1
|
Broder KC, Matrosova VY, Tkavc R, Gaidamakova EK, Ho LTVT, Macintyre AN, Soc A, Diallo A, Darnell SC, Bash S, Daly MJ, Jerse AE, Liechti GW. Irradiated whole cell Chlamydia vaccine confers significant protection in a murine genital tract challenge model. NPJ Vaccines 2024; 9:207. [PMID: 39528548 PMCID: PMC11554809 DOI: 10.1038/s41541-024-00968-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 09/11/2024] [Indexed: 11/16/2024] Open
Abstract
Chlamydia trachomatis infections are the most common bacterial STIs globally and can lead to serious morbidity if untreated. Development of a killed, whole-cell vaccine has been stymied by coincident epitope destruction during inactivation. Here, we present a prototype Chlamydia vaccine composed of elementary bodies (EBs) from the related mouse pathogen, Chlamydia muridarum (Cm). EBs inactivated by gamma rays (Ir-Cm) in the presence of the antioxidant Mn2+-Decapeptide (DEHGTAVMLK) Phosphate (MDP) are protected from epitope damage but not DNA damage. Cm EBs gamma-inactivated with MDP retain their structure and provide significant protection in a murine genital tract infection model. Mice vaccinated with Ir-Cm (+MDP) exhibited elevated levels of Cm-specific IgG and IgA antibodies, reduced bacterial burdens, accelerated clearance, and distinctive cytokine responses compared to unvaccinated controls and animals vaccinated with EBs irradiated without MDP. Preserving EB epitopes with MDP during gamma inactivation offers the potential for a polyvalent, whole-cell vaccine against C. trachomatis.
Collapse
Affiliation(s)
- Kieran C Broder
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
- Department of Microbiology, Immunology, and Pathology, College of Veterinary Medicine and Biological Sciences, Colorado State University, Ft. Collins, CO, USA
| | - Vera Y Matrosova
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
- Department of Pathology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Rok Tkavc
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Elena K Gaidamakova
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
- Department of Pathology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Lam Thuy Vi Tran Ho
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | | | - Anthony Soc
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Aissata Diallo
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Stephen C Darnell
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Sarah Bash
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Michael J Daly
- Department of Pathology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.
| | - Ann E Jerse
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.
| | - George W Liechti
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.
| |
Collapse
|
2
|
Sundaram B, Tweedell RE, Prasanth Kumar S, Kanneganti TD. The NLR family of innate immune and cell death sensors. Immunity 2024; 57:674-699. [PMID: 38599165 PMCID: PMC11112261 DOI: 10.1016/j.immuni.2024.03.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 03/07/2024] [Accepted: 03/12/2024] [Indexed: 04/12/2024]
Abstract
Nucleotide-binding oligomerization domain (NOD)-like receptors, also known as nucleotide-binding leucine-rich repeat receptors (NLRs), are a family of cytosolic pattern recognition receptors that detect a wide variety of pathogenic and sterile triggers. Activation of specific NLRs initiates pro- or anti-inflammatory signaling cascades and the formation of inflammasomes-multi-protein complexes that induce caspase-1 activation to drive inflammatory cytokine maturation and lytic cell death, pyroptosis. Certain NLRs and inflammasomes act as integral components of larger cell death complexes-PANoptosomes-driving another form of lytic cell death, PANoptosis. Here, we review the current understanding of the evolution, structure, and function of NLRs in health and disease. We discuss the concept of NLR networks and their roles in driving cell death and immunity. An improved mechanistic understanding of NLRs may provide therapeutic strategies applicable across infectious and inflammatory diseases and in cancer.
Collapse
Affiliation(s)
- Balamurugan Sundaram
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Rebecca E Tweedell
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | | | | |
Collapse
|
3
|
Cocomazzi G, Del Pup L, Contu V, Maggio G, Parmegiani L, Ciampaglia W, De Ruvo D, Faioli R, Maglione A, Baldini GM, Baldini D, Pazienza V. Gynecological Cancers and Microbiota Dynamics: Insights into Pathogenesis and Therapy. Int J Mol Sci 2024; 25:2237. [PMID: 38396914 PMCID: PMC10889201 DOI: 10.3390/ijms25042237] [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: 01/05/2024] [Revised: 02/07/2024] [Accepted: 02/10/2024] [Indexed: 02/25/2024] Open
Abstract
In recent years, the relationship between the microbiota and various aspects of health has become a focal point of scientific investigation. Although the most studied microbiota concern the gastrointestinal tract, recently, the interest has also been extended to other body districts. Female genital tract dysbiosis and its possible impact on pathologies such as endometriosis, polycystic ovary syndrome (PCOS), pelvic inflammatory disease (PID), and gynecological cancers have been unveiled. The incursion of pathogenic microbes alters the ecological equilibrium of the vagina, triggering inflammation and compromising immune defense, potentially fostering an environment conducive to cancer development. The most common types of gynecological cancer include cervical, endometrial, and ovarian cancer, which occur in women of any age but especially in postmenopausal women. Several studies highlighted that a low presence of lactobacilli at the vaginal level, and consequently, in related areas (such as the endometrium and ovary), correlates with a higher risk of gynecological pathology and likely contributes to increased incidence and worse prognosis of gynecological cancers. The complex interplay between microbial communities and the development, progression, and treatment of gynecologic malignancies is a burgeoning field not yet fully understood. The intricate crosstalk between the gut microbiota and systemic inflammation introduces a new dimension to our understanding of gynecologic cancers. The objective of this review is to focus attention on the association between vaginal microbiota and gynecological malignancies and provide detailed knowledge for future diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Giovanna Cocomazzi
- Division of Gastroenterology, Fondazione IRCCS-Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, FG, Italy;
| | - Lino Del Pup
- Gynecological Endocrinology and Fertility, University Sanitary Agency Friuli Central (ASUFC), Via Pozzuolo, 330, 33100 Udine, FVG, Italy;
| | - Viviana Contu
- Integrative Medicine Unit, Humanitas Gradenigo, Corso Regina Margherita 8/10, 10153 Torino, FC, Italy;
| | - Gabriele Maggio
- Pia Fondazione Cardinale Giovanni Panico, Via S. Pio X, 4, 73039 Tricase, LE, Italy;
| | - Lodovico Parmegiani
- Next Fertility GynePro, NextClinics International Via T. Cremona 8, 40137 Bologna, RE, Italy; (L.P.); (W.C.)
| | - Walter Ciampaglia
- Next Fertility GynePro, NextClinics International Via T. Cremona 8, 40137 Bologna, RE, Italy; (L.P.); (W.C.)
| | - Daniele De Ruvo
- Gynaecology, Obstetrics and Reproductive Medicine Affidea Promea, Via Menabrea 14, 10126 Torino, TO, Italy;
| | - Raffaele Faioli
- Gynecology and Obstetrics, IRCCS “Casa Sollievo della Sofferenza”, 71013 San Giovanni Rotondo, FG, Italy; (R.F.); (A.M.)
| | - Annamaria Maglione
- Gynecology and Obstetrics, IRCCS “Casa Sollievo della Sofferenza”, 71013 San Giovanni Rotondo, FG, Italy; (R.F.); (A.M.)
| | - Giorgio Maria Baldini
- IVF Center, Momò Fertilife, 76011 Bisceglie Via Cala dell’Arciprete, 76011 Bisceglie, BT, Italy; (G.M.B.); (D.B.)
| | - Domenico Baldini
- IVF Center, Momò Fertilife, 76011 Bisceglie Via Cala dell’Arciprete, 76011 Bisceglie, BT, Italy; (G.M.B.); (D.B.)
| | - Valerio Pazienza
- Division of Gastroenterology, Fondazione IRCCS-Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, FG, Italy;
| |
Collapse
|
4
|
Wang X, Wu H, Fang C, Li Z. Insights into innate immune cell evasion by Chlamydia trachomatis. Front Immunol 2024; 15:1289644. [PMID: 38333214 PMCID: PMC10850350 DOI: 10.3389/fimmu.2024.1289644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 01/11/2024] [Indexed: 02/10/2024] Open
Abstract
Chlamydia trachomatis, is a kind of obligate intracellular pathogen. The removal of C. trachomatis relies primarily on specific cellular immunity. It is currently considered that CD4+ Th1 cytokine responses are the major protective immunity against C. trachomatis infection and reinfection rather than CD8+ T cells. The non-specific immunity (innate immunity) also plays an important role in the infection process. To survive inside the cells, the first process that C. trachomatis faces is the innate immune response. As the "sentry" of the body, mast cells attempt to engulf and remove C. trachomatis. Dendritic cells present antigen of C. trachomatis to the "commanders" (T cells) through MHC-I and MHC-II. IFN-γ produced by activated T cells and natural killer cells (NK) further activates macrophages. They form the body's "combat troops" and produce immunity against C. trachomatis in the tissues and blood. In addition, the role of eosinophils, basophils, innate lymphoid cells (ILCs), natural killer T (NKT) cells, γδT cells and B-1 cells should not be underestimated in the infection of C. trachomatis. The protective role of innate immunity is insufficient, and sexually transmitted diseases (STDs) caused by C. trachomatis infections tend to be insidious and recalcitrant. As a consequence, C. trachomatis has developed a unique evasion mechanism that triggers inflammatory immunopathology and acts as a bridge to protective to pathological adaptive immunity. This review focuses on the recent advances in how C. trachomatis evades various innate immune cells, which contributes to vaccine development and our understanding of the pathophysiologic consequences of C. trachomatis infection.
Collapse
Affiliation(s)
| | | | | | - Zhongyu Li
- Institute of Pathogenic Biology, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, School of Nursing, Hengyang Medical College, University of South China, Hengyang, China
| |
Collapse
|
5
|
Armitage CW, O'Meara CP, Bryan ER, Kollipara A, Trim LK, Hickey D, Carey AJ, Huston WM, Donnelly G, Yazdani A, Blumberg RS, Beagley KW. IgG exacerbates genital chlamydial pathology in females by enhancing pathogenic CD8 + T cell responses. Scand J Immunol 2024; 99:e13331. [PMID: 38441219 PMCID: PMC10909563 DOI: 10.1111/sji.13331] [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: 05/30/2023] [Revised: 08/25/2023] [Accepted: 09/11/2023] [Indexed: 03/07/2024]
Abstract
Chlamydia trachomatis infections are an important sexually transmitted infection that can lead to inflammation, scarring and hydrosalpinx/infertility. However, infections are commonly clinically asymptomatic and do not receive treatment. The underlying cause of asymptomatic immunopathology remains unknown. Here, we demonstrate that IgG produced during male infection enhanced the incidence of immunopathology and infertility in females. Human endocervical cells expressing the neonatal Fc Receptor (FcRn) increased translocation of human IgG-opsonized C. trachomatis. Using total IgG purified from infected male mice, we opsonized C. muridarum and then infected female mice, mimicking sexual transmission. Following infection, IgG-opsonized Chlamydia was found to transcytose the epithelial barrier in the uterus, where it was phagocytosed by antigen-presenting cells (APCs) and trafficked to the draining lymph nodes. APCs then expanded both CD4+ and CD8+ T cell populations and caused significantly more infertility in female mice infected with non-opsonized Chlamydia. Enhanced phagocytosis of IgG-opsonized Chlamydia significantly increased pro-inflammatory signalling and T cell proliferation. As IgG is transcytosed by FcRn, we utilized FcRn-/- mice and observed that shedding kinetics of Chlamydia were only affected in FcRn-/- mice infected with IgG-opsonized Chlamydia. Depletion of CD8+ T cells in FcRn-/- mice lead to a significant reduction in the incidence of infertility. Taken together, these data demonstrate that IgG seroconversion during male infection can amplify female immunopathology, dependent on FcRn transcytosis, APC differentiation and enhanced CD8 T cell responses.
Collapse
Affiliation(s)
- Charles W. Armitage
- Centre for Immunology and Infection Control and School of Biomedical SciencesQueensland University of Technology (QUT)BrisbaneQueenslandAustralia
| | - Connor P. O'Meara
- Centre for Immunology and Infection Control and School of Biomedical SciencesQueensland University of Technology (QUT)BrisbaneQueenslandAustralia
- Drop Bio Ltd, School of Biotechnology and Biomolecular Sciences (BABS)University of New South WalesSydneyNew South WalesAustralia
| | - Emily R. Bryan
- Centre for Immunology and Infection Control and School of Biomedical SciencesQueensland University of Technology (QUT)BrisbaneQueenslandAustralia
| | - Avinash Kollipara
- Centre for Immunology and Infection Control and School of Biomedical SciencesQueensland University of Technology (QUT)BrisbaneQueenslandAustralia
| | - Logan K. Trim
- Centre for Immunology and Infection Control and School of Biomedical SciencesQueensland University of Technology (QUT)BrisbaneQueenslandAustralia
| | - Danica Hickey
- Centre for Immunology and Infection Control and School of Biomedical SciencesQueensland University of Technology (QUT)BrisbaneQueenslandAustralia
| | - Alison J. Carey
- Centre for Immunology and Infection Control and School of Biomedical SciencesQueensland University of Technology (QUT)BrisbaneQueenslandAustralia
| | - Wilhelmina M. Huston
- School of Life SciencesUniversity of Technology (UTS) SydneyUltimoNew South WalesAustralia
| | - Gavin Donnelly
- Queensland Fertility Group (QFG)BrisbaneQueenslandAustralia
| | - Anusch Yazdani
- Queensland Fertility Group (QFG)BrisbaneQueenslandAustralia
| | - Richard S. Blumberg
- Division of Gastroenterology, Department of MedicineBrigham & Women's Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Kenneth W. Beagley
- Centre for Immunology and Infection Control and School of Biomedical SciencesQueensland University of Technology (QUT)BrisbaneQueenslandAustralia
| |
Collapse
|
6
|
Capitani N, Baldari CT. The Immunological Synapse: An Emerging Target for Immune Evasion by Bacterial Pathogens. Front Immunol 2022; 13:943344. [PMID: 35911720 PMCID: PMC9325968 DOI: 10.3389/fimmu.2022.943344] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 06/20/2022] [Indexed: 11/13/2022] Open
Abstract
Similar to other pathogens, bacteria have developed during their evolution a variety of mechanisms to overcome both innate and acquired immunity, accounting for their ability to cause disease or chronic infections. The mechanisms exploited for this critical function act by targeting conserved structures or pathways that regulate the host immune response. A strategic potential target is the immunological synapse (IS), a highly specialized structure that forms at the interface between antigen presenting cells (APC) and T lymphocytes and is required for the establishment of an effective T cell response to the infectious agent and for the development of long-lasting T cell memory. While a variety of bacterial pathogens are known to impair or subvert cellular processes essential for antigen processing and presentation, on which IS assembly depends, it is only recently that the possibility that IS may be a direct target of bacterial virulence factors has been considered. Emerging evidence strongly supports this notion, highlighting IS targeting as a powerful, novel means of immune evasion by bacterial pathogens. In this review we will present a brief overview of the mechanisms used by bacteria to affect IS assembly by targeting APCs. We will then summarize what has emerged from the current handful of studies that have addressed the direct impact of bacterial virulence factors on IS assembly in T cells and, based on the strategic cellular processes targeted by these factors in other cell types, highlight potential IS-related vulnerabilities that could be exploited by these pathogens to evade T cell mediated immunity.
Collapse
Affiliation(s)
- Nagaja Capitani
- Department of Life Sciences, University of Siena, Siena, Italy
| | | |
Collapse
|
7
|
Nie MZ, Zhang RQ, Zhao MC, Tan H, Hu YX, Fan GH, Li JY, He AN, Tian FY, Li FY, Zheng YH, Shen XX, Tie YQ, Ma XJ. Development of a duplex recombinase-aided amplification assay for direct detection of Mycoplasma pneumoniae and Chlamydia trachomatis in clinical samples. METHODS IN MICROBIOLOGY 2022; 198:106504. [PMID: 35654228 DOI: 10.1016/j.mimet.2022.106504] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 05/25/2022] [Accepted: 05/25/2022] [Indexed: 12/15/2022]
Abstract
BACKGROUND Pneumonia caused by Mycoplasma pneumoniae is common in the elderly and children, and pneumonia caused by Chlamydia trachomatis is prevalent in newborns. This study aimed to establish a rapid, sensitive, and simple method for the direct detection of M. pneumoniae and C. trachomatis in clinical samples without DNA extraction. METHODS We established a duplex recombinase-aided amplification (RAA) assay with the RNAseP gene as an internal control for detecting the P1 gene of M. pneumoniae and the ORF8 gene of C. trachomatis, respectively. The results were obtained at 39 °C within 15-20 min. A total of 130 clinical samples suspected of M. pneumoniae or C. trachomatis infection were collected and tested by duplex RAA and PCR. DNA extracted via a commercial kit or treated with a nucleic acid-releasing agent was used and compared, respectively. Standard recombinant plasmids were used to test the sensitivity of the duplex RAA assay. In addition, other similar common pathogens were used to verify the specificity of the duplex RAA assay. RESULTS The sensitivity of the duplex RAA assay for detecting M. pneumoniae and C. trachomatis was 10 copies/μL using recombinant plasmids. Compared with PCR, the sensitivity and specificity of duplex RAA assays for M. pneumoniae and C. trachomatis was 100% using clinical DNA samples extracted using a commercial kit and a nucleic acid-releasing agent, and the Kappa value was 1. CONCLUSION The advantages of this duplex RAA assay include high sensitivity and specificity, short duration, and simple extraction steps, with potential for use in the on-site detection of M. pneumoniae and C. trachomatis in resource-limited settings.
Collapse
Affiliation(s)
- Ming-Zhu Nie
- Hebei Medical University, Shijiazhuang 050031, Hebei, China; NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, No. 155, Changbai Street, Changping District, Beijing 102206, China.
| | - Rui-Qing Zhang
- NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, No. 155, Changbai Street, Changping District, Beijing 102206, China.
| | - Meng-Chuan Zhao
- Children's Hospital of Hebei Province, Shijiazhuang 050031, Hebei, China
| | - He Tan
- Hebei General Hospital, Shijiazhuang 050051, Hebei, China
| | - Ya-Xin Hu
- NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, No. 155, Changbai Street, Changping District, Beijing 102206, China; North China University of Science and Technology, Tangshan 063210, Hebei, China
| | - Guo-Hao Fan
- NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, No. 155, Changbai Street, Changping District, Beijing 102206, China
| | - Jing-Yi Li
- Hebei Medical University, Shijiazhuang 050031, Hebei, China; NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, No. 155, Changbai Street, Changping District, Beijing 102206, China
| | - An-Na He
- NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, No. 155, Changbai Street, Changping District, Beijing 102206, China; North China University of Science and Technology, Tangshan 063210, Hebei, China
| | - Feng-Yu Tian
- Hebei Medical University, Shijiazhuang 050031, Hebei, China; NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, No. 155, Changbai Street, Changping District, Beijing 102206, China
| | - Feng-Yu Li
- Hebei Medical University, Shijiazhuang 050031, Hebei, China; NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, No. 155, Changbai Street, Changping District, Beijing 102206, China
| | - Ye-Huan Zheng
- Autobio Diagnostics CO., Ltd, Zhengzhou 450000, Henan, China
| | - Xin-Xin Shen
- NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, No. 155, Changbai Street, Changping District, Beijing 102206, China.
| | - Yan-Qing Tie
- Hebei General Hospital, Shijiazhuang 050051, Hebei, China.
| | - Xue-Jun Ma
- NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, No. 155, Changbai Street, Changping District, Beijing 102206, China.
| |
Collapse
|
8
|
Sontyana B, Shrivastava R, Battu S, Ghosh S, Mukhopadhyay S. Phagosome maturation and modulation of macrophage effector function by intracellular pathogens: target for therapeutics. Future Microbiol 2021; 17:59-76. [PMID: 34877879 DOI: 10.2217/fmb-2021-0101] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Macrophages are important cells that regulate various innate functions. Macrophages after engulfment of pathogens proceed for phagosome maturation and finally fuse with lysosomes to kill pathogens. Although pathogen degradation is one of the important functions of phagosomes, various immune-effector functions of macrophages are also dependent on the phagosome maturation process. This review discusses signaling processes regulating phagosome maturation as well as various effector functions of macrophages such as apoptosis, antigen presentation, autophagy and inflammasome that are dependent on the phagosome maturation process. It also discusses strategies adopted by various intracellular pathogens to counteract these functions to evade intracellular destruction mechanisms. These studies may give direction for the development of new therapeutics to control various intracellular infections.
Collapse
Affiliation(s)
- Brahmaji Sontyana
- Laboratory of Molecular Cell Biology, Centre for DNA Fingerprinting & Diagnostics (CDFD), Inner Ring Road, Uppal, Hyderabad, 500039, Telangana, India.,Graduate Studies, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Rohini Shrivastava
- Laboratory of Molecular Cell Biology, Centre for DNA Fingerprinting & Diagnostics (CDFD), Inner Ring Road, Uppal, Hyderabad, 500039, Telangana, India.,Graduate Studies, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Srikanth Battu
- Laboratory of Molecular Cell Biology, Centre for DNA Fingerprinting & Diagnostics (CDFD), Inner Ring Road, Uppal, Hyderabad, 500039, Telangana, India
| | - Sudip Ghosh
- Molecular Biology Unit, ICMR-National Institute of Nutrition, Jamai Osmania PO, Hyderabad, 500007, Telangana, India
| | - Sangita Mukhopadhyay
- Laboratory of Molecular Cell Biology, Centre for DNA Fingerprinting & Diagnostics (CDFD), Inner Ring Road, Uppal, Hyderabad, 500039, Telangana, India
| |
Collapse
|
9
|
Kiritsy MC, Ankley LM, Trombley J, Huizinga GP, Lord AE, Orning P, Elling R, Fitzgerald KA, Olive AJ. A genetic screen in macrophages identifies new regulators of IFNγ-inducible MHCII that contribute to T cell activation. eLife 2021; 10:65110. [PMID: 34747695 PMCID: PMC8598162 DOI: 10.7554/elife.65110] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 11/03/2021] [Indexed: 12/26/2022] Open
Abstract
Cytokine-mediated activation of host immunity is central to the control of pathogens. Interferon-gamma (IFNγ) is a key cytokine in protective immunity that induces major histocompatibility complex class II molecules (MHCII) to amplify CD4+ T cell activation and effector function. Despite its central role, the dynamic regulation of IFNγ-induced MHCII is not well understood. Using a genome-wide CRISPR-Cas9 screen in murine macrophages, we identified genes that control MHCII surface expression. Mechanistic studies uncovered two parallel pathways of IFNγ-mediated MHCII control that require the multifunctional glycogen synthase kinase three beta (GSK3β) or the mediator complex subunit 16 (MED16). Both pathways control distinct aspects of the IFNγ response and are necessary for IFNγ-mediated induction of the MHCII transactivator Ciita, MHCII expression, and CD4+ T cell activation. Our results define previously unappreciated regulation of MHCII expression that is required to control CD4+ T cell responses.
Collapse
Affiliation(s)
- Michael C Kiritsy
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, United States
| | - Laurisa M Ankley
- Department of Microbiology & Molecular Genetics, College of Osteopathic Medicine, Michigan State University, East Lansing, United States
| | - Justin Trombley
- Department of Microbiology & Molecular Genetics, College of Osteopathic Medicine, Michigan State University, East Lansing, United States
| | - Gabrielle P Huizinga
- Department of Microbiology & Molecular Genetics, College of Osteopathic Medicine, Michigan State University, East Lansing, United States
| | - Audrey E Lord
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, United States
| | - Pontus Orning
- Division of Infectious Disease and Immunology, Department of Medicine, University of Massachusetts Medical School, Worcester, United States
| | - Roland Elling
- Division of Infectious Disease and Immunology, Department of Medicine, University of Massachusetts Medical School, Worcester, United States
| | - Katherine A Fitzgerald
- Division of Infectious Disease and Immunology, Department of Medicine, University of Massachusetts Medical School, Worcester, United States
| | - Andrew J Olive
- Department of Microbiology & Molecular Genetics, College of Osteopathic Medicine, Michigan State University, East Lansing, United States
| |
Collapse
|
10
|
Chen H, Peng B, Yang C, Xie L, Zhong S, Sun Z, Li Z, Wang C, Liu X, Tang X, Zhong G, Lu C. The role of an enzymatically inactive CPAF mutant vaccination in Chlamydia muridarum genital tract infection. Microb Pathog 2021; 160:105137. [PMID: 34390765 DOI: 10.1016/j.micpath.2021.105137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 08/06/2021] [Accepted: 08/09/2021] [Indexed: 10/20/2022]
Abstract
Chlamydia trachomatis urogenital tract infection causes pelvic inflammatory disease and infertility, increases the risk of co-infection with HPV and HIV. Chlamydial vaccination is considered the most promising approach to prevent and control its infection. Among various chlamydial vaccine candidates, chlamydial protease-like activity factor (CPAF) have been reported to provide robust protective immunity against genital chlamydial infection in mice with reduced vaginal shedding and oviduct pathology. However, CPAF is a serine protease which has enzymatical activity to degrade a large number of substrates. In order to increase the safety of CPAF vaccine, in this study, we used a mutant CPAF that is deficient in enzymatical activity to determine whether proteolytic activity of CPAF affect its vaccine efficacy. The wild type or mutant CPAF immunization causes a significant lower chlamydial shedding from the vaginal and resolve the infection as early as day 20, compared to day 28 in adjuvant control mice. More important, reduced upper reproductive tract pathology were also observed in these two groups. The mutant or wild type CPAF immunization induced not only robust splenic IFN-γ and serum IgG2a but also sIgA secretion in the vaginal fluids. Furthermore, neutralization of chlamydia with immune sera did not provide protection against oviduct pathology. However, adoptive transfer of CD4+ splenocytes isolated from the mutant or wild type CPAF immunized mice resulted in a significant and comparable reduced oviduct pathology. Our results indicate mutant CPAF vaccination is as same efficacy as wild type, and the protection relies on CD4+ T cells, which will further promote the development of CPAF as clinical chlamydial vaccine.
Collapse
Affiliation(s)
- Hui Chen
- Institute of Pathogenic Biology and Key Laboratory of Special Pathogen Prevention and Control of Hunan Province, Hengyang Medical College, University of South China, Hengyang, Hunan, China
| | - Bo Peng
- Cancer Research Institute, University of South China, Hengyang, Hunan, China
| | - Chunfen Yang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of University of South China, Hengyang, Hunan, China
| | - Lijuan Xie
- Institute of Pathogenic Biology and Key Laboratory of Special Pathogen Prevention and Control of Hunan Province, Hengyang Medical College, University of South China, Hengyang, Hunan, China
| | - Shufang Zhong
- Institute of Pathogenic Biology and Key Laboratory of Special Pathogen Prevention and Control of Hunan Province, Hengyang Medical College, University of South China, Hengyang, Hunan, China
| | - Zhenjie Sun
- Institute of Pathogenic Biology and Key Laboratory of Special Pathogen Prevention and Control of Hunan Province, Hengyang Medical College, University of South China, Hengyang, Hunan, China
| | - Zhongyu Li
- Institute of Pathogenic Biology and Key Laboratory of Special Pathogen Prevention and Control of Hunan Province, Hengyang Medical College, University of South China, Hengyang, Hunan, China
| | - Chuan Wang
- Institute of Pathogenic Biology and Key Laboratory of Special Pathogen Prevention and Control of Hunan Province, Hengyang Medical College, University of South China, Hengyang, Hunan, China
| | - Xiao Liu
- Institute of Pathogenic Biology and Key Laboratory of Special Pathogen Prevention and Control of Hunan Province, Hengyang Medical College, University of South China, Hengyang, Hunan, China
| | - Xin Tang
- Cancer Research Institute, University of South China, Hengyang, Hunan, China
| | - Guangming Zhong
- Department of Microbiology, Immunology, and Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Chunxue Lu
- Institute of Pathogenic Biology and Key Laboratory of Special Pathogen Prevention and Control of Hunan Province, Hengyang Medical College, University of South China, Hengyang, Hunan, China.
| |
Collapse
|
11
|
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: 2.3] [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
|
12
|
Heuberger C, Pott J, Maloy KJ. Why do intestinal epithelial cells express MHC class II? Immunology 2021; 162:357-367. [PMID: 32966619 PMCID: PMC7968399 DOI: 10.1111/imm.13270] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 08/19/2020] [Accepted: 08/20/2020] [Indexed: 12/23/2022] Open
Abstract
Intestinal epithelial cells (IECs) constitute the border between the vast antigen load present in the intestinal lumen and the mucosal immune compartment. Their ability to express antigen processing and presentation machinery evokes the question whether IECs function as non-conventional antigen-presenting cells. Major histocompatibility complex (MHC) class II expression by non-haematopoietic cells, such as IECs, is tightly regulated by the class II transactivator (CIITA) and is classically induced by IFN-γ. As MHC class II expression by IECs is upregulated under inflammatory conditions, it has been proposed to activate effector CD4+ T (Teff) cells. However, other studies have reported contradictory results and instead suggested a suppressive role of antigen presentation by IECs, through regulatory T (Treg)-cell activation. Recent studies investigating the role of MHC class II + exosomes released by IECs also reported conflicting findings of either immune enhancing or immunosuppressive activities. Moreover, in addition to modulating inflammatory responses, recent findings suggest that MHC class II expression by intestinal stem cells may elicit crosstalk that promotes epithelial renewal. A more complete understanding of the different consequences of IEC MHC class II antigen presentation will guide future efforts to modulate this pathway to selectively invoke protective immunity while maintaining tolerance to beneficial antigens.
Collapse
Affiliation(s)
- Cornelia Heuberger
- Sir William Dunn School of PathologyUniversity of OxfordOxfordUK
- Kennedy Institute of RheumatologyUniversity of OxfordOxfordUK
| | - Johanna Pott
- Sir William Dunn School of PathologyUniversity of OxfordOxfordUK
- Hubrecht Organoid TechnologyUtrechtNetherlands
| | - Kevin Joseph Maloy
- Sir William Dunn School of PathologyUniversity of OxfordOxfordUK
- Institute of Infection, Immunity and InflammationUniversity of GlasgowGlasgowUK
| |
Collapse
|
13
|
León Machado JA, Steimle V. The MHC Class II Transactivator CIITA: Not (Quite) the Odd-One-Out Anymore among NLR Proteins. Int J Mol Sci 2021; 22:1074. [PMID: 33499042 PMCID: PMC7866136 DOI: 10.3390/ijms22031074] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 01/15/2021] [Accepted: 01/19/2021] [Indexed: 12/14/2022] Open
Abstract
In this review, we discuss the major histocompatibility complex (MHC) class II transactivator (CIITA), which is the master regulator of MHC class II gene expression. CIITA is the founding member of the mammalian nucleotide-binding and leucine-rich-repeat (NLR) protein family but stood apart for a long time as the only transcriptional regulator. More recently, it was found that its closest homolog, NLRC5 (NLR protein caspase activation and recruitment domain (CARD)-containing 5), is a regulator of MHC-I gene expression. Both act as non-DNA-binding activators through multiple protein-protein interactions with an MHC enhanceosome complex that binds cooperatively to a highly conserved combinatorial cis-acting module. Thus, the regulation of MHC-II expression is regulated largely through the differential expression of CIITA. In addition to the well-defined role of CIITA in MHC-II GENE regulation, we will discuss several other aspects of CIITA functions, such as its role in cancer, its role as a viral restriction element contributing to intrinsic immunity, and lastly, its very recently discovered role as an inhibitor of Ebola and SARS-Cov-2 virus replication. We will briefly touch upon the recently discovered role of NLRP3 as a transcriptional regulator, which suggests that transcriptional regulation is, after all, not such an unusual feature for NLR proteins.
Collapse
Affiliation(s)
| | - Viktor Steimle
- Département de Biologie, Université de Sherbrooke, 2500 Boul., Sherbrooke, QC J1K 2R1, Canada;
| |
Collapse
|
14
|
A novel protein upstream stimulatory factor 2 identified in lamprey, Lethenteron reissneri. Dev Genes Evol 2020; 230:347-357. [PMID: 32852621 DOI: 10.1007/s00427-020-00666-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 07/29/2020] [Indexed: 10/23/2022]
Abstract
Upstream stimulatory factors are kinds of multi-functional transcription factors, which are expressed in eukaryotes widely, including Upstream stimulatory factor 1 (USFl) and upstream stimulatory factor 2 (USF2). USF protein has a typical basic helix-loop-helix leucine zipper (b-HLH-LZ) structure, which is involved in cell cycle, cell proliferations, glucose and lipid metabolism, and other biochemical processes. Although the USF family is an important regulator of cellular processes, little is known about the USF genes of lampreys, especially their evolutionary relationships, expression profiles, and biological functions. Here, an upstream stimulatory factor 2 (USF2) homolog from lamprey (Lethenteron reissneri) was identified and characterized (designated as L-USF2) because it is closer to USF2 subfamily than to USF1 subfamily. The cDNA fragment of L-USF2 has an open reading frame (ORF) of 765-bp length, encodes 254 amino acids, and contains an HLH domain at the c-terminal of amino acids. Meanwhile, motifs and genetic structure analysis reveal that USF2 gene exons are conserved. Moreover, the 3D structure analysis indicates that L-USF2 adopts the general USF2 folding and has a high structural similarity with H-USF2. The synteny results showed that the L-USF2 adjacent gene changed greatly compared with the jaw vertebrates. By real-time quantitative experiment and Western blot analysis, we found that L-USF2 gene played a significant role in the immune responses. This study not only provides us with a further understanding of the evolution and function of the USF gene family but also provides a basis for exploring its immune responses and immune defenses in lampreys.
Collapse
|
15
|
McQueen BE, Kiatthanapaiboon A, Fulcher ML, Lam M, Patton K, Powell E, Kollipara A, Madden V, Suchland RJ, Wyrick P, O'Connell CM, Reidel B, Kesimer M, Randell SH, Darville T, Nagarajan UM. Human Fallopian Tube Epithelial Cell Culture Model To Study Host Responses to Chlamydia trachomatis Infection. Infect Immun 2020; 88:e00105-20. [PMID: 32601108 PMCID: PMC7440757 DOI: 10.1128/iai.00105-20] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 06/23/2020] [Indexed: 12/20/2022] Open
Abstract
Chlamydia trachomatis infection of the human fallopian tubes can lead to damaging inflammation and scarring, ultimately resulting in infertility. To study the human cellular responses to chlamydial infection, researchers have frequently used transformed cell lines that can have limited translational relevance. We developed a primary human fallopian tube epithelial cell model based on a method previously established for culture of primary human bronchial epithelial cells. After protease digestion and physical dissociation of excised fallopian tubes, epithelial cell precursors were expanded in growth factor-containing medium. Expanded cells were cryopreserved to generate a biobank of cells from multiple donors and cultured at an air-liquid interface. Culture conditions stimulated cellular differentiation into polarized mucin-secreting and multiciliated cells, recapitulating the architecture of human fallopian tube epithelium. The polarized and differentiated cells were infected with a clinical isolate of C. trachomatis, and inclusions containing chlamydial developmental forms were visualized by fluorescence and electron microscopy. Apical secretions from infected cells contained increased amounts of proteins associated with chlamydial growth and replication, including transferrin receptor protein 1, the amino acid transporters SLC3A2 and SLC1A5, and the T-cell chemoattractants CXCL10, CXCL11, and RANTES. Flow cytometry revealed that chlamydial infection induced cell surface expression of T-cell homing and activation proteins, including ICAM-1, VCAM-1, HLA class I and II, and interferon gamma receptor. This human fallopian tube epithelial cell culture model is an important tool with translational potential for studying cellular responses to Chlamydia and other sexually transmitted pathogens.
Collapse
Affiliation(s)
- Bryan E McQueen
- Department of Pediatrics, University of North Carolina, Chapel Hill, North Carolina, USA
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Amy Kiatthanapaiboon
- Department of Pediatrics, University of North Carolina, Chapel Hill, North Carolina, USA
| | - M Leslie Fulcher
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Mariam Lam
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Kate Patton
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Emily Powell
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Avinash Kollipara
- Department of Pediatrics, University of North Carolina, Chapel Hill, North Carolina, USA
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Victoria Madden
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Robert J Suchland
- University of Washington, Division of Allergy and Infectious Diseases, Department of Medicine, Seattle, Washington, USA
| | - Priscilla Wyrick
- Department of Pediatrics, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Catherine M O'Connell
- Department of Pediatrics, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Boris Reidel
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Mehmet Kesimer
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Scott H Randell
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Toni Darville
- Department of Pediatrics, University of North Carolina, Chapel Hill, North Carolina, USA
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Uma M Nagarajan
- Department of Pediatrics, University of North Carolina, Chapel Hill, North Carolina, USA
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, North Carolina, USA
| |
Collapse
|
16
|
Fighting Persistence: How Chronic Infections with Mycobacterium tuberculosis Evade T Cell-Mediated Clearance and New Strategies To Defeat Them. Infect Immun 2020; 88:IAI.00916-19. [PMID: 32094248 DOI: 10.1128/iai.00916-19] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Chronic bacterial infections are caused by pathogens that persist within their hosts and avoid clearance by the immune system. Treatment and/or detection of such pathogens is difficult, and the resulting pathologies are often deleterious or fatal. There is an urgent need to develop protective vaccines and host-directed therapies that synergize with antibiotics to prevent pathogen persistence and infection-associated pathologies. However, many persistent pathogens, such as Mycobacterium tuberculosis, actively target the very host pathways activated by vaccination. These immune evasion tactics blunt the effectiveness of immunization strategies and are impeding progress to control these infections throughout the world. Therefore, it is essential that M. tuberculosis immune evasion-related pathogen virulence strategies are considered to maximize the effectiveness of potential new treatments. In this review, we focus on how Mycobacterium tuberculosis infects antigen-presenting cells and evades effective immune clearance by the adaptive response through (i) manipulating antigen presentation, (ii) repressing T cell-activating costimulatory molecules, and (iii) inducing ligands that drive T cell exhaustion. In this context, we will examine the challenges that bacterial virulence strategies pose to developing new vaccines. We will then discuss new approaches that will help dissect M. tuberculosis immune evasion mechanisms and devise strategies to bypass them to promote long-term protection and prevent disease progression.
Collapse
|
17
|
Pekmezovic M, Mogavero S, Naglik JR, Hube B. Host-Pathogen Interactions during Female Genital Tract Infections. Trends Microbiol 2019; 27:982-996. [PMID: 31451347 DOI: 10.1016/j.tim.2019.07.006] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 06/25/2019] [Accepted: 07/22/2019] [Indexed: 12/11/2022]
Abstract
Dysbiosis in the female genital tract (FGT) is characterized by the overgrowth of pathogenic bacterial, fungal, or protozoan members of the microbiota, leading to symptomatic or asymptomatic infections. In this review, we discuss recent advances in studies dealing with molecular mechanisms of pathogenicity factors of Gardnerella vaginalis, Mycoplasma genitalium, Mycoplasma hominis, Neisseria gonorrhoeae, Streptococcus agalactiae, Chlamydia trachomatis, Trichomonas vaginalis, and Candida spp., as well as their interactions with the host and microbiota in the various niches of the FGT. Taking a holistic approach to identifying fundamental commonalities and differences during these infections could help us to better understand reproductive tract health and improve current prevention and treatment strategies.
Collapse
Affiliation(s)
- Marina Pekmezovic
- Department of Microbial Pathogenicity Mechanisms, Hans Knoell Institute, Jena, Germany
| | - Selene Mogavero
- Department of Microbial Pathogenicity Mechanisms, Hans Knoell Institute, Jena, Germany
| | - Julian R Naglik
- Centre for Host-Microbiome Interactions, Faculty of Dental, Oral, and Craniofacial Sciences, King's College London, SE1 1UL, UK
| | - Bernhard Hube
- Department of Microbial Pathogenicity Mechanisms, Hans Knoell Institute, Jena, Germany; Institute of Microbiology, Friedrich Schiller University, Jena, Germany. @leibniz-hki.de
| |
Collapse
|
18
|
Chlamydia and Its Many Ways of Escaping the Host Immune System. J Pathog 2019; 2019:8604958. [PMID: 31467721 PMCID: PMC6699355 DOI: 10.1155/2019/8604958] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Accepted: 07/02/2019] [Indexed: 12/18/2022] Open
Abstract
The increasing number of new cases of Chlamydia infection worldwide may be attributed to the pathogen's ability to evade various host immune responses. Summarized here are means of evasion utilized by Chlamydia enabling survival in a hostile host environment. The pathogen's persistence involves a myriad of molecular interactions manifested in a variety of ways, e.g., formation of membranous intracytoplasmic inclusions and cytokine-induced amino acid synthesis, paralysis of phagocytic neutrophils, evasion of phagocytosis, inhibition of host cell apoptosis, suppression of antigen presentation, and induced expression of a check point inhibitor of programmed host cell death. Future studies could focus on the targeting of these molecules associated with immune evasion, thus limiting the spread and tissue damage caused by this pathogen.
Collapse
|
19
|
Kumar R, Gong H, Liu L, Ramos-Solis N, Seye CI, Derbigny WA. TLR3 deficiency exacerbates the loss of epithelial barrier function during genital tract Chlamydia muridarum infection. PLoS One 2019; 14:e0207422. [PMID: 30625140 PMCID: PMC6326510 DOI: 10.1371/journal.pone.0207422] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Accepted: 12/22/2018] [Indexed: 12/18/2022] Open
Abstract
Problem Chlamydia trachomatis infections are often associated with acute syndromes including cervicitis, urethritis, and endometritis, which can lead to chronic sequelae such as pelvic inflammatory disease (PID), chronic pelvic pain, ectopic pregnancy, and tubal infertility. As epithelial cells are the primary cell type productively infected during genital tract Chlamydia infections, we investigated whether Chlamydia has any impact on the integrity of the host epithelial barrier as a possible mechanism to facilitate the dissemination of infection, and examined whether TLR3 function modulates its impact. Method of study We used wild-type and TLR3-deficient murine oviduct epithelial (OE) cells to ascertain whether C. muridarum infection had any effect on the epithelial barrier integrity of these cells as measured by transepithelial resistance (TER) and cell permeability assays. We next assessed whether infection impacted the transcription and protein function of the cellular tight-junction (TJ) genes for claudins1-4, ZO-1, JAM1 and occludin via quantitative real-time PCR (qPCR) and western blot. Results qPCR, immunoblotting, transwell permeability assays, and TER studies show that Chlamydia compromises cellular TJ function throughout infection in murine OE cells and that TLR3 deficiency significantly exacerbates this effect. Conclusion Our data show that TLR3 plays a role in modulating epithelial barrier function during Chlamydia infection of epithelial cells lining the genital tract. These findings propose a role for TLR3 signaling in maintaining the integrity of epithelial barrier function during genital tract Chlamydia infection, a function that we hypothesize is important in helping limit the chlamydial spread and subsequent genital tract pathology.
Collapse
Affiliation(s)
- Ramesh Kumar
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Haoli Gong
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
- Xiangya Second Hospital, Central South University, Changsha, Hunan Province, People’s Republic of China
| | - Luyao Liu
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
- Xiangya Second Hospital, Central South University, Changsha, Hunan Province, People’s Republic of China
| | - Nicole Ramos-Solis
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Cheikh I. Seye
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Wilbert A. Derbigny
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
- * E-mail:
| |
Collapse
|
20
|
Ibana JA, Sherchand SP, Fontanilla FL, Nagamatsu T, Schust DJ, Quayle AJ, Aiyar A. Chlamydia trachomatis-infected cells and uninfected-bystander cells exhibit diametrically opposed responses to interferon gamma. Sci Rep 2018; 8:8476. [PMID: 29855501 PMCID: PMC5981614 DOI: 10.1038/s41598-018-26765-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Accepted: 05/18/2018] [Indexed: 12/13/2022] Open
Abstract
The intracellular bacterial pathogen, Chlamydia trachomatis, is a tryptophan auxotroph. Therefore, induction of the host tryptophan catabolizing enzyme, indoleamine-2,3-dioxgenase-1 (IDO1), by interferon gamma (IFNγ) is one of the primary protective responses against chlamydial infection. However, despite the presence of a robust IFNγ response, active and replicating C. trachomatis can be detected in cervical secretions of women. We hypothesized that a primary C. trachomatis infection may evade the IFNγ response, and that the protective effect of this cytokine results from its activation of tryptophan catabolism in bystander cells. To test this hypothesis, we developed a novel method to separate a pool of cells exposed to C. trachomatis into pure populations of live infected and bystander cells and applied this technique to distinguish between the effects of IFNγ on infected and bystander cells. Our findings revealed that the protective induction of IDO1 is suppressed specifically within primary infected cells because Chlamydia attenuates the nuclear import of activated STAT1 following IFNγ exposure, without affecting STAT1 levels or phosphorylation. Critically, the IFNγ-mediated induction of IDO1 activity is unhindered in bystander cells. Therefore, the IDO1-mediated tryptophan catabolism is functional in these cells, transforming these bystander cells into inhospitable hosts for a secondary C. trachomatis infection.
Collapse
Affiliation(s)
- Joyce A Ibana
- Immunopharmacology Research Laboratory, Institute of Biology, College of Science, University of the Philippines, Diliman, Quezon City, 1101, Philippines.
| | - Shardulendra P Sherchand
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA, 70112, USA
| | - Francis L Fontanilla
- Immunopharmacology Research Laboratory, Institute of Biology, College of Science, University of the Philippines, Diliman, Quezon City, 1101, Philippines
| | - Takeshi Nagamatsu
- Department of Obstetrics and Gynecology, Faculty Medicine, University of Tokyo, Tokyo, Japan
| | - Danny J Schust
- Department of Obstetrics, Gynecology, and Women's Health, University of Missouri, Columbia, MO, 15276, USA
| | - Alison J Quayle
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA, 70112, USA
| | - Ashok Aiyar
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA, 70112, USA
| |
Collapse
|
21
|
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
|
22
|
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
|
23
|
Cerny KL, Van Fleet M, Slepenkin A, Peterson EM, Bridges PJ. Differential Expression of mRNA Encoding Cytokines and Chemokines in the Reproductive Tract after Infection of Mice with Chlamydia trachomatis. ACTA ACUST UNITED AC 2015; 4. [PMID: 26779389 PMCID: PMC4712740 DOI: 10.4172/2161-038x.1000152] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Infection with Chlamydia trachomatis targets epithelial cells within the genital tract which respond by secreting chemokines and cytokines. Persistent inflammation can lead to fibrosis, tubal infertility and/or ectopic pregnancy; many infections are asymptomatic. Most studies have investigated the inflammatory response in the initial stages of infection, less is known about the later stages of infection, especially with a low, potentially asymptomatic, bacterial load. Our objective was to determine the inflammatory mediators involved in clearance of low-grade infection and the potential involvement in chronic inflammation. Six to eight week old C3H/HeJ mice were pretreated with 2.5 mg medroxyprogesterone acetate on day -10 and -3 before infection. Mice (n=3 for 28 d, n=3 for 35 d) were infected with 5 × 102 inclusion-forming units of C. trachomatis, serovar D; vaginal cultures were obtained weekly to monitor infection. Control mice (n=3 for 28 d, n=3 for 35 d) were sham infected. Mice were killed on day 28 (experiment 1) and day 35 (experiment 2) post-infection and vaginal tissue, uterine horns and oviducts collected for analysis of mRNAs encoding inflammatory cytokines and chemokines. Total RNA was isolated and a superarray analysis performed using mouse Cytokines and Chemokines PCR arrays (Qiagen, Valencia, CA). Statistical differences in gene expression were determined using a paired Students t-test. At 28 days after infection, the expression of mRNA encoding 6, 35 and 3 inflammatory genes differed from controls in vaginal, uterine and oviductal tissues, respectively (P<0.05). At 35 days after infection, the expression of mRNA encoding 16, 38 and 14 inflammatory genes differed from controls in vaginal, uterine and oviductal tissues, respectively (P<0.05). Understanding the mechanisms involved in the inflammatory response at later stages of infection should aid in the development of treatment options that minimize the development of asymptomatic, chronic inflammation-induced infertility.
Collapse
Affiliation(s)
- Katheryn L Cerny
- Department of Animal and Food Sciences, University of Kentucky, Lexington, KY, USA
| | - Maranda Van Fleet
- Department of Animal and Food Sciences, University of Kentucky, Lexington, KY, USA
| | - Anatoly Slepenkin
- Department of Pathology and Laboratory Medicine, University of California Irvine, Irvine, CA, USA
| | - Ellena M Peterson
- Department of Pathology and Laboratory Medicine, University of California Irvine, Irvine, CA, USA
| | - Phillip J Bridges
- Department of Animal and Food Sciences, University of Kentucky, Lexington, KY, USA
| |
Collapse
|
24
|
Beiting DP, Hidano S, Baggs JE, Geskes JM, Fang Q, Wherry EJ, Hunter CA, Roos DS, Cherry S. The Orphan Nuclear Receptor TLX Is an Enhancer of STAT1-Mediated Transcription and Immunity to Toxoplasma gondii. PLoS Biol 2015. [PMID: 26196739 PMCID: PMC4509904 DOI: 10.1371/journal.pbio.1002200] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
The protozoan parasite, Toxoplasma, like many intracellular pathogens, suppresses interferon gamma (IFN-γ)-induced signal transducer and activator of transcription 1 (STAT1) activity. We exploited this well-defined host–pathogen interaction as the basis for a high-throughput screen, identifying nine transcription factors that enhance STAT1 function in the nucleus, including the orphan nuclear hormone receptor TLX. Expression profiling revealed that upon IFN-γ treatment TLX enhances the output of a subset of IFN-γ target genes, which we found is dependent on TLX binding at those loci. Moreover, infection of TLX deficient mice with the intracellular parasite Toxoplasma results in impaired production of the STAT1-dependent cytokine interleukin-12 by dendritic cells and increased parasite burden in the brain during chronic infection. These results demonstrate a previously unrecognized role for this orphan nuclear hormone receptor in regulating STAT1 signaling and host defense and reveal that STAT1 activity can be modulated in a context-specific manner by such “modifiers.” Exploitation of the parasite Toxoplasma gondii identifies the host orphan nuclear hormone receptor TLX as a key enhancer of STAT1-dependent immune signaling and host defense. Immune responses are orchestrated by a diverse array of secreted ligands, yet the downstream transcriptional responses are coordinated by a relatively small set of key transcription factors, including nuclear factor kappa B (NF-κB) and signal transducers and activators of transcription (STATs). The molecular mechanisms that tailor the output of these immune signaling pathways to generate cell-, tissue-, or context-specific responses are poorly understood. In this study, we exploit a host–pathogen interaction, Toxoplasma gondii infection in mice, using a genetic screen to identify host factors that overcome parasite suppression of STAT1 signaling. We show that the orphan nuclear receptor TLX, a key regulator of brain development, enhances expression of a subset of STAT1-dependent genes in response to IFN-γ stimulation. Through genetic and pharmacological studies, we show that endogenous TLX function is required for triggering appropriate responses to IFN-γ in astrocytes. Moreover, we found that genetic disruption of TLX in mice impairs their ability to mount an effective immune response and control T. gondii infection in the brain. These data suggest that natural or synthetic ligands for TLX might be effective tools for modulating immune responses, particularly in the brain where TLX expression is highest.
Collapse
Affiliation(s)
- Daniel P. Beiting
- Department of Pathobiology, University of Pennsylvania, Philadelphia, Pennsylvania, Philadelphia, United States of America
| | - Shinya Hidano
- Department of Pathobiology, University of Pennsylvania, Philadelphia, Pennsylvania, Philadelphia, United States of America
| | - Julie E. Baggs
- Department of Pharmacology and the Institute for Translational Medicine and Therapeutics, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Jeanne M. Geskes
- Department of Pharmacology and the Institute for Translational Medicine and Therapeutics, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Qun Fang
- Department of Pathobiology, University of Pennsylvania, Philadelphia, Pennsylvania, Philadelphia, United States of America
| | - E. John Wherry
- Department of Microbiology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Christopher A. Hunter
- Department of Pathobiology, University of Pennsylvania, Philadelphia, Pennsylvania, Philadelphia, United States of America
| | - David S. Roos
- Department of Biology, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- * E-mail: (DSR); (SC)
| | - Sara Cherry
- Department of Microbiology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
- * E-mail: (DSR); (SC)
| |
Collapse
|
25
|
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
|
26
|
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
|
27
|
Leroux LP, Dasanayake D, Rommereim LM, Fox BA, Bzik DJ, Jardim A, Dzierszinski FS. Secreted Toxoplasma gondii molecules interfere with expression of MHC-II in interferon gamma-activated macrophages. Int J Parasitol 2015; 45:319-32. [PMID: 25720921 DOI: 10.1016/j.ijpara.2015.01.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2014] [Revised: 01/16/2015] [Accepted: 01/18/2015] [Indexed: 11/15/2022]
Abstract
The obligate intracellular protozoan parasite Toxoplasma gondii interferes with major histocompatibility complex class II antigen presentation to dampen host CD4(+) T cell responses. While it is known that T. gondii inhibits major histocompatibility complex class II gene transcription and expression in infected host cells, the mechanism of this host manipulation is unknown. Here, we show that soluble parasite proteins inhibit IFNγ-induced expression of major histocompatibility complex class II on the surface of the infected cell in a dose-dependent response that was abolished by protease treatment. Subcellular fractionation of T. gondii tachyzoites revealed that the major histocompatibility complex class II inhibitory activity co-partitioned with rhoptries and/or dense granules. However, parasite mutants deleted for single rhoptries or dense granules genes (ROP1, 4/7, 14, 16 and 18 or GRA 2-9 and 12 knock-out strains) retained the ability to inhibit expression of major histocompatibility complex class II. In addition, excreted/secreted antigens released by extracellular tachyzoites displayed immunomodulatory activity characterized by an inhibition of major histocompatibility complex class II expression, and reduced expression and release of TNFα by macrophages. Tandem MS analysis of parasite excreted/secreted antigens generated a list of T. gondii secreted proteins that may participate in major histocompatibility complex class II inhibition and the modulation of host immune functions.
Collapse
Affiliation(s)
- Louis-Philippe Leroux
- Institute of Parasitology, McGill University, Parasitology Building, 21,111 Lakeshore Road, Sainte-Anne-de-Bellevue, QC H9X 3V9, Canada; Centre for Host-Parasite Interaction, McGill University, Parasitology Building, 21,111 Lakeshore Road, Sainte-Anne-de-Bellevue, QC H9X 3V9, Canada
| | - Dayal Dasanayake
- Institute of Parasitology, McGill University, Parasitology Building, 21,111 Lakeshore Road, Sainte-Anne-de-Bellevue, QC H9X 3V9, Canada; Centre for Host-Parasite Interaction, McGill University, Parasitology Building, 21,111 Lakeshore Road, Sainte-Anne-de-Bellevue, QC H9X 3V9, Canada
| | - Leah M Rommereim
- Geisel School of Medicine at Dartmouth, Borwell Research Building, 1 Medical Center Drive, Lebanon, NH 03756, USA
| | - Barbara A Fox
- Geisel School of Medicine at Dartmouth, Borwell Research Building, 1 Medical Center Drive, Lebanon, NH 03756, USA
| | - David J Bzik
- Geisel School of Medicine at Dartmouth, Borwell Research Building, 1 Medical Center Drive, Lebanon, NH 03756, USA
| | - Armando Jardim
- Institute of Parasitology, McGill University, Parasitology Building, 21,111 Lakeshore Road, Sainte-Anne-de-Bellevue, QC H9X 3V9, Canada; Centre for Host-Parasite Interaction, McGill University, Parasitology Building, 21,111 Lakeshore Road, Sainte-Anne-de-Bellevue, QC H9X 3V9, Canada.
| | - Florence S Dzierszinski
- Institute of Parasitology, McGill University, Parasitology Building, 21,111 Lakeshore Road, Sainte-Anne-de-Bellevue, QC H9X 3V9, Canada; Carleton University Research Office, Dunton Tower, 1125 Colonel By Drive, Ottawa, ON K1S 5B6, Canada
| |
Collapse
|
28
|
Redgrove KA, McLaughlin EA. The Role of the Immune Response in Chlamydia trachomatis Infection of the Male Genital Tract: A Double-Edged Sword. Front Immunol 2014; 5:534. [PMID: 25386180 PMCID: PMC4209867 DOI: 10.3389/fimmu.2014.00534] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Accepted: 10/09/2014] [Indexed: 01/16/2023] Open
Abstract
Chlamydia trachomatis (CT) is the most prevalent bacterial sexually transmitted infection in the world, with more than 100 million cases reported annually. While there have been extensive studies into the adverse effects that CT infection has on the female genital tract, and on the subsequent ability of these women to conceive, studies into the consequences on male fertility have been limited and controversial. This is in part due to the asymptomatic nature of the infection, where it is estimated that 50% of men with Chlamydia fail to show any symptoms. It is accepted, however, that acute and/or persistent CT infection is the causative agent for conditions such as urethritis, epididymitis, epididymo-orchitis, and potentially prostatitis. As with most infections, the immune system plays a fundamental role in the body’s attempts to eradicate the infection. The first and most important immune response to Chlamydia infection is a local one, whereby immune cells such as leukocytes are recruited to the site of infections, and subsequently secrete pro-inflammatory cytokines and chemokines such as interferon gamma. Immune cells also work to initiate and potentiate chronic inflammation through the production of reactive oxygen species (ROS), and the release of molecules with degradative properties including defensins, elastase, collagenase, cathespins, and lysozyme. This long-term inflammation can lead to cell proliferation (a possible precursor to cancer), tissue remodeling, and scarring, as well as being linked to the onset of autoimmune responses in genetically disposed individuals. This review will focus on the ability of the immune system to recognize and clear acute and persistent chlamydial infections in the male genital tract, and on the paradoxical damage that chronic inflammation resulting from the infection can cause on the reproductive health of the individual.
Collapse
Affiliation(s)
- Kate A Redgrove
- Priority Research Centre in Reproductive Biology and Chemical Biology, University of Newcastle , Callaghan, NSW , Australia ; School of Environmental and Life Science, University of Newcastle , Callaghan, NSW , Australia
| | - Eileen A McLaughlin
- Priority Research Centre in Reproductive Biology and Chemical Biology, University of Newcastle , Callaghan, NSW , Australia ; School of Environmental and Life Science, University of Newcastle , Callaghan, NSW , Australia
| |
Collapse
|
29
|
Gupta R, Arkatkar T, Yu JJ, Wali S, Haskins WE, Chambers JP, Murthy AK, Bakar SA, Guentzel MN, Arulanandam BP. Chlamydia muridarum infection associated host MicroRNAs in the murine genital tract and contribution to generation of host immune response. Am J Reprod Immunol 2014; 73:126-40. [PMID: 24976530 DOI: 10.1111/aji.12281] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Accepted: 05/21/2014] [Indexed: 12/23/2022] Open
Abstract
PROBLEM Chlamydia trachomatis (CT) is the leading sexually transmitted bacterial infection in humans and is associated with reproductive tract damage. However, little is known about the involvement and regulation of microRNAs (miRs) in genital CT. METHODS We analyzed miRs in the genital tract (GT) following C. muridarum (murine strain of CT) challenge of wild type (WT) and CD4(+) T-cell deficient (CD4(-/-)) C57BL/6 mice at days 6 and 12 post-challenge. RESULTS At day 6, miRs significantly downregulated in the lower GT were miR-125b-5p, -16, -214, -23b, -135a, -182, -183, -30c, and -30e while -146 and -451 were significantly upregulated, profiles not exhibited at day 12 post-bacterial challenge. Significant differences in miR-125b-5p (+5.06-fold change), -135a (+4.9), -183 (+7.9), and -182 (+3.2) were observed in C. muridarum-infected CD4(-/-) compared to WT mice. In silico prediction and mass spectrometry revealed regulation of miR-135a and -182 and associated proteins, that is, heat-shock protein B1 and alpha-2HS-glycoprotein. CONCLUSION This study provides evidence on regulation of miRs following genital chlamydial infection suggesting a role in pathogenesis and host immunity.
Collapse
Affiliation(s)
- Rishein Gupta
- South Texas Center for Emerging Infectious Diseases and Center of Excellence in Infection Genomics, University of Texas at San Antonio, San Antonio, TX, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Hafner LM, Wilson DP, Timms P. Development status and future prospects for a vaccine against Chlamydia trachomatis infection. Vaccine 2013; 32:1563-71. [PMID: 23973245 DOI: 10.1016/j.vaccine.2013.08.020] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2013] [Revised: 07/04/2013] [Accepted: 08/11/2013] [Indexed: 01/08/2023]
Abstract
Chlamydia trachomatis continues to be the most commonly reported sexually transmitted bacterial infection in many countries with more than 100 million new cases estimated annually. These acute infections translate into significant downstream health care costs, particularly for women, where complications can include pelvic inflammatory disease and other disease sequelae such as tubal factor infertility. Despite years of research, the immunological mechanisms responsible for protective immunity versus immunopathology are still not well understood, although it is widely accepted that T cell driven IFN-g and Th17 responses are critical for clearing infection. While antibodies are able to neutralize infections in vitro, alone they are not protective, indicating that any successful vaccine will need to elicit both arms of the immune response. In recent years, there has been an expansion in the number and types of antigens that have been evaluated as vaccines, and combined with the new array of mucosal adjuvants, this aspect of chlamydial vaccinology is showing promise. Most recently, the opportunities to develop successful vaccines have been given a significant boost with the development of a genetic transformation system for Chlamydia, as well as the identification of the key role of the chlamydial plasmid in virulence. While still remaining a major challenge, the development of a successful C. trachomatis vaccine is starting to look more likely.
Collapse
Affiliation(s)
- Louise M Hafner
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Australia
| | - David P Wilson
- The Kirby Institute, University of New South Wales, Sydney, Australia
| | - Peter Timms
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Australia.
| |
Collapse
|
31
|
Alvarez-Navarro C, Cragnolini JJ, Dos Santos HG, Barnea E, Admon A, Morreale A, López de Castro JA. Novel HLA-B27-restricted epitopes from Chlamydia trachomatis generated upon endogenous processing of bacterial proteins suggest a role of molecular mimicry in reactive arthritis. J Biol Chem 2013; 288:25810-25825. [PMID: 23867464 DOI: 10.1074/jbc.m113.493247] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Reactive arthritis (ReA) is an HLA-B27-associated spondyloarthropathy that is triggered by diverse bacteria, including Chlamydia trachomatis, a frequent intracellular parasite. HLA-B27-restricted T-cell responses are elicited against this bacterium in ReA patients, but their pathogenetic significance, autoimmune potential, and relevant epitopes are unknown. High resolution and sensitivity mass spectrometry was used to identify HLA-B27 ligands endogenously processed and presented by HLA-B27 from three chlamydial proteins for which T-cell epitopes were predicted. Fusion protein constructs of ClpC, Na(+)-translocating NADH-quinone reductase subunit A, and DNA primase were expressed in HLA-B27(+) cells, and their HLA-B27-bound peptidomes were searched for endogenous bacterial ligands. A non-predicted peptide, distinct from the predicted T-cell epitope, was identified from ClpC. A peptide recognized by T-cells in vitro, NQRA(330-338), was detected from the reductase subunit. This is the second HLA-B27-restricted T-cell epitope from C. trachomatis with relevance in ReA demonstrated to be processed and presented in live cells. A novel peptide from the DNA primase, DNAP(211-223), was also found. This was a larger variant of a known epitope and was highly homologous to a self-derived natural ligand of HLA-B27. All three bacterial peptides showed high homology with human sequences containing the binding motif of HLA-B27. Molecular dynamics simulations further showed a striking conformational similarity between DNAP(211-223) and its homologous and much more flexible human-derived HLA-B27 ligand. The results suggest that molecular mimicry between HLA-B27-restricted bacterial and self-derived epitopes is frequent and may play a role in ReA.
Collapse
Affiliation(s)
- Carlos Alvarez-Navarro
- From the Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas and Universidad Autónoma, Madrid, Spain and
| | - Juan J Cragnolini
- From the Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas and Universidad Autónoma, Madrid, Spain and
| | - Helena G Dos Santos
- From the Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas and Universidad Autónoma, Madrid, Spain and
| | - Eilon Barnea
- the Faculty of Biology, Technion-Israel Institute of Technology, Haifa 32000, Israel
| | - Arie Admon
- the Faculty of Biology, Technion-Israel Institute of Technology, Haifa 32000, Israel
| | - Antonio Morreale
- From the Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas and Universidad Autónoma, Madrid, Spain and
| | - José A López de Castro
- From the Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas and Universidad Autónoma, Madrid, Spain and.
| |
Collapse
|
32
|
Chlamydia trachomatis infection results in a modest pro-inflammatory cytokine response and a decrease in T cell chemokine secretion in human polarized endocervical epithelial cells. Cytokine 2013; 63:151-65. [PMID: 23673287 DOI: 10.1016/j.cyto.2013.04.022] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2013] [Revised: 04/11/2013] [Accepted: 04/12/2013] [Indexed: 01/19/2023]
Abstract
The endocervical epithelium is a major reservoir for Chlamydia trachomatis in women, and genital infections are extended in their duration. Epithelial cells act as mucosal sentinels by secreting cytokines and chemokines in response to pathogen challenge and infection. We therefore determined the signature cytokine and chemokine response of primary-like endocervix-derived epithelial cells in response to a common genital serovar (D) of C. trachomatis. For these studies, we used a recently-established polarized, immortalized, endocervical epithelial cell model (polA2EN) that maintains, in vitro, the architectural and functional characteristics of endocervical epithelial cells in vivo including the production of pro-inflammatory cytokines. PolA2EN cells were susceptible to C. trachomatis infection, and chlamydiae in these cells underwent a normal developmental cycle as determined by a one-step growth curve. IL1α protein levels were increased in both apical and basolateral secretions of C. trachomatis infected polA2EN cells, but this response did not occur until 72h after infection. Furthermore, protein levels of the pro-inflammatory cytokines and chemokines IL6, TNFα and CXCL8 were not significantly different between C. trachomatis infected polA2EN cells and mock infected cells at any time during the chlamydial developmental cycle up to 120h post-infection. Intriguingly, C. trachomatis infection resulted in a significant decrease in the constitutive secretion of T cell chemokines IP10 and RANTES, and this required a productive C. trachomatis infection. Examination of anti-inflammatory cytokines revealed a high constitutive apical secretion of IL1ra from polA2EN cells that was not significantly modulated by C. trachomatis infection. IL-11 was induced by C. trachomatis, although only from the basolateral membrane. These results suggest that C. trachomatis can use evasion strategies to circumvent a robust pro-inflammatory cytokine and chemokine response. These evasion strategies, together with the inherent immune repertoire of endocervical epithelial cells, may aid chlamydiae in establishing, and possibly sustaining, an intracellular niche in microenvironments of the endocervix in vivo.
Collapse
|
33
|
Bastidas RJ, Elwell CA, Engel JN, Valdivia RH. Chlamydial intracellular survival strategies. Cold Spring Harb Perspect Med 2013; 3:a010256. [PMID: 23637308 DOI: 10.1101/cshperspect.a010256] [Citation(s) in RCA: 160] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Chlamydia trachomatis is the most common sexually transmitted bacterial pathogen and the causative agent of blinding trachoma. Although Chlamydia is protected from humoral immune responses by residing within remodeled intracellular vacuoles, it still must contend with multilayered intracellular innate immune defenses deployed by its host while scavenging for nutrients. Here we provide an overview of Chlamydia biology and highlight recent findings detailing how this vacuole-bound pathogen manipulates host-cellular functions to invade host cells and maintain a replicative niche.
Collapse
Affiliation(s)
- Robert J Bastidas
- Department of Molecular Genetics and Microbiology, Center for Microbial Pathogenesis, Duke University Medical Center, Durham, NC 27710, USA
| | | | | | | |
Collapse
|
34
|
Alibek K, Karatayeva N, Bekniyazov I. The role of infectious agents in urogenital cancers. Infect Agent Cancer 2012; 7:35. [PMID: 23198689 PMCID: PMC3626724 DOI: 10.1186/1750-9378-7-35] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2012] [Accepted: 11/20/2012] [Indexed: 02/07/2023] Open
Abstract
Since the late 1990s, infectious agents have been thought to play a role in the pathogenesis of approximately 15% of cancers. It is now widely accepted that infection of stomach tissue with the bacteria Helicobacter pylori is an important cause of stomach adenocarcinoma. In addition, oncogenic viruses, such as papilloma viruses, herpes viruses, and hepadnaviruses are strongly associated with increased risk of cervical cancer, lymphomas, liver cancer, amongst others. However, in the scientific community the percentage of cancers caused by pathogens is believed to be far higher than 15%. A significant volume of data collected to date show an association between infectious agents and urogenital cancers. These agents include Chlamydia trachomatis, Neisseria gonorrhoea, Mycoplasma genitalium and certain viruses that have been implicated in ovarian cancer. Other pathogens include the hepatitis C and Epstein-Barr viruses, which are potentially involved in kidney cancer. In addition, infections with Schistosoma haematobium, the human papillomavirus, and human polyomaviruses are strongly associated with an increased risk of urinary bladder cancer. This article reviews publications available to date on the role of infectious agents in urogenital cancers. A greater understanding of the role of such agents could aid the identification of novel methods of urogenital cancer treatment.
Collapse
Affiliation(s)
- Kenneth Alibek
- Nazarbayev University, 53 Kabanbay Batyr Avenue, Astana 010000, Kazakhstan
- Republican Scientific Center for Emergency Care, 3 Kerey and Zhanibek Khan Street, Astana 010000, Kazakhstan
| | - Nargis Karatayeva
- Nazarbayev University, 53 Kabanbay Batyr Avenue, Astana 010000, Kazakhstan
| | - Ildar Bekniyazov
- Nazarbayev University, 53 Kabanbay Batyr Avenue, Astana 010000, Kazakhstan
| |
Collapse
|
35
|
Axtner J, Sommer S. The functional importance of sequence versus expression variability of MHC alleles in parasite resistance. Genetica 2012. [PMID: 23180005 DOI: 10.1007/s10709-012-9689-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Understanding selection processes driving the pronounced allelic polymorphism of the major histocompatibility complex (MHC) genes and its functional associations to parasite load have been the focus of many recent wildlife studies. Two main selection scenarios are currently debated which explain the susceptibility or resistance to parasite infections either by the effects of (1) specific MHC alleles which are selected frequency-dependent in space and time or (2) a heterozygote or divergent allele advantage. So far, most studies have focused only on structural variance in co-evolutionary processes although this might not be the only trait subject to natural selection. In the present study, we analysed structural variance stretching from exon1 through exon3 of MHC class II DRB genes as well as genotypic expression variance in relation to the gastrointestinal helminth prevalence and infection intensity in wild yellow-necked mice (Apodemus flavicollis). We found support for the functional importance of specific alleles both on the sequence and expression level. By resampling a previously investigated study population we identified specific MHC alleles affected by temporal shifts in parasite pressure and recorded associated changes in allele frequencies. The allele Apfl-DRB*23 was associated with resistance to infections by the oxyurid nematode Syphacia stroma and at the same time with susceptibility to cestode infection intensity. In line with our expectation, MHC mRNA transcript levels tended to be higher in cestode-infected animals carrying the allele Apfl-DRB*23. However, no support for a heterozygote or divergent allele advantage on the sequence or expression level was detected. The individual amino acid distance of genotypes did not explain individual differences in parasite loads and the genetic distance had no effect on MHC genotype expression. For ongoing studies on the functional importance of expression variance in parasite resistance, allele-specific expression data would be preferable.
Collapse
Affiliation(s)
- Jan Axtner
- Evolutionary Genetics, Leibniz Institute for Zoo and Wildlife Research, Alfred-Kowalke-Str. 15, 10315, Berlin, Germany.
| | | |
Collapse
|
36
|
Chen AL, Johnson KA, Lee JK, Sütterlin C, Tan M. CPAF: a Chlamydial protease in search of an authentic substrate. PLoS Pathog 2012; 8:e1002842. [PMID: 22876181 PMCID: PMC3410858 DOI: 10.1371/journal.ppat.1002842] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2012] [Accepted: 06/22/2012] [Indexed: 01/13/2023] Open
Abstract
Bacteria in the genus Chlamydia are major human pathogens that cause an intracellular infection. A chlamydial protease, CPAF, has been proposed as an important virulence factor that cleaves or degrades at least 16 host proteins, thereby altering multiple cellular processes. We examined 11 published CPAF substrates and found that there was no detectable proteolysis when CPAF activity was inhibited during cell processing. We show that the reported proteolysis of these putative CPAF substrates was due to enzymatic activity in cell lysates rather than in intact cells. Nevertheless, Chlamydia-infected cells displayed Chlamydia-host interactions, such as Golgi reorganization, apoptosis resistance, and host cytoskeletal remodeling, that have been attributed to CPAF-dependent proteolysis of host proteins. Our findings suggest that other mechanisms may be responsible for these Chlamydia-host interactions, and raise concerns about all published CPAF substrates and the proposed roles of CPAF in chlamydial pathogenesis. Chlamydia are bacteria that invade eukaryotic host cells and live within a membrane-bound compartment called the chlamydial inclusion. Growth and survival of these important human and animal pathogens depends on extensive interactions with the host cell, which allow chlamydiae to acquire critical nutrients and to avoid host anti-microbial defenses. Chlamydiae are proposed to cause many of these host-pathogen interactions through the cleavage or degradation of host proteins by the chlamydial protease CPAF, which is secreted into the host cytoplasm. Here, we raise questions about the proposed roles of this virulence factor during infection, as well as its published substrates. We found that there was no detectable cleavage or degradation of 11 previously reported CPAF substrates in Chlamydia-infected cells and that CPAF-mediated proteolysis of these host proteins occurs during cell harvest and lysis. However, we still observed host-pathogen interactions previously attributed to CPAF proteolysis of these proteins, suggesting that Chlamydia is likely to cause these effects on the host cell through other mechanisms. Our findings call for a re-evaluation of all published CPAF substrates as well as the proposed roles of this protease in chlamydial pathogenesis.
Collapse
Affiliation(s)
- Allan L. Chen
- Department of Microbiology and Molecular Genetics, University of California at Irvine, Irvine, California, United States of America
| | - Kirsten A. Johnson
- Department of Microbiology and Molecular Genetics, University of California at Irvine, Irvine, California, United States of America
- Department of Developmental and Cell Biology, University of California at Irvine, Irvine, California, United States of America
| | - Jennifer K. Lee
- Department of Microbiology and Molecular Genetics, University of California at Irvine, Irvine, California, United States of America
- Department of Developmental and Cell Biology, University of California at Irvine, Irvine, California, United States of America
| | - Christine Sütterlin
- Department of Developmental and Cell Biology, University of California at Irvine, Irvine, California, United States of America
- * E-mail: (CS); (MT)
| | - Ming Tan
- Department of Microbiology and Molecular Genetics, University of California at Irvine, Irvine, California, United States of America
- Department of Medicine, University of California at Irvine, Irvine, California, United States of America
- * E-mail: (CS); (MT)
| |
Collapse
|
37
|
Ibana JA, Aiyar A, Quayle AJ, Schust DJ. Modulation of MICA on the surface of Chlamydia trachomatis-infected endocervical epithelial cells promotes NK cell-mediated killing. FEMS IMMUNOLOGY AND MEDICAL MICROBIOLOGY 2012; 65:32-42. [PMID: 22251247 PMCID: PMC5029121 DOI: 10.1111/j.1574-695x.2012.00930.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2011] [Revised: 01/09/2012] [Accepted: 01/09/2012] [Indexed: 12/24/2022]
Abstract
Chlamydia trachomatis serovars D-K are obligate intracellular bacteria that have tropism for the columnar epithelial cells of the genital tract. Chlamydia trachomatis infection has been reported to induce modifications in immune cell ligand expression on epithelial host cells. In this study, we used an in vitro infection model that resulted in a partial infection of C. trachomatis-exposed primary-like immortalized endocervical epithelial cells (A2EN). Using this model, we demonstrated that expression of the natural killer (NK) cell activating ligand, MHC class I-related protein A (MICA), was upregulated on C. trachomatis-infected, but not on noninfected bystander cells. MICA upregulation was concomitant with MHC class I downregulation and impacted the susceptibility of C. trachomatis-infected cells to NK cell activity. The specificity of MICA upregulation was reflected by a higher cytolytic activity of an NK cell line (NK92MI) against C. trachomatis-infected cells compared with uninfected control cells. Significantly, data also indicated that NK cells exerted a partial, but incomplete sterilizing effect on C. trachomatis as shown by the reduction in recoverable inclusion forming units (IFU) when cocultured with C. trachomatis-infected cells. Taken together, our data suggest that NK cells may play a significant role in the ability of the host to counter C. trachomatis infection.
Collapse
Affiliation(s)
- Joyce Altamarino Ibana
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Science Center, New Orleans, LA, USA
| | - Ashok Aiyar
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Science Center, New Orleans, LA, USA
| | - Alison Jane Quayle
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Science Center, New Orleans, LA, USA
| | - Danny Joseph Schust
- Department of Obstetrics, Gynecology and Women’s Health, University of Missouri School of Medicine, Columbia, MO, USA
| |
Collapse
|
38
|
Lu C, Holland MJ, Gong S, Peng B, Bailey RL, Mabey DW, Wu Y, Zhong G. Genome-wide identification of Chlamydia trachomatis antigens associated with trachomatous trichiasis. Invest Ophthalmol Vis Sci 2012; 53:2551-9. [PMID: 22427578 DOI: 10.1167/iovs.11-9212] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
PURPOSE Chlamydia trachomatis is the leading infectious cause of blindness. The goal of the current study was to search for biomarkers associated with C. trachomatis-induced ocular pathologies. METHODS We used a whole genome scale proteome array to systematically profile antigen specificities of antibody responses to C. trachomatis infection in individuals from trachoma-endemic communities with or without end-stage trachoma (trichiasis) in The Gambia. RESULTS When 61 trichiasis patients were compared with their control counterparts for overall antibody reactivity with organisms of different chlamydial species, no statistically significant difference was found. Both groups developed significantly higher titers of antibodies against C. trachomatis ocular serovars A and B than ocular serovar C, genital serovar D, or Chlamydia psittaci, whereas the titers of anti-Chlamydia pneumoniae antibodies were the highest. When antisera from 33 trichiasis and 26 control patients (with relatively high titers of antibodies to C. trachomatis ocular serovars) were reacted with 908 C. trachomatis proteins, 447 antigens were recognized by at least 1 of the 59 antisera, and 10 antigens by 50% or more antisera, the latter being designated as immunodominant antigens. More importantly, four antigens were preferentially recognized by the trichiasis group, with antigens CT414, CT667, and CT706 collectively reacting with 30% of trichiasis antisera but none from the normal group, and antigen CT695 reacting with 61% of trichiasis but only 31% of normal antisera. On the other hand, eight antigens were preferentially recognized by the control group, with antigens CT019, CT117, CT301, CT553, CT556, CT571, and CT709 together reacting with 46% of normal antisera and none from the trichiasis group, whereas antigen CT442 reacted with 35% of normal and 19% of trichiasis antisera respectively. CONCLUSIONS The current study, by mapping immunodominant C. trachomatis antigens and identifying antigens associated with both ocular pathology and protection, has provided important information for further understanding chlamydial pathogenesis and the development of subunit vaccines.
Collapse
Affiliation(s)
- Chunxue Lu
- Department of Microbiology and Immunology, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Li J, Li J, You Y, Chen S. The role of upstream stimulatory factor 1 in the transcriptional regulation of the human TBX21 promoter mediated by the T-1514C polymorphism associated with systemic lupus erythematosus. Immunogenetics 2012; 64:361-70. [PMID: 22258560 DOI: 10.1007/s00251-011-0597-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2011] [Accepted: 12/23/2011] [Indexed: 01/11/2023]
Abstract
T-bet is a key regulator for the lineage commitment in CD4+ T helper (Th) 1 cells by activating the hallmark production of interferon-γ. Previously, two single nucleotide polymorphisms (SNPs) in the TBX21 promoter, T-1993C and T-1514C, have been shown by statistic studies to associate with systemic lupus erythematosus (SLE). The effect of -1993 SNP on the Yin Yang 1 transcription factor-mediated promoter activity has been already indicated. This study aimed to investigate roles of the T-1514C SNP on TBX21 transcription and its functional effect by luciferase reporter, electrophoretic mobility shift assay (EMSA), chromatin immunoprecipitation (ChIP) assay, and flow cytometric analysis of intracellular T-bet, IFN-γ, and IL-4 expression in activated CD4+ T cells. The TBX21 promoter carrying -1514C possessed significantly lower transcriptional activity than that of -1514T and was markedly downregulated by the overexpression of upstream stimulatory factor 1 (USF-1) when compared with the promoter carrying -1514T. EMSA indicated that the transcription factor USF-1 was bound to the -1514C allele probe with the affinity higher than that to the -1514T allele probe. ChIP assay suggested that USF-1 bound around -1514 of TBX21 genomic DNA in vivo in the human T cell line Jurkat with -1514C/T. The individuals carrying -1514C allele were determined to have significantly diminished expression of T-bet and IFN-γ and increased IL-4 production in CD4+ T cells compared with those of -1514T allele. The findings demonstrate that the T-1514C polymorphism affects TBX21 gene expression and Th1 cytokine production by binding USF-1 to the SNP site.
Collapse
Affiliation(s)
- Junggang Li
- Institute of Infectious Diseases, Southwest Hospital, Third Military Medical University, Chongqing, 400038, China
| | | | | | | |
Collapse
|
40
|
Axtner J, Sommer S. Heligmosomoides polygyrus infection is associated with lower MHC class II gene expression in Apodemus flavicollis: indication for immune suppression? INFECTION GENETICS AND EVOLUTION 2011; 11:2063-71. [PMID: 21983561 DOI: 10.1016/j.meegid.2011.09.020] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2011] [Revised: 09/22/2011] [Accepted: 09/22/2011] [Indexed: 01/16/2023]
Abstract
Due to their key role in recognizing foreign antigens and triggering the subsequent immune response the genes of the major histocompatibility complex (MHC) provide a potential target for parasites to attack in order to evade detection and expulsion from the host. A diminished MHC gene expression results in less activated T cells and might serve as a gateway for pathogens and parasites. Some parasites are suspected to be immune suppressors and promote co-infections of other parasites even in other parts of the body. In our study we found indications that the gut dwelling nematode Heligmosomoides polygyrus might exert a systemic immunosuppressive effect in yellow-necked mice (Apodemus flavicollis). The amount of hepatic MHC class II DRB gene RNA transcripts in infected mice was negatively associated with infection intensity with H. polygyrus. The hepatic expression of immunosuppressive cytokines, such as transforming growth factor β and interleukin 10 was not associated with H. polygyrus infection. We did not find direct positive associations of H. polygyrus with other helminth species. But the prevalence and infection intensity of the nematodes Syphacia stroma and Trichuris muris were higher in multiple infected individuals. Furthermore, our data indicated antagonistic effects in the helminth community of A. flavicollis as cestode infection correlated negatively with H. polygyrus and helminth species richness. Our study shows that expression analyses of immune relevant genes can also be performed in wildlife, opening new aspects and possibilities for future ecological and evolutionary research.
Collapse
Affiliation(s)
- Jan Axtner
- Evolutionary Genetics, Leibniz Institute for Zoo and Wildlife Research, Alfred-Kowalke-Str 15, 10315 Berlin, Germany
| | | |
Collapse
|
41
|
Antichlamydial antibodies, human fertility, and pregnancy wastage. Infect Dis Obstet Gynecol 2011; 2011:525182. [PMID: 21949601 PMCID: PMC3178110 DOI: 10.1155/2011/525182] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2011] [Accepted: 06/21/2011] [Indexed: 11/17/2022] Open
Abstract
Genital infections with Chlamydia trachomatis (C. trachomatis) continue to be a worldwide epidemic. Immune response to chlamydia is important to both clearance of the disease and disease pathogenesis. Interindividual responses and current chlamydial control programs will have enormous effects on this disease and its control strategies. Humoral immune response to C. trachomatis occurs in humans and persistent antibody levels appear to be most directly correlated with more severe and longstanding disease and with reinfection. There is a close correlation between the presence of antichlamydial antibodies in females and tubal factor infertility; the closest associations have been found for antibodies against chlamydial heat shock proteins. The latter antibodies have also been shown to be useful among infertile patients with prior ectopic pregnancy, and their presence has been correlated with poor IVF outcomes, including early pregnancy loss. We review the existing literature on chlamydial antibody testing in infertile patients and present an algorithm for such testing in the infertile couple.
Collapse
|
42
|
Srivastava P, Vardhan H, Bhengraj AR, Jha R, Singh LC, Salhan S, Mittal A. Azithromycin Treatment Modulates the Extracellular Signal-Regulated Kinase Mediated Pathway and Inhibits Inflammatory Cytokines and Chemokines in Epithelial Cells from Infertile Women with RecurrentChlamydia trachomatisInfection. DNA Cell Biol 2011; 30:545-54. [DOI: 10.1089/dna.2010.1167] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Pragya Srivastava
- Institute of Pathology, Indian Council of Medical Research, New Delhi, India
| | - Harsh Vardhan
- Institute of Pathology, Indian Council of Medical Research, New Delhi, India
| | | | - Rajneesh Jha
- Institute of Pathology, Indian Council of Medical Research, New Delhi, India
| | | | - Sudha Salhan
- Department of Gynecology and Obstetrics, Safdarjung Hospital, New Delhi, India
| | - Aruna Mittal
- Institute of Pathology, Indian Council of Medical Research, New Delhi, India
| |
Collapse
|
43
|
Carey A, Cunningham K, Andrew D, Hafner L, Timms P, Beagley K. A comparison of the effects of a chlamydial vaccine administered during or after a C. muridarum urogenital infection of female mice. Vaccine 2011; 29:6505-13. [PMID: 21767592 DOI: 10.1016/j.vaccine.2011.07.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2010] [Revised: 06/20/2011] [Accepted: 07/05/2011] [Indexed: 10/18/2022]
Abstract
Research into an efficacious Chlamydia trachomatis vaccine is ongoing, however, there has been no examination into the timing of vaccine administration to either asymptomatically or previously infected individuals. Using the female Chlamydia muridarum genital tract mouse model, we examined this aspect of vaccine development. Our results show timing of vaccination affected the production of systemic antibodies, but had minimal effects on mucosal antibody production. Vaccination during an active infection or after a resolved infection did not provide protection against re-exposure to Chlamydia, and did not exacerbate the development of pathological sequelae in infected mice. This demonstrates that vaccination may not be protective in individuals who are seropositive for an acute or previous chlamydial infection.
Collapse
Affiliation(s)
- Alison Carey
- Institute of Health & Biomedical Innovation, Cell & Molecular Biosciences, Faculty of Science, Queensland University of Technology, 60 Musk Ave, Kelvin Grove, QLD 4059, Australia
| | | | | | | | | | | |
Collapse
|
44
|
Wang J, Frohlich KM, Buckner L, Quayle AJ, Luo M, Feng X, Beatty W, Hua Z, Rao X, Lewis ME, Sorrells K, Santiago K, Zhong G, Shen L. Altered protein secretion of Chlamydia trachomatis in persistently infected human endocervical epithelial cells. MICROBIOLOGY-SGM 2011; 157:2759-2771. [PMID: 21737500 DOI: 10.1099/mic.0.044917-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Chlamydia trachomatis is the most common bacterial infection of the human reproductive tract globally; however, the mechanisms underlying the adaptation of the organism to its natural target cells, human endocervical epithelial cells, are not clearly understood. To secure its intracellular niche, C. trachomatis must modulate the host cellular machinery by secreting virulence factors into the host cytosol to facilitate bacterial growth and survival. Here we used primary human endocervical epithelial cells and HeLa cells infected with C. trachomatis to examine the secretion of bacterial proteins during productive growth and persistent growth induced by ampicillin. Specifically, we observed a decrease in secretable chlamydial protease-like activity factor (CPAF) in the cytosol of host epithelial cells exposed to ampicillin with no evident reduction of CPAF product by C. trachomatis. In contrast, the expression of CopN and Tarp was downregulated, suggesting that C. trachomatis responds to ampicillin exposure by selectively altering the expression of secretable proteins. In addition, we observed a greater accumulation of outer-membrane vesicles from C. trachomatis in persistently infected cells. Taken together, these results suggest that the regulation of both gene expression and the secretion of chlamydial virulence proteins is involved in the adaptation of the bacteria to a persistent infection state in human genital epithelial cells.
Collapse
Affiliation(s)
- Jin Wang
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Kyla M Frohlich
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Lyndsey Buckner
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Alison J Quayle
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Miao Luo
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Xiaogeng Feng
- Department of Molecular Biology and Biochemistry, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Wandy Beatty
- Department of Molecular Microbiology, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Ziyu Hua
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Xiancai Rao
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Maria E Lewis
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Kelly Sorrells
- Department of Pathology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Kerri Santiago
- Department of Pathology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Guangming Zhong
- Department of Microbiology and Immunology, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Li Shen
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| |
Collapse
|
45
|
Abstract
As biomedical research becomes increasingly data-intensive, it is increasingly essential to integrate genomic-scale datasets, so as to generate a more holistic picture of complex biological processes. The systems biology paradigm may differ in strategy from traditional reductionist scientific methods, but the goal remains the same: to generate tenable hypotheses driving the experimental elucidation of biological mechanisms. Intracellular pathogens provide an excellent opportunity for systems analysis, as many of these organisms are amenable to genetic manipulation, allowing their biology to be played off against that of the host. Moreover, many of the most fundamental biological properties of these microbes (host cell invasion, immune evasion, intracellular replication, long-term persistence) are directly linked to pathogenesis and readily quantifiable using genomic-scale technologies. In this review, we summarize and discuss some of the available and foreseeable functional genomics datasets pertaining to host-pathogen interactions and suggest that the host-pathogen interface represents a promising, tractable challenge for systems biological analysis. Success will require developing and leveraging new technologies, expanding data acquisition, and increasing public access to comprehensive datasets, to assemble quantitative and testable models of the host-pathogen relationship.
Collapse
Affiliation(s)
- Daniel P Beiting
- Department of Biology, Penn Genome Frontiers Institute, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | |
Collapse
|
46
|
Ibana JA, Schust DJ, Sugimoto J, Nagamatsu T, Greene SJ, Quayle AJ. Chlamydia trachomatis immune evasion via downregulation of MHC class I surface expression involves direct and indirect mechanisms. Infect Dis Obstet Gynecol 2011; 2011:420905. [PMID: 21747639 PMCID: PMC3123996 DOI: 10.1155/2011/420905] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2010] [Accepted: 02/15/2011] [Indexed: 11/10/2022] Open
Abstract
Genital C. trachomatis infections typically last for many months in women. This has been attributed to several strategies by which C. trachomatis evades immune detection, including well-described methods by which C. trachomatis decreases the cell surface expression of the antigen presenting molecules major histocompatibility complex (MHC) class I, MHC class II, and CD1d in infected genital epithelial cells. We have harnessed new methods that allow for separate evaluation of infected and uninfected cells within a mixed population of chlamydia-infected endocervical epithelial cells to demonstrate that MHC class I downregulation in the presence of C. trachomatis is mediated by direct and indirect (soluble) factors. Such indirect mechanisms may aid in priming surrounding cells for more rapid immune evasion upon pathogen entry and help promote unfettered spread of C. trachomatis genital infections.
Collapse
Affiliation(s)
- Joyce A. Ibana
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Science Center, New Orleans, LA 70112, USA
| | - Danny J. Schust
- Department of Obstetrics, Gynecology and Women's Health, University of Missouri School of Medicine, Columbia, MO 65202, USA
| | - Jun Sugimoto
- Department of Obstetrics, Gynecology and Women's Health, University of Missouri School of Medicine, Columbia, MO 65202, USA
| | - Takeshi Nagamatsu
- Department of Obstetrics, Gynecology and Women's Health, University of Missouri School of Medicine, Columbia, MO 65202, USA
| | - Sheila J. Greene
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Science Center, New Orleans, LA 70112, USA
| | - Alison J. Quayle
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Science Center, New Orleans, LA 70112, USA
| |
Collapse
|
47
|
Lei L, Qi M, Budrys N, Schenken R, Zhong G. Localization of Chlamydia trachomatis hypothetical protein CT311 in host cell cytoplasm. Microb Pathog 2011; 51:101-9. [PMID: 21605656 DOI: 10.1016/j.micpath.2011.05.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2011] [Revised: 05/02/2011] [Accepted: 05/04/2011] [Indexed: 11/28/2022]
Abstract
The chlamydia-specific hypothetical protein CT311 was detected both inside and outside of the chlamydial inclusions in Chlamydia trachomatis-infected cells. The extra-inclusion CT311 molecules were distributed in the host cell cytoplasm with a pattern similar to that of CPAF, a known Chlamydia-secreted protease. The detection of CT311 was specific since the anti-CT311 antibody labeling was only removed by absorption with CT311 but not CPAF fusion proteins. In addition, both anti-CT311 and anti-CPAF antibodies only detected their corresponding endogenous proteins without cross-reacting with each other or any other antigens in the whole cell lysates of C. trachomatis-infected cells. Although both CT311 and CPAF proteins were first detected 12 h after infection, localization of CT311 into host cell cytosol was delayed until 24 h while CPAF secretion into host cell cytosol was already obvious by 18 h after infection. The host cell cytosolic localization of CT311 was further confirmed in human primary cells. CT311 was predicted to contain an N-terminal secretion signal sequence and the CT311 signal sequence directed secretion of PhoA into bacterial periplasmic region in a heterologous assay system, suggesting that a sec-dependent pathway may play a role in the secretion of CT311 into host cell cytosol. This hypothesis is further supported by the observation that secretion of CT311 in Chlamydia-infected cells was blocked by a C16 compound known to inhibit signal peptidase I. These findings have provided important molecular information for further understanding the C. trachomatis pathogenic mechanisms.
Collapse
Affiliation(s)
- Lei Lei
- Department of Microbiology and Immunology, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA
| | | | | | | | | |
Collapse
|
48
|
The chlamydial periplasmic stress response serine protease cHtrA is secreted into host cell cytosol. BMC Microbiol 2011; 11:87. [PMID: 21527029 PMCID: PMC3107777 DOI: 10.1186/1471-2180-11-87] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2010] [Accepted: 04/28/2011] [Indexed: 01/05/2023] Open
Abstract
Background The periplasmic High Temperature Requirement protein A (HtrA) plays important roles in bacterial protein folding and stress responses. However, the role of chlamydial HtrA (cHtrA) in chlamydial pathogenesis is not clear. Results The cHtrA was detected both inside and outside the chlamydial inclusions. The detection was specific since both polyclonal and monoclonal anti-cHtrA antibodies revealed similar intracellular labeling patterns that were only removed by absorption with cHtrA but not control fusion proteins. In a Western blot assay, the anti-cHtrA antibodies detected the endogenous cHtrA in Chlamydia-infected cells without cross-reacting with any other chlamydial or host cell antigens. Fractionation of the infected cells revealed cHtrA in the host cell cytosol fraction. The periplasmic cHtrA protein appeared to be actively secreted into host cell cytosol since no other chlamydial periplasmic proteins were detected in the host cell cytoplasm. Most chlamydial species secreted cHtrA into host cell cytosol and the secretion was not inhibitable by a type III secretion inhibitor. Conclusion Since it is hypothesized that chlamydial organisms possess a proteolysis strategy to manipulate host cell signaling pathways, secretion of the serine protease cHtrA into host cell cytosol suggests that the periplasmic cHtrA may also play an important role in chlamydial interactions with host cells.
Collapse
|
49
|
Chlamydia trachomatis secretion of an immunodominant hypothetical protein (CT795) into host cell cytoplasm. J Bacteriol 2011; 193:2498-509. [PMID: 21441519 DOI: 10.1128/jb.01301-10] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The Chlamydia-specific hypothetical protein CT795 was dominantly recognized by human antisera produced during C. trachomatis infection but not by animal antisera raised against dead chlamydia organisms. The immundominant region recognized by the human antibodies was mapped to the N-terminal fragment T22-S69. The endogenous CT795 was detected in the cytoplasm of host cells during C. trachomatis infection and was highly enriched in the host cytosolic fraction but absent in the purified chlamydia organisms, suggesting that CT795 is synthesized and secreted into host cell cytoplasm without incorporation into the organisms. All C. trachomatis serovars tested secreted CT795. A predicted signal peptide of CT795 directed the mature PhoA to cross Escherichia coli inner membranes. The secretion of CT795 in Chlamydia-infected cells was inhibited by a C(16) compound targeting signal peptidase I, but not by a C(1) compound known to block the type III secretion pathway. These results suggest that CT795, like CPAF (a Chlamydia-secreted virulence factor), is secreted into the host cell cytoplasm via a sec-dependent mechanism and not by a type III secretion pathway. The above characterizations of CT795 have provided important information for further understanding the potential roles of CT795 in C. trachomatis pathogenesis.
Collapse
|
50
|
Zhong G. Chlamydia trachomatis secretion of proteases for manipulating host signaling pathways. Front Microbiol 2011; 2:14. [PMID: 21687409 PMCID: PMC3109274 DOI: 10.3389/fmicb.2011.00014] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2011] [Accepted: 01/19/2011] [Indexed: 12/23/2022] Open
Abstract
The human pathogen Chlamydia trachomatis secretes numerous effectors into host cells in order to successfully establish and complete the intracellular growth cycle. Three C. trachomatis proteases [chlamydial proteasome/protease-like activity factor (CPAF), tail-specific protease (Tsp), and chlamydial high temperature requirement protein A (cHtrA)] have been localized in the cytosol of the infected cells either by direct immunofluorescence visualization or functional implication. Both CPAF and Tsp have been found to play important roles in C. trachomatis interactions with host cells although the cellular targets of cHtrA have not been identified. All three proteases contain a putative N-terminal signal sequence, suggesting that they may be secreted via a sec-dependent pathway. However, these proteases are also found in chlamydial organism-free vesicles in the lumen of the chlamydial inclusions before they are secreted into host cell cytosol, suggesting that these proteases may first be translocated into the periplasmic region via a sec-dependent pathway and then exported outside of the organisms via an outer membrane vesicles (OMVs) budding mechanism. The vesiculized proteases in the inclusion lumen can finally enter host cell cytosol via vesicle fusing with or passing through the inclusion membrane. Continuing identification and characterization of the C. trachomatis-secreted proteins (CtSPs) will not only promote our understanding of C. trachomatis pathogenic mechanisms but also allow us to gain novel insights into the OMV pathway, a well-known mechanism used by bacteria to export virulence factors although its mechanism remains elusive.
Collapse
Affiliation(s)
- Guangming Zhong
- Department of Microbiology and Immunology, University of Texas Health Science Center at San Antonio San Antonio, TX, USA
| |
Collapse
|