1
|
Jin Y, Li W, Ba X, Li Y, Wang Y, Zhang H, Li Z, Zhou J. Gut microbiota changes in horses with Chlamydia. BMC Microbiol 2023; 23:246. [PMID: 37660043 PMCID: PMC10474637 DOI: 10.1186/s12866-023-02986-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 08/18/2023] [Indexed: 09/04/2023] Open
Abstract
BACKGROUND Zoonotic diseases pose a significant threat to public health. Chlamydia, as an intracellular pathogen, can colonize the intestinal tract of humans and animals, changing the gut microbiota. However, only a few studies have evaluated alterations in the gut microbiota of horses infected with Chlamydia. Therefore, this study aimed to investigate gut microbiota and serum biochemical indicators in horses with Chlamydial infection (IG) and healthy horses (HG). Fecal and blood samples were collected from 16 horses (IG: 10; HG: 6) before morning feeding for the determination of gut microbiota and serum biochemical parameters. RESULTS The results showed that total globulin (GLB), alanine aminotransferase (ALT), and creatine kinase (CK) levels were significantly increased in IG compared with HG. Notably, the gut microbial diversity increased in IG compared with HG. Furthermore, Moraxellaceae and Akkermanisa abundance decreased in IG, while Streptococcus, Treponema, Prevotella, and Paraprevotella abundances (13 genera of bacterial species) increased. Compared with HG, carbohydrate metabolism increased in IG while amino acid metabolism decreased. In addition, the abundance of 18 genera of bacteria was associated with the level of five serum biochemical indicators. CONCLUSIONS In summary, this study elucidated the influence of Chlamydia infection in horses on the gut microbiota, unraveling consequential alterations in its composition and metabolic profile. Therefore, this study improves the understanding of Chlamydia-induced intestinal infections.
Collapse
Affiliation(s)
- Youshun Jin
- State Key Laboratory for Animal Disease Control and Prevention, Key Laboratory of Veterinary Public Health of Agriculture Ministry Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730046, China
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou Veterinary Research Institute, Lanzhou University, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, China
| | - Wei Li
- State Key Laboratory for Animal Disease Control and Prevention, Key Laboratory of Veterinary Public Health of Agriculture Ministry Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730046, China
| | - Xuli Ba
- State Key Laboratory for Animal Disease Control and Prevention, Key Laboratory of Veterinary Public Health of Agriculture Ministry Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730046, China
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou Veterinary Research Institute, Lanzhou University, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, China
| | - Yunhui Li
- Animal Pathology Laboratory, College of Veterinary Medicine, Northwest A&F University, Xianyang, 712100, China
| | - Yanyan Wang
- State Key Laboratory for Animal Disease Control and Prevention, Key Laboratory of Veterinary Public Health of Agriculture Ministry Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730046, China
| | - Huaiyu Zhang
- Animal Pathology Laboratory, College of Veterinary Medicine, Northwest A&F University, Xianyang, 712100, China
| | - Zhaocai Li
- State Key Laboratory for Animal Disease Control and Prevention, Key Laboratory of Veterinary Public Health of Agriculture Ministry Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730046, China
| | - Jizhang Zhou
- State Key Laboratory for Animal Disease Control and Prevention, Key Laboratory of Veterinary Public Health of Agriculture Ministry Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730046, China.
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou Veterinary Research Institute, Lanzhou University, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, China.
| |
Collapse
|
2
|
Intracellular lifestyle of Chlamydia trachomatis and host-pathogen interactions. Nat Rev Microbiol 2023:10.1038/s41579-023-00860-y. [PMID: 36788308 DOI: 10.1038/s41579-023-00860-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/23/2023] [Indexed: 02/16/2023]
Abstract
In recent years, substantial progress has been made in the understanding of the intracellular lifestyle of Chlamydia trachomatis and how the bacteria establish themselves in the human host. As an obligate intracellular pathogenic bacterium with a strongly reduced coding capacity, C. trachomatis depends on the provision of nutrients from the host cell. In this Review, we summarize the current understanding of how C. trachomatis establishes its intracellular replication niche, how its metabolism functions in the host cell, how it can defend itself against the cell autonomous and innate immune response and how it overcomes adverse situations through the transition to a persistent state. In particular, we focus on those processes for which a mechanistic understanding has been achieved.
Collapse
|
3
|
Mayavannan A, Shantz E, Haidl ID, Wang J, Marshall JS. Mast cells selectively produce inflammatory mediators and impact the early response to Chlamydia reproductive tract infection. Front Immunol 2023; 14:1166068. [PMID: 37138882 PMCID: PMC10150091 DOI: 10.3389/fimmu.2023.1166068] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 03/28/2023] [Indexed: 05/05/2023] Open
Abstract
Introduction Chlamydia trachomatis (C. trachomatis) is a Gram-negative obligate intracellular bacterium that causes reproductive tract complications in women, including ectopic pregnancies and tubal factor infertility. We hypothesized that mast cells, which are common at mucosal barriers, may contribute to responses to Chlamydia infection and aimed to define human mast cell responses to C. trachomatis. Methods Human cord blood-derived mast cells (CBMCs) were exposed to C. trachomatis to assess bacterial uptake, mast cell degranulation, gene expression, and production of inflammatory mediators. The role of formyl peptide receptors and Toll-like receptor 2 (TLR2) were investigated using pharmacological inhibitors and soluble TLR2. Mast cell-deficient mice and littermate controls were used to examine the in vivo role of mast cells in influencing the immune response to Chlamydia infection in the female reproductive tract. Results C. trachomatis bacteria were taken up by human mast cells but did not replicate efficiently inside CBMCs. C. trachomatis-activated mast cells did not degranulate but maintained viability and exhibited cellular activation with homotypic aggregation and upregulation of ICAM-1. However, they significantly enhanced the gene expression of IL1B, CCL3, NFKB1, CXCL8, and IL6. Inflammatory mediators were produced, including TNF, IL-1β, IL-1RA, IL-6, GM-CSF, IL-23, CCL3, CCL5, and CXCL8. Endocytic blockade resulted in reduced gene expression of IL6, IL1B, and CCL3, suggesting C. trachomatis induced mast cell activation in both extracellular and intracellular locations. The IL-6 response to C. trachomatis was reduced when CBMCs were treated with C. trachomatis coated with soluble TLR2. Mast cells derived from TLR2-deficient mice also demonstrated a reduced IL-6 response to C. muridarum. Five days following C. muridarum infection, mast cell-deficient mice showed attenuated CXCL2 production and significantly reduced numbers of neutrophils, eosinophils, and B cells in the reproductive tract when compared with mast cell-containing littermates. Discussion Taken together, these data demonstrate that mast cells are reactive to Chlamydia spp. through multiple mechanisms that include TLR2-dependent pathways. Mast cells also play an important role in shaping in vivo immune responses in Chlamydia reproductive tract infection through both effector cell recruitment and modification of the chemokine microenvironment.
Collapse
Affiliation(s)
- Animamalar Mayavannan
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada
| | - Emily Shantz
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada
| | - Ian D. Haidl
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada
| | - Jun Wang
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada
- Canadian Center for Vaccinology, Izaak Walton Killam (IWK) Health Centre, Halifax, NS, Canada
| | - Jean S. Marshall
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada
- Department of Pathology, Dalhousie University, Halifax, NS, Canada
- *Correspondence: Jean S. Marshall,
| |
Collapse
|
4
|
Scharbaai-Vázquez R, J. López Font F, A. Zayas Rodríguez F. Persistence in Chlamydia. Infect Dis (Lond) 2022. [DOI: 10.5772/intechopen.109299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Chlamydia spp. are important causes of acute and persistent/chronic infections. All Chlamydia spp. display a unique biphasic developmental cycle alternating between an infectious elementary body (EB) and a replicative form, the reticulate body (RB), followed by the multiplication of RBs by binary fission and progressive differentiation back into EBs. During its intracellular life, Chlamydia employs multiple mechanisms to ensure its persistence inside the host. These include evasion of diverse innate immune responses, modulation of host cell structure and endocytosis, inhibition of apoptosis, activation of pro-signaling pathways, and conversion to enlarged, non-replicative but viable “aberrant bodies” (ABs). Early research described several systems for Chlamydial persistence with a significant number of variables that make a direct comparison of results difficult. Now, emerging tools for genetic manipulations in Chlamydia and advances in global microarray, transcriptomics, and proteomics have opened new and exciting opportunities to understand the persistent state of Chlamydia and link the immune and molecular events of persistence with the pathogenesis of recurrent and chronic Chlamydial infections. This chapter reviews our current understanding and advances in the molecular biology of Chlamydia persistence.
Collapse
|
5
|
Klasinc R, Battin C, Paster W, Reiter M, Schatzlmaier P, Rhein P, Spittler A, Steinberger P, Stockinger H. TLR4/CD14/MD2 Revealed as the Limited Toll-like Receptor Complex for Chlamydia trachomatis-Induced NF-κB Signaling. Microorganisms 2022; 10:microorganisms10122489. [PMID: 36557742 PMCID: PMC9783372 DOI: 10.3390/microorganisms10122489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 12/10/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022] Open
Abstract
Chlamydia trachomatis (Ct) is the most common cause of genital tract infections as well as preventable blindness worldwide. Pattern recognition receptors such as toll-like receptors (TLRs) represent the initial step in recognizing pathogenic microorganisms and are crucial for the initiation of an appropriate immune response. However, our understanding of TLR-signaling in Chlamydia-infected immune cells is incomplete. For a better comprehension of pathological inflammatory responses, robust models for interrogating TLR-signaling upon chlamydial infections are needed. To analyze the TLR response, we developed and utilized a highly sensitive and selective fluorescent transcriptional cellular reporter system to measure the activity of the transcription factor NF-κB. Upon incubation of the reporter cells with different preparations of Ct, we were able to pinpoint which components of TLRs are involved in the recognition of Ct. We identified CD14 associated with unique characteristics of different serovars as the crucial factor of the TLR4/CD14/MD2 complex for Ct-mediated activation of the NF-κB pathway. Furthermore, we found the TLR4/CD14/MD2 complex to be decisive for the uptake of Ct-derived lipopolysaccharides but not for infection and replication of Ct. Imaging flow cytometry provided information about inclusion formation in myeloid- as well as lymphocytic cells and was highest for Ct L2 with at least 25% of inclusion forming cells. Ct E inclusion formation was eminent in Jurkat cells without CD14 expression (11.1%). Thus, our model enables to determine Ct uptake and signal induction by pinpointing individual components of the recognition and signaling pathways to better understand the immune response towards infectious pathogens.
Collapse
Affiliation(s)
- Romana Klasinc
- Medical University of Vienna, Center for Pathophysiology, Infectiology and Immunology, Institute for Hygiene and Applied Immunology, 1090 Vienna, Austria
- Correspondence:
| | - Claire Battin
- Medical University of Vienna, Center for Pathophysiology, Infectiology and Immunology, Institute of Immunology, Division of Immune Receptors and T Cell Activation, 1090 Vienna, Austria
| | - Wolfgang Paster
- Medical University of Vienna, Center for Pathophysiology, Infectiology and Immunology, Institute of Immunology, Division of Immune Receptors and T Cell Activation, 1090 Vienna, Austria
- St. Anna Children’s Cancer Research Institute (CCRI), 1090 Vienna, Austria
| | - Michael Reiter
- Medical University of Vienna, Center for Pathophysiology, Infectiology and Immunology, Institute for Hygiene and Applied Immunology, 1090 Vienna, Austria
| | - Philipp Schatzlmaier
- Medical University of Vienna, Center for Pathophysiology, Infectiology and Immunology, Institute for Hygiene and Applied Immunology, 1090 Vienna, Austria
| | - Peter Rhein
- Luminex B.V., A DiaSorin Company, 5215 MV ‘s-Hertogenbosch, The Netherlands
| | - Andreas Spittler
- Medical University of Vienna, Core Facility Flow Cytometry and Department of Surgery, Research Laboratories, 1090 Vienna, Austria
| | - Peter Steinberger
- Medical University of Vienna, Center for Pathophysiology, Infectiology and Immunology, Institute of Immunology, Division of Immune Receptors and T Cell Activation, 1090 Vienna, Austria
| | - Hannes Stockinger
- Medical University of Vienna, Center for Pathophysiology, Infectiology and Immunology, Institute for Hygiene and Applied Immunology, 1090 Vienna, Austria
| |
Collapse
|
6
|
Xu Y, Wang J. Chlamydia
transmitting from the genital to gastrointestinal tract and inducing tubal disease: Double attack pattern. ZHONG NAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF CENTRAL SOUTH UNIVERSITY. MEDICAL SCIENCES 2022; 47:1275-1280. [PMID: 36411712 PMCID: PMC10930326 DOI: 10.11817/j.issn.1672-7347.2022.220023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Indexed: 06/16/2023]
Abstract
Chlamydia trachomatis ( CT ) genital tract infection is insidious, and patients often have no conscious symptoms.Delayed treatment after infection can lead to serious complications. Chlamydia muridarum ( CM ) genital tract infection in female mice can simulate CT genital tract infection in women, which is an ideal model to investigate the pathogenesis of CT . CM plasmid protein pGP3, chromosomal protein TC0237/TC0668, CM -specific CD8 + T cells, TNF-α, and IL-13 can induce genital tract inflammation, CD4 + T cells are responsible for CM clearance. However, tubal inflammation persists after genital tract CM is removed. Genital tract CM can spread spontaneously in vivo and colonize the gastrointestinal (GI) tract, but the GI tract CM cannot reverse spread to the genital tract. The survival time and number of CM transmitted from genital tract to GI tract are positively correlated with the long-term lesion of oviduct, while the CM inoculated directly into the GI tract has no pathogenicity in both the genital and GI tract. The double attack pattern of Chlamydia -induced genital tract inflammatory lesions is as follows: CM infection of oviduct epithelial cells initiates the process of oviduct repair as the first attack. After genital CM spreads to the GI tract, activated chlamydia-specific CD8 + T cells are recruited to the genital tract and secreted pro-fibrotic cytokines such as TNF-α and IL-13. This process is called the second attack which transform tubal repair initiated by the first attack into long-term tubal fibrosis/hydrosalpinx. Elucidating the pathogenic mechanism of Chlamydia infection can provide new ideas for the development of Chlamydia vaccine, which is expected to solve the problems of infertility caused by repeated CT infection in women.
Collapse
Affiliation(s)
- Ying Xu
- Department of Immunology, School of Basic Medical Science, Central South University, Changsha 410078, China.
| | - Jie Wang
- Department of Immunology, School of Basic Medical Science, Central South University, Changsha 410078, China.
| |
Collapse
|
7
|
Del Balzo D, Capmany A, Cebrian I, Damiani MT. Chlamydia trachomatis Infection Impairs MHC-I Intracellular Trafficking and Antigen Cross-Presentation by Dendritic Cells. Front Immunol 2021; 12:662096. [PMID: 33936099 PMCID: PMC8082151 DOI: 10.3389/fimmu.2021.662096] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 03/26/2021] [Indexed: 11/21/2022] Open
Abstract
During cross-presentation, exogenous antigens (i.e. intracellular pathogens or tumor cells) are internalized and processed within the endocytic system and also by the proteasome in the cytosol. Then, antigenic peptides are associated with Major Histocompatibility Complex (MHC) class I molecules and these complexes transit to the plasma membrane in order to trigger cytotoxic immune responses through the activation of CD8+ T lymphocytes. Dendritic cells (DCs) are particularly adapted to achieve efficient antigen cross-presentation and their endocytic network displays important roles during this process, including a sophisticated MHC-I transport dependent on recycling compartments. In this study, we show that C. trachomatis, an obligate intracellular pathogen that exhibits multiple strategies to evade the immune system, is able to induce productive infections in the murine DC line JAWS-II. Our results show that when C. trachomatis infects these cells, the bacteria-containing vacuole strongly recruits host cell recycling vesicles, but no other endosomal compartments. Furthermore, we found that chlamydial infection causes significant alterations of MHC-I trafficking in JAWS-II DCs: reduced levels of MHC-I expression at the cell surface, disruption of the perinuclear MHC-I intracellular pool, and impairment of MHC-I endocytic recycling to the plasma membrane. We observed that all these modifications lead to a hampered cross-presentation ability of soluble and particulate antigens by JAWS-II DCs and primary bone marrow-derived DCs. In summary, our findings provide substantial evidence that C. trachomatis hijacks the DC endocytic recycling system, causing detrimental changes on MHC-I intracellular transport, which are relevant for competent antigen cross-presentation.
Collapse
Affiliation(s)
- Diego Del Balzo
- Biochemistry and Immunity Laboratory, School of Medicine, University of Cuyo, IMBECU-CONICET, Centro Universitario, Mendoza, Argentina
| | - Anahí Capmany
- Biochemistry and Immunity Laboratory, School of Medicine, University of Cuyo, IMBECU-CONICET, Centro Universitario, Mendoza, Argentina
| | - Ignacio Cebrian
- Instituto de Histología y Embriología de Mendoza (IHEM)-CONICET, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina
| | - María Teresa Damiani
- Biochemistry and Immunity Laboratory, School of Medicine, University of Cuyo, IMBECU-CONICET, Centro Universitario, Mendoza, Argentina
| |
Collapse
|
8
|
Mott PD, Taylor CM, Lillis RA, Ardizzone CM, Albritton HL, Luo M, Calabresi KG, Martin DH, Myers L, Quayle AJ. Differences in the Genital Microbiota in Women Who Naturally Clear Chlamydia trachomatis Infection Compared to Women Who Do Not Clear; A Pilot Study. Front Cell Infect Microbiol 2021; 11:615770. [PMID: 33912473 PMCID: PMC8072278 DOI: 10.3389/fcimb.2021.615770] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 03/03/2021] [Indexed: 02/06/2023] Open
Abstract
In vitro studies indicate IFNγ is central to Chlamydia trachomatis (Ct) eradication, but its function may be compromised by anaerobes typically associated with bacterial vaginosis (BV), a frequent co-morbidity in women with Ct. Here we investigated the associations between natural clearance of cervical Ct infection, the vaginal microbiome, and the requirements for IFNγ by evaluating the vaginal microbial and cytokine composition of Ct treatment visit samples from women who cleared Ct infection in the interim between their Ct screening and Ct treatment visit. The pilot cohort was young, predominantly African American, and characterized by a high rate of BV that was treated with metronidazole at the Ct screening visit. The rate of natural Ct clearance was 23.6% by the Ct treatment visit (median 9 days). 16S rRNA gene sequencing revealed that metronidazole-treated women who had a Lactobacillus spp.-dominant vaginal microbiota (CST 2 or 3) at the Ct treatment visit, were more prevalent in the Ct clearing population than the non-clearing population (86% v. 50%). L. iners (CST2) was the major Lactobacillus spp. present in Ct clearers, and 33% still remained anaerobe-dominant (CST1). Vaginal IFNγ levels were not significantly different in Ct clearers and non-clearers and were several logs lower than that required for killing Ct in vitro. An expanded panel of IFNγ-induced and proinflammatory cytokines and chemokines also did not reveal differences between Ct clearers and non-clearers, but, rather, suggested signatures better associated with specific CSTs. Taken together, these findings suggest that BV-associated bacteria may impede Ct clearance, but a Lactobacillus spp.-dominant microbiome is not an absolute requirement to clear. Further, IFNγ may be required at lower concentrations than in vitro modeling indicates, suggesting it may act together with other factors in vivo. Data also revealed that the vaginal bacteria-driven inflammation add complexity to the genital cytokine milieu, but changes in this microbiota may contribute to, or provide cytokine biomarkers, for a shift to Ct clearance.
Collapse
Affiliation(s)
- Patricia Dehon Mott
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Christopher M. Taylor
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Rebecca A. Lillis
- Division of Infectious Diseases, Department of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Caleb M. Ardizzone
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Hannah L. Albritton
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Meng Luo
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Kaitlyn G. Calabresi
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - David H. Martin
- Division of Infectious Diseases, Department of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Leann Myers
- Department of Biostatistics and Data Science, Tulane University School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, United States
| | - Alison J. Quayle
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| |
Collapse
|
9
|
Karunakaran KP, Yu H, Jiang X, Chan QWT, Foster LJ, Johnson RM, Brunham RC. Discordance in the Epithelial Cell-Dendritic Cell Major Histocompatibility Complex Class II Immunoproteome: Implications for Chlamydia Vaccine Development. J Infect Dis 2020; 221:841-850. [PMID: 31599954 DOI: 10.1093/infdis/jiz522] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 10/05/2019] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Chlamydia trachomatis and Chlamydia muridarum are intracellular bacterial pathogens of mucosal epithelial cells. CD4 T cells and major histocompatibility complex (MHC) class II molecules are essential for protective immunity against them. Antigens presented by dendritic cells (DCs) expand naive pathogen-specific T cells (inductive phase), whereas antigens presented by epithelial cells identify infected epithelial cells as targets during the effector phase. We previously showed that DCs infected by C trachomatis or C muridarum present epitopes from a limited spectrum of chlamydial proteins recognized by Chlamydia-specific CD4 T cells from immune mice. METHODS We hypothesized that Chlamydia-infected DCs and epithelial cells present overlapping sets of Chlamydia-MHC class II epitopes to link inductive and effector phases to generate protective immunity. We tested that hypothesis by infecting an oviductal epithelial cell line with C muridarum, followed by immunoaffinity isolation and sequencing of MHC class I- and II-bound peptides. RESULTS We identified 26 class I-bound and 4 class II-bound Chlamydia-derived peptides from infected epithelial cells. We were surprised to find that none of the epithelial cell class I- and class II-bound chlamydial peptides overlapped with peptides presented by DCs. CONCLUSIONS We suggest the discordance between the DC and epithelial cell immunoproteomes has implications for delayed clearance of Chlamydia and design of a Chlamydia vaccine.
Collapse
Affiliation(s)
- Karuna P Karunakaran
- Vaccine Research Laboratory, University of British Columbia Centre for Disease Control, Vancouver, British Columbia, Canada
| | - Hong Yu
- Vaccine Research Laboratory, University of British Columbia Centre for Disease Control, Vancouver, British Columbia, Canada
| | - Xiaozhou Jiang
- Vaccine Research Laboratory, University of British Columbia Centre for Disease Control, Vancouver, British Columbia, Canada
| | - Queenie W T Chan
- Department of Biochemistry and Molecular Biology, Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada
| | - Leonard J Foster
- Department of Biochemistry and Molecular Biology, Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada
| | - Raymond M Johnson
- Section of Infectious Diseases, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Robert C Brunham
- Vaccine Research Laboratory, University of British Columbia Centre for Disease Control, Vancouver, British Columbia, Canada
| |
Collapse
|
10
|
Yadav S, Verma V, Singh Dhanda R, Yadav M. Insights into the toll-like receptors in sexually transmitted infections. Scand J Immunol 2020; 93:e12954. [PMID: 32762084 DOI: 10.1111/sji.12954] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 07/10/2020] [Accepted: 08/01/2020] [Indexed: 12/12/2022]
Abstract
Toll-like receptors (TLRs) are like soldiers of an innate immune system, which protects vital biological processes against invading pathogens. TLR signalling pathways help in the removal of pathogens and mediate well-established inflammatory processes. However, these processes may also aid in the development or augmentation of an infection or an autoimmune disease. Recent studies have delineated TLR polymorphism's role in the loss of function, making hosts more resistant or vulnerable to the development of an infection. In this review, we have discussed the association of TLRs with sexually transmitted infections (STIs), especially to the pathogen-specific ligands. We have also assessed the impact on TLR downstream signalling and the maintenance of cellular homeostasis during immune responses. Besides, we have discussed the role of TLRs single nucleotide polymorphisms in various STIs. Since TLRs are known to play a part in defence mechanisms and in aiding infections therefore, a thorough understanding of TLRs structure and molecular mechanisms is required to explain how they can influence the outcome of an STI. Such a strategy may lead to the development of novel and useful immunotherapeutic approaches to control pathogen progression and prevent transmission.
Collapse
Affiliation(s)
- Sonal Yadav
- Dr. B. R. Ambedkar Center for Biomedical Research (ACBR), University of Delhi, Delhi, India
| | - Vivek Verma
- Dr. B. R. Ambedkar Center for Biomedical Research (ACBR), University of Delhi, Delhi, India
| | | | - Manisha Yadav
- Dr. B. R. Ambedkar Center for Biomedical Research (ACBR), University of Delhi, Delhi, India.,Department of Clinical Microbiology, Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
11
|
Cheok YY, Lee CYQ, Cheong HC, Looi CY, Wong WF. Chronic Inflammatory Diseases at Secondary Sites Ensuing Urogenital or Pulmonary Chlamydia Infections. Microorganisms 2020; 8:microorganisms8010127. [PMID: 31963395 PMCID: PMC7022716 DOI: 10.3390/microorganisms8010127] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Revised: 01/02/2020] [Accepted: 01/08/2020] [Indexed: 12/11/2022] Open
Abstract
Chlamydia trachomatis and C. pneumoniae are members of the Chlamydiaceae family of obligate intracellular bacteria. The former causes diseases predominantly at the mucosal epithelial layer of the urogenital or eye, leading to pelvic inflammatory diseases or blindness; while the latter is a major causative agent for pulmonary infection. On top of these well-described diseases at the respective primary infection sites, Chlamydia are notoriously known to migrate and cause pathologies at remote sites of a host. One such example is the sexually acquired reactive arthritis that often occurs at few weeks after genital C. trachomatis infection. C. pneumoniae, on the other hand, has been implicated in an extensive list of chronic inflammatory diseases which include atherosclerosis, multiple sclerosis, Alzheimer’s disease, asthma, and primary biliary cirrhosis. This review summarizes the Chlamydia infection associated diseases at the secondary sites of infection, and describes the potential mechanisms involved in the disease migration and pathogenesis.
Collapse
Affiliation(s)
- Yi Ying Cheok
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, Kuala Lumpur 50603, Malaysia; (Y.Y.C.); (C.Y.Q.L.); (H.C.C.)
| | - Chalystha Yie Qin Lee
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, Kuala Lumpur 50603, Malaysia; (Y.Y.C.); (C.Y.Q.L.); (H.C.C.)
| | - Heng Choon Cheong
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, Kuala Lumpur 50603, Malaysia; (Y.Y.C.); (C.Y.Q.L.); (H.C.C.)
| | - Chung Yeng Looi
- School of Biosciences, Faculty of Health and Medical Sciences, Taylor’s University, Subang Jaya 47500, Malaysia;
| | - Won Fen Wong
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, Kuala Lumpur 50603, Malaysia; (Y.Y.C.); (C.Y.Q.L.); (H.C.C.)
- Correspondence: ; Tel.: +603-7967-6672
| |
Collapse
|
12
|
Faris R, Andersen SE, McCullough A, Gourronc F, Klingelhutz AJ, Weber MM. Chlamydia trachomatis Serovars Drive Differential Production of Proinflammatory Cytokines and Chemokines Depending on the Type of Cell Infected. Front Cell Infect Microbiol 2019; 9:399. [PMID: 32039039 PMCID: PMC6988789 DOI: 10.3389/fcimb.2019.00399] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 11/07/2019] [Indexed: 12/22/2022] Open
Abstract
Chlamydia trachomatis serovars A-C infect conjunctival epithelial cells and untreated infection can lead to blindness. D-K infect genital tract epithelial cells resulting in pelvic inflammatory disease, ectopic pregnancy, and sterility while L1-L3 infect epithelial cells and macrophages, causing an invasive infection. Despite some strains of Chlamydia sharing high nucleotide sequence similarity, the bacterial and host factors that govern tissue and cellular tropism remain largely unknown. Following introduction of C. trachomatis via intercourse, epithelial cells of the vagina, foreskin, and ectocervix are exposed to large numbers of the pathogen, yet their response to infection and the dynamics of chlamydial growth in these cells has not been well-characterized compared to growth in more permissive cell types that harbor C. trachomatis. We compared intracellular replication and inclusion development of representative C. trachomatis serovars in immortalized human conjunctival epithelial, urogenital epithelial, PMA stimulated THP-1 (macrophages), and HeLa cells. We demonstrate that urogenital epithelial cells of the vagina, ectocervix, and foreskin restrict replication of serovar A while promoting robust replication and inclusion development of serovar D and L2. Macrophages restrict serovars D and A while L2 proliferates in these cells. Furthermore, we show that GM-CSF, RANTES, GROα, IL-1α, IL-1β, IP-10, IL-8, and IL-18 are produced in a cell-type and serovar-specific manner. Collectively we have established a series of human cell lines that represent some of the first cell types to encounter C. trachomatis following exposure and show that differential production of key cytokines early during infection could regulate serovar-host cell specificity.
Collapse
Affiliation(s)
- Robert Faris
- Department of Microbiology and Immunology, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Shelby E Andersen
- Department of Microbiology and Immunology, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Alix McCullough
- Department of Microbiology and Immunology, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Françoise Gourronc
- Department of Microbiology and Immunology, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Aloysius J Klingelhutz
- Department of Microbiology and Immunology, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Mary M Weber
- Department of Microbiology and Immunology, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| |
Collapse
|
13
|
Ziklo N, Huston WM, Taing K, Timms P. High expression of IDO1 and TGF-β1 during recurrence and post infection clearance with Chlamydia trachomatis, are independent of host IFN-γ response. BMC Infect Dis 2019; 19:218. [PMID: 30832593 PMCID: PMC6398247 DOI: 10.1186/s12879-019-3843-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 02/21/2019] [Indexed: 11/11/2022] Open
Abstract
Background Chlamydia trachomatis infections in women continue to be a major public health concern due to their high prevalence and consequent reproductive morbidities. While antibiotics are usually efficient to clear the Chlamydia, repeat infections are common and may contribute to pathological outcomes. Interferon-gamma (IFN-γ)-mediated immunity has been suggested to be protective against reinfection, and represent an important anti-chlamydial agent, primarily via the induction of indoleamine-2,3 dioxygenase 1 (IDO1) enzyme. IDO1 catalyzes the degradation of tryptophan, which can eliminate C. trachomatis infection in vitro. Here, we sought to measure IDO1 expression levels and related immune markers during different C. trachomatis infection statuses (repeated vs single infection vs post antibiotic treatment), in vitro and in vivo. Methods In this study, we measured the expression levels of IDO1 and immune regulatory markers, transforming growth factor β1 (TGF-β1) and forkhead box P3 (FoxP3), in vaginal swab samples of C. trachomatis-infected women, with either single or repeated infection. In addition, we used an in vitro co-culture model of endometrial carcinoma cell-line and peripheral blood mononuclear cells (PBMCs) to measure the same immune markers. Results We found that in women with repeated C. trachomatis infections vaginal IDO1 and TGF-β1 expression levels were significantly increased. Whereas, women who cleared their infection post antibiotic treatment, had increased levels of IDO1 and TGF-β1, as well as FoxP3. Similarly, using the in vitro model, we found significant upregulation of IDO1 and TGF-β1 levels in the co-culture infected with C. trachomatis. Furthermore, we found that in PBMCs infected with C. trachomatis there was a significant upregulation in IDO1 levels, which was independent of IFN-γ. In fact, C. trachomatis infection in PBMCs failed to induce IFN-γ levels in comparison to the uninfected culture. Conclusions Our data provide evidence for a regulatory immune response comprised of IDO1, TGF-β1 and FoxP3 in women post antibiotic treatment. In this study, we demonstrated a significant increase in IDO1 expression levels in response to C. trachomatis infection, both in vivo and in vitro, without elevated IFN-γ levels. This study implicates IDO1 and TGF-β1 as part of the immune response to repeated C. trachomatis infections, independently of IFN-γ. Electronic supplementary material The online version of this article (10.1186/s12879-019-3843-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Noa Ziklo
- Faculty of Science, Health, Education & Engineering, University of the Sunshine Coast, Sippy Downs, Sunshine Coast, QLD, Australia.
| | - Wilhelmina M Huston
- School of Life Sciences, Faculty of Science, University of Technology, Sydney, Australia
| | - Kuong Taing
- Sunshine Coast Sexual Health and HIV Service (Clinic 87), Nambour, Sunshine Coast, QLD, Australia
| | - Peter Timms
- Faculty of Science, Health, Education & Engineering, University of the Sunshine Coast, Sippy Downs, Sunshine Coast, QLD, Australia
| |
Collapse
|
14
|
Rothschild P, Chen Y, Wells K, Sherwin JC, Crock C. Has the increasing incidence of chlamydia and gonorrhoea resulted in increased chlamydial and gonococcal conjunctivitis presentations? Results from Melbourne, Australia, from 2000 to 2017. Clin Exp Ophthalmol 2019; 47:289-291. [DOI: 10.1111/ceo.13373] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Accepted: 08/04/2018] [Indexed: 11/27/2022]
Affiliation(s)
- Philip Rothschild
- Launceston Clinical SchoolThe University of Tasmania Launceston Tasmania Australia
| | - Ye Chen
- Emergency DepartmentRoyal Victorian Eye and Ear Hospital Melbourne Victoria Australia
| | - Kristen Wells
- Emergency DepartmentRoyal Victorian Eye and Ear Hospital Melbourne Victoria Australia
| | - Justin C. Sherwin
- Emergency DepartmentRoyal Victorian Eye and Ear Hospital Melbourne Victoria Australia
| | - Carmel Crock
- Emergency DepartmentRoyal Victorian Eye and Ear Hospital Melbourne Victoria Australia
| |
Collapse
|
15
|
Li LY, Zhang HR, Jiang ZL, Chang YZ, Shao CZ. Overexpression of Dendritic Cell-Specific Intercellular Adhesion Molecule-3-Grabbing Nonintegrin in Dendritic Cells Protecting against Aspergillosis. Chin Med J (Engl) 2019; 131:2575-2582. [PMID: 30381591 PMCID: PMC6213851 DOI: 10.4103/0366-6999.244103] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Background: Dendritic cells (DCs) play an important role in host defense against pathogen infection. DC-specific intercellular adhesion molecule-3-grabbing nonintegrin (SIGN) is a group II C-type lectin receptor and specifically expressed on the surface of DCs. This study aimed to determine whether DC-SIGN affects intracellular signaling activation, Th1/Th2 imbalance and aspergillus immune evasion in aspergillus infection, and explore the application of DC-SIGN-modified DCs in immunotherapy. Methods: DCs were first obtained from the mononuclear cells of peripheral blood. The interferon (IFN)-γ and dexamethasone (Dex) were used to stimulate DCs. The expression of DC-SIGN, Th1 and Th2 cytokines, and the capacity of DCs in stimulating T cells proliferation and phagocytosis, and nuclear factor (NF)-κB activation were analyzed. In addition, adenovirus expression vector Ad-DC-SIGN was generated to transfect DCs. Mannan was used to block DC-SIGN signaling for confirming the involvement of DC-SIGN function in Aspergillus fumigatus (Af)-induced DCs maturation. The unpaired, two-tailed Student's t-test was used in the comparisons between two groups. Results: Exogenous IFN-γ could activate Af-induced DCs and promote the Th0 cells toward Th1 profile (interleukin [IL]-12 in IFN-γ/Af group: 50.96 ± 4.38 pg/ml; control/Af group: 29.70 ± 2.00 pg/ml, t = 10.815, P < 0.001). On the other hand, Dex inhibited the secretion of Th2 cytokines (IL-10 in Dex/Af group: 5.27 ± 0.85 pg/ml; control/Af group: 15.14 ± 1.40 pg/ml, t = 14.761, P < 0.001)), and successfully caused immunosuppression. After transfection with Ad-DC-SIGN, DCs have improved phagocytosis (phagocytosis rates in Ad-DC-SIGN group: 74.0% ± 3.4%; control group: 64.7% ± 6.8%, t = 3.104, P = 0.013). There was more Th1 cytokine secreted in the Af-induced DC-SIGN modified DCs (IL-12 in Ad-DC-SIGN/Af group: 471.98 ± 166.31 pg/ml; control/Af group: 33.35 ± 5.98 pg/ml, t = 6.456, P = 0.001), correlated to the enhanced NF-κB activation. Conclusion: Overexpressing DC-SIGN in DCs had a protective function on aspergillosis.
Collapse
Affiliation(s)
- Li-Yang Li
- Department of Pulmonary Medicine, Shanghai Institute of Respiratory Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Hao-Ru Zhang
- Laboratory of Molecular Iron Metabolism, The Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life Science, Hebei Normal University, Shijiazhuang, Hebei 050024, China
| | - Zhi-Long Jiang
- Department of Pulmonary Medicine, Shanghai Institute of Respiratory Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yan-Zhong Chang
- Laboratory of Molecular Iron Metabolism, The Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life Science, Hebei Normal University, Shijiazhuang, Hebei 050024, China
| | - Chang-Zhou Shao
- Department of Pulmonary Medicine, Shanghai Institute of Respiratory Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| |
Collapse
|
16
|
Lausen M, Christiansen G, Bouet Guldbæk Poulsen T, Birkelund S. Immunobiology of monocytes and macrophages during Chlamydia trachomatis infection. Microbes Infect 2018; 21:73-84. [PMID: 30528899 DOI: 10.1016/j.micinf.2018.10.007] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 10/11/2018] [Accepted: 10/11/2018] [Indexed: 02/03/2023]
Abstract
Infections caused by the intracellular bacterium Chlamydia trachomatis are a global health burden affecting more than 100 million people annually causing damaging long-lasting infections. In this review, we will present and discuss important aspects of the interaction between C. trachomatis and monocytes/macrophages.
Collapse
Affiliation(s)
- Mads Lausen
- Department of Health Science and Technology, Aalborg University, Fredrik Bajers Vej 3b, 9220 Aalborg Ø, Denmark.
| | - Gunna Christiansen
- Department of Biomedicine, Aarhus University, Wilhelms Meyers Allé 4, 8000 Aarhus, Denmark
| | | | - Svend Birkelund
- Department of Health Science and Technology, Aalborg University, Fredrik Bajers Vej 3b, 9220 Aalborg Ø, Denmark
| |
Collapse
|
17
|
Lantos I, Virok DP, Mosolygó T, Rázga Z, Burián K, Endrész V. Growth characteristics of Chlamydia trachomatis in human intestinal epithelial Caco-2 cells. Pathog Dis 2018; 76:4939475. [PMID: 29635314 DOI: 10.1093/femspd/fty024] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Accepted: 03/15/2018] [Indexed: 12/13/2022] Open
Abstract
Chlamydia trachomatis is an obligate intracellular bacterium causing infections of the eyes, urogenital and respiratory tracts. Asymptomatic, repeat and chronic infections with C. trachomatis are common in the urogenital tract potentially causing severe reproductive pathology. Animal models of infection and epidemiological studies suggested the gastrointestinal tract as a reservoir of chlamydiae and as a source of repeat urogenital infections. Thus, we investigated the growth characteristics of C. trachomatis in human intestinal epithelial Caco-2 cells and the infection-induced defensin production. Immunofluorescence staining and transmission electron microscopy showed the presence of chlamydial inclusions in the cells. Chlamydial DNA and viable C. trachomatis were recovered from Caco-2 cells in similar quantity compared to that detected in the usual in vitro host cell of this bacterium. The kinetics of expression of selected C. trachomatis genes in Caco-2 cells indicated prolonged replication with persisting high expression level of late genes and of heat shock protein gene groEL. Replication of C. trachomatis induced moderate level of β-defensin-2 production by Caco-2 cells, which might contribute to avoidance of immune recognition in the intestine. According to our results, Caco-2 cells support C. trachomatis replication, suggesting that the gastrointestinal tract is a site of residence for these bacteria.
Collapse
Affiliation(s)
- Ildikó Lantos
- Department of Medical Microbiology and Immunobiology, University of Szeged, H-6720, Szeged, Dóm Sq. 10, Hungary
| | - Dezso P Virok
- Department of Medical Microbiology and Immunobiology, University of Szeged, H-6720, Szeged, Dóm Sq. 10, Hungary
| | - Tímea Mosolygó
- Department of Medical Microbiology and Immunobiology, University of Szeged, H-6720, Szeged, Dóm Sq. 10, Hungary
| | - Zsolt Rázga
- Department of Pathology, University of Szeged, H-6720, Állomás Str. 2, Hungary
| | - Katalin Burián
- Department of Medical Microbiology and Immunobiology, University of Szeged, H-6720, Szeged, Dóm Sq. 10, Hungary
| | - Valéria Endrész
- Department of Medical Microbiology and Immunobiology, University of Szeged, H-6720, Szeged, Dóm Sq. 10, Hungary
| |
Collapse
|
18
|
Mackern-Oberti JP, Motrich RD, Damiani MT, Saka HA, Quintero CA, Sánchez LR, Moreno-Sosa T, Olivera C, Cuffini C, Rivero VE. Male genital tract immune response against Chlamydia trachomatis infection. Reproduction 2018; 154:R99-R110. [PMID: 28878094 DOI: 10.1530/rep-16-0561] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Revised: 07/01/2017] [Accepted: 07/11/2017] [Indexed: 01/01/2023]
Abstract
Chlamydia trachomatis is the most commonly reported agent of sexually transmitted bacterial infections worldwide. This pathogen frequently leads to persistent, long-term, subclinical infections, which in turn may cause severe pathology in susceptible hosts. This is in part due to the strategies that Chlamydia trachomatis uses to survive within epithelial cells and to evade the host immune response, such as subverting intracellular trafficking, interfering signaling pathways and preventing apoptosis. Innate immune receptors such as toll-like receptors expressed on epithelial and immune cells in the genital tract mediate the recognition of chlamydial molecular patterns. After bacterial recognition, a subset of pro-inflammatory cytokines and chemokines are continuously released by epithelial cells. The innate immune response is followed by the initiation of the adaptive response against Chlamydia trachomatis, which in turn may result in T helper 1-mediated protection or in T helper 2-mediated immunopathology. Understanding the molecular mechanisms developed by Chlamydia trachomatis to avoid killing and host immune response would be crucial for designing new therapeutic approaches and developing protective vaccines. In this review, we focus on chlamydial survival strategies and the elicited immune responses in male genital tract infections.
Collapse
Affiliation(s)
- Juan Pablo Mackern-Oberti
- Instituto de Medicina y Biología Experimental de Cuyo. IMBECU-CONICETMendoza, Argentina .,Instituto de Fisiología. Facultad de Ciencias MédicasUniversidad Nacional de Cuyo, Mendoza, Argentina
| | - Rubén Darío Motrich
- Centro de Investigaciones en Bioquímica Clínica e Inmunología CIBICI-CONICETDepartamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Maria Teresa Damiani
- Instituto de Histología y Embriología de Mendoza. IHEM-CONICETFacultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Héctor Alex Saka
- Centro de Investigaciones en Bioquímica Clínica e Inmunología CIBICI-CONICETDepartamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | | | - Leonardo Rodolfo Sánchez
- Centro de Investigaciones en Bioquímica Clínica e Inmunología CIBICI-CONICETDepartamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Tamara Moreno-Sosa
- Instituto de Medicina y Biología Experimental de Cuyo. IMBECU-CONICETMendoza, Argentina
| | - Carolina Olivera
- Centro de Investigaciones en Bioquímica Clínica e Inmunología CIBICI-CONICETDepartamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Cecilia Cuffini
- Instituto de Virología Dr. J. M. VanellaFacultad de Ciencias Médicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Virginia Elena Rivero
- Centro de Investigaciones en Bioquímica Clínica e Inmunología CIBICI-CONICETDepartamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| |
Collapse
|
19
|
Lausen M, Christiansen G, Karred N, Winther R, Poulsen TBG, Palarasah Y, Birkelund S. Complement C3 opsonization of Chlamydia trachomatis facilitates uptake in human monocytes. Microbes Infect 2018; 20:328-336. [PMID: 29729435 DOI: 10.1016/j.micinf.2018.04.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2017] [Revised: 03/13/2018] [Accepted: 04/23/2018] [Indexed: 01/08/2023]
Abstract
Chlamydia trachomatis is an obligate intracellular bacterium that causes severe infections, which can lead to infertility and ectopic pregnancy. Although both innate and adaptive immune responses are elicited during chlamydial infection the bacterium succeeds to evade host defense mechanisms establishing chronic infections. Thus, studying the host-pathogen interaction during chlamydial infection is of importance to understand how C. trachomatis can cause chronic infections. Both the complement system and monocytes play essential roles in anti-bacterial defense, and, therefore, we investigated the interaction between the complement system and the human pathogens C. trachomatis D and L2. Complement competent serum facilitated rapid uptake of both chlamydial serovars into monocytes. Using immunoelectron microscopy, we showed that products of complement C3 were loosely deposited on the bacterial surface in complement competent serum and further characterization demonstrated that the deposited C3 product was the opsonin iC3b. Using C3-depleted serum we confirmed that complement C3 facilitates rapid uptake of chlamydiae into monocytes in complement competent serum. Complement facilitated uptake did not influence intracellular survival of C. trachomatis or C. trachomatis-induced cytokine secretion. Hence, C. trachomatis D and L2 activate the complement system leading to chlamydial opsonization by iC3b and subsequent phagocytosis, activation and bacterial elimination by human monocytes.
Collapse
Affiliation(s)
- Mads Lausen
- Department of Health Science and Technology, Aalborg University, Fredrik Bajers Vej 3b, 9220, Aalborg Ø, Denmark
| | - Gunna Christiansen
- Department of Biomedicine, Aarhus University, Wilhelms Meyers Allé 4, 8000, Aarhus, Denmark
| | - Nichlas Karred
- Department of Health Science and Technology, Aalborg University, Fredrik Bajers Vej 3b, 9220, Aalborg Ø, Denmark
| | - Robert Winther
- Department of Health Science and Technology, Aalborg University, Fredrik Bajers Vej 3b, 9220, Aalborg Ø, Denmark
| | - Thomas Bouet Guldbæk Poulsen
- Department of Health Science and Technology, Aalborg University, Fredrik Bajers Vej 3b, 9220, Aalborg Ø, Denmark
| | - Yaseelan Palarasah
- Unit for Thrombosis Research, Institute of Public Health, University of Southern Denmark, Esbjerg, Denmark
| | - Svend Birkelund
- Department of Health Science and Technology, Aalborg University, Fredrik Bajers Vej 3b, 9220, Aalborg Ø, Denmark.
| |
Collapse
|
20
|
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: 35] [Impact Index Per Article: 5.0] [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
|
21
|
Leonard CA, Schoborg RV, Borel N. Productive and Penicillin-Stressed Chlamydia pecorum Infection Induces Nuclear Factor Kappa B Activation and Interleukin-6 Secretion In Vitro. Front Cell Infect Microbiol 2017; 7:180. [PMID: 28553623 PMCID: PMC5425588 DOI: 10.3389/fcimb.2017.00180] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 04/25/2017] [Indexed: 11/13/2022] Open
Abstract
Nuclear factor kappa B (NFκB) is an inflammatory transcription factor that plays an important role in the host immune response to infection. The potential for chlamydiae to activate NFκB has been an area of interest, however most work has focused on chlamydiae impacting human health. Given that inflammation characteristic of chlamydial infection may be associated with severe disease outcomes or contribute to poor overall fitness in farmed animals, we evaluated the ability of porcine chlamydiae to induce NFκB activation in vitro. C. pecorum infection induced both NFκB nuclear translocation and activation at 2 hours post infection (hpi), an effect strongly enhanced by suppression of host de novo protein synthesis. C. suis and C. trachomatis showed less capacity for NFκB activation compared to C. pecorum, suggesting a species-specific variation in NFκB activation. At 24 hpi, C. pecorum induced significant NFκB activation, an effect not abolished by penicillin (beta lactam)-induced chlamydial stress. C. pecorum-dependent secretion of interleukin 6 was also detected in the culture supernatant of infected cells at 24 hpi, and this effect, too, was unchanged by penicillin-induced chlamydial stress. Taken together, these results suggest that NFκB participates in the early inflammatory response to C. pecorum and that stressed chlamydiae can promote inflammation.
Collapse
Affiliation(s)
- Cory A Leonard
- Department of Pathobiology, Institute of Veterinary Pathology, University of ZurichZurich, Switzerland
| | - Robert V Schoborg
- Department of Biomedical Sciences, Center for Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State UniversityJohnson City, TN, USA
| | - Nicole Borel
- Department of Pathobiology, Institute of Veterinary Pathology, University of ZurichZurich, Switzerland
| |
Collapse
|
22
|
Intravenous Inoculation with Chlamydia muridarum Leads to a Long-Lasting Infection Restricted to the Gastrointestinal Tract. Infect Immun 2016; 84:2382-2388. [PMID: 27271744 DOI: 10.1128/iai.00432-16] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Accepted: 06/01/2016] [Indexed: 12/16/2022] Open
Abstract
Chlamydia has been detected in the gastrointestinal tracts of both animals and humans. However, it remains unclear whether the chlamydial organisms can be introduced into the gastrointestinal tract via pathways independent of the oral and anal routes. We have recently shown that Chlamydia muridarum spreads from the genital tract to the gastrointestinal tract potentially via the circulatory system. To test whether hematogenous C. muridarum can spread to and establish a long-lasting colonization in the mouse gastrointestinal tract, we inoculated mice intravenously with a luciferase-expressing C. muridarum strain and monitored its distribution. After tail vein inoculation, most luciferase-generated bioluminescence signals were detected in the mouse abdominal area throughout the experiment. The ex vivo imaging revealed that the abdominal signals came from the gastrointestinal tract tissues. Simultaneous monitoring of chlamydial organisms in individual organs or tissues revealed an initial stage of systemic spreading followed by a long-lasting infection in the gastrointestinal tract. A retro-orbital vein inoculation of the C. muridarum organisms at a lower dose in a different mouse strain also led to colonization of the gastrointestinal tract. We have demonstrated that intravenous C. muridarum inoculation can result in colonization of the gastrointestinal tract, suggesting that the chlamydial organisms may use the sexual behavior-independent circulation pathway to infect the gastrointestinal tract.
Collapse
|
23
|
Buchacher T, Ohradanova-Repic A, Stockinger H, Fischer MB, Weber V. M2 Polarization of Human Macrophages Favors Survival of the Intracellular Pathogen Chlamydia pneumoniae. PLoS One 2015; 10:e0143593. [PMID: 26606059 PMCID: PMC4659546 DOI: 10.1371/journal.pone.0143593] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Accepted: 11/06/2015] [Indexed: 12/12/2022] Open
Abstract
Intracellular pathogens have developed various strategies to escape immunity to enable their survival in host cells, and many bacterial pathogens preferentially reside inside macrophages, using diverse mechanisms to penetrate their defenses and to exploit their high degree of metabolic diversity and plasticity. Here, we characterized the interactions of the intracellular pathogen Chlamydia pneumoniae with polarized human macrophages. Primary human monocytes were pre-differentiated with granulocyte macrophage colony-stimulating factor or macrophage colony-stimulating factor for 7 days to yield M1-like and M2-like macrophages, which were further treated with interferon-γ and lipopolysaccharide or with interleukin-4 for 48 h to obtain fully polarized M1 and M2 macrophages. M1 and M2 cells exhibited distinct morphology with round or spindle-shaped appearance for M1 and M2, respectively, distinct surface marker profiles, as well as different cytokine and chemokine secretion. Macrophage polarization did not influence uptake of C. pneumoniae, since comparable copy numbers of chlamydial DNA were detected in M1 and M2 at 6 h post infection, but an increase in chlamydial DNA over time indicating proliferation was only observed in M2. Accordingly, 72±5% of M2 vs. 48±7% of M1 stained positive for chlamydial lipopolysaccharide, with large perinuclear inclusions in M2 and less clearly bordered inclusions for M1. Viable C. pneumoniae was present in lysates from M2, but not from M1 macrophages. The ability of M1 to restrict chlamydial replication was not observed in M1-like macrophages, since chlamydial load showed an equal increase over time for M1-like and M2-like macrophages. Our findings support the importance of macrophage polarization for the control of intracellular infection, and show that M2 are the preferred survival niche for C. pneumoniae. M1 did not allow for chlamydial proliferation, but failed to completely eliminate chlamydial infection, giving further evidence for the ability of C. pneumoniae to evade cellular defense and to persist in human macrophages.
Collapse
Affiliation(s)
- Tanja Buchacher
- Christian Doppler Laboratory for Innovative Therapy Approaches in Sepsis, Danube University Krems, Krems, Austria
| | - Anna Ohradanova-Repic
- Molecular Immunology Unit, Institute for Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Hannes Stockinger
- Molecular Immunology Unit, Institute for Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Michael B. Fischer
- Department of Blood Group Serology and Transfusion Medicine, Medical University of Vienna, Vienna, Austria
- Department for Health Sciences and Biomedicine, Danube University Krems, Krems, Austria
| | - Viktoria Weber
- Christian Doppler Laboratory for Innovative Therapy Approaches in Sepsis, Danube University Krems, Krems, Austria
- Department for Health Sciences and Biomedicine, Danube University Krems, Krems, Austria
- * E-mail:
| |
Collapse
|
24
|
Enhanced Direct Major Histocompatibility Complex Class I Self-Antigen Presentation Induced by Chlamydia Infection. Infect Immun 2015; 84:480-90. [PMID: 26597986 DOI: 10.1128/iai.01254-15] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Accepted: 11/17/2015] [Indexed: 11/20/2022] Open
Abstract
The direct major histocompatibility complex (MHC) class I antigen presentation pathway ensures intracellular peptides are displayed at the cellular surface for recognition of infected or transformed cells by CD8(+) cytotoxic T lymphocytes. Chlamydia spp. are obligate intracellular bacteria and, as such, should be targeted by CD8(+) T cells. It is likely that Chlamydia spp. have evolved mechanisms to avoid the CD8(+) killer T cell responses by interfering with MHC class I antigen presentation. Using a model system of self-peptide presentation which allows for posttranslational control of the model protein's stability, we tested the ability of various Chlamydia species to alter direct MHC class I antigen presentation. Infection of the JY lymphoblastoid cell line limited the accumulation of a model host protein and increased presentation of the model-protein-derived peptides. Enhanced self-peptide presentation was detected only when presentation was restricted to defective ribosomal products, or DRiPs, and total MHC class I levels remained unaltered. Skewed antigen presentation was dependent on a bacterial synthesized component, as evidenced by reversal of the observed phenotype upon preventing bacterial transcription, translation, and the inhibition of bacterial lipooligosaccharide synthesis. These data suggest that Chlamydia spp. have evolved to alter the host antigen presentation machinery to favor presentation of defective and rapidly degraded forms of self-antigen, possibly as a mechanism to diminish the presentation of peptides derived from bacterial proteins.
Collapse
|
25
|
Buchacher T, Wiesinger-Mayr H, Vierlinger K, Rüger BM, Stanek G, Fischer MB, Weber V. Human blood monocytes support persistence, but not replication of the intracellular pathogen C. pneumoniae. BMC Immunol 2014; 15:60. [PMID: 25488836 PMCID: PMC4268907 DOI: 10.1186/s12865-014-0060-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Accepted: 12/01/2014] [Indexed: 01/09/2023] Open
Abstract
Background Intracellular pathogens have devised various mechanisms to subvert the host immune response in order to survive and replicate in host cells. Here, we studied the infection of human blood monocytes with the intracellular pathogen C. pneumoniae and the effect on cytokine and chemokine profiles in comparison to stimulation with LPS. Results Monocytes purified from peripheral blood mononuclear cells by negative depletion were infected with C. pneumoniae. While immunofluorescence confirmed the presence of chlamydial lipopolysaccharide (LPS) in the cytoplasm of infected monocytes, real-time PCR did not provide evidence for replication of the intracellular pathogen. Complementary to PCR, C. pneumoniae infection was confirmed by an oligonucleotide DNA microarray for the detection of intracellular pathogens. Raman microspectroscopy revealed different molecular fingerprints for infected and non-infected monocytes, which were mainly due to changes in lipid and fatty acid content. Stimulation of monocytes with C. pneumoniae or with LPS induced similar profiles of tumor necrosis factor-alpha (TNF-α) and interleukin (IL)-6, but higher levels of IL-1β, IL-12p40 and IL-12p70 for C. pneumoniae which were statistically significant. C. pneumoniae also induced release of the chemokines MCP-1, MIP-1α and MIP-1β, and CXCL-8, which correlated with TNF-α secretion. Conclusion Infection of human blood monocytes with intracellular pathogens triggers altered cytokine and chemokine pattern as compared to stimulation with extracellular ligands such as LPS. Complementing conventional methods, an oligonucleotide DNA microarray for the detection of intracellular pathogens as well as Raman microspectroscopy provide useful tools to trace monocyte infection. Electronic supplementary material The online version of this article (doi:10.1186/s12865-014-0060-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Tanja Buchacher
- Christian Doppler Laboratory for Innovative Therapy Approaches in Sepsis, Danube University Krems, Krems, Austria.
| | | | | | - Beate M Rüger
- Department of Blood Group Serology and Transfusion Medicine, Medical University of Vienna, Vienna, Austria.
| | - Gerold Stanek
- Institute for Hygiene and Applied Immunology, Medical University of Vienna, Vienna, Austria.
| | - Michael B Fischer
- Department of Blood Group Serology and Transfusion Medicine, Medical University of Vienna, Vienna, Austria. .,Department for Health Sciences and Biomedicine, Danube University Krems, Krems, Austria.
| | - Viktoria Weber
- Christian Doppler Laboratory for Innovative Therapy Approaches in Sepsis, Danube University Krems, Krems, Austria. .,Department for Health Sciences and Biomedicine, Danube University Krems, Krems, Austria.
| |
Collapse
|