1
|
Karunakaran KP, Yu H, Jiang X, Chan QWT, Sigola L, Millis LA, Chen J, Tang P, Foster LJ, Brunham RC. Immunoproteomic discovery of Mycobacterium bovis antigens, including the surface lipoprotein Mpt83 as a T cell antigen useful for vaccine development. Vaccine 2024; 42:126266. [PMID: 39232399 DOI: 10.1016/j.vaccine.2024.126266] [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: 02/10/2024] [Revised: 08/12/2024] [Accepted: 08/21/2024] [Indexed: 09/06/2024]
Abstract
Tuberculosis (TB) is one of the leading causes of death from infectious diseases, killing approximately 1.3 million people worldwide in 2022 alone. The current vaccine for TB contains a live attenuated bacterium, Mycobacterium bovis BCG (Bacille Calmette-Guérin). The BCG vaccine is highly effective in preventing severe forms of childhood TB but does not protect against latent infection or disease in older age groups. A new or improved BCG vaccine for prevention of pulmonary TB is urgently needed. In this study, we infected murine bone marrow derived dendritic cells from C57BL/6 mice with M. bovis BCG followed by elution and identification of BCG-derived MHC class I and class II-bound peptides using tandem mass spectrometry. We identified 1436 MHC-bound peptides of which 94 were derived from BCG. Fifty-five peptides were derived from MHC class I molecules and 39 from class II molecules. We tested the 94 peptides for their immunogenicity using IFN- γ ELISPOT assay with splenocytes purified from BCG immunized mice and 10 showed positive responses. Seven peptides were derived from MHC II and three from MHC class I. In particular, MHC class II binding peptides derived from the mycobacterial surface lipoprotein Mpt83 were highly antigenic. Further evaluations of these immunogenic BCG peptides may identify proteins useful as new TB vaccine candidates.
Collapse
Affiliation(s)
- Karuna P Karunakaran
- Vaccine Research Laboratory, University of British Columbia Centre for Disease Control, Vancouver, B.C., Canada; Department of Biology, Faculty of Science and Technology, Douglas College, New Westminster, B.C., Canada
| | - Hong Yu
- Vaccine Research Laboratory, University of British Columbia Centre for Disease Control, Vancouver, B.C., Canada
| | - Xiaozhou Jiang
- Vaccine Research Laboratory, University of British Columbia Centre for Disease Control, Vancouver, B.C., Canada
| | - Queenie W T Chan
- Department of Biochemistry and Molecular Biology, Michael Smith Laboratories, University of British Columbia, Vancouver, B.C., Canada
| | - Lynette Sigola
- Department of Biology, Faculty of Science and Technology, Douglas College, New Westminster, B.C., Canada
| | - Leonard A Millis
- Department of Biology, Faculty of Science and Technology, Douglas College, New Westminster, B.C., Canada
| | - Jiaqi Chen
- Department of Biochemistry and Molecular Biology, Michael Smith Laboratories, University of British Columbia, Vancouver, B.C., Canada
| | | | - Leonard J Foster
- Department of Biochemistry and Molecular Biology, Michael Smith Laboratories, University of British Columbia, Vancouver, B.C., Canada
| | - Robert C Brunham
- Vaccine Research Laboratory, University of British Columbia Centre for Disease Control, Vancouver, B.C., Canada.
| |
Collapse
|
2
|
Dziadula J, Sabbaj S, Gupta K, Cutter G, Yu H, Brunham RC, Geisler WM. Interferon-γ Responses to Chlamydia trachomatis Vaccine Candidate Proteins in Women With Different Chlamydia Outcomes. J Infect Dis 2024; 230:569-577. [PMID: 38394609 PMCID: PMC11420703 DOI: 10.1093/infdis/jiae092] [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: 10/27/2023] [Revised: 01/31/2024] [Accepted: 02/21/2024] [Indexed: 02/25/2024] Open
Abstract
BACKGROUND Chlamydia trachomatis testing and treatment strategies have not decreased infection rates, justifying need for a chlamydia vaccine. A murine study showed that a vaccine consisting of major outer membrane protein (MOMP) and polymorphic membrane proteins (Pmps) E, F, G, and H elicited protective immunity; studies on human cellular immune responses to Pmps are sparse. METHODS Interferon gamma (IFN-γ) responses to these 5 proteins were measured by ELISPOT in peripheral blood mononuclear cells from women returning for treatment of a positive chlamydia test. Responses were compared in those with spontaneous chlamydia clearance versus persisting infection at baseline and no reinfection versus reinfection at a 3-month follow-up visit. RESULTS IFN-γ response to 1 or more proteins was detected in 39% at baseline and 51.5% at follow-up, most often to PmpE and MOMP. IFN-γ responses to MOMP were detected less often at follow-up versus baseline in women with reinfection, but were maintained in those without reinfection. Women with spontaneous clearance had a higher magnitude of IFN-γ response to PmpE and MOMP. CONCLUSIONS IFN-γ responses to these 5 C. trachomatis vaccine candidate proteins were heterogenous and primarily directed against MOMP and PmpE. Spontaneous chlamydia clearance and absence of reinfection may be clinical correlates of protection.
Collapse
Affiliation(s)
- Jacob Dziadula
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Steffanie Sabbaj
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Kanupriya Gupta
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Gary Cutter
- Department of Biostatistics, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Hong Yu
- British Columbia Centre for Disease Control, University of British Columbia, Vancouver, British Columbia, Canada
| | - Robert C Brunham
- British Columbia Centre for Disease Control, University of British Columbia, Vancouver, British Columbia, Canada
| | - William M Geisler
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
3
|
Yount KS, Darville T. Immunity to Sexually Transmitted Bacterial Infections of the Female Genital Tract: Toward Effective Vaccines. Vaccines (Basel) 2024; 12:863. [PMID: 39203989 PMCID: PMC11359697 DOI: 10.3390/vaccines12080863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 07/19/2024] [Accepted: 07/27/2024] [Indexed: 09/03/2024] Open
Abstract
Sexually transmitted infections (STIs) caused by bacterial pathogens Chlamydia trachomatis, Neisseria gonorrhoeae, and Treponema pallidum present significant public health challenges. These infections profoundly impact reproductive health, leading to pelvic inflammatory disease, infertility, and increased susceptibility to other infections. Prevention measures, including antibiotic treatments, are limited by the often-asymptomatic nature of these infections, the need for repetitive and continual screening of sexually active persons, antibiotic resistance for gonorrhea, and shortages of penicillin for syphilis. While vaccines exist for viral STIs like human papillomavirus (HPV) and hepatitis B virus (HBV), there are no vaccines available for bacterial STIs. This review examines the immune responses in the female genital tract to these bacterial pathogens and the implications for developing effective vaccines against bacterial STIs.
Collapse
Affiliation(s)
| | - Toni Darville
- Department of Pediatrics, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA;
| |
Collapse
|
4
|
Murthy AK, Wright-McAfee E, Warda K, Moy LN, Bui N, Musunuri T, Manam S, Chako CZ, Ramsey KH, Li W. Protective anti-chlamydial vaccine regimen-induced CD4+ T cell response mediates early inhibition of pathogenic CD8+ T cell response following genital challenge. Pathog Dis 2024; 82:ftae008. [PMID: 38684476 PMCID: PMC11149721 DOI: 10.1093/femspd/ftae008] [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: 03/05/2024] [Revised: 04/19/2024] [Accepted: 04/26/2024] [Indexed: 05/02/2024] Open
Abstract
We have demonstrated previously that TNF-α-producing CD8+ T cells mediate chlamydial pathogenesis, likely in an antigen (Ag)-specific fashion. Here we hypothesize that inhibition of Ag-specific CD8+ T cell response after immunization and/or challenge would correlate with protection against oviduct pathology induced by a protective vaccine regimen. Intranasal (i.n.) live chlamydial elementary body (EB), intramuscular (i.m.) live EB, or i.n. irrelevant antigen, bovine serum albumin (BSA), immunized animals induced near-total protection, 50% protection, or no protection, respectively against oviduct pathology following i.vag. C. muridarum challenge. In these models, we evaluated Ag-specific CD8+ T cell cytokine response at various time-periods after immunization or challenge. The results show protective efficacy of vaccine regimens correlated with reduction of Ag-specific CD8+ T cell TNF-α responses following i.vag. chlamydial challenge, not after immunization. Depletion of CD4+ T cells abrogated, whereas adoptive transfer of Ag-specific CD4+ T cells induced the significant reduction of Ag-specific CD8+ T cell TNF-α response after chlamydial challenge. In conclusion, protective anti-chlamydial vaccine regimens induce Ag-specific CD4+ T cell response that mediate early inhibition of pathogenic CD8+ T cell response following challenge and may serve as a predictive biomarker of protection against Chlamydia -induced chronic pathologies.
Collapse
Affiliation(s)
- Ashlesh K Murthy
- College of Veterinary Medicine, Midwestern University, Glendale, AZ 85308, USA
| | - Erika Wright-McAfee
- College of Veterinary Medicine, Midwestern University, Glendale, AZ 85308, USA
| | - Katerina Warda
- Arizona College of Osteopathic Medicine, Midwestern University, Glendale, AZ 85308, USA
| | - Lindsay N Moy
- Arizona College of Osteopathic Medicine, Midwestern University, Glendale, AZ 85308, USA
| | - Nhi Bui
- Arizona College of Osteopathic Medicine, Midwestern University, Glendale, AZ 85308, USA
| | - Tarakarama Musunuri
- Arizona College of Osteopathic Medicine, Midwestern University, Glendale, AZ 85308, USA
| | - Srikanth Manam
- College of Veterinary Medicine, Midwestern University, Glendale, AZ 85308, USA
| | - Clemence Z Chako
- College of Veterinary Medicine, Midwestern University, Glendale, AZ 85308, USA
| | - Kyle H Ramsey
- College of Veterinary Medicine, Midwestern University, Glendale, AZ 85308, USA
- College of Graduate Studies, Midwestern University, Glendale, AZ 85308, USA
| | - Weidang Li
- College of Veterinary Medicine, Midwestern University, Glendale, AZ 85308, USA
| |
Collapse
|
5
|
Poston TB. Advances in vaccine development for Chlamydia trachomatis. Pathog Dis 2024; 82:ftae017. [PMID: 39043447 PMCID: PMC11338180 DOI: 10.1093/femspd/ftae017] [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: 04/25/2024] [Revised: 06/18/2024] [Accepted: 07/25/2024] [Indexed: 07/25/2024] Open
Abstract
Chlamydia trachomatis is the most prevalent bacterial sexually transmitted infection globally. Antibiotic treatment is highly effective, but infection is often asymptomatic resulting in most individuals going undetected and untreated. This untreated infection can ascend to the upper female genital tract to cause pelvic inflammatory disease, tubal factor infertility, and ectopic pregnancy. Chlamydia screening and treatment programs have failed to control this epidemic and demonstrate the need for an efficacious vaccine to prevent transmission and disease. Animal models and human epidemiological data reveal that natural immunity can provide partial or short-lived sterilizing immunity. These data further demonstrate the importance of eliciting interferon gamma (IFNγ)-producing cluster of differentiation 4 (CD4) T cells (Th1 and Th1/17 cells) that can likely synergize with antibody-mediated opsonophagocytosis to provide optimal protection. These studies have guided preclinical rational vaccine design for decades and the first Phase 1 clinical trials have recently been completed. Recent advances have led to improvements in vaccine platforms and clinically safe adjuvants that help provide a path forward. This review describes vaccine models, correlates of immunity, antigen and adjuvant selection, and future clinical testing for Chlamydia vaccine development.
Collapse
Affiliation(s)
- Taylor B Poston
- Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
| |
Collapse
|
6
|
Yu H, Geisler WM, Dai C, Gupta K, Cutter G, Brunham RC. Antibody responses to Chlamydia trachomatis vaccine candidate antigens in Chlamydia-infected women and correlation with antibody-mediated phagocytosis of elementary bodies. Front Cell Infect Microbiol 2024; 14:1342621. [PMID: 38371301 PMCID: PMC10869445 DOI: 10.3389/fcimb.2024.1342621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 01/16/2024] [Indexed: 02/20/2024] Open
Abstract
Murine research has revealed a significant role for antibody responses in protection against Chlamydia reinfection. To explore potential humoral immune markers of protection elicited by Chlamydia trachomatis (CT) antigens in humans in the context of presumed clinical correlates of protection, we used both an IgG1-based ELISA and a conventional total IgG ELISA to evaluate antibody responses. We evaluated responses to five CT outer membrane proteins (PmpE, PmpF, PmpG, PmpH, and MOMP), along with other promising CT antigens (Pgp3 and HSP60), negative control antigens (RecO and AtpE), and CT elementary bodies (EBs) in sera from a well-characterized cohort of 60 women with different CT infection outcomes, including two outcomes that are likely clinical correlates of protective immunity: spontaneous resolution of infection and absence of reinfection after treatment. Furthermore, we used a flow cytometry-based assay to measure antibody-mediated phagocytosis by neutrophils in these sera. Results demonstrated that IgG1 ELISA displayed higher sensitivity than conventional total IgG ELISA in assessing antibody responses to CT EBs and antigens. Pgp3 IgG1 ELISA exhibited the highest sensitivity compared to IgG1 ELISA incorporating CT EBs or other antigens, confirming Pgp3 IgG1 ELISA as an ideal assay for CT antibody detection. Most (95%) sera from women with CT infection outcomes exhibited antibody-mediated phagocytosis of CT EBs, which was significantly correlated with IgG1 antibody responses to MOMP, Pgp3, HSP60, and PmpF. However, neither IgG1 responses to CT antigens and EBs nor antibody-mediated phagocytosis were associated with clinical correlates of protection. These findings suggest that neither CT IgG1 antibody detection nor antibody-mediated phagocytosis will be useful as immune correlates of protection against CT infection in humans.
Collapse
Affiliation(s)
- Hong Yu
- Department of Medicine, British Columbia Centre for Disease Control, University of British Columbia, Vancouver, BC, Canada
| | - William M. Geisler
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Chuanbin Dai
- Department of Medicine, British Columbia Centre for Disease Control, University of British Columbia, Vancouver, BC, Canada
| | - Kanupriya Gupta
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Gary Cutter
- Department of Biostatistics, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Robert C. Brunham
- Department of Medicine, British Columbia Centre for Disease Control, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
7
|
Lu C, Wang J, Zhong G. Preclinical screen for protection efficacy of chlamydial antigens that are immunogenic in humans. Infect Immun 2023; 91:e0034923. [PMID: 37889004 PMCID: PMC10652899 DOI: 10.1128/iai.00349-23] [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: 08/29/2023] [Accepted: 10/02/2023] [Indexed: 10/28/2023] Open
Abstract
To search for subunit vaccine candidates, immunogenic chlamydial antigens identified in humans were evaluated for protection against both infection and pathology in a mouse genital tract infection model under three different immunization regimens. The intramuscular immunization regimen was first used to evaluate 106 chlamydial antigens, which revealed that two antigens significantly reduced while 11 increased genital chlamydial burden. The two infection-reducing antigens failed to prevent pathology and 23 additional antigens even exacerbated pathology. Thus, intranasal mucosal immunization was tested next since intranasal inoculation with live Chlamydia muridarum prevented both genital infection and pathology. Two of the 29 chlamydial antigens evaluated were found to prevent genital infection but not pathology and three exacerbate pathology. To further improve protection efficacy, a combinational regimen (intranasal priming + intramuscular boosting + a third intraperitoneal/subcutaneous boost) was tested. This regimen identified four infection-reducing antigens, but only one of them prevented pathology. Unfortunately, this protective antigen was not advanced further due to its amino acid sequence homology with several human molecules. Two pathology-exacerbating antigens were also found. Nevertheless, intranasal mucosal priming with viable C. muridarum in control groups consistently prevented both genital infection and pathology regardless of the subsequent boosters. Thus, screening 140 different chlamydial antigens with 21 repeated multiple times in 17 experiments failed to identify a subunit vaccine candidate but demonstrated the superiority of viable chlamydial organisms in inducing immunity against both genital infection and pathology, laying the foundation for developing a live-attenuated Chlamydia vaccine.
Collapse
Affiliation(s)
- Chunxue Lu
- Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hengyang, Hunan, China
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center, San Antonio, Texas, USA
| | - Jie Wang
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center, San Antonio, Texas, USA
- Department of Immunology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Guangming Zhong
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center, San Antonio, Texas, USA
| |
Collapse
|
8
|
Li Q, Chen S, Yan Z, Fang H, Wang Z, He C. A Novel Intranasal Vaccine With PmpGs + MOMP Induces Robust Protections Both in Respiratory Tract and Genital System Post Chlamydia psittaci Infection. Front Vet Sci 2022; 9:855447. [PMID: 35529835 PMCID: PMC9072866 DOI: 10.3389/fvets.2022.855447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Accepted: 02/14/2022] [Indexed: 11/13/2022] Open
Abstract
Chlamydia psittaci (C. psittaci) is a crucial zoonotic pathogen that causes severe respiratory and reproductive system disease in humans and animals. In our pioneer study, polymorphic membrane protein G (PmpG) mediated attachment to host cells as the adhesions and induced immunity against C. psittaci infection. We hypothesize that multiple PmpG antigens adjuvanted with Vibrio cholerae ghost (VCG) and chitosan gel might trigger full protection via the intranasal route (i.n). In the present study, 40 SPF chickens were randomly divided into four groups, including the PmpGs + MOMP group (i.n), major outer membrane protein (MOMP) group (i.n), PmpGs (Pmp17G + Pmp20G + Pmp21G) group (i.n), and control groups (VCG + chitosan gel) (i.n). Post twice immunizations, the PmpGs + MOMP group yielded highly level-specific IgG, IgA antibodies, and lymphocyte proliferation. As for cytokines, IFN-γ expression was upregulated significantly, while IL-10 concentration was downregulated in the PmpGs + MOMP group compared with other groups. Post challenge, exudate inflammations in air sacs, bacterial loads in lungs, and bacterial shedding in throat swabs were reduced significantly in the PmpGs + MOMP group. In the second experiment, 100 breeder ducks were divided into the PmpGs + MOMP group (i.n), the commercial MOMP group (via intramuscular injection, i.m), the inactivated EBs group (i.n), and the control group (i.n), 25 ducks per group. Post challenge, the reduced egg production recovered soon in the inactivated EBs group and the PmpGs + MOMP group. Moreover, the aforementioned two groups induced higher robust IgG antibodies, lymphocyte proliferation, and IFN-γ secretions than the commercial MOMP vaccine did. Postmortem, lower bacterial loads of spleens were determined in the PmpGs + MOMP group and the inactivated EBs group. However, bacterial clearance of follicular membranes and shedding from the vaginal tract were not significant differences among the three tested groups. Furthermore, the PmpGs + MOMP group induced lower inflammations in the follicles and oviducts. Based on the above evidence, the combination of PmpGs and MOMP adjuvanted with chitosan gel and VCG via intranasal route could induce full protection both in the respiratory system and genital tract post C. psittaci infection. More importantly, the combination antigens are superior to the inactivated EBs antigen due to no contamination to the environment and less genital inflammation. The combination of PmpGs + MOMP adjuvanted with VCG and chitosan gel might be a promising novel vaccine by blocking C. psittaci infection from animals to human beings.
Collapse
Affiliation(s)
- Qiang Li
- College of Life Science and Engineering, Foshan University, Foshan, China
- Key Lab of Animal Epidemiology and Zoonoses of Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Siyu Chen
- Key Lab of Animal Epidemiology and Zoonoses of Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Zhuanqiang Yan
- Wen's Group Academy, Wen's Foodstuffs Group Co., Ltd., Yunfu, China
| | - Huanxin Fang
- Wen's Group Academy, Wen's Foodstuffs Group Co., Ltd., Yunfu, China
| | - Zhanxin Wang
- Wen's Group Academy, Wen's Foodstuffs Group Co., Ltd., Yunfu, China
- *Correspondence: Zhanxin Wang
| | - Cheng He
- College of Life Science and Engineering, Foshan University, Foshan, China
- Key Lab of Animal Epidemiology and Zoonoses of Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, China Agricultural University, Beijing, China
- Cheng He
| |
Collapse
|
9
|
Johnson RM, Asashima H, Mohanty S, Shaw AC. Combining Cellular Immunology With RNAseq to Identify Novel Chlamydia T-Cell Subset Signatures. J Infect Dis 2022; 225:2033-2042. [PMID: 35172331 PMCID: PMC9159333 DOI: 10.1093/infdis/jiac051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 02/14/2022] [Indexed: 11/14/2022] Open
Abstract
Chlamydia trachomatis serovars A-L cause important diseases of the eyes and reproductive tract by infecting epithelium lining those organs. A major hurdle for vaccine trials is finding a surrogate biomarker for protective immunity. Investigational data argues for T-cell biomarker(s) reflecting mucosal adaption, cytokine polarization, B-cell help, antibacterial effector mechanisms, or some combination thereof. A human investigation and 2 mouse studies link IL-13 to protection from infection/immunopathology. We performed RNAseq on T cells resident in spleens and genital tracts of naturally immune mice. CD4 signatures were consistent with helper function that differed by site including a genital tract-specific Fgl2 signal. The genital tract CD8 signature featured IL-10 and promotion of healing/scarring with a unique transcription of granzyme A. The RNAseq data was used to refine previously published CD4γ13 and CD8γ13 transcriptomes derived from protective T-cell clones, potentially identifying practicable T-cell subset signatures for assessing Chlamydia vaccine candidates.
Collapse
Affiliation(s)
- Raymond M Johnson
- Correspondence: Raymond M. Johnson, MD, PhD, Department of Internal Medicine, Yale University School of Medicine, PO Box 208022, TAC s169, New Haven, CT 06520-8022 ()
| | - Hiromitsu Asashima
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Subhasis Mohanty
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Albert C Shaw
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
10
|
Du JJ, Zhou SH, Cheng ZR, Xu WB, Zhang RY, Wang LS, Guo J. MUC1 Specific Immune Responses Enhanced by Coadministration of Liposomal DDA/MPLA and Lipoglycopeptide. Front Chem 2022; 10:814880. [PMID: 35186882 PMCID: PMC8854779 DOI: 10.3389/fchem.2022.814880] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 01/17/2022] [Indexed: 12/11/2022] Open
Abstract
Mucin 1 (MUC1), a well-known tumor-associated antigen and attractive target for tumor immunotherapy, is overexpressed in most human epithelial adenomas with aberrant glycosylation. However, its low immunogenicity impedes the development of MUC1-targeted antitumor vaccines. In this study, we investigated three liposomal adjuvant systems containing toll-like receptor 4 (TLR4) agonist monophosphoryl lipid A (MPLA) and auxiliary lipids of different charges: cationic lipid dimethyldioctadecylammonium (DDA), neutral lipid distearoylglycerophosphocholine (DSPC) or anionic lipid dioleoylphosphatidylglycerol (DOPG), respectively. ELISA assay evidenced that the positively charged DDA/MPLA liposomes are potent immune activators, which induced remarkable levels of anti-MUC1 antibodies and exhibited robust Th1-biased immune responses. Importantly, the antibodies induced by DDA/MPLA liposomes efficiently recognized and killed MUC1-positive tumor cells through complement-mediated cytotoxicity. In addition, antibody titers in mice immunized with P2-MUC1 vaccine were significantly higher than those from mice immunized with P1-MUC1 or MUC1 vaccine, which indicated that the lipid conjugated on MUC1 antigen also played important role for immunomodulation. This study suggested that the liposomal DDA/MPLA with lipid-MUC1 is a promising antitumor vaccine, which can be used for the immunotherapy of various epithelial carcinomas represented by breast cancer.
Collapse
Affiliation(s)
- Jing-Jing Du
- Hubei Key Laboratory of Kidney Disease Pathogenesis and Intervention, College of Medicine, Hubei Polytechnic University, Huangshi, China
| | - Shi-Hao Zhou
- Key Laboratory of Pesticide and Chemical Biology of Ministry of Education, Hubei International Scientific and Technological Cooperation Base of Pesticide and Green Synthesis, International Joint Research Center for Intelligent Biosensing Technology and Health, College of Chemistry, Central China Normal University, Wuhan, China
| | - Zi-Ru Cheng
- Hubei Key Laboratory of Kidney Disease Pathogenesis and Intervention, College of Medicine, Hubei Polytechnic University, Huangshi, China
| | - Wen-Bo Xu
- Key Laboratory of Pesticide and Chemical Biology of Ministry of Education, Hubei International Scientific and Technological Cooperation Base of Pesticide and Green Synthesis, International Joint Research Center for Intelligent Biosensing Technology and Health, College of Chemistry, Central China Normal University, Wuhan, China
| | - Ru-Yan Zhang
- Key Laboratory of Pesticide and Chemical Biology of Ministry of Education, Hubei International Scientific and Technological Cooperation Base of Pesticide and Green Synthesis, International Joint Research Center for Intelligent Biosensing Technology and Health, College of Chemistry, Central China Normal University, Wuhan, China
| | - Long-Sheng Wang
- Hubei Provincial Key Laboratory of Green Materials for Light Industry, School of Materials and Chemical Engineering, Hubei University of Technology, Wuhan, China
- *Correspondence: Long-Sheng Wang, ; Jun Guo,
| | - Jun Guo
- Key Laboratory of Pesticide and Chemical Biology of Ministry of Education, Hubei International Scientific and Technological Cooperation Base of Pesticide and Green Synthesis, International Joint Research Center for Intelligent Biosensing Technology and Health, College of Chemistry, Central China Normal University, Wuhan, China
- *Correspondence: Long-Sheng Wang, ; Jun Guo,
| |
Collapse
|
11
|
Nguyen NDNT, Guleed S, Olsen AW, Follmann F, Christensen JP, Dietrich J. Th1/Th17 T cell Tissue-Resident Immunity Increases Protection, But Is Not Required in a Vaccine Strategy Against Genital Infection With Chlamydia trachomatis. Front Immunol 2021; 12:790463. [PMID: 34925371 PMCID: PMC8674352 DOI: 10.3389/fimmu.2021.790463] [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: 10/06/2021] [Accepted: 11/04/2021] [Indexed: 11/13/2022] Open
Abstract
The requirement for vaccine-induced tissue-resident immunity for protection against one or repeated infections with Chlamydia trachomatis (C.t.) is still not fully resolved. In this study, our aim was to investigate to which degree tissue-resident Th1/Th17 T cells in the genital tract (GT) could add to the protection mediated by circulating immunity. Out of several mucosal vaccine strategies, a strategy termed SIM (for simultaneous intrauterine and parenteral immunization with CAF01 adjuvanted CTH522), was superior in generating genital tract tissue-resident Th1/Th17 T cell immunity. This led to a faster and stronger local CD4 T cell response post infection, consisting of multifunctional IFNγ/TNFα-producing Th1 T cells and IFNγ/TNFα/IL-17-producing Th17 T cells, and a faster recruitment of innate immune cells. Post infection, SIM animals showed an additional significant reduction in bacterial levels compared to mice having received only a parenteral vaccine. Nevertheless, the parenteral strategy reduced bacterial levels by 75%, and interestingly, post infection, these mice generated their own vaccine-derived genital tract tissue-resident memory Th1/Th17 T cells, which upon a subsequent infection showed as fast an activation in the genital tract, as observed in SIM mice. Furthermore, in contrast to after the first infection, both groups of mice now showed a similar infection-induced boost in local vaginal IgA and IgG titers. Thus, vaccine-induced resident immunity, generated pre-infection, led to an advantage in the response against the first infection, but not the second infection, suggesting that a parenteral vaccine strategy is a suitable vaccine strategy against infections with Chlamydia trachomatis.
Collapse
Affiliation(s)
| | - Safia Guleed
- Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark
| | - Anja Weinreich Olsen
- Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark
| | - Frank Follmann
- Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark
| | | | - Jes Dietrich
- Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark
| |
Collapse
|
12
|
Darville T. Pelvic Inflammatory Disease Due to Neisseria gonorrhoeae and Chlamydia trachomatis: Immune Evasion Mechanisms and Pathogenic Disease Pathways. J Infect Dis 2021; 224:S39-S46. [PMID: 34396413 DOI: 10.1093/infdis/jiab031] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Pelvic inflammatory disease (PID) results from ascension of sexually transmitted pathogens from the lower genital tract to the uterus and/or fallopian tubes in women, with potential spread to neighboring pelvic organs. Patients may present acutely with lower abdominal or pelvic pain and pelvic organ tenderness. Many have subtle symptoms or are asymptomatic and present later with tubal factor infertility, ectopic pregnancy, or chronic pelvic pain. Neisseria gonorrhoeae and Chlamydia trachomatis are the 2 most commonly recognized PID pathogens. Their ability to survive within host epithelial cells and neutrophils highlights a need for T-cell-mediated production of interferon γ in protection. Data indicate that for both pathogens, antibody can accelerate clearance by enhancing opsonophagocytosis and bacterial killing when interferon γ is present. A study of women with N. gonorrhoeae- and/or C. trachomatis-induced PID with histologic endometritis revealed activation of myeloid cell, cell death, and innate inflammatory pathways in conjunction with dampening of T-cell activation pathways. These findings are supported by multiple studies in mouse models of monoinfection with N. gonorrhoeae or Chlamydia spp. Both pathogens exert multiple mechanisms of immune evasion that benefit themselves and each other at the expense of the host. However, similarities in host immune mechanisms that defend against these 2 bacterial pathogens instill optimism for the prospects of a combined vaccine for prevention of PID and infections in both women and men.
Collapse
Affiliation(s)
- Toni Darville
- Departments of Pediatrics and Microbiology & Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
13
|
Lanfermann C, Kohn M, Laudeley R, Rheinheimer C, Klos A. Chlamydia trachomatis Cross-Serovar Protection during Experimental Lung Reinfection in Mice. Vaccines (Basel) 2021; 9:vaccines9080871. [PMID: 34451996 PMCID: PMC8402589 DOI: 10.3390/vaccines9080871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 08/02/2021] [Accepted: 08/04/2021] [Indexed: 12/02/2022] Open
Abstract
Chlamydia trachomatis causes most bacterial sexually transmitted diseases worldwide. Different major outer membrane proteins (MOMPs) define various serovars of this intracellular pathogen: In women, D to L3 can cause urethritis, cervicitis, salpingitis, and oophoritis, and, thus, infertility. Protective immunity might be serovar-specific since chlamydial infection does not appear to induce an effective acquired immunity and reinfections occur. A better understanding of induced cross-serovar protection is essential for the selection of suitable antigens in vaccine development. In our mouse lung infection screening model, we evaluated the urogenital serovars D, E, and L2 in this regard. Seven weeks after primary infection or mock-infection, respectively, mice were infected a second time with the identical or one of the other serovars. Body weight and clinical score were monitored for 7 days. Near the peak of the second lung infection, bacterial load, myeloperoxidase, IFN-γ, and TNF-α in lung homogenate, as well as chlamydia-specific IgG levels in blood were determined. Surprisingly, compared with mice that were infected then for the first time, almost independent of the serovar combination used, all acquired parameters of disease were similarly diminished. Our reinfection study suggests that efficient cross-serovar protection could be achieved by a vaccine combining chlamydial antigens that do not include nonconserved MOMP regions.
Collapse
|
14
|
Richardson S, Medhavi F, Tanner T, Lundy S, Omosun Y, Igietseme JU, Carroll D, Eko FO. Cellular Basis for the Enhanced Efficacy of the Fms-Like Tyrosine Kinase 3 Ligand (FL) Adjuvanted VCG-Based Chlamydia abortus Vaccine. Front Immunol 2021; 12:698737. [PMID: 34249004 PMCID: PMC8264281 DOI: 10.3389/fimmu.2021.698737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 06/11/2021] [Indexed: 12/02/2022] Open
Abstract
Efficacious vaccines are needed to control genital chlamydial diseases in humans and the veterinary industry. We previously reported a C. abortus (Cab) vaccine comprising recombinant Vibrio cholerae ghosts (rVCG) expressing the conserved and immunogenic N-terminal region of the Cab polymorphic membrane protein D (rVCG-Pmp18.1) protein that protected mice against intravaginal challenge. In this study, we investigated the immunomodulatory effect of the hematopoietic progenitor activator cytokine, Fms-like tyrosine kinase 3-ligand (FL) when co-administered with the rVCG-Pmp18.1 vaccine as a strategy to enhance the protective efficacy and the potential mechanism of immunomodulation. Groups of female C57BL/6J mice were immunized and boosted twice intranasally (IN) with rVCG-PmpD18.1 with and without FL or purified rPmp18.1 or rVCG-gD2 (antigen control) or PBS (medium) per mouse. The results revealed that co-administration of the vaccine with FL enhanced antigen-specific cellular and humoral immune responses and protected against live Cab genital infection. Comparative analysis of immune cell phenotypes infiltrating mucosal and systemic immune inductive tissue sites following immunization revealed that co-administration of rVCG-Pmp18.1 with FL significantly enhanced the number of macrophages, dendritic and NK cells, γδ and NK T cells in the spleen (systemic) and iliac lymph nodes (ILN) draining the genital tract (mucosal) tissues compared to rVCG-Pmp18.1 alone. Furthermore, FL enhanced monocyte infiltration in the ILN, while CD19+ B cells and CD4+ T cells were enhanced in the spleen. These results indicate that the immunomodulatory effect of FL is associated with its ability to mobilize innate immune cells and subsequent activation of robust antigen-specific immune effectors in mucosal and systemic lymphoid tissues.
Collapse
Affiliation(s)
- Shakyra Richardson
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA, United States
| | - Fnu Medhavi
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA, United States
| | - Tayhlor Tanner
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA, United States
| | - Stephanie Lundy
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA, United States
| | - Yusuf Omosun
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA, United States
| | - Joseph U. Igietseme
- National Center for Emerging Zoonotic and Infectious Diseases, Centers for Disease Control and Prevention (CDC), Atlanta, GA, United States
| | - Darin Carroll
- National Center for Emerging Zoonotic and Infectious Diseases, Centers for Disease Control and Prevention (CDC), Atlanta, GA, United States
| | - Francis O. Eko
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA, United States
| |
Collapse
|
15
|
Pedraza L, Camargo M, Moreno-Pérez DA, Sánchez R, Del Río-Ospina L, Báez-Murcia IM, Patarroyo ME, Patarroyo MA. Identifying HLA DRB1-DQB1 alleles associated with Chlamydia trachomatis infection and in silico prediction of potentially-related peptides. Sci Rep 2021; 11:12837. [PMID: 34145318 PMCID: PMC8213839 DOI: 10.1038/s41598-021-92294-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 06/09/2021] [Indexed: 02/05/2023] Open
Abstract
HLA class II (HLA-II) genes' polymorphism influences the immune response to Chlamydia trachomatis (Ct), it is considered a sexually transmitted infection. However, associations between HLA-II alleles and Ct-infection have been little explored in humans; this study was thus aimed at determining HLA-DRB1-DQB1 alleles/haplotypes' effect on Ct-infection outcome in a cohort of Colombian women. Cervical sample DNA was used as template for detecting Ct by PCR and typing HLA-DRB1-DQB1 alleles/haplotypes by Illumina MiSeq sequencing. Survival models were adjusted for identifying the alleles/haplotypes' effect on Ct-outcome; bioinformatics tools were used for predicting secreted bacterial protein T- and B-cell epitopes. Sixteen HLA-DRB1 alleles having a significant effect on Ct-outcome were identified in the 262 women analysed. DRB1*08:02:01G and DRB1*12:01:01G were related to infection-promoting events. Only the DQB1*05:03:01G allele related to clearance/persistence events was found for HLA-DQB1. HLA-DRB1 allele homozygous women were associated with events having a lower probability of clearance and/or early occurrence of persistence. Twenty-seven peptides predicted in silico were associated with protective immunity against Ct; outer membrane and polymorphic membrane protein-derived peptides had regions having dual potential for being T- or B-cell epitopes. This article describes HLA-DRB1-DQB1 alleles/haplotypes related to Ct-infection resolution and the peptides predicted in silico which might probably be involved in host immune response. The data provides base information for developing future studies leading to the development of effective prevention measures against Ct-infection.
Collapse
Affiliation(s)
- Leidy Pedraza
- Molecular Biology and Immunology Department, Fundación Instituto de Inmunología de Colombia (FIDIC), 111321, Bogotá D.C., Colombia
- MSc Programme in Microbiology, Universidad Nacional de Colombia, 111321, Bogotá D.C., Colombia
| | - Milena Camargo
- Molecular Biology and Immunology Department, Fundación Instituto de Inmunología de Colombia (FIDIC), 111321, Bogotá D.C., Colombia
- Animal Science Faculty, Universidad de Ciencias Aplicadas y Ambientales (U.D.C.A), 111166, Bogotá D.C., Colombia
| | - Darwin A Moreno-Pérez
- Molecular Biology and Immunology Department, Fundación Instituto de Inmunología de Colombia (FIDIC), 111321, Bogotá D.C., Colombia
- Animal Science Faculty, Universidad de Ciencias Aplicadas y Ambientales (U.D.C.A), 111166, Bogotá D.C., Colombia
| | - Ricardo Sánchez
- Molecular Biology and Immunology Department, Fundación Instituto de Inmunología de Colombia (FIDIC), 111321, Bogotá D.C., Colombia
- Faculty of Medicine, Universidad Nacional de Colombia, 111321, Bogotá D.C., Colombia
| | - Luisa Del Río-Ospina
- Molecular Biology and Immunology Department, Fundación Instituto de Inmunología de Colombia (FIDIC), 111321, Bogotá D.C., Colombia
| | - Indira M Báez-Murcia
- Molecular Biology and Immunology Department, Fundación Instituto de Inmunología de Colombia (FIDIC), 111321, Bogotá D.C., Colombia
| | - Manuel E Patarroyo
- Molecular Biology and Immunology Department, Fundación Instituto de Inmunología de Colombia (FIDIC), 111321, Bogotá D.C., Colombia
- Faculty of Medicine, Universidad Nacional de Colombia, 111321, Bogotá D.C., Colombia
- Health Sciences Division, Main Campus, Universidad Santo Tomás, 110231, Bogotá D.C., Colombia
| | - Manuel A Patarroyo
- Molecular Biology and Immunology Department, Fundación Instituto de Inmunología de Colombia (FIDIC), 111321, Bogotá D.C., Colombia.
- Faculty of Medicine, Universidad Nacional de Colombia, 111321, Bogotá D.C., Colombia.
- Health Sciences Division, Main Campus, Universidad Santo Tomás, 110231, Bogotá D.C., Colombia.
| |
Collapse
|
16
|
Prophylactic Multi-Subunit Vaccine against Chlamydia trachomatis: In Vivo Evaluation in Mice. Vaccines (Basel) 2021; 9:vaccines9060609. [PMID: 34204170 PMCID: PMC8226540 DOI: 10.3390/vaccines9060609] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 05/31/2021] [Accepted: 06/03/2021] [Indexed: 01/08/2023] Open
Abstract
Chlamydia trachomatis is the most frequent sexually-transmitted disease-causing bacterium. Urogenital serovars of this intracellular pathogen lead to urethritis and cervicitis. Ascending infections result in pelvic inflammatory disease, salpingitis, and oophoritis. One of 200 urogenital infections leads to tubal infertility. Serovars A–C cause trachoma with visual impairment. There is an urgent need for a vaccine. We characterized a new five-component subunit vaccine in a mouse vaccination-lung challenge infection model. Four recombinant Pmp family-members and Ctad1 from C. trachomatis serovar E, all of which participate in adhesion and binding of chlamydial elementary bodies to host cells, were combined with the mucosal adjuvant cyclic-di-adenosine monophosphate. Intranasal application led to a high degree of cross-serovar protection against urogenital and ocular strains of C. trachomatis, which lasted at least five months. Critical evaluated parameters were body weight, clinical score, chlamydial load, a granulocyte marker and the cytokines IFN-γ/TNF-α in lung homogenate. Vaccine antigen-specific antibodies and a mixed Th1/Th2/Th17 T cell response with multi-functional CD4+ and CD8+ T cells correlate with protection. However, serum-transfer did not protect the recipients suggesting that circulating antibodies play only a minor role. In the long run, our new vaccine might help to prevent the feared consequences of human C. trachomatis infections.
Collapse
|
17
|
Karunakaran KP, Yu H, Jiang X, Chan QWT, Foster LJ, Johnson RM, Brunham RC. Discordance in the Epithelial Cell-Dendritic Cell Major Histocompatibility Complex Class II Immunoproteome: Implications for Chlamydia Vaccine Development. J Infect Dis 2020; 221:841-850. [PMID: 31599954 DOI: 10.1093/infdis/jiz522] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 10/05/2019] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Chlamydia trachomatis and Chlamydia muridarum are intracellular bacterial pathogens of mucosal epithelial cells. CD4 T cells and major histocompatibility complex (MHC) class II molecules are essential for protective immunity against them. Antigens presented by dendritic cells (DCs) expand naive pathogen-specific T cells (inductive phase), whereas antigens presented by epithelial cells identify infected epithelial cells as targets during the effector phase. We previously showed that DCs infected by C trachomatis or C muridarum present epitopes from a limited spectrum of chlamydial proteins recognized by Chlamydia-specific CD4 T cells from immune mice. METHODS We hypothesized that Chlamydia-infected DCs and epithelial cells present overlapping sets of Chlamydia-MHC class II epitopes to link inductive and effector phases to generate protective immunity. We tested that hypothesis by infecting an oviductal epithelial cell line with C muridarum, followed by immunoaffinity isolation and sequencing of MHC class I- and II-bound peptides. RESULTS We identified 26 class I-bound and 4 class II-bound Chlamydia-derived peptides from infected epithelial cells. We were surprised to find that none of the epithelial cell class I- and class II-bound chlamydial peptides overlapped with peptides presented by DCs. CONCLUSIONS We suggest the discordance between the DC and epithelial cell immunoproteomes has implications for delayed clearance of Chlamydia and design of a Chlamydia vaccine.
Collapse
Affiliation(s)
- Karuna P Karunakaran
- Vaccine Research Laboratory, University of British Columbia Centre for Disease Control, Vancouver, British Columbia, Canada
| | - Hong Yu
- Vaccine Research Laboratory, University of British Columbia Centre for Disease Control, Vancouver, British Columbia, Canada
| | - Xiaozhou Jiang
- Vaccine Research Laboratory, University of British Columbia Centre for Disease Control, Vancouver, British Columbia, Canada
| | - Queenie W T Chan
- Department of Biochemistry and Molecular Biology, Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada
| | - Leonard J Foster
- Department of Biochemistry and Molecular Biology, Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada
| | - Raymond M Johnson
- Section of Infectious Diseases, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Robert C Brunham
- Vaccine Research Laboratory, University of British Columbia Centre for Disease Control, Vancouver, British Columbia, Canada
| |
Collapse
|
18
|
Comparison of Chlamydia outer membrane complex to recombinant outer membrane proteins as vaccine. Vaccine 2020; 38:3280-3291. [PMID: 32151463 DOI: 10.1016/j.vaccine.2020.02.059] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 01/02/2020] [Accepted: 02/18/2020] [Indexed: 12/22/2022]
Abstract
The Chlamydial outer membrane complex (COMC) from the elementary body (EB) is a protein rich insoluble outer membrane shell from which cytosolic proteins have been extracted with detergent. In this study we conducted mass spectrometry experiments to detect proteins in the COMC prepared from C. muridarum EB. Proteomic analysis showed that the COMC contained 75 proteins that included 10 outer membrane proteins (OMPs) such as the major outer membrane protein (MOMP) and polymorphic membrane proteins (Pmps) that were previously identified as CD4 T cell vaccine candidates. We tested the vaccine efficacy of COMC in comparison to individual or combination of recombinant OMPs formulated with Th1 polarizing adjuvant DDA/MPL in two murine genital tract models (C. muridarum and C. trachomatis) by measuring organismal shedding, tubal pathology and immune responses including neutralizing antibodies. In the C. muridarum model, the COMC vaccine generated broadly reactive immune responses against multiple outer membrane proteins, high levels of EB binding and neutralizing antibody and exhibited superior protection against genital infection and pathology when compared to the recombinant PmpG vaccine. Denaturing the COMC by boiling significantly reduced protection. In the C. trachomatis model, the COMC vaccine also conferred greater protection compared to individual or multiple recombinant outer membrane proteins. Immunization with multiple COMCs from C. trachomatis serovars D, F and J generated neutralizing antibodies against multiple C. trachomatis serovars. We conclude that broader immunogenicity and generation of neutralizing antibody may explain the superior efficacy of COMC vaccine. The study suggests that conformationally intact proteins will be necessary for a successful recombinant OMP vaccine.
Collapse
|
19
|
Nguyen NDNT, Olsen AW, Lorenzen E, Andersen P, Hvid M, Follmann F, Dietrich J. Parenteral vaccination protects against transcervical infection with Chlamydia trachomatis and generate tissue-resident T cells post-challenge. NPJ Vaccines 2020; 5:7. [PMID: 31993218 PMCID: PMC6978417 DOI: 10.1038/s41541-020-0157-x] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 12/20/2019] [Indexed: 11/21/2022] Open
Abstract
The optimal protective immunity against Chlamydia trachomatis (C.t.) is still not fully resolved. One of the unresolved issues concerns the importance of resident immunity, since a recent study showed that optimal protection against a transcervical (TC) infection required genital tissue-resident memory T cells. An important question in the Chlamydia field is therefore if a parenteral vaccine strategy, inducing only circulating immunity primed at a nonmucosal site, should be pursued by Chlamydia vaccine developers. To address this question we studied the protective efficacy of a parenteral Chlamydia vaccine, formulated in the Th1/Th17 T cell-inducing adjuvant CAF01. We found that a parenteral vaccination induced significant protection against a TC infection and against development of chronic pathology. Protection correlated with rapid recruitment of Th1/Th17 T cells to the genital tract (GT), which efficiently prevented infection-driven generation of low quality Th1 or Th17 T cells, and instead maintained a pool of high quality multifunctional Th1/Th17 T cells in the GT throughout the infection. After clearance of the infection, a pool of these cells settled in the GT as tissue-resident Th1 and Th17 cells expressing CD69 but not CD103, CD49d, or CCR7, where they responded rapidly to a reinfection. These results show that a nonmucosal parenteral strategy inducing Th1 and Th17 T cells mediates protection against both infection with C.t. as well as development of chronic pathology, and lead to post-challenge protective tissue-resident memory immunity in the genital tract.
Collapse
Affiliation(s)
| | - Anja W Olsen
- 1Statens Serum Institut, Department for Infectious Disease Immunology, Copenhagen, Denmark
| | - Emma Lorenzen
- 1Statens Serum Institut, Department for Infectious Disease Immunology, Copenhagen, Denmark
| | - Peter Andersen
- 1Statens Serum Institut, Department for Infectious Disease Immunology, Copenhagen, Denmark
| | - Malene Hvid
- 2Department of Biomedicine and Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Frank Follmann
- 1Statens Serum Institut, Department for Infectious Disease Immunology, Copenhagen, Denmark
| | - Jes Dietrich
- 1Statens Serum Institut, Department for Infectious Disease Immunology, Copenhagen, Denmark
| |
Collapse
|
20
|
Pais R, Omosun Y, Igietseme JU, Fujihashi K, Eko FO. Route of Vaccine Administration Influences the Impact of Fms-Like Tyrosine Kinase 3 Ligand (Flt3L) on Chlamydial-Specific Protective Immune Responses. Front Immunol 2019; 10:1577. [PMID: 31333682 PMCID: PMC6621642 DOI: 10.3389/fimmu.2019.01577] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 06/24/2019] [Indexed: 11/24/2022] Open
Abstract
We tested the hypothesis that the impact of the Fms-like tyrosine kinase 3-ligand (Flt3L; FL) on recombinant Vibrio cholerae ghost (rVCG) vaccine-induced chlamydial immunity is influenced by route of vaccine delivery. Female C57BL/6J mice were immunized rectally (IR) or intramuscularly (IM) with rVCG co-expressing the Chlamydia trachomatis PmpD and PorB proteins (rVCG- PmpD/PorB) with and without FL or glycoprotein D of HSV-2 (rVCG-gD2) as antigen control. Vaccine evaluation was based on measurement of T cell proliferation, Th1/Th2 cytokine, and humoral responses at systemic and mucosal compartments, and protection against intravaginal challenge infection. Results revealed that high levels of CD4+ T cell-mediated and humoral immune responses, were elicited in mice as a function of both IR and IM immunization. Unexpectedly, co-administration of vaccine with FL enhanced specific Th1-type cytokine levels and T cell proliferative responses following IR but not IM immunization. While administration of vaccine with FL enhanced the specific mucosal and systemic IgA antibody responses following both immunization routes, IgG2c responses were not enhanced following IR delivery. The vaccine-induced immune effectors protected mice against live heterologous C. muridarum infection irrespective of route of vaccine administration, with the regimen incorporating FL having a protective advantage. Further evaluation showed that protection afforded by the FL adjuvanted vaccine was facilitated by CD4+ T cells, as indicated by reduction in the intensity and duration of genital chlamydial shedding by naïve mice following adoptive transfer of immune CD4+ T cells. Taken together, the results indicate that comparable protective immunity, which is enhanced by co-delivery with FL, is elicited in the female genital tract against Chlamydia infection after mucosal and systemic administration, highlighting the ability of FL to function as an effective immunostimulator at both mucosal and systemic sites. The differential modulation of humoral and cellular immune responses, and protective immunity afforded by the FL adjuvanted vaccine following IR administration indicates that the immunomodulatory impact of FL on chlamydial-specific immunity is influenced by the route of vaccine administration. Thus, targeting of VCG-based vaccines to antigen presenting cells by co-delivery with FL is a feasible immunization approach for inducing effective chlamydial immunity in the female genital tract.
Collapse
Affiliation(s)
- Roshan Pais
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA, United States
| | - Yusuf Omosun
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA, United States
| | - Joseph U. Igietseme
- Molecular Pathogenesis Laboratory, Centers for Disease Control and Prevention, Atlanta, GA, United States
| | - Kohtaro Fujihashi
- Department of Pediatric Dentistry, Institute of Oral Health Research, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Francis O. Eko
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA, United States
| |
Collapse
|
21
|
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]
|
22
|
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
|
23
|
|
24
|
Brunham RC. Perspective: my 37 year journey through Chlamydia research: Chlamydia antigen analysis using monoclonal antibodies and major histocompatibility complex molecules. Pathog Dis 2018; 75:4056143. [PMID: 28961985 DOI: 10.1093/femspd/ftx089] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 07/27/2017] [Indexed: 01/08/2023] Open
Abstract
Chlamydia antigen analysis enables understanding of disease pathogenesis, facilitates development of diagnostic immunoassays and is essential to the design of a subunit Chlamydia trachomatis vaccine. Using an autobiographical narrative, I review over three decades of antigen analysis research findings coming from my research laboratory and provide an outlook to the broader field of Chlamydia seroepidemiology and vaccinology. Based on the experiences of my scientific career I conclude with thoughts for young scientists newly entering the Chlamydia research field.
Collapse
Affiliation(s)
- Robert C Brunham
- British Columbia Centre for Disease Control, University of British Columbia, Vancouver, BC V6 1N4, Canada
| |
Collapse
|
25
|
B Cell Presentation of Chlamydia Antigen Selects Out Protective CD4γ13 T Cells: Implications for Genital Tract Tissue-Resident Memory Lymphocyte Clusters. Infect Immun 2018; 86:IAI.00614-17. [PMID: 29158429 DOI: 10.1128/iai.00614-17] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Accepted: 11/10/2017] [Indexed: 12/31/2022] Open
Abstract
Surveillance and defense of the enormous mucosal interface with the nonsterile world are critical to protecting the host from a wide range of pathogens. Chlamydia trachomatis is an intracellular bacterial pathogen that replicates almost exclusively in the epithelium of the reproductive tract. The fallopian tubes and vagina are poorly suited to surveillance and defense, with limited immune infrastructure positioned near the epithelium. However, a dynamic process during clearing primary infections leaves behind new lymphoid clusters immediately beneath the epithelium. These memory lymphocyte clusters (MLCs) harboring tissue-resident memory (Trm) T cells are presumed to play an important role in protection from subsequent infections. Histologically, human Chlamydia MLCs have prominent B cell populations. We investigated the status of genital tract B cells during C. muridarum infections and the nature of T cells recovered from immune mice using immune B cells as antigen-presenting cells (APCs). These studies revealed a genital tract plasma B cell population and a novel genital tract CD4 T cell subset producing both gamma interferon (IFN-γ) and interleukin-13 (IL-13). A panel of CD4 T cell clones and microarray analysis showed that the molecular fingerprint of CD4γ13 T cells includes a Trm-like transcriptome. Adoptive transfer of a Chlamydia-specific CD4γ13 T cell clone completely prevented oviduct immunopathology without accelerating bacterial clearance. Existence of a CD4γ13 T cell subset provides a plausible explanation for the observation that human peripheral blood mononuclear cell (PBMC) Chlamydia-specific IFN-γ and IL-13 responses predict resistance to reinfection.
Collapse
|
26
|
Pan Q, Zhang Q, Chu J, Pais R, Liu S, He C, Eko FO. Chlamydia abortus Pmp18.1 Induces IL-1β Secretion by TLR4 Activation through the MyD88, NF-κB, and Caspase-1 Signaling Pathways. Front Cell Infect Microbiol 2017; 7:514. [PMID: 29326885 PMCID: PMC5741698 DOI: 10.3389/fcimb.2017.00514] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Accepted: 11/29/2017] [Indexed: 12/16/2022] Open
Abstract
The polymorphic membrane protein D (Pmp18D) is a 160-kDa outer membrane protein that is conserved and plays an important role in Chlamydia abortus pathogenesis. We have identified an N-terminal fragment of Pmp18D (designated Pmp18.1) as a possible subunit vaccine antigen. In this study, we evaluated the vaccine potential of Pmp18.1 by investigating its ability to induce innate immune responses in dendritic cells and the signaling pathway(s) involved in rPmp18.1-induced IL-1β secretion. We next investigated the immunomodulatory impact of VCG, in comparison with the more established Th1-promoting adjuvants, CpG and FL, on rPmp18.1-mediated innate immune activation. Finally, the effect of siRNA targeting TLR4, MyD88, NF-κB p50, and Caspase-1 mRNA in DCs on IL-1β cytokine secretion was also investigated. Bone marrow-derived dendritic cells (BMDCs) were stimulated with rPmp18.1 in the presence or absence of VCG or CpG or FL and the magnitude of cytokines produced was assessed using a multiplex cytokine ELISA assay. Expression of costimulatory molecules and Toll-like receptors (TLRs) was analyzed by flow cytometry. Quantitation of intracellular levels of myeloid differentiation factor 88 (MyD88), nuclear factor kappa beta (NF-κB p50/p65), and Caspase-1 was evaluated by Western immunoblotting analysis while NF-κB p65 nuclear translocation was assessed by confocal microscopy. The results showed DC stimulation with rPmp18.1 provoked the secretion of proinflammatory cytokines and upregulated expression of TLRs and co-stimulatory molecules associated with DC maturation. These responses were significantly (p ≤ 0.001) enhanced by VCG but not CpG or FL. In addition, rPmp18.1 activated the expression of MyD88, NF-κB p50, and Caspase-1 as well as the nuclear expression of NF-κB p65 in treated DCs. Furthermore, targeting TLR4, MyD88, NF-κB p50, and Caspase-1 mRNA in BMDCs with siRNA significantly reduced their expression levels, resulting in decreased IL-1β cytokine secretion, strongly suggesting their involvement in the rPmp18.1-induced IL-1β cytokine secretion. Taken together, these results indicate that C. abortus Pmp18.1 induces IL-1β secretion by TLR4 activation through the MyD88, NF-κB as well as the Caspase-1 signaling pathways and may be a potential C. abortus vaccine candidate. The vaccine potential of Pmp18.1 will subsequently be evaluated in an appropriate animal model, using VCG as an immunomodulator, following immunization and challenge.
Collapse
Affiliation(s)
- Qing Pan
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA, United States.,Key Lab of Animal Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Qiang Zhang
- Key Lab of Animal Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Jun Chu
- Key Lab of Animal Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Roshan Pais
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA, United States
| | - Shanshan Liu
- Key Lab of Animal Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Cheng He
- Key Lab of Animal Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Francis O Eko
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA, United States
| |
Collapse
|
27
|
Poston TB, Qu Y, Girardi J, O'Connell CM, Frazer LC, Russell AN, Wall M, Nagarajan UM, Darville T. A Chlamydia-Specific TCR-Transgenic Mouse Demonstrates Th1 Polyfunctionality with Enhanced Effector Function. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2017; 199:2845-2854. [PMID: 28855311 PMCID: PMC5770186 DOI: 10.4049/jimmunol.1700914] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 08/07/2017] [Indexed: 02/06/2023]
Abstract
Chlamydia is responsible for millions of new infections annually, and current efforts focus on understanding cellular immunity for targeted vaccine development. The Chlamydia-specific CD4 T cell response is characterized by the production of IFN-γ, and polyfunctional Th1 responses are associated with enhanced protection. A major limitation in studying these responses is the paucity of tools available for detection, quantification, and characterization of polyfunctional Ag-specific T cells. We addressed this problem by developing a TCR-transgenic (Tg) mouse with CD4 T cells that respond to a common Ag in Chlamydia muridarum and Chlamydia trachomatis Using an adoptive-transfer approach, we show that naive Tg CD4 T cells become activated, proliferate, migrate to the infected tissue, and acquire a polyfunctional Th1 phenotype in infected mice. Polyfunctional Tg Th1 effectors demonstrated enhanced IFN-γ production compared with polyclonal cells, protected immune-deficient mice against lethality, mediated bacterial clearance, and orchestrated an anamnestic response. Adoptive transfer of Chlamydia-specific CD4 TCR-Tg T cells with polyfunctional capacity offers a powerful approach for analysis of protective effector and memory responses against chlamydial infection and demonstrates that an effective monoclonal CD4 T cell response may successfully guide subunit vaccination strategies.
Collapse
Affiliation(s)
- Taylor B Poston
- Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599; and
| | - Yanyan Qu
- Department of Pediatrics, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA 15224
| | - Jenna Girardi
- Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599; and
| | - Catherine M O'Connell
- Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599; and
| | - Lauren C Frazer
- Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599; and
| | - Ali N Russell
- Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599; and
| | - McKensie Wall
- Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599; and
| | - Uma M Nagarajan
- Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599; and
| | - Toni Darville
- Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599; and
| |
Collapse
|
28
|
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
|
29
|
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
|
30
|
Müller T, Becker E, Stallmann S, Waldhuber A, Römmler-Dreher F, Albrecht S, Mohr F, Hegemann JH, Miethke T. Vaccination with the polymorphic membrane protein A reduces Chlamydia muridarum induced genital tract pathology. Vaccine 2017; 35:2801-2810. [PMID: 28413133 DOI: 10.1016/j.vaccine.2017.04.017] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Revised: 03/22/2017] [Accepted: 04/09/2017] [Indexed: 01/16/2023]
Abstract
Chlamydia trachomatis serovars D-K are one of the most frequent causes of sexually transmitted infections of the female genital tract, with possible complications such as hydrosalpinx, pelvic inflammatory disease, extra-uterine gravidity or infertility. We used the murine genital tract infection model with C. muridarum for vaccination studies and found that more than 70% of the infected mice suffered from uterus dilatations and/or hydrosalpinx. Systemic consequences of the vaginal infection were apparent by splenomegaly ten to fifteen days post infection. While cultivable microorganisms were detectable for the first 23days post infection, the first lesions of the genital tract developed at day 15, however, many lesions occurred later in the absence of cultivable bacteria. Lesions were not accompanied by pro-inflammatory cytokines such as IFNɣ, TNF and IL-6, since these cytokines were almost undetectable in the genital tract 43days post infection. To prevent genital tract lesions, we vaccinated mice with the polymorphic membrane protein (Pmp) A in combination with CpG-ODN 1826 as adjuvant. The vaccine lowered the chlamydial burden and the differences were significant at day 10 post infection but not later. More importantly the vaccine decreased the rate and severity of genital tract lesions. Interestingly, control vaccination with the protein ovalbumin plus CpG-ODN 1826 enhanced significantly the severity but not the rate of pathologic lesions, which was presumably caused by the activation of innate immune responses by the adjuvant in the absence of a C. muridarum-specific adaptive immune response. In summary, vaccination with recombinant PmpA plus CpG-ODN 1826 significantly reduced C. muridarum-induced tissue damage, however, CpG-ODN 1826 may aggravate C. muridarum-induced tissue injuries in the absence of a protective antigen.
Collapse
Affiliation(s)
- Tina Müller
- Institut für Medizinische Mikrobiologie, Immunologie und Hygiene, Technische Universität München, Trogerstr. 30, 81675 München, Germany
| | - Elisabeth Becker
- Funktionelle Genomforschung der Mikroorganismen, Heinrich-Heine-Universität Düsseldorf, Universitätsstraβe 1, 40225 Düsseldorf, Germany
| | - Sonja Stallmann
- Funktionelle Genomforschung der Mikroorganismen, Heinrich-Heine-Universität Düsseldorf, Universitätsstraβe 1, 40225 Düsseldorf, Germany
| | - Anna Waldhuber
- Institut für Medizinische Mikrobiologie, Immunologie und Hygiene, Technische Universität München, Trogerstr. 30, 81675 München, Germany
| | - Franziska Römmler-Dreher
- Institut für Medizinische Mikrobiologie, Immunologie und Hygiene, Technische Universität München, Trogerstr. 30, 81675 München, Germany
| | - Simone Albrecht
- Institute of Medical Microbiology and Hygiene, Medical Faculty of Mannheim, University of Heidelberg, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany
| | - Fabian Mohr
- Institut für Medizinische Mikrobiologie, Immunologie und Hygiene, Technische Universität München, Trogerstr. 30, 81675 München, Germany
| | - Johannes H Hegemann
- Funktionelle Genomforschung der Mikroorganismen, Heinrich-Heine-Universität Düsseldorf, Universitätsstraβe 1, 40225 Düsseldorf, Germany
| | - Thomas Miethke
- Institute of Medical Microbiology and Hygiene, Medical Faculty of Mannheim, University of Heidelberg, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany.
| |
Collapse
|
31
|
Update on Chlamydia trachomatis Vaccinology. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2017; 24:CVI.00543-16. [PMID: 28228394 DOI: 10.1128/cvi.00543-16] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Attempts to produce a vaccine to protect against Chlamydia trachomatis-induced trachoma were initiated more than 100 years ago and continued for several decades. Using whole organisms, protective responses were obtained. However, upon exposure to C. trachomatis, disease exacerbation developed in some immunized individuals, precluding the implementation of the vaccine. Evidence of the role of C. trachomatis as a sexually transmitted pathogen started to emerge in the 1960s, and it soon became evident that it can cause acute infections and long-term sequelae in women, men, and newborns. The main focus of this minireview is to summarize recent findings and discuss formulations, including antigens, adjuvants, routes, and delivery systems for immunization, primarily explored in the female mouse model, with the goal of implementing a vaccine against C. trachomatis genital infections.
Collapse
|
32
|
Pal S, Favaroni A, Tifrea DF, Hanisch PT, Luczak SET, Hegemann JH, de la Maza LM. Comparison of the nine polymorphic membrane proteins of Chlamydia trachomatis for their ability to induce protective immune responses in mice against a C. muridarum challenge. Vaccine 2017; 35:2543-2549. [PMID: 28385608 DOI: 10.1016/j.vaccine.2017.03.070] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 03/06/2017] [Accepted: 03/22/2017] [Indexed: 01/24/2023]
Abstract
OBJECTIVES To test vaccines, formulated with novel antigens, to protect mice against Chlamydia infections. METHODS To determine the ability of polymorphic membrane proteins (Pmps) to induce cross-species protective immune responses, recombinant fragments from all nine C. trachomatis serovar E Pmps were used to vaccinate BALB/c mice utilizing CpG-1826 and Montanide ISA 720 as adjuvants. C. muridarum recombinant MOMP and PBS, formulated with the same adjuvants, were used as positive and negative controls, respectively. Mice were challenged intranasally with 104 inclusion-forming units (IFU) of C. muridarum. Animals were weighed daily and at 10days post-challenge, they were euthanized, their lungs harvested, weighed and the number of chlamydial IFU counted. RESULTS Following vaccination the nine Pmps elicited immune responses. Based on body weight changes, or number of IFU recovered from lungs, mice vaccinated with Pmp C, G or H were the best protected. For example, over the 10-day period, the negative control group vaccinated with PBS lost significantly more body weight than mice immunized with PmpC or G (P<0.05). C. muridarum MOMP vaccinated mice were better protected against body weight losses than any group immunized with Pmps. Also, the median number of IFU recovered from the lungs of mice vaccinated with PmpC (72×106) or PmpH (61×106) was significantly less than from mice immunized with PBS (620×106; P<0.05). As determined by the number of IFU, all Pmps elicited less protection than C. muridarum MOMP (0.078×106 IFU; P<0.05). CONCLUSIONS This is the first time PmpC has been shown to elicit cross-species protection against a respiratory challenge. Additional work with Pmps C, G and H is recommended to determine their ability to protect animal models against genital and ocular challenges.
Collapse
Affiliation(s)
- Sukumar Pal
- Department of Pathology and Laboratory Medicine, University of California, Irvine, Irvine, CA, USA
| | - Alison Favaroni
- Institut für Funktionelle Genomforschung der Mikroorganismen, Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany
| | - Delia F Tifrea
- Department of Pathology and Laboratory Medicine, University of California, Irvine, Irvine, CA, USA
| | - Philipp T Hanisch
- Institut für Funktionelle Genomforschung der Mikroorganismen, Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany
| | - Sören E T Luczak
- Institut für Funktionelle Genomforschung der Mikroorganismen, Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany
| | - Johannes H Hegemann
- Institut für Funktionelle Genomforschung der Mikroorganismen, Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany
| | - Luis M de la Maza
- Department of Pathology and Laboratory Medicine, University of California, Irvine, Irvine, CA, USA.
| |
Collapse
|
33
|
Pal S, Tifrea DF, Follmann F, Andersen P, de la Maza LM. The cationic liposomal adjuvants CAF01 and CAF09 formulated with the major outer membrane protein elicit robust protection in mice against a Chlamydia muridarum respiratory challenge. Vaccine 2017; 35:1705-1711. [PMID: 28238632 DOI: 10.1016/j.vaccine.2017.02.020] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Revised: 01/20/2017] [Accepted: 02/09/2017] [Indexed: 12/31/2022]
Abstract
Two cationic liposomal adjuvants CAF01 and CAF09 were formulated with the native or the recombinant Chlamydia muridarum major outer membrane protein (nMOMP and rMOMP). BALB/c mice were immunized with the four vaccine formulations using the subcutaneous followed by the intranasal (i.n.) routes. As positive controls mice were inoculated i.n. with live C. muridarum and negative controls received i.n. minimal essential medium (MEM). Four weeks after the last immunization mice were challenged i.n. with 104 inclusion forming units (IFU) of C. muridarum. Following the challenge the mice were weighed daily. At 10days post-challenge the mice were euthanized, their lungs weighed and the number of C. muridarum IFU determined. Serum collected the day before the challenge showed that all four groups of mice immunized with CAF01, or CAF09 and MOMP had significant C. muridarum-specific antibody titers. As determined by a T-cell lymphoproliferative assay, these four groups of mice also mounted robust cell mediated immune responses with high production of IFN-γ and IL17 and low levels of IL-4. Following the challenge the four groups of mice lost significantly less body weight than the MEM-immunized group. Lungs of mice vaccinated with CAF01, or CAF09, and nMOMP were significantly lighter than those from mice immunized using rMOMP. The number of IFU recovered from the lungs of mice vaccinated with CAF01, or CAF09, and nMOMP was similar to the number of IFU recovered from mice immunized with live EB. Mice that received rMOMP had significantly higher numbers of IFU than other groups. In conclusion, CAF01 and CAF09 elicited very robust protective humoral and cellular immune responses and were equally effective at adjuntavizing the C. muridarum MOMP. Mice vaccinated with nMOMP were significantly better protected than those immunized with rMOMP, indicative of the importance of the structural conformation of this antigen in protection.
Collapse
MESH Headings
- Adjuvants, Immunologic/administration & dosage
- Administration, Intranasal
- Animals
- Antibodies, Bacterial/blood
- Bacterial Outer Membrane Proteins/immunology
- Bacterial Vaccines/administration & dosage
- Bacterial Vaccines/immunology
- Body Weight
- Cell Proliferation
- Chlamydia Infections/pathology
- Chlamydia Infections/prevention & control
- Chlamydia muridarum/immunology
- Cytokines/metabolism
- Disease Models, Animal
- Injections, Subcutaneous
- Liposomes/administration & dosage
- Lung/pathology
- Mice, Inbred BALB C
- Pneumonia, Bacterial/pathology
- Pneumonia, Bacterial/prevention & control
- T-Lymphocytes/immunology
- Treatment Outcome
- Vaccines, Subunit/administration & dosage
- Vaccines, Subunit/immunology
Collapse
Affiliation(s)
- Sukumar Pal
- Department of Pathology and Laboratory Medicine, Medical Sciences I, Room D440, University of California, Irvine, Irvine, CA 92697-4800, USA
| | - Delia F Tifrea
- Department of Pathology and Laboratory Medicine, Medical Sciences I, Room D440, University of California, Irvine, Irvine, CA 92697-4800, USA
| | - Frank Follmann
- Department of Infectious Disease Immunology, Adjuvant Research, Staten Serum Institute, Copenhagen, Denmark
| | - Peter Andersen
- Department of Infectious Disease Immunology, Adjuvant Research, Staten Serum Institute, Copenhagen, Denmark
| | - Luis M de la Maza
- Department of Pathology and Laboratory Medicine, Medical Sciences I, Room D440, University of California, Irvine, Irvine, CA 92697-4800, USA.
| |
Collapse
|
34
|
Shaw JH, Behar AR, Snider TA, Allen NA, Lutter EI. Comparison of Murine Cervicovaginal Infection by Chlamydial Strains: Identification of Extrusions Shed In vivo. Front Cell Infect Microbiol 2017; 7:18. [PMID: 28217555 PMCID: PMC5289954 DOI: 10.3389/fcimb.2017.00018] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Accepted: 01/16/2017] [Indexed: 12/23/2022] Open
Abstract
Chlamydia trachomatis is the leading cause of bacterial sexually transmitted infections (STIs) and preventable blindness. Untreated, asymptomatic infection as well as frequent re-infection are common and may drive pelvic inflammatory disease, ectopic pregnancy, and infertility. In vivo models of chlamydial infection continue to be instrumental in progress toward a vaccine and further elucidating the pathogenesis of this intracellular bacterium, however significant gaps in our understanding remain. Chlamydial host cell exit occurs via two mechanisms, lysis and extrusion, although the latter has yet to be reported in vivo and its biological role is unclear. The objective of this study was to investigate whether chlamydial extrusions are shed in vivo following infection with multiple strains of Chlamydia. We utilized an established C3H/HeJ murine cervicovaginal infection model with C. trachomatis serovars D and L2 and the Chlamydia muridarum strain MoPn to monitor the (i) time course of infection and mode of host cell exit, (ii) mucosal and systemic immune response to infection, and (iii) gross and histopathology following clearance of active infection. The key finding herein is the first identification of chlamydial extrusions shed from host cells in an in vivo model. Extrusions, a recently appreciated mode of host cell exit and potential means of dissemination, had been previously observed solely in vitro. The results of this study demonstrate that chlamydial extrusions exist in vivo and thus warrant further investigation to determine their role in chlamydial pathogenesis.
Collapse
Affiliation(s)
- Jennifer H Shaw
- Department of Integrative Biology, Oklahoma State University Stillwater, OK, USA
| | - Amanda R Behar
- Department of Microbiology and Molecular Genetics, Oklahoma State University Stillwater, OK, USA
| | - Timothy A Snider
- Department of Veterinary Pathobiology, Oklahoma State University Stillwater, OK, USA
| | - Noah A Allen
- Department of Integrative Biology, Oklahoma State University Stillwater, OK, USA
| | - Erika I Lutter
- Department of Microbiology and Molecular Genetics, Oklahoma State University Stillwater, OK, USA
| |
Collapse
|
35
|
Jiang J, Liu G, Kickhoefer VA, Rome LH, Li LX, McSorley SJ, Kelly KA. A Protective Vaccine against Chlamydia Genital Infection Using Vault Nanoparticles without an Added Adjuvant. Vaccines (Basel) 2017; 5:vaccines5010003. [PMID: 28106821 PMCID: PMC5371739 DOI: 10.3390/vaccines5010003] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2016] [Revised: 12/23/2016] [Accepted: 01/06/2017] [Indexed: 12/01/2022] Open
Abstract
Chlamydia trachomatis genital infection is the most common sexually transmitted bacterial disease, causing a significant burden to females due to reproductive dysfunction. Intensive screening and antibiotic treatment are unable to completely prevent female reproductive dysfunction, thus, efforts have become focused on developing a vaccine. A major impediment is identifying a safe and effective adjuvant which induces cluster of differentiation 4 (CD4) cells with attributes capable of halting genital infection and inflammation. Previously, we described a natural nanocapsule called the vault which was engineered to contain major outer membrane protein (MOMP) and was an effective vaccine which significantly reduced early infection and favored development of a cellular immune response in a mouse model. In the current study, we used another chlamydial antigen, a polymorphic membrane protein G-1 (PmpG) peptide, to track antigen-specific cells and evaluate, in depth, the vault vaccine for its protective capacity in the absence of an added adjuvant. We found PmpG-vault immunized mice significantly reduced the genital bacterial burden and histopathologic parameters of inflammation following a C. muridarum challenge. Immunization boosted antigen-specific CD4 cells with a multiple cytokine secretion pattern and reduced the number of inflammatory cells in the genital tract making the vault vaccine platform safe and effective for chlamydial genital infection. We conclude that vaccination with a Chlamydia-vault vaccine boosts antigen-specific immunities that are effective at eradicating infection and preventing reproductive tract inflammation.
Collapse
Affiliation(s)
- Janina Jiang
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, 10833 Le Conte Ave. CHS 1P-177, Los Angeles, CA 90095, USA.
| | - Guangchao Liu
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, 10833 Le Conte Ave. CHS 1P-177, Los Angeles, CA 90095, USA.
| | - Valerie A Kickhoefer
- Department of Biological Chemistry, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA.
| | - Leonard H Rome
- Department of Biological Chemistry, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA.
| | - Lin-Xi Li
- Center for Comparative Medicine, Department of Anatomy, Physiology & Cell Biology, School of Veterinary Medicine, University of California-Davis, Davis, CA 95616, USA.
| | - Stephen J McSorley
- Center for Comparative Medicine, Department of Anatomy, Physiology & Cell Biology, School of Veterinary Medicine, University of California-Davis, Davis, CA 95616, USA.
| | - Kathleen A Kelly
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, 10833 Le Conte Ave. CHS 1P-177, Los Angeles, CA 90095, USA.
- Department of Biological Chemistry, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA.
| |
Collapse
|
36
|
Abstract
Vaccines based on humoral immunity alone are unlikely to protect against infections caused by intracellular pathogens and today's most pressing infectious diseases of public health importance are caused by intracellular infections that include tuberculosis, malaria, HIV/AIDS, and others such as Chlamydia trachomatis. For these infections, vaccines that induce cellular immune responses are essential. Major impediments in developing such vaccines include difficulty in identifying relevant T cell antigens and delivering them in ways that elicit protective cellular immunity. Genomics and proteomics now provide tools to allow unbiased empirical identification of candidate T cell antigens. This approach represents an advance on bioinformatic searches for candidate T cell antigens. This chapter discusses an immunoproteomic approach we have used to identify Chlamydia T cell antigens. We further discuss how these T cell antigens can be developed into a human vaccine.
Collapse
|
37
|
Wali S, Gupta R, Yu JJ, Lanka GKK, Chambers JP, Guentzel MN, Zhong G, Murthy AK, Arulanandam BP. Chlamydial protease-like activity factor mediated protection against C. trachomatis in guinea pigs. Immunol Cell Biol 2016; 95:454-460. [PMID: 27990018 PMCID: PMC5449249 DOI: 10.1038/icb.2016.122] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2015] [Revised: 12/02/2016] [Accepted: 12/06/2016] [Indexed: 01/30/2023]
Abstract
We have comprehensively demonstrated using the mouse model that intranasal immunization with recombinant chlamydial protease-like activity factor (rCPAF) leads to a significant reduction in bacterial burden, genital tract pathology and preserves fertility following intravaginal genital chlamydial challenge. In the present report, we evaluated the protective efficacy of rCPAF immunization in guinea pigs, a second animal model for genital chlamydial infection. Using a vaccination strategy similar to the mouse model, we intranasally immunized female guinea pigs with rCPAF plus CpG deoxynucleotides (CpG; as an adjuvant), and challenged intravaginally with C. trachomatis serovar D (CT-D). Immunization with rCPAF/CpG significantly reduced vaginal CT-D shedding and induced resolution of infection by day 24, compared to day 33 in CpG alone treated and challenged animals. Immunization induced robust anti-rCPAF serum IgG 2 weeks following the last immunization, and was sustained at a high level 4 weeks post challenge. Upregulation of antigen specific IFN-γ gene expression was observed in rCPAF/CpG vaccinated splenocytes. Importantly, a significant reduction in inflammation in the genital tissue in rCPAF/CpG-immunized guinea pigs compared to CpG-immunized animals was observed. Taken together, this study provides evidence of the protective efficacy of rCPAF as a vaccine candidate in a second animal model of genital chlamydial infection.
Collapse
Affiliation(s)
- Shradha Wali
- South Texas Center for Emerging Infectious Diseases and Center for Excellence in Infection Genomics, University of Texas at San Antonio, One UTSA Circle, San Antonio, TX, USA
| | - Rishein Gupta
- South Texas Center for Emerging Infectious Diseases and Center for Excellence in Infection Genomics, University of Texas at San Antonio, One UTSA Circle, San Antonio, TX, USA
| | - Jieh-Juen Yu
- South Texas Center for Emerging Infectious Diseases and Center for Excellence in Infection Genomics, University of Texas at San Antonio, One UTSA Circle, San Antonio, TX, USA
| | - Gopala Krishna Koundinya Lanka
- South Texas Center for Emerging Infectious Diseases and Center for Excellence in Infection Genomics, University of Texas at San Antonio, One UTSA Circle, San Antonio, TX, USA
| | - James P Chambers
- South Texas Center for Emerging Infectious Diseases and Center for Excellence in Infection Genomics, University of Texas at San Antonio, One UTSA Circle, San Antonio, TX, USA
| | - M Neal Guentzel
- South Texas Center for Emerging Infectious Diseases and Center for Excellence in Infection Genomics, University of Texas at San Antonio, One UTSA Circle, San Antonio, TX, USA
| | - Guangming Zhong
- Department of Microbiology and Immunology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Ashlesh K Murthy
- Department of Pathology, Midwestern University, Downers Grove, IL, USA
| | - Bernard P Arulanandam
- South Texas Center for Emerging Infectious Diseases and Center for Excellence in Infection Genomics, University of Texas at San Antonio, One UTSA Circle, San Antonio, TX, USA
| |
Collapse
|
38
|
Liang S, Bulir D, Kaushic C, Mahony J. Considerations for the rational design of a Chlamydia vaccine. Hum Vaccin Immunother 2016; 13:831-835. [PMID: 27835064 PMCID: PMC5404357 DOI: 10.1080/21645515.2016.1252886] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
Chlamydia trachomatis is the leading cause of preventable blindness and the most common bacterial sexually transmitted infection. Remarkable progress in vaccine research over the past six decades has led to the advancement of novel C. trachomatis vaccine candidates into clinical trials. However, many questions regarding the role of specific cellular populations and molecular mechanisms in protective immunity against human C. trachomatis genital tract infections remain unanswered. Biomarkers of vaccine induced protective immunity are elusive in humans, while a cautionary message on the translatability of data obtained from current animal models has emanated from vaccine research and development efforts against other important human pathogens. In this commentary, we highlight recent advances in Chlamydia vaccine development and discuss their implications in the context of a rational approach to the design of a human C. trachomatis vaccine.
Collapse
Affiliation(s)
- Steven Liang
- a Michael G. DeGroote Institute for Infectious Disease Research , McMaster University , Hamilton , Ontario , Canada.,b St. Joseph's Research Institute , St. Joseph's Healthcare , Hamilton , Ontario , Canada
| | - David Bulir
- a Michael G. DeGroote Institute for Infectious Disease Research , McMaster University , Hamilton , Ontario , Canada.,b St. Joseph's Research Institute , St. Joseph's Healthcare , Hamilton , Ontario , Canada
| | - Charu Kaushic
- a Michael G. DeGroote Institute for Infectious Disease Research , McMaster University , Hamilton , Ontario , Canada.,c Department of Pathology and Molecular Medicine , McMaster University , Hamilton , Ontario , Canada
| | - James Mahony
- a Michael G. DeGroote Institute for Infectious Disease Research , McMaster University , Hamilton , Ontario , Canada.,b St. Joseph's Research Institute , St. Joseph's Healthcare , Hamilton , Ontario , Canada.,c Department of Pathology and Molecular Medicine , McMaster University , Hamilton , Ontario , Canada
| |
Collapse
|
39
|
Karunakaran KP, Yu H, Jiang X, Chan Q, Goldberg MF, Jenkins MK, Foster LJ, Brunham RC. Identification of MHC-Bound Peptides from Dendritic Cells Infected with Salmonella enterica Strain SL1344: Implications for a Nontyphoidal Salmonella Vaccine. J Proteome Res 2016; 16:298-306. [PMID: 27802388 DOI: 10.1021/acs.jproteome.6b00926] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Worldwide Salmonella enterica infections result in substantial morbidity and mortality and are the major cause of infant bacteremia in Sub-Saharan Africa. Diseases caused by Salmonella are treatable with antibiotics, but successful antibiotic treatment has become difficult due to antimicrobial resistance and collateral effects on the microbiome. An effective vaccine together with public health efforts may be a better strategy to control these infections. Protective immunity against Salmonella depends primarily on CD4 T-cell-mediated immune responses; therefore, identifying relevant T-cell antigens is necessary for Salmonella vaccine development. We previously used a dendritic-cell-based immunoproteomics approach in our laboratory to identify T-cell antigens. The testing of these antigens as vaccine candidates against Chlamydia infection in mice yielded positive results. We applied this technology in the present study by infecting murine bone-marrow-derived dendritic cells from C57BL/6 mice with Salmonella enterica strain SL1344, followed by immunoaffinity isolation of MHC class I and II molecules and elution of bound peptides. The sequences of the peptides were identified using tandem mass spectrometry. We identified 87 MHC class-II- and 23 MHC class-I-binding Salmonella-derived peptides. Four of the 12 highest scoring class-II-binding Salmonella peptides stimulated IFN-γ production by CD4+ T cells from the spleens of mice with persistent Salmonella infection. We conclude that antigens identified by MHC immunoproteomics will be useful for Salmonella immunobiology studies and are potential Salmonella vaccine candidates. Data have been deposited to the ProteomeXchange Consortium via the PRIDE partner repository with the data set identifier PXD004451.
Collapse
Affiliation(s)
- Karuna P Karunakaran
- Vaccine Research Laboratory, University of British Columbia Centre for Disease Control , Vancouver, British Columbia V5Z 4R4, Canada
| | - Hong Yu
- Vaccine Research Laboratory, University of British Columbia Centre for Disease Control , Vancouver, British Columbia V5Z 4R4, Canada
| | - Xiaozhou Jiang
- Vaccine Research Laboratory, University of British Columbia Centre for Disease Control , Vancouver, British Columbia V5Z 4R4, Canada
| | - Queenie Chan
- Department of Biochemistry and Molecular Biology, Centre for High-Throughput Biology, University of British Columbia , Vancouver, British Columbia V6T 1Z3, Canada
| | - Michael F Goldberg
- Center for Immunology, Department of Microbiology and Immunology, University of Minnesota Medical School , Minneapolis, Minnesota 55455, United States
| | - Marc K Jenkins
- Center for Immunology, Department of Microbiology and Immunology, University of Minnesota Medical School , Minneapolis, Minnesota 55455, United States
| | - Leonard J Foster
- Department of Biochemistry and Molecular Biology, Centre for High-Throughput Biology, University of British Columbia , Vancouver, British Columbia V6T 1Z3, Canada
| | - Robert C Brunham
- Vaccine Research Laboratory, University of British Columbia Centre for Disease Control , Vancouver, British Columbia V5Z 4R4, Canada
| |
Collapse
|
40
|
Testing the H56 Vaccine Delivered in 4 Different Adjuvants as a BCG-Booster in a Non-Human Primate Model of Tuberculosis. PLoS One 2016; 11:e0161217. [PMID: 27525651 PMCID: PMC4985151 DOI: 10.1371/journal.pone.0161217] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Accepted: 08/02/2016] [Indexed: 11/19/2022] Open
Abstract
The search for new and improved tuberculosis (TB) vaccines has focused on IFN-γ both for selecting antigens and for evaluating vaccine delivery strategies. The essential role of IFN-γ in endogenous host protection is well established, but it is still uncertain whether this also holds true for vaccine protection. Here we evaluate the H56 fusion protein vaccine as a BCG booster in a non-human primate (NHP) model of TB that closely recapitulates human TB pathogenesis. To date, only a handful of novel adjuvants have been tested in the NHP model of TB, and therefore we administered H56 in 3 novel cationic liposome adjuvants of increasing immunogenicity (CAF01, CAF04, CAF05) and compared them to H56 in the IC31® adjuvant previously reported to promote protection in this model. The individual clinical parameters monitored during infection (weight, ESR, X-ray) all correlated with survival, and boosting BCG with H56 in all adjuvants resulted in better survival rates compared to BCG alone. The adjuvants promoted IFN-γ-responses of increasing intensity as measured by ELISPOT in the peripheral blood, but the level of vaccine-specific IFN-γ production did not correlate with or predict disease outcome. This study’s main outcome underscores the importance of the choice of adjuvant for TB subunit vaccines, and secondly it highlights the need for better correlates of protection in preclinical models of TB.
Collapse
|
41
|
Carneiro C, Correia A, Lima T, Vilanova M, Pais C, Gomes AC, Real Oliveira MEC, Sampaio P. Protective effect of antigen delivery using monoolein-based liposomes in experimental hematogenously disseminated candidiasis. Acta Biomater 2016; 39:133-145. [PMID: 27150234 DOI: 10.1016/j.actbio.2016.05.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Revised: 04/22/2016] [Accepted: 05/01/2016] [Indexed: 01/08/2023]
Abstract
UNLABELLED We evaluated the potential of a liposomal antigen delivery system (ADS) containing Candida albicans cell wall surface proteins (CWSP) in mediating protection against systemic candidiasis. Treatment of bone-marrow-derived dendritic cells with CWSP-loaded dioctadecyldimethylammonium bromide:monoolein (DODAB:MO) liposomes enhanced and prolonged their activation comparatively to free antigen, indicating that liposome-entrapped CWSP were released more sustainable. Therefore, we immunized mice with CWSP either in a free form or loaded into two different DODAB:MO liposome formulations, respectively designated as ADS1 and ADS2, prior to intravenous C. albicans infection. Immunization with ADS1, but not with ADS2, conferred significant protection to infected mice, comparatively to immunization with CWSP or empty liposomes as control. ADS1-immunized mice presented significantly higher serum levels of C. albicans-specific antibodies that enhanced phagocytosis of this fungus. In these mice, a mixed cytokine production profile was observed encompassing IFN-γ, IL-4, IL-17A and IL-10. Nevertheless, only production of IL-4, IL-17 and IL-10 was higher than in controls. In this study we demonstrated that DODAB:MO liposomes enhance the immunogenicity of C. albicans antigens and host protection in a murine model of systemic candidiasis. Therefore, this liposomal adjuvant could be a promising candidate to assess in vaccination against this pathogenic fungus. STATEMENT OF SIGNIFICANCE This work describes the immunomodulation capacity of the previously validated antigen delivery system (ADS) composed by dioctadecyldimethylammonium bromide (DODAB) and monoolein (MO) lipids incorporating the cell wall surface proteins (CWSP) from C. albicans. Here, we not only present the ability of this system in facilitating antigen uptake by DCs in vitro, but also that this system induces higher levels of pro-inflammatory cytokines and opsonizing specific IgG antibodies in serum of mice immunized subcutaneously. We show that the ADS are efficient nanocarrier and modulate the immune response against intravenous C. albicans infection favoring mouse protection. In sum, we show that the incorporation of C. albicans antigens in DODAB:MO nanocarries are a promising vaccine strategy against C. albicans fungal infection.
Collapse
|
42
|
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
|
43
|
Designing liposomal adjuvants for the next generation of vaccines. Adv Drug Deliv Rev 2016; 99:85-96. [PMID: 26576719 DOI: 10.1016/j.addr.2015.11.005] [Citation(s) in RCA: 88] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Revised: 10/09/2015] [Accepted: 11/05/2015] [Indexed: 12/12/2022]
Abstract
Liposomes not only offer the ability to enhance drug delivery, but can effectively act as vaccine delivery systems and adjuvants. Their flexibility in size, charge, bilayer rigidity and composition allow for targeted antigen delivery via a range of administration routes. In the development of liposomal adjuvants, the type of immune response promoted has been linked to their physico-chemical characteristics, with the size and charge of the liposomal particles impacting on liposome biodistribution, exposure in the lymph nodes and recruitment of the innate immune system. The addition of immunostimulatory agents can further potentiate their immunogenic properties. Here, we outline the attributes that should be considered in the design and manufacture of liposomal adjuvants for the delivery of sub-unit and nucleic acid based vaccines.
Collapse
|
44
|
Abstract
For almost 2 decades, results from Chlamydia pathogenesis investigations have been conceptualized using a cytokine polarization narrative. Recent viral immunity studies identifying protective tissue-resident memory T cells (Trm) suggest an alternative paradigm based on localized immune networks. As Chlamydia vaccines enter the preclinical pipeline and, in the case of an attenuated trachoma vaccine, are given to human subjects, it may be useful to ask whether cytokine polarization is the appropriate framework for understanding and evaluating vaccine efficacy. In this review, we revisit C. trachomatis pathogenesis data from mice and humans using a Trm narrative and note a comfortable concordance with the Chlamydia pathogenesis literature.
Collapse
|
45
|
Yu H, Karunakaran KP, Jiang X, Brunham RC. Subunit vaccines for the prevention of mucosal infection with Chlamydia trachomatis. Expert Rev Vaccines 2016; 15:977-88. [PMID: 26938202 DOI: 10.1586/14760584.2016.1161510] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Chlamydia trachomatis is the most common preventable cause of tubal infertility in women. In high-income countries, despite public health control efforts, C. trachomatis case rates continue to rise. Most medium and low-income countries lack any Chlamydia control program; therefore, a vaccine is essential for the control of Chlamydia infections. A rationally designed Chlamydia vaccine requires understanding of the immunological correlates of protective immunity, pathological responses to this mucosal pathogen, identification of optimal vaccine antigens and selection of suitable adjuvant delivery systems that engender protective immunity. Fortunately, Chlamydia vaccinology is facilitated by genomic knowledge and by murine models that reproduce many of the features of human C. trachomatis infection. This article reviews recent progress in these areas with a focus on subunit vaccine development.
Collapse
Affiliation(s)
- Hong Yu
- a Vaccine Research Laboratory , University of British Columbia Centre for Disease Control , Vancouver , British Columbia , Canada
| | - Karuna P Karunakaran
- a Vaccine Research Laboratory , University of British Columbia Centre for Disease Control , Vancouver , British Columbia , Canada
| | - Xiaozhou Jiang
- a Vaccine Research Laboratory , University of British Columbia Centre for Disease Control , Vancouver , British Columbia , Canada
| | - Robert C Brunham
- a Vaccine Research Laboratory , University of British Columbia Centre for Disease Control , Vancouver , British Columbia , Canada
| |
Collapse
|
46
|
Vasilevsky S, Stojanov M, Greub G, Baud D. Chlamydial polymorphic membrane proteins: regulation, function and potential vaccine candidates. Virulence 2015; 7:11-22. [PMID: 26580416 DOI: 10.1080/21505594.2015.1111509] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Pmps (Polymorphic Membrane Proteins) are a group of membrane bound surface exposed chlamydial proteins that have been characterized as autotransporter adhesins and are important in the initial phase of chlamydial infection. These proteins all contain conserved GGA (I, L, V) and FxxN tetrapeptide motifs in the N-terminal portion of each protein. All chlamydial species express Pmps. Even in the chlamydia-related bacteria Waddlia chondrophila, a Pmp-like adhesin has been identified, demonstrating the importance of Pmps in Chlamydiales biology. Chlamydial species vary in the number of pmp genes and their differentially regulated expression during the infectious cycle or in response to stress. Studies have also demonstrated that Pmps are able to induce innate immune functional responses in infected cells, including production of IL-8, IL-6 and MCP-1, by activating the transcription factor NF-κB. Human serum studies have indicated that although anti-Pmp specific antibodies are produced in response to a chlamydial infection, the response is variable depending on the Pmp protein. In C. trachomatis, PmpB, PmpC, PmpD and PmpI were the proteins eliciting the strongest immune response among adolescents with and without pelvic inflammatory disease (PID). In contrast, PmpA and PmpE elicited the weakest antibody response. Interestingly, there seems to be a gender bias for Pmp recognition with a stronger anti-Pmp reactivity in male patients. Furthermore, anti-PmpA antibodies might contribute to adverse pregnancy outcomes, at least among women with PID. In vitro studies indicated that dendritic cells infected with C. muridarum were able to present PmpG and PmpF on their MHC class II receptors and T cells were able to recognize the MHC class-II bound peptides. In addition, vaccination with PmpEFGH and Major Outer Membrane Protein (MOMP) significantly protected mice against a genital tract C. muridarum infection, suggesting that Pmps may be an important component of a multi-subunit chlamydial vaccine. Thus, Pmps might be important not only for the pathogenesis of chlamydial infection, but also as potential candidate vaccine proteins.
Collapse
Affiliation(s)
- Sam Vasilevsky
- a Materno-fetal and Obstetrics Research Unit ; Department of Obstetrics and Gynecology; Maternity; University Hospital ; Lausanne , Switzerland
| | - Milos Stojanov
- a Materno-fetal and Obstetrics Research Unit ; Department of Obstetrics and Gynecology; Maternity; University Hospital ; Lausanne , Switzerland
| | - Gilbert Greub
- b Center for Research on Intracellular Bacteria; Institute of Microbiology; Faculty of Biology and Medicine; University of Lausanne and University Hospital ; Lausanne , Switzerland
| | - David Baud
- a Materno-fetal and Obstetrics Research Unit ; Department of Obstetrics and Gynecology; Maternity; University Hospital ; Lausanne , Switzerland
| |
Collapse
|
47
|
Nunes A, Gomes JP, Karunakaran KP, Brunham RC. Bioinformatic Analysis of Chlamydia trachomatis Polymorphic Membrane Proteins PmpE, PmpF, PmpG and PmpH as Potential Vaccine Antigens. PLoS One 2015; 10:e0131695. [PMID: 26131720 PMCID: PMC4488443 DOI: 10.1371/journal.pone.0131695] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Accepted: 06/05/2015] [Indexed: 12/31/2022] Open
Abstract
Chlamydia trachomatis is the most important infectious cause of infertility in women with important implications in public health and for which a vaccine is urgently needed. Recent immunoproteomic vaccine studies found that four polymorphic membrane proteins (PmpE, PmpF, PmpG and PmpH) are immunodominant, recognized by various MHC class II haplotypes and protective in mouse models. In the present study, we aimed to evaluate genetic and protein features of Pmps (focusing on the N-terminal 600 amino acids where MHC class II epitopes were mapped) in order to understand antigen variation that may emerge following vaccine induced immune selection. We used several bioinformatics platforms to study: i) Pmps' phylogeny and genetic polymorphism; ii) the location and distribution of protein features (GGA(I, L)/FxxN motifs and cysteine residues) that may impact pathogen-host interactions and protein conformation; and iii) the existence of phase variation mechanisms that may impact Pmps' expression. We used a well-characterized collection of 53 fully-sequenced strains that represent the C. trachomatis serovars associated with the three disease groups: ocular (N=8), epithelial-genital (N=25) and lymphogranuloma venereum (LGV) (N=20). We observed that PmpF and PmpE are highly polymorphic between LGV and epithelial-genital strains, and also within populations of the latter. We also found heterogeneous representation among strains for GGA(I, L)/FxxN motifs and cysteine residues, suggesting possible alterations in adhesion properties, tissue specificity and immunogenicity. PmpG and, to a lesser extent, PmpH revealed low polymorphism and high conservation of protein features among the genital strains (including the LGV group). Uniquely among the four Pmps, pmpG has regulatory sequences suggestive of phase variation. In aggregate, the results suggest that PmpG may be the lead vaccine candidate because of sequence conservation but may need to be paired with another protective antigen (like PmpH) in order to prevent immune selection of phase variants.
Collapse
Affiliation(s)
- Alexandra Nunes
- Bioinformatics Unit, Department of Infectious Diseases, National Institute of Health, Lisbon, Portugal
| | - João P. Gomes
- Bioinformatics Unit, Department of Infectious Diseases, National Institute of Health, Lisbon, Portugal
| | - Karuna P. Karunakaran
- Vaccine Research Laboratory, University of British Columbia Centre for Disease Control, Vancouver, Canada
| | - Robert C. Brunham
- Vaccine Research Laboratory, University of British Columbia Centre for Disease Control, Vancouver, Canada
- * E-mail:
| |
Collapse
|
48
|
Karunakaran KP, Yu H, Jiang X, Chan Q, Moon KM, Foster LJ, Brunham RC. Outer membrane proteins preferentially load MHC class II peptides: implications for a Chlamydia trachomatis T cell vaccine. Vaccine 2015; 33:2159-66. [PMID: 25738816 PMCID: PMC4390527 DOI: 10.1016/j.vaccine.2015.02.055] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Revised: 02/11/2015] [Accepted: 02/19/2015] [Indexed: 02/03/2023]
Abstract
CD4 T cell immune responses such as interferon-γ and tumor necrosis factor-α secretion are necessary for Chlamydia immunity. We used an immunoproteomic approach in which Chlamydia trachomatis and Chlamydia muridarum-derived peptides presented by MHC class II molecules on the surface of infected dendritic cells (DCs) were identified by tandem mass spectrometry using bone marrow derived DCs (BMDCs) from mice of different MHC background. We first compared the C. muridarum immunoproteome in C3H mice to that previously identified in C57BL/6 mice. Fourteen MHC class II binding peptides from 11 Chlamydia proteins were identified from C3H infected BMDCs. Two C. muridarum proteins overlapped between C3H and C57B/6 mice and both were polymorphic membrane proteins (Pmps) which presented distinct class II binding peptides. Next we studied DCs from C57BL/6 mice infected with the human strain, C. trachomatis serovar D. Sixty MHC class II binding peptides derived from 27 C. trachomatis proteins were identified. Nine proteins were orthologous T cell antigens between C. trachomatis and C. muridarum and 2 of the nine were Pmps which generated MHC class II binding epitopes at distinct sequences within the proteins. As determined by antigen specific splenocyte responses outer membrane proteins PmpF, -G and -H and the major outer membrane protein (MOMP) were antigenic in mice previously infected with C. muridarum or C. trachomatis. Furthermore a recombinant protein vaccine consisting of the four Pmps (PmpEFGH) with MOMP formulated with a Th1 polarizing adjuvant significantly accelerated (p<0.001) clearance in the C57BL/6 mice C. trachomatis transcervical infection model. We conclude that Chlamydia outer membrane proteins are important T cell antigens useful in the development of a C. trachomatis subunit vaccine.
Collapse
Affiliation(s)
- Karuna P Karunakaran
- Vaccine Research Laboratory, University of British Columbia Centre for Disease Control, Vancouver, BC, Canada
| | - Hong Yu
- Vaccine Research Laboratory, University of British Columbia Centre for Disease Control, Vancouver, BC, Canada
| | - Xiaozhou Jiang
- Vaccine Research Laboratory, University of British Columbia Centre for Disease Control, Vancouver, BC, Canada
| | - Queenie Chan
- Department of Biochemistry and Molecular Biology, Centre for High-Throughput Biology, University of British Columbia, Vancouver, BC, Canada
| | - Kyung-Mee Moon
- Department of Biochemistry and Molecular Biology, Centre for High-Throughput Biology, University of British Columbia, Vancouver, BC, Canada
| | - Leonard J Foster
- Department of Biochemistry and Molecular Biology, Centre for High-Throughput Biology, University of British Columbia, Vancouver, BC, Canada
| | - Robert C Brunham
- Vaccine Research Laboratory, University of British Columbia Centre for Disease Control, Vancouver, BC, Canada.
| |
Collapse
|
49
|
Li LX, McSorley SJ. A re-evaluation of the role of B cells in protective immunity to Chlamydia infection. Immunol Lett 2015; 164:88-93. [PMID: 25704502 DOI: 10.1016/j.imlet.2015.02.004] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Revised: 02/06/2015] [Accepted: 02/12/2015] [Indexed: 01/04/2023]
Abstract
Chlamydia trachomatis is the etiological agent of the most commonly reported bacterial sexual transmitted infection (STI) in North America and Europe. The control of Chlamydia infection is hindered by the asymptomatic nature of initial infection but the consequence of untreated infection seriously threatens the reproductive health of young women. Unfortunately, there is no licensed vaccine for Chlamydia vaccine, in part due to our incomplete understanding of the immune response to Chlamydia urogenital infection. It has been well established that T cell-mediated immunity plays a dominant role in protective immunity against Chlamydia and thus the importance of B cells is somewhat underappreciated. Here, we summarize recent progress on understanding the role of B cells during Chlamydia genital tract infections and discuss how B cells and humoral immunity make an effective contribution to host defense against important intracellular pathogens, including Chlamydia.
Collapse
Affiliation(s)
- Lin-Xi Li
- Center for Comparative Medicine, Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California, Davis, Davis, CA 95616, United States.
| | - Stephen J McSorley
- Center for Comparative Medicine, Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California, Davis, Davis, CA 95616, United States
| |
Collapse
|
50
|
|