Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 26
1.
STAR Protoc ; 3(2): 101256, 2022 06 17.
Article En | MEDLINE | ID: mdl-35345596

The multilayered meninges surrounding the brain and spinal cord harbor distinct immune cell populations with prominent roles in health and diseases. Here we present an optimized protocol for RNA fluorescence in situ hybridization (RNA FISH) in meningeal whole mounts, allowing the visualization of gene expression. We also describe the combination of this protocol with immunohistochemistry for simultaneous visualization of mRNA and proteins. This protocol can be used for assessing spatial gene expression within the meninges.


Brain , RNA , Animals , Immunohistochemistry , In Situ Hybridization, Fluorescence/methods , Mice , RNA, Messenger/genetics
2.
Cell Host Microbe ; 29(7): 1177-1185.e6, 2021 07 14.
Article En | MEDLINE | ID: mdl-34043959

Persistent and intermittent fecal shedding, hallmarks of Salmonella infections, are important for fecal-oral transmission. In the intestine, Salmonella enterica serovar Typhimurium (STm) actively invades intestinal epithelial cells (IECs) and survives in the Salmonella-containing vacuole (SCV) and the cell cytosol. Cytosolic STm replicate rapidly, express invasion factors, and induce extrusion of infected epithelial cells into the intestinal lumen. Here, we engineered STm that self-destruct in the cytosol (STmCytoKill), but replicates normally in the SCV, to examine the role of cytosolic STm in infection. Intestinal expansion and fecal shedding of STmCytoKill are impaired in mouse models of infection. We propose a model whereby repeated rounds of invasion, cytosolic replication, and release of invasive STm from extruded IECs fuels the high luminal density required for fecal shedding.


Cytosol/microbiology , Epithelial Cells/microbiology , Feces/microbiology , Salmonella Infections/microbiology , Salmonella typhimurium/physiology , Animals , Female , HeLa Cells , Humans , Intestines/microbiology , Male , Mice , Mice, Inbred C57BL , Salmonella typhimurium/genetics , Salmonella typhimurium/growth & development , Vacuoles/microbiology
3.
Nat Commun ; 12(1): 348, 2021 01 13.
Article En | MEDLINE | ID: mdl-33441540

In the enteric pathogen Salmonella enterica serovar Typhimurium, invasion and motility are coordinated by the master regulator HilD, which induces expression of the type III secretion system 1 (T3SS1) and motility genes. Methyl-accepting chemotaxis proteins (MCPs) detect specific ligands and control the direction of the flagellar motor, promoting tumbling and changes in direction (if a repellent is detected) or smooth swimming (in the presence of an attractant). Here, we show that HilD induces smooth swimming by upregulating an uncharacterized MCP (McpC), and this is important for invasion of epithelial cells. Remarkably, in vitro assays show that McpC can suppress tumbling and increase smooth swimming in the absence of exogenous ligands. Expression of mcpC is repressed by the universal regulator H-NS, which can be displaced by HilD. Our results highlight the importance of smooth swimming for Salmonella Typhimurium invasiveness and indicate that McpC can act via a ligand-independent mechanism when incorporated into the chemotactic receptor array.


Bacterial Proteins/metabolism , Chemotaxis/physiology , Methyl-Accepting Chemotaxis Proteins/metabolism , Salmonella typhimurium/metabolism , Transcription Factors/metabolism , Animals , Bacterial Proteins/genetics , Caco-2 Cells , Cattle , Cells, Cultured , Chemotaxis/genetics , Gene Expression Regulation, Bacterial , HeLa Cells , Humans , Methyl-Accepting Chemotaxis Proteins/genetics , Mice, Inbred C57BL , Movement/physiology , Mutation , Salmonella Infections/microbiology , Salmonella typhimurium/genetics , Salmonella typhimurium/physiology , Transcription Factors/genetics
4.
mBio ; 11(4)2020 08 18.
Article En | MEDLINE | ID: mdl-32817110

Chlamydia trachomatis is an obligate intracellular bacterial pathogen that causes blinding trachoma and sexually transmitted disease afflicting hundreds of millions of people globally. A fundamental but poorly understood pathophysiological characteristic of chlamydial infection is the propensity to cause persistent infection that drives damaging inflammatory disease. The chlamydial plasmid is a virulence factor, but its role in the pathogenesis of persistent infection capable of driving immunopathology is unknown. Here, we show by using mouse and nonhuman primate infection models that the secreted plasmid gene protein 3 (Pgp3) is essential for establishing persistent infection. Ppg3-dependent persistent genital tract infection resulted in a severe endometritis caused by an intense infiltration of endometrial submucosal macrophages. Pgp3 released from the cytosol of lysed infected oviduct epithelial cells, not organism outer membrane-associated Pgp3, inhibited the chlamydial killing activity of antimicrobial peptides. Genetic Pgp3 rescue experiments in cathelin-related antimicrobial peptide (CRAMP)-deficient mice showed Pgp3-targeted antimicrobial peptides to subvert innate immunity as a pathogenic strategy to establish persistent infection. These findings provide important advances in understanding the role of Pgp3 in the pathogenesis of persistent chlamydial infection and associated immunopathology.IMPORTANCEChlamydia trachomatis can cause persistent infection that drives damaging inflammatory responses resulting in infertility and blindness. Little is known about chlamydial genes that cause persistence or factors that drive damaging pathology. In this work, we show that the C. trachomatis plasmid protein gene 3 (Pgp3) is the essential virulence factor for establishing persistent female genital tract infection and provide supportive evidence that Pgp3 functions similarly in a nonhuman primate trachoma model. We further show that persistent Ppg3-dependent infection drives damaging immunopathology. These results are important advances in understanding the pathophysiology of chlamydial persistence.


Antigens, Bacterial/genetics , Bacterial Proteins/genetics , Chlamydia Infections/immunology , Chlamydia trachomatis/genetics , Chlamydia trachomatis/pathogenicity , Virulence Factors/genetics , Animals , Antigens, Bacterial/metabolism , Bacterial Proteins/metabolism , Cytokines/immunology , Epithelial Cells/microbiology , Female , HeLa Cells , Humans , Macaca , Mice , Mice, Inbred C57BL
5.
PLoS One ; 14(8): e0215190, 2019.
Article En | MEDLINE | ID: mdl-31393874

The bacterial pathogen Salmonella enterica serovar Typhimurium is one of the most common causes of foodborne disease in humans and is also an important model system for bacterial pathogenesis. Oral inoculation of C57Bl/6 mice, which are genetically susceptible to Salmonella, results in systemic infection but the murine intestine is not efficiently colonized unless the intestinal microbiota is disrupted. Pretreatment of C57Bl/6 mice with streptomycin, followed by oral inoculation with Salmonella Typhimurium results in colitis resembling human intestinal Salmonellosis. The predominant method of delivery of bacteria is oral gavage, during which organisms are deposited directly into the stomach via a feeding needle. Although convenient, this method can be stressful for mice, and may lead to unwanted tracheal or systemic introduction of bacteria. Here, we developed a method for oral infection of mice by voluntary consumption of regular mouse chow inoculated with bacteria. Mice readily ate chow fragments containing up to 108 CFU Salmonella, allowing for a wide range of infectious doses. In mice pretreated with streptomycin, infection with inoculated chow resulted in reproducible infections with doses as low as 103 CFU. Mice not treated with streptomycin, as well as resistant Nramp1 reconstituted C57Bl/6J mice, were also readily infected using this method. In summary, voluntary consumption of chow inoculated with Salmonella represents a natural route of infection for foodborne salmonellosis and a viable alternative to oral gavage.


Salmonella Food Poisoning/metabolism , Salmonella Infections, Animal/microbiology , Animals , Colitis/pathology , Disease Models, Animal , Intestines/microbiology , Mice , Mice, Inbred C57BL , Salmonella typhimurium/pathogenicity , Streptomycin/administration & dosage
6.
Infect Immun ; 86(5)2018 05.
Article En | MEDLINE | ID: mdl-29463617

The Chlamydia trachomatis plasmid and inclusion membrane protein CT135 are virulence factors in the pathogenesis of murine female genital tract infection. To determine if these virulence factors play a similar role in female nonhuman primates, we infected pig-tailed macaques with the same C. trachomatis strains shown to be important in the murine model. Wild-type C. trachomatis and its isogenic mutant strain deficient in both plasmid and CT135 were used to infect macaques. Macaques were given primary and repeated cervicovaginal challenges with the wild-type and mutant strains. The infection rate, infection duration, and antibody response were similar among macaques infected with both strains. Unexpectedly, colposcopy, laparoscopy, and histologic analysis revealed no substantial genital tract pathology following either primary or repeated cervicovaginal challenges. Cytokine analysis of cervicovaginal secretions from both challenged groups revealed low concentrations of interleukin 1ß (IL-1ß) and elevated levels of the interleukin 1 receptor agonist (IL-1RA). We propose that an imbalance of IL-1ß and IL-1RA in macaques is the reason for the mild inflammatory responses observed in infected urogenital tissues. Thus, understanding the pathobiology of chlamydial infection requires a better understanding of host epigenetic and chlamydial genetic factors. Our findings also have implications for understanding the high frequency of asymptomatic infections in humans.


Chlamydia Infections/immunology , Chlamydia trachomatis/genetics , Chlamydia trachomatis/immunology , Macaca/immunology , Plasmids/immunology , Reproductive Tract Infections/immunology , Virulence Factors/immunology , Animals , Female , Humans , Mice , Plasmids/genetics , Virulence Factors/genetics
7.
Genome Res ; 27(7): 1220-1229, 2017 07.
Article En | MEDLINE | ID: mdl-28588068

Chlamydia trachomatis is the world's most prevalent bacterial sexually transmitted infection and leading infectious cause of blindness, yet it is one of the least understood human pathogens, in part due to the difficulties of in vitro culturing and the lack of available tools for genetic manipulation. Genome sequencing has reinvigorated this field, shedding light on the contemporary history of this pathogen. Here, we analyze 563 full genomes, 455 of which are novel, to show that the history of the species comprises two phases, and conclude that the currently circulating lineages are the result of evolution in different genomic ecotypes. Temporal analysis indicates these lineages have recently expanded in the space of thousands of years, rather than the millions of years as previously thought, a finding that dramatically changes our understanding of this pathogen's history. Finally, at a time when almost every pathogen is becoming increasingly resistant to antimicrobials, we show that there is no evidence of circulating genomic resistance in C. trachomatis.


Chlamydia trachomatis/genetics , Drug Resistance, Bacterial/genetics , Ecotype , Evolution, Molecular , Genome, Bacterial , Chlamydia trachomatis/isolation & purification , Female , Humans , Male
8.
Pathog Dis ; 75(3)2017 04 01.
Article En | MEDLINE | ID: mdl-28369275

Chlamydia trachomatis is an obligate intracellular pathogen characterized by a unique biphasic developmental cycle that alternates between infectious and non-infectious organisms. Chlamydial ChxR is a transcriptional activator that has been implicated in the regulation of the development cycle. We used a reverse genetics approach to generate three chxR null mutants. All three mutants grew normally in cultured mammalian cells. Whole genome sequencing identified SNPs in other genes; however, none of the mutated genes were common to all three ChxR null mutants arguing against a genetic compensatory mechanism that would explain the non-essential in vitro growth phenotype. Comparative proteomics identified five proteins, CT005, CT214, CT565, CT694 and CT695, that were significantly downregulated in all ChxR null mutants. This group includes established inclusion membrane and type III secreted proteins. ChxR transcriptional regulation of these genes was confirmed by qRT-PCR. Importantly, while ChxR null mutants exhibited no growth deficiencies in in vitro, they did show significant differences in in vivo growth using a mouse genital tract model. Collectively, our findings demonstrated that ChxR is a transcriptional activator that regulates the expression of virulence genes whose functions are restricted to in vivo infection.


Bacterial Proteins/metabolism , Chlamydia Infections/genetics , Chlamydia Infections/microbiology , Chlamydia trachomatis/metabolism , Host-Pathogen Interactions , Transcription Factors/metabolism , Alleles , Animals , Bacterial Proteins/genetics , Cell Line , Chlamydia trachomatis/genetics , Chlamydia trachomatis/growth & development , Disease Models, Animal , Female , Gene Expression , Gene Expression Regulation , Genome, Bacterial , Humans , Mice , Mutation , Sequence Analysis, DNA , Transcription Factors/genetics , Virulence Factors/genetics , Virulence Factors/metabolism
9.
Pathog Dis ; 73(6): ftv043, 2015 Aug.
Article En | MEDLINE | ID: mdl-26109550

Chlamydia trachomatis is an important human pathogen causing both ocular and sexually transmitted disease. Recently, we identified CT135 as an important virulence determinant in a mouse infection model. Results from CEL 1 digestion assays and sequencing analyses indicated that CT135 was much more polymorphic in high in vitro passage reference serovars than it was in clinical strains that had undergone limited passaging. Herein, we used targeted next-generation sequencing of the CT134-135 locus, from reference strains and clinical isolates, enabling accurate discovery of single nucleotide polymorphisms and other population genetic variations. Our results indicate that CT134 is stable in all C. trachomatis serovars examined. In contrast, CT135 is highly polymorphic in high-passaged reference ocular and non-LGV genital serovars, with the majority of the mutations resulting in gene disruption. In low-passaged ocular clinical isolates, CT135 was frequently disrupted, whereas in genital clinical isolates CT135 was intact in almost all instances. When a serovar K isolate, with an intact CT134 and CT135, was subjected to serial passage in vitro CT134 remained invariable, while numerous gene interrupting mutations rapidly accumulated in CT135. Collectively, our data indicate that, for genital serovars, CT135 is under strong positive selection in vivo, and negative selection in vitro.


Chlamydia trachomatis/genetics , Genetic Variation , Genomic Instability , Virulence Factors/genetics , Animals , Chlamydia Infections/microbiology , Chlamydia trachomatis/isolation & purification , Chlamydia trachomatis/pathogenicity , DNA, Bacterial/genetics , Gene Deletion , High-Throughput Nucleotide Sequencing , Humans , Mice , Mutation , Selection, Genetic , Sequence Analysis, DNA , Serial Passage
10.
Infect Immun ; 83(7): 2870-81, 2015 Jul.
Article En | MEDLINE | ID: mdl-25939505

Pathogenically diverse Chlamydia spp. can have surprisingly similar genomes. Chlamydia trachomatis isolates that cause trachoma, sexually transmitted genital tract infections (chlamydia), and invasive lymphogranuloma venereum (LGV) and the murine strain Chlamydia muridarum share 99% of their gene content. A region of high genomic diversity between Chlamydia spp. termed the plasticity zone (PZ) may encode niche-specific virulence determinants that dictate pathogenic diversity. We hypothesized that PZ genes might mediate the greater virulence and gamma interferon (IFN-γ) resistance of C. muridarum compared to C. trachomatis in the murine genital tract. To test this hypothesis, we isolated and characterized a series of C. muridarum PZ nonsense mutants. Strains with nonsense mutations in chlamydial cytotoxins, guaBA-add, and a phospholipase D homolog developed normally in cell culture. Two of the cytotoxin mutants were less cytotoxic than the wild type, suggesting that the cytotoxins may be functional. However, none of the PZ nonsense mutants exhibited increased IFN-γ sensitivity in cell culture or were profoundly attenuated in a murine genital tract infection model. Our results suggest that C. muridarum PZ genes are transcribed--and some may produce functional proteins--but are dispensable for infection of the murine genital tract.


Chlamydia Infections/microbiology , Chlamydia muridarum/genetics , Chlamydia muridarum/physiology , Genome, Bacterial , Virulence Factors/genetics , Animals , Chlamydia Infections/pathology , Chlamydia trachomatis/genetics , Chlamydia trachomatis/physiology , Codon, Nonsense , DNA Mutational Analysis , Female , Mice, Inbred C57BL , Virulence
11.
J Immunol ; 192(10): 4648-54, 2014 May 15.
Article En | MEDLINE | ID: mdl-24711617

Trachoma, caused by the obligate intracellular organism Chlamydia trachomatis, is the world's leading cause of preventable blindness for which a vaccine is needed. We have previously shown that a plasmid-deficient live-attenuated trachoma vaccine delivered ocularly to macaques elicited either solid or partial protective immunity against a virulent ocular challenge. Solidly protected macaques shared the same MHC class II alleles implicating CD4(+) T cells in superior protective immunity. Understandably, we sought to define T cell immune correlates in these animals to potentially improve vaccine efficacy. In this study, following a 2-y resting period, these macaques were boosted i.m. with the live-attenuated trachoma vaccine and their peripheral T cell anamnestic responses studied. Both solidly and partially protected macaques exhibited a CD4(+) and CD8(+) T cell anamnestic response following booster immunization. CD8(+) but not CD4(+) T cells from solidly protected macaques proliferated against soluble chlamydial Ag. We observed a more rapid T cell inflammatory cytokine response in tears of solidly protected animals following ocular rechallenge. Most notably, depletion of CD8(+) T cells in solidly protected macaques completely abrogated protective immunity. Collectively, our findings support the conclusion that CD8(+) T cells play an important but unexpected role in live-attenuated trachoma vaccine-mediated protective immunity.


Bacterial Vaccines/pharmacology , CD8-Positive T-Lymphocytes/immunology , Chlamydia trachomatis/immunology , Trachoma/prevention & control , Animals , Bacterial Vaccines/immunology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/pathology , Cell Proliferation/drug effects , Female , Macaca nemestrina , Male , Trachoma/immunology , Trachoma/pathology , Vaccines, Attenuated/pharmacology
12.
Infect Immun ; 82(7): 2756-62, 2014 Jul.
Article En | MEDLINE | ID: mdl-24733093

Chlamydia trachomatis is an obligate intracellular mucosotropic pathogen of significant medical importance. It is the etiological agent of blinding trachoma and bacterial sexually transmitted diseases, infections that afflict hundreds of millions of people globally. The C. trachomatis polymorphic membrane protein D (PmpD) is a highly conserved autotransporter and the target of broadly cross-reactive neutralizing antibodies; however, its role in host-pathogen interactions is unknown. Here we employed a targeted reverse genetics approach to generate a pmpD null mutant that was used to define the role of PmpD in the pathogenesis of chlamydial infection. We show that pmpD is not an essential chlamydial gene and the pmpD null mutant has no detectable deficiency in cultured murine cells or in a murine mucosal infection model. Notably, however, the pmpD null mutant was significantly attenuated for macaque eyes and cultured human cells. A reduction in pmpD null infection of human endocervical cells was associated with a deficiency in chlamydial attachment to cells. Collectively, our results show that PmpD is a chlamydial virulence factor that functions in early host-cell interactions. This study is the first of its kind using reverse genetics to evaluate the contribution of a C. trachomatis gene to disease pathogenesis.


Bacterial Proteins/metabolism , Chlamydia Infections/microbiology , Chlamydia trachomatis/metabolism , Membrane Proteins/metabolism , Virulence Factors/metabolism , Animals , Bacterial Proteins/genetics , Cell Line , Female , Gene Expression Regulation, Bacterial , Host-Pathogen Interactions , Humans , Macaca fascicularis , Male , Membrane Proteins/genetics , Mice , Mice, Inbred C3H , Mutation
13.
PLoS Negl Trop Dis ; 7(5): e2248, 2013.
Article En | MEDLINE | ID: mdl-23738030

BACKGROUND: Chlamydia trachomatis is the etiological agent of trachoma the world's leading cause of infectious blindness. Here, we investigate whether protracted clearance of a primary infection in nonhuman primates is attributable to antigenic variation or related to the maturation of the anti-chlamydial humoral immune response specific to chlamydial antigens. METHODOLOGY/PRINCIPAL FINDINGS: Genomic sequencing of organisms isolated throughout the protracted primary infection revealed that antigenic variation was not related to the inability of monkeys to efficiently resolve their infection. To explore the maturation of the humoral immune response as a possible reason for delayed clearance, sera were analyzed by radioimmunoprecipitation using intrinsically radio-labeled antigens prepared under non-denaturing conditions. Antibody recognition was restricted to the antigenically variable major outer membrane protein (MOMP) and a few antigenically conserved antigens. Recognition of MOMP occurred early post-infection and correlated with reduction in infectious ocular burdens but not with infection eradication. In contrast, antibody recognition of conserved antigens, identified as PmpD, Hsp60, CPAF and Pgp3, appeared late and correlated with infection eradication. Partial immunity to re-challenge was associated with a discernible antibody recall response against all antigens. Antibody recognition of PmpD and CPAF was destroyed by heat treatment while MOMP and Pgp3 were partially affected, indicating that antibody specific to conformational epitopes on these proteins may be important to protective immunity. CONCLUSIONS/SIGNIFICANCE: Our findings suggest that delayed clearance of chlamydial infection in NHP is not the result of antigenic variation but rather a consequence of the gradual maturation of the C. trachomatis antigen-specific humoral immune response. However, we cannot conclude that antibodies specific for these proteins play the primary role in host protective immunity as they could be surrogate markers of T cell immunity. Collectively, our results argue that an efficacious subunit trachoma vaccine might require a combination of these antigens delivered in their native conformation.


Antibodies, Bacterial/blood , Chlamydia trachomatis/immunology , Secondary Prevention , Trachoma/immunology , Trachoma/prevention & control , Animals , Antigenic Variation , Chlamydia trachomatis/genetics , Chlamydia trachomatis/isolation & purification , Disease Models, Animal , Macaca fascicularis , Male , Radioimmunoprecipitation Assay , Serum/immunology
14.
Infect Immun ; 81(3): 636-44, 2013 Mar.
Article En | MEDLINE | ID: mdl-23319558

Chlamydia trachomatis causes chronic inflammatory diseases of the eye and genital tract and has global medical importance. The chlamydial plasmid plays an important role in the pathophysiology of these diseases, as plasmid-deficient organisms are highly attenuated. The cryptic plasmid carries noncoding RNAs and eight conserved open reading frames (ORFs). To understand plasmid gene function, we generated plasmid shuttle vectors with deletions in each of the eight ORFs. The individual deletion mutants were used to transform chlamydiae and the transformants were characterized phenotypically and at the transcriptional level. We show that pgp1, -2, -6, and -8 are essential for plasmid maintenance, while the other ORFs can be deleted and the plasmid stably maintained. We further show that a pgp4 knockout mutant exhibits an in vitro phenotype similar to its isogenic plasmidless strain, in terms of abnormal inclusion morphology and lack of glycogen accumulation. Microarray and qRT-PCR analysis revealed that Pgp4 is a transcriptional regulator of plasmid-encoded pgp3 and multiple chromosomal genes, including the glycogen synthase gene glgA, that are likely important in chlamydial virulence. Our findings have major implications for understanding the plasmid's role in chlamydial pathogenesis at the molecular level.


Bacterial Proteins/metabolism , Chlamydia trachomatis/metabolism , Gene Expression Regulation, Bacterial/physiology , Plasmids/metabolism , Transcription, Genetic/physiology , Animals , Bacterial Proteins/genetics , Cell Line , Chlamydia trachomatis/cytology , Chlamydia trachomatis/genetics , Chromosomes, Bacterial , Gene Deletion , Mice , Plasmids/genetics , Protein Array Analysis , Reverse Transcriptase Polymerase Chain Reaction , Virulence
15.
J Exp Med ; 208(11): 2217-23, 2011 Oct 24.
Article En | MEDLINE | ID: mdl-21987657

Blinding trachoma is an ancient neglected tropical disease caused by Chlamydia trachomatis for which a vaccine is needed. We describe a live-attenuated vaccine that is safe and efficacious in preventing trachoma in nonhuman primates, a model with excellent predictive value for humans. Cynomolgus macaques infected ocularly with a trachoma strain deficient for the 7.5-kb conserved plasmid presented with short-lived infections that resolved spontaneously without ocular pathology. Multiple infections with the attenuated plasmid-deficient strain produced no inflammatory ocular pathology but induced an anti-chlamydial immune response. Macaques vaccinated with the attenuated strain were either solidly or partially protected after challenge with virulent plasmid-bearing organisms. Partially protected macaques shed markedly less infectious organisms than controls. Immune correlates of protective immunity were not identified, but we did detect a correlation between MHC class II alleles and solid versus partial protection. Epidemiological models of trachoma control indicate that a vaccine with this degree of efficacy would significantly reduce the prevalence of infection and rates of reinfection, known risk factors which drive blinding disease.


Chlamydia trachomatis/immunology , Chlamydia trachomatis/pathogenicity , Trachoma/microbiology , Trachoma/prevention & control , Vaccines, Attenuated/therapeutic use , Animals , Chlamydia trachomatis/genetics , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/immunology , Humans , Macaca fascicularis/genetics , Macaca fascicularis/immunology , Major Histocompatibility Complex , Trachoma/epidemiology
16.
Proc Natl Acad Sci U S A ; 108(17): 7189-93, 2011 Apr 26.
Article En | MEDLINE | ID: mdl-21482792

Chlamydia trachomatis is an obligate intracellular bacterial pathogen that infects hundreds of millions of individuals globally, causing blinding trachoma and sexually transmitted disease. More effective chlamydial control measures are needed, but progress toward this end has been severely hampered by the lack of a tenable chlamydial genetic system. Here, we describe a reverse-genetic approach to create isogenic C. trachomatis mutants. C. trachomatis was subjected to low-level ethyl methanesulfonate mutagenesis to generate chlamydiae that contained less then one mutation per genome. Mutagenized organisms were expanded in small subpopulations that were screened for mutations by digesting denatured and reannealed PCR amplicons of the target gene with the mismatch specific endonuclease CEL I. Subpopulations with mutations were then sequenced for the target region and plaque-cloned if the desired mutation was detected. We demonstrate the utility of this approach by isolating a tryptophan synthase gene (trpB) null mutant that was otherwise isogenic to its parental clone as shown by de novo genome sequencing. The mutant was incapable of avoiding the anti-microbial effect of IFN-γ-induced tryptophan starvation. The ability to genetically manipulate chlamydiae is a major advancement that will enhance our understanding of chlamydial pathogenesis and accelerate the development of new anti-chlamydial therapeutic control measures. Additionally, this strategy could be applied to other medically important bacterial pathogens with no or difficult genetic systems.


Chlamydia trachomatis/genetics , Mutagenesis , Mutation , Tryptophan Synthase/genetics , Antineoplastic Agents, Alkylating/pharmacology , Chlamydia Infections/enzymology , Chlamydia Infections/genetics , Chlamydia trachomatis/enzymology , Ethyl Methanesulfonate/pharmacology , Humans , Tryptophan Synthase/metabolism
17.
Infect Immun ; 78(9): 3660-8, 2010 Sep.
Article En | MEDLINE | ID: mdl-20547745

Chlamydia trachomatis is a human pathogen of global importance. An obstacle to studying the pathophysiology of human chlamydial disease is the lack of a suitable murine model of C. trachomatis infection. Mice are less susceptible to infection with human isolates due in part to innate mouse-specific host defense mechanisms to which human strains are sensitive. Another possible factor that influences the susceptibility of mice to infection is that human isolates are commonly cultivated in vitro prior to infection of mice; therefore, virulence genes could be lost as a consequence of negative selective pressure. We tested this hypothesis by infecting innate immunity-deficient C3H/HeJ female mice intravaginally with a human serovar D urogenital isolate that had undergone multiple in vitro passages. We observed early and late infection clearance phenotypes. Strains of each phenotype were isolated and then used to reinfect naïve mice. Following infection, the late-clearance strain was significantly more virulent. It caused unvarying infections of much longer durations with greater infectious burdens that naturally ascended to the upper genital tract, causing salpingitis. Despite contrasting in vivo virulence characteristics, the strains exhibited no differences in the results of in vitro infectivity assays or sensitivities to gamma interferon. Genome sequencing of the strains revealed mutations that localized to a single gene (CT135), implicating it as a critical virulence factor. Mutations in CT135 were not unique to serovar D but were also found in multiple oculogenital reference strains. Our findings provide new information about the pathogenomics of chlamydial infection and insights for improving murine models of infection using human strains.


Chlamydia Infections/etiology , Chlamydia trachomatis/pathogenicity , Frameshift Mutation , Genital Diseases, Female/etiology , Virulence Factors/genetics , Animals , Base Sequence , Chlamydia Infections/pathology , Chlamydia trachomatis/genetics , Female , Genital Diseases, Female/pathology , Mice , Mice, Inbred C3H , Molecular Sequence Data , Phenotype , Polymorphism, Genetic
18.
J Immunol ; 182(12): 8063-70, 2009 Jun 15.
Article En | MEDLINE | ID: mdl-19494332

A vaccine is likely the most effective strategy for controlling human chlamydial infections. Recent studies have shown immunization with Chlamydia muridarum major outer membrane protein (MOMP) can induce significant protection against infection and disease in mice if its native trimeric structure is preserved (nMOMP). The objective of this study was to investigate the immunogenicity and vaccine efficacy of Chlamydia trachomatis nMOMP in a nonhuman primate trachoma model. Cynomolgus monkeys (Macaca fascicularis) were immunized systemically with nMOMP, and monkeys were challenged ocularly. Immunization induced high serum IgG and IgA ELISA Ab titers, with Abs displaying high strain-specific neutralizing activity. The PBMCs of immunized monkeys produced a broadly cross-reactive, Ag-specific IFN-gamma response equivalent to that induced by experimental infection. Immunized monkeys exhibited a significant decrease in infectious burden during the early peak shedding periods (days 3-14). However, at later time points, they exhibited no difference from control animals in either burden or duration of infection. Immunization had no effect on the progression of ocular disease. These results show that systemically administered nMOMP is highly immunogenic in nonhuman primates and elicits partially protective immunity against ocular chlamydial challenge. This is the first time a subunit vaccine has shown a significant reduction in ocular shedding in nonhuman primates. A partially protective vaccine, particularly one that reduces infectious burden after primary infection of children, could interrupt the natural trachoma reinfection cycle. This would have a beneficial effect on the transmission between children and sensitized adults which drives blinding inflammatory disease.


Bacterial Outer Membrane Proteins/immunology , Bacterial Vaccines/immunology , Chlamydia Infections/immunology , Chlamydia Infections/prevention & control , Chlamydia trachomatis/immunology , Macaca fascicularis/immunology , Animals , Antibodies, Bacterial/blood , Antibodies, Bacterial/immunology , Antibody Specificity , Chlamydia Infections/pathology , Chlamydia Infections/transmission , Cytokines/immunology , Cytokines/metabolism , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , HeLa Cells , Humans , Kinetics , Leukocytes/immunology , Leukocytes/metabolism , Male , Protein Denaturation , Titrimetry
19.
Infect Immun ; 76(6): 2273-83, 2008 Jun.
Article En | MEDLINE | ID: mdl-18347045

Chlamydia trachomatis possesses a cryptic 7.5-kb plasmid of unknown function. Here, we describe a comprehensive molecular and biological characterization of the naturally occurring plasmidless human C. trachomatis strain L2(25667R). We found that despite minimal chromosomal polymorphisms, the LGV strain L2(25667R) was indistinguishable from plasmid-positive strain L2(434) with regard to its in vitro infectivity characteristics such as growth kinetics, plaquing efficiency, and plaque size. The only in vitro phenotypic differences between L2(434) and L2(25667R) were the accumulation of glycogen granules in the inclusion matrix and the lack of the typical intrainclusion Brownian-like movement characteristic of C. trachomatis strains. Conversely, we observed a marked difference between the two strains in their abilities to colonize and infect the female mouse genital tract. The 50% infective dose of plasmidless strain L2(25667R) was 400-fold greater (4 x 10(6) inclusion-forming units [IFU]) than that of plasmid-bearing strain L2(434) (1 x 10(4) IFU). Transcriptome analysis of the two strains demonstrated a decrease in the transcript levels of a subset of chromosomal genes for strain L2(25667R). Among those genes was glgA, encoding glycogen synthase, a finding consistent with the failure of L2(25667R) to accumulate glycogen granules. These findings support a primary role for the plasmid in in vivo infectivity and suggest that virulence is controlled, at least in part, by the plasmid's ability to regulate the expression of chromosomal genes. Our findings have important implications in understanding a role for the plasmid in the pathogenesis of human infection and disease.


Chlamydia trachomatis/physiology , Chromosomes, Bacterial/genetics , Plasmids/physiology , Transcription, Genetic/physiology , Virulence Factors/physiology , Animals , Bacteriological Techniques , Chlamydia Infections/microbiology , Chlamydia trachomatis/classification , Chlamydia trachomatis/cytology , Chlamydia trachomatis/genetics , Female , Gene Expression Profiling , Gene Expression Regulation, Bacterial/physiology , Mice , Mice, Inbred C3H , Plasmids/genetics , Polymorphism, Genetic , Protein Array Analysis , Vaginosis, Bacterial/microbiology , Virulence Factors/genetics
20.
J Infect Dis ; 197(3): 449-56, 2008 Feb 01.
Article En | MEDLINE | ID: mdl-18199030

Chlamydia trachomatis is the etiological agent of trachoma, the leading cause of preventable blindness. Trachoma presents distinct clinical syndromes ranging from mild and self-limiting to severe inflammatory disease. The underlying host and pathogen factors responsible for these diverse clinical outcomes are unclear. To assess the role played by pathogen variation in disease outcome, we analyzed the genomes of 4 trachoma strains representative of the 3 major trachoma serotypes, using microarray-based comparative genome sequencing. Outside of ompA, trachoma strains differed primarily in a very small subset of genes (n = 22). These subtle genetic variations were manifested in profound differences in virulence as measured by in vitro growth rate, burst size, plaque morphology, and interferon-gamma sensitivity but most importantly in virulence as shown by ocular infection of nonhuman primates. Our findings are the first to identify genes that correlate with differences in pathogenicity among trachoma strains.


Chlamydia trachomatis/genetics , Genetic Variation , Genome, Bacterial , Primates/microbiology , Animals , Chlamydia trachomatis/isolation & purification , Chlamydia trachomatis/pathogenicity , HeLa Cells , Humans , Macaca fascicularis , Male , Polymorphism, Genetic , Trachoma/microbiology
...