51
|
Zou Y, Dai W, Lei W, Su S, Huang Q, Zhou Z, Chen C, Li Z. Identification of proteins interacting with pORF5 in the pathogenesis of C. trachomatis. Am J Transl Res 2018; 10:1633-1647. [PMID: 30018706 PMCID: PMC6038076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Accepted: 03/21/2017] [Indexed: 06/08/2023]
Abstract
OBJECTIVE This study is to identify and investigate the proteins interacting with pORF5 implicated in the pathogenesis of C. trachomatis. METHODS The isobaric tags for relative and absolute quantitation (iTRAQ) approach combined with nano liquid chromatography-tandem mass spectrometry (NanoLC-MS/MS) analysis was applied to identify and quantify the differentially expressed proteins in the pORF5-transfected HeLa (pORF5-HeLa) cells and the control vector-transfected HeLa (vector-HeLa) cells. Quantitative real-time PCR (qRT-PCR) and Western blot analysis were performed to detect the mRNA and protein expression levels. RESULTS Totally 3355 proteins were quantified by employing biological replicates, 314 of which were differentially expressed between the pORF5-HeLa and vector-HeLa cells. Nine differentially expressed proteins (HIST1H1C, HBA1, PARK7, HMGB1, HMGB2, CLIC1, KRT7, SFN, and CDKN2A) were subjected to qRT-PCR, and two over-expressed proteins (HMGB1 and PRAK7) were subjected to the Western blot analysis, to validate the proteomic results. The results from the qRT-PCR and Western blot analysis were consistent with the findings from the proteomic analysis. Moreover, pORF5 could inhibit the TNF-α-induced apoptosis in HeLa cells. Through siRNA-mediated functional screening, the high-mobility group box 1 (HMGB1) was shown to be relevant to the inhibition of the apoptotic response in the host cells. CONCLUSION Identification of key proteins interacting with pORF5 could contribute to the understanding and further exploration of the function of pORF5 in the pathogenic mechanisms of C. trachomatis.
Collapse
Affiliation(s)
- Yan Zou
- Pathogenic Biology Institute, School of Medicine, University of South ChinaHengyang 421001, Hunan, China
| | - Wenting Dai
- Pathogenic Biology Institute, School of Medicine, University of South ChinaHengyang 421001, Hunan, China
| | - Wenbo Lei
- Pathogenic Biology Institute, School of Medicine, University of South ChinaHengyang 421001, Hunan, China
| | - Shengmei Su
- Pathogenic Biology Institute, School of Medicine, University of South ChinaHengyang 421001, Hunan, China
| | - Qiulin Huang
- Department of General Surgery, The First Affiliated Hospital of University of South ChinaHengyang 421001, Hunan, China
| | - Zhou Zhou
- Pathogenic Biology Institute, School of Medicine, University of South ChinaHengyang 421001, Hunan, China
| | - Chaoqun Chen
- Pathogenic Biology Institute, School of Medicine, University of South ChinaHengyang 421001, Hunan, China
| | - Zhongyu Li
- Pathogenic Biology Institute, School of Medicine, University of South ChinaHengyang 421001, Hunan, China
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug StudyHengyang 421001, Hunan, China
| |
Collapse
|
52
|
Versteeg B, Bruisten SM, Pannekoek Y, Jolley KA, Maiden MCJ, van der Ende A, Harrison OB. Genomic analyses of the Chlamydia trachomatis core genome show an association between chromosomal genome, plasmid type and disease. BMC Genomics 2018; 19:130. [PMID: 29426279 PMCID: PMC5810182 DOI: 10.1186/s12864-018-4522-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Accepted: 01/31/2018] [Indexed: 12/02/2022] Open
Abstract
Background Chlamydia trachomatis (Ct) plasmid has been shown to encode genes essential for infection. We evaluated the population structure of Ct using whole-genome sequence data (WGS). In particular, the relationship between the Ct genome, plasmid and disease was investigated. Results WGS data from 157 Ct isolates deposited in the Chlamydiales pubMLST database (http://pubMLST.org/chlamydiales/) were annotated with 902 genes including the core and accessory genome. Plasmid associated genes were annotated and a plasmid MLST scheme was defined allowing plasmid sequence types to be determined. Plasmid allelic variation was investigated. Phylogenetic relationships were examined using the Genome Comparator tool available in pubMLST. Phylogenetic analyses identified four distinct Ct core genome clusters and six plasmid clusters, with a strong association between the chromosomal genotype and plasmid. This in turn was linked to ompA genovars and disease phenotype. Horizontal genetic transfer of plasmids was observed for three urogenital-associated isolates, which possessed plasmids more commonly found in isolates resulting from ocular infections. The pgp3 gene was identified as the most polymorphic plasmid gene and pgp4 was the most conserved. Conclusion A strong association between chromosomal genome, plasmid type and disease was observed, consistent with previous studies. This suggests co-evolution of the Ct chromosome and their plasmids, but we confirmed that plasmid transfer can occur between isolates. These data provide a better understanding of the genetic diversity occurring across the Ct genome in association with the plasmid content. Electronic supplementary material The online version of this article (10.1186/s12864-018-4522-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Bart Versteeg
- Public Health Laboratory, Department of Infectious Diseases, Public Health Service Amsterdam, Amsterdam, the Netherlands.
| | - Sylvia M Bruisten
- Public Health Laboratory, Department of Infectious Diseases, Public Health Service Amsterdam, Amsterdam, the Netherlands.,Amsterdam Infection & Immunity Institute, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Yvonne Pannekoek
- Amsterdam Infection & Immunity Institute, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands.,Department of Medical Microbiology, Academic Medical Center, Amsterdam, the Netherlands
| | - Keith A Jolley
- Peter Medawar building, Department of Zoology, University of Oxford, Oxford, UK
| | - Martin C J Maiden
- Peter Medawar building, Department of Zoology, University of Oxford, Oxford, UK
| | - Arie van der Ende
- Amsterdam Infection & Immunity Institute, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands.,Department of Medical Microbiology, Academic Medical Center, Amsterdam, the Netherlands
| | - Odile B Harrison
- Peter Medawar building, Department of Zoology, University of Oxford, Oxford, UK
| |
Collapse
|
53
|
Plasmid Negative Regulation of CPAF Expression Is Pgp4 Independent and Restricted to Invasive Chlamydia trachomatis Biovars. mBio 2018; 9:mBio.02164-17. [PMID: 29382731 PMCID: PMC5790913 DOI: 10.1128/mbio.02164-17] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
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. 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.
Collapse
|
54
|
Zhong G. Chlamydia Spreading from the Genital Tract to the Gastrointestinal Tract - A Two-Hit Hypothesis. Trends Microbiol 2017; 26:611-623. [PMID: 29289422 DOI: 10.1016/j.tim.2017.12.002] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 11/21/2017] [Accepted: 12/06/2017] [Indexed: 12/18/2022]
Abstract
Chlamydia trachomatis, a leading bacterial cause of sexually transmitted infection-induced infertility, is frequently detected in the gastrointestinal tract. Chlamydia muridarum, a model pathogen for investigating C. trachomatis pathogenesis, readily spreads from the mouse genital tract to the gastrointestinal tract, establishing long-lasting colonization. C. muridarum mutants, despite their ability to activate acute oviduct inflammation, are attenuated in inducing tubal fibrosis and are no longer able to colonize the gastrointestinal tract, suggesting that the spread of C. muridarum to the gastrointestinal tract may contribute to its pathogenicity in the upper genital tract. However, gastrointestinal C. muridarum cannot directly autoinoculate the genital tract. Both antigen-specific CD8+ T cells and profibrotic cytokines, such as TNFα and IL-13, are essential for C. muridarum to induce tubal fibrosis; this may be induced by the gastrointestinal C. muridarum, as a second hit, to transmucosally convert tubal repairing - initiated by C. muridarum infection of tubal epithelial cells (serving as the first hit) - into pathogenic fibrosis. Testing the two-hit mouse model should both add new knowledge to the growing list of mechanisms by which gastrointestinal microbes contribute to pathologies in extragastrointestinal tissues and provide information for investigating the potential role of gastrointestinal C. trachomatis in human chlamydial pathogenesis.
Collapse
Affiliation(s)
- Guangming Zhong
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health, Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA.
| |
Collapse
|
55
|
The Genital Tract Virulence Factor pGP3 Is Essential for Chlamydia muridarum Colonization in the Gastrointestinal Tract. Infect Immun 2017; 86:IAI.00429-17. [PMID: 29038127 DOI: 10.1128/iai.00429-17] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Accepted: 10/04/2017] [Indexed: 02/06/2023] Open
Abstract
The cryptic plasmid is essential for Chlamydia muridarum dissemination from the genital tract to the gastrointestinal (GI) tract. Following intravaginal inoculation, a C. muridarum strain deficient in plasmid-encoded pGP3 or pGP4 but not pGP5, pGP7, or pGP8 failed to spread to the mouse gastrointestinal tract, although mice infected with these strains developed productive genital tract infections. pGP3- or pGP4-deficient strains also failed to colonize the gastrointestinal tract when delivered intragastrically. pGP4 regulates pGP3, while pGP3 does not affect pGP4 expression, indicating that pGP3 is critical for C. muridarum colonization of the gastrointestinal tract. Mutants deficient in GlgA, a chromosome-encoded protein regulated by pGP4, also consistently colonized the mouse gastrointestinal tract. Interestingly, C. muridarum colonization of the gastrointestinal tract positively correlated with pathogenicity in the upper genital tract. pGP3-deficient C. muridarum strains did not induce hydrosalpinx or spread to the GI tract even when delivered to the oviduct by intrabursal inoculation. Thus, the current study not only has revealed that pGP3 is a novel chlamydial colonization factor in the gastrointestinal tract but also has laid a foundation for investigating the significance of gastrointestinal Chlamydia.
Collapse
|
56
|
Lei W, Li Q, Su S, Bu J, Huang Q, Li Z. Chlamydia trachomatis plasmid-encoded protein pORF5 protects mitochondrial function by inducing mitophagy and increasing HMGB1 expression. Pathog Dis 2017; 75:4443198. [PMID: 29040500 DOI: 10.1093/femspd/ftx111] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2017] [Accepted: 10/10/2017] [Indexed: 12/22/2022] Open
Affiliation(s)
- Wenbo Lei
- Institute of Pathogenic Biology, Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, No. 28, Changsheng West Road Hengyang, Hunan 421001, P.R. China
| | - Qun Li
- Institute of Pathogenic Biology, Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, No. 28, Changsheng West Road Hengyang, Hunan 421001, P.R. China
| | - Shengmei Su
- Institute of Pathogenic Biology, Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, No. 28, Changsheng West Road Hengyang, Hunan 421001, P.R. China
| | - Jichang Bu
- Institute of Pathogenic Biology, Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, No. 28, Changsheng West Road Hengyang, Hunan 421001, P.R. China
| | - Qiulin Huang
- Department of General Surgery, the First Affiliated Hospital of University of South China, No. 69, Chuanshan Road, Hengyang, Hunan 421001, P.R. China
| | - Zhongyu Li
- Institute of Pathogenic Biology, Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, No. 28, Changsheng West Road Hengyang, Hunan 421001, P.R. China
| |
Collapse
|
57
|
Shao L, Melero J, Zhang N, Arulanandam B, Baseman J, Liu Q, Zhong G. The cryptic plasmid is more important for Chlamydia muridarum to colonize the mouse gastrointestinal tract than to infect the genital tract. PLoS One 2017; 12:e0177691. [PMID: 28542376 PMCID: PMC5444651 DOI: 10.1371/journal.pone.0177691] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Accepted: 05/02/2017] [Indexed: 02/07/2023] Open
Abstract
Chlamydia has been detected in the gastrointestinal tracts of both animals and humans. However, the mechanism by which Chlamydia colonizes the gut remains unclear. Chlamydia muridarum is known to spread from the genital to the gastrointestinal tracts hematogenously. The C. muridarum plasmid is a key pathogenic determinant in the mouse upper genital tract although plasmid-deficient C. muridarum is still able to colonize the upper genital tract. We now report that plasmid-deficient C. muridarum exhibits significantly delayed/reduced spreading from the mouse genital to the gastrointestinal tracts. C. muridarum with or without plasmid maintained similar levels in the mouse circulatory system following intravenous inoculation but the hematogenous plasmid-deficient C. muridarum was significantly less efficient in colonizing the gastrointestinal tract. Consistently, plasmid-deficient C. muridarum failed to restore normal colonization in the gastrointestinal tract even after intragastric inoculation at a high dose. Thus, we have demonstrated a plasmid-dependent colonization of C. muridarum in the gastrointestinal tract, supporting the concept that C. muridarum may have acquired the plasmid for adaptation to the mouse gastrointestinal tract during oral-fecal transmission. Since the plasmid is more important for C. muridarum to colonize the gastrointestinal tract than to infect the genital tract, the current study has laid a foundation for further defining the host pathways targeted by the plasmid-encoded or -regulated chlamydial effectors.
Collapse
Affiliation(s)
- Lili Shao
- Department of Microbiology and Immunology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
- Department of Dermatovenereology, Tianjin Medical University General Hospital, Tianjin, China
| | - Jose Melero
- Department of Microbiology and Immunology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Nu Zhang
- Department of Microbiology and Immunology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Bernard Arulanandam
- Department of Biology, University of Texas at San Antonio, San Antonio, Texas, United States of America
| | - Joel Baseman
- Department of Microbiology and Immunology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Quanzhong Liu
- Department of Dermatovenereology, Tianjin Medical University General Hospital, Tianjin, China
| | - Guangming Zhong
- Department of Microbiology and Immunology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| |
Collapse
|
58
|
Update on Chlamydia trachomatis Vaccinology. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2017; 24:CVI.00543-16. [PMID: 28228394 DOI: 10.1128/cvi.00543-16] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Attempts to produce a vaccine to protect against Chlamydia trachomatis-induced trachoma were initiated more than 100 years ago and continued for several decades. Using whole organisms, protective responses were obtained. However, upon exposure to C. trachomatis, disease exacerbation developed in some immunized individuals, precluding the implementation of the vaccine. Evidence of the role of C. trachomatis as a sexually transmitted pathogen started to emerge in the 1960s, and it soon became evident that it can cause acute infections and long-term sequelae in women, men, and newborns. The main focus of this minireview is to summarize recent findings and discuss formulations, including antigens, adjuvants, routes, and delivery systems for immunization, primarily explored in the female mouse model, with the goal of implementing a vaccine against C. trachomatis genital infections.
Collapse
|
59
|
Winstanley CE, Ramsey KH, Marsh P, Clarke IN. Development and evaluation of an enzyme-linked immunosorbent assay for the detection of antibodies to a common urogenital derivative of Chlamydia trachomatis plasmid-encoded PGP3. J Immunol Methods 2017; 445:23-30. [PMID: 28283408 DOI: 10.1016/j.jim.2017.03.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Revised: 02/19/2017] [Accepted: 03/03/2017] [Indexed: 10/20/2022]
Abstract
BACKGROUND Urogenital infection with Chlamydia trachomatis is the most commonly diagnosed sexually transmitted infection in the developed world. Accurate measurement and therefore understanding the seroprevalence of urogenital C. trachomatis infections requires a rigorously optimised and validated ELISA. Previous ELISAs based on the C. trachomatis plasmid-encoded protein, PGP3, have been described but lack standardisation and critical controls or use a less common PGP3 as the capture antigen. METHODOLOGY/PRINCIPAL FINDINGS A sensitive and specific indirect ELISA was developed based on recombinant PGP3 derived from a urogenital strain of C. trachomatis, serovar E (pSW2), using a rigorous validation protocol. Serum samples were collected from 166 genitourinary medicine (GUM) clinic patients diagnosed as positive or negative for urogenital C. trachomatis infection by nucleic acid amplification testing (NAATs). Overall sensitivity and specificity compared to NAATs was 68.18% and 98.0%, respectively. Sensitivities for female and male samples were 71.93% and 64.15%, respectively. Comparison of samples from these patients diagnosed positive for C. trachomatis by NAAT and patients diagnosed negative by NAAT revealed statistical significance (p≤0.0001). CONCLUSIONS We have developed and validated a sensitive and specific ELISA to detect anti-PGP3 antibodies as an indicator of past and current infection to C. trachomatis using PGP3 from a common urogenital strain. It is anticipated that this assay will be used for seroepidemiological analysis of urogenital C. trachomatis in populations.
Collapse
Affiliation(s)
- Catherine E Winstanley
- Molecular Microbiology Group, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, Hampshire, United Kingdom.
| | - Kyle H Ramsey
- Department of Microbiology & Immunology, Chicago College of Osteopathic Medicine, Midwestern University, Downers Grove, IL, USA
| | - Peter Marsh
- Public Health England Regional Microbiology Laboratory, Southampton General Hospital, Southampton, Hampshire, United Kingdom
| | - Ian N Clarke
- Molecular Microbiology Group, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, Hampshire, United Kingdom
| |
Collapse
|
60
|
Vouga M, Baud D, Greub G. Simkania negevensis, an insight into the biology and clinical importance of a novel member of the Chlamydiales order. Crit Rev Microbiol 2016; 43:62-80. [PMID: 27786615 DOI: 10.3109/1040841x.2016.1165650] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Simkania negevensis is a Chlamydia-related bacterium discovered in 1993 and represents the founding member of the Simkaniaceae family within the Chlamydiales order. As other Chlamydiales, it is an obligate intracellular bacterium characterized by a biphasic developmental cycle. Its similarities with the pathogenic Chlamydia trachomatis and Chlamydia pneumoniae make it an interesting bacterium. So far, little is known about its biology, but S. negevensis harbors various microbiological characteristics of interest, including a strong association of the Simkania-containing vacuole with the ER and the presence of an intron in the 23S rRNA encoding gene. Evidence of human exposition has been reported worldwide. However, there is a lack of robust clinical studies evaluating its implication in human diseases; current data suggest an association with pneumonia and bronchiolitis making S. negevensis a potential emerging pathogen. Owing to its fastidious growth requirements, the clinical relevance of S. negevensis is probably underestimated. In this review, we summarize the current knowledge on S. negevensis and explore future research challenges.
Collapse
Affiliation(s)
- Manon Vouga
- a Institute of Microbiology , Center for Research on Intracellular Bacteria, Faculty of Biology and Medicine, University and University Hospital of Lausanne , Lausanne , Switzerland.,b Department "Femme-Mère-Enfant" , Materno-Fetal and Obstetrics Research Unit, University Hospital , Lausanne , Switzerland
| | - David Baud
- a Institute of Microbiology , Center for Research on Intracellular Bacteria, Faculty of Biology and Medicine, University and University Hospital of Lausanne , Lausanne , Switzerland.,b Department "Femme-Mère-Enfant" , Materno-Fetal and Obstetrics Research Unit, University Hospital , Lausanne , Switzerland
| | - Gilbert Greub
- a Institute of Microbiology , Center for Research on Intracellular Bacteria, Faculty of Biology and Medicine, University and University Hospital of Lausanne , Lausanne , Switzerland.,c Infectious Diseases Unit , University hospital , Lausanne , Switzerland
| |
Collapse
|
61
|
Zhong G. Chlamydial Plasmid-Dependent Pathogenicity. Trends Microbiol 2016; 25:141-152. [PMID: 27712952 DOI: 10.1016/j.tim.2016.09.006] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Revised: 09/11/2016] [Accepted: 09/19/2016] [Indexed: 10/20/2022]
Abstract
Most Chlamydia species carry a 7.5kb plasmid encoding eight open reading frames conventionally called plasmid glycoproteins 1-8 or pGP1-8. Although the plasmid is not critical for chlamydial growth in vitro, its role in chlamydial pathogenesis is clearly demonstrated in the genital tracts of mice infected with Chlamydia muridarum, a model for investigating the human pathogen Chlamydia trachomatis. Plasmid-free C. trachomatis is also attenuated in both the mouse genital tract and nonhuman primate ocular tissue. Deficiency in pGP3 alone, which is regulated by pGP4, largely reproduced the in vivo but not in vitro phenotypes of the plasmid-free organisms, suggesting that pGP3 is a key in vivo virulence factor. The positive and negative regulations of some chromosomal genes by pGP4 and pGP5, respectively, may allow the plasmid to promote chlamydial adaptation to varied animal tissue environments. The focus of this review is to summarize the progress on the pathogenic functions of the plasmid-encoded open reading frames, which may motivate further investigation of the molecular mechanisms of chlamydial pathogenicity and development of medical utility of the chlamydial plasmid system.
Collapse
Affiliation(s)
- Guangming Zhong
- Department of Microbiology and Immunology, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA.
| |
Collapse
|
62
|
Pettengill MA, Abdul-Sater A, Coutinho-Silva R, Ojcius DM. Danger signals, inflammasomes, and the intricate intracellular lives of chlamydiae. Biomed J 2016; 39:306-315. [PMID: 27884377 PMCID: PMC6138786 DOI: 10.1016/j.bj.2016.07.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Accepted: 07/13/2016] [Indexed: 12/20/2022] Open
Abstract
Chlamydiae are obligate intracellular bacterial pathogens, and as such are sensitive to alterations in the cellular physiology of their hosts. Chlamydial infections often cause pathologic consequences due to prolonged localized inflammation. Considerable advances have been made in the last few years regarding our understanding of how two key inflammation-associated signaling pathways influence the biology of Chlamydia infections: inflammation regulating purinergic signaling pathways significantly impact intracellular chlamydial development, and inflammasome activation modulates both chlamydial growth and infection mediated pro-inflammatory cytokine production. We review here elements of both pathways, presenting the latest developments contributing to our understanding of how chlamydial infections are influenced by inflammasomes and purinergic signaling.
Collapse
Affiliation(s)
| | - Ali Abdul-Sater
- School of Kinesiology and Health Science, York University, Toronto, Ontario, Canada
| | - Robson Coutinho-Silva
- Laboratory of Immunophysiology, Carlos Chagas Filho Biophysics Institute of the Federal University of Rio de Janeiro, Brazil
| | - David M Ojcius
- Laboratory of Immunophysiology, Carlos Chagas Filho Biophysics Institute of the Federal University of Rio de Janeiro, Brazil; Department of Biomedical Sciences, University of the Pacific, Arthur Dugoni School of Dentistry, San Francisco, USA.
| |
Collapse
|
63
|
Abstract
Etiology, transmission and protection: Chlamydia
trachomatis is the leading cause of bacterial sexually transmitted
infection (STI) globally. However, C. trachomatis also causes
trachoma in endemic areas, mostly Africa and the Middle East, and is a leading
cause of preventable blindness worldwide. Epidemiology, incidence and
prevalence: The World Health Organization estimates 131 million
new cases of C. trachomatis genital infection occur annually.
Globally, infection is most prevalent in young women and men (14-25 years),
likely driven by asymptomatic infection, inadequate partner treatment and
delayed development of protective immunity.
Pathology/Symptomatology: C.
trachomatis infects susceptible squamocolumnar or transitional
epithelial cells, leading to cervicitis in women and urethritis in men. Symptoms
are often mild or absent but ascending infection in some women may lead to
Pelvic Inflammatory Disease (PID), resulting in reproductive sequelae such as
ectopic pregnancy, infertility and chronic pelvic pain. Complications of
infection in men include epididymitis and reactive arthritis.
Molecular mechanisms of infection: Chlamydiae
manipulate an array of host processes to support their obligate intracellular
developmental cycle. This leads to activation of signaling pathways resulting in
disproportionate influx of innate cells and the release of tissue damaging
proteins and pro-inflammatory cytokines. Treatment and
curability: Uncomplicated urogenital infection is treated with
azithromycin (1 g, single dose) or doxycycline (100 mg twice daily x 7 days).
However, antimicrobial treatment does not ameliorate established disease. Drug
resistance is rare but treatment failures have been described. Development of an
effective vaccine that protects against upper tract disease or that limits
transmission remains an important goal.
Collapse
Affiliation(s)
- Catherine M O'Connell
- Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Morgan E Ferone
- Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
64
|
Yang Z, Tang L, Shao L, Zhang Y, Zhang T, Schenken R, Valdivia R, Zhong G. The Chlamydia-Secreted Protease CPAF Promotes Chlamydial Survival in the Mouse Lower Genital Tract. Infect Immun 2016; 84:2697-702. [PMID: 27382018 PMCID: PMC4995919 DOI: 10.1128/iai.00280-16] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2016] [Accepted: 06/30/2016] [Indexed: 12/16/2022] Open
Abstract
Despite the extensive in vitro characterization of CPAF (chlamydial protease/proteasome-like activity factor), its role in chlamydial infection and pathogenesis remains unclear. We now report that a Chlamydia trachomatis strain deficient in expression of CPAF (L2-17) is no longer able to establish a successful infection in the mouse lower genital tract following an intravaginal inoculation. The L2-17 organisms were cleared from the mouse lower genital tract within a few days, while a CPAF-sufficient C. trachomatis strain (L2-5) survived in the lower genital tract for more than 3 weeks. However, both the L2-17 and L2-5 organisms maintained robust infection courses that lasted up to 4 weeks when they were directly delivered into the mouse upper genital tract. The CPAF-dependent chlamydial survival in the lower genital tract was confirmed in multiple strains of mice. Thus, we have demonstrated a critical role of CPAF in promoting C. trachomatis survival in the mouse lower genital tracts. It will be interesting to further investigate the mechanisms of the CPAF-dependent chlamydial pathogenicity.
Collapse
Affiliation(s)
- Zhangsheng Yang
- Department of Microbiology & Immunology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Lingli Tang
- Department of Clinic Diagnosis, Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Lili Shao
- Department of Microbiology & Immunology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Yuyang Zhang
- Department of Microbiology & Immunology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Tianyuan Zhang
- Department of Microbiology & Immunology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Robert Schenken
- Department of Obstetrics and Gynecology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Raphael Valdivia
- Duke Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Guangming Zhong
- Department of Microbiology & Immunology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| |
Collapse
|
65
|
Yang Z, Tang L, Zhou Z, Zhong G. Neutralizing antichlamydial activity of complement by chlamydia-secreted protease CPAF. Microbes Infect 2016; 18:669-674. [PMID: 27436813 DOI: 10.1016/j.micinf.2016.07.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Revised: 06/22/2016] [Accepted: 07/05/2016] [Indexed: 01/19/2023]
Abstract
Ascending infection by sexually transmitted Chlamydia trachomatis is required for chlamydial induction of tubal pathology. To achieve ascension, the C. trachomatis organisms may have to spread from cell to cell, which inevitably exposes the organisms to extracellular mucosal effectors such as complement factors that are known to possess strong antichlamydial activities. Here, we report that the chlamydia-secreted protease CPAF efficiently neutralized complement factor C3-dependent antichlamydial activity. The neutralization was dependent on the proteolytic activity of CPAF and correlated with the CPAF-mediated degradation of complement factor C3 and factor B. As a result, CPAF preferentially inhibited the alternative complement activation pathway. The significance and limitation of these observations were discussed.
Collapse
Affiliation(s)
- Zhangsheng Yang
- Department of Microbiology & Immunology, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA
| | - Lingli Tang
- Department of Clinic Diagnosis, Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhiguang Zhou
- Department of Endocrinology, Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Guangming Zhong
- Department of Microbiology & Immunology, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA.
| |
Collapse
|
66
|
Liang M, Wen Y, Ran O, Chen L, Wang C, Li L, Xie Y, Zhang Y, Chen C, Wu Y. Protective immunity induced by recombinant protein CPSIT_p8 of Chlamydia psittaci. Appl Microbiol Biotechnol 2016; 100:6385-6393. [PMID: 27052378 DOI: 10.1007/s00253-016-7494-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Revised: 03/17/2016] [Accepted: 03/22/2016] [Indexed: 10/22/2022]
Abstract
Chlamydia psittaci is a zoonotic pathogen with a broad host range that can lead to severe respiratory and systemic disease in humans. Currently, an effective commercial vaccine against C. psittaci infection is not available. The chlamydial plasmid is an important virulence factor and encodes plasmid proteins that play important roles in chlamydial infection and the corresponding immune response. In this study, we assessed the efficacy of vaccination with plasmid proteins at preventing C. psittaci lung infection in a murine model. BALB/c mice were immunized intraperitoneally, three times at 2-week intervals, with purified recombinant CPSIT_p8 protein and then infected with C. psittaci. Immunization significantly decreased chlamydial load in the lungs of infected mice, resulted in a lower level of IFN-γ, and reduced the extent of inflammation. In vivo or in vitro neutralization of C. psittaci with sera collected from immunized mice did not reduce the amount of viable C. psittaci in the lungs of mice, indicating that CPSIT_p8-specific antibodies do not have neutralizing capacity. Furthermore, confocal fluorescence microscopy using a mouse anti-CPSIT_p8 antibody revealed that CPSIT_p8 was localized inside the inclusion of C. psittaci 6BC-infected cells. Our results demonstrate that CPSIT_p8 protein induces significant protective immunity against challenge with C. psittaci in mice and represents a promising new vaccine candidate for the prevention of C. psittaci infection.
Collapse
Affiliation(s)
- Mingxing Liang
- Pathogenic Biology Institute, Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, 421001, China.,Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang, 421001, China
| | - Yating Wen
- Pathogenic Biology Institute, Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, 421001, China
| | - Ou Ran
- Pathogenic Biology Institute, Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, 421001, China.,Department of Clinical Laboratory, Central Hospital of Xiangtan, Xiangtan, 411100, China
| | - Liesong Chen
- Pathogenic Biology Institute, Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, 421001, China
| | - Chuan Wang
- Pathogenic Biology Institute, Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, 421001, China
| | - Li Li
- Pathogenic Biology Institute, Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, 421001, China
| | - Yafeng Xie
- Pathogenic Biology Institute, Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, 421001, China
| | - Yang Zhang
- Department of Pathology, University of South China, Hengyang, 421001, China
| | - Chaoqun Chen
- Pathogenic Biology Institute, Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, 421001, China. .,Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang, 421001, China.
| | - Yimou Wu
- Pathogenic Biology Institute, Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, 421001, China. .,Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang, 421001, China.
| |
Collapse
|
67
|
Jelocnik M, Bachmann NL, Seth-Smith H, Thomson NR, Timms P, Polkinghorne AM. Molecular characterisation of the Chlamydia pecorum plasmid from porcine, ovine, bovine, and koala strains indicates plasmid-strain co-evolution. PeerJ 2016; 4:e1661. [PMID: 26870613 PMCID: PMC4748734 DOI: 10.7717/peerj.1661] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Accepted: 01/14/2016] [Indexed: 01/07/2023] Open
Abstract
Background. Highly stable, evolutionarily conserved, small, non-integrative plasmids are commonly found in members of the Chlamydiaceae and, in some species, these plasmids have been strongly linked to virulence. To date, evidence for such a plasmid in Chlamydia pecorum has been ambiguous. In a recent comparative genomic study of porcine, ovine, bovine, and koala C. pecorum isolates, we identified plasmids (pCpec) in a pig and three koala strains, respectively. Screening of further porcine, ovine, bovine, and koala C. pecorum isolates for pCpec showed that pCpec is common, but not ubiquitous in C. pecorum from all of the infected hosts. Methods. We used a combination of (i) bioinformatic mining of previously sequenced C. pecorum genome data sets and (ii) pCpec PCR-amplicon sequencing to characterise a further 17 novel pCpecs in C. pecorum isolates obtained from livestock, including pigs, sheep, and cattle, as well as those from koala. Results and Discussion. This analysis revealed that pCpec is conserved with all eight coding domain sequences (CDSs) present in isolates from each of the hosts studied. Sequence alignments revealed that the 21 pCpecs show 99% nucleotide sequence identity, with 83 single nucleotide polymorphisms (SNPs) shown to differentiate all of the plasmids analysed in this study. SNPs were found to be mostly synonymous and were distributed evenly across all eight pCpec CDSs as well as in the intergenic regions. Although conserved, analyses of the 21 pCpec sequences resolved plasmids into 12 distinct genotypes, with five shared between pCpecs from different isolates, and the remaining seven genotypes being unique to a single pCpec. Phylogenetic analysis revealed congruency and co-evolution of pCpecs with their cognate chromosome, further supporting polyphyletic origin of the koala C. pecorum. This study provides further understanding of the complex epidemiology of this pathogen in livestock and koala hosts and paves the way for studies to evaluate the function of this putative C. pecorum virulence factor.
Collapse
Affiliation(s)
- Martina Jelocnik
- Centre for Animal Health Innovation, University of the Sunshine Coast , Sippy Downs, Queensland , Australia
| | - Nathan L Bachmann
- Centre for Animal Health Innovation, University of the Sunshine Coast , Sippy Downs, Queensland , Australia
| | - Helena Seth-Smith
- Functional Genomics Center Zurich, University of Zurich , Zurich , Switzerland
| | - Nicholas R Thomson
- Infection Genomics, The Wellcome Trust Sanger Institute , Cambridge , United Kingdom
| | - Peter Timms
- Centre for Animal Health Innovation, University of the Sunshine Coast , Sippy Downs, Queensland , Australia
| | - Adam M Polkinghorne
- Centre for Animal Health Innovation, University of the Sunshine Coast , Sippy Downs, Queensland , Australia
| |
Collapse
|
68
|
Cao W, Zou Y, Su S, He Z, Liu Y, Huang Q, Li Z. Chlamydial plasmid-encoded protein pORF5 induces production of IL-1β and IL-18 via NALP3 inflammasome activation and p38 MAPK pathway. Int J Clin Exp Med 2015; 8:20368-20379. [PMID: 26884953 PMCID: PMC4723798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Accepted: 11/06/2015] [Indexed: 06/05/2023]
Abstract
The pathogenesis of Chlamydia-induced inflammation is poorly understood. pORF5 is the only secreted protein encoded by Chlamydial plasmid. This study aims to investigate the effects of pORF5 on the production of interleukin-1β (IL-1β) and interleukin-18 (IL-18) and the underlying mechanisms of these effects. THP-1 (a human acute monocytic leukemia cell line) cells were stimulated by pORF5 with or without pretreatment with Natch domain, Leucine-rich repeat and PYD-containing protein 3 (NALP3) siRNA, apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC) siRNA, cysteine aspartate-specific protease-1 (caspase-1) specific inhibitor and p38 mitogen-activated protein kinase (p38 MAPK) inhibitor. IL-1β, IL-18 and caspase-1 expression was detected through both ELISA and qRT-PCR. NALP3 and ASC expression was detected by qRT-PCR. The expression of caspase-1 and phosphorylated-p38 MAPK was detected by western blot analysis. pORF5 induced IL-1β, IL-18, caspase-1 and NALP3 inflammasome expression in THP-1 cells. Caspase-1 inhibitor significantly reduced pORF5-induced IL-1β and IL-18 expression. The siRNAs for NALP3 inflammasome significantly reduced pORF5-induced IL-1β, IL-18 and caspase-1 expression. Furthermore, p38 MAPK inhibitor significantly reduced pORF5-induced IL-1β, IL-18, caspase-1 and NALP3 inflammasome expression. pORF5 could induce production of IL-1β and IL-18 via NALP3 inflammasome activation and p38MAPK pathway. pORF5 protein might play an important role in Chlamydia pathogenesis. This study provides a new insight into the molecular pathogenesis of Chlamydial diseases.
Collapse
Affiliation(s)
- Wenjuan Cao
- Pathogenic Biology Institute, School of Medicine, University of South ChinaHengyang 421001, P. R. China
| | - Yan Zou
- Pathogenic Biology Institute, School of Medicine, University of South ChinaHengyang 421001, P. R. China
| | - Shengmei Su
- Pathogenic Biology Institute, School of Medicine, University of South ChinaHengyang 421001, P. R. China
| | - Zhansheng He
- Pathogenic Biology Institute, School of Medicine, University of South ChinaHengyang 421001, P. R. China
| | - Yan Liu
- Pathogenic Biology Institute, School of Medicine, University of South ChinaHengyang 421001, P. R. China
| | - Qiulin Huang
- Department of General Surgery, The First Affiliated Hospital of University of South ChinaHengyang 421001, P. R. China
| | - Zhongyu Li
- Pathogenic Biology Institute, School of Medicine, University of South ChinaHengyang 421001, P. R. China
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug StudyHengyang 421001, P. R. China
| |
Collapse
|
69
|
Jelocnik M, Bachmann NL, Kaltenboeck B, Waugh C, Woolford L, Speight KN, Gillett A, Higgins DP, Flanagan C, Myers GSA, Timms P, Polkinghorne A. Genetic diversity in the plasticity zone and the presence of the chlamydial plasmid differentiates Chlamydia pecorum strains from pigs, sheep, cattle, and koalas. BMC Genomics 2015; 16:893. [PMID: 26531162 PMCID: PMC4632680 DOI: 10.1186/s12864-015-2053-8] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Accepted: 10/08/2015] [Indexed: 12/22/2022] Open
Abstract
Background Chlamydia pecorum is a globally recognised pathogen of livestock and koalas. To date, comparative genomics of C. pecorum strains from sheep, cattle and koalas has revealed that only single nucleotide polymorphisms (SNPs) and a limited number of pseudogenes appear to contribute to the genetic diversity of this pathogen. No chlamydial plasmid has been detected in these strains despite its ubiquitous presence in almost all other chlamydial species. Genomic analyses have not previously included C. pecorum from porcine hosts. We sequenced the genome of three C. pecorum isolates from pigs with differing pathologies in order to re-evaluate the genetic differences and to update the phylogenetic relationships between C. pecorum from each of the hosts. Methods Whole genome sequences for the three porcine C. pecorum isolates (L1, L17 and L71) were acquired using C. pecorum-specific sequence capture probes with culture-independent methods, and assembled in CLC Genomics Workbench. The pairwise comparative genomic analyses of 16 pig, sheep, cattle and koala C. pecorum genomes were performed using several bioinformatics platforms, while the phylogenetic analyses of the core C. pecorum genomes were performed with predicted recombination regions removed. Following the detection of a C. pecorum plasmid, a newly developed C. pecorum-specific plasmid PCR screening assay was used to evaluate the plasmid distribution in 227 C. pecorum samples from pig, sheep, cattle and koala hosts. Results Three porcine C. pecorum genomes were sequenced using C. pecorum-specific sequence capture probes with culture-independent methods. Comparative genomics of the newly sequenced porcine C. pecorum genomes revealed an increased average number of SNP differences (~11 500) between porcine and sheep, cattle, and koala C. pecorum strains, compared to previous C. pecorum genome analyses. We also identified a third copy of the chlamydial cytotoxin gene, found only in porcine C. pecorum isolates. Phylogenetic analyses clustered porcine isolates into a distinct clade, highlighting the polyphyletic origin of C. pecorum in livestock. Most surprising, we also discovered a plasmid in the porcine C. pecorum genome. Using this novel C. pecorum plasmid (pCpec) sequence, a) we developed a pCpec screening assay to evaluate the plasmid distribution in C. pecorum from different hosts; and b) to characterise the pCpec sequences from available previously sequenced C. pecorum genome data. pCpec screening showed that the pCpec is common in all hosts of C. pecorum, however not all C. pecorum strains carry pCpec. Conclusions This study provides further insight into the complexity of C. pecorum epidemiology and novel genomic regions that may be linked to host specificity. C. pecorum plasmid characterisation may aid in improving our understanding of C. pecorum pathogenesis across the variety of host species this animal pathogen infects. Electronic supplementary material The online version of this article (doi:10.1186/s12864-015-2053-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Martina Jelocnik
- Faculty of Science, Health, Education and Engineering, University of the Sunshine Coast, Sippy Downs, QLD, 4558, Australia
| | - Nathan L Bachmann
- Faculty of Science, Health, Education and Engineering, University of the Sunshine Coast, Sippy Downs, QLD, 4558, Australia
| | | | - Courtney Waugh
- Faculty of Science, Health, Education and Engineering, University of the Sunshine Coast, Sippy Downs, QLD, 4558, Australia
| | - Lucy Woolford
- School of Animal and Veterinary Sciences, University of Adelaide, Roseworthy, South Australia, 5371, Australia
| | - K Natasha Speight
- School of Animal and Veterinary Sciences, University of Adelaide, Roseworthy, South Australia, 5371, Australia
| | - Amber Gillett
- Australia Zoo Wildlife Hospital, Beerwah, QLD, 4519, Australia
| | - Damien P Higgins
- Faculty of Veterinary Science, The University of Sydney, New South Wales, 2006, Australia
| | - Cheyne Flanagan
- Port Macquarie Koala Hospital, Port Macquarie, NSW, 2444, Australia
| | - Garry S A Myers
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Peter Timms
- Faculty of Science, Health, Education and Engineering, University of the Sunshine Coast, Sippy Downs, QLD, 4558, Australia
| | - Adam Polkinghorne
- Faculty of Science, Health, Education and Engineering, University of the Sunshine Coast, Sippy Downs, QLD, 4558, Australia.
| |
Collapse
|
70
|
Badamchi-Zadeh A, McKay PF, Holland MJ, Paes W, Brzozowski A, Lacey C, Follmann F, Tregoning JS, Shattock RJ. Intramuscular Immunisation with Chlamydial Proteins Induces Chlamydia trachomatis Specific Ocular Antibodies. PLoS One 2015; 10:e0141209. [PMID: 26501198 PMCID: PMC4621052 DOI: 10.1371/journal.pone.0141209] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Accepted: 10/05/2015] [Indexed: 10/29/2022] Open
Abstract
BACKGROUND Ocular infection with Chlamydia trachomatis can cause trachoma, which is the leading cause of blindness due to infection worldwide. Despite the large-scale implementation of trachoma control programmes in the majority of countries where trachoma is endemic, there remains a need for a vaccine. Since C. trachomatis infects the conjunctival epithelium and stimulates an immune response in the associated lymphoid tissue, vaccine regimens that enhance local antibody responses could be advantageous. In experimental infections of non-human primates (NHPs), antibody specificity to C. trachomatis antigens was found to change over the course of ocular infection. The appearance of major outer membrane protein (MOMP) specific antibodies correlated with a reduction in ocular chlamydial burden, while subsequent generation of antibodies specific for PmpD and Pgp3 correlated with C. trachomatis eradication. METHODS We used a range of heterologous prime-boost vaccinations with DNA, Adenovirus, modified vaccinia Ankara (MVA) and protein vaccines based on the major outer membrane protein (MOMP) as an antigen, and investigated the effect of vaccine route, antigen and regimen on the induction of anti-chlamydial antibodies detectable in the ocular lavage fluid of mice. RESULTS Three intramuscular vaccinations with recombinant protein adjuvanted with MF59 induced significantly greater levels of anti-MOMP ocular antibodies than the other regimens tested. Intranasal delivery of vaccines induced less IgG antibody in the eye than intramuscular delivery. The inclusion of the antigens PmpD and Pgp3, singly or in combination, induced ocular antigen-specific IgG antibodies, although the anti-PmpD antibody response was consistently lower and attenuated by combination with other antigens. CONCLUSIONS If translatable to NHPs and/or humans, this investigation of the murine C. trachomatis specific ocular antibody response following vaccination provides a potential mouse model for the rapid and high throughput evaluation of future trachoma vaccines.
Collapse
Affiliation(s)
- Alexander Badamchi-Zadeh
- Mucosal Infection & Immunity Group, Section of Virology, Imperial College London, St Mary’s Campus, London, United Kingdom
| | - Paul F. McKay
- Mucosal Infection & Immunity Group, Section of Virology, Imperial College London, St Mary’s Campus, London, United Kingdom
| | - Martin J. Holland
- London School of Hygiene and Tropical Medicine, Keppel St, London, United Kingdom
| | - Wayne Paes
- Centre for Immunology and Infection, Hull York Medical School, University of York, York, United Kingdom
- York Structural Biology Laboratory, Department of Chemistry, University of York, York, United Kingdom
| | - Andrzej Brzozowski
- York Structural Biology Laboratory, Department of Chemistry, University of York, York, United Kingdom
| | - Charles Lacey
- Centre for Immunology and Infection, Hull York Medical School, University of York, York, United Kingdom
| | - Frank Follmann
- Chlamydia Vaccine Research, Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark
| | - John S. Tregoning
- Mucosal Infection & Immunity Group, Section of Virology, Imperial College London, St Mary’s Campus, London, United Kingdom
| | - Robin J. Shattock
- Mucosal Infection & Immunity Group, Section of Virology, Imperial College London, St Mary’s Campus, London, United Kingdom
- * E-mail:
| |
Collapse
|
71
|
Chlamydial plasmid-encoded virulence factor Pgp3 neutralizes the antichlamydial activity of human cathelicidin LL-37. Infect Immun 2015; 83:4701-9. [PMID: 26416907 DOI: 10.1128/iai.00746-15] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Accepted: 09/16/2015] [Indexed: 11/20/2022] Open
Abstract
Chlamydia trachomatis infection in the lower genital tract can ascend to and cause pathologies in the upper genital tract, potentially leading to severe complications, such as tubal infertility. However, chlamydial organisms depleted of plasmid or deficient in the plasmid-encoded Pgp3 are attenuated in ascending infection and no longer are able to induce the upper genital tract pathologies, indicating a significant role of Pgp3 in chlamydial pathogenesis. We now report that C. trachomatis Pgp3 can neutralize the antichlamydial activity of human cathelicidin LL-37, a host antimicrobial peptide secreted by both genital tract epithelial cells and infiltrating neutrophils. Pgp3 bound to and formed stable complexes with LL-37. We further showed that the middle region of Pgp3 (Pgp3m) was responsible for both the binding to and neutralization of LL-37, suggesting that Pgp3m can be targeted for attenuating chlamydial pathogenicity or developed for blocking LL-37-involved non-genital-tract pathologies, such as rosacea and psoriasis. Thus, the current study has provided significant information for both understanding the mechanisms of chlamydial pathogenesis and developing novel therapeutic agents.
Collapse
|
72
|
Plasmid-Encoded Pgp5 Is a Significant Contributor to Chlamydia muridarum Induction of Hydrosalpinx. PLoS One 2015; 10:e0124840. [PMID: 25915629 PMCID: PMC4411118 DOI: 10.1371/journal.pone.0124840] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Accepted: 03/18/2015] [Indexed: 12/22/2022] Open
Abstract
We have previously shown that the plasmid-encoded Pgp3 is a major virulence factor for C. muridarum induction of hydrosalpinx. We now report that Pgp5 also plays a significant role in the development of hydrosalpinx following C. muridarum induction. Pgp5 deficiency was introduced via either in-frame deletion (CM-Δpgp5) or premature stop codon installation (CM-pgp5S). Mice infected with either CM-Δpgp5 or CM-pgp5S developed hydrosalpinges at significantly reduced levels with an incidence rate of <40% and a mean severity score of 2 or less. In contrast, 80% or more mice developed hydrosalpinx with a severity score of >3 when mice were infected with Pgp5-sufficient C. muridarum (plasmid-competent wild type or plasmid-free C. muridarum transformed with a full plasmid or depleted of pgp7 gene). The attenuated pathogenicity of the Pgp5-deficient C. muridarum correlated with a significantly reduced level of ascending infection in the oviduct tissue despite the similar overall shedding courses between mice infected with Pgp5-deficeint versus sufficient C. muridarum. Furthermore, in the oviducts of mice infected with Pgp5-deficient C. muridarum, significantly lower levels of inflammatory cell infiltration and cytokine production were detected. Thus, Pgp5 is a significant plasmid-encoded virulence factor for C. muridarum pathogenicity in the upper genital tract.
Collapse
|
73
|
Intrauterine infection with plasmid-free Chlamydia muridarum reveals a critical role of the plasmid in chlamydial ascension and establishes a model for evaluating plasmid-independent pathogenicity. Infect Immun 2015; 83:2583-92. [PMID: 25870225 DOI: 10.1128/iai.00353-15] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Accepted: 04/06/2015] [Indexed: 12/18/2022] Open
Abstract
Intravaginal infection with plasmid-competent but not plasmid-free Chlamydia muridarum induces hydrosalpinx in mouse upper genital tract, indicating a critical role of the plasmid in chlamydial pathogenicity. To evaluate the contribution of the plasmid to chlamydial ascension and activation of tubal inflammation, we delivered plasmid-free C. muridarum directly into the endometrium by intrauterine inoculation. We found that three of the six mouse strains tested, including CBA/J, C3H/HeJ, and C57BL/6J, developed significant hydrosalpinges when 1 × 10(7) inclusion-forming units (IFU) of plasmid-free C. muridarum were intrauterinally inoculated. Even when the inoculum was reduced to 1 × 10(4) IFU, the CBA/J mice still developed robust hydrosalpinx. The hydrosalpinx development in CBA/J mice correlated with increased organism ascension to the oviduct following the intrauterine inoculation. The CBA/J mice intravaginally infected with the same plasmid-free C. muridarum strain displayed reduced ascending infection and failed to develop hydrosalpinx. These observations have demonstrated a critical role of the plasmid in chlamydial ascending infection. The intrauterine inoculation of the CBA/J mice with plasmid-free C. muridarum not only resulted in more infection in the oviduct but also stimulated more inflammatory infiltration and cytokine production in the oviduct than the intravaginal inoculation, suggesting that the oviduct inflammation can be induced by plasmid-independent factors, which makes the hydrosalpinx induction in CBA/J mice by intrauterine infection with plasmid-free C. muridarum a suitable model for investigating plasmid-independent pathogenic mechanisms.
Collapse
|
74
|
Characterization of CPAF critical residues and secretion during Chlamydia trachomatis infection. Infect Immun 2015; 83:2234-41. [PMID: 25776755 DOI: 10.1128/iai.00275-15] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2015] [Accepted: 03/11/2015] [Indexed: 11/20/2022] Open
Abstract
CPAF (chlamydial protease-like activity factor), a Chlamydia serine protease, is activated via proximity-induced intermolecular dimerization that triggers processing and removal of an inhibitory peptide occupying the CPAF substrate-binding groove. An active CPAF is a homodimer of two identical intramolecular heterodimers, each consisting of 29-kDa N-terminal and 35-kDa C-terminal fragments. However, critical residues for CPAF intermolecular dimerization, catalytic activity, and processing were defined in cell-free systems. Complementation of a CPAF-deficient chlamydial organism with a plasmid-encoded CPAF has enabled us to characterize CPAF during infection. The transformants expressing CPAF mutated at intermolecular dimerization, catalytic, or cleavage residues still produced active CPAF, although at a lower efficiency, indicating that CPAF can tolerate more mutations inside Chlamydia-infected cells than in cell-free systems. Only by simultaneously mutating both intermolecular dimerization and catalytic residues was CPAF activation completely blocked during infection, both indicating the importance of the critical residues identified in the cell-free systems and exploring the limit of CPAF's tolerance for mutations in the intracellular environment. We further found that active CPAF was always detected in the host cell cytoplasm while nonactive CPAF was restricted to within the chlamydial inclusions, regardless of how the infected cell samples were treated. Thus, CPAF translocation into the host cell cytoplasm correlates with CPAF enzymatic activity and is not altered by sample treatment conditions. These observations have provided new evidence for CPAF activation and translocation, which should encourage continued investigation of CPAF in chlamydial pathogenesis.
Collapse
|
75
|
Tang L, Chen J, Zhou Z, Yu P, Yang Z, Zhong G. Chlamydia-secreted protease CPAF degrades host antimicrobial peptides. Microbes Infect 2015; 17:402-8. [PMID: 25752416 DOI: 10.1016/j.micinf.2015.02.005] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Revised: 02/25/2015] [Accepted: 02/26/2015] [Indexed: 12/15/2022]
Abstract
Chlamydia trachomatis infection in the lower genital tract, if untreated, can ascend to the upper genital tract, potentially leading to complications such as tubal factor infertility. The ascension involves cell-to-cell spreading, which may require C. trachomatis organisms to overcome mucosal extracellular effectors such as antimicrobial peptides. We found that among the 8 antimicrobial peptides tested, the cathelicidin LL-37 that is produced by both urogenital epithelial cells and the recruited neutrophils possessed a most potent antichlamydial activity. Interestingly, this antichlamydial activity was completely inhibited by CPAF, a C. trachomatis-secreted serine protease. The inhibition was dependent on CPAF's proteolytic activity. CPAF selectively degraded LL-37 and other antimicrobial peptides with an antichlamydial activity. CPAF is known to secrete into and accumulate in the infected host cell cytoplasm at the late stage of chlamydial intracellular growth and may be released to confront the extracellular antimicrobial peptides before the intra-inclusion organisms are exposed to extracellular environments during host cell lysis and chlamydial spreading. Thus, the finding that CPAF selectively targets host antimicrobial peptides that possess antichlamydial activities for proteolysis suggests that CPAF may contribute to C. trachomatis pathogenicity by aiding in ascending infection.
Collapse
Affiliation(s)
- Lingli Tang
- Department of Microbiology and Immunology, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA; Department of Clinic Diagnosis, Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jianlin Chen
- Department of Microbiology and Immunology, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA; Department of Obstetrics and Gynecology, Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhiguang Zhou
- Department of Endocrinology, Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ping Yu
- Department of Immunology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Zhangsheng Yang
- Department of Microbiology and Immunology, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA
| | - Guangming Zhong
- Department of Microbiology and Immunology, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA.
| |
Collapse
|
76
|
In vitro passage selects for Chlamydia muridarum with enhanced infectivity in cultured cells but attenuated pathogenicity in mouse upper genital tract. Infect Immun 2015; 83:1881-92. [PMID: 25712926 DOI: 10.1128/iai.03158-14] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2014] [Accepted: 02/13/2015] [Indexed: 12/30/2022] Open
Abstract
Although modern Chlamydia muridarum has been passaged for decades, there are no reports on the consequences of serial passage with strong selection pressure on its fitness. In order to explore the potential for Pasteurian selection to induce genomic and phenotypic perturbations to C. muridarum, a starter population was passaged in cultured cells for 28 generations without standard infection assistance. The resultant population, designated CMG28, displays markedly reduced in vitro dependence on centrifugation for infection and low incidence and severity of upper genital tract pathology following intravaginal inoculation into mice compared to the parental C. muridarum population, CMG0. Deep sequencing of CMG0 and CMG28 revealed novel protein variants in the hypothetical genes TC0237 (Q117E) and TC0668 (G322R). In vitro attachment assays of isogenic plaque clone pairs with mutations in either TC0237 and TC0668 or only TC0237 reveal that TC0237(Q117E) is solely responsible for enhanced adherence to host cells. Paradoxically, double mutants, but not TC0237(Q117E) single mutants, display severely attenuated in vivo pathogenicity. These findings implicate TC0237 and TC0668 as novel genetic factors involved in chlamydial attachment and pathogenicity, respectively, and show that serial passage under selection pressure remains an effective tool for studying Chlamydia pathogenicity.
Collapse
|
77
|
Plasmid-encoded Pgp3 is a major virulence factor for Chlamydia muridarum to induce hydrosalpinx in mice. Infect Immun 2014; 82:5327-35. [PMID: 25287930 DOI: 10.1128/iai.02576-14] [Citation(s) in RCA: 103] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Hydrosalpinx induction in mice by Chlamydia muridarum infection, a model that has been used to study C. trachomatis pathogenesis in women, is known to depend on the cryptic plasmid that encodes eight genes designated pgp1 to pgp8. To identify the plasmid-encoded pathogenic determinants, we evaluated C. muridarum transformants deficient in the plasmid-borne gene pgp3, -4, or -7 for induction of hydrosalpinx. C. muridarum transformants with an in-frame deletion of either pgp3 or -4 but not -7 failed to induce hydrosalpinx. The deletion mutant phenotype was reproduced by using transformants with premature termination codon insertions in the corresponding pgp genes (to minimize polar effects inherent in the deletion mutants). Pgp4 is known to regulate pgp3 expression, while lack of Pgp3 does not significantly affect Pgp4 function. Thus, we conclude that Pgp3 is an effector virulence factor and that lack of Pgp3 may be responsible for the attenuation in C. muridarum pathogenicity described above. This attenuated pathogenicity was further correlated with a rapid decrease in chlamydial survival in the lower genital tract and reduced ascension to the upper genital tract in mice infected with C. muridarum deficient in Pgp3 but not Pgp7. The Pgp3-deficient C. muridarum organisms were also less invasive when delivered directly to the oviduct on day 7 after inoculation. These observations demonstrate that plasmid-encoded Pgp3 is required for C. muridarum survival in the mouse genital tract and represents a major virulence factor in C. muridarum pathogenesis in mice.
Collapse
|
78
|
Mosolygó T, Szabó AM, Balogh EP, Faludi I, Virók DP, Endrész V, Samu A, Krenács T, Burián K. Protection promoted by pGP3 or pGP4 against Chlamydia muridarum is mediated by CD4(+) cells in C57BL/6N mice. Vaccine 2014; 32:5228-33. [PMID: 25077421 DOI: 10.1016/j.vaccine.2014.07.039] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2014] [Revised: 06/26/2014] [Accepted: 07/09/2014] [Indexed: 01/03/2023]
Abstract
Urogenital tract infection with Chlamydia trachomatis is a leading cause of sexually transmitted infections. There is currently no commercially available vaccine against C. trachomatis. The highly conserved plasmid of chlamydiae has been considered to be a virulence factor and the plasmid proteins have important roles in the Chlamydia-specific immune response. This study was designed to evaluate the efficacy of vaccination with plasmid proteins in the prevention of C. muridarum lung infection in a mouse model. C57BL/6N mice were immunised 3 times subcutaneously with recombinant pGP3 or pGP4 and infected with C. muridarum. Immunisation of the mice with recombinant pGP3 or pGP4 protein caused a significantly lower chlamydial burden in the lungs of the infected mice; the lower IFN-γ level indicated a reduced extent of inflammation. In vitro or in vivo neutralisation of C. muridarum with sera obtained from immunised mice did not reduce the number of viable C. muridarum in the lungs of mice. However, adoptive transfer of the CD4(+) spleen cells isolated from the immunised mice resulted in a significantly reduced bacterial burden. Our results indicate that it is not the pGP3- and pGP4-specific antibodies, but the CD4(+) cells that are responsible for the protective effect of the immune response to plasmid proteins.
Collapse
Affiliation(s)
- Tímea Mosolygó
- Department of Medical Microbiology and Immunobiology, University of Szeged, Szeged, Hungary
| | - Agnes M Szabó
- Department of Medical Microbiology and Immunobiology, University of Szeged, Szeged, Hungary
| | - Emese P Balogh
- Department of Medical Microbiology and Immunobiology, University of Szeged, Szeged, Hungary
| | - Ildikó Faludi
- Department of Medical Microbiology and Immunobiology, University of Szeged, Szeged, Hungary
| | - Dezső P Virók
- Institute of Clinical Microbiology, University of Szeged, Szeged, Hungary
| | - Valéria Endrész
- Department of Medical Microbiology and Immunobiology, University of Szeged, Szeged, Hungary
| | - Alíz Samu
- Ist Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Tibor Krenács
- Ist Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Katalin Burián
- Department of Medical Microbiology and Immunobiology, University of Szeged, Szeged, Hungary.
| |
Collapse
|
79
|
Plasmid CDS5 influences infectivity and virulence in a mouse model of Chlamydia trachomatis urogenital infection. Infect Immun 2014; 82:3341-9. [PMID: 24866804 DOI: 10.1128/iai.01795-14] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
The native plasmid of both Chlamydia muridarum and Chlamydia trachomatis has been shown to control virulence and infectivity in mice and in lower primates. We recently described the development of a plasmid-based genetic transformation protocol for Chlamydia trachomatis that for the first time provides a platform for the molecular dissection of the function of the chlamydial plasmid and its individual genes or coding sequences (CDS). In the present study, we transformed a plasmid-free lymphogranuloma venereum isolate of C. trachomatis, serovar L2, with either the original shuttle vector (pGFP::SW2) or a derivative of pGFP::SW2 carrying a deletion of the plasmid CDS5 gene (pCDS5KO). Female mice were inoculated with these strains either intravaginally or transcervically. We found that transformation of the plasmid-free isolate with the intact pGFP::SW2 vector significantly enhanced infectivity and induction of host inflammatory responses compared to the plasmid-free parental isolate. Transformation with pCDS5KO resulted in infection courses and inflammatory responses not significantly different from those observed in mice infected with the plasmid-free isolate. These results indicate a critical role of plasmid CDS5 in in vivo fitness and in induction of inflammatory responses. To our knowledge, these are the first in vivo observations ascribing infectivity and virulence to a specific plasmid gene.
Collapse
|
80
|
Wang Y, Cutcliffe LT, Skilton RJ, Ramsey KH, Thomson NR, Clarke IN. The genetic basis of plasmid tropism between Chlamydia trachomatis and Chlamydia muridarum. Pathog Dis 2014; 72:19-23. [PMID: 24700815 PMCID: PMC4314687 DOI: 10.1111/2049-632x.12175] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2014] [Revised: 03/26/2014] [Accepted: 03/26/2014] [Indexed: 11/30/2022] Open
Abstract
The development of genetic transformation technology for Chlamydia trachomatis using its endogenous plasmid has recently been described. Chlamydia muridarum cannot be transformed by the C. trachomatis plasmid, indicating a barrier between chlamydial species. To determine which regions of the plasmid conferred the species specificity, we used the novel approach of transforming wild-type C. muridarum carrying the endogenous plasmid pNigg and forced recombination with the C. trachomatis vector pGFP::SW2 which carries the complete C. trachomatis plasmid (pSW2). Penicillin and chloramphenicol-resistant transformants expressing the green fluorescent protein were selected. Recovery of plasmids from these transformants showed they were recombinants. The differences between the pSW2 and pNigg allowed identification of the recombination breakpoints and showed that pGFP::SW2 had exchanged a ∼ 1 kbp region with pNigg covering CDS 2. The recombinant plasmid (pSW2NiggCDS2) is maintained under antibiotic selection when transformed into plasmid-cured C. muridarum. The ability to select for recombinants in C. muridarum shows that the barrier is not at transformation, but at the level of plasmid replication or maintenance. Our studies show that CDS 2, together with adjoining sequences, is the main determinant of plasmid tropism.
Collapse
Affiliation(s)
- Yibing Wang
- Molecular Microbiology Group, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, UK
| | | | | | | | | | | |
Collapse
|
81
|
Mosolygó T, Faludi I, Balogh EP, Szabó ÁM, Karai A, Kerekes F, Virók DP, Endrész V, Burián K. Expression of Chlamydia muridarum plasmid genes and immunogenicity of pGP3 and pGP4 in different mouse strains. Int J Med Microbiol 2014; 304:476-83. [DOI: 10.1016/j.ijmm.2014.02.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2013] [Revised: 11/25/2013] [Accepted: 02/09/2014] [Indexed: 12/01/2022] Open
|
82
|
da Cunha M, Milho C, Almeida F, Pais SV, Borges V, Maurício R, Borrego MJ, Gomes JP, Mota LJ. Identification of type III secretion substrates of Chlamydia trachomatis using Yersinia enterocolitica as a heterologous system. BMC Microbiol 2014; 14:40. [PMID: 24533538 PMCID: PMC3931295 DOI: 10.1186/1471-2180-14-40] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2013] [Accepted: 02/10/2014] [Indexed: 12/18/2022] Open
Abstract
Background Chlamydia trachomatis is an obligate intracellular human pathogen causing ocular and urogenital infections that are a significant clinical and public health concern. This bacterium uses a type III secretion (T3S) system to manipulate host cells, through the delivery of effector proteins into their cytosol, membranes, and nucleus. In this work, we aimed to find previously unidentified C. trachomatis T3S substrates. Results We first analyzed the genome of C. trachomatis L2/434 strain for genes encoding mostly uncharacterized proteins that did not appear to possess a signal of the general secretory pathway and which had not been previously experimentally shown to be T3S substrates. We selected several genes with these characteristics and analyzed T3S of the encoding proteins using Yersinia enterocolitica as a heterologous system. We identified 23 C. trachomatis proteins whose first 20 amino acids were sufficient to drive T3S of the mature form of β-lactamase TEM-1 by Y. enterocolitica. We found that 10 of these 23 proteins were also type III secreted in their full-length versions by Y. enterocolitica, providing additional support that they are T3S substrates. Seven of these 10 likely T3S substrates of C. trachomatis were delivered by Y. enterocolitica into host cells, further suggesting that they could be effectors. Finally, real-time quantitative PCR analysis of expression of genes encoding the 10 likely T3S substrates of C. trachomatis showed that 9 of them were clearly expressed during infection of host cells. Conclusions Using Y. enterocolitica as a heterologous system, we identified 10 likely T3S substrates of C. trachomatis (CT053, CT105, CT142, CT143, CT144, CT161, CT338, CT429, CT656, and CT849) and could detect translocation into host cells of CT053, CT105, CT142, CT143, CT161, CT338, and CT429. Therefore, we revealed several C. trachomatis proteins that could be effectors subverting host cell processes.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Luís Jaime Mota
- Infection Biology Laboratory, Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal.
| |
Collapse
|
83
|
Sturdevant GL, Zhou B, Carlson JH, Whitmire WM, Song L, Caldwell HD. Infectivity of urogenital Chlamydia trachomatis plasmid-deficient, CT135-null, and double-deficient strains in female mice. Pathog Dis 2014; 71:90-2. [PMID: 24376189 DOI: 10.1111/2049-632x.12121] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2013] [Revised: 12/04/2013] [Accepted: 12/12/2013] [Indexed: 11/28/2022] Open
Abstract
Chlamydia trachomatis is the most common cause of human bacterial sexually transmitted infections and is the world's leading cause of infectious preventable blindness. The chlamydial 7.5-kb plasmid and chromosomal gene CT135 have been shown to be important virulence factors in both nonhuman primate and mouse infection models. Chlamydia trachomatis plasmid-deficient urogenital isolates and a predicted CT135 null mutant have been evaluated independently in the female mouse genital tract model and both have been shown to reduce infectivity and virulence. However, these attenuating phenotypes have not been evaluated collectively in the murine model. Here, we test the infectivity of C. trachomatis serovar D strains in the mouse model that are plasmid-deficient, CT135 disrupted, or possess a combination of these attenuating genotypes. We find that the presence of the plasmid results in infections with higher infectious burdens, whereas CT135 facilitates a more protracted or chronic infection. Not unexpectedly, a combination of these genetic deficiencies resulted in a strain with enhanced infection attenuation characteristics.
Collapse
Affiliation(s)
- Gail L Sturdevant
- Laboratory of Intracellular Parasites, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | | | | | | | | | | |
Collapse
|
84
|
Transformation of Chlamydia muridarum reveals a role for Pgp5 in suppression of plasmid-dependent gene expression. J Bacteriol 2013; 196:989-98. [PMID: 24363344 DOI: 10.1128/jb.01161-13] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Transformation of Chlamydia trachomatis should greatly advance the chlamydial research. However, significant progress has been hindered by the failure of C. trachomatis to induce clinically relevant pathology in animal models. Chlamydia muridarum, which naturally infects mice, can induce hydrosalpinx in mice, a tubal pathology also seen in women infected with C. trachomatis. We have developed a C. muridarum transformation system and confirmed Pgp1, -2, -6, and -8 as plasmid maintenance factors, Pgp3, -5, and -7 as dispensable for in vitro growth, and Pgp4 as a positive regulator of genes that are dependent on plasmid for expression. More importantly, we have discovered that Pgp5 can negatively regulate the same plasmid-dependent genes. Deletion of Pgp5 led to a significant increase in expression of the plasmid-dependent genes, suggesting that Pgp5 can suppress the expression of these genes. Replacement of pgp5 with a mCherry gene, or premature termination of pgp5 translation, also increased expression of the plasmid-dependent genes, indicating that Pgp5 protein but not its DNA sequence is required for the inhibitory effect. Replacing C. muridarum pgp5 with a C. trachomatis pgp5 still inhibited the plasmid-dependent gene expression, indicating that the negative regulation of plasmid-dependent genes is a common feature of all Pgp5 regardless of its origin. Nevertheless, C. muridarum Pgp5 is more potent than C. trachomatis Pgp5 in suppressing gene expression. Thus, we have uncovered a novel function of Pgp5 and developed a C. muridarum transformation system for further mapping chlamydial pathogenic and protective determinants in animal models.
Collapse
|
85
|
Song L, Carlson JH, Zhou B, Virtaneva K, Whitmire WM, Sturdevant GL, Porcella SF, McClarty G, Caldwell HD. Plasmid-mediated transformation tropism of chlamydial biovars. Pathog Dis 2013; 70:189-93. [PMID: 24214488 DOI: 10.1111/2049-632x.12104] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2013] [Accepted: 10/04/2013] [Indexed: 11/28/2022] Open
Abstract
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.
Collapse
Affiliation(s)
- Lihua Song
- Laboratory of Intracellular Parasites, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA; State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Beijing, China
| | | | | | | | | | | | | | | | | |
Collapse
|
86
|
A Conrad T, Yang Z, Ojcius D, Zhong G. A path forward for the chlamydial virulence factor CPAF. Microbes Infect 2013; 15:1026-32. [PMID: 24141088 DOI: 10.1016/j.micinf.2013.09.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2013] [Revised: 08/26/2013] [Accepted: 09/02/2013] [Indexed: 12/14/2022]
Abstract
CPAF is a conserved and secreted protease from obligate intracellular bacteria of the order Chlamydiales. Recently, it was demonstrated that most of its host targets are an artifact of inaccurate methods. This review aims to summarize key features of CPAF and propose new approaches for evaluating its role in chlamydial pathogenesis.
Collapse
Affiliation(s)
- Turner A Conrad
- Department of Microbiology and Immunology, University of Texas Health Science Center San Antonio, San Antonio, TX 78229, USA
| | | | | | | |
Collapse
|
87
|
Gong S, Yang Z, Lei L, Shen L, Zhong G. Characterization of Chlamydia trachomatis plasmid-encoded open reading frames. J Bacteriol 2013; 195:3819-26. [PMID: 23794619 PMCID: PMC3754608 DOI: 10.1128/jb.00511-13] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2013] [Accepted: 06/12/2013] [Indexed: 11/20/2022] Open
Abstract
The recent success in transformation of Chlamydia trachomatis represents a major advancement in Chlamydia research. Plasmid-free C. trachomatis serovar L2 organisms can be transformed with chlamydial plasmid-based shuttle vectors pGFP::SW2 and pBRCT. Deletion of plasmid genes coding for Pgp1 to Pgp8 in pBRCT led to the identification of Pgp1, -2, -6, and -8 as plasmid maintenance factors; Pgp4 as a transcriptional regulator of chlamydial virulence-associated gene expression; and Pgp3, -5, and -7 as being dispensable for chlamydial growth in vitro. Using the pGFP::SW2 vector system, we confirmed these findings in the current report. To further dissect the roles of pgp coding sequences and Pgp proteins in plasmid maintenance, we introduced premature stop codons into the pgp genes. Stable transformants were obtained with pGFP::SW2 derivatives carrying premature stop codons in pgp8 but not in pgp1, pgp2, and pgp6, suggesting that the pgp8 coding sequence but not the Pgp8 protein is required for maintaining the plasmid, while Pgp1, -2, and -6 proteins are necessary for plasmid maintenance. We also found that a minimum of 30 nucleotides in the pgp3 coding region was required for pgp4 expression. Finally, mCherry red fluorescent protein was successfully expressed when the mCherry gene was used to replace the pgp3, pgp4, or pgp5 coding region, indicating that these regions can be used to express nonchlamydial genes in chlamydial organisms. These novel observations have provided information for further use of chlamydial plasmid shuttle vectors as genetic tools to understand chlamydial biology and pathogenicity as well as to develop attenuated chlamydial vaccines.
Collapse
Affiliation(s)
- Siqi Gong
- Department of Microbiology and Immunology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Zhangsheng Yang
- Department of Microbiology and Immunology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Lei Lei
- Department of Microbiology and Immunology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Li Shen
- Department of Microbiology, Immunology and Parasitology, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA
| | - Guangming Zhong
- Department of Microbiology and Immunology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| |
Collapse
|
88
|
Oviduct infection and hydrosalpinx in DBA1/j mice is induced by intracervical but not intravaginal inoculation with Chlamydia muridarum. PLoS One 2013; 8:e71649. [PMID: 23940777 PMCID: PMC3734308 DOI: 10.1371/journal.pone.0071649] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2013] [Accepted: 07/01/2013] [Indexed: 11/19/2022] Open
Abstract
Intravaginal infection with C. muridarum in mice often results in hydrosalpinx similar to that found in women urogenitally infected with C. trachomatis, making the C. muridarum lower genital tract infection murine model suitable for studying C. trachomatis pathogenesis. To our surprise, DBA1/j mice were highly resistant to hydrosalpinx following an intravaginal infection with C. muridarum although these mice were as susceptible to lower genital tract infection as other mouse strains. A significantly lower level of C. muridarum organisms was recovered from the oviduct of DBA1/j mice, correlating the resistance to hydrosalpinx with reduced ascension of C. muridarum to the oviduct. The DBA1/j resistance to hydrosalpinx was effectively overcome by intracervical inoculation with C. muridarum. The intracervically inoculated DBA1/j mice developed severe hydrosalpinx with the highest levels of live C. muridarum organisms recovered from uterine tissue on day 3 and oviduct tissue on day 7 post inoculation while in intravaginally inoculated DBA1/j mice, the peak of live organism recovery from uterine tissue was delayed to day 7 with no rise in the amount of live organisms recovered from the oviduct. These observations have not only validated the correlation between hydrosalpinx and live organism invasion in the oviduct but also demonstrated that the intracervical inoculation, by promoting rapid chlamydial replication in the uterine epithelial cells and ascension to the oviduct of DBA1/j mice, may be used for further understanding chlamydial pathogenic mechanisms. The above findings also suggest that strategies aimed at reducing tubal infection may be most effective in blocking tubal pathology.
Collapse
|
89
|
Lu C, Lei L, Peng B, Tang L, Ding H, Gong S, Li Z, Wu Y, Zhong G. Chlamydia trachomatis GlgA is secreted into host cell cytoplasm. PLoS One 2013; 8:e68764. [PMID: 23894341 PMCID: PMC3722199 DOI: 10.1371/journal.pone.0068764] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2012] [Accepted: 06/04/2013] [Indexed: 12/03/2022] Open
Abstract
Glycogen has been localized both inside and outside Chlamydia trachomatis organisms. We now report that C. trachomatis glycogen synthase (GlgA) was detected in both chlamydial organism-associated and -free forms. The organism-free GlgA molecules were localized both in the lumen of chlamydial inclusions and in the cytosol of host cells. The cytosolic GlgA displayed a distribution pattern similar to that of a known C. trachomatis-secreted protease, CPAF. The detection of GlgA was specific since the anti-GlgA antibody labeling was only removed by preabsorption with GlgA but not CPAF fusion proteins. GlgA was detectable at 12h and its localization into host cell cytosol only became apparent at 24h after infection. The cytosolic localization of GlgA was conserved among all C. trachomatis serovars. However, the significance of the GlgA secretion into host cell cytoplasm remains unclear since, while expression of chlamydial GlgA in HeLa cells increased glycogen stores, it did not affect a subsequent infection with C. trachomatis. Similar to several other C. trachomatis-secreted proteins, GlgA is immunogenic in women urogenitally infected with C. trachomatis, suggesting that GlgA is expressed and may be secreted into host cell cytosol during C. trachomatis infection in humans. These findings have provided important information for further understanding C. trachomatis pathogenic mechanisms.
Collapse
Affiliation(s)
- Chunxue Lu
- Department of Pathogen Biology, University of South China, Hengyang, Hunan, China
- Department of Microbiology and Immunology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Lei Lei
- Department of Microbiology and Immunology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Bo Peng
- Department of Pathogen Biology, University of South China, Hengyang, Hunan, China
- Department of Microbiology and Immunology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Lingli Tang
- Department of Microbiology and Immunology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Honglei Ding
- Department of Microbiology and Immunology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Siqi Gong
- Department of Microbiology and Immunology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Zhongyu Li
- Department of Pathogen Biology, University of South China, Hengyang, Hunan, China
| | - Yimou Wu
- Department of Pathogen Biology, University of South China, Hengyang, Hunan, China
- * E-mail: (YW); (GZ)
| | - Guangming Zhong
- Department of Microbiology and Immunology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
- * E-mail: (YW); (GZ)
| |
Collapse
|
90
|
Antibody signature of spontaneous clearance of Chlamydia trachomatis ocular infection and partial resistance against re-challenge in a nonhuman primate trachoma model. PLoS Negl Trop Dis 2013; 7:e2248. [PMID: 23738030 PMCID: PMC3667776 DOI: 10.1371/journal.pntd.0002248] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2013] [Accepted: 04/18/2013] [Indexed: 11/19/2022] Open
Abstract
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. Chlamydia trachomatis is the etiological agent of trachoma the world's leading cause of infectious blindness. In this study, we investigated whether delayed clearance of a primary infection in nonhuman primates was attributable to antigenic variation or related to gradual changes in the humoral immune response specific to chlamydial antigens. We found that antigenic variation was not related to the inability of monkeys to efficiently resolve their infection. However, exploring changes in the immune response as a possible reason for delayed clearance revealed that antibody recognition was restricted to the antigenically variable major surface protein and a few conserved polypeptides. Antibody recognition of the major antigenically variable surface protein correlated with the initial reduction in infectious burdens while recognition of conserved chlamydial antigens occurred late and correlated with infection eradication. These findings suggest that delayed clearance of chlamydial infection is not the result of antigenic variation but a consequence of a gradually evolving humoral immune response specific to different chlamydial antigens. Antibody recognition was at least partially directed against conformational epitopes, indicating that an efficacious subunit trachoma vaccine might require a combination of antigens delivered in their native conformation.
Collapse
|
91
|
Lei L, Dong X, Li Z, Zhong G. Identification of a novel nuclear localization signal sequence in Chlamydia trachomatis-secreted hypothetical protein CT311. PLoS One 2013; 8:e64529. [PMID: 23717625 PMCID: PMC3662721 DOI: 10.1371/journal.pone.0064529] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2013] [Accepted: 04/16/2013] [Indexed: 12/03/2022] Open
Abstract
We previously reported that Chlamydia trachomatis hypothetical protein CT311 was secreted out of chlamydial inclusion and into host cell cytosol. We now found that CT311 further entered host cell nucleus at the late stage of infection and continued to accumulate in the nucleus of C. trachomatis-infected cells. When CT311 was expressed via a transgene in mammalian cells, CT311 protein was exclusively detected in the nucleus, suggesting that CT311 by itself is sufficient for nuclear targeting. However, preexisting nuclear CT311 did not affect subsequent chlamydial infection. Using deletion constructs, we mapped a nuclear localization signal sequence of CT311 to residues 21 to 63 (21AVEGKPLSRAAQLRERRKDLHVSGKPSPRYALKKRALEAKKNK63). This sequence was sufficient for targeting a heterologous protein into mammalian cell nucleus and it contains two independent clusters of basic residues (34RERRK38 and 53KKRALEAKKNK63 respectively). Deletion or alanine substitution of the basic residues in either cluster led to loss of nuclear targeting activity, suggesting that both clusters are critical for the nuclear targeting function. These observations have demonstrated that the hypothetical protein CT311 possesses a novel nuclear localization signal sequence with dual modules of basic residues for targeting host cell nucleus during Chlamydia trachomatis infection.
Collapse
Affiliation(s)
- Lei Lei
- Department of Microbiology and Immunology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Xiaohua Dong
- Department of Pharmacology, School of Pharmacy, Hebei Northern University, Zhangjiakou, Hebei, P. R. China
| | - Zhongyu Li
- Department of Microbiology, University of South China, Hengyang, Hunan, P. R. China
| | - Guangming Zhong
- Department of Microbiology and Immunology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
- * E-mail:
| |
Collapse
|
92
|
Galaleldeen A, Taylor AB, Chen D, Schuermann JP, Holloway SP, Hou S, Gong S, Zhong G, Hart PJ. Structure of the Chlamydia trachomatis immunodominant antigen Pgp3. J Biol Chem 2013; 288:22068-79. [PMID: 23703617 DOI: 10.1074/jbc.m113.475012] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Chlamydia trachomatis infection is the most common sexually transmitted bacterial disease. Left untreated, it can lead to ectopic pregnancy, pelvic inflammatory disease, and infertility. Here we present the structure of the secreted C. trachomatis protein Pgp3, an immunodominant antigen and putative virulence factor. The ∼84-kDa Pgp3 homotrimer, encoded on a cryptic plasmid, consists of globular N- and C-terminal assemblies connected by a triple-helical coiled-coil. The C-terminal domains possess folds similar to members of the TNF family of cytokines. The closest Pgp3 C-terminal domain structural homologs include a lectin from Burkholderia cenocepacia, the C1q component of complement, and a portion of the Bacillus anthracis spore surface protein BclA, all of which play roles in bioadhesion. The N-terminal domain consists of a concatenation of structural motifs typically found in trimeric viral proteins. The central parallel triple-helical coiled-coil contains an unusual alternating pattern of apolar and polar residue pairs that generate a rare right-handed superhelical twist. The unique architecture of Pgp3 provides the basis for understanding its role in chlamydial pathogenesis and serves as the platform for its optimization as a potential vaccine antigen candidate.
Collapse
Affiliation(s)
- Ahmad Galaleldeen
- Department of Biochemistry, University of Texas Health Science Center, San Antonio, Texas 78229, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
93
|
Ferreira R, Borges V, Nunes A, Borrego MJ, Gomes JP. Assessment of the load and transcriptional dynamics of Chlamydia trachomatis plasmid according to strains' tissue tropism. Microbiol Res 2013; 168:333-339. [PMID: 23590987 DOI: 10.1016/j.micres.2013.02.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2013] [Revised: 01/28/2013] [Accepted: 02/06/2013] [Indexed: 10/27/2022]
Abstract
Chlamydia trachomatis maintain a conserved plasmid, which is a primary regulator of chromosomal genes, but there is no experimental evidences associating it with the strains' differential tissue tropism (ocular and genital mucosae, and lymph nodes). We investigated if the number of plasmids per strain correlate with expression profiles of plasmid ORFs and small anti-sense RNAs (sRNAs), and also if these molecular features underlie tropism dissimilarities. We performed absolute and relative qPCR to determine both the plasmid load and expression throughout C. trachomatis development. Our findings suggest that plasmid load (never exceeding 8 copies) is not a function of expression needs and does not reflect tissue tropism. However, for most ORFs, ocular strains presented lower expression than genital or lymphogranuloma venereum (LGV) strains, and ORF6/pgp4 (transcriptional regulator of virulence associated genes) presented the highest mean expression among strains, followed by the virulence factor ORF5/pgp3 (also regulated by ORF6/pgp4). More, the mean expression levels of the sRNA-2 (anti-sense to ORF2/pgp8) were up to 100-fold higher than those of the ORFs, and up to 12-fold higher than that of sRNA-7 (anti-sense to ORF7/pgp5) for the LGV strains. Overall, besides the known regulatory role of C. trachomatis plasmid, its transcriptional dynamics sustains tropism differences.
Collapse
Affiliation(s)
- Rita Ferreira
- Department of Infectious Diseases, National Institute of Health, Av. Padre Cruz, 1649-016 Lisbon, Portugal
| | - Vítor Borges
- Department of Infectious Diseases, National Institute of Health, Av. Padre Cruz, 1649-016 Lisbon, Portugal
| | - Alexandra Nunes
- Department of Infectious Diseases, National Institute of Health, Av. Padre Cruz, 1649-016 Lisbon, Portugal
| | - Maria José Borrego
- Department of Infectious Diseases, National Institute of Health, Av. Padre Cruz, 1649-016 Lisbon, Portugal
| | - João Paulo Gomes
- Department of Infectious Diseases, National Institute of Health, Av. Padre Cruz, 1649-016 Lisbon, Portugal.
| |
Collapse
|
94
|
Zhou H, Huang Q, Li Z, Wu Y, Xie X, Ma K, Cao W, Zhou Z, Lu C, Zhong G. PORF5 plasmid protein of Chlamydia trachomatis induces MAPK-mediated pro-inflammatory cytokines via TLR2 activation in THP-1 cells. SCIENCE CHINA-LIFE SCIENCES 2013; 56:460-6. [PMID: 23546865 DOI: 10.1007/s11427-013-4470-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2013] [Accepted: 03/13/2013] [Indexed: 11/28/2022]
Abstract
Infection with Chlamydia trachomatis induces inflammatory pathologies in the urogenital tract that can lead to infertility and ectopic pregnancy. Pathogenesis of infection has been mostly attributed to excessive cytokine production. However, precise mechanisms on how C. trachomatis triggers this production, and which protein(s) stimulate inflammatory cytokines remains unknown. In the present study, the C. trachomatis pORF5 protein induced tumor necrosis factor alpha (TNF-α), interleukin-1 beta (IL-1β) and interleukin-8 (IL-8) in dose- and time-dependent manners in the THP-1 human monocyte cell line. We found that intracellular p38/mitogen-activated protein kinase (MAPK) and extracellular signal-regulated kinase (ERK)/MAPK signaling pathways were required for the induction of TNF-α, IL-1β and IL-8. Blockade of toll-like receptor 2 (TLR2) signaling reduced induction levels of TNF-α, IL-8 and IL-1β. We concluded that the C. trachomatis pORF5 protein might contribute to the inflammatory processes associated with chlamydial infections.
Collapse
Affiliation(s)
- Hui Zhou
- Pathogenic Biology Institute, University of South China, Hengyang 421001, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
95
|
Wang Y, Kahane S, Cutcliffe LT, Skilton RJ, Lambden PR, Persson K, Bjartling C, Clarke IN. Genetic transformation of a clinical (genital tract), plasmid-free isolate of Chlamydia trachomatis: engineering the plasmid as a cloning vector. PLoS One 2013; 8:e59195. [PMID: 23527131 PMCID: PMC3601068 DOI: 10.1371/journal.pone.0059195] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2012] [Accepted: 02/11/2013] [Indexed: 11/18/2022] Open
Abstract
Our study had three objectives: to extend the plasmid-based transformation protocol to a clinical isolate of C. trachomatis belonging to the trachoma biovar, to provide “proof of principle” that it is possible to “knock out” selected plasmid genes (retaining a replication competent plasmid) and to investigate the plasticity of the plasmid. A recently developed, plasmid-based transformation protocol for LGV isolates of C. trachomatis was modified and a plasmid-free, genital tract C. trachomatis isolate from Sweden (SWFP-) was genetically transformed. Transformation of this non-LGV C. trachomatis host required a centrifugation step, but the absence of the natural plasmid removed the need for plaque purification of transformants. Transformants expressed GFP, were penicillin resistant and iodine stain positive for accumulated glycogen. The transforming plasmid did not recombine with the host chromosome. A derivative of pGFP::SW2 carrying a deletion of the plasmid CDS5 gene was engineered. CDS5 encodes pgp3, a protein secreted from the inclusion into the cell cytoplasm. This plasmid (pCDS5KO) was used to transform C. trachomatis SWFP-, and established that pgp3 is dispensable for plasmid function. The work shows it is possible to selectively delete segments of the chlamydial plasmid, and this is the first step towards a detailed molecular dissection of the role of the plasmid. The 3.6 kb β-galactosidase cassette was inserted into the deletion site of CDS5 to produce plasmid placZ-CDS5KO. Transformants were penicillin resistant, expressed GFP and stained for glycogen. In addition, they expressed β-galactosidase showing that the lacZ cassette was functional in C. trachomatis. An assay was developed that allowed the visualisation of individual inclusions by X-gal staining. The ability to express active β-galactosidase within chlamydial inclusions is an important advance as it allows simple, rapid assays to measure directly chlamydial infectivity without the need for plaquing, fluorescence or antibody staining.
Collapse
Affiliation(s)
- Yibing Wang
- Molecular Microbiology Group, University of Southampton, Southampton, United Kingdom
| | - Simona Kahane
- Department of Virology, Ben Gurion University of the Negev, Beer Sheva, Israel
| | - Lesley T. Cutcliffe
- Molecular Microbiology Group, University of Southampton, Southampton, United Kingdom
| | - Rachel J. Skilton
- Molecular Microbiology Group, University of Southampton, Southampton, United Kingdom
| | - Paul R. Lambden
- Molecular Microbiology Group, University of Southampton, Southampton, United Kingdom
| | - Kenneth Persson
- Department of Laboratory Medicine, Malmo University Hospital, Malmo, Sweden
| | - Carina Bjartling
- Department of Obstetrics and Gynaecology, Malmo University Hospital, Malmo, Sweden
| | - Ian N. Clarke
- Molecular Microbiology Group, University of Southampton, Southampton, United Kingdom
- * E-mail:
| |
Collapse
|
96
|
A major advance in elucidating the biology/pathobiology of Chlamydia trachomatis. Infect Immun 2013; 81:622-4. [PMID: 23319560 DOI: 10.1128/iai.00012-13] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
97
|
Chlamydia trachomatis plasmid-encoded Pgp4 is a transcriptional regulator of virulence-associated genes. Infect Immun 2013; 81:636-44. [PMID: 23319558 DOI: 10.1128/iai.01305-12] [Citation(s) in RCA: 139] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
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.
Collapse
|
98
|
Igietseme JU, Omosun Y, Partin J, Goldstein J, He Q, Joseph K, Ellerson D, Ansari U, Eko FO, Bandea C, Zhong G, Black CM. Prevention of Chlamydia-induced infertility by inhibition of local caspase activity. J Infect Dis 2013; 207:1095-104. [PMID: 23303804 DOI: 10.1093/infdis/jit009] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Tubal factor infertility (TFI) represents 36% of female infertility and genital infection by Chlamydia trachomatis (C. trachomatis) is a major cause. Although TFI is associated with host inflammatory responses to bacterial components, the molecular pathogenesis of Chlamydia-induced infertility remains poorly understood. We investigated the hypothesis that activation of specific cysteine proteases, the caspases, during C. trachomatis genital infection causes the disruption of key fertility-promoting molecules required for embryo development and implantation. We analyzed the effect of caspase inhibition on infertility and the integrity of Dicer, a caspase-sensitive, fertility-promoting ribonuclease III enzyme, and key micro-RNAs in the reproductive system. Genital infection with the inflammation- and caspase-inducing, wild-type C. trachomatis serovar L2 led to infertility, but the noninflammation-inducing, plasmid-free strain did not. We confirmed that caspase-mediated apoptotic tissue destruction may contribute to chlamydial pathogenesis. Caspase-1 or -3 deficiency, or local administration of the pan caspase inhibitor, Z-VAD-FMK into normal mice protected against Chlamydia-induced infertility. Finally, the oviducts of infected infertile mice showed evidence of caspase-mediated cleavage inactivation of Dicer and alteration in critical miRNAs that regulate growth, differentiation, and development, including mir-21. These results provide new insight into the molecular pathogenesis of TFI with significant implications for new strategies for treatment and prevention of chlamydial complications.
Collapse
Affiliation(s)
- Joseph U Igietseme
- National Center for Emerging Zoonotic and Infectious Diseases, Centers for Disease Control and Prevention (CDC), Atlanta, GA 30333, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
99
|
Schust DJ, Ibana JA, Buckner LR, Ficarra M, Sugimoto J, Amedee AM, Quayle AJ. Potential mechanisms for increased HIV-1 transmission across the endocervical epithelium during C. trachomatis infection. Curr HIV Res 2012; 10:218-27. [PMID: 22384841 DOI: 10.2174/157016212800618093] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2011] [Revised: 12/18/2011] [Accepted: 12/28/2011] [Indexed: 11/22/2022]
Abstract
Among the now pandemic sexually transmitted infections (STIs), Chlamydia trachomatis (C. trachomatis) is the predominant bacterial pathogen and human immunodeficiency virus type 1 (HIV-1) is the most lethal of the viral pathogens. The female genital tract is the primary site for heterosexual transmission of both C. trachomatis and HIV-1. Infection with C. trachomatis, and with a variety of other STIs, increases the risk for transmission of HIV-1, although the mechanisms for this finding remain unclear. We have used in vitro modeling to assess the mechanisms by which infection with genital C. trachomatis serovars might increase the transmission of HIV-1 across the female genital tract. C. trachomatis infection of an immortalized endocervical epithelial cell line (A2EN) increases the cell surface expression of the HIV-1 alternative primary receptor, galactosyl ceramide (GalCer), and of the HIV-1 co-receptors, CXCR4 and CCR5. C. trachomatis infection also increases the binding of HIV-1 to A2EN cells, and, subsequently, increases levels of virus in co-cultures of HIV-exposed A2EN and susceptible MT4-R5 T cells. Finally, in vivo endocervical cell sampling reveals a dramatic increase in the number of CD4+, CXCR4 and/or CCR5 positive T cell targets in the endocervix of C. trachomatis positive women when compared to those who are C. trachomatis negative. This combination of in vitro and in vivo results suggests several mechanisms for increased transmission of HIV-1 across the endocervices of C. trachomatis-infected women.
Collapse
Affiliation(s)
- Danny J Schust
- Department of Obstetrics, Gynecology and Women's Health, University of Missouri School of Medicine, Columbia, MO, USA.
| | | | | | | | | | | | | |
Collapse
|
100
|
Isolation of Chlamydia trachomatis and membrane vesicles derived from host and bacteria. J Microbiol Methods 2012; 91:222-30. [PMID: 22960504 DOI: 10.1016/j.mimet.2012.08.012] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2012] [Revised: 08/21/2012] [Accepted: 08/24/2012] [Indexed: 02/06/2023]
Abstract
The study of intracellular bacteria and nanometer-size membrane vesicles within infected host cells poses an important challenge as it is difficult to identify each distinct population in the context of the complex populations generated from active host-pathogen interactions. Here, suspension cultures of L929 cells infected with the prevalent obligate intracellular bacterium Chlamydia trachomatis strain F/Cal-IC-13 are utilized for the large scale preparation and isolation of natural membrane vesicles and bacterial forms. Cell lysis with nitrogen cavitation in combination with differential centrifugation, OptiPrep™ density gradient separation, and immunoenrichment using anti-chlamydial lipopolysaccharide antibodies and MagnaBind beads allows for the isolation of both productive and persistent bacterial forms, as well as membrane vesicles derived from the host and pathogen. We have evaluated these populations by electron microscopy and Western blot analysis for identification of biomarkers. In addition, purified persistent forms of C. trachomatis induced by ampicillin display adenosine-5'-triphosphate (ATP) transport activity, suggesting that ampicillin-induced persistent C. trachomatis organisms, at least in part, rely upon host ATP as an energy source. Importantly, several chlamydial cytotoxic and/or secreted proteins are demonstrated to be associated with these vesicles, supporting the idea that membrane vesicles are generated by Chlamydia as a means of carrying and delivering virulence factors necessary for pathogenesis. The ability to produce large-scale infections and generate distinct bacteria and host-derived populations for biochemical analysis, while reducing the burdens of time and cost have implications in all areas of chlamydiology. These protocols can be applied to other strains of C. trachomatis or other intracellular bacteria.
Collapse
|