Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters











Publication year range
1.
Infect Immun ; 92(7): e0004824, 2024 Jul 11.
Article in English | MEDLINE | ID: mdl-38814083

ABSTRACT

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


Subject(s)
Neisseria , Animals , Mice , Neisseria/immunology , Host-Pathogen Interactions/immunology , Female , Mice, Inbred C57BL , Disease Models, Animal , Mouth/microbiology , Mouth/immunology
2.
Infect Immun ; 92(5): e0000424, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38563734

ABSTRACT

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


Subject(s)
Neisseria , Nod1 Signaling Adaptor Protein , Nod2 Signaling Adaptor Protein , Peptidoglycan , Animals , Humans , Mice , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Membrane Transport Proteins , Neisseria/genetics , Neisseria gonorrhoeae/immunology , Neisseria gonorrhoeae/genetics , Neisseria gonorrhoeae/metabolism , Nod1 Signaling Adaptor Protein/metabolism , Nod1 Signaling Adaptor Protein/genetics , Nod2 Signaling Adaptor Protein/metabolism , Nod2 Signaling Adaptor Protein/genetics , Peptidoglycan/metabolism
3.
Microbiol Resour Announc ; 10(23): e0045221, 2021 Jun 10.
Article in English | MEDLINE | ID: mdl-34110239

ABSTRACT

Neisseria musculi is an oral commensal of wild-caught mice. Here, we report the complete genome sequence of N. musculi strain NW831, generated using a combination of the Illumina and PacBio platforms.

4.
Article in English | MEDLINE | ID: mdl-32423958

ABSTRACT

Pharyngeal infections by Neisseria gonorrhoeae are often asymptomatic, making them difficult to treat. However, in vivo animal modeling of human pharyngeal infections by pathogenic Neisseria species is challenging due to numerous host tropism barriers. We have relied on rhesus macaques to investigate pharyngeal persistence of naturally occurring Neisseria species in response to antibiotics. These species include Neisseria mucosa, Neisseria oralis, and a species unique to macaques. Four animals previously treated intramuscularly with the fluoroquinolone enrofloxacin for 2 weeks were monitored for persistence of their preexisting Neisseria populations for a period of 10 weeks. Enrofloxacin exposure did not eliminate preexisting flora from two of the four animals. Characterization of a collection of macaque Neisseria isolates supported the hypothesis that pharyngeal persistence was linked to reduced enrofloxacin susceptibility conferred by mutations in either gyrA or parC Interestingly, we observed a change in neisserial population dynamics for several weeks following enrofloxacin exposure. Enrofloxacin appeared to promote competition between strains for dominance in the pharyngeal niche. Specifically, following enrofloxacin treatment, strains bearing single gyrA mutations and low MICs persisted long-term. In contrast, strains with both gyrA and parC mutations and high MICs became culturally undetectable, consistent with the hypothesis that they were less fit. Our study has provided insight into pharyngeal persistence dynamics of Neisseria species bearing fluoroquinolone resistance determinants. The rhesus macaque provides a valuable host animal that may be used in the future to simulate treatment failures associated with the presence of antimicrobial-resistant Neisseria spp. in the human pharynx.


Subject(s)
DNA Gyrase , DNA Topoisomerase IV , Animals , Anti-Bacterial Agents/pharmacology , DNA Gyrase/genetics , DNA Topoisomerase IV/genetics , Fluoroquinolones/pharmacology , Humans , Macaca mulatta , Microbial Sensitivity Tests , Mutation , Neisseria , Neisseria gonorrhoeae/genetics , Pharynx
5.
PLoS Pathog ; 15(2): e1007495, 2019 02.
Article in English | MEDLINE | ID: mdl-30753248

ABSTRACT

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.


Subject(s)
Gonorrhea/metabolism , Membrane Cofactor Protein/physiology , Neisseria gonorrhoeae/pathogenicity , Autophagy/physiology , Bacterial Adhesion , Cell Line , Cervix Uteri , Down-Regulation , Epithelial Cells , Female , Fimbriae, Bacterial , Gonorrhea/physiopathology , Humans , Lysosomes , Membrane Cofactor Protein/immunology , Membrane Glycoproteins , Membrane Proteins/metabolism , Neisseria gonorrhoeae/metabolism , Primary Cell Culture , Protein Isoforms
6.
J Bacteriol ; 200(16)2018 08 15.
Article in English | MEDLINE | ID: mdl-29866804

ABSTRACT

The affordability of bacterial genome sequencing has provided a helpful tool for sequencing large strain collections. Bente Børud (J. Bacteriol. 200:e00794-17, 2018, https://doi.org/doi:10.1128/JB.00794-17) recently led an effort to analyze the genomes of a collection of oropharyngeal Neisseria meningitidis isolates from 50 healthy individuals. Paired longitudinal isolates from each individual were sequenced. Genome analyses focused on (i) predicting the expression state of phase-variable loci that encode enzymes important for O-linked protein glycosylation and (ii) correlating specific genotypes with glycosylation phenotypes.


Subject(s)
Meningococcal Infections , Neisseria meningitidis , Genotype , Glycosylation , Humans , Phenotype , Polysaccharides
7.
Infect Immun ; 86(5)2018 05.
Article in English | MEDLINE | ID: mdl-29440372

ABSTRACT

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.


Subject(s)
Gram-Negative Bacterial Infections/immunology , Gram-Negative Bacterial Infections/transmission , Host-Pathogen Interactions , Immunity, Innate , Mice/microbiology , Neisseria/pathogenicity , Symbiosis , Animals , Disease Models, Animal
8.
Pathog Dis ; 75(3)2017 Apr 01.
Article in English | MEDLINE | ID: mdl-28369241

ABSTRACT

The Gram-negative bacteria genus Neisseria includes both pathogenic and commensal species that are found primarily in the upper respiratory tract of humans and animals. The development of animal models to study neisserial pathogenesis has focused almost exclusively on two species that cause disease in humans. These include Neisseria meningitidis, an obligate commensal that can cause invasive disease, and N. gonorrhoeae, the causative agent of gonorrhea. Both pathogens can persist in the upper respiratory tract. This article will give a brief overview of the genus Neisseria. The anatomy of the upper respiratory tract and its use as a niche for bacteria will be discussed. Next, studies that provide insight about the first stage of upper respiratory tract infection, namely colonization, will be reviewed. Most studies of upper respiratory tract infection have focused on N. meningitidis infections of laboratory mice. This review will also discuss models of respiratory tract persistence by Neisseria species, including commensals, in mice, non-human primates and human volunteers. The article includes a section that discusses the future utility of upper respiratory tract models in informing the development of effective antimicrobial therapies. Such knowledge is needed to minimize the dissemination of antimicrobial resistance from respiratory reservoirs.


Subject(s)
Neisseria/physiology , Respiratory System/microbiology , Respiratory Tract Infections/microbiology , Animals , Animals, Genetically Modified , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Disease Models, Animal , Drug Resistance, Bacterial , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Humans , Immunity , Neisseria/drug effects , Respiratory System/anatomy & histology , Respiratory System/immunology , Respiratory System/pathology , Respiratory Tract Infections/diagnosis , Respiratory Tract Infections/drug therapy , Respiratory Tract Infections/immunology , Risk Factors
9.
Int J Syst Evol Microbiol ; 66(9): 3585-3593, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27298306

ABSTRACT

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.


Subject(s)
Mice/microbiology , Neisseria/classification , Phylogeny , Animals , Bacterial Typing Techniques , Base Composition , DNA, Bacterial/genetics , Genes, Bacterial , Mouth/microbiology , Multilocus Sequence Typing , Neisseria/genetics , Neisseria/isolation & purification , North America , Nucleic Acid Hybridization , RNA, Ribosomal, 16S/genetics , Sequence Analysis, DNA
10.
Proc Natl Acad Sci U S A ; 110(8): 3059-64, 2013 Feb 19.
Article in English | MEDLINE | ID: mdl-23382234

ABSTRACT

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.


Subject(s)
Gene Transfer, Horizontal , Gram-Negative Bacterial Infections/microbiology , Neisseria/pathogenicity , Animals , Genetic Markers , Gram-Negative Bacterial Infections/genetics , Gram-Negative Bacterial Infections/transmission , Host-Pathogen Interactions , Macaca mulatta , Molecular Sequence Data , Neisseria/classification , Neisseria/genetics , Phylogeny , Virulence
11.
PLoS One ; 6(6): e21373, 2011.
Article in English | MEDLINE | ID: mdl-21731720

ABSTRACT

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.


Subject(s)
Fimbriae, Bacterial/metabolism , Gene Transfer, Horizontal/genetics , Genes, Bacterial/genetics , Neisseria elongata/metabolism , Neisseria gonorrhoeae/genetics , Base Sequence , DNA, Bacterial/metabolism , Drug Resistance, Bacterial/drug effects , Epithelial Cells/drug effects , Epithelial Cells/microbiology , Epithelial Cells/ultrastructure , Fimbriae, Bacterial/drug effects , Fimbriae, Bacterial/genetics , Fimbriae, Bacterial/ultrastructure , Humans , Mutation/genetics , Neisseria elongata/drug effects , Neisseria elongata/genetics , Neisseria elongata/ultrastructure , Neisseria gonorrhoeae/drug effects , Neisseria gonorrhoeae/ultrastructure , Rifampin/pharmacology , Species Specificity , Surface Properties/drug effects , Transcription, Genetic/drug effects , Transformation, Bacterial/drug effects , Transformation, Bacterial/genetics
12.
PLoS One ; 6(1): e16287, 2011 Jan 19.
Article in English | MEDLINE | ID: mdl-21283821

ABSTRACT

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.


Subject(s)
Lymphocyte Activation , Membrane Cofactor Protein/metabolism , T-Lymphocytes/immunology , Cell Differentiation/immunology , Cells, Cultured , Humans , Intracellular Space/metabolism , Presenilins/metabolism , Protein Structure, Tertiary , T-Lymphocytes/cytology , T-Lymphocytes, Regulatory/cytology
13.
PLoS One ; 5(7): e11835, 2010 Jul 28.
Article in English | MEDLINE | ID: mdl-20676376

ABSTRACT

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.


Subject(s)
Gene Transfer, Horizontal/genetics , Genome, Bacterial/genetics , Neisseria/genetics , Virulence/genetics , Humans , Neisseria/pathogenicity , Neisseria gonorrhoeae/genetics , Neisseria gonorrhoeae/pathogenicity , Neisseria lactamica/genetics , Neisseria lactamica/pathogenicity , Neisseria meningitidis/genetics , Neisseria meningitidis/pathogenicity
14.
J Immunol ; 184(2): 694-701, 2010 Jan 15.
Article in English | MEDLINE | ID: mdl-20018629

ABSTRACT

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.


Subject(s)
Amyloid Precursor Protein Secretases/metabolism , Gonorrhea/metabolism , Membrane Cofactor Protein/metabolism , Meningococcal Infections/metabolism , Presenilins/metabolism , Fimbriae, Bacterial , Humans , Mechanotransduction, Cellular , Membrane Cofactor Protein/physiology , Neisseria gonorrhoeae , Neisseria meningitidis , Protein Isoforms , Signal Transduction
15.
Microbiology (Reading) ; 155(Pt 12): 4084-4092, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19762436

ABSTRACT

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.


Subject(s)
Fimbriae, Bacterial/ultrastructure , Neisseria gonorrhoeae/pathogenicity , Neisseria gonorrhoeae/ultrastructure , Bacterial Adhesion/physiology , Cell Line , Fimbriae, Bacterial/physiology , Humans , Imaging, Three-Dimensional , Microscopy, Electron, Scanning , Microscopy, Electron, Transmission , Neisseria gonorrhoeae/physiology , Virulence/physiology
16.
Infect Immun ; 75(10): 4743-53, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17682045

ABSTRACT

Neisseria gonorrhoeae is the bacterium that causes gonorrhea, a major sexually transmitted disease and a significant cofactor for human immunodeficiency virus transmission. The retactile N. gonorrhoeae type IV pilus (Tfp) mediates twitching motility and attachment. Using live-cell microscopy, we reveal for the first time the dynamics of twitching motility by N. gonorrhoeae in its natural environment, human epithelial cells. Bacteria aggregate into microcolonies on the cell surface and induce a massive remodeling of the microvillus architecture. Surprisingly, the microcolonies are motile, and they fuse to form progressively larger structures that undergo rapid reorganization, suggesting that bacteria communicate with each other during infection. As reported, actin plaques form beneath microcolonies. Here, we show that cortical plaques comigrate with motile microcolonies. These activities are dependent on pilT, the Tfp retraction locus. Cultures infected with a pilT mutant have significantly higher numbers of apoptotic cells than cultures infected with the wild-type strain. Inducing pilT expression with isopropyl-beta-D-thiogalactopyranoside partially rescues cells from infection-induced apoptosis, demonstrating that Tfp retraction is intrinsically cytoprotective for the host. Tfp-mediated attachment is therefore a continuum of microcolony motility and force stimulation of host cell signaling, leading to a cytoprotective effect.


Subject(s)
Bacterial Adhesion/physiology , Cytoprotection , Epithelial Cells/microbiology , Fimbriae, Bacterial/physiology , Neisseria gonorrhoeae/physiology , Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/physiology , Apoptosis , Bacterial Proteins/genetics , Bacterial Proteins/physiology , Cell Line, Tumor , Humans , Locomotion/physiology , Microscopy/methods , Molecular Motor Proteins/genetics , Molecular Motor Proteins/physiology , Mutation
17.
Infect Immun ; 74(4): 2428-35, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16552073

ABSTRACT

CD46 (membrane cofactor protein), a complement-regulatory protein that participates in innate and acquired immunity, also serves as a receptor for viral and bacterial pathogens. CD46 isoforms terminate in one of two cytoplasmic tails, Cyt1 or Cyt2, which differ in signaling and trafficking properties. Dissecting the functions of the two cytoplasmic tails in these cellular processes has been hampered by the absence of specific reagents. Here we report the construction of Cyt1- and Cyt2-specific monoclonal antibodies (MAbs). These MAbs recognize unique epitopes within the tails and can be used for immunofluorescence microscopy, immunoblotting, and immunoprecipitation. Studies of Neisseria gonorrhoeae-infected cells with the CD46 tail MAbs demonstrate the differential recruitment of Cyt1 and Cyt2 to the cortical plaque.


Subject(s)
Antibodies, Monoclonal/metabolism , Membrane Cofactor Protein/immunology , Membrane Cofactor Protein/metabolism , Neisseria gonorrhoeae/immunology , Neisseria gonorrhoeae/isolation & purification , Amino Acid Sequence , Antibodies, Monoclonal/biosynthesis , Bacterial Adhesion/immunology , Cell Line, Tumor , Cell Polarity/immunology , Cytoplasm/immunology , Epithelial Cells/immunology , Epithelial Cells/metabolism , Humans , Membrane Cofactor Protein/genetics , Microscopy, Fluorescence , Molecular Sequence Data , Neisseria gonorrhoeae/metabolism , Protein Isoforms/genetics , Protein Isoforms/immunology , Protein Isoforms/metabolism
18.
BMC Microbiol ; 5: 50, 2005 Sep 01.
Article in English | MEDLINE | ID: mdl-16137322

ABSTRACT

BACKGROUND: The sexually transmitted disease, gonorrhea, is a serious health problem in developed as well as in developing countries, for which treatment continues to be a challenge. The recent completion of the genome sequence of the causative agent, Neisseria gonorrhoeae, opens up an entirely new set of approaches for studying this organism and the diseases it causes. Here, we describe the initial phases of the construction of an expression-capable clone set representing the protein-coding ORFs of the gonococcal genome using a recombination-based cloning system. RESULTS: The clone set thus far includes 1672 of the 2250 predicted ORFs of the N. gonorrhoeae genome, of which 1393 (83%) are sequence-validated. Included in this set are 48 of the 61 ORFs of the gonococcal genetic island of strain MS11, not present in the sequenced genome of strain FA1090. L-arabinose-inducible glutathione-S-transferase (GST)-fusions were constructed from random clones and each was shown to express a fusion protein of the predicted size following induction, demonstrating the use of the recombination cloning system. PCR amplicons of each ORF used in the cloning reactions were spotted onto glass slides to produce DNA microarrays representing 2035 genes of the gonococcal genome. Pilot experiments indicate that these arrays are suitable for the analysis of global gene expression in gonococci. CONCLUSION: This archived set of Gateway entry clones will facilitate high-throughput genomic and proteomic studies of gonococcal genes using a variety of expression and analysis systems. In addition, the DNA arrays produced will allow us to generate gene expression profiles of gonococci grown in a wide variety of conditions. Together, the resources produced in this work will facilitate experiments to dissect the molecular mechanisms of gonococcal pathogenesis on a global scale, and ultimately lead to the determination of the functions of unknown genes in the genome.


Subject(s)
Gene Library , Neisseria gonorrhoeae/genetics , Cloning, Molecular , Gene Expression Regulation, Bacterial , Genome, Bacterial , Neisseria gonorrhoeae/classification , Oligonucleotide Array Sequence Analysis , Open Reading Frames
SELECTION OF CITATIONS
SEARCH DETAIL