Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
1.
mBio ; 15(1): e0279223, 2024 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-38084997

RESUMEN

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.


Asunto(s)
Proteínas Fimbrias , Fimbrias Bacterianas , Ratones , Animales , Humanos , Neisseria/genética , Simbiosis , Neisseria gonorrhoeae , Proteínas Bacterianas
2.
PLoS Pathog ; 18(5): e1010497, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35580146

RESUMEN

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.


Asunto(s)
Elementos Transponibles de ADN , Interacciones Microbiota-Huesped , Neisseria , Animales , Portador Sano/microbiología , Portador Sano/fisiopatología , Elementos Transponibles de ADN/genética , Biblioteca de Genes , Interacciones Microbiota-Huesped/genética , Interacciones Microbiota-Huesped/fisiología , Ratones , Microbiota/genética , Membrana Mucosa/microbiología , Neisseria/genética , Neisseria/patogenicidad , Neisseria gonorrhoeae/genética , Neisseria gonorrhoeae/patogenicidad , Neisseria meningitidis/genética , Neisseria meningitidis/patogenicidad , Simbiosis/genética , Simbiosis/fisiología , Factores de Virulencia/genética
3.
Microb Cell ; 6(12): 544-546, 2019 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-31832426

RESUMEN

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.

4.
Cell Host Microbe ; 26(2): 228-239.e8, 2019 08 14.
Artículo en Inglés | MEDLINE | ID: mdl-31378677

RESUMEN

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.


Asunto(s)
ADN Bacteriano/metabolismo , Neisseria gonorrhoeae/crecimiento & desarrollo , Neisseria/fisiología , Simbiosis , Animales , Antibiosis/fisiología , Técnicas de Cocultivo , Recuento de Colonia Microbiana , Daño del ADN , Metilación de ADN , Femenino , Ratones , Ratones Endogámicos BALB C , Modelos Animales , Neisseria/genética , Neisseria gonorrhoeae/genética
5.
Methods Mol Biol ; 1997: 403-412, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31119636

RESUMEN

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.


Asunto(s)
Modelos Animales de Enfermedad , Gonorrea/microbiología , Neisseria/patogenicidad , Animales , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Microbioma Gastrointestinal/inmunología , Gonorrea/terapia , Humanos , Ratones/microbiología , Mucosa Bucal/inmunología , Mucosa Bucal/microbiología , Neisseria/genética , Neisseria/inmunología , Simbiosis/inmunología , Factores de Virulencia/genética , Factores de Virulencia/metabolismo
6.
PLoS Pathog ; 15(2): e1007495, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30753248

RESUMEN

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.


Asunto(s)
Gonorrea/metabolismo , Proteína Cofactora de Membrana/fisiología , Neisseria gonorrhoeae/patogenicidad , Autofagia/fisiología , Adhesión Bacteriana , Línea Celular , Cuello del Útero , Regulación hacia Abajo , Células Epiteliales , Femenino , Fimbrias Bacterianas , Gonorrea/fisiopatología , Humanos , Lisosomas , Proteína Cofactora de Membrana/inmunología , Glicoproteínas de Membrana , Proteínas de la Membrana/metabolismo , Neisseria gonorrhoeae/metabolismo , Cultivo Primario de Células , Isoformas de Proteínas
7.
Microbiologyopen ; 8(5): e00713, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30079633

RESUMEN

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.


Asunto(s)
Proteínas Fimbrias/biosíntesis , Regulación Bacteriana de la Expresión Génica , Histidina Quinasa/metabolismo , Neisseria/genética , Transducción de Señal , Factores de Transcripción/metabolismo , Transcripción Genética , ARN Polimerasas Dirigidas por ADN/metabolismo , Proteínas Fimbrias/genética , Neisseria/metabolismo
8.
Mol Microbiol ; 110(5): 677-688, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-29719082

RESUMEN

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.


Asunto(s)
Proteínas Fimbrias , Proteínas Motoras Moleculares , Neisseria gonorrhoeae/genética , Neisseria gonorrhoeae/metabolismo , Acetilación , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Proteínas Fimbrias/genética , Proteínas Fimbrias/metabolismo , Fimbrias Bacterianas/metabolismo , Proteínas Motoras Moleculares/genética , Proteínas Motoras Moleculares/metabolismo , Mutación , Procesamiento Proteico-Postraduccional
9.
Infect Immun ; 86(5)2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29440372

RESUMEN

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.


Asunto(s)
Infecciones por Bacterias Gramnegativas/inmunología , Infecciones por Bacterias Gramnegativas/transmisión , Interacciones Huésped-Patógeno , Inmunidad Innata , Ratones/microbiología , Neisseria/patogenicidad , Simbiosis , Animales , Modelos Animales de Enfermedad
10.
Immunohorizons ; 2(9): 305-313, 2018 10 31.
Artículo en Inglés | MEDLINE | ID: mdl-31022695

RESUMEN

Neisseria musculi, isolated from the oral cavity of wild-caught mice, does not colonize most inbred mouse strains. N. musculi does weakly (50%) colonize C57BL/6J (B6) mice but readily colonizes CAST/EiJ (CAST) mice. In this study, we examined whether differences in the CAST and B6 host response could elucidate mechanisms governing N. musculi colonization. In vivo stimulation of B6 or CAST splenocytes with wild type (WT) Neisseria or Escherichia coli LPS showed that CAST mice had a blunted inflammatory response, producing significantly lower levels of IL-6 than B6 mice. The use of specific genetic knockouts highlighted a need for an intact innate immune system to prevent colonization. B6-RAG-1-/- mice were colonized at a similar rate as WT B6 mice, whereas B6-MyD88-/- and TLR4-/- mice were readily colonized like CAST (100%) mice. Sequence analysis revealed a unique point mutation in TLR4 in CAST mice. However, crosses to TLR4-/- mice and analysis of recombinant inbred Collaborative Cross mice showed that TLR4 from CAST mice was not sufficient to allow Neisseria colonization. In vitro stimulation of B6 bone marrow-derived macrophages or splenocytes with WT Neisseria yielded low levels of IL-6 compared with LPS stimulation. Surprisingly, UV-inactivated Neisseria induced high levels of IL-6, suggesting suppression of IL-6 production is an active bacterial process. Consistent with a critical role for IL-6 in preventing colonization, mice deficient for the IL-6 receptor were efficiently colonized, indicating host IL-6 production plays a critical role in determining host colonization susceptibility.


Asunto(s)
Infecciones por Bacterias Gramnegativas/inmunología , Neisseria/inmunología , Inmunidad Adaptativa , Animales , Escherichia coli , Interacciones Microbiota-Huesped , Inmunidad Innata , Interleucina-6/deficiencia , Interleucina-6/inmunología , Macrófagos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos , Ratones Noqueados , Simbiosis/inmunología , Receptor Toll-Like 4
11.
Infect Immun ; 85(3)2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28052997

RESUMEN

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.


Asunto(s)
Gonorrea/microbiología , Interacciones Huésped-Patógeno , Membrana Mucosa/microbiología , Neisseria gonorrhoeae/fisiología , Vagina/microbiología , Técnicas de Cultivo de Célula , Línea Celular , Quimiocinas/metabolismo , Citocinas/metabolismo , Femenino , Humanos , Técnicas In Vitro , Mucinas/metabolismo , Membrana Mucosa/ultraestructura , Mutación
12.
mBio ; 7(6)2016 12 06.
Artículo en Inglés | MEDLINE | ID: mdl-27923924

RESUMEN

Retraction of the type IV pilus (Tfp) mediates DNA uptake, motility, and social and infection behavior in a wide variety of prokaryotes. To date, investigations into Tfp retraction-dependent activities have used a mutant deleted of PilT, the ATPase motor protein that causes the pilus fiber to retract. ΔpilT cells are nontransformable, nonmotile, and cannot aggregate into microcolonies. We tested the hypothesis that these retraction-dependent activities are sensitive to the strength of PilT enzymatic activity by using the pathogen Neisseria gonorrhoeae as a model. We constructed an N. gonorrhoeae mutant with an amino acid substitution in the PilT Walker B box (a substitution of cysteine for leucine at position 201, encoded by pilTL201C). Purified PilTL201C forms a native hexamer, but mutant hexamers hydrolyze ATP at half the maximal rate. N. gonorrhoeae pilTL201C cells produce Tfp fibers, crawl at the same speed as the wild-type (wt) parent, and are equally transformable. However, the social behavior of pilTL201C cells is intermediate between the behaviors of wt and ΔpilT cells. The infection behavior of pilTL201C is also defective, due to its failure to activate the epidermal growth factor receptor (EGFR)-heparin-binding EGF-like growth factor (HB-EGF) pathway. Our study indicates that pilus retraction, per se, is not sufficient for N. gonorrhoeae microcolony formation or infectivity; rather, these activities are sensitive to the strength of PilT enzymatic activity. We discuss the implications of these findings for Neisseria pathogenesis in the context of mechanobiology. IMPORTANCE: Type IV pili are fibers expressed on the surface of many bacteria. Neisseria gonorrhoeae cells crawl, take up DNA, and communicate with each other and with human cells by retracting these fibers. Here, we show that an N. gonorrhoeae mutant expressing an enzymatically weakened type IV pilus retraction motor still crawls and takes up DNA normally. However, mutant cells exhibit abnormal social behavior, and they are less infective because they fail to activate the epidermal growth factor receptor. Our study shows that N. gonorrhoeae social and infection behaviors are sensitive to the strength of the retraction motor enzyme.


Asunto(s)
Proteínas Fimbrias/genética , Proteínas Fimbrias/metabolismo , Fimbrias Bacterianas/fisiología , Interacciones Microbianas , Neisseria gonorrhoeae/fisiología , Neisseria gonorrhoeae/patogenicidad , Adenosina Trifosfato/metabolismo , Sustitución de Aminoácidos , Adhesión Bacteriana , Análisis Mutacional de ADN , Competencia de la Transformación por ADN , Hidrólisis , Locomoción , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Neisseria gonorrhoeae/genética
13.
Int J Syst Evol Microbiol ; 66(9): 3585-3593, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27298306

RESUMEN

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.


Asunto(s)
Ratones/microbiología , Neisseria/clasificación , Filogenia , Animales , Técnicas de Tipificación Bacteriana , Composición de Base , ADN Bacteriano/genética , Genes Bacterianos , Boca/microbiología , Tipificación de Secuencias Multilocus , Neisseria/genética , Neisseria/aislamiento & purificación , América del Norte , Hibridación de Ácido Nucleico , ARN Ribosómico 16S/genética , Análisis de Secuencia de ADN
14.
Mol Microbiol ; 90(1): 103-13, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23899162

RESUMEN

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.


Asunto(s)
Proteínas Fimbrias/biosíntesis , Regulación Bacteriana de la Expresión Génica , Neisseria elongata/genética , ARN Polimerasa Sigma 54/metabolismo , Factores de Integración del Huésped/metabolismo , Factores de Transcripción/metabolismo , Transcripción Genética
15.
Cell Microbiol ; 15(11): 1837-50, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23648135

RESUMEN

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.


Asunto(s)
Factor de Transcripción Activador 3/metabolismo , Interacciones Huésped-Patógeno , Evasión Inmune , Interleucina-6/antagonistas & inhibidores , Neisseria gonorrhoeae/fisiología , Línea Celular Tumoral , Células Epiteliales/inmunología , Células Epiteliales/microbiología , Regulación de la Expresión Génica , Humanos
16.
Proc Natl Acad Sci U S A ; 110(8): 3059-64, 2013 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-23382234

RESUMEN

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.


Asunto(s)
Transferencia de Gen Horizontal , Infecciones por Bacterias Gramnegativas/microbiología , Neisseria/patogenicidad , Animales , Marcadores Genéticos , Infecciones por Bacterias Gramnegativas/genética , Infecciones por Bacterias Gramnegativas/transmisión , Interacciones Huésped-Patógeno , Macaca mulatta , Datos de Secuencia Molecular , Neisseria/clasificación , Neisseria/genética , Filogenia , Virulencia
17.
Methods Mol Biol ; 799: 197-216, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-21993648

RESUMEN

The importance of physical forces in biology is becoming more appreciated. Neisseria gonorrhoeaehas become a paradigm for the study of physical forces in the bacterial world. Cycles of elongations and retractions of Type IV pili enables N. gonorrhoeaebacteria to exert forces on its environment, forces that play major roles in the life cycle of this pathogen. In order to better understand the role of these forces, there is a need to fully characterize them. Here, we present two different techniques, optical tweezers and Polyacrylamide MicroPillars (PoMPs), for measuring pilus retraction forces. Initially designed for N. gonorrhoeae, these assays can be readily modified to study other pilus-bearing bacteria including Neisseria meningitidis.


Asunto(s)
Fimbrias Bacterianas/fisiología , Movimiento/fisiología , Neisseria/fisiología , Pinzas Ópticas , Resinas Acrílicas , Fenómenos Biomecánicos
18.
PLoS One ; 6(6): e21373, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21731720

RESUMEN

The genus Neisseria contains at least eight commensal and two pathogenic species. According to the Neisseria phylogenetic tree, commensals are basal to the pathogens. N. elongata, which is at the opposite end of the tree from N. gonorrhoeae, has been observed to be fimbriated, and these fimbriae are correlated with genetic competence in this organism. We tested the hypothesis that the fimbriae of N. elongata are Type IV pili (Tfp), and that Tfp functions in genetic competence. We provide evidence that the N. elongata fimbriae are indeed Tfp. Tfp, as well as the DNA Uptake Sequence (DUS), greatly enhance N. elongata DNA transformation. Tfp allows N. elongata to make intimate contact with N. gonorrhoeae and to mediate the transfer of antibiotic resistance markers between these two species. We conclude that Tfp functional for genetic competence is a trait of a commensal member of the Neisseria genus. Our findings provide a mechanism for the horizontal gene transfer that has been observed among Neisseria species.


Asunto(s)
Fimbrias Bacterianas/metabolismo , Transferencia de Gen Horizontal/genética , Genes Bacterianos/genética , Neisseria elongata/metabolismo , Neisseria gonorrhoeae/genética , Secuencia de Bases , ADN Bacteriano/metabolismo , Farmacorresistencia Bacteriana/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Células Epiteliales/microbiología , Células Epiteliales/ultraestructura , Fimbrias Bacterianas/efectos de los fármacos , Fimbrias Bacterianas/genética , Fimbrias Bacterianas/ultraestructura , Humanos , Mutación/genética , Neisseria elongata/efectos de los fármacos , Neisseria elongata/genética , Neisseria elongata/ultraestructura , Neisseria gonorrhoeae/efectos de los fármacos , Neisseria gonorrhoeae/ultraestructura , Rifampin/farmacología , Especificidad de la Especie , Propiedades de Superficie/efectos de los fármacos , Transcripción Genética/efectos de los fármacos , Transformación Bacteriana/efectos de los fármacos , Transformación Bacteriana/genética
19.
PLoS One ; 6(1): e16287, 2011 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-21283821

RESUMEN

BACKGROUND: Adequate termination of an immune response is as important as the induction of an appropriate response. CD46, a regulator of complement activity, promotes T cell activation and differentiation towards a regulatory Tr1 phenotype. This Tr1 differentiation pathway is defective in patients with MS, asthma and rheumatoid arthritis, underlying its importance in controlling T cell function and the need to understand its regulatory mechanisms. CD46 has two cytoplasmic tails, Cyt1 and Cyt2, derived from alternative splicing, which are co-expressed in all nucleated human cells. The regulation of their expression and precise functions in regulating human T cell activation has not been fully elucidated. METHODOLOGY/PRINCIPAL FINDINGS: Here, we first report the novel role of CD46 in terminating T cell activation. Second, we demonstrate that its functions as an activator and inhibitor of T cell responses are mediated through the temporal processing of its cytoplasmic tails. Cyt1 processing is required to turn T cell activation on, while processing of Cyt2 switches T cell activation off, as demonstrated by proliferation, CD25 expression and cytokine secretion. Both tails require processing by Presenilin/γSecretase (P/γS) to exert these functions. This was confirmed by expressing wild-type Cyt1 and Cyt2 tails and uncleavable mutant tails in primary T cells. The role of CD46 tails was also demonstrated with T cells expressing CD19 ectodomain-CD46 C-Terminal Fragment (CTF) fusions, which allowed specific triggering of each tail individually. CONCLUSIONS/SIGNIFICANCE: We conclude that CD46 acts as a molecular rheostat to control human T cell activation through the regulation of processing of its cytoplasmic tails.


Asunto(s)
Activación de Linfocitos , Proteína Cofactora de Membrana/metabolismo , Linfocitos T/inmunología , Diferenciación Celular/inmunología , Células Cultivadas , Humanos , Espacio Intracelular/metabolismo , Presenilinas/metabolismo , Estructura Terciaria de Proteína , Linfocitos T/citología , Linfocitos T Reguladores/citología
20.
PLoS One ; 5(7): e11835, 2010 Jul 28.
Artículo en Inglés | MEDLINE | ID: mdl-20676376

RESUMEN

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.


Asunto(s)
Transferencia de Gen Horizontal/genética , Genoma Bacteriano/genética , Neisseria/genética , Virulencia/genética , Humanos , Neisseria/patogenicidad , Neisseria gonorrhoeae/genética , Neisseria gonorrhoeae/patogenicidad , Neisseria lactamica/genética , Neisseria lactamica/patogenicidad , Neisseria meningitidis/genética , Neisseria meningitidis/patogenicidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA