1
|
Medhavi F, Tanner T, Richardson S, Lundy S, Omosun Y, Eko FO. A VCG-Based Multiepitope Chlamydia Vaccine Incorporating the Cholera Toxin A1 Subunit (MECA) Confers Protective Immunity Against Transcervical Challenge. Biomedicines 2025; 13:288. [PMID: 40002702 PMCID: PMC11852492 DOI: 10.3390/biomedicines13020288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Revised: 01/19/2025] [Accepted: 01/22/2025] [Indexed: 02/27/2025] Open
Abstract
Background/Objectives: We generated a novel recombinant Vibrio cholerae ghost (rVCG)-based subunit vaccine incorporating the A1 subunit of cholera toxin (CTA1) and a multiepitope Chlamydia trachomatis (CT) antigen (MECA) derived from five chlamydial outer membrane proteins (rVCG-MECA). The ability of this vaccine to protect against a CT transcervical challenge was evaluated. Methods: Female C57BL/6J mice were immunized thrice at two-week intervals with rVCG-MECA or rVCG-gD2 (antigen control) via the intramuscular (IM) or intranasal (IN) route. PBS-immunized mice or mice immunized with live CT served as negative and positive controls, respectively. Results: Vaccine delivery stimulated robust humoral and cell-mediated immune effectors, characterized by local mucosal and systemic CT-specific IgG, IgG2c, and IgA antibody and IFN-γ (Th1 cytokine) responses. The elicited mucosal and systemic IgG2c and IgA antibody responses persisted for 16 weeks post-immunization. Immunization with rVCG-MECA afforded protection comparable to that provided by IN immunization with live CT EBs without any side effects, irrespective of route of vaccine delivery. Conclusions: The results underline the potential of a multiepitope vaccine as a promising resource for protecting against CT genital infection and the potential of CTA1 on the VCG platform as a mucosal and systemic adjuvant for developing CT vaccines.
Collapse
Affiliation(s)
| | | | | | | | | | - Francis O. Eko
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA 30310, USA; (F.M.); (Y.O.)
| |
Collapse
|
2
|
Wilton ZER, Jamus AN, Core SB, Frietze KM. Pathogenic and Protective Roles of Neutrophils in Chlamydia trachomatis Infection. Pathogens 2025; 14:112. [PMID: 40005489 PMCID: PMC11858174 DOI: 10.3390/pathogens14020112] [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: 12/13/2024] [Revised: 01/14/2025] [Accepted: 01/20/2025] [Indexed: 02/27/2025] Open
Abstract
Chlamydia trachomatis (Ct) is an obligate intracellular pathogen that causes the most commonly diagnosed bacterial sexually transmitted infection (STI) and is a leading cause of preventable blindness globally. Ct infections can generate a strong pro-inflammatory immune response, leading to immune-mediated pathology in infected tissues. Neutrophils play an important role in mediating both pathology and protection during infection. Excessive neutrophil activation, migration, and survival are associated with host tissue damage during Chlamydia infections. In contrast, neutrophils also perform phagocytic killing of Chlamydia in the presence of IFN-γ and anti-Chlamydia antibodies. Neutrophil extracellular traps (NETs) and many neutrophil degranulation products have also demonstrated strong anti-Chlamydia functions. To counteract this neutrophil-mediated protection, Chlamydia has developed several evasion strategies. Various Chlamydia proteins can limit potentially protective neutrophil responses by directly targeting receptors present on the surface of neutrophils or neutrophil degranulation products. In this review, we provide a survey of current knowledge regarding the role of neutrophils in pathogenesis and protection, including the ways that Chlamydia circumvents neutrophil functions, and we propose critical areas for future research.
Collapse
Affiliation(s)
| | | | | | - Kathryn M. Frietze
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences, Albuquerque, NM 87131, USA
| |
Collapse
|
3
|
Pal S, Carmichael JR, Tifrea DF, Tatarenkova O, de la Maza LM. Chlamydia trachomatis Serovars from the C-Complex and the B- and C-Related Complexes Are Significantly More Pathogenic than Those from the B-Complex in C3H/HeN but Not in BALB/c Mice. Pathogens 2025; 14:97. [PMID: 39861058 PMCID: PMC11768385 DOI: 10.3390/pathogens14010097] [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: 11/27/2024] [Revised: 01/16/2025] [Accepted: 01/17/2025] [Indexed: 01/27/2025] Open
Abstract
Studies in humans indicate that certain Chlamydia trachomatis serovars are more pathogenic than others. Specifically, several studies concluded that serovars from the C-complex are more pathogenic than those from the B-complex, although there are reports that do not support this finding. To investigate these results in an animal model, the eight genitourinary C. trachomatis serovars were tested in two strains of mice: C3H/HeN and BALB/c. These two strains of mice were investigated because C3H/HeN is more susceptible to Chlamydia muridarum infections than BALB/c, indicative of differences in their immunogenetic background. Mice were infected transcervically with 105 inclusion forming units of each of the C. trachomatis serovars, and vaginal cultures were collected. To determine the pathogenicity and its impact on fertility, at week seven post-infection, female mice were caged with male mice. In the C3H/HeN mice, significant differences in vaginal shedding and fertility were observed between serovars from the B-complex (D and E) and those from the C-complex (H, I, J) and B- and C-related complexes (G, F, and K). The animals infected with serovars F, G, H, I, J, and K shed less but had significantly more infertility than the mice infected with serovars D or E. The experiments in the BALB/c mice, however, did not show major differences in pathogenicity between the eight C. trachomatis serovars. These results support the findings in humans and emphasize the critical importance of the immunogenetic background of the host on the outcome of C. trachomatis infections. The data imply that management of C. trachomatis-infected patients may require a more personalized approach.
Collapse
Affiliation(s)
| | | | | | | | - 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; (S.P.); (J.R.C.); (D.F.T.); (O.T.)
| |
Collapse
|
4
|
Huynh DT, Nolfi E, Guleed S, Medfai L, Wolf N, Uijen RF, de Jonge MI, van Ulsen P, Dietrich J, Luirink J, Sijts AJAM, Jong WSP. Intradermal administration of novel particulate Chlamydia trachomatis vaccine candidates drives protective immune responses. Biomed Pharmacother 2024; 180:117563. [PMID: 39405914 DOI: 10.1016/j.biopha.2024.117563] [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: 06/25/2024] [Revised: 10/07/2024] [Accepted: 10/08/2024] [Indexed: 11/14/2024] Open
Abstract
Chlamydia trachomatis causes the most prevalent bacterial sexually transmitted infection worldwide. Its complex lifecycle and the lack of appropriate antigen delivery vehicles make it difficult to develop an effective C. trachomatis vaccine. Recently, bacterial protein bodies (PBs) have emerged as promising bioparticles for vaccine antigen delivery. By developing a PB-tag for translational fusion, we were able to induce the aggregation of recombinant antigens expressed in Escherichia coli into PBs. Here, we investigated the immunogenicity and efficacy of PBs containing either the C. trachomatis MOMP-derived CTH522-SP or HtrA antigen in mice. Intradermal administration of c-di-AMP-adjuvanted PB-CTH522-SP and PB-HtrA vaccines, produced in an LPS-detoxified E. coli strain, induced antigen-specific cellular immunity, as measured by significant release of IFN-γ and IL17a in draining cervical lymph node and splenic cell cultures. Moreover, significant induction of HtrA-specific IFN-γ expressing CD4+ and CD8+ T cells was detected in the spleens. While immunization with the two PB vaccines led to prominent levels of specific antibodies in both serum and vaginal compartments, only antiserum against PB-CTH522-SP exhibited C. trachomatis-specific neutralization activity. Importantly, intradermal immunization with PB-CTH522-SP significantly reduced bacterial counts following C. trachomatis genital challenge. These data highlight the potential of the PB-based platform for the development of C. trachomatis vaccines.
Collapse
Affiliation(s)
- Dung T Huynh
- Abera Bioscience AB, Uppsala, Sweden; Group of Molecular Microbiology, Amsterdam Institute for Life and Environment (A-LIFE), Vrije Universiteit, Amsterdam, the Netherlands
| | - Emanuele Nolfi
- Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | - Safia Guleed
- Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark
| | - Lobna Medfai
- Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | | | - Rienke F Uijen
- Laboratory of Medical Immunology, Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Marien I de Jonge
- Laboratory of Medical Immunology, Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Peter van Ulsen
- Group of Molecular Microbiology, Amsterdam Institute for Life and Environment (A-LIFE), Vrije Universiteit, Amsterdam, the Netherlands
| | - Jes Dietrich
- Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark
| | - Joen Luirink
- Abera Bioscience AB, Uppsala, Sweden; Group of Molecular Microbiology, Amsterdam Institute for Life and Environment (A-LIFE), Vrije Universiteit, Amsterdam, the Netherlands
| | - Alice J A M Sijts
- Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands.
| | | |
Collapse
|
5
|
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
|
6
|
Toumasis P, Vrioni G, Tsinopoulos IT, Exindari M, Samonis G. Insights into Pathogenesis of Trachoma. Microorganisms 2024; 12:1544. [PMID: 39203386 PMCID: PMC11355952 DOI: 10.3390/microorganisms12081544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 05/31/2024] [Accepted: 07/25/2024] [Indexed: 09/03/2024] Open
Abstract
Trachoma is the most common infectious cause of blindness worldwide. This review investigates the pathogenesis of trachoma, focusing on its causative agent, transmission pathways, disease progression, and immune responses. Trachoma is caused by serovars A-C of the bacterium Chlamydia trachomatis (Ct). Transmission occurs through direct or indirect exchanges of ocular and nasal secretions, especially in regions with poor hygiene and overcrowded living conditions. The disease is initiated in early childhood by repeated infection of the ocular surface by Ct. This triggers recurrent chronic inflammatory episodes, leading to the development of conjunctival scarring and potentially to trichiasis, corneal opacity, and visual impairment. Exploring the pathogenesis of trachoma not only unveils the intricate pathways and mechanisms underlying this devastating eye disease but also underscores the multifaceted dimensions that must be considered in its management.
Collapse
Affiliation(s)
- Panagiotis Toumasis
- MSc in Ocular Surgery, School of Medicine, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece; (P.T.); (I.T.T.); (M.E.)
| | - Georgia Vrioni
- Department of Microbiology, School of Medicine, National and Kapodistrian University of Athens, 115 25 Athens, Greece;
| | - Ioannis T. Tsinopoulos
- MSc in Ocular Surgery, School of Medicine, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece; (P.T.); (I.T.T.); (M.E.)
- Second Department of Ophthalmology, School of Medicine, Aristotle University of Thessaloniki, Papageorgiou General Hospital, 564 29 Thessaloniki, Greece
| | - Maria Exindari
- MSc in Ocular Surgery, School of Medicine, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece; (P.T.); (I.T.T.); (M.E.)
- Department of Microbiology, School of Medicine, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece
| | - George Samonis
- School of Medicine, University of Crete, 715 00 Heraklion, Greece
- Metropolitan Hospital, 185 47 Piraeus, Greece
| |
Collapse
|
7
|
Armitage CW, Bryan ER, Trim L, Palframan E, Wager L, Beagley KW, Carey AJ. Haematopoietic innate interleukin 17A production drives immunopathology in female mouse genital Chlamydia muridarum infection. Scand J Immunol 2024; 99:e13359. [PMID: 38605527 DOI: 10.1111/sji.13359] [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: 10/05/2023] [Revised: 01/02/2024] [Accepted: 01/09/2024] [Indexed: 04/13/2024]
Abstract
Chlamydia trachomatis infection is the leading cause of bacterial urogenital infection and has been demonstrated to drive inflammation and scarring of the reproductive tract. Recent studies have identified key triggers of proinflammatory adaptive immune responses driven by innate leukocytes and epithelia driving immunopathology. Utilizing chimeric mouse models, we investigated the definitive source and role of IL17 and IL17 signalling receptors during early Chlamydia muridarum infection of the female urogenital tract. Bone marrow transplants from wild-type (WT) and IL17A-/- mice to recipients demonstrated equivocal infection kinetics in the reproductive tract, but interestingly, adoptive transfer of IL17A-/- immune cells to WT recipients resulted in no infertility, suggesting a haematopoietic (as opposed to tissue) source of IL17 driving immunopathology. To further delineate the role of IL17 in immunopathology, we infected WT and IL17 receptor A (IL17RA)-/- female mice and observed a significant reduction in immunopathology in IL17RA-/- mice. WT bone marrow transplants to IL17RA-/- recipient mice prevented hydrosalpinx, suggesting signalling through IL17RA drives immunopathology. Furthermore, early chemical inhibition of IL17 signalling significantly reduced hydrosalpinx, suggesting IL17 acts as an innate driver of disease. Early during the infection, IL17 was produced by γδ T cells in the cervico-vagina, but more importantly, by neutrophils at the site of infertility in the oviducts. Taken together, these data suggest innate production of IL17 by haematopoietic leukocytes drives immunopathology in the epithelia during early C. muridarum infection of the female reproductive tract.
Collapse
Affiliation(s)
- Charles W Armitage
- School of Biomedical Science and Centre for Immunology and Infection Control, Faculty of Health, Queensland University of Technology, Brisbane, Queensland, Australia
- Peter Goher Department of Immunobiology, Kings College London, London, UK
| | - Emily R Bryan
- School of Biomedical Science and Centre for Immunology and Infection Control, Faculty of Health, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Logan Trim
- School of Biomedical Science and Centre for Immunology and Infection Control, Faculty of Health, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Ella Palframan
- School of Biomedical Science and Centre for Immunology and Infection Control, Faculty of Health, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Lucas Wager
- School of Biomedical Science and Centre for Immunology and Infection Control, Faculty of Health, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Kenneth W Beagley
- School of Biomedical Science and Centre for Immunology and Infection Control, Faculty of Health, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Alison J Carey
- School of Biomedical Science and Centre for Immunology and Infection Control, Faculty of Health, Queensland University of Technology, Brisbane, Queensland, Australia
| |
Collapse
|
8
|
Sahu R, Verma R, Egbo TE, Giambartolomei GH, Singh SR, Dennis VA. Effects of prime-boost strategies on the protective efficacy and immunogenicity of a PLGA (85:15)-encapsulated Chlamydia recombinant MOMP nanovaccine. Pathog Dis 2024; 82:ftae004. [PMID: 38862192 PMCID: PMC11186516 DOI: 10.1093/femspd/ftae004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 01/08/2024] [Accepted: 06/10/2024] [Indexed: 06/13/2024] Open
Abstract
To begin to optimize the immunization routes for our reported PLGA-rMOMP nanovaccine [PLGA-encapsulated Chlamydia muridarum (Cm) recombinant major outer membrane protein (rMOMP)], we compared two prime-boost immunization strategies [subcutaneous (SC) and intramuscular (IM-p) prime routes followed by two SC-boosts)] to evaluate the nanovaccine-induced protective efficacy and immunogenicity in female BALB/c mice. Our results showed that mice immunized via the SC and IM-p routes were protected against a Cm genital challenge by a reduction in bacterial burden and with fewer bacteria in the SC mice. Protection of mice correlated with rMOMP-specific Th1 (IL-2 and IFN-γ) and not Th2 (IL-4, IL-9, and IL-13) cytokines, and CD4+ memory (CD44highCD62Lhigh) T-cells, especially in the SC mice. We also observed higher levels of IL-1α, IL-6, IL-17, CCL-2, and G-CSF in SC-immunized mice. Notably, an increase of cytokines/chemokines was seen after the challenge in the SC, IM-p, and control mice (rMOMP and PBS), suggesting a Cm stimulation. In parallel, rMOMP-specific Th1 (IgG2a and IgG2b) and Th2 (IgG1) serum, mucosal, serum avidity, and neutralizing antibodies were more elevated in SC than in IM-p mice. Overall, the homologous SC prime-boost immunization of mice induced enhanced cellular and antibody responses with better protection against a genital challenge compared to the heterologous IM-p.
Collapse
Affiliation(s)
- Rajnish Sahu
- Center for NanoBiotechnology Research, Department of Biological Sciences, 1627 Harris Way, Alabama State University, Montgomery AL, 36104, United States
| | - Richa Verma
- Center for NanoBiotechnology Research, Department of Biological Sciences, 1627 Harris Way, Alabama State University, Montgomery AL, 36104, United States
| | - Timothy E Egbo
- US Army Medical Research Institute of Infectious Diseases, Unit 8900, DPO, AE, Box 330, 09831, United States
| | - Guillermo H Giambartolomei
- Instituto de Inmunología, Genética y Metabolismo (INIGEM). CONICET. AV. Cordoba 2351, Universidad de Buenos Aires, Buenos Aires, C1120AAR, Argentina
| | - Shree R Singh
- Center for NanoBiotechnology Research, Department of Biological Sciences, 1627 Harris Way, Alabama State University, Montgomery AL, 36104, United States
| | - Vida A Dennis
- Center for NanoBiotechnology Research, Department of Biological Sciences, 1627 Harris Way, Alabama State University, Montgomery AL, 36104, United States
| |
Collapse
|
9
|
Borges ÁH, Follmann F, Dietrich J. Chlamydia trachomatis vaccine development - a view on the current challenges and how to move forward. Expert Rev Vaccines 2022; 21:1555-1567. [PMID: 36004386 DOI: 10.1080/14760584.2022.2117694] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Chlamydia trachomatis is the most common sexually transmitted bacterial pathogen in the world. A licensed vaccine is not yet available, but the first vaccines have entered clinical trials. AREAS COVERED : We describe the progress that has been made in our understanding of the type of immunity that a protective vaccine should induce, and the challenges that vaccine developers face. We also focus on the clinical development of a chlamydia vaccine. The first chlamydia vaccine candidate has now been tested in a clinical phase-I trial, and another phase-I trial is currently running. We discuss what it will take to continue this development and what future trial setups could look like. EXPERT OPINION The chlamydia field is coming of age and the first phase I clinical trial of a C. trachomatis vaccine has been successfully completed. We expect and hope that this will motivate various stakeholders to support further development of chlamydia vaccines in humans.
Collapse
Affiliation(s)
- Álvaro H Borges
- Statens Serum Institut, Department of Infectious Diseases Immunology, Kobenhavn, 2300 Denmark
| | | | - Jes Dietrich
- Statens Serum Institut, Department of Infectious Diseases Immunology, Kobenhavn, 2300 Denmark
| |
Collapse
|
10
|
Abstract
Trachoma is a neglected tropical disease caused by infection with conjunctival strains of Chlamydia trachomatis. It can result in blindness. Pathophysiologically, trachoma is a disease complex composed of two linked chronic processes: a recurrent, generally subclinical infectious-inflammatory disease that mostly affects children, and a non-communicable, cicatricial and, owing to trichiasis, eventually blinding disease that supervenes in some individuals later in life. At least 150 infection episodes over an individual's lifetime are needed to precipitate trichiasis; thus, opportunity exists for a just global health system to intervene to prevent trachomatous blindness. Trachoma is found at highest prevalence in the poorest communities of low-income countries, particularly in sub-Saharan Africa; in June 2021, 1.8 million people worldwide were going blind from the disease. Blindness attributable to trachoma can appear in communities many years after conjunctival C. trachomatis transmission has waned or ceased; therefore, the two linked disease processes require distinct clinical and public health responses. Surgery is offered to individuals with trichiasis and antibiotic mass drug administration and interventions to stimulate facial cleanliness and environmental improvement are designed to reduce infection prevalence and transmission. Together, these interventions comprise the SAFE strategy, which is achieving considerable success. Although much work remains, a continuing public health problem from trachoma in the year 2030 will be difficult for the world to excuse.
Collapse
|
11
|
Greydanus DE, Cabral MD, Patel DR. Pelvic inflammatory disease in the adolescent and young adult: An update. Dis Mon 2021; 68:101287. [PMID: 34521505 DOI: 10.1016/j.disamonth.2021.101287] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Pelvic inflammatory disease (PID) is an infection of the female upper genital tract that is typically polymicrobial with classic core involvement of Neisseria gonorrhoeae and/or Chlamydia trachomatis, though other endogenous flora from the vagino-cervical areas can be involved as well. It is often a sexually transmitted disease but other etiologic routes are also noted. A variety of risk factors have been identified including adolescence, young adulthood, adolescent cervical ectropion, multiple sexual partners, immature immune system, history of previous PID, risky contraceptive practices and others. An early diagnosis and prompt treatment are necessary to reduce risks of PID complications such as chronic pelvic pain, ectopic pregnancy and infertility. Current management principles of PID are also reviewed. It is important for clinicians to screen sexually active females for common sexually transmitted infections such as Chlamydia trachomatis and provide safer sex education to their adolescent and young adult patients. Clinicians should provide comprehensive management to persons with PID and utilize established guidelines such as those from the US Centers for Disease Control and Prevention (CDC).
Collapse
Affiliation(s)
- Donald E Greydanus
- Department of Pediatric and Adolescent Medicine, Western Michigan University, Homer Stryker M.D. School of Medicine, 1000 Oakland Drive, Kalamazoo, Michigan, 49008, United States of America.
| | - Maria Demma Cabral
- Department of Pediatric and Adolescent Medicine, Western Michigan University, Homer Stryker M.D. School of Medicine, 1000 Oakland Drive, Kalamazoo, Michigan, 49008, United States of America.
| | - Dilip R Patel
- Department of Pediatric and Adolescent Medicine, Western Michigan University, Homer Stryker M.D. School of Medicine, 1000 Oakland Drive, Kalamazoo, Michigan, 49008, United States of America.
| |
Collapse
|
12
|
Chen H, Wang L, Zhao L, Luo L, Min S, Wen Y, Lei W, Shu M, Li Z. Alterations of Vaginal Microbiota in Women With Infertility and Chlamydia trachomatis Infection. Front Cell Infect Microbiol 2021; 11:698840. [PMID: 34414130 PMCID: PMC8370387 DOI: 10.3389/fcimb.2021.698840] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 07/19/2021] [Indexed: 01/16/2023] Open
Abstract
Chlamydia trachomatis (C. trachomatis) is the most common etiological agent of bacterial sexually transmitted infections (STIs) worldwide and causes serious health sequelae such as cervicitis, pelvic inflammatory disease, and even infertility if ascending from the lower to the upper female genital tract. Previous studies have revealed the pivotal role of vaginal microbiota in susceptibility to STIs. However, alterations in the vaginal microbiota in women who are infertile and infected with C. trachomatis remain unknown. This study used metagenomic analysis of sequenced 16S rRNA gene amplicons to examine the vaginal microbial profiles of women with tubal infertility who were C. trachomatis-negative and those who were C. trachomatis-positive pre- and post-antibiotic treatment. Women who were C. trachomatis-negative and deemed healthy were recruited as references of eubiosis and dysbiosis. Women with tubal infertility and C. trachomatis infection presented a unique Lactobacillus iners-dominated vaginal microbiota rather than one dominated by Lactobacillus crispatus and displayed a decrease in Lactobacillus, Bifidobacterium, Enterobacter, Atopobium, and Streptococcus, accompanied by decreased levels of cytokines such as interferon (IFN)-γ and interleukin (IL)-10. This altered vaginal microbiota could be restored with varying degrees after standard treatment for C. trachomatis. This shift could be a predictive vaginal microbiota signature for C. trachomatis infection among females with tubal infertility, while no significant differences in phylum, class, and operational taxonomic unit (OTU) levels were observed between women with tubal infertility who were C. trachomatis-negative and healthy controls. This is the first study to provide data on the association of vaginal microbiota with C. trachomatis infection among women with tubal infertility and highlights unprecedented potential opportunities to predict C. trachomatis infection.
Collapse
Affiliation(s)
- Hongliang Chen
- Institute of Pathogenic Biology, Hengyang Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China.,Chenzhou No.1 People's Hospital, The First School of Clinical Medicine, Southern Medical University, Chenzhou, China
| | - Li Wang
- Chenzhou No.1 People's Hospital, The First School of Clinical Medicine, Southern Medical University, Chenzhou, China
| | - Lanhua Zhao
- Institute of Pathogenic Biology, Hengyang Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China
| | - Lipei Luo
- Chenzhou No.1 People's Hospital, The First School of Clinical Medicine, Southern Medical University, Chenzhou, China
| | - Shuling Min
- Institute of Pathogenic Biology, Hengyang Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China.,Chenzhou No.1 People's Hospital, The First School of Clinical Medicine, Southern Medical University, Chenzhou, China
| | - Yating Wen
- Institute of Pathogenic Biology, Hengyang Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China
| | - Wenbo Lei
- Institute of Pathogenic Biology, Hengyang Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China
| | - Mingyi Shu
- Institute of Pathogenic Biology, Hengyang Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China
| | - Zhongyu Li
- Institute of Pathogenic Biology, Hengyang Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China
| |
Collapse
|
13
|
Swedik S, Madola A, Levine A. IL-17C in human mucosal immunity: More than just a middle child. Cytokine 2021; 146:155641. [PMID: 34293699 DOI: 10.1016/j.cyto.2021.155641] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 06/29/2021] [Accepted: 06/30/2021] [Indexed: 02/07/2023]
Abstract
Interleukin-17C (IL-17C) is an understudied member of the IL-17 family of cytokines. Its synthesis is induced by both cytokines and pathogenic stimuli in a variety of cell types, most often expressed at mucosal and barrier surfaces. IL-17C expression is dysregulated in a variety of autoinflammatory and autoimmune diseases including inflammatory bowel disease, psoriasis, and atopic dermatitis, yet it is protective against bacterial infections of the gut, skin, and lungs. In this review we highlight studies on IL-17C regulation and its function at human mucosal surfaces. Understanding the relationship between IL-17C and autoinflammatory and autoimmune diseases of the mucosa and defining the beneficial and pathogenic functions of the cytokine in inflammatory responses are the first steps in determining the potential for IL-17C as a therapeutic target.
Collapse
Affiliation(s)
- Stephanie Swedik
- Department of Molecular Biology and Microbiology, Case Western Reserve University, 10900 Euclid Ave, Cleveland, OH 44106, United States
| | - Abson Madola
- Department of Biology, Case Western Reserve University, 10900 Euclid Ave, Cleveland, OH 44106, United States
| | - Alan Levine
- Department of Molecular Biology and Microbiology, Case Western Reserve University, 10900 Euclid Ave, Cleveland, OH 44106, United States; Departments of Pathology, Pharmacology, Medicine, and Pediatrics, Case Western Reserve University, 10900 Euclid Ave, Cleveland, OH 44106, United States.
| |
Collapse
|
14
|
Reduced uterine tissue damage during Chlamydia muridarum infection in TREM-1,3 deficient mice. Infect Immun 2021; 89:e0007221. [PMID: 34125599 DOI: 10.1128/iai.00072-21] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Genital infections with Chlamydia trachomatis can lead to uterine and oviduct tissue damage in the female reproductive tract. Neutrophils are strongly associated with tissue damage during chlamydial infection, while an adaptive CD4 T cell response is necessary to combat infection. Activation of triggering receptor expressed on myeloid cells-1 (TREM-1) on neutrophils has previously been shown to induce and/or enhance degranulation synergistically with TLR-signaling. Additionally, TREM-1 can promote neutrophil transepithelial migration. In this study, we sought to determine the contribution of TREM-1,3 in immunopathology in the female mouse genital tract during Chlamydia muridarum infection. Relative to control mice, trem1,3-/- mice had no difference in chlamydial burden or duration of lower genital tract infection. We also observed a similar incidence of oviduct hydrosalpinx 45 days post-infection in trem1,3-/- compared to WT mice. However, compared to WT, trem1,3-/- mice developed significantly fewer uterine horn hydrometra. Early in infection, trem1,3-/- mice displayed a notable decrease in the number of uterine glands containing polymorphonuclear cells and uterine horn lumens had fewer neutrophils, with increased G-CSF. Trem1,3-/- mice also had reduced erosion of the luminal epithelium. These data indicate TREM-1,3 contributes to transepithelial neutrophil migration in the uterus and uterine glands, promoting the development of uterine hydrometra in infected mice.
Collapse
|
15
|
Xiang W, Yu N, Lei A, Li X, Tan S, Huang L, Zhou Z. Insights Into Host Cell Cytokines in Chlamydia Infection. Front Immunol 2021; 12:639834. [PMID: 34093528 PMCID: PMC8176227 DOI: 10.3389/fimmu.2021.639834] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 05/05/2021] [Indexed: 01/08/2023] Open
Abstract
Chlamydial infection causes a number of clinically relevant diseases and induces significant morbidity in humans. Immune and inflammatory responses contribute to both the clearance of Chlamydia infection and pathology in host tissues. Chlamydia infection stimulates host cells to produce a large number of cytokines that trigger and regulate host immune responses against Chlamydia. However, inappropriate responses can occur with excessive production of cytokines, resulting in overreactive inflammatory responses and alterations in host or Chlamydia metabolism. As a result, Chlamydia persists and causes wound healing delays, leading to more severe tissue damage and triggering long-lasting fibrotic sequelae. Here, we summarize the roles of cytokines in Chlamydia infection and pathogenesis, thus advancing our understanding chlamydial infection biology and the pathogenic mechanisms involved.
Collapse
Affiliation(s)
- Wenjing Xiang
- Institute of Pathogenic Biology, Hengyang Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China
| | - Nanyan Yu
- Institute of Pathogenic Biology, Hengyang Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China
| | - Aihua Lei
- Institute of Pathogenic Biology, Hengyang Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China
| | - Xiaofang Li
- Institute of Pathogenic Biology, Hengyang Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China
| | - Shui Tan
- Institute of Pathogenic Biology, Hengyang Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China
| | - Lijun Huang
- Institute of Pathogenic Biology, Hengyang Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China.,Nanyue Biopharmaceutical Co. Ltd., Hunan Province Innovative Training Base for Postgraduates, University of South China and Nanyue Biopharmaceutical Co. Ltd., Hengyang, China
| | - Zhou Zhou
- Institute of Pathogenic Biology, Hengyang Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China
| |
Collapse
|
16
|
de la Maza LM, Darville TL, Pal S. Chlamydia trachomatis vaccines for genital infections: where are we and how far is there to go? Expert Rev Vaccines 2021; 20:421-435. [PMID: 33682583 DOI: 10.1080/14760584.2021.1899817] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
INTRODUCTION Chlamydia trachomatis is the most common sexually transmitted bacterial pathogen in the world. Antibiotic treatment does not prevent against reinfection and a vaccine is not yet available. AREAS COVERED We focus the review on the progress made of our understanding of the immunological responses required for a vaccine to elicit protection, and on the antigens, adjuvants, routes of immunization and delivery systems that have been tested in animal models. PubMed and Google Scholar were used to search publication on these topics for the last 5 years and recent Reviews were examined. EXPERT OPINION The first Phase 1 clinical trial of a C. trachomatis vaccine to protect against genital infections was successfully completed. We expect that, in the next five years, additional vaccine clinical trials will be implemented.
Collapse
Affiliation(s)
- Luis M de la Maza
- Department of Pathology and Laboratory Medicine Medical Sciences, I, Room D440 University of California, Irvine, California, USA
| | - Toni L Darville
- Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Sukumar Pal
- Department of Pathology and Laboratory Medicine Medical Sciences, I, Room D440 University of California, Irvine, California, USA
| |
Collapse
|
17
|
Dockterman J, Coers J. Immunopathogenesis of genital Chlamydia infection: insights from mouse models. Pathog Dis 2021; 79:ftab012. [PMID: 33538819 PMCID: PMC8189015 DOI: 10.1093/femspd/ftab012] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Accepted: 02/02/2021] [Indexed: 12/13/2022] Open
Abstract
Chlamydiae are pathogenic intracellular bacteria that cause a wide variety of diseases throughout the globe, affecting the eye, lung, coronary arteries and female genital tract. Rather than by direct cellular toxicity, Chlamydia infection generally causes pathology by inducing fibrosis and scarring that is largely mediated by host inflammation. While a robust immune response is required for clearance of the infection, certain elements of that immune response may also damage infected tissue, leading to, in the case of female genital infection, disease sequelae such as pelvic inflammatory disease, infertility and ectopic pregnancy. It has become increasingly clear that the components of the immune system that destroy bacteria and those that cause pathology only partially overlap. In the ongoing quest for a vaccine that prevents Chlamydia-induced disease, it is important to target mechanisms that can achieve protective immunity while preventing mechanisms that damage tissue. This review focuses on mouse models of genital Chlamydia infection and synthesizes recent studies to generate a comprehensive model for immunity in the murine female genital tract, clarifying the respective contributions of various branches of innate and adaptive immunity to both host protection and pathogenic genital scarring.
Collapse
Affiliation(s)
- Jacob Dockterman
- Department of Immunology, Duke University Medical Center, Durham, NC 22710, USA
| | - Jörn Coers
- Department of Immunology, Duke University Medical Center, Durham, NC 22710, USA
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 22710, USA
| |
Collapse
|
18
|
Howard S, Richardson S, Benyeogor I, Omosun Y, Dye K, Medhavi F, Lundy S, Adebayo O, Igietseme JU, Eko FO. Differential miRNA Profiles Correlate With Disparate Immunity Outcomes Associated With Vaccine Immunization and Chlamydial Infection. Front Immunol 2021; 12:625318. [PMID: 33692799 PMCID: PMC7937703 DOI: 10.3389/fimmu.2021.625318] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 01/29/2021] [Indexed: 12/12/2022] Open
Abstract
Vaccine-induced immune responses following immunization with promising Chlamydia vaccines protected experimental animals from Chlamydia-induced upper genital tract pathologies and infertility. In contrast, primary genital infection with live Chlamydia does not protect against these pathologies. We hypothesized that differential miRNA profiles induced in the upper genital tracts (UGT) of mice correlate with the disparate immunity vs. pathologic outcomes associated with vaccine immunization and chlamydial infection. Thus, miRNA expression profiles in the UGT of mice after Chlamydia infection (Live EB) and immunization with dendritic cell (DC)-based vaccine (DC vaccine) or VCG-based vaccine (VCG vaccine) were compared using the NanoString nCounter Mouse miRNA assay. Of the 602 miRNAs differentially expressed (DE) in the UGT of immunized and infected mice, we selected 58 with counts >100 and p-values < 0.05 for further analysis. Interestingly, vaccine immunization and Chlamydia infection induced the expression of distinct miRNA profiles with a higher proportion in vaccine-immunized compared to Chlamydia infected mice; DC vaccine (41), VCG vaccine (23), and Live EB (15). Hierarchical clustering analysis showed notable differences in the uniquely DE miRNAs for each experimental group, with DC vaccine showing the highest number (21 up-regulated, five down-regulated), VCG vaccine (two up-regulated, five down-regulated), and live EB (two up-regulated, four down-regulated). The DC vaccine-immunized group showed the highest number (21 up-regulated and five down-regulated compared to two up-regulated and four down-regulated in the live Chlamydia infected group). Pathway analysis showed that the DE miRNAs target genes that regulate several biological processes and functions associated with immune response and inflammation. These results suggest that the induction of differential miRNA expression plays a significant role in the disparate immunity outcomes associated with Chlamydia infection and vaccination.
Collapse
Affiliation(s)
- Simone Howard
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA, United States
| | - Shakyra Richardson
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA, United States
| | - Ifeyinwa Benyeogor
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, United States
| | - Yusuf Omosun
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA, United States
| | - Kamran Dye
- Department of Chemistry, Morehouse College, Atlanta, GA, United States
| | - Fnu Medhavi
- 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
| | - Olayinka Adebayo
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA, United States
| | - Joseph U. Igietseme
- 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
|
19
|
Innate IFN-γ Is Essential for Systemic Chlamydia muridarum Control in Mice, While CD4 T Cell-Dependent IFN-γ Production Is Highly Redundant in the Female Reproductive Tract. Infect Immun 2021; 89:IAI.00541-20. [PMID: 33257535 PMCID: PMC8097277 DOI: 10.1128/iai.00541-20] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 11/17/2020] [Indexed: 12/15/2022] Open
Abstract
Protective immunity against the obligate intracellular bacterium Chlamydia has long been thought to rely on CD4 T cell-dependent gamma interferon (IFN-γ) production. Nevertheless, whether IFN-γ is produced by other cellular sources during Chlamydia infection and how CD4 T cell-dependent and -independent IFN-γ contribute differently to host resistance have not been carefully evaluated. In this study, we dissected the requirements of IFN-γ produced by innate immune cells and CD4 T cells for resolution of Chlamydia muridarum female reproductive tract (FRT) infection. After C. muridarum intravaginal infection, IFN-γ-deficient and T cell-deficient mice exhibited opposite phenotypes for survival and bacterial shedding at the FRT mucosa, demonstrating the distinct requirements for IFN-γ and CD4 T cells in host defense against Chlamydia In Rag1-deficient mice, IFN-γ produced by innate lymphocytes (ILCs) accounted for early bacterial control and prolonged survival in the absence of adaptive immunity. Although type I ILCs are potent IFN-γ producers, we found that mature NK cells and ILC1s were not the sole sources of innate IFN-γ in response to Chlamydia By conducting T cell adoptive transfer, we showed definitively that IFN-γ-deficient CD4 T cells were sufficient for effective bacterial killing in the FRT during the first 21 days of infection and reduced bacterial burden more than 1,000-fold, although mice receiving IFN-γ-deficient CD4 T cells failed to completely eradicate the bacteria from the FRT like their counterparts receiving wild-type (WT) CD4 T cells. Together, our results revealed that innate IFN-γ is essential for preventing systemic Chlamydia dissemination, whereas IFN-γ produced by CD4 T cells is largely redundant at the FRT mucosa.
Collapse
|
20
|
Quispe Calla NE, Vicetti Miguel RD, Aceves KM, Huang H, Howitt B, Cherpes TL. Ovariectomized mice and postmenopausal women exhibit analogous loss of genital epithelial integrity. Tissue Barriers 2021; 9:1865760. [PMID: 33427560 DOI: 10.1080/21688370.2020.1865760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
Abstract
Roughly half of all postmenopausal women are affected by the genitourinary syndrome of menopause (GSM). Symptoms of GSM, including vaginal irritation and dyspareunia, occur as reduced estrogen (E) production elicits loss of elasticity and other changes in genital tract tissue. While the use of the injectable contraceptive depot-medroxyprogesterone acetate (DMPA) likewise lowers serum E concentrations in reproductive age women and is associated with decreased genital levels of the cell-cell adhesion molecules desmoglein-1 (DSG1) and desmocollin-1 (DSC1) and impaired genital epithelial barrier function, the relevance of these findings to women in menopause is uncertain. Exploring the impact of menopause on genital epithelial integrity herein, we detected significantly lower levels of DSG1 and DSC1 in ectocervical tissue from menopausal and postmenopausal vs premenopausal women. Using ovariectomized (OVX) mice as a menopause model, we comparably saw significantly lower vaginal tissue levels of DSG1 and DSC1 in OVX mice vs. mice in estrus. Compared to estrus-stage mice and E-treated OVX mice, DMPA-treated ovary-intact mice and OVX mice also exhibited significantly reduced genital epithelial barrier function, greater susceptibility to genital herpes simplex virus type 2 infection, and delayed clearance of genital Chlamydia trachomatis infection. Current studies thus identify analogous loss of genital epithelial integrity in OVX mice and menopausal and postmenopausal women. By showing that loss of genital epithelial integrity is associated with increased mouse susceptibility to bacterial and viral pathogens, our findings also prioritize the need to resolve if reduced genital epithelial integrity in postmenopausal women is a significant risk factor for genital infection.
Collapse
Affiliation(s)
- Nirk E Quispe Calla
- Department of Comparative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Kristen M Aceves
- Department of Comparative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Huijie Huang
- Department of Comparative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Brooke Howitt
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Thomas L Cherpes
- Department of Comparative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
21
|
Chetty A, Omondi MA, Butters C, Smith KA, Katawa G, Ritter M, Layland L, Horsnell W. Impact of Helminth Infections on Female Reproductive Health and Associated Diseases. Front Immunol 2020; 11:577516. [PMID: 33329545 PMCID: PMC7719634 DOI: 10.3389/fimmu.2020.577516] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 10/27/2020] [Indexed: 12/25/2022] Open
Abstract
A growing body of knowledge exists on the influence of helminth infections on allergies and unrelated infections in the lung and gastrointestinal (GI) mucosa. However, the bystander effects of helminth infections on the female genital mucosa and reproductive health is understudied but important considering the high prevalence of helminth exposure and sexually transmitted infections in low- and middle-income countries (LMICs). In this review, we explore current knowledge about the direct and systemic effects of helminth infections on unrelated diseases. We summarize host disease-controlling immunity of important sexually transmitted infections and introduce the limited knowledge of how helminths infections directly cause pathology to female reproductive tract (FRT), alter susceptibility to sexually transmitted infections and reproduction. We also review work by others on type 2 immunity in the FRT and hypothesize how these insights may guide future work to help understand how helminths alter FRT health.
Collapse
Affiliation(s)
- Alisha Chetty
- Institute of Infectious Disease and Molecular Medicine and Division of Immunology, University of Cape Town, Cape Town, South Africa
| | - Millicent A Omondi
- Institute of Infectious Disease and Molecular Medicine and Division of Immunology, University of Cape Town, Cape Town, South Africa
| | - Claire Butters
- Institute of Infectious Disease and Molecular Medicine and Division of Immunology, University of Cape Town, Cape Town, South Africa
| | - Katherine Ann Smith
- Institute of Infectious Disease and Molecular Medicine and Division of Immunology, University of Cape Town, Cape Town, South Africa.,School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | - Gnatoulma Katawa
- Ecole Supérieure des Techniques Biologiques et Alimentaires, Université de Lomé, Lomé, Togo
| | - Manuel Ritter
- Institute for Medical Microbiology, Immunology and Parasitology (IMMIP), University Hospital Bonn (UKB), Bonn, Germany
| | - Laura Layland
- Institute for Medical Microbiology, Immunology and Parasitology (IMMIP), University Hospital Bonn (UKB), Bonn, Germany
| | - William Horsnell
- Institute of Infectious Disease and Molecular Medicine and Division of Immunology, University of Cape Town, Cape Town, South Africa.,Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
22
|
Stone BJ, Steele KH, Men H, Srodulski SJ, Bryda EC, Fath-Goodin A. A Nonsurgical Embryo Transfer Technique for Fresh and Cultured Blastocysts in Rats. JOURNAL OF THE AMERICAN ASSOCIATION FOR LABORATORY ANIMAL SCIENCE 2020; 59:488-495. [PMID: 32787997 DOI: 10.30802/aalas-jaalas-19-000163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
The use of a nonsurgical embryo transfer technique in rodents eliminates the potential pain, distress, and health complications that may result from a surgical procedure and as such, represents a refinement in rodent assisted reproductive techniques. A nonsurgical technique has not been previously developed for use with rat embryos. Here we describe an efficient method to deliver either fresh or cultured blastocyst stage embryos to the uterine horn of pseudopregnant female rats using a rat nonsurgical embryo transfer (rNSET) device. The rNSET device is composed of a Teflon catheter and a hub that attaches to a 2 μL pipette. Oxytocin is used to dilate the cervix before the delivery of blastocysts, allowing passage of the rNSET catheter directly into the uterine horn for embryo delivery. The efficiency of recovery of pups after nonsurgical embryo transfer is similar to the efficiency after surgical embryo transfer. Furthermore, the technique is not stressful to the subjects, as demonstrated by the absence of a decrease in weight or increase in fecal corticosterone level in recipients of embryos delivered nonsurgically, without the use of anesthesia or analgesia.
Collapse
Affiliation(s)
| | | | - Hongsheng Men
- Rat Resource and Research Center, University of Missouri, Columbia, Missouri; Department of Veterinary Pathobiology, University of Missouri, Columbia, Missouri
| | | | - Elizabeth C Bryda
- Rat Resource and Research Center, University of Missouri, Columbia, Missouri; Department of Veterinary Pathobiology, University of Missouri, Columbia, Missouri
| | | |
Collapse
|
23
|
Modulation of T helper 1 and T helper 2 immune balance in a murine stress model during Chlamydia muridarum genital infection. PLoS One 2020; 15:e0226539. [PMID: 32413046 PMCID: PMC7228091 DOI: 10.1371/journal.pone.0226539] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 04/22/2020] [Indexed: 02/08/2023] Open
Abstract
A murine model to study the effect of cold-induced stress (CIS) on Chlamydia muridarum genital infection and immune response has been developed in our laboratory. Previous results in the lab show that CIS increases the intensity of chlamydia genital infection, but little is known about the effects and mechanisms of CIS on the differentiation and activities of CD4+ T cell subpopulations and bone marrow-derived dendritic cells (BMDCs). The factors that regulate the production of T helper 1 (Th1) or T helper 2 (Th2) cytokines are not well defined. In this study, we examined whether CIS modulates the expressions of beta-adrenergic receptor (β-AR), transcription factors, hallmark cytokines of Th1 and Th2, and differentiation of BMDCs during C. muridarum genital infection in the murine model. Our results show that the mRNA level of the beta2-adrenergic receptor (β2-AR) compared to β1-AR and β3-AR was high in the mixed populations of CD4+ T cells and BMDCs. Furthermore, we observed decreased expression of T-bet, low level of Interferon-gamma (IFN-γ) production, increased expression of GATA-3, and Interleukin-4 (IL-4) production in CD4+ T cells of stressed mice. Exposure of BMDCs to Fenoterol, β2-AR agonist, or ICI118,551, β2-AR antagonist, revealed significant β2-AR stimulation or inhibition, respectively, in stressed mice. Moreover, co-culturing of mature BMDCs and naïve CD4+ T cells increased the production of IL-4, IL-10, L-17, and IL-23 cytokines, suggesting that stimulation of β2-AR leads to the increased production of Th2 cytokines. Overall, our results show for the first time that CIS promotes the switching from a Th1 to Th2 cytokine environment. This was evidenced in the murine stress model by the overexpression of GATA-3 concurrent with elevated IL-4 production, reduced T-bet expression, and IFN-γ secretion.
Collapse
|
24
|
Alyautdina OS, Esina EV. Immunological Methods for Treatment of Vulvovaginal Infections in the Preconception Period. J Med Life 2020; 12:368-373. [PMID: 32025255 PMCID: PMC6993294 DOI: 10.25122/jml-2019-0068] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Traditional therapy and extensive use of medications and intravaginal autolymphocyte therapy show different results of the treatment of vulvovaginal infections. The purpose of the article was to explore safe and highly effective methods to treat vulvovaginal infections and diseases of the pelvic organs. The standard clinical and laboratory screening of 70 patients of reproductive age was carried out to diagnose the diseases of the reproductive tract. The screening included the description of quantitative and qualitative characteristics of vaginal discharge, examining the mucous covering of the vulva and vagina, microscopic examination of Gram-stained vaginal swabs, endocervical cultures, and diagnosis of sexually transmitted infections using polymerase chain reaction. Intravaginal autolymphocyte therapy was used together with traditionally-accepted treatment schemes (etiotropic antibacterial and antifungal therapy) in the treatment of the main group (40 patients). Traditional treatment methods depending on the etiology of the development of infection were used in the control group (30 patients). The IgM, IgA, and IgG levels were also observed because of the possibility of causing embryo rejection. This study shows that in case of relapsing vulvovaginitis and mixed infections accompanied by disorders of the immune system at different levels, the use of intravaginal autolymphocyte therapy in a comprehensive therapy can be assessed as advisable and pathogenetically substantiated.
Collapse
Affiliation(s)
- Olga S Alyautdina
- Department of Obstetrics and Gynecology, Sechenov First Moscow State Medical University, Moscow, Russian Federation
| | | |
Collapse
|
25
|
Malaviarachchi PA, Mercado MAB, McSorley SJ, Li LX. Antibody, but not B-cell-dependent antigen presentation, plays an essential role in preventing Chlamydia systemic dissemination in mice. Eur J Immunol 2020; 50:676-684. [PMID: 32026472 DOI: 10.1002/eji.201948391] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 12/06/2019] [Indexed: 12/16/2022]
Abstract
The obligate intracellular bacterium Chlamydia trachomatis causes the most prevalent bacterial sexually transmitted infection worldwide. CD4 T cells play a central role in the protective immunity against Chlamydia female reproductive tract (FRT) infection, while B cells are thought to be dispensable for resolution of primary Chlamydia infection in mouse models. We recently reported an unexpected requirement of B cells in local Chlamydia-specific CD4 T-cell priming and bacterial containment within the FRT. Here, we sought to tackle the precise effector function of B cells during Chlamydia primary infection. Using mixed bone marrow chimeras that lack B-cell-dependent Ag presentation (MHCIIB - / - ) or devoid of circulating antibodies (AID-/- × μS-/- ), we show that Chlamydia-specific CD4 T-cell expansion does not rely on Ag presentation by B cells. Importantly, we demonstrate that antibody, but not B-cell-dependent Ag presentation, is required for preventing systemic bacterial dissemination following Chlamydia FRT infection.
Collapse
Affiliation(s)
- Priyangi A Malaviarachchi
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Miguel A B Mercado
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Stephen J McSorley
- Center for Comparative Medicine, Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California, Davis, CA, USA
| | - Lin-Xi Li
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| |
Collapse
|
26
|
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
|
27
|
Boddicker MA, Kaufhold RM, Cox KS, Lucas BJ, Xie J, Nahas DD, Touch S, Espeseth AS, Vora KA, Skinner JM. A Novel LNP-Based <i>Chlamydia</i> Subunit Vaccine Formulation That Induces Th1 Responses without Upregulating IL-17 Provides Equivalent Protection in Mice as Formulations That Induced IL-17 and Th1 Cytokines. ACTA ACUST UNITED AC 2020. [DOI: 10.4236/wjv.2020.104005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
28
|
Quispe Calla NE, Vicetti Miguel RD, Glick ME, Kwiek JJ, Gabriel JM, Cherpes TL. Exogenous oestrogen inhibits genital transmission of cell-associated HIV-1 in DMPA-treated humanized mice. J Int AIDS Soc 2019; 21. [PMID: 29334191 PMCID: PMC5810324 DOI: 10.1002/jia2.25063] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Accepted: 01/02/2018] [Indexed: 12/11/2022] Open
Abstract
Introduction HIV affects more women than any other life‐threatening infectious agent, and most infections are sexually transmitted. HIV must breach the female genital tract mucosal barrier to establish systemic infection, and clinical studies indicate virus more easily evades this barrier in women using depot‐medroxyprogesterone acetate (DMPA) and other injectable progestins for contraception. Identifying a potential mechanism for this association, we learned DMPA promotes susceptibility of wild‐type mice to genital herpes simplex virus type 2 (HSV‐2) infection by reducing genital tissue expression of the cell‐cell adhesion molecule desmoglein‐1 (DSG‐1) and increasing genital mucosal permeability. Conversely, DMPA‐mediated increases in genital mucosal permeability and HSV‐2 susceptibility were eliminated in mice concomitantly administered exogenous oestrogen (E). To confirm and extend these findings, herein we used humanized mice to define effects of systemic DMPA and intravaginal (ivag) E administration on susceptibility to genital infection with cell‐associated HIV‐1. Methods Effects of DMPA or an intravaginal (ivag) E cream on engraftment of NOD‐scid‐IL‐2Rgcnull (NSG) mice with human peripheral blood mononuclear cells (hPBMCs) were defined with flow cytometry. Confocal microscopy was used to evaluate effects of DMPA, DMPA and E cream, or DMPA and the pharmacologically active component of the cream on vaginal tissue DSG‐1 expression and genital mucosal permeability to low molecular weight (LMW) molecules and hPBMCs. In other studies, hPBMC‐engrafted NSG mice (hPBMC‐NSG) received DMPA or DMPA and ivag E cream before genital inoculation with 106 HIV‐1‐infected hPBMCs. Mice were euthanized 10 days after infection, and plasma HIV‐1 load quantified by qRT‐PCR and splenocytes used to detect HIV‐1 p24 antigen via immunohistochemistry and infectious virus via TZM‐bl luciferase assay. Results Whereas hPBMC engraftment was unaffected by DMPA or E treatment, mice administered DMPA and E (cream or the pharmacologically active cream component) displayed greater vaginal tissue expression of DSG‐1 protein and decreased vaginal mucosal permeability to LMW molecules and hPBMCs versus DMPA‐treated mice. DMPA‐treated hPBMC‐NSG mice were also uniformly susceptible to genital transmission of cell‐associated HIV‐1, while no animal concomitantly administered DMPA and E cream acquired systemic HIV‐1 infection. Conclusion Exogenous E administration reduces susceptibility of DMPA‐treated humanized mice to genital HIV‐1 infection.
Collapse
Affiliation(s)
- Nirk E Quispe Calla
- Department of Comparative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Melissa E Glick
- The Ohio State University (OSU) College of Veterinary Medicine, Columbus, OH, USA
| | - Jesse J Kwiek
- Department of Microbiology, OSU College of Arts and Sciences, Columbus, OH, USA
| | | | - Thomas L Cherpes
- Department of Comparative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
29
|
Jolly AL, Rau S, Chadha AK, Abdulraheem EA, Dean D. Stromal Fibroblasts Drive Host Inflammatory Responses That Are Dependent on Chlamydia trachomatis Strain Type and Likely Influence Disease Outcomes. mBio 2019; 10:e00225-19. [PMID: 30890604 PMCID: PMC6426598 DOI: 10.1128/mbio.00225-19] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 02/01/2019] [Indexed: 01/11/2023] Open
Abstract
Chlamydia trachomatis ocular strains cause a blinding disease known as trachoma. These strains rarely cause urogenital infections and are not found in the upper genital tract or rectum. Urogenital strains are responsible for a self-limited conjunctivitis and the sequelae of infertility, ectopic pregnancy, and hemorrhagic proctitis. However, the differential cellular responses that drive these clinically observed disease outcomes are not completely understood. Primary conjunctival, endocervical, and endometrial epithelial and stromal fibroblast cells, HeLa229 cells, and immortalized conjunctival epithelial (HCjE) cells were infected with the ocular A/Har-13 (A) and Ba/Apache-2 (Ba) strains and urogenital D/UW-3 (D) and E/Bour (E) strains. Infection rates, progeny production, and cytokine/chemokine secretion levels were evaluated in comparison with those in uninfected cells. All strain types infected all cell types with similar levels of efficacy and development. However, progeny production levels differed among primary cells: Ba produced significantly more progeny than E in endocervical and endometrial fibroblasts, while A progeny were less abundant than E progeny. C.trachomatis infection of primary epithelial cells elicited an increase in pro- and anti-inflammatory mediators compared to levels in uninfected cells, but there were no significant differences by strain type. In contrast, for primary fibroblasts, ocular strains elicited significant increases in the pro- and anti-inflammatory mediators macrophage inflammatory protein (MIP)-1β, thymus- and activation-regulated chemokine (TARC), interleukin (IL)-2, IL-12p70, and interferon gamma-induced protein 10 (IP-10) compared to levels in urogenital strains, while urogenital strains elicited a distinct and significant increase in the proinflammatory mediators IL-1α, IL-1β, IL-8, gamma interferon (IFN-γ), and granulocyte-macrophage colony-stimulating factor (GM-CSF). Our data indicate that primary fibroblasts, not epithelial cells, drive host inflammatory responses that are dependent on strain type and likely influence disease outcomes, establishing their importance as a novel model for studies of C. trachomatis disease pathogenesis.IMPORTANCEChlamydia trachomatis is a human pathogen and the leading cause of preventable blindness and sexually transmitted diseases in the world. Certain C. trachomatis strains cause ocular disease, while others cause upper genital tract pathology. However, little is known about the cellular or immunologic basis for these differences. Here, we compared the abilities of the strain types to infect, replicate, and initiate an immune response in primary human ocular and urogenital epithelial cells, as well as in fibroblasts from the underlying stroma. While there were no significant differences in infection rates or intracellular growth for any strain in any cell type, proinflammatory responses were driven not by the epithelial cells but by fibroblasts and were distinct between ocular and urogenital strains. Our findings suggest that primary fibroblasts are a novel and more appropriate model for studies of immune responses that will expand our understanding of the differential pathological disease outcomes caused by various C. trachomatis strain types.
Collapse
Affiliation(s)
- Amber Leah Jolly
- Center for Immunobiology and Vaccine Development, UCSF Benioff Children's Hospital Oakland Research Institute, Oakland, California, USA
| | - Sameeha Rau
- Center for Immunobiology and Vaccine Development, UCSF Benioff Children's Hospital Oakland Research Institute, Oakland, California, USA
| | - Anmol K Chadha
- Center for Immunobiology and Vaccine Development, UCSF Benioff Children's Hospital Oakland Research Institute, Oakland, California, USA
| | - Ekhlas Ahmed Abdulraheem
- Center for Immunobiology and Vaccine Development, UCSF Benioff Children's Hospital Oakland Research Institute, Oakland, California, USA
| | - Deborah Dean
- Center for Immunobiology and Vaccine Development, UCSF Benioff Children's Hospital Oakland Research Institute, Oakland, California, USA
- Department of Bioengineering, University of California at Berkeley, Berkeley, California, USA
- Department of Medicine and Pediatrics, University of California at San Francisco, San Francisco, California, USA
| |
Collapse
|
30
|
Kaufhold RM, Boddicker MA, Field JA, Lucas BJ, Antonello JM, Espeseth AS, Skinner JM, Heinrichs JH, Smith JG. Evaluating Potential Vaccine Antigens in both the <i>Chlamydia trachomatis</i> and <i>Chlamydia muridarum</i> Intravaginal Mouse Challenge Models. ACTA ACUST UNITED AC 2019. [DOI: 10.4236/wjv.2019.92004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
31
|
Vicetti Miguel RD, Quispe Calla NE, Cherpes TL. Levonorgestrel and Female Genital Tract Immunity: Time for a Closer Look. J Infect Dis 2018; 218:1517-1518. [PMID: 29917111 PMCID: PMC6151086 DOI: 10.1093/infdis/jiy363] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 06/12/2018] [Indexed: 01/09/2023] Open
Affiliation(s)
| | - Nirk E Quispe Calla
- Department of Comparative Medicine, Stanford University School of Medicine, California
| | - Thomas L Cherpes
- Department of Comparative Medicine, Stanford University School of Medicine, California
| |
Collapse
|
32
|
Bakshi RK, Gupta K, Jordan SJ, Chi X, Lensing SY, Press CG, Geisler WM. An Adaptive Chlamydia trachomatis-Specific IFN-γ-Producing CD4 + T Cell Response Is Associated With Protection Against Chlamydia Reinfection in Women. Front Immunol 2018; 9:1981. [PMID: 30245688 PMCID: PMC6137090 DOI: 10.3389/fimmu.2018.01981] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 08/13/2018] [Indexed: 01/20/2023] Open
Abstract
Background: Adaptive immune responses that mediate protection against Chlamydia trachomatis (CT) remain poorly defined in humans. Animal chlamydia models have demonstrated that CD4+ Th1 cytokine responses mediate protective immunity against reinfection. To better understand protective immunity to CT in humans, we investigated whether select CT-specific CD4+ Th1 and CD8+ T cell cytokine responses were associated with protection against CT reinfection in women. Methods: Peripheral blood mononuclear cells were collected from 135 CT-infected women at treatment and follow-up visits and stimulated with CT antigens. CD4+ and CD8+ T-cells expressing IFN-γ, TNF-α, and/or IL-2 were assessed using intracellular cytokine staining and cytokine responses were compared between visits and between women with vs. without CT reinfection at follow-up. Results: A CD4+TNF-α response was detected in the majority (77%) of study participants at the treatment visit, but a lower proportion had this response at follow-up (62%). CD4+ IFN-γ and CD4+ IL-2 responses occurred less frequently at the treatment visit (32 and 18%, respectively), but increased at follow-up (51 and 41%, respectively). CD8+ IFN-γ and CD8+ TNF-α responses were detected more often at follow-up (59% for both responses) compared to the treatment visit (30% for both responses). At follow-up, a CD4+IFN-γ response was detected more often in women without vs. with reinfection (60 vs. 33%, P = 0.005). Conclusions: Our findings suggest that a CT-specific CD4+ IFN-γ response is associated with protective immunity against CT reinfection and is thus an important component of adaptive immunity to CT in women.
Collapse
Affiliation(s)
- Rakesh K Bakshi
- Division of Infectious Diseases, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Kanupriya Gupta
- Division of Infectious Diseases, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Stephen J Jordan
- Division of Infectious Diseases, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Xiaofei Chi
- Department of Biostatistics, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Shelly Y Lensing
- Department of Biostatistics, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Christen G Press
- Division of Infectious Diseases, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - William M Geisler
- Division of Infectious Diseases, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
33
|
Labuda JC, McSorley SJ. Diversity in the T cell response to Chlamydia-sum are better than one. Immunol Lett 2018; 202:59-64. [PMID: 30179654 DOI: 10.1016/j.imlet.2018.08.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 08/10/2018] [Accepted: 08/15/2018] [Indexed: 02/06/2023]
Abstract
Chlamydia trachomatis is responsible for an increasing number of sexually transmitted infections in the United States and is a common cause of serious pathology in the female reproductive tract (FRT). Given the impact and incidence of these infections, the production of an effective Chlamydia vaccine is a public health priority. Mouse models of Chlamydia infection have been utilized to develop a detailed and mechanistic understanding of protective immunity in the FRT. These studies reveal that MHC class-II restricted Chlamydia-specific CD4 T cells are critical for primary bacterial clearance and provide effective protection against secondary infection in the FRT. Despite the clear importance of IFN- γ produced by CD4 Th1 cells, there are also suggestions of wider functional heterogeneity in the CD4 T cell response to Chlamydia infection. Understanding the role of this diversity in the CD4 T helper cell response in the FRT should allow a more nuanced view of CD4 T cell biology in the context of Chlamydia infection and may be critical for vaccine development. Here, we summarize our current understanding of CD4 T helper subsets in the clearance of Chlamydia and discuss some areas where knowledge needs to be further extended by additional experimentation.
Collapse
Affiliation(s)
- Jasmine C Labuda
- 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
|
34
|
Stanfield BA, Rider PJF, Caskey J, Del Piero F, Kousoulas KG. Intramuscular vaccination of guinea pigs with the live-attenuated human herpes simplex vaccine VC2 stimulates a transcriptional profile of vaginal Th17 and regulatory Tr1 responses. Vaccine 2018; 36:2842-2849. [PMID: 29655629 DOI: 10.1016/j.vaccine.2018.03.075] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 03/23/2018] [Accepted: 03/27/2018] [Indexed: 12/31/2022]
Abstract
Herpes simplex virus is a common causative agent of oral and genital diseases. Novel vaccines and therapeutics are needed to combat herpes infections especially after the failure of subunit vaccines in human clinical trials. We have shown that the live-attenuated HSV-1 VC2 vaccine strain is unable to establish latency in vaccinated animals and produces a robust immune response capable of completely protecting mice against lethal vaginal HSV-1 or HSV-2 infections. The guinea pig represents the best small animal model of genital HSV-2 disease. Reported here, twenty-one female Hartley guinea pigs received intramuscular injection with either the VC2 vaccine, or equal volume of conditioned tissue culture media. Animals received 2 booster vaccinations at 21 day intervals following the initial vaccination. After vaccination, animals were challenged with the highly virulent HSV-2 (G) strain. Histologically, VC2 vaccinated animals had little to no apparent inflammation/disease following challenge. Unvaccinated animals developed moderate to severe erosive and ulcerative vaginitis. Quantitative reverse-transcriptase PCR analysis in VC2 vaccinated and challenged animals identified transcriptional signatures of Th17 and regulatory Tr1 cells associated with the inflammatory response primed by VC2 vaccination. Treatment of cultured human vaginal epithelial cells (VK2 cells) with a combination of IL-17A and IL-22 resulted in the significant induction of beta-defensin 3 expression. Further, treatment of VK2 cells with IL-17A, IL-22, IL-36 or beta-defensin 3 resulted in diminished HSV-2 replication. Overall, these results suggest that intramuscular vaccination with the live-attenuated vaccine VC2 primes a mucosal immune response predisposing the adaptive expression of transcripts associated with a Th17 response to challenge and these responses contribute to antiviral immunity.
Collapse
Affiliation(s)
- Brent A Stanfield
- Department of Pathobiological Sciences and Division of Biotechnology and Molecular Medicine, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Paul J F Rider
- Department of Pathobiological Sciences and Division of Biotechnology and Molecular Medicine, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA
| | - John Caskey
- Department of Pathobiological Sciences and Division of Biotechnology and Molecular Medicine, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Fabio Del Piero
- Department of Pathobiological Sciences and Division of Biotechnology and Molecular Medicine, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Konstantin G Kousoulas
- Department of Pathobiological Sciences and Division of Biotechnology and Molecular Medicine, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA.
| |
Collapse
|
35
|
V γ4+ T Cells: A Novel IL-17-Producing γδ T Subsets during the Early Phase of Chlamydial Airway Infection in Mice. Mediators Inflamm 2018; 2018:6265746. [PMID: 29670466 PMCID: PMC5835244 DOI: 10.1155/2018/6265746] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Accepted: 12/18/2017] [Indexed: 12/16/2022] Open
Abstract
Our previous studies showed that γδ T cells provided immune protection against Chlamydial muridarum (Cm), an obligate intracellular strain of chlamydia trachomatis, lung infection by producing abundant IL-17. In this study, we investigated the proliferation and activation of lung γδ T cell subsets, specifically the IL-17 and IFNγ production by them following Cm lung infection. Our results found that five γδ T cell subsets, Vγ1+ T, Vγ2+ T, Vγ4+ T, Vγ5+ T, and Vγ6+ T, expressed in lungs of naïve mice, while Cm lung infection mainly induced the proliferation and activation of Vγ4+ T cells at day 3 p.i., following Vγ1+ T cells at day 7 p.i. Cytokine detection showed that Cm lung infection induced IFNγ secretion firstly by Vγ4+ T cells at very early stage (day 3) and changed to Vγ1+ T cells at midstage (day 7). Furthermore, Vγ4+ T cell is the main γδ T cell subset that secretes IL-17 at the very early stage of Cm lung infection and Vγ1+ T cell did not secrete IL-17 during the infection. These findings provide in vivo evidence that Vγ4+T cells are the major IL-17 and IFNγ-producing γδ T cell subsets at the early period of Cm lung infection.
Collapse
|
36
|
Novel Role for Interleukin-17 in Enhancing Type 1 Helper T Cell Immunity in the Female Genital Tract following Mucosal Herpes Simplex Virus 2 Vaccination. J Virol 2017; 91:JVI.01234-17. [PMID: 28956763 DOI: 10.1128/jvi.01234-17] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 09/18/2017] [Indexed: 12/17/2022] Open
Abstract
It is well established that interferon gamma (IFN-γ) production by CD4+ T cells is critical for antiviral immunity against herpes simplex virus 2 (HSV-2) genital infection. However, the role of interleukin-17A (IL-17A) production by CD4+ T cells in HSV-2 antiviral immunity is yet to be elucidated. Here we demonstrate that IL-17A plays an important role in enhancing antiviral T helper type 1 (Th1) responses in the female genital tract (FGT) and is essential for effective protection conferred by HSV-2 vaccination. While IL-17A did not play a critical role during primary genital HSV-2 infection, seen by lack of differences in susceptibility between IL-17A-deficient (IL-17A-/-) and wild-type (WT) C57BL/6 mice, it was critical for mediating antiviral responses after challenge/reexposure. Compared to WT mice, IL-17A-/- mice (i) infected intravaginally and reexposed or (ii) vaccinated intranasally and challenged intravaginally demonstrated poor outcomes. Following intravaginal HSV-2 reexposure or challenge, vaccinated IL-17A-/- mice had significantly higher mortality, greater disease severity, higher viral shedding, and higher levels of proinflammatory cytokines and chemokines in vaginal secretions. Furthermore, IL-17A-/- mice had impaired Th1 cell responses after challenge/reexposure, with significantly lower proportions of vaginal IFN-γ+ CD4+ T cells. The impaired Th1 cell responses in IL-17A-/- mice coincided with smaller populations of IFN-γ+ CD4+ tissue resident memory T (TRM) cells in the genital tract postimmunization. Taken together, these findings describe a novel role for IL-17A in regulating antiviral IFN-γ+ Th1 cell immunity in the vaginal tract. This strategy could be exploited to enhance antiviral immunity following HSV-2 vaccination.IMPORTANCE T helper type 1 (Th1) immunity, specifically interferon gamma (IFN-γ) production by CD4+ T cells, is critical for protection against genital herpesvirus (HSV-2) infection, and enhancing this response can potentially help improve disease outcomes. Our study demonstrated that interleukin-17A (IL-17A) plays an essential role in enhancing antiviral Th1 responses in the female genital tract (FGT). We found that in the absence of IL-17A, preexposed and vaccinated mice showed poor disease outcomes and were unable to overcome HSV-2 reexposure/challenge. IL-17A-deficient mice (IL-17A-/-) had smaller populations of IFN-γ+ CD4+ tissue resident memory T (TRM) cells in the genital tract postimmunization than did wild-type (WT) mice, which coincided with attenuated Th1 responses postchallenge. This has important implications for developing effective vaccines against HSV-2, as we propose that strategies inducing IL-17A in the genital tract may promote more effective Th1 cell immunity and better overall protection.
Collapse
|
37
|
Ramadhani AM, Derrick T, Macleod D, Massae P, Mtuy T, Jeffries D, Roberts CH, Bailey RL, Mabey DCW, Holland MJ, Burton MJ. Immunofibrogenic Gene Expression Patterns in Tanzanian Children with Ocular Chlamydia trachomatis Infection, Active Trachoma and Scarring: Baseline Results of a 4-Year Longitudinal Study. Front Cell Infect Microbiol 2017; 7:406. [PMID: 28966918 PMCID: PMC5605569 DOI: 10.3389/fcimb.2017.00406] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 08/31/2017] [Indexed: 12/11/2022] Open
Abstract
Trachoma, caused by Chlamydia trachomatis, is the world's leading infectious cause of blindness and remains a significant public health problem. Much of trachomatous disease pathology is thought to be caused indirectly by host cellular and immune responses, however the immune response during active trachoma and how this initiates progressive scarring is not clearly understood. Defining protective vs. pathogenic immune response to C. trachomatis is important for vaccine design and evaluation. This study reports the baseline results of a longitudinal cohort of Tanzanian children, who were monitored for 4 years in order to determine the immunofibrogenic and infectious correlates of progressive scarring trachoma. In this cohort baseline, 506 children aged 6-10 years were assessed for clinical signs, infection status and the expression of 91 genes of interest prior to mass azithromycin administration for trachoma control. C. trachomatis was detected using droplet digital PCR and gene expression was measured using quantitative real-time PCR. The prevalence of follicles, papillary inflammation and scarring were 33.6, 31.6, and 28.5%, respectively. C. trachomatis was detected in 78/506 (15.4%) individuals, 62/78 of whom also had follicles. C. trachomatis infection was associated with a strong upregulation of IFNG and IL22, the enrichment of Th1 and NK cell pathways and Th17 cell-associated cytokines. In individuals with inflammation in the absence of infection the IFNG/IL22 and NK cell response was reduced, however, pro-inflammatory, growth and matrix factors remained upregulated and mucins were downregulated. Our data suggest that, strong IFNG/IL22 responses, probably related to Th1 and NK cell involvement, is important for clearance of C. trachomatis and that the residual pro-inflammatory and pro-fibrotic phenotype that persists after infection might contribute to pathological scarring. Interestingly, females appear more susceptible to developing papillary inflammation and scarring than males, even at this young age, despite comparable levels of C. trachomatis infection. Females also had increased expression of a number of IFNγ pathway related genes relative to males, suggesting that overexpression of this pathway in response to infection might contribute to more severe scarring. Longitudinal investigation of these factors will reveal their relative contributions to protection from C. trachomatis infection and development of scarring complications.
Collapse
Affiliation(s)
- Athumani M Ramadhani
- Clinical Research Department, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical MedicineLondon, United Kingdom
- Kilimanjaro Christian Medical CentreMoshi, Tanzania
| | - Tamsyn Derrick
- Clinical Research Department, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical MedicineLondon, United Kingdom
- Kilimanjaro Christian Medical CentreMoshi, Tanzania
| | - David Macleod
- Department of Infectious Disease Epidemiology, Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical MedicineLondon, United Kingdom
| | | | - Tara Mtuy
- Clinical Research Department, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical MedicineLondon, United Kingdom
- Kilimanjaro Christian Medical CentreMoshi, Tanzania
| | - David Jeffries
- Support Services (Statistics), Medical Research Council Unit The GambiaFajara, Gambia
| | - Chrissy H Roberts
- Clinical Research Department, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical MedicineLondon, United Kingdom
| | - Robin L Bailey
- Clinical Research Department, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical MedicineLondon, United Kingdom
| | - David C W Mabey
- Clinical Research Department, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical MedicineLondon, United Kingdom
| | - Martin J Holland
- Clinical Research Department, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical MedicineLondon, United Kingdom
| | - Matthew J Burton
- Clinical Research Department, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical MedicineLondon, United Kingdom
| |
Collapse
|
38
|
IL-4-secreting eosinophils promote endometrial stromal cell proliferation and prevent Chlamydia-induced upper genital tract damage. Proc Natl Acad Sci U S A 2017; 114:E6892-E6901. [PMID: 28765368 DOI: 10.1073/pnas.1621253114] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Genital Chlamydia trachomatis infections in women typically are asymptomatic and do not cause permanent upper genital tract (UGT) damage. Consistent with this presentation, type 2 innate and TH2 adaptive immune responses associated with dampened inflammation and tissue repair are elicited in the UGT of Chlamydia-infected women. Primary C. trachomatis infection of mice also causes no genital pathology, but unlike women, does not generate Chlamydia-specific TH2 immunity. Herein, we explored the significance of type 2 innate immunity for restricting UGT tissue damage in Chlamydia-infected mice, and in initial studies intravaginally infected wild-type, IL-10-/-, IL-4-/-, and IL-4Rα-/- mice with low-dose C. trachomatis inoculums. Whereas Chlamydia was comparably cleared in all groups, IL-4-/- and IL-4Rα-/- mice displayed endometrial damage not seen in wild-type or IL-10-/- mice. Congruent with the aberrant tissue repair in mice with deficient IL-4 signaling, we found that IL-4Rα and STAT6 signaling mediated IL-4-induced endometrial stromal cell (ESC) proliferation ex vivo, and that genital administration of an IL-4-expressing adenoviral vector greatly increased in vivo ESC proliferation. Studies with IL-4-IRES-eGFP (4get) reporter mice showed eosinophils were the main IL-4-producing endometrial leukocyte (constitutively and during Chlamydia infection), whereas studies with eosinophil-deficient mice identified this innate immune cell as essential for endometrial repair during Chlamydia infection. Together, our studies reveal IL-4-producing eosinophils stimulate ESC proliferation and prevent Chlamydia-induced endometrial damage. Based on these results, it seems possible that the robust type 2 immunity elicited by Chlamydia infection of human genital tissue may analogously promote repair processes that reduce phenotypic disease expression.
Collapse
|
39
|
Pais R, Omosun Y, He Q, Blas-Machado U, Black C, Igietseme JU, Fujihashi K, Eko FO. Rectal administration of a chlamydial subunit vaccine protects against genital infection and upper reproductive tract pathology in mice. PLoS One 2017; 12:e0178537. [PMID: 28570663 PMCID: PMC5453548 DOI: 10.1371/journal.pone.0178537] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Accepted: 05/15/2017] [Indexed: 11/18/2022] Open
Abstract
In this study, we tested the hypothesis that rectal immunization with a VCG-based chlamydial vaccine would cross-protect mice against heterologous genital Chlamydia trachomatis infection and Chlamydia-induced upper genital tract pathologies in mice. Female mice were immunized with a C. trachomatis serovar D-derived subunit vaccine or control or live serovar D elementary bodies (EBs) and the antigen-specific mucosal and systemic immune responses were characterized. Vaccine efficacy was determined by evaluating the intensity and duration of genital chlamydial shedding following intravaginal challenge with live serovar E chlamydiae. Protection against upper genital tract pathology was determined by assessing infertility and tubal inflammation. Rectal immunization elicited high levels of chlamydial-specific IFN-gamma-producing CD4 T cells and humoral immune responses in mucosal and systemic tissues. The elicited immune effectors cross-reacted with the serovar E chlamydial antigen and reduced the length and intensity of genital chlamydial shedding. Furthermore, immunization with the VCG-vaccine but not the rVCG-gD2 control reduced the incidence of tubal inflammation and protected mice against Chlamydia-induced infertility. These results highlight the potential of rectal immunization as a viable mucosal route for inducing protective immunity in the female genital tract.
Collapse
Affiliation(s)
- Roshan Pais
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, Georgia, United States of America
| | - Yusuf Omosun
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, Georgia, United States of America
| | - Qing He
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, Georgia, United States of America
| | - Uriel Blas-Machado
- Veterinary Diagnostic Laboratory, College of Veterinary Medicine, The University of Georgia, Athens, Georgia, United States of America
| | - Carolyn Black
- Centers for Disease Control and Prevention (CDC), Atlanta, Georgia, United States of America
| | - Joseph U. Igietseme
- Centers for Disease Control and Prevention (CDC), Atlanta, Georgia, United States of America
| | - Kohtaro Fujihashi
- Department of Pediatric Dentistry, Immunobiology Vaccine Center, The University of Alabama at Birmingham, Birmingham Alabama, United States of America
| | - Francis O. Eko
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, Georgia, United States of America
| |
Collapse
|