1
|
Medhavi F, Tanner T, Richardson S, Lundy S, Omosun Y, Eko FO. A VCG-Based Multiepitope Chlamydia Vaccine Incorporating the Cholera Toxin A1 Subunit (MECA) Confers Protective Immunity Against Transcervical Challenge. Biomedicines 2025; 13:288. [PMID: 40002702 PMCID: PMC11852492 DOI: 10.3390/biomedicines13020288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Revised: 01/19/2025] [Accepted: 01/22/2025] [Indexed: 02/27/2025] Open
Abstract
Background/Objectives: We generated a novel recombinant Vibrio cholerae ghost (rVCG)-based subunit vaccine incorporating the A1 subunit of cholera toxin (CTA1) and a multiepitope Chlamydia trachomatis (CT) antigen (MECA) derived from five chlamydial outer membrane proteins (rVCG-MECA). The ability of this vaccine to protect against a CT transcervical challenge was evaluated. Methods: Female C57BL/6J mice were immunized thrice at two-week intervals with rVCG-MECA or rVCG-gD2 (antigen control) via the intramuscular (IM) or intranasal (IN) route. PBS-immunized mice or mice immunized with live CT served as negative and positive controls, respectively. Results: Vaccine delivery stimulated robust humoral and cell-mediated immune effectors, characterized by local mucosal and systemic CT-specific IgG, IgG2c, and IgA antibody and IFN-γ (Th1 cytokine) responses. The elicited mucosal and systemic IgG2c and IgA antibody responses persisted for 16 weeks post-immunization. Immunization with rVCG-MECA afforded protection comparable to that provided by IN immunization with live CT EBs without any side effects, irrespective of route of vaccine delivery. Conclusions: The results underline the potential of a multiepitope vaccine as a promising resource for protecting against CT genital infection and the potential of CTA1 on the VCG platform as a mucosal and systemic adjuvant for developing CT vaccines.
Collapse
Affiliation(s)
| | | | | | | | | | - Francis O. Eko
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA 30310, USA; (F.M.); (Y.O.)
| |
Collapse
|
2
|
Sahu R, Verma R, Egbo TE, Giambartolomei GH, Singh SR, Dennis VA. Effects of prime-boost strategies on the protective efficacy and immunogenicity of a PLGA (85:15)-encapsulated Chlamydia recombinant MOMP nanovaccine. Pathog Dis 2024; 82:ftae004. [PMID: 38862192 PMCID: PMC11186516 DOI: 10.1093/femspd/ftae004] [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: 10/30/2023] [Revised: 01/08/2024] [Accepted: 06/10/2024] [Indexed: 06/13/2024] Open
Abstract
To begin to optimize the immunization routes for our reported PLGA-rMOMP nanovaccine [PLGA-encapsulated Chlamydia muridarum (Cm) recombinant major outer membrane protein (rMOMP)], we compared two prime-boost immunization strategies [subcutaneous (SC) and intramuscular (IM-p) prime routes followed by two SC-boosts)] to evaluate the nanovaccine-induced protective efficacy and immunogenicity in female BALB/c mice. Our results showed that mice immunized via the SC and IM-p routes were protected against a Cm genital challenge by a reduction in bacterial burden and with fewer bacteria in the SC mice. Protection of mice correlated with rMOMP-specific Th1 (IL-2 and IFN-γ) and not Th2 (IL-4, IL-9, and IL-13) cytokines, and CD4+ memory (CD44highCD62Lhigh) T-cells, especially in the SC mice. We also observed higher levels of IL-1α, IL-6, IL-17, CCL-2, and G-CSF in SC-immunized mice. Notably, an increase of cytokines/chemokines was seen after the challenge in the SC, IM-p, and control mice (rMOMP and PBS), suggesting a Cm stimulation. In parallel, rMOMP-specific Th1 (IgG2a and IgG2b) and Th2 (IgG1) serum, mucosal, serum avidity, and neutralizing antibodies were more elevated in SC than in IM-p mice. Overall, the homologous SC prime-boost immunization of mice induced enhanced cellular and antibody responses with better protection against a genital challenge compared to the heterologous IM-p.
Collapse
Affiliation(s)
- Rajnish Sahu
- Center for NanoBiotechnology Research, Department of Biological Sciences, 1627 Harris Way, Alabama State University, Montgomery AL, 36104, United States
| | - Richa Verma
- Center for NanoBiotechnology Research, Department of Biological Sciences, 1627 Harris Way, Alabama State University, Montgomery AL, 36104, United States
| | - Timothy E Egbo
- US Army Medical Research Institute of Infectious Diseases, Unit 8900, DPO, AE, Box 330, 09831, United States
| | - Guillermo H Giambartolomei
- Instituto de Inmunología, Genética y Metabolismo (INIGEM). CONICET. AV. Cordoba 2351, Universidad de Buenos Aires, Buenos Aires, C1120AAR, Argentina
| | - Shree R Singh
- Center for NanoBiotechnology Research, Department of Biological Sciences, 1627 Harris Way, Alabama State University, Montgomery AL, 36104, United States
| | - Vida A Dennis
- Center for NanoBiotechnology Research, Department of Biological Sciences, 1627 Harris Way, Alabama State University, Montgomery AL, 36104, United States
| |
Collapse
|
3
|
Arasu Y, Bryan E, Russell FA, Huettner N, Carey AJ, Boyd BJ, Beagley KW, Dargaville TR. Enhanced clearance of C. muridarum infection using azithromycin-loaded liposomes. Int J Pharm 2024; 650:123709. [PMID: 38101758 DOI: 10.1016/j.ijpharm.2023.123709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 12/11/2023] [Accepted: 12/12/2023] [Indexed: 12/17/2023]
Abstract
Chlamydia trachomatis is an intracellular bacterium which infects around 129 million people annually. Despite similar infection rates between sexes, most research investigating the effects of chlamydial infection on fertility has focused on females. There is now emerging evidence of a potential link between Chlamydia and impaired male fertility. The only treatments for chlamydial infection are antibiotics, with azithromycin (AZI) being one of the commonly used drugs. However, recent studies have suggested that optimizing the treatment regime is necessary, as higher concentrations of AZI may be required to effectively clear the infection in certain cell types, particularly testicular macrophages. To address this challenge, we have prepared liposomes consisting of 1,2-dioleoyl-sn-glycero-3-phosphocholine (DOPC) and D-α-tocopheryl polyethylene glycol 1000 succinate (TPGS) loaded with AZI for clearing Chlamydia. These liposomes exhibited stability over time and were readily taken up by both macrophages and epithelial cells. Moreover, they demonstrated significant enhancement of chlamydial clearance in both cell types. In a mouse model, the drug-loaded liposomes cleared Chlamydia within the penile urethra more efficiently than the same dose of unencapsulated drug. Furthermore, the liposome-drug treatment showed significant protective effects on sperm motility and morphology, suggesting potential benefits in reducing sperm damage caused by the infection.
Collapse
Affiliation(s)
- Yanushia Arasu
- School of Chemistry and Physics and Centre for Materials Science, Faculty of Science, Queensland University of Technology, Brisbane, Australia; School of Biomedical Sciences and Centre for Immunology and Infection Control, Queensland University of Technology, Brisbane, Australia
| | - Emily Bryan
- School of Biomedical Sciences and Centre for Immunology and Infection Control, Queensland University of Technology, Brisbane, Australia
| | - Freya A Russell
- School of Biomedical Sciences and Centre for Immunology and Infection Control, Queensland University of Technology, Brisbane, Australia
| | - Nick Huettner
- School of Chemistry and Physics and Centre for Materials Science, Faculty of Science, Queensland University of Technology, Brisbane, Australia
| | - Alison J Carey
- School of Biomedical Sciences and Centre for Immunology and Infection Control, Queensland University of Technology, Brisbane, Australia
| | - Ben J Boyd
- Drug Delivery, Disposition & Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Clayton, Australia; Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Kenneth W Beagley
- School of Biomedical Sciences and Centre for Immunology and Infection Control, Queensland University of Technology, Brisbane, Australia
| | - Tim R Dargaville
- School of Chemistry and Physics and Centre for Materials Science, Faculty of Science, Queensland University of Technology, Brisbane, Australia.
| |
Collapse
|
4
|
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
|
5
|
Guo X, Pan X, Sun Q, Hu Y, Shi J. Design of a novel multiepitope vaccine against Chlamydia pneumoniae using the extracellular protein as a target. Sci Rep 2023; 13:15070. [PMID: 37700027 PMCID: PMC10497608 DOI: 10.1038/s41598-023-42222-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 09/06/2023] [Indexed: 09/14/2023] Open
Abstract
Chlamydia pneumoniae (C. pneumoniae) infection in humans is universal and causes various respiratory infectious diseases, making a safe and effective preventive vaccine essential. In this study, a multi-epitope vaccine with CTLA-4 extracellular structure was constructed by an immunoinformatics approach. Since MOMP protein is the major extracellular protein in C. pneumoniae and has good immunogenicity and high conservation, we selected the MOMP protein of C. pneumoniae as the antigen target, predicted the T and B cell epitopes of the MOMP protein and then connected the CTLA-4 extracellular structure with the predicted dominant epitopes by various linkers to construct a multi-epitope vaccine. The biochemical characterization of the multi-epitope vaccine showed its immunogenicity and anti-allergic properties. The tertiary structure of this vaccine, along with molecular docking, molecular dynamics simulation, and principal component analysis, showed that the multi-epitope vaccine structure interacted with B7 (B7-1, B7-2) and toll-like receptors (TLR-2, TLR-4). Ultimately, the vaccine was cloned and effectively expressed in silico on an insect baculovirus expression vector (pFastBac1). These analyses showed that the designed vaccine could potentially target antigen-presenting cells and was immune to C. pneumoniae, which provided novel strategies for developing the vaccine.
Collapse
Affiliation(s)
- Xiaomei Guo
- Yunnan Provincial Key Laboratory of Vector-Borne Diseases Control and Research, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, 935 Jiaoling Road, Kunming, 650118, Yunnan, China
- Kunming Medical University, Kunming, Yunnan, China
| | - Xiaohong Pan
- Yunnan Provincial Key Laboratory of Vector-Borne Diseases Control and Research, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, 935 Jiaoling Road, Kunming, 650118, Yunnan, China
| | - Qiangming Sun
- Yunnan Provincial Key Laboratory of Vector-Borne Diseases Control and Research, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, 935 Jiaoling Road, Kunming, 650118, Yunnan, China.
- National Kunming High-Level Biosafety Primate Research Center, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, Yunnan, China.
| | - Yunzhang Hu
- Yunnan Provincial Key Laboratory of Vector-Borne Diseases Control and Research, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, 935 Jiaoling Road, Kunming, 650118, Yunnan, China.
- Kunming Medical University, Kunming, Yunnan, China.
| | - Jiandong Shi
- Yunnan Provincial Key Laboratory of Vector-Borne Diseases Control and Research, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, 935 Jiaoling Road, Kunming, 650118, Yunnan, China.
- National Kunming High-Level Biosafety Primate Research Center, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, Yunnan, China.
| |
Collapse
|
6
|
Pal S, Slepenkin A, Felgner J, Huw Davies D, Felgner P, de la Maza LM. Evaluation of Four Adjuvant Combinations, IVAX-1, IVAX-2, CpG-1826+Montanide ISA 720 VG and CpG-1018+Montanide ISA 720 VG, for Safety and for Their Ability to Elicit Protective Immune Responses in Mice against a Respiratory Challenge with Chlamydia muridarum. Pathogens 2023; 12:863. [PMID: 37513710 PMCID: PMC10383793 DOI: 10.3390/pathogens12070863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 05/17/2023] [Accepted: 06/14/2023] [Indexed: 07/30/2023] Open
Abstract
There is an urgent need to produce a vaccine for Chlamydia trachomatis infections. Here, using the Chlamydia muridarum major outer membrane protein (MOMP) as an antigen, four adjuvant combinations IVAX-1 (MPLA+CpG-1018+AddaVax), IVAX-2 (MPLA+CpG-1018+AS03), CpG-1826+Montanide ISA 720 VG (CpG-1826+Mont) and CpG-1018+Montanide ISA 720 VG (CpG-1018+Mont), were tested for their local reactogenicity and ability to elicit protection in BALB/c mice against a respiratory challenge with C. muridarum. Immunization with IVAX-1 or IVAX-2 induced no significant local reactogenicity following intramuscular immunization. In contrast, vaccines containing Montanide resulted in the formation of a local granuloma. Based on the IgG2a/IgG1 ratio in serum, the four adjuvant combinations elicited Th1-biased responses. IVAX-1 induced the highest in vitro neutralization titers while CpG-1018+Mont stimulated the lowest. As determined by the levels of IFN-γ produced by T-cells, the most robust cellular immune responses were elicited in mice immunized with CpG-1018+Mont, while the weakest responses were mounted by mice receiving IVAX-1. Following the respiratory challenge, mice immunized with CpG-1018+Mont lost the least amount of body weight and had the lowest number of C. muridarum inclusion-forming units (IFUs) in the lungs, while those receiving IVAX-2 had lost the most weight and had the highest number of IFUs in their lungs. Animals vaccinated with CpG-1826+Mont had the lightest lungs while those immunized using IVAX-2 had the heaviest. To conclude, due to their safety and adjuvanticity, IVAX formulations should be considered for inclusion in human vaccines against Chlamydia.
Collapse
Affiliation(s)
- Sukumar Pal
- Department of Pathology and Laboratory Medicine, University of California, Irvine, CA 92697, USA
| | - Anatoli Slepenkin
- Department of Pathology and Laboratory Medicine, University of California, Irvine, CA 92697, USA
| | - Jiin Felgner
- Vaccine Research and Development Center, Department of Physiology and Biophysics, University of California, Irvine, CA 92697, USA
| | - D Huw Davies
- Vaccine Research and Development Center, Department of Physiology and Biophysics, University of California, Irvine, CA 92697, USA
| | - Philip Felgner
- Vaccine Research and Development Center, Department of Physiology and Biophysics, University of California, Irvine, CA 92697, USA
| | - Luis M de la Maza
- Department of Pathology and Laboratory Medicine, University of California, Irvine, CA 92697, USA
| |
Collapse
|
7
|
Bryan ER, Redgrove KA, Mooney AR, Mihalas BP, Sutherland JM, Carey AJ, Armitage CW, Trim LK, Kollipara A, Mulvey PBM, Palframan E, Trollope G, Bogoevski K, McLachlan R, McLaughlin EA, Beagley KW. Chronic testicular Chlamydia muridarum infection impairs mouse fertility and offspring development†. Biol Reprod 2021; 102:888-901. [PMID: 31965142 PMCID: PMC7124966 DOI: 10.1093/biolre/ioz229] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2019] [Revised: 11/28/2019] [Accepted: 01/12/2020] [Indexed: 12/26/2022] Open
Abstract
With approximately 131 million new genital tract infections occurring each year, Chlamydia is the most common sexually transmitted bacterial pathogen worldwide. Male and female infections occur at similar rates and both cause serious pathological sequelae. Despite this, the impact of chlamydial infection on male fertility has long been debated, and the effects of paternal chlamydial infection on offspring development are unknown. Using a male mouse chronic infection model, we show that chlamydial infection persists in the testes, adversely affecting the testicular environment. Infection increased leukocyte infiltration, disrupted the blood:testis barrier and reduced spermiogenic cell numbers and seminiferous tubule volume. Sperm from infected mice had decreased motility, increased abnormal morphology, decreased zona-binding capacity, and increased DNA damage. Serum anti-sperm antibodies were also increased. When both acutely and chronically infected male mice were bred with healthy female mice, 16.7% of pups displayed developmental abnormalities. Female offspring of chronically infected sires had smaller reproductive tracts than offspring of noninfected sires. The male pups of infected sires displayed delayed testicular development, with abnormalities in sperm vitality, motility, and sperm-oocyte binding evident at sexual maturity. These data suggest that chronic testicular Chlamydia infection can contribute to male infertility, which may have an intergenerational impact on sperm quality.
Collapse
Affiliation(s)
- Emily R Bryan
- School of Biomedical Sciences and Institute of Health & Biomedical Innovation, Queensland University of Technology, Herston, Queensland, Australia
| | - Kate A Redgrove
- School of Environmental and Life Sciences, Faculty of Science, The University of Newcastle, University Drive, Callaghan, New South Wales, Australia
| | - Alison R Mooney
- School of Biomedical Sciences and Institute of Health & Biomedical Innovation, Queensland University of Technology, Herston, Queensland, Australia
| | - Bettina P Mihalas
- School of Environmental and Life Sciences, Faculty of Science, The University of Newcastle, University Drive, Callaghan, New South Wales, Australia
| | - Jessie M Sutherland
- School of Environmental and Life Sciences, Faculty of Science, The University of Newcastle, University Drive, Callaghan, New South Wales, Australia
| | - Alison J Carey
- School of Biomedical Sciences and Institute of Health & Biomedical Innovation, Queensland University of Technology, Herston, Queensland, Australia
| | - Charles W Armitage
- School of Biomedical Sciences and Institute of Health & Biomedical Innovation, Queensland University of Technology, Herston, Queensland, Australia.,Peter Goher Department of Immunobiology, King's College London, London, United Kingdom
| | - Logan K Trim
- School of Biomedical Sciences and Institute of Health & Biomedical Innovation, Queensland University of Technology, Herston, Queensland, Australia
| | - Avinash Kollipara
- School of Biomedical Sciences and Institute of Health & Biomedical Innovation, Queensland University of Technology, Herston, Queensland, Australia
| | - Peter B M Mulvey
- School of Biomedical Sciences and Institute of Health & Biomedical Innovation, Queensland University of Technology, Herston, Queensland, Australia
| | - Ella Palframan
- School of Biomedical Sciences and Institute of Health & Biomedical Innovation, Queensland University of Technology, Herston, Queensland, Australia
| | - Gemma Trollope
- School of Biomedical Sciences and Institute of Health & Biomedical Innovation, Queensland University of Technology, Herston, Queensland, Australia
| | - Kristofor Bogoevski
- Scientific Services, Histology Services, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Robert McLachlan
- Department of Obstetrics and Gynaecology, Hudson Institute of Medical Research, Monash Medical Centre, Monash University, Clayton, Victoria, Australia
| | - Eileen A McLaughlin
- School of Environmental and Life Sciences, Faculty of Science, The University of Newcastle, University Drive, Callaghan, New South Wales, Australia.,School of Science, Western Sydney University, Richmond, New South Wales, Australia.,School of Life Sciences, The University of Auckland, Auckland, New Zealand
| | - Kenneth W Beagley
- School of Biomedical Sciences and Institute of Health & Biomedical Innovation, Queensland University of Technology, Herston, Queensland, Australia
| |
Collapse
|
8
|
Nazareth L, Walkden H, Chacko A, Delbaz A, Shelper T, Armitage CW, Reshamwala R, Trim LK, St John JA, Beagley KW, Ekberg JAK. Chlamydia muridarum Can Invade the Central Nervous System via the Olfactory and Trigeminal Nerves and Infect Peripheral Nerve Glial Cells. Front Cell Infect Microbiol 2021; 10:607779. [PMID: 33489937 PMCID: PMC7819965 DOI: 10.3389/fcimb.2020.607779] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 11/23/2020] [Indexed: 12/11/2022] Open
Abstract
Chlamydia pneumoniae can infect the brain and has been linked to late-onset dementia. Chlamydia muridarum, which infects mice, is often used to model human chlamydial infections. While it has been suggested to be also important for modelling brain infection, nervous system infection by C. muridarum has not been reported in the literature. C. pneumoniae has been shown to infect the olfactory bulb in mice after intranasal inoculation, and has therefore been suggested to invade the brain via the olfactory nerve; however, nerve infection has not been shown to date. Another path by which certain bacteria can reach the brain is via the trigeminal nerve, but it remains unknown whether Chlamydia species can infect this nerve. Other bacteria that can invade the brain via the olfactory and/or trigeminal nerve can do so rapidly, however, whether Chlamydia spp. can reach the brain earlier than one-week post inoculation remains unknown. In the current study, we showed that C. muridarum can within 48 h invade the brain via the olfactory nerve, in addition to infecting the trigeminal nerve. We also cultured the glial cells of the olfactory and trigeminal nerves and showed that C. muridarum readily infected the cells, constituting a possible cellular mechanism explaining how the bacteria can invade the nerves without being eliminated by glial immune functions. Further, we demonstrated that olfactory and trigeminal glia differed in their responses to C. muridarum, with olfactory glia showing less infection and stronger immune response than trigeminal glia.
Collapse
Affiliation(s)
- Lynn Nazareth
- Menzies Health Institute Queensland, Griffith University, Southport, QLD, Australia.,Clem Jones Centre for Neurobiology and Stem Cell Research, Griffith University, Nathan, QLD, Australia
| | - Heidi Walkden
- Menzies Health Institute Queensland, Griffith University, Southport, QLD, Australia.,Clem Jones Centre for Neurobiology and Stem Cell Research, Griffith University, Nathan, QLD, Australia
| | - Anu Chacko
- Menzies Health Institute Queensland, Griffith University, Southport, QLD, Australia.,Clem Jones Centre for Neurobiology and Stem Cell Research, Griffith University, Nathan, QLD, Australia
| | - Ali Delbaz
- Menzies Health Institute Queensland, Griffith University, Southport, QLD, Australia.,Clem Jones Centre for Neurobiology and Stem Cell Research, Griffith University, Nathan, QLD, Australia
| | - Todd Shelper
- Menzies Health Institute Queensland, Griffith University, Southport, QLD, Australia.,Clem Jones Centre for Neurobiology and Stem Cell Research, Griffith University, Nathan, QLD, Australia
| | - Charles W Armitage
- Centre for Immunology and Infection Control, School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, Australia
| | - Ronak Reshamwala
- Menzies Health Institute Queensland, Griffith University, Southport, QLD, Australia.,Clem Jones Centre for Neurobiology and Stem Cell Research, Griffith University, Nathan, QLD, Australia
| | - Logan K Trim
- Centre for Immunology and Infection Control, School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, Australia
| | - James A St John
- Menzies Health Institute Queensland, Griffith University, Southport, QLD, Australia.,Clem Jones Centre for Neurobiology and Stem Cell Research, Griffith University, Nathan, QLD, Australia.,Griffith Institute for Drug Discovery, Griffith University, Nathan, QLD, Australia
| | - Kenneth W Beagley
- Centre for Immunology and Infection Control, School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, Australia
| | - Jenny A K Ekberg
- Menzies Health Institute Queensland, Griffith University, Southport, QLD, Australia.,Clem Jones Centre for Neurobiology and Stem Cell Research, Griffith University, Nathan, QLD, Australia.,Griffith Institute for Drug Discovery, Griffith University, Nathan, QLD, Australia
| |
Collapse
|
9
|
Chlamydia-Specific IgA Secretion in the Female Reproductive Tract Induced via Per-Oral Immunization Confers Protection against Primary Chlamydia Challenge. Infect Immun 2020; 89:IAI.00413-20. [PMID: 33139380 DOI: 10.1128/iai.00413-20] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Accepted: 10/20/2020] [Indexed: 12/12/2022] Open
Abstract
Chlamydia trachomatis is an obligate intracellular pathogen that causes sexually transmitted disease. In women, chlamydial infections may cause pelvic inflammatory disease (PID), ectopic pregnancy, and infertility. The role of antibodies in protection against a primary Chlamydia infection is unclear and was a focus of this work. Using the C. muridarum mouse infection model, we show that intestinal mucosa is infected via intranasal (i.n.) or per-oral (p.o.) Chlamydia inoculation and that unlike the female reproductive tract (FRT) mucosa, it halts systemic Chlamydia dissemination. Moreover, p.o. immunization or infection with Chlamydia confers protection against per-vaginal (p.v.) challenge, resulting in significantly decreased bacterial burden in the FRT, accelerated Chlamydia clearance, and reduced hydrosalpinx pathology. In contrast, subcutaneous (s.c.) immunization conferred no protection against the p.v. challenge. Both p.o. and s.c. immunizations induced Chlamydia-specific serum IgA. However, IgA was found only in the vaginal washes and fecal extracts of p.o.-immunized animals. Following a p.v. challenge, unimmunized control and s.c.-s.c.-immunized animals developed Chlamydia-specific intestinal IgA yet failed to develop IgA in the FRT, indicating that IgA response in the FRT relies on the FRT to gastrointestinal tract (GIT) antigen transport. Vaginal secretions of p.o.-immunized animals neutralize Chlamydia in vivo, resulting in significantly lower Chlamydia burden in the FRT and Chlamydia transport to the GIT. We also show that infection of the GIT is not necessary for induction of protective immunity in the FRT, a finding that is important for the development of p.o. subunit vaccines to target Chlamydia and possibly other sexually transmitted pathogens.
Collapse
|
10
|
Bryan ER, Kollipara A, Trim LK, Armitage CW, Carey AJ, Mihalas B, Redgrove KA, McLaughlin EA, Beagley KW. Hematogenous dissemination of Chlamydia muridarum from the urethra in macrophages causes testicular infection and sperm DNA damage†. Biol Reprod 2020; 101:748-759. [PMID: 31373361 DOI: 10.1093/biolre/ioz146] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 05/27/2019] [Accepted: 07/25/2019] [Indexed: 12/17/2022] Open
Abstract
The incidence of Chlamydia infection, in both females and males, is increasing worldwide. Male infections have been associated clinically with urethritis, epididymitis, and orchitis, believed to be caused by ascending infection, although the impact of infection on male fertility remains controversial. Using a mouse model of male chlamydial infection, we show that all the major testicular cell populations, germ cells, Sertoli cells, Leydig cells, and testicular macrophages can be productively infected. Furthermore, sperm isolated from vas deferens of infected mice also had increased levels of DNA damage as early as 4 weeks post-infection. Bilateral vasectomy, prior to infection, did not affect the chlamydial load recovered from testes at 2, 4, and 8 weeks post-infection, and Chlamydia-infected macrophages were detectable in blood and the testes as soon as 3 days post-infection. Partial depletion of macrophages with clodronate liposomes significantly reduced the testicular chlamydial burden, consistent with a hematogenous route of infection, with Chlamydia transported to the testes in infected macrophages. These data suggest that macrophages serve as Trojan horses, transporting Chlamydia from the penile urethra to the testes within 3 days of infection, bypassing the entire male reproductive tract. In the testes, infected macrophages likely transfer infection to Leydig, Sertoli, and germ cells, causing sperm DNA damage and impaired spermatogenesis.
Collapse
Affiliation(s)
- Emily R Bryan
- School of Biomedical Sciences and Institute of Health & Biomedical Innovation, Queensland University of Technology, Herston, QLD, Australia
| | - Avinash Kollipara
- School of Biomedical Sciences and Institute of Health & Biomedical Innovation, Queensland University of Technology, Herston, QLD, Australia
| | - Logan K Trim
- School of Biomedical Sciences and Institute of Health & Biomedical Innovation, Queensland University of Technology, Herston, QLD, Australia
| | - Charles W Armitage
- School of Biomedical Sciences and Institute of Health & Biomedical Innovation, Queensland University of Technology, Herston, QLD, Australia
| | - Alison J Carey
- School of Biomedical Sciences and Institute of Health & Biomedical Innovation, Queensland University of Technology, Herston, QLD, Australia
| | - Bettina Mihalas
- School of Environmental and Life Sciences, Faculty of Science, The University of Newcastle, Callaghan, NSW, Australia
| | - Kate A Redgrove
- School of Environmental and Life Sciences, Faculty of Science, The University of Newcastle, Callaghan, NSW, Australia
| | - Eileen A McLaughlin
- School of Environmental and Life Sciences, Faculty of Science, The University of Newcastle, Callaghan, NSW, Australia.,Science and Technology Office, University of Canberra, Bruce, ACT, Australia
| | - Kenneth W Beagley
- School of Biomedical Sciences and Institute of Health & Biomedical Innovation, Queensland University of Technology, Herston, QLD, Australia
| |
Collapse
|
11
|
Tang Y, Guo F, Lei A, Xiang J, Liu P, Ten W, Dai G, Li R. GrpE Immunization Protects Against Ureaplasma urealyticum Infection in BALB/C Mice. Front Immunol 2020; 11:1495. [PMID: 32849509 PMCID: PMC7411329 DOI: 10.3389/fimmu.2020.01495] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 06/08/2020] [Indexed: 12/28/2022] Open
Abstract
Nucleotide exchange factor (GrpE), a highly conserved antigen, is rapidly expressed and upregulated when Ureaplasma urealyticum infects a host, which could act as a candidative vaccine if it can induce an anti-U. urealyticum immune reaction. Here, we evaluated the vaccine potential of recombinant GrpE protein adjuvanted by Freund's adjuvant (FA), to protect against U. urealyticum genital tract infection in a mouse model. After booster immunization in mice with FA, the GrpE can induced both humoral and cellular immune response after intramuscular injection into BALB/c mice. A strong humoral immune response was detected in the GrpE-immunized mice characterized by production of high titers of antigen-specific serum IgG (IgG1, IgG2a, and IgG3) antibodies. At the same time, the GrpE also induced a Th1-biased cytokine spectrum with high levels of IFN-γ and TNF-α after re-stimulation with immunogen GrpE in vitro, suggesting that GrpE could trigger the Th1 response when used for vaccination in the presence of FA. Although GrpE vaccination in the presence of a Th1-type adjuvant-induced had readily detectable Th1 responses, there wasn't increase inflammation in response to the infection. More importantly, the robust immune responses in mice after immunization with GrpE showed a significantly reduced U. urealyticum burden in cervical tissues. Histopathological analysis confirmed that tissues of GrpE-immunized BALB/c mice were protected against the pathological effects of U. urealyticum infection. In conclusion, this study preliminarily reveals GrpE protein as a promising new candidate vaccine for preventing U. urealyticum reproductive tract infection.
Collapse
Affiliation(s)
- Yanhong Tang
- Chenzhou Hospital Affiliated to University of South China, Hunan, China.,Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Institute of Pathogenic Biology, Pathogenic Biology Institute, Medical College, University of South China, Hunan, China.,The First People's Hospital of Chenzhou, Hunan, China
| | - Fangyi Guo
- Chenzhou Hospital Affiliated to University of South China, Hunan, China.,Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Institute of Pathogenic Biology, Pathogenic Biology Institute, Medical College, University of South China, Hunan, China.,The First People's Hospital of Chenzhou, Hunan, China
| | - Aihua Lei
- Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Institute of Pathogenic Biology, Pathogenic Biology Institute, Medical College, University of South China, Hunan, China
| | - Jing Xiang
- The First People's Hospital of Chenzhou, Hunan, China
| | - Pengqin Liu
- The First People's Hospital of Huaihua, Hunan, China
| | - Wenyou Ten
- The First People's Hospital of Chenzhou, Hunan, China
| | - Guozhi Dai
- Chenzhou Hospital Affiliated to University of South China, Hunan, China.,Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Institute of Pathogenic Biology, Pathogenic Biology Institute, Medical College, University of South China, Hunan, China.,The First People's Hospital of Chenzhou, Hunan, China
| | - Ranhui Li
- Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Institute of Pathogenic Biology, Pathogenic Biology Institute, Medical College, University of South China, Hunan, China
| |
Collapse
|
12
|
Phillips S, Quigley BL, Timms P. Seventy Years of Chlamydia Vaccine Research - Limitations of the Past and Directions for the Future. Front Microbiol 2019; 10:70. [PMID: 30766521 PMCID: PMC6365973 DOI: 10.3389/fmicb.2019.00070] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 01/15/2019] [Indexed: 11/30/2022] Open
Abstract
Chlamydia is a major bacterial pathogen that infects humans, as well as a wide range of animals, including marsupials, birds, cats, pigs, cattle, and sheep. Antibiotics are the only treatment currently available, however, with high rates of re-infection, there is mounting pressure to develop Chlamydia vaccines. In this review, we analyzed how Chlamydia vaccine trials have developed over the past 70 years and identified where future trials need to be focused. There has been a strong bias toward studies targeting C. muridarum and C. trachomatis within mice and a lack of studies matching chlamydial species to their end target host. Even though a large number of specific antigenic targets have been studied, the results from whole-cell vaccine targets show slightly more promising results overall. There has also been a strong bias toward systemic vaccine delivery systems, despite the finding that mucosal delivery systems have shown more promising outcomes. However, the only successful vaccines with matched chlamydial species/infecting host are based on systemic vaccine delivery methods. We highlight the extensive work done with mouse model trials and indicate that whole cell antigenic targets are capable of inducing an effective response, protecting from disease and reducing shedding rates. However, replication of these results using antigen preparations more conducive to commercial vaccine production has proven difficult. To date, the Major Outer Membrane Protein (MOMP) has emerged as the most suitable substitute for whole cell targets and its delivery as a combined systemic and mucosal vaccine is most effective. Finally, although mouse model trials are useful, differences between hosts and infecting chlamydial strains are preventing vaccine formulations from mouse models to be translated into larger animals or intended hosts.
Collapse
Affiliation(s)
- Samuel Phillips
- Genecology Research Centre, The University of the Sunshine Coast, Maroochydore, QLD, Australia
| | - Bonnie L Quigley
- Genecology Research Centre, The University of the Sunshine Coast, Maroochydore, QLD, Australia
| | - Peter Timms
- Genecology Research Centre, The University of the Sunshine Coast, Maroochydore, QLD, Australia
| |
Collapse
|
13
|
A recombinant multi-epitope peptide vaccine based on MOMP and CPSIT_p6 protein protects against Chlamydia psittaci lung infection. Appl Microbiol Biotechnol 2018; 103:941-952. [DOI: 10.1007/s00253-018-9513-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 11/05/2018] [Accepted: 11/09/2018] [Indexed: 12/27/2022]
|
14
|
Verma R, Sahu R, Dixit S, Duncan SA, Giambartolomei GH, Singh SR, Dennis VA. The Chlamydia M278 Major Outer Membrane Peptide Encapsulated in the Poly(lactic acid)-Poly(ethylene glycol) Nanoparticulate Self-Adjuvanting Delivery System Protects Mice Against a Chlamydia muridarum Genital Tract Challenge by Stimulating Robust Systemic and Local Mucosal Immune Responses. Front Immunol 2018; 9:2369. [PMID: 30374357 PMCID: PMC6196261 DOI: 10.3389/fimmu.2018.02369] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Accepted: 09/24/2018] [Indexed: 12/26/2022] Open
Abstract
Recently, we reported that our PPM chlamydial nanovaccine [a biodegradable co-polymeric PLA-PEG (poly(lactic acid)-poly(ethylene glycol))-encapsulated M278 peptide (derived from the major outer membrane protein (MOMP) of Chlamydia)] exploits the caveolin-mediated endocytosis pathway for endosomal processing and MHC class II presentation to immune-potentiate Chlamydia-specific CD4+ T-cell immune effector responses. In the present study, we employed the Chlamydia muridarum mouse infection model to evaluate the protective efficacy of PPM against a genital tract challenge. Our results show that mice immunized with PPM were significantly protected against a homologous genital tract challenge evidently by reduced vaginal bacterial loads. Protection of mice correlated with enhanced Chlamydia-specific adaptive immune responses predominated by IFN-γ along with CD4+ T-cells proliferation and their differentiation to CD4+ memory (CD44high CD62Lhigh) and effector (CD44high CD62Llow) T-cell phenotypes. We observed the elevation of M278- and MOMP-specific serum antibodies with high avidity in the ascending order IgG1 > IgG2b > IgG2a. A key finding was the elevated mucosal IgG1 and IgA antibody titers followed by an increase in MOMP-specific IgA after the challenge. The Th1/Th2 antibody titer ratios (IgG2a/IgG1 and IgG2b/IgG1) revealed that PPM evoked a Th2-directed response, which skewed to a Th1-dominated antibody response after the bacterial challenge of mice. In addition, PPM immune sera neutralized the infectivity of C. muridarum in McCoy cells, suggesting the triggering of functional neutralizing antibodies. Herein, we reveal for the first time that subcutaneous immunization with the self-adjuvanting biodegradable co-polymeric PPM nanovaccine immune-potentiated robust CD4+ T cell-mediated immune effector responses; a mixed Th1 and Th2 antibody response and local mucosal IgA to protect mice against a chlamydial genital tract challenge.
Collapse
Affiliation(s)
- Richa Verma
- Center for NanoBiotechnology Research, Alabama State University, Montgomery, AL, United States
| | - Rajnish Sahu
- Center for NanoBiotechnology Research, Alabama State University, Montgomery, AL, United States
| | - Saurabh Dixit
- Center for NanoBiotechnology Research, Alabama State University, Montgomery, AL, United States
| | - Skyla A Duncan
- Center for NanoBiotechnology Research, Alabama State University, Montgomery, AL, United States
| | - Guillermo H Giambartolomei
- Instituto de Inmunología, Genética y Metabolismo (INIGEM), CONICET, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Shree R Singh
- Center for NanoBiotechnology Research, Alabama State University, Montgomery, AL, United States
| | - Vida A Dennis
- Center for NanoBiotechnology Research, Alabama State University, Montgomery, AL, United States
| |
Collapse
|
15
|
Sahu R, Verma R, Dixit S, Igietseme JU, Black CM, Duncan S, Singh SR, Dennis VA. Future of human Chlamydia vaccine: potential of self-adjuvanting biodegradable nanoparticles as safe vaccine delivery vehicles. Expert Rev Vaccines 2018; 17:217-227. [PMID: 29382248 PMCID: PMC6330895 DOI: 10.1080/14760584.2018.1435279] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Accepted: 01/29/2018] [Indexed: 01/12/2023]
Abstract
INTRODUCTION There is a persisting global burden and considerable public health challenge by the plethora of ocular, genital and respiratory diseases caused by members of the Gram-negative bacteria of the genus Chlamydia. The major diseases are conjunctivitis and blinding trachoma, non-gonococcal urethritis, cervicitis, pelvic inflammatory disease, ectopic pregnancy, tubal factor infertility, and interstitial pneumonia. The failures in screening and other prevention programs led to the current medical opinion that an efficacious prophylactic vaccine is the best approach to protect humans from chlamydial infections. Unfortunately, there is no human Chlamydia vaccine despite successful veterinary vaccines. A major challenge has been the effective delivery of vaccine antigens to induce safe and effective immune effectors to confer long-term protective immunity. The dawn of the era of biodegradable polymeric nanoparticles and the adjuvanted derivatives may accelerate the realization of the dream of human vaccine in the foreseeable future. AREAS COVERED This review focuses on the current status of human chlamydial vaccine research, specifically the potential of biodegradable polymeric nanovaccines to provide efficacious Chlamydia vaccines in the near future. EXPERT COMMENTARY The safety of biodegradable polymeric nanoparticles-based experimental vaccines with or without adjuvants and the array of available chlamydial vaccine candidates would suggest that clinical trials in humans may be imminent. Also, the promising results from vaccine testing in animal models could lead to human vaccines against trachoma and reproductive diseases simultaneously.
Collapse
Affiliation(s)
- Rajnish Sahu
- Department of Biological Sciences, Alabama State University, Montgomery, AL, USA
| | - Richa Verma
- Department of Biological Sciences, Alabama State University, Montgomery, AL, USA
| | - Saurabh Dixit
- Department of Biological Sciences, Alabama State University, Montgomery, AL, USA
| | - Joseph U. Igietseme
- National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control & Prevention (CDC), Atlanta, GA, USA
| | - Carolyn M Black
- National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control & Prevention (CDC), Atlanta, GA, USA
| | - Skyla Duncan
- Department of Biological Sciences, Alabama State University, Montgomery, AL, USA
| | - Shree R Singh
- Department of Biological Sciences, Alabama State University, Montgomery, AL, USA
| | - Vida A Dennis
- Department of Biological Sciences, Alabama State University, Montgomery, AL, USA
| |
Collapse
|
16
|
O'Meara CP, Armitage CW, Andrew DW, Kollipara A, Lycke NY, Potter AA, Gerdts V, Petrovsky N, Beagley KW. Multistage vaccines containing outer membrane, type III secretion system and inclusion membrane proteins protects against a Chlamydia genital tract infection and pathology. Vaccine 2017; 35:3883-3888. [PMID: 28602608 DOI: 10.1016/j.vaccine.2017.05.063] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Revised: 05/20/2017] [Accepted: 05/23/2017] [Indexed: 01/07/2023]
Abstract
Pathogens with a complex lifecycles can effectively evade host immunity in part due to each developmental stage expressing unique sets of antigens. Multisubunit vaccines incorporating signature antigens reflecting distinct developmental stages (multistage vaccines) have proven effective against viral, bacterial and parasitic infection at preventing pathogen evasion of host immunity. Chlamydia trachomatis is characterized by a biphasic extra/intracellular developmental cycle and an acute/persistent (latent) metabolic state; hence a multistage vaccine may prevent immune evasion and enhance clearance. Here we tested the efficacy of a multistage vaccine containing outer membrane (MOMP and PmpG), type three secretion system (T3SS) (CdsF and TC0873) and inclusion membrane proteins (IncA and TC0500) in mice against an intravaginal challenge with Chlamydia muridarum. Comparison of single (eg. MOMP) and double antigen vaccines (eg. MOMP and PmpG), largely targeting the extracellular stage, elicited significant yet comparable protection against vaginal shedding when compared to unimmunized control mice. Utilization of different adjuvants (ISCOMATRIX - IMX, PCEP/polyI:C/IDR1002 - VIDO, CTA1-DD and ADVAX) and numerous immunization routes (subcutaneous - SQ and intranasal - IN) further enhanced protection against infection. However, a multistage vaccine elicited significantly greater protection against vaginal shedding and upper genital tract pathology than vaccines targeting only extra- or intracellular stages. This indicates that protection elicited by a vaccine targeting extracellular chlamydial antigens could be improved by including chlamydial antigen expressed during intracellular phase.
Collapse
Affiliation(s)
- Connor P O'Meara
- Institute of Health and Biomedical Innovation (IHBI) and School of Biomedical Sciences, Queensland University of Technology (QUT), Brisbane, Queensland, Australia; Department of Developmental Immunology, Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Baden-Wüttemburg, Germany
| | - Charles W Armitage
- Institute of Health and Biomedical Innovation (IHBI) and School of Biomedical Sciences, Queensland University of Technology (QUT), Brisbane, Queensland, Australia
| | - Dean W Andrew
- Department of Biochemistry and Molecular Biology, University of Melbourne, Melbourne, Victoria, Australia
| | - Avinash Kollipara
- Institute of Health and Biomedical Innovation (IHBI) and School of Biomedical Sciences, Queensland University of Technology (QUT), Brisbane, Queensland, Australia
| | - Nils Y Lycke
- Mucosal Immunobiology and Vaccine Centre, University of Gothenburg, Sweden
| | - Andrew A Potter
- Vaccine and Infectious Disease Organization - International Vaccine Centre, University of Saskatchewan, Saskatoon, Canada
| | - Volker Gerdts
- Vaccine and Infectious Disease Organization - International Vaccine Centre, University of Saskatchewan, Saskatoon, Canada
| | - Nikolai Petrovsky
- Vaxine Pty Ltd, Adelaide, Australia; Department of Endocrinology, Flinders Medical Centre/Flinders University, Adelaide, Australia
| | - Kenneth W Beagley
- Institute of Health and Biomedical Innovation (IHBI) and School of Biomedical Sciences, Queensland University of Technology (QUT), Brisbane, Queensland, Australia.
| |
Collapse
|
17
|
Immunization of a wild koala population with a recombinant Chlamydia pecorum Major Outer Membrane Protein (MOMP) or Polymorphic Membrane Protein (PMP) based vaccine: New insights into immune response, protection and clearance. PLoS One 2017; 12:e0178786. [PMID: 28575080 PMCID: PMC5456371 DOI: 10.1371/journal.pone.0178786] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Accepted: 05/18/2017] [Indexed: 12/11/2022] Open
Abstract
We assessed the effects of two different single-dose anti-Chlamydia pecorum (C. pecorum) vaccines (containing either Major Outer Membrane Protein (3MOMP) or Polymorphic Membrane Protein (Pmp) as antigens) on the immune response of a group of wild koalas. Both vaccines elicited a systemic humoral response as seen by the production of anti-chlamydial IgG antibodies in more than 90% of vaccinated koalas. A mucosal immune response was also observed, with an increase in Chlamydia-specific mucosal IgG and/or IgA antibodies in some koalas post-vaccination. Both vaccines elicited a cell-mediated immune response as measured by the production of the cytokines IFN-γ and IL-17 post-vaccination. To determine the level of protection provided by the vaccines under natural conditions we assessed C. pecorum infection loads and chlamydial disease status of all vaccinated koalas pre- and post-vaccination, compared to a non-vaccinated cohort from the same habitat. The MOMP vaccinated koalas that were infected on the day of vaccination showed significant clearance of their infection at 6 months post-vaccination. In contrast, the number of new infections in the PMP vaccine was similar to the control group, with some koalas progressing to disease. Genotyping of the ompA gene from the C. pecorum strains infecting the vaccinated animals, identified genetic variants of ompA-F genotype and a new genotype ompA-O. We found that those animals that were the least well protected became infected with strains of C. pecorum not covered by the vaccine. In conclusion, a single dose vaccine formulated with either recombinant PmpG or MOMP can elicit both cell-mediated and humoral (systemic and mucosal) immune responses, with the MOMP vaccine showing clearance of infection in all infected koalas. Although the capability of our vaccines to stimulate an adaptive response and be protective needs to be fully evaluated, this work illustrates the necessity to combine epitopes most relevant to a large panel of variable strains with an efficient adjuvant.
Collapse
|
18
|
Hadad R, Marks E, Kalbina I, Schön K, Unemo M, Lycke N, Strid Å, Andersson S. Protection against genital tract Chlamydia trachomatis infection following intranasal immunization with a novel recombinant MOMP VS2/4 antigen. APMIS 2017; 124:1078-1086. [PMID: 27859689 DOI: 10.1111/apm.12605] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2016] [Accepted: 08/18/2016] [Indexed: 12/01/2022]
Abstract
The asymptomatic nature of most Chlamydia trachomatis infections and the lack of appropriate effects by current prevention and management call for vaccine development. We evaluated a recombinant subunit vaccine candidate based on the major outer membrane protein variable segments 2 and 4 (MOMP VS2/4). To achieve maximal immunogenicity and ease of production and purification, MOMP VS2/4 was constructed by using highly immunogenic sequences of MOMP only, thereby minimizing the presence of hydrophobic regions, and spacing the immunogenic epitopes with a flexible amino acid sequence. A purification tag was also added. The MOMP VS2/4 was given intranasally, with or without intravaginal boost, with cholera toxin (CT) adjuvant to C57BL/6 mice, which were screened for immunogenicity and protection against a live challenge infection with C. trachomatis serovar D. Bacterial shedding, cell-mediated responses, and antibody responses were monitored. Immunized mice exhibited significantly less bacterial shedding and were better protected against infertility as compared to unimmunized control mice. Immunizations stimulated both systemic and local specific antibody (IgG1, IgG2c, and IgA) responses, and primed T cells that produced interferon-γ and interleukins 13 and 17 upon challenge with recall antigen. Thus, MOMP VS2/4, in combination with CT adjuvant, stimulated Th1, Th2, and Th17 effector cells, and generated protective immunity associated with less pathology. We regard MOMP VS2/4 as a promising candidate for further development into a mucosal chlamydial vaccine.
Collapse
Affiliation(s)
- Ronza Hadad
- Örebro Life Science Center, School of Science and Technology, Örebro University, Örebro, Sweden.,Department of Laboratory Medicine, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Ellen Marks
- Department of Medical Microbiology and Immunology, University of Gothenburg, Gothenburg, Sweden
| | - Irina Kalbina
- Örebro Life Science Center, School of Science and Technology, Örebro University, Örebro, Sweden
| | - Karin Schön
- Department of Medical Microbiology and Immunology, University of Gothenburg, Gothenburg, Sweden
| | - Magnus Unemo
- Department of Laboratory Medicine, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Nils Lycke
- Department of Medical Microbiology and Immunology, University of Gothenburg, Gothenburg, Sweden
| | - Åke Strid
- Örebro Life Science Center, School of Science and Technology, Örebro University, Örebro, Sweden
| | - Sören Andersson
- Department of Laboratory Medicine, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| |
Collapse
|
19
|
Armitage CW, O'Meara CP, Beagley KW. Chlamydial infection enhances expression of the polymeric immunoglobulin receptor (pIgR) and transcytosis of IgA. Am J Reprod Immunol 2016; 77. [PMID: 27868280 DOI: 10.1111/aji.12611] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Accepted: 11/03/2016] [Indexed: 11/27/2022] Open
Abstract
PROBLEM The pIgR mediates transport of IgA into the lumen of mucosal tissues preventing pathogenic infection. Despite this, the expression of pIgR during chlamydial infections of the male and female reproductive tracts remains poorly understood. METHOD OF STUDY The expression of pIgR in response to hormone cycling or over the course of chlamydial infection was determined in vitro and in vivo by Western blot or immunohistochemistry. RESULTS PIgR was upregulated in response to Chlamydia spp. infection of human epithelia, in both male and female mouse reproductive tracts. PIgR expression was found to be highest during estrus in the cervicovaginal and uterine epithelia and lowest during diestrus or following hormonal synchronization with Depo-Provera. Chlamydial infection of mice mediates upregulation of pIgR and transcytosis of IgA into the lumen. CONCLUSIONS Our results suggest that chlamydial infection enhances IgA secretion and pIgR expression by epithelia in the lower reproductive tracts of females and males, and hormone synchronization downregulates pIgR expression and transcytosis of IgA prior to challenge.
Collapse
Affiliation(s)
- Charles W Armitage
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Qld, Australia
| | - Connor P O'Meara
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Qld, Australia.,Department of Developmental Immunology, Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Baden-Wurttemberg, Germany
| | - Kenneth W Beagley
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Qld, Australia
| |
Collapse
|
20
|
O'Meara CP, Armitage CW, Kollipara A, Andrew DW, Trim L, Plenderleith MB, Beagley KW. Induction of partial immunity in both males and females is sufficient to protect females against sexual transmission of Chlamydia. Mucosal Immunol 2016; 9:1076-88. [PMID: 26647717 DOI: 10.1038/mi.2015.125] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2015] [Accepted: 10/22/2015] [Indexed: 02/04/2023]
Abstract
Sexually transmitted Chlamydia trachomatis causes infertility, and because almost 90% of infections are asymptomatic, a vaccine is required for its eradication. Mathematical modeling studies have indicated that a vaccine eliciting partial protection (non-sterilizing) may prevent Chlamydia infection transmission, if administered to both sexes before an infection. However, reducing chlamydial inoculum transmitted by males and increasing infection resistance in females through vaccination to elicit sterilizing immunity has yet to be investigated experimentally. Here we show that a partially protective vaccine (chlamydial major outer membrane protein (MOMP) and ISCOMATRIX (IMX) provided sterilizing immunity against sexual transmission between immunized mice. Immunizing male or female mice before an infection reduced chlamydial burden and disease development, but did not prevent infection. However, infection and inflammatory disease responsible for infertility were absent in 100% of immunized female mice challenged intravaginally with ejaculate collected from infected immunized males. In contrast to the sterilizing immunity generated following recovery from a previous chlamydial infection, protective immunity conferred by MOMP/IMX occurred independent of resident memory T cells. Our results demonstrate that vaccination of males or females can further protect the opposing sex, whereas vaccination of both sexes can synergize to elicit sterilizing immunity against Chlamydia sexual transmission.
Collapse
Affiliation(s)
- C P O'Meara
- Department of Infectious Diseases, Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology (QUT), Brisbane, Queensland, Australia.,Department of Developmental Immunology, Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Baden-Wüttemburg, Germany
| | - C W Armitage
- Department of Infectious Diseases, Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology (QUT), Brisbane, Queensland, Australia
| | - A Kollipara
- Department of Infectious Diseases, Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology (QUT), Brisbane, Queensland, Australia
| | - D W Andrew
- Department of Infectious Diseases, Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology (QUT), Brisbane, Queensland, Australia.,Department of Infectious Diseases, Centre for Biomedical Research, Burnet Institute, Melbourne, Victoria, Australia
| | - L Trim
- Department of Infectious Diseases, Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology (QUT), Brisbane, Queensland, Australia
| | - M B Plenderleith
- Department of Biomedical Sciences, Neuroscience Laboratory-School of Biomedical Science, Queensland University of Technology (QUT), Brisbane, Queensland, Australia
| | - K W Beagley
- Department of Infectious Diseases, Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology (QUT), Brisbane, Queensland, Australia
| |
Collapse
|
21
|
Genital Chlamydia trachomatis: understanding the roles of innate and adaptive immunity in vaccine research. Clin Microbiol Rev 2016; 27:346-70. [PMID: 24696438 DOI: 10.1128/cmr.00105-13] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Chlamydia trachomatis is the leading cause of bacterial sexually transmitted disease worldwide, and despite significant advances in chlamydial research, a prophylactic vaccine has yet to be developed. This Gram-negative obligate intracellular bacterium, which often causes asymptomatic infection, may cause pelvic inflammatory disease (PID), ectopic pregnancies, scarring of the fallopian tubes, miscarriage, and infertility when left untreated. In the genital tract, Chlamydia trachomatis infects primarily epithelial cells and requires Th1 immunity for optimal clearance. This review first focuses on the immune cells important in a chlamydial infection. Second, we summarize the research and challenges associated with developing a chlamydial vaccine that elicits a protective Th1-mediated immune response without inducing adverse immunopathologies.
Collapse
|
22
|
Knudsen NPH, Olsen A, Buonsanti C, Follmann F, Zhang Y, Coler RN, Fox CB, Meinke A, D'Oro U, Casini D, Bonci A, Billeskov R, De Gregorio E, Rappuoli R, Harandi AM, Andersen P, Agger EM. Different human vaccine adjuvants promote distinct antigen-independent immunological signatures tailored to different pathogens. Sci Rep 2016; 6:19570. [PMID: 26791076 PMCID: PMC4726129 DOI: 10.1038/srep19570] [Citation(s) in RCA: 191] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Accepted: 12/15/2015] [Indexed: 01/20/2023] Open
Abstract
The majority of vaccine candidates in clinical development are highly purified proteins and peptides relying on adjuvants to enhance and/or direct immune responses. Despite the acknowledged need for novel adjuvants, there are still very few adjuvants in licensed human vaccines. A vast number of adjuvants have been tested pre-clinically using different experimental conditions, rendering it impossible to directly compare their activity. We performed a head-to-head comparison of five different adjuvants Alum, MF59®, GLA-SE, IC31® and CAF01 in mice and combined these with antigens from M. tuberculosis, influenza, and chlamydia to test immune-profiles and efficacy in infection models using standardized protocols. Regardless of antigen, each adjuvant had a unique immunological signature suggesting that the adjuvants have potential for different disease targets. Alum increased antibody titers; MF59® induced strong antibody and IL-5 responses; GLA-SE induced antibodies and Th1; CAF01 showed a mixed Th1/Th17 profile and IC31® induced strong Th1 responses. MF59® and GLA-SE were strong inducers of influenza HI titers while CAF01, GLA-SE and IC31® enhanced protection to TB and chlamydia. Importantly, this is the first extensive attempt to categorize clinical-grade adjuvants based on their immune profiles and protective efficacy to inform a rational development of next generation vaccines for human use.
Collapse
Affiliation(s)
- Niels Peter H Knudsen
- Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark
| | - Anja Olsen
- Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark
| | - Cecilia Buonsanti
- Novartis Vaccines and Diagnostics s.r.l (a GSK Company), Siena, Italy
| | - Frank Follmann
- Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark
| | - Yuan Zhang
- Department of Microbiology and Immunology, University of Gothenburg, Gothenburg, Sweden
| | - Rhea N Coler
- Infectious Disease Research Institute, Seattle, WA, USA
| | | | | | - Ugo D'Oro
- Novartis Vaccines and Diagnostics s.r.l (a GSK Company), Siena, Italy
| | - Daniele Casini
- Novartis Vaccines and Diagnostics s.r.l (a GSK Company), Siena, Italy
| | - Alessandra Bonci
- Novartis Vaccines and Diagnostics s.r.l (a GSK Company), Siena, Italy
| | - Rolf Billeskov
- Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark
| | - Ennio De Gregorio
- Novartis Vaccines and Diagnostics s.r.l (a GSK Company), Siena, Italy
| | - Rino Rappuoli
- Novartis Vaccines and Diagnostics s.r.l (a GSK Company), Siena, Italy
| | - Ali M Harandi
- Department of Microbiology and Immunology, University of Gothenburg, Gothenburg, Sweden
| | - Peter Andersen
- Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark
| | - Else Marie Agger
- Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark
| |
Collapse
|
23
|
Baillet AC, Rehaume LM, Benham H, O'Meara CP, Armitage CW, Ruscher R, Brizard G, Harvie MCG, Velasco J, Hansbro PM, Forrester JV, Degli-Esposti MA, Beagley KW, Thomas R. High Chlamydia Burden Promotes Tumor Necrosis Factor-Dependent Reactive Arthritis in SKG Mice. Arthritis Rheumatol 2015; 67:1535-47. [PMID: 25624153 DOI: 10.1002/art.39041] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Accepted: 01/15/2015] [Indexed: 12/14/2022]
Abstract
OBJECTIVE Chlamydia trachomatis is a sexually transmitted obligate intracellular pathogen that causes inflammatory reactive arthritis, spondylitis, psoriasiform dermatitis, and conjunctivitis in some individuals after genital infection. The immunologic basis for this inflammatory response in susceptible hosts is poorly understood. As ZAP-70(W163C) -mutant BALB/c (SKG) mice are susceptible to spondylo-arthritis after systemic exposure to microbial β-glucan, we undertook the present study to compare responses to infection with Chlamydia muridarum in SKG mice and BALB/c mice. METHODS After genital or respiratory infection with C muridarum, conjunctivitis and arthritis were assessed clinically, and eye, skin, and joint specimens were analyzed histologically. Chlamydial major outer membrane protein antigen-specific responses were assessed in splenocytes. Treg cells were depleted from FoxP3-DTR BALB/c or SKG mice, and chlamydial DNA was quantified by polymerase chain reaction. RESULTS Five weeks after vaginal infection with live C muridarum, arthritis, spondylitis, and psoriasiform dermatitis developed in female SKG mice, but not in BALB/c mice. Inflammatory bowel disease did not occur in mice of either strain. The severity of inflammatory disease was correlated with C muridarum inoculum size and vaginal burden postinoculation. Treatment with combination antibiotics starting 1 day postinoculation prevented disease. Chlamydial antigen was present in macrophages and spread from the infection site to lymphoid organs and peripheral tissue. In response to chlamydial antigen, production of interferon-γ and interleukin-17 was impaired in T cells from SKG mice but tumor necrosis factor (TNF) responses were exaggerated, compared to findings in T cells from BALB/c mice. Unlike previous observations in arthritis triggered by β-glucan, no autoantibodies developed. Accelerated disease triggered by depletion of Treg cells was TNF dependent. CONCLUSION In the susceptible SKG strain, Chlamydia-induced reactive arthritis develops as a result of deficient intracellular pathogen control, with antigen-specific TNF production upon dissemination of antigen, and TNF-dependent inflammatory disease.
Collapse
Affiliation(s)
- Athan C Baillet
- University of Queensland Diamantina Institute, Translational Research Institute and Princess Alexandra Hospital, Brisbane, Queensland, Australia
| | - Linda M Rehaume
- University of Queensland Diamantina Institute, Translational Research Institute and Princess Alexandra Hospital, Brisbane, Queensland, Australia
| | - Helen Benham
- University of Queensland Diamantina Institute, Translational Research Institute and Princess Alexandra Hospital, Brisbane, Queensland, Australia
| | - Connor P O'Meara
- Queensland University of Technology, Brisbane, Queensland, Australia
| | | | - Roland Ruscher
- University of Queensland Diamantina Institute, Translational Research Institute and Princess Alexandra Hospital, Brisbane, Queensland, Australia
| | | | - Marina C G Harvie
- Queensland University of Technology, Brisbane, Queensland, Australia
| | - Jared Velasco
- University of Queensland Diamantina Institute, Translational Research Institute and Princess Alexandra Hospital, Brisbane, Queensland, Australia
| | - Phillip M Hansbro
- Hunter Medical Research Institute and University of Newcastle, Newcastle, New South Wales, Australia
| | - John V Forrester
- Lions Eye Institute, Nedlands, West Australia, Australia, and University of Aberdeen Medical School, Aberdeen, Scotland
| | - Mariapia A Degli-Esposti
- Lions Eye Institute, Nedlands, West Australia, Australia, and University of West Australia, Crawley, West Australia, Australia
| | - Kenneth W Beagley
- Queensland University of Technology, Brisbane, Queensland, Australia
| | - Ranjeny Thomas
- University of Queensland Diamantina Institute, Translational Research Institute and Princess Alexandra Hospital, Brisbane, Queensland, Australia
| |
Collapse
|
24
|
Armitage CW, O'Meara CP, Harvie MCG, Timms P, Wijburg OL, Beagley KW. Evaluation of intra- and extra-epithelial secretory IgA in chlamydial infections. Immunology 2015; 143:520-30. [PMID: 24827556 DOI: 10.1111/imm.12317] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Revised: 05/07/2014] [Accepted: 05/09/2014] [Indexed: 12/20/2022] Open
Abstract
Immunoglobulin A is an important mucosal antibody that can neutralize mucosal pathogens by either preventing attachment to epithelia (immune exclusion) or alternatively inhibit intra-epithelial replication following transcytosis by the polymeric immunoglobulin receptor (pIgR). Chlamydia trachomatis is a major human pathogen that initially targets the endocervical or urethral epithelium in women and men, respectively. As both tissues contain abundant secretory IgA (SIgA) we assessed the protection afforded by IgA targeting different chlamydial antigens expressed during the extra- and intra-epithelial stages of infection. We developed an in vitro model using polarizing cells expressing the murine pIgR together with antigen-specific mouse IgA, and an in vivo model using pIgR(-/-) mice. Secretory IgA targeting the extra-epithelial chlamydial antigen, the major outer membrane protein, significantly reduced infection in vitro by 24% and in vivo by 44%. Conversely, pIgR-mediated delivery of IgA targeting the intra-epithelial inclusion membrane protein A bound to the inclusion but did not reduce infection in vitro or in vivo. Similarly, intra-epithelial IgA targeting the secreted protease Chlamydia protease-like activity factor also failed to reduce infection. Together, these data suggest the importance of pIgR-mediated delivery of IgA targeting extra-epithelial, but not intra-epithelial, chlamydial antigens for protection against a genital tract infection.
Collapse
Affiliation(s)
- Charles W Armitage
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Kelvin Grove, Qld, Australia
| | | | | | | | | | | |
Collapse
|
25
|
Sobinoff AP, Dando SJ, Redgrove KA, Sutherland JM, Stanger SJ, Armitage CW, Timms P, McLaughlin EA, Beagley KW. Chlamydia muridarum infection-induced destruction of male germ cells and sertoli cells is partially prevented by Chlamydia major outer membrane protein-specific immune CD4 cells. Biol Reprod 2014; 92:27. [PMID: 25472923 DOI: 10.1095/biolreprod.114.124180] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
Chlamydia trachomatis infections are increasingly prevalent worldwide. Male chlamydial infections are associated with urethritis, epididymitis, and orchitis; however, the role of Chlamydia in prostatitis and male factor infertility remains controversial. Using a model of Chlamydia muridarum infection in male C57BL/6 mice, we investigated the effects of chlamydial infection on spermatogenesis and determined the potential of immune T cells to prevent infection-induced outcomes. Antigen-specific CD4 T cells significantly reduced the infectious burden in the penile urethra, epididymis, and vas deferens. Infection disrupted seminiferous tubules, causing loss of germ cells at 4 and 8 wk after infection, with the most severely affected tubules containing only Sertoli cells. Increased mitotic proliferation, DNA repair, and apoptosis in spermatogonial cells and damaged germ cells were evident in atrophic tubules. Activated caspase 3 (casp3) staining revealed increased (6-fold) numbers of Sertoli cells with abnormal morphology that were casp3 positive in tubules of infected mice, indicating increased levels of apoptosis. Sperm count and motility were both decreased in infected mice, and there was a significant decrease in morphologically normal spermatozoa. Assessment of the spermatogonial stem cell population revealed a decrease in promyelocytic leukemia zinc finger (PLZF)-positive cells in the seminiferous tubules. Interestingly, adoptive transfer of antigen-specific CD4 cells, particularly T-helper 2-like cells, prior to infection prevented these effects in spermatogenesis and Sertoli cells. These data suggest that chlamydial infection adversely affects spermatogenesis and male fertility, and that vaccination can potentially prevent the spread of infection and these adverse outcomes.
Collapse
Affiliation(s)
- Alexander P Sobinoff
- Priority Research Centres in Chemical Biology and Reproductive Science, School of Environmental & Life Sciences, University of Newcastle, Newcastle, New South Wales, Australia
| | - Samantha J Dando
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Kate A Redgrove
- Priority Research Centres in Chemical Biology and Reproductive Science, School of Environmental & Life Sciences, University of Newcastle, Newcastle, New South Wales, Australia
| | - Jessie M Sutherland
- Priority Research Centres in Chemical Biology and Reproductive Science, School of Environmental & Life Sciences, University of Newcastle, Newcastle, New South Wales, Australia
| | - Simone J Stanger
- Priority Research Centres in Chemical Biology and Reproductive Science, School of Environmental & Life Sciences, University of Newcastle, Newcastle, New South Wales, Australia
| | - Charles W Armitage
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Peter Timms
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Eileen A McLaughlin
- Priority Research Centres in Chemical Biology and Reproductive Science, School of Environmental & Life Sciences, University of Newcastle, Newcastle, New South Wales, Australia
| | - Kenneth W Beagley
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia
| |
Collapse
|
26
|
McNeilly CL, Crichton ML, Primiero CA, Frazer IH, Roberts MS, Kendall MAF. Microprojection arrays to immunise at mucosal surfaces. J Control Release 2014; 196:252-60. [PMID: 25285611 DOI: 10.1016/j.jconrel.2014.09.028] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Revised: 09/22/2014] [Accepted: 09/29/2014] [Indexed: 12/18/2022]
Abstract
The buccal mucosa (inner cheek) is an attractive site for delivery of immunotherapeutics, due to its ease of access and rich antigen presenting cell (APC) distribution. However, to date, most delivery methods to the buccal mucosa have only been topical-with the challenges of: 1) an environment where significant biomolecule degradation may occur; 2) inability to reach the APCs that are located deep in the epithelium and lamina propria; and 3) salivary flow and mucous secretion that may result in removal of the therapeutic agent before absorption has taken place. To overcome these challenges and achieve consistent, repeatable targeted delivery of immunotherapeutics to within the buccal mucosa (not merely on to the surface), we utilised microprojection arrays (Nanopatches-110 μm length projections, 3364 projections, 16 mm2 surface area) with a purpose built clip applicator. The mechanical application of Nanopatches bearing a dry-coated vaccine (commercial influenza vaccine, as a test case immunotherapeutic) released the vaccine to a depth of 47.8±14.8 μm (mean±SD, n=4), in the mouse buccal mucosa (measured using fluorescent delivered dyes and CryoSEM). This location is in the direct vicinity of APCs, facilitating antigenic uptake. Resultant systemic immune responses were similar to systemic immunization methods, and superior to comparative orally immunised mice. This confirms the Nanopatch administered vaccine was delivered into the buccal mucosa and not ingested. This study demonstrates a minimally-invasive delivery device with rapid (2 min of application time), accurate and consistent release of immunotherapeutics in to the buccal mucosa-that conceptually can be extended in to human use for broad and practical utility.
Collapse
Affiliation(s)
- Celia L McNeilly
- The University of Queensland, Delivery of Drugs and Genes Group (D(2)G(2)), The Australian Institute for Bioengineering and Nanotechnology, St Lucia, QLD 4072, Australia
| | - Michael L Crichton
- The University of Queensland, Delivery of Drugs and Genes Group (D(2)G(2)), The Australian Institute for Bioengineering and Nanotechnology, St Lucia, QLD 4072, Australia; Vaxxas Pty Ltd, Australian Institute for Bioengineering and Nanotechnology, Brisbane, Queensland, Australia
| | - Clare A Primiero
- The University of Queensland, Delivery of Drugs and Genes Group (D(2)G(2)), The Australian Institute for Bioengineering and Nanotechnology, St Lucia, QLD 4072, Australia
| | - Ian H Frazer
- The University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, Queensland, Australia
| | - Michael S Roberts
- Therapeutics Research Centre, School of Medicine, Princess Alexandra Hospital, The University of Queensland, Brisbane, Queensland, Australia; School of Pharmacy and Medical Science, University of South Australia, Adelaide, Australia
| | - Mark A F Kendall
- The University of Queensland, Delivery of Drugs and Genes Group (D(2)G(2)), The Australian Institute for Bioengineering and Nanotechnology, St Lucia, QLD 4072, Australia; Vaxxas Pty Ltd, Australian Institute for Bioengineering and Nanotechnology, Brisbane, Queensland, Australia; The University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, Queensland, Australia.
| |
Collapse
|
27
|
Mathew M, Waugh C, Beagley KW, Timms P, Polkinghorne A. Interleukin 17A is an immune marker for chlamydial disease severity and pathogenesis in the koala (Phascolarctos cinereus). DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2014; 46:423-429. [PMID: 24915607 DOI: 10.1016/j.dci.2014.05.015] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2014] [Revised: 05/22/2014] [Accepted: 05/22/2014] [Indexed: 06/03/2023]
Abstract
The koala (Phascolarctos cinereus) is an iconic Australian marsupial species that is facing many threats to its survival. Chlamydia pecorum infections are a significant contributor to this ongoing decline. A major limiting factor in our ability to manage and control chlamydial disease in koalas is a limited understanding of the koala's cell-mediated immune response to infections by this bacterial pathogen. To identify immunological markers associated with chlamydial infection and disease in koalas, we used koala-specific Quantitative Real Time PCR (qrtPCR) assays to profile the cytokine responses of Peripheral Blood Mononuclear Cells (PBMCs) collected from 41 koalas with different stages of chlamydial disease. Target cytokines included the principal Th1 (Interferon gamma; IFNγ), Th2 (Interleukin 10; IL10), and pro-inflammatory cytokines (Tumor Necrosis Factor alpha; TNFα). A novel koala-specific IL17A qrtPCR assay was also developed as part of this study to quantitate the gene expression of this Th17 cytokine in koalas. A statistically significant higher IL17A gene expression was observed in animals with current chlamydial disease compared to animals with asymptomatic chlamydial infection. A modest up-regulation of pro-inflammatory cytokines, such as TNFα and IFNγ, was also observed in these animals with signs of current chlamydial disease. IL10 gene expression was not evident in the majority of animals from both groups. Future longitudinal studies are now required to confirm the role played by cytokines in pathology and/or protection against C. pecorum infection in the koala.
Collapse
Affiliation(s)
- Marina Mathew
- Institute of Health & Biomedical Innovation, Queensland University of Technology, 60 Musk Avenue, Kelvin Grove 4059, Brisbane, Australia
| | - Courtney Waugh
- Institute of Health & Biomedical Innovation, Queensland University of Technology, 60 Musk Avenue, Kelvin Grove 4059, Brisbane, Australia; Faculty of Science, Health, Education and Engineering, University of the Sunshine Coast, 90 Sippy Downs Dr, Sippy Downs 4558, QLD, Australia
| | - Kenneth W Beagley
- Institute of Health & Biomedical Innovation, Queensland University of Technology, 60 Musk Avenue, Kelvin Grove 4059, Brisbane, Australia
| | - Peter Timms
- Institute of Health & Biomedical Innovation, Queensland University of Technology, 60 Musk Avenue, Kelvin Grove 4059, Brisbane, Australia; Faculty of Science, Health, Education and Engineering, University of the Sunshine Coast, 90 Sippy Downs Dr, Sippy Downs 4558, QLD, Australia
| | - Adam Polkinghorne
- Institute of Health & Biomedical Innovation, Queensland University of Technology, 60 Musk Avenue, Kelvin Grove 4059, Brisbane, Australia; Faculty of Science, Health, Education and Engineering, University of the Sunshine Coast, 90 Sippy Downs Dr, Sippy Downs 4558, QLD, Australia.
| |
Collapse
|
28
|
Divergent outcomes following transcytosis of IgG targeting intracellular and extracellular chlamydial antigens. Immunol Cell Biol 2014; 92:417-26. [PMID: 24445600 DOI: 10.1038/icb.2013.110] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Revised: 12/11/2013] [Accepted: 12/12/2013] [Indexed: 12/12/2022]
Abstract
Antibodies can have a protective but non-essential role in natural chlamydial infections dependent on antigen specificity and antibody isotype. IgG is the dominant antibody in both male and female reproductive tract mucosal secretions, and is bi-directionally trafficked across epithelia by the neonatal Fc receptor (FcRn). Using pH-polarized epididymal epithelia grown on Transwells, IgG specifically targeted at an extracellular chlamydial antigen; the major outer membrane protein (MOMP), enhanced uptake and translocation of infection at pH 6-6.5 but not at neutral pH. This was dependent on FcRn expression. Conversely, FcRn-mediated transport of IgG targeting the intracellular chlamydial inclusion membrane protein A (IncA), induced aberrant inclusion morphology, recruited autophagic proteins independent of lysosomes and significantly reduced infection. Challenge of female mice with MOMP-specific IgG-opsonized Chlamydia muridarum delayed infection clearance but exacerbated oviduct occlusion. In male mice, MOMP-IgG elicited by immunization afforded no protection against testicular chlamydial infection, whereas the transcytosis of IncA-IgG significantly reduced testicular chlamydial burden. Together these data show that the protective and pathological effects of IgG are dependent on FcRn-mediated transport as well as the specificity of IgG for intracellular or extracellular antigens.
Collapse
|
29
|
Immunity against a Chlamydia infection and disease may be determined by a balance of IL-17 signaling. Immunol Cell Biol 2013; 92:287-97. [PMID: 24366518 DOI: 10.1038/icb.2013.92] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2013] [Revised: 11/03/2013] [Accepted: 11/07/2013] [Indexed: 01/05/2023]
Abstract
Most vaccines developed against Chlamydia using animal models provide partial protection against a genital tract infection. However, protection against the oviduct pathology associated with infertility is highly variable and often has no defining immunological correlate. When comparing two adjuvants (CTA1-DD and a combination of Cholera toxin plus CpG-oligodeoxynucleotide-CT/CpG) combined with the chlamydial major outer membrane protein (MOMP) antigen and delivered via the intranasal (IN), sublingual (SL) or transcutaneous (TC) routes, we identified two vaccine groups with contrasting outcomes following infection. SL immunization with MOMP/CTA1-DD induced a 70% reduction in the incidence of oviduct pathology, without significantly altering the course of infection. Conversely, IN immunization with MOMP/CT/CpG prevented an ascending infection, but not the oviduct pathology. This anomaly presented a unique opportunity to study the mechanisms by which vaccines can prevent oviduct pathology, other than by controlling the infection. The IL-17 signaling in the oviducts was found to associate with both the enhancement of immunity to infection and the development of oviduct pathology. This conflicting role of IL-17 may provide some explanation for the discordance in protection between infection and disease and suggests that controlling immunopathology, as opposed to the rapid eradication of the infection, may be essential for an effective human chlamydial vaccine that prevents infertility.
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
|