Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Pathog Dis ; 812023 Jan 17.
Artigo em Inglês | MEDLINE | ID: mdl-37804183

RESUMO

Chlamydia trachomatis is an obligate intracellular bacterium that causes blinding trachoma and sexually transmitted disease. The chlamydial plasmid is a critical virulence factor in the pathogenesis of these diseases. Plasmid gene protein 4 (Pgp4) plays a major role in chlamydial virulence by regulating the expression of both chromosomal genes and Pgp3. Despite the importance of Pgp4 in mediating lytic exit from host cells the pathogenic mechanism by which it functions is unknown. CT084 is a highly conserved chromosomal gene with homology to phospholipase D. We showed CT084 expression is regulated by Pgp4 and expressed late in the chlamydial developmental cycle. To investigate the function of CT084 in chlamydial lytic exit from infected cells, we made a CT084 null strain (ct084::bla) by using Targetron. The ct084::bla strain grew normally in vitro compared to wild-type strain; however, the strain did not lyse infected cells and produced significantly less and smaller plaques. Collectively, our finding shows Pgp4-regulated CT084-mediated chlamydia lytic exit from infected host cells.


Assuntos
Infecções por Chlamydia , Tracoma , Humanos , Chlamydia trachomatis/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Plasmídeos/genética , Fenótipo , Infecções por Chlamydia/microbiologia
2.
Infect Immun ; 91(7): e0009623, 2023 07 18.
Artigo em Inglês | MEDLINE | ID: mdl-37255490

RESUMO

All members of the family Chlamydiaceae have lipopolysaccharides (LPS) that possess a shared carbohydrate trisaccharide antigen, 3-deoxy-d-manno-oct-2-ulosonic acid (Kdo) that is functionally uncharacterized. A single gene, genus-specific epitope (gseA), is responsible for attaching the tri-Kdo to lipid IVA. To investigate the function of Kdo in chlamydial host cell interactions, we made a gseA-null strain (L2ΔgseA) by using TargeTron mutagenesis. Immunofluorescence microscopy and immunoblotting with a Kdo-specific monoclonal antibody demonstrated that L2ΔgseA lacked Kdo. L2ΔgseA reacted by immunoblotting with a monoclonal antibody specific for a conserved LPS glucosamine-PO4 epitope, indicating that core lipid A was retained by the mutant. The mutant strain produced a similar number of inclusions as the parental strain but yielded lower numbers of infectious elementary bodies. Transmission electron microscopy of L2ΔgseA-infected cells showed atypical developmental forms and a reduction in the number of elementary bodies. Immunoblotting of dithiothreitol-treated L2ΔgseA-infected cells lysates revealed a marked reduction in outer membrane OmcB disulfide cross-linking, suggesting that the elementary body outer membrane structure was affected by the lack of Kdo. Notably, lactic acid dehydrogenase release by infected cells demonstrated that L2ΔgseA was significantly more cytotoxic to host cells than the wild type. The cytotoxic phenotype may result from an altered outer membrane biogenesis structure and/or function or, conversely, from a direct pathobiological effect of Kdo on an unknown host cell target. These findings implicate a previously unrecognized role for Kdo in host cell interactions that facilitates postinfection host cell survival.


Assuntos
Chlamydia trachomatis , Lipopolissacarídeos , Lipopolissacarídeos/metabolismo , Sequência de Carboidratos , Epitopos , Açúcares Ácidos , Anticorpos Monoclonais
3.
Nat Commun ; 12(1): 5454, 2021 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-34526512

RESUMO

Chlamydia trachomatis infection causes severe inflammatory disease resulting in blindness and infertility. The pathophysiology of these diseases remains elusive but myeloid cell-associated inflammation has been implicated. Here we show NLRP3 inflammasome activation is essential for driving a macrophage-associated endometritis resulting in infertility by using a female mouse genital tract chlamydial infection model. We find the chlamydial parasitophorous vacuole protein CT135 triggers NLRP3 inflammasome activation via TLR2/MyD88 signaling as a pathogenic strategy to evade neutrophil host defense. Paradoxically, a consequence of CT135 mediated neutrophil killing results in a submucosal macrophage-associated endometritis driven by ATP/P2X7R induced NLRP3 inflammasome activation. Importantly, macrophage-associated immunopathology occurs independent of macrophage infection. We show chlamydial infection of neutrophils and epithelial cells produce elevated levels of extracellular ATP. We propose this source of ATP serves as a DAMP to activate submucosal macrophage NLRP3 inflammasome that drive damaging immunopathology. These findings offer a paradigm of sterile inflammation in infectious disease pathogenesis.


Assuntos
Infecções por Chlamydia/imunologia , Chlamydia/imunologia , Inflamação/imunologia , Células Mieloides/imunologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/imunologia , Neutrófilos/imunologia , Receptores Purinérgicos P2X7/imunologia , Trifosfato de Adenosina/imunologia , Trifosfato de Adenosina/metabolismo , Animais , Células Cultivadas , Chlamydia/fisiologia , Infecções por Chlamydia/metabolismo , Infecções por Chlamydia/microbiologia , Modelos Animais de Doenças , Feminino , Células HeLa , Interações Hospedeiro-Patógeno/imunologia , Humanos , Evasão da Resposta Imune/imunologia , Inflamação/metabolismo , Inflamação/microbiologia , Macrófagos/imunologia , Macrófagos/metabolismo , Macrófagos/microbiologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células Mieloides/metabolismo , Células Mieloides/microbiologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Neutrófilos/metabolismo , Neutrófilos/microbiologia , Receptores Purinérgicos P2X7/genética , Receptores Purinérgicos P2X7/metabolismo
4.
mBio ; 12(3): e0117921, 2021 06 29.
Artigo em Inglês | MEDLINE | ID: mdl-34101486

RESUMO

Chlamydia are obligate intracellular Gram-negative bacteria distinguished by a unique developmental biology confined within a parasitophorous vacuole termed an inclusion. The chlamydial plasmid is a central virulence factor in the pathogenesis of infection. Plasmid gene protein 4 (Pgp4) regulates the expression of plasmid gene protein 3 (Pgp3) and chromosomal glycogen synthase (GlgA), virulence factors secreted from the inclusion to the host cytosol by an unknown mechanism. Here, we identified a plasmid-dependent secretion system for the cytosolic delivery of Pgp3 and GlgA. The secretion system consisted of a segregated population of globular structures originating from midcycle reticulate bodies. Globular structures contained the Pgp4-regulated proteins CT143, CT144, and CT050 in addition to Pgp3 and GlgA. Genetic replacement of Pgp4 with Pgp3 or GlgA negated the formation of globular structures, resulting in retention of Pgp3 and GlgA in chlamydial organisms. The generation of globular structures and secretion of virulence factors occurred independently of type 2 and type 3 secretion systems. Globular structures were enriched with lipopolysaccharide but lacked detectable major outer membrane protein and heat shock protein 60, implicating them as outer membrane vesicles. Thus, we have discovered a novel chlamydial plasmid-dependent secretion system that transports virulence factor cargo from the chlamydial inclusion to the host cytosol. IMPORTANCE The Chlamydia trachomatis plasmid regulates the expression and secretion of immune evasion virulence factors to the host cytosol by an unknown mechanism. In this study, we identified a novel plasmid gene protein 4 (Pgp4)-dependent secretion system. The system consists of globular structures distinct from typical chlamydial developmental forms that export Pgp3 and GlgA to the host cytosol. Globular structures emerged at mid-chlamydial growth cycle from distinct populations of reticulate bodies. The formation of globular structures occurred independently of known chlamydial secretion systems. These results identify a Pgp4-dependent secretory system required for exporting plasmid regulated virulence factors to the host cytosol.


Assuntos
Sistemas de Secreção Bacterianos/genética , Chlamydia trachomatis/genética , Citosol/metabolismo , Plasmídeos/genética , Fatores de Virulência/genética , Antígenos de Bactérias/genética , Sistemas de Secreção Bacterianos/metabolismo , Linhagem Celular , Técnicas de Transferência de Genes , Humanos
5.
mBio ; 11(4)2020 08 18.
Artigo em Inglês | MEDLINE | ID: mdl-32817110

RESUMO

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.


Assuntos
Antígenos de Bactérias/genética , Proteínas de Bactérias/genética , Infecções por Chlamydia/imunologia , Chlamydia trachomatis/genética , Chlamydia trachomatis/patogenicidade , Fatores de Virulência/genética , Animais , Antígenos de Bactérias/metabolismo , Proteínas de Bactérias/metabolismo , Citocinas/imunologia , Células Epiteliais/microbiologia , Feminino , Células HeLa , Humanos , Macaca , Camundongos , Camundongos Endogâmicos C57BL
6.
mBio ; 10(2)2019 04 23.
Artigo em Inglês | MEDLINE | ID: mdl-31015326

RESUMO

Chlamydia trachomatis is the most common bacterial cause of sexually transmitted infections. C. trachomatis sexually transmitted infections are commonly asymptomatic, implying a pathogenic strategy for the evasion of innate inflammatory immune responses, a paradox as the C. trachomatis outer membrane contains lipopolysaccharide (LPS), a known potent agonist of inflammatory innate immunity. Here, we studied the ability of chlamydial LPS to activate the proinflammatory canonical and noncanonical inflammasome pathways in mouse bone marrow-derived macrophages (BMDM). We show, in comparison to Escherichiacoli LPS, that C. trachomatis LPS-treated BMDM produce significantly less IL-6, TNF, and type I interferon mRNA, indicating that downstream signaling through the canonical TLR4 myddosome and triffosome pathways was blocked. We confirmed this in C. trachomatis LPS-treated BMDM by showing the lack of NF-κB and IRF3 phosphorylation, respectively. Interestingly, C. trachomatis LPS bound CD14 and promoted its endocytosis; however; it did not promote efficient TLR4/MD-2 dimerization or endocytosis, known requirements for myddosome and triffosome signaling pathways. We further found that transfection of BMDM with C. trachomatis LPS did not cause pyroptotic cell ballooning, cytotoxicity, or IL-1ß secretion, all characteristic features of noncanonical inflammasome activation. Western blotting confirmed that cytosolic C. trachomatis LPS failed to signal through caspase-11, as shown by the lack of gasdermin D, caspase-1, or IL-1ß proteolytic cleavage. We propose that chlamydiae evolved a unique LPS structure as a pathogenic strategy to avoid canonical and noncanonical innate immune signaling and conclude that this strategy might explain the high incidence of asymptomatic infections.IMPORTANCEChlamydia trachomatis is the most common bacterial cause of sexually transmitted infections (STI). C. trachomatis STI are commonly asymptomatic, implying a pathogenic strategy for the evasion of innate inflammatory immune responses, a paradox as the C. trachomatis outer membrane contains lipopolysaccharide (LPS), a known potent agonist of inflammatory innate immunity. Here, we found that C. trachomatis LPS is not capable of engaging the canonical TLR4/MD-2 or noncanonical caspase-11 inflammatory pathways. The inability of C. trachomatis LPS to trigger innate immunity inflammatory pathways is related to its unique fatty acid structure. Evolutionary modification of the LPS structure likely evolved as a pathogenic strategy to silence innate host defense mechanisms. The findings might explain the high incidence of asymptomatic chlamydial genital infection.


Assuntos
Chlamydia trachomatis/imunologia , Chlamydia trachomatis/patogenicidade , Evasão da Resposta Imune , Imunidade Inata , Lipopolissacarídeos/metabolismo , Fatores de Virulência/metabolismo , Animais , Citocinas/biossíntese , Escherichia coli/imunologia , Escherichia coli/patogenicidade , Perfilação da Expressão Gênica , Macrófagos/imunologia , Camundongos Endogâmicos C57BL
7.
mBio ; 9(1)2018 01 30.
Artigo em Inglês | MEDLINE | ID: mdl-29382731

RESUMO

Chlamydia trachomatis is an obligate intracellular bacterial pathogen that causes blinding trachoma and sexually transmitted disease. C. trachomatis isolates are classified into 2 biovars-lymphogranuloma venereum (LGV) and trachoma-which are distinguished biologically by their natural host cell infection tropism. LGV biovars infect macrophages and are invasive, whereas trachoma biovars infect oculo-urogenital epithelial cells and are noninvasive. The C. trachomatis plasmid is an important virulence factor in the pathogenesis of these infections. Central to its pathogenic role is the transcriptional regulatory function of the plasmid protein Pgp4, which regulates the expression of plasmid and chromosomal virulence genes. As many gene regulatory functions are post-transcriptional, we employed a comparative proteomic study of cells infected with plasmid-cured C. trachomatis serovars A and D (trachoma biovar), a L2 serovar (LGV biovar), and the L2 serovar transformed with a plasmid containing a nonsense mutation in pgp4 to more completely elucidate the effects of the plasmid on chlamydial infection biology. Our results show that the Pgp4-dependent elevations in the levels of Pgp3 and a conserved core set of chromosomally encoded proteins are remarkably similar for serovars within both C. trachomatis biovars. Conversely, we found a plasmid-dependent, Pgp4-independent, negative regulation in the expression of the chlamydial protease-like activity factor (CPAF) for the L2 serovar but not the A and D serovars. The molecular mechanism of plasmid-dependent negative regulation of CPAF expression in the LGV serovar is not understood but is likely important to understanding its macrophage infection tropism and invasive infection nature.IMPORTANCE The Chlamydia trachomatis plasmid is an important virulence factor in the pathogenesis of chlamydial infection. It is known that plasmid protein 4 (Pgp4) functions in the transcriptional regulation of the plasmid virulence protein 3 (Pgp3) and multiple chromosomal loci of unknown function. Since many gene regulatory functions can be post-transcriptional, we undertook a comparative proteomic analysis to better understand the plasmid's role in chlamydial and host protein expression. We report that Pgp4 is a potent and specific master positive regulator of a common core of plasmid and chromosomal virulence genes shared by multiple C. trachomatis serovars. Notably, we show that the plasmid is a negative regulator of the expression of the chlamydial virulence factor CPAF. The plasmid regulation of CPAF is independent of Pgp4 and restricted to a C. trachomatis macrophage-tropic strain. These findings are important because they define a previously unknown role for the plasmid in the pathophysiology of invasive chlamydial infection.


Assuntos
Chlamydia trachomatis/genética , Chlamydia trachomatis/metabolismo , Endopeptidases/biossíntese , Regulação Bacteriana da Expressão Gênica , Plasmídeos , Fatores de Transcrição/metabolismo , Chlamydia trachomatis/química , Células Epiteliais/microbiologia , Células HeLa , Humanos , Proteoma/análise
8.
Pathog Dis ; 75(3)2017 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-28369275

RESUMO

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.


Assuntos
Proteínas de Bactérias/metabolismo , Infecções por Chlamydia/genética , Infecções por Chlamydia/microbiologia , Chlamydia trachomatis/metabolismo , Interações Hospedeiro-Patógeno , Fatores de Transcrição/metabolismo , Alelos , Animais , Proteínas de Bactérias/genética , Linhagem Celular , Chlamydia trachomatis/genética , Chlamydia trachomatis/crescimento & desenvolvimento , Modelos Animais de Doenças , Feminino , Expressão Gênica , Regulação da Expressão Gênica , Genoma Bacteriano , Humanos , Camundongos , Mutação , Análise de Sequência de DNA , Fatores de Transcrição/genética , Fatores de Virulência/genética , Fatores de Virulência/metabolismo
9.
mBio ; 7(5)2016 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-27677792

RESUMO

The chlamydial protease-like activity factor (CPAF) is hypothesized to be an important secreted virulence factor; however, challenges in denaturing its proteolytic activity have hampered attempts to identify its legitimate targets. Here, we use a genetic and proteomic approach to identify authentic CPAF targets. Human epithelial cells infected with CPAF-sufficient and CPAF-deficient chlamydiae were lysed using known CPAF-denaturing conditions. Their protein profiles were analyzed using isobaric mass tags and liquid chromatography-tandem mass spectrometry. Comparative analysis of CPAF-sufficient and CPAF-deficient infections identified a limited number of CPAF host and chlamydial protein targets. Host targets were primarily interferon-stimulated gene products, whereas chlamydial targets were type III secreted proteins. We provide evidence supporting a cooperative role for CPAF and type III secreted effectors in blocking NF-κB p65 nuclear translocation, resulting in decreased beta interferon and proinflammatory cytokine synthesis. Genetic complementation of null organisms with CPAF restored p65 nuclear translocation inhibition and proteolysis of chlamydial type III secreted effector proteins (T3SEs). We propose that CPAF and T3SEs cooperate in the inhibition of host innate immunity. IMPORTANCE: Chlamydia trachomatis is an important human pathogen responsible for over 100 million infections each year worldwide. Its success as an intracellular pathogen revolves around its ability to evade host immunity. The chlamydial protease-like activity factor (CPAF) is a conserved serine protease secreted into the host cytosol of infected cells that is thought to play an important role in immune evasion. Currently, CPAF's authentic in situ target(s) and mechanism of action in immune evasion are poorly characterized. Using a CPAF-deficient strain and high-throughput proteomics, we report novel CPAF host and chlamydial targets. Host targets were primarily interferon-stimulated genes, whereas chlamydial targets were exclusively type III secreted proteins. We propose a novel mechanism for CPAF and type III secreted proteins in the evasion of host innate immune responses. These findings provide new insights into CPAF's function as a virulence factor and a better understanding of how chlamydiae evade host immunity.

10.
Pathog Dis ; 73(6): ftv043, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26109550

RESUMO

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.


Assuntos
Chlamydia trachomatis/genética , Variação Genética , Instabilidade Genômica , Fatores de Virulência/genética , Animais , Infecções por Chlamydia/microbiologia , Chlamydia trachomatis/isolamento & purificação , Chlamydia trachomatis/patogenicidade , DNA Bacteriano/genética , Deleção de Genes , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Camundongos , Mutação , Seleção Genética , Análise de Sequência de DNA , Inoculações Seriadas
11.
Pathog Dis ; 70(2): 189-93, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24214488

RESUMO

Chlamydia trachomatis and C. muridarum are human and mouse pathogens, respectively, which show high conservation of gene order and content. Both species contain a common 7.5-kb plasmid that is an important virulence factor. Recently described transformation systems have been used to characterize C. trachomatis L2 plasmid gene functions; however, similar studies have not been reported for C. trachomatis ocular tropic serovar A or the mouse strain, C. muridarum. Here, we have conducted genetic experiments with C. trachomatis serovar A and C. muridarum and report the following: (1) successful transformation of C. muridarum and C. trachomatis serovar A is restricted to a shuttle vector with a C. muridarum or C. trachomatis serovar A plasmid backbone, respectively; (2) transformation of plasmid-deficient C. muridarum with the C. muridarum-based shuttle vector complement glycogen accumulation and inclusion morphology; and (3) C. muridarum plasmid-encoded Pgp4 is a regulator of chromosomal (glgA) and plasmid (pgp3) virulence genes. In summary, our findings show a previously unrecognized and unexpected role for the chlamydial plasmid in its transformation tropism and confirm the plasmids regulatory role of virulence genes in C. muridarum.


Assuntos
Chlamydia muridarum/genética , Chlamydia trachomatis/genética , Transferência Genética Horizontal , Especificidade de Hospedeiro , Plasmídeos , Transformação Bacteriana , Animais , Regulação Bacteriana da Expressão Gênica , Vetores Genéticos , Humanos , Camundongos , Fatores de Virulência/biossíntese
12.
Infect Immun ; 81(3): 636-44, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23319558

RESUMO

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.


Assuntos
Proteínas de Bactérias/metabolismo , Chlamydia trachomatis/metabolismo , Regulação Bacteriana da Expressão Gênica/fisiologia , Plasmídeos/metabolismo , Transcrição Gênica/fisiologia , Animais , Proteínas de Bactérias/genética , Linhagem Celular , Chlamydia trachomatis/citologia , Chlamydia trachomatis/genética , Cromossomos Bacterianos , Deleção de Genes , Camundongos , Plasmídeos/genética , Análise Serial de Proteínas , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Virulência
13.
Proc Natl Acad Sci U S A ; 108(17): 7189-93, 2011 Apr 26.
Artigo em Inglês | MEDLINE | ID: mdl-21482792

RESUMO

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.


Assuntos
Chlamydia trachomatis/genética , Mutagênese , Mutação , Triptofano Sintase/genética , Antineoplásicos Alquilantes/farmacologia , Infecções por Chlamydia/enzimologia , Infecções por Chlamydia/genética , Chlamydia trachomatis/enzimologia , Metanossulfonato de Etila/farmacologia , Humanos , Triptofano Sintase/metabolismo
14.
Infect Immun ; 78(9): 3678-88, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20605986

RESUMO

Chlamydia trachomatis is the leading cause of infectious blindness worldwide and is the most commonly reported pathogen causing sexually transmitted infections. Tarp (translocated actin recruiting phosphoprotein), a type III secreted effector that mediates actin nucleation, is central to C. trachomatis infection. The phylogenetic analysis of tarP from reference strains as well as ocular, genital, and lymphogranuloma venereum (LGV) clinical isolates demonstrated an evolutionary relationship with disease phenotype, with LGV and ocular isolates branched into clades that were separate from the urogenital isolates. The sequence analysis of Tarp indicated a high degree of variability and identified trends within clinical groupings. Tarps from LGV strains contained the highest number of tyrosine-rich repeat regions (up to nine) and the fewest (two) predicted actin binding domains. The converse was noted for Tarp proteins from ocular isolates that contained up to four actin binding domains and as few as one tyrosine-rich repeat region. The results suggest that Tarp is among the few known genes to play a role in C. trachomatis adaptations to specific niches within the host.


Assuntos
Actinas/metabolismo , Proteínas de Bactérias/genética , Chlamydia trachomatis/classificação , Proteínas de Bactérias/química , Chlamydia trachomatis/genética , Feminino , Humanos , Masculino , Fenótipo , Filogenia , Polimorfismo de Nucleotídeo Único , Porinas/genética , Estrutura Terciária de Proteína , Sequências Repetitivas de Aminoácidos
15.
J Infect Dis ; 197(3): 449-56, 2008 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-18199030

RESUMO

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.


Assuntos
Chlamydia trachomatis/genética , Variação Genética , Genoma Bacteriano , Primatas/microbiologia , Animais , Chlamydia trachomatis/isolamento & purificação , Chlamydia trachomatis/patogenicidade , Células HeLa , Humanos , Macaca fascicularis , Masculino , Polimorfismo Genético , Tracoma/microbiologia
16.
Biochim Biophys Acta ; 1774(10): 1254-63, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17827077

RESUMO

A recently discovered subgroup of class I ribonucleotide reductase (RNR) found in the infectious bacterium Chlamydia trachomatis (C. trachomatis) was shown to exhibit a high-valent Fe(III)Fe(IV) center instead of the tyrosyl radical observed normally in all class I RNRs. The X-ray structure showed that C. trachomatis WT RNR has a phenylalanine at the position of the active tyrosine in Escherichia coli RNR. In this paper the X-ray structure of variant F127Y is presented, where the tyrosine is restored. Using (1)H- and (57)Fe-ENDOR spectroscopy it is shown, that in WT and variants F127Y and Y129F of C. trachomatis RNR, the Fe(III)Fe(IV) center is virtually identical with the short-lived intermediate X observed during the iron oxygen reconstitution reaction in class I RNR from E. coli. The experimental data are consistent with a recent theoretical model for X, proposing two bridging oxo ligands and one terminal water ligand. A surprising extension of the lifetime of the Fe(III)Fe(IV) state in C. trachomatis from a few seconds to several hours at room temperature was observed under catalytic conditions in the presence of substrate. These findings suggest a possible new role for the Fe(III)Fe(IV) state also in other class I RNR, during the catalytic radical transfer reaction, by which the substrate turnover is started.


Assuntos
Chlamydia trachomatis/enzimologia , Compostos Férricos/química , Compostos Ferrosos/química , Ribonucleotídeo Redutases/química , Substituição de Aminoácidos/genética , Chlamydia trachomatis/genética , Cristalografia por Raios X , Espectroscopia de Ressonância de Spin Eletrônica , Mutação Puntual , Ribonucleotídeo Redutases/genética , Especificidade por Substrato/genética
17.
Cell Microbiol ; 9(9): 2289-98, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17501981

RESUMO

Chlamydia trachomatis and C. muridarum, human and mouse pathogens, respectively, share more than 99% of open reading frames (ORFs) but differ in a cytotoxin locus. Presence or absence of cytotoxin gene(s) in these strains correlates with their ability to grow in IFN-gamma treated mouse cells. Growth of toxin-positive C. muridarum is not affected in IFN-gamma treated cells, whereas growth of toxin-negative C. trachomatis is inhibited. We previously reported that this difference in IFN-gamma sensitivity is important to the in vivo infection tropism of these pathogens. Here we describe a phenotypic rescue assay that utilizes C. muridarum gamma irradiated killed elementary bodies (iEB) to rescue C. trachomatis infectivity in IFN-gamma treated mouse cells. Rescue by iEB was temporal, maximal early post infection, directly related to multiplicity of iEB infection, and was independent of de novo chlamydial transcription. Lastly, C. muridarum iEB vacuoles and C. trachomatis inclusions were not fusogenic, suggesting the factor(s) responsible for rescue was secreted or exposed to the cytosol where it inactivated IFN-gamma induced effectors. Chlamydial phenotypic rescue may have broader utility for the study of other EB associated virulence factors that function early in the interaction of chlamydiae with host cells.


Assuntos
Chlamydia muridarum/fisiologia , Chlamydia trachomatis/fisiologia , Interferon gama/metabolismo , Animais , Chlamydia muridarum/citologia , Chlamydia muridarum/genética , Chlamydia muridarum/efeitos da radiação , Chlamydia trachomatis/citologia , Chlamydia trachomatis/genética , Raios gama , Genoma Bacteriano , Células HeLa , Humanos , Camundongos , Fenótipo , Vacúolos/metabolismo
18.
Curr Opin Microbiol ; 10(1): 47-51, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17208039

RESUMO

Chlamydia trachomatis is a human pathogen and Chlamydia muridarum is a mouse pathogen but paradoxically, they share near genomic synteny. The majority of strain-variable genes are located primarily in a hyper-variable region termed the plasticity zone. Tryptophan synthase and cytotoxin are plasticity zone genes unique to the human and murine strains, respectively. Tryptophan synthase is a virulence factor that differentiates C. trachomatis strains into genital and ocular disease pathotypes, whereas cytotoxin(s) is a virulence factor linked to murine infection tropism. Divergence in these loci is strongly correlated with host-specific interferon gamma effector activities, suggesting that these virulence genes have co-evolved with their respective hosts as a primary mechanism to evade innate immunity. These findings have important implications for chlamydial animal modeling studies.


Assuntos
Infecções por Chlamydia/microbiologia , Chlamydia/imunologia , Chlamydia/patogenicidade , Interferon gama/imunologia , Animais , Toxinas Bacterianas/genética , Toxinas Bacterianas/metabolismo , Chlamydia/genética , Infecções por Chlamydia/imunologia , Humanos , Camundongos , Óperon , Triptofano Sintase/genética , Triptofano Sintase/fisiologia , Virulência/genética
19.
Mol Microbiol ; 59(6): 1678-91, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16553875

RESUMO

We previously reported that Chlamydia trachomatis expresses the genes encoding tryptophan synthase (trpBA) and the tryptophan repressor (trpR). Here we employ primer extension analysis to identify the transcriptional origins of both trpR and trpBA, allowing for the identification of the putative operator sequences for both trpR and trpBA. Moreover we demonstrate that native recombinant chlamydial TrpR binds to the predicted operator sequence upstream of trpR. A restriction endonuclease protection assay was designed and used to demonstrate that 5-fluorotryptophan was the only tryptophan analogue capable of activating binding of native recombinant chlamydial TrpR to its operator. Additionally, 5-fluorotryptophan was the only analogue that repressed expression of trpBA at a level analogous to L-tryptophan itself. Based on these findings, a mutant selection protocol was designed and a C. trachomatis isolate containing a frameshift mutation in trpR was isolated. This chlamydial mutant synthesizes a truncated TrpR protein that cannot regulate expression of trpBA and trpR in response to changes in tryptophan levels. These findings provide the first genetic proof that TrpR acts as a negative regulator of transcription in C. trachomatis.


Assuntos
Proteínas de Bactérias/metabolismo , Chlamydia trachomatis/genética , Regulação Bacteriana da Expressão Gênica , Regiões Operadoras Genéticas , Regulon/genética , Proteínas Repressoras/metabolismo , Sequência de Aminoácidos , Proteínas de Bactérias/genética , Sequência de Bases , Chlamydia trachomatis/efeitos dos fármacos , Chlamydia trachomatis/metabolismo , DNA Bacteriano/metabolismo , Expressão Gênica/efeitos dos fármacos , Dados de Sequência Molecular , Mutagênese , Mutação , Proteínas Repressoras/genética , Triptofano/análogos & derivados , Triptofano/metabolismo , Triptofano/farmacologia , Triptofano Sintase/genética
20.
Infect Immun ; 74(1): 225-38, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16368976

RESUMO

Gamma interferon (IFN-gamma)-induced effector mechanisms have potent antichlamydial activities that are critical to host defense. The most prominent and well-studied effectors are indoleamine dioxygenase (IDO) and nitric oxide (NO) synthase. The relative contributions of these mechanisms as inhibitors of chlamydial in vitro growth have been extensively studied using different host cells, induction mechanisms, and chlamydial strains with conflicting results. Here, we have undertaken a comparative analysis of cytokine- and lipopolysaccharide (LPS)-induced IDO and NO using an extensive assortment of human and murine host cells infected with human and murine chlamydial strains. Following cytokine (IFN-gamma or tumor necrosis factor alpha) and/or LPS treatment, the majority of human cell lines induced IDO but failed to produce NO. Conversely, the majority of mouse cell lines studied produced NO, not IDO. Induction of IDO in human cell lines inhibited growth of L2 and mouse pneumonitis agent, now referred to as Chlamydia muridarum MoPn equally in all but two lines, and inhibition was completely reversible by the addition of tryptophan. IFN-gamma treatment of mouse cell lines resulted in substantially greater reduction of L2 than MoPn growth. However, despite elevated NO production by murine cells, blockage of NO synthesis with the l-arginine analogue N-monomethyl-l-arginine only partially rescued chlamydial growth, suggesting the presence of another IFN-gamma-inducible antichlamydial mechanism unique to murine cells. Moreover, NO generated from the chemical nitric oxide donor sodium nitroprusside showed little direct effect on chlamydial infectivity or growth, indicating a natural resistance to NO. Finally, IFN-gamma-inducible IDO expression in human HeLa cells was inhibited following exogenous NO treatment, resulting in a permissive environment for chlamydial growth. In summary, cytokine- and LPS-inducible effectors produced by human and mouse cells differ and, importantly, these host-specific effector responses result in chlamydial strain-specific antimicrobial activities.


Assuntos
Chlamydia muridarum/crescimento & desenvolvimento , Chlamydia muridarum/imunologia , Chlamydia trachomatis/crescimento & desenvolvimento , Chlamydia trachomatis/imunologia , Interferon gama/fisiologia , Animais , Linhagem Celular , Linhagem Celular Tumoral , Infecções por Chlamydia/enzimologia , Infecções por Chlamydia/imunologia , Infecções por Chlamydia/prevenção & controle , Células HeLa , Humanos , Indolamina-Pirrol 2,3,-Dioxigenase/biossíntese , Indolamina-Pirrol 2,3,-Dioxigenase/fisiologia , Interferon gama/genética , Lipopolissacarídeos/farmacologia , Camundongos , Doadores de Óxido Nítrico/farmacologia , Óxido Nítrico Sintase Tipo II/biossíntese , Óxido Nítrico Sintase Tipo II/metabolismo , Óxido Nítrico Sintase Tipo II/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...