the wild-type Cyt2 tail but not the mutant derivative in which tyrosine 354 has been substituted with alanine. We conclude that GC infection leads to rapid tyrosine phosphorylation of the CD46 Cyt2 tail and that the Src kinase c-Yes is involved in this reaction. Together, the findings reported here and elsewhere strongly suggest that pilus binding to CD46 is not a simple static process. Rather, they support a model in which pilus interaction with CD46 promotes signaling cascades important for Neisseria infectivity.


Assuntos
Antígenos CD/metabolismo , Adesão Celular/genética , Células Epiteliais/metabolismo , Fímbrias Bacterianas/metabolismo , Glicoproteínas de Membrana/metabolismo , Neisseria gonorrhoeae/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Tirosina/metabolismo , Antígenos CD/genética , Adesão Celular/efeitos dos fármacos , Células Cultivadas , Células Epiteliais/microbiologia , Fímbrias Bacterianas/ultraestrutura , Imunofluorescência , Humanos , Proteína Cofatora de Membrana , Glicoproteínas de Membrana/genética , Neisseria gonorrhoeae/citologia , Infecções por Neisseriaceae/genética , Infecções por Neisseriaceae/metabolismo , Estrutura Terciária de Proteína/efeitos dos fármacos , Estrutura Terciária de Proteína/genética , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-yes , Quinases da Família src/antagonistas & inibidores , Quinases da Família src/metabolismo
17.
Microb Cell ; 6(12): 544-546, 2019 Oct 19.
Artigo em Inglês | MEDLINE | ID: mdl-31832426

RESUMO

It is now abundantly clear that our microbiota (commensals) are critical for many physiological and developmental processes. They have also been shown to inhibit pathogen colonization, through a variety of means including nutrient competition and secretion of microbicidal or biofilm-inhibiting proteins/peptides. Our recent study, Kim et al., (2019), adds a new dimension to the concept of commensal protection. It shows that commensal Neisseria kill the closely related pathogen N. gonorrhoeae through an unexpected mechanism, one that involves genetic competence, DNA methylation state and recombination. This microreview summarizes the report and discusses questions and lines of research arising from the study. Further investigation into this DNA-based killing mechanism will provide a better understanding of Neisseria biology and commensal-pathogen interactions on the mucosa, and identify strategies for preventing pathogenic Neisseria transmission.

18.
Methods Mol Biol ; 1997: 403-412, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31119636

RESUMO

We have developed a natural mouse model to study persistent colonization by commensal Neisseria. The system couples the ordinary lab mouse with Neisseria musculi (Nmus), a commensal in the oral cavity and gut of the wild mouse, Mus musculus. The pairing of Nmus with its natural reservoir circumvents host restriction barriers that have impeded previous studies of Neisseria in vivo behavior. The model allows, for the first time, for the dissection of host and neisserial determinants of asymptomatic colonization. Inoculation procedures are noninvasive and susceptibility to Nmus colonization varies with host genetic background. In colonized mice, bacterial burdens are detectable up to 1-year post inoculation, making it an ideal model for the study of persistence. As Nmus encodes several Neisseria gonorrhoeae (and Neisseria meningitidis) host interaction factors, the system can be used to query the in vivo functions of these commonly held genes and factors. Nmus also encodes many pathogenic Neisseria vaccine targets including a polysaccharide capsule, making the model potentially useful for vaccine development. The ease of genetic manipulation of Nmus enhances the feasibility of such studies.


Assuntos
Modelos Animais de Doenças , Gonorreia/microbiologia , Neisseria/patogenicidade , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Microbioma Gastrointestinal/imunologia , Gonorreia/terapia , Humanos , Camundongos/microbiologia , Mucosa Bucal/imunologia , Mucosa Bucal/microbiologia , Neisseria/genética , Neisseria/imunologia , Simbiose/imunologia , Fatores de Virulência/genética , Fatores de Virulência/metabolismo
19.
Microbiologyopen ; 8(5): e00713, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30079633

RESUMO

Over 20 genes are involved in the biogenesis and function of the Neisseria Type IV pilus (Tfp). In the pathogenic species, RpoD and the integration host factor (IHF) protein regulate expression of pilE, encoding the Tfp structural subunit. We previously reported that in commensal species, pilE transcription is regulated by RpoN, IHF, and activator Npa. Npa has many hallmarks of response regulators in two-component regulatory systems, leading us to search for its response regulator partner. We report that Npa partners with sensor kinase Nps to control pilE transcription. Among the genes involved in Tfp biogenesis and function, only pilE is controlled by RpoN and Npa/Nps. We summarize our findings in a model, and discuss the implications of the differential regulation of pilE the context of Neisseria Tfp biogenesis.


Assuntos
Proteínas de Fímbrias/biossíntese , Regulação Bacteriana da Expressão Gênica , Histidina Quinase/metabolismo , Neisseria/genética , Transdução de Sinais , Fatores de Transcrição/metabolismo , Transcrição Gênica , RNA Polimerases Dirigidas por DNA/metabolismo , Proteínas de Fímbrias/genética , Neisseria/metabolismo
20.
Cell Host Microbe ; 26(2): 228-239.e8, 2019 08 14.
Artigo em Inglês | MEDLINE | ID: mdl-31378677

RESUMO

The mucosa is colonized with commensal Neisseria. Some of these niches are sites of infection for the STD pathogen Neisseria gonorrhoeae (Ngo). Given the antagonistic behavior of commensal bacteria toward their pathogenic relatives, we hypothesized that commensal Neisseria may negatively affect Ngo colonization. Here, we report that commensal species of Neisseria kill Ngo through a mechanism based on genetic competence and DNA methylation state. Specifically, commensal-triggered killing occurs when the pathogen takes up commensal DNA containing a methylation pattern that it does not recognize. Indeed, any DNA will kill Ngo if it can enter the cell, is differentially methylated, and has homology to the pathogen genome. Consistent with these findings, commensal Neisseria elongata accelerates Ngo clearance from the mouse in a DNA-uptake-dependent manner. Collectively, we propose that commensal Neisseria antagonizes Ngo infection through a DNA-mediated mechanism and that DNA is a potential microbicide against this highly drug-resistant pathogen.


Assuntos
DNA Bacteriano/metabolismo , Neisseria gonorrhoeae/crescimento & desenvolvimento , Neisseria/fisiologia , Simbiose , Animais , Antibiose/fisiologia , Técnicas de Cocultura , Contagem de Colônia Microbiana , Dano ao DNA , Metilação de DNA , Feminino , Camundongos , Camundongos Endogâmicos BALB C , Modelos Animais , Neisseria/genética , Neisseria gonorrhoeae/genética
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa