1
|
Chen X, Fu Y, Deng H, Li P, Zhao W, Shao L, Liu Y, Wang H, Hou S. Pgp3 monoclonal antibody inhibits the pathogenicity of Chlamydia muridarum to the genital tract of mice. Int Immunopharmacol 2025; 148:114039. [PMID: 39837015 DOI: 10.1016/j.intimp.2025.114039] [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: 07/30/2024] [Revised: 11/28/2024] [Accepted: 01/05/2025] [Indexed: 01/23/2025]
Abstract
BACKGROUND Chlamydia trachomatis (Ct) is the leading cause of tubal inflammation in women, with a high tendency for persistent asymptomatic infections. Antibiotics are currently the primary treatment for Ct infections of the reproductive tract. However, mounting evidence indicates an increasing incidence of persistent infections and recurrence due to antibiotic treatment failure, highlighting the urgent need for novel therapeutic approaches. METHODS In this study, a monoclonal antibody against plasmid-encoded protein Pgp3 was prepared using hybridoma technology and its effects on the pathogenicity of Ct were investigated both in vitro and in vivo. RESULTS Infectivity of Chlamydia muridarum (Cm) elementary bodies (EBs) increased after incubation with His-Pgp3. When Pgp3mAb-pretreated Ct EBs or Cm-infected cell lysates were used to inoculate HeLa cells, a significantly reduced number of inclusions was observed compared with untreated controls. Cm-infected HeLa cells began to secrete Pgp3 after 6 h. Infection with Cm progeny was significantly inhibited by the addition of Pgp3mAb co-cultured during the first developmental cycle of Cm. Immunofluorescence assays revealed that Pgp3mAb could not enter the host cells. His-Pgp3 stimulated the secretion of IL-6 and IL-8 in human fallopian tube epithelial cells, while Pgp3mAb inhibited this pro-inflammatory effect of His-Pgp3. Cm-infected mice subcutaneously injected with Pgp3mAb demonstrated reduced shedding of live organisms in the lower genital tract, shorter infection cycles, reduced hydrosalpinx, and a reduced inflammatory response. CONCLUSION Pgp3 enhanced Cm infectivity in host cells. In vitro, Pgp3mAb inhibited Cm infection by binding to secreted Pgp3 and membrane-bound Pgp3, with a more pronounced effect on secreted Pgp3. Furthermore, Pgp3mAb inhibited the pro-inflammatory effects of Pgp3, thereby attenuating the inflammatory response. Subcutaneous administration of Pgp3mAb effectively reduced Cm-induced pathogenicity in the murine reproductive tract.
Collapse
Affiliation(s)
- Xiuqi Chen
- Department of Dermatovenereology, Tianjin Medical University General Hospital/Tianjin Institute of Sexually Transmitted Disease, Tianjin 300052, China
| | - Yujie Fu
- Department of Dermatovenereology, Tianjin Medical University General Hospital/Tianjin Institute of Sexually Transmitted Disease, Tianjin 300052, China
| | - Han Deng
- Department of Dermatovenereology, Tianjin Medical University General Hospital/Tianjin Institute of Sexually Transmitted Disease, Tianjin 300052, China
| | - Pinglu Li
- Department of Dermatovenereology, Tianjin Medical University General Hospital/Tianjin Institute of Sexually Transmitted Disease, Tianjin 300052, China
| | - Wanxing Zhao
- Department of Dermatovenereology, Tianjin Medical University General Hospital/Tianjin Institute of Sexually Transmitted Disease, Tianjin 300052, China
| | - Lili Shao
- Department of Dermatovenereology, Tianjin Medical University General Hospital/Tianjin Institute of Sexually Transmitted Disease, Tianjin 300052, China
| | - Yuanjun Liu
- Department of Dermatovenereology, Tianjin Medical University General Hospital/Tianjin Institute of Sexually Transmitted Disease, Tianjin 300052, China
| | - Huiping Wang
- Department of Dermatovenereology, Tianjin Medical University General Hospital/Tianjin Institute of Sexually Transmitted Disease, Tianjin 300052, China
| | - Shuping Hou
- Department of Dermatovenereology, Tianjin Medical University General Hospital/Tianjin Institute of Sexually Transmitted Disease, Tianjin 300052, China.
| |
Collapse
|
2
|
Tian Q, Zhang T, Shu C, Han Z, Huang Y, Wan J, Wang L, Sun X. Diverse animal models for Chlamydia infections: unraveling pathogenesis through the genital and gastrointestinal tracts. Front Microbiol 2024; 15:1386343. [PMID: 38605708 PMCID: PMC11007077 DOI: 10.3389/fmicb.2024.1386343] [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: 02/15/2024] [Accepted: 03/18/2024] [Indexed: 04/13/2024] Open
Abstract
Chlamydia trachomatis is responsible for infections in various mucosal tissues, including the eyes, urogenital, respiratory, and gastrointestinal tracts. Chronic infections can result in severe consequences such as blindness, ectopic pregnancy, and infertility. The underlying mechanisms leading to these diseases involve sustained inflammatory responses, yet thorough comprehension of the underlying mechanisms remains elusive. Chlamydial biologists employ in multiple methods, integrating biochemistry, cell biology, and genetic tools to identify bacterial factors crucial for host cell interactions. While numerous animal models exist to study chlamydial pathogenesis and assess vaccine efficacy, selecting appropriate models for biologically and clinically relevant insights remains a challenge. Genital infection models in animals have been pivotal in unraveling host-microbe dynamics, identifying potential chlamydial virulence factors influencing genital pathogenicity. However, the transferability of this knowledge to human pathogenic mechanisms remains uncertain. Many putative virulence factors lack assessment in optimal animal tissue microenvironments, despite the diverse chlamydial infection models available. Given the propensity of genital Chlamydia to spread to the gastrointestinal tract, investigations into the pathogenicity and immunological impact of gut Chlamydia become imperative. Notably, the gut emerges as a promising site for both chlamydial infection vaccination and pathogenesis. This review elucidates the pathogenesis of Chlamydia infections and delineates unique features of prevalent animal model systems. The primary focus of this review is to consolidate and summarize current animal models utilized in Chlamydia researches, presenting findings, discussions on their contributions, and suggesting potential directions for further studies.
Collapse
Affiliation(s)
- Qi Tian
- Department of Obstetrics & Gynecology, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, Hunan, China
- National Health Commission Key Laboratory for Birth Defect Research and Prevention, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, Hunan, China
| | - Tianyuan Zhang
- Key Lab of Molecular Virology and Immunology, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai, China
| | - Chuqiang Shu
- Department of Obstetrics & Gynecology, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, Hunan, China
- National Health Commission Key Laboratory for Birth Defect Research and Prevention, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, Hunan, China
| | - Zixuan Han
- Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - Youyou Huang
- Department of Obstetrics & Gynecology, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, Hunan, China
| | - Jiao Wan
- Department of Obstetrics & Gynecology, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, Hunan, China
| | - Luying Wang
- Department of Obstetrics and Gynecology, 3rd Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xin Sun
- Department of Obstetrics and Gynecology, 3rd Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
3
|
He Y, Wang Y, He R, Abdelsalam AM, Zhong G. IL-23 receptor signaling licenses group 3-like innate lymphoid cells to restrict a live-attenuated oral Chlamydia vaccine in the gut. Infect Immun 2023; 91:e0037123. [PMID: 37850749 PMCID: PMC10652955 DOI: 10.1128/iai.00371-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Accepted: 09/14/2023] [Indexed: 10/19/2023] Open
Abstract
An IFNγ-susceptible mutant of Chlamydia muridarum is attenuated in pathogenicity in the genital tract and was recently licensed as an intracellular Oral vaccine vector or intrOv. Oral delivery of intrOv induces transmucosal protection in the genital tract, but intrOv itself is cleared from the gut (without shedding any infectious particles externally) by IFNγ from group 3-like innate lymphoid cells (ILC3s). We further characterized the intrOv interactions with ILC3s in the current study, since the interactions may impact both the safety and efficacy of intrOv as an oral Chlamydia vaccine. Intracolonic inoculation with intrOv induced IFNγ that in return inhibited intrOv. The intrOv-IFNγ interactions were dependent on RORγt, a signature transcriptional factor of ILC3s. Consistently, the transfer of oral intrOv-induced ILC3s from RORγt-GFP reporter mice to IFNγ-deficient mice rescued the inhibition of intrOv. Thus, IFNγ produced by intrOv-induced ILC3s is likely responsible for inhibiting intrOv, which is further supported by the observation that oral intrOv did induce significant levels of IFNγ-producing LC3s (IFNγ+ILC3s). Interestingly, IL-23 receptor knockout (IL-23R-/-) mice no longer inhibited intrOv, which was accompanied by reduced colonic IFNγ. Transfer of oral intrOv-induced ILC3s rescued the IL-23R-/- mice to inhibit intrOv, validating the dependence of ILC3s on IL-23R signaling for inhibiting intrOv. Clearly, intrOv induces intestinal IFNγ+ILC3s for its own inhibition in the gut, which is facilitated by IL-23R signaling. These findings have provided a mechanism for ensuring the safety of intrOv as an oral Chlamydia vaccine and a platform for investigating how oral intrOv induces transmucosal protection in the genital tract.
Collapse
Affiliation(s)
- Ying He
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yihui Wang
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Rongze He
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Ahmed Mohamed Abdelsalam
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Guangming Zhong
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| |
Collapse
|
4
|
Lu C, Wang J, Zhong G. Preclinical screen for protection efficacy of chlamydial antigens that are immunogenic in humans. Infect Immun 2023; 91:e0034923. [PMID: 37889004 PMCID: PMC10652899 DOI: 10.1128/iai.00349-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 10/02/2023] [Indexed: 10/28/2023] Open
Abstract
To search for subunit vaccine candidates, immunogenic chlamydial antigens identified in humans were evaluated for protection against both infection and pathology in a mouse genital tract infection model under three different immunization regimens. The intramuscular immunization regimen was first used to evaluate 106 chlamydial antigens, which revealed that two antigens significantly reduced while 11 increased genital chlamydial burden. The two infection-reducing antigens failed to prevent pathology and 23 additional antigens even exacerbated pathology. Thus, intranasal mucosal immunization was tested next since intranasal inoculation with live Chlamydia muridarum prevented both genital infection and pathology. Two of the 29 chlamydial antigens evaluated were found to prevent genital infection but not pathology and three exacerbate pathology. To further improve protection efficacy, a combinational regimen (intranasal priming + intramuscular boosting + a third intraperitoneal/subcutaneous boost) was tested. This regimen identified four infection-reducing antigens, but only one of them prevented pathology. Unfortunately, this protective antigen was not advanced further due to its amino acid sequence homology with several human molecules. Two pathology-exacerbating antigens were also found. Nevertheless, intranasal mucosal priming with viable C. muridarum in control groups consistently prevented both genital infection and pathology regardless of the subsequent boosters. Thus, screening 140 different chlamydial antigens with 21 repeated multiple times in 17 experiments failed to identify a subunit vaccine candidate but demonstrated the superiority of viable chlamydial organisms in inducing immunity against both genital infection and pathology, laying the foundation for developing a live-attenuated Chlamydia vaccine.
Collapse
Affiliation(s)
- Chunxue Lu
- Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hengyang, Hunan, China
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center, San Antonio, Texas, USA
| | - Jie Wang
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center, San Antonio, Texas, USA
- Department of Immunology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Guangming Zhong
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center, San Antonio, Texas, USA
| |
Collapse
|
5
|
Wang Y, He R, Winner H, Gauduin MC, Zhang N, He C, Zhong G. Induction of Transmucosal Protection by Oral Vaccination with an Attenuated Chlamydia. Infect Immun 2023; 91:e0004323. [PMID: 37036335 PMCID: PMC10187116 DOI: 10.1128/iai.00043-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 03/14/2023] [Indexed: 04/11/2023] Open
Abstract
Chlamydia muridarum has been used to study chlamydial pathogenesis because it induces mice to develop hydrosalpinx, a pathology observed in C. trachomatis-infected women. We identified a C. muridarum mutant that is no longer able to induce hydrosalpinx. In the current study, we evaluated the mutant as an attenuated vaccine. Following an intravaginal immunization with the mutant, mice were protected from hydrosalpinx induced by wild-type C. muridarum. However, the mutant itself productively colonized the mouse genital tract and produced infectious organisms in vaginal swabs. Nevertheless, the mutant failed to produce infectious shedding in the rectal swabs following an oral inoculation. Importantly, mice orally inoculated with the mutant mounted transmucosal immunity against challenge infection of wild-type C. muridarum in the genital tract. The protection was detected as early as day 3 following the genital challenge infection and the orally immunized mice were protected from any significant pathology in the upper genital tract. However, the same orally immunized mice failed to prevent the colonization of wild-type C. muridarum in the gastrointestinal tract. The transmucosal immunity induced by the oral mutant was further validated in the airway. The orally vaccinated mice were protected from both lung infection and systemic toxicity caused by intranasally inoculated wild-type C. muridarum although the same mice still permitted the gastrointestinal colonization by the wild-type C. muridarum. These observations suggest that the mutant C. muridarum may be developed into an intracellular oral vaccine vector (or IntrOv) for selectively inducing transmucosal immunity in extra-gut tissues.
Collapse
Affiliation(s)
- Yihui Wang
- Key Laboratory of Animal Epidemiology and Zoonosis, Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, People’s Republic of China
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, Texas, USA
| | - Rongze He
- Key Laboratory of Animal Epidemiology and Zoonosis, Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, People’s Republic of China
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, Texas, USA
| | - Halah Winner
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, Texas, USA
| | - Marie-Claire Gauduin
- Department of Virology, Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Nu Zhang
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, Texas, USA
| | - Cheng He
- Key Laboratory of Animal Epidemiology and Zoonosis, Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, People’s Republic of China
| | - Guangming Zhong
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, Texas, USA
| |
Collapse
|
6
|
Winner H, Friesenhahn A, Wang Y, Stanbury N, Wang J, He C, Zhong G. Regulation of chlamydial colonization by IFNγ delivered via distinct cells. Trends Microbiol 2023; 31:270-279. [PMID: 36175276 PMCID: PMC9974551 DOI: 10.1016/j.tim.2022.09.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 08/31/2022] [Accepted: 09/06/2022] [Indexed: 12/18/2022]
Abstract
The mouse-adapted pathogen Chlamydia muridarum (CM) induces pathology in the mouse genital tract but fails to do so in the gastrointestinal tract. CM is cleared from both the genital tract and small intestine by IFNγ delivered by antigen-specific CD4+ T cells but persists for a long period in the large intestine. The long-lasting colonization of CM in the large intestine is regulated by IFNγ delivered by group 3 innate lymphoid cells (ILC3s). Interestingly, the ILC3-delivered IFNγ can inhibit the human pathogen Chlamydia trachomatis (CT) in the mouse endometrium. Thus, IFNγ produced/delivered by different cells may selectively restrict chlamydial colonization in different tissues. Revealing the underlying mechanisms of chlamydial interactions with IFNγ produced by different cells may yield new insights into both chlamydial pathogenicity and mucosal immunity.
Collapse
Affiliation(s)
- Halah Winner
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78230, USA
| | - Ann Friesenhahn
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78230, USA
| | - Yihui Wang
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78230, USA; College of Veterinary Medicine, China Agricultural University, Two Yuanmingyuan Xi Lu, Haidian District, Beijing 100193, PR China
| | - Nicholas Stanbury
- Department of Obstetrics and Gynecology, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78230, USA
| | - Jie Wang
- Department of Immunology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, PR China
| | - Cheng He
- College of Veterinary Medicine, China Agricultural University, Two Yuanmingyuan Xi Lu, Haidian District, Beijing 100193, PR China
| | - Guangming Zhong
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78230, USA.
| |
Collapse
|
7
|
Wang Y, He R, Winner H, Gauduin MC, Zhang N, He C, Zhong G. Induction of transmucosal protection by oral vaccination with an attenuated Chlamydia. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.30.526385. [PMID: 36778293 PMCID: PMC9915634 DOI: 10.1101/2023.01.30.526385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/15/2023]
Abstract
Chlamydia muridarum has been used to study chlamydial pathogenesis since it induces mice to develop hydrosalpinx, a pathology observed in C. trachomatis -infected women. We identified a C. muridarum mutant that is no longer able to induce hydrosalpinx. In the current study, we evaluated the mutant as an attenuated vaccine. Following an intravaginal immunization with the mutant, mice were protected from hydrosalpinx induced by wild type C. muridarum . However, the mutant itself productively colonized the mouse genital tract and produced infectious organisms in vaginal swabs. Nevertheless, the mutant failed to produce infectious shedding in the rectal swabs following an oral inoculation. Importantly, mice orally inoculated with the mutant mounted transmucosal immunity against challenge infection of wild type C. muridarum in the genital tract. The protection was detected as early as day 3 following the challenge infection and the immunized mice were protected from any significant pathology in the upper genital tract. However, the same orally immunized mice failed to prevent the colonization of wild type C. muridarum in the gastrointestinal tract. The transmucosal immunity induced by the oral mutant was further validated in the airway. The orally vaccinated mice were protected from both lung infection and systemic toxicity caused by intranasally inoculated wild type C. muridarum although the same mice still permitted the gastrointestinal colonization by the wild type C. muridarum . These observations suggest that the mutant C. muridarum may be developed into an intr acellular o ral v accine vector (or IntrOv) for selectively inducing transmucosal immunity in extra-gut tissues.
Collapse
|
8
|
Tian Q, Zhang T, Wang L, Ma J, Sun X. Gut dysbiosis contributes to chlamydial induction of hydrosalpinx in the upper genital tract. Front Microbiol 2023; 14:1142283. [PMID: 37125189 PMCID: PMC10133527 DOI: 10.3389/fmicb.2023.1142283] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 03/23/2023] [Indexed: 05/02/2023] Open
Abstract
Chlamydia trachomatis is one of the most common sexually infections that cause infertility, and its genital infection induces tubal adhesion and hydrosalpinx. Intravaginal Chlamydia muridarum infection in mice can induce hydrosalpinx in the upper genital tract and it has been used for studying C. trachomatis pathogenicity. DBA2/J strain mice were known to be resistant to the chlamydial induction of hydrosalpinx. In this study, we took advantage of this feature of DBA2/J mice to evaluate the role of antibiotic induced dysbiosis in chlamydial pathogenicity. Antibiotics (vancomycin and gentamicin) were orally administrated to induce dysbiosis in the gut of DBA2/J mice. The mice with or without antibiotic treatment were evaluated for gut and genital dysbiosis and then intravaginally challenged by C. muridarum. Chlamydial burden was tested and genital pathologies were evaluated. We found that oral antibiotics significantly enhanced chlamydial induction of genital hydrosalpinx. And the antibiotic treatment induced severe dysbiosis in the GI tract, including significantly reduced fecal DNA and increased ratios of firmicutes over bacteroidetes. The oral antibiotic did not alter chlamydial infection or microbiota in the mouse genital tracts. Our study showed that the oral antibiotics-enhanced hydrosalpinx correlated with dysbiosis in gut, providing the evidence for associating gut microbiome with chlamydial genital pathogenicity.
Collapse
Affiliation(s)
- Qi Tian
- Department of Obstetrics and Gynecology, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, Hunan, China
- National Health Commission Key Laboratory for Birth Defect Research and Prevention, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, Hunan, China
- *Correspondence: Qi Tian,
| | - Tianyuan Zhang
- Key Lab of Molecular Virology and Immunology, Institute Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
- Tianyuan Zhang,
| | - Luying Wang
- Department of Obstetrics and Gynecology, Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jingyue Ma
- Department of Dermatovenereology, Tianjin Medical University General Hospital, Tianjin, China
| | - Xin Sun
- Department of Obstetrics and Gynecology, Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
9
|
Cortes E, Montero B, Yuguero O. Chlamydia trachomatis Infection Could Be a Risk Factor for Infertility in Women: A Prospective Descriptive Study. Cureus 2022; 14:e30697. [DOI: 10.7759/cureus.30697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/26/2022] [Indexed: 11/06/2022] Open
|
10
|
Xu Y, Wang J. Chlamydia
transmitting from the genital to gastrointestinal tract and inducing tubal disease: Double attack pattern. ZHONG NAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF CENTRAL SOUTH UNIVERSITY. MEDICAL SCIENCES 2022; 47:1275-1280. [PMID: 36411712 PMCID: PMC10930326 DOI: 10.11817/j.issn.1672-7347.2022.220023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Indexed: 06/16/2023]
Abstract
Chlamydia trachomatis ( CT ) genital tract infection is insidious, and patients often have no conscious symptoms.Delayed treatment after infection can lead to serious complications. Chlamydia muridarum ( CM ) genital tract infection in female mice can simulate CT genital tract infection in women, which is an ideal model to investigate the pathogenesis of CT . CM plasmid protein pGP3, chromosomal protein TC0237/TC0668, CM -specific CD8 + T cells, TNF-α, and IL-13 can induce genital tract inflammation, CD4 + T cells are responsible for CM clearance. However, tubal inflammation persists after genital tract CM is removed. Genital tract CM can spread spontaneously in vivo and colonize the gastrointestinal (GI) tract, but the GI tract CM cannot reverse spread to the genital tract. The survival time and number of CM transmitted from genital tract to GI tract are positively correlated with the long-term lesion of oviduct, while the CM inoculated directly into the GI tract has no pathogenicity in both the genital and GI tract. The double attack pattern of Chlamydia -induced genital tract inflammatory lesions is as follows: CM infection of oviduct epithelial cells initiates the process of oviduct repair as the first attack. After genital CM spreads to the GI tract, activated chlamydia-specific CD8 + T cells are recruited to the genital tract and secreted pro-fibrotic cytokines such as TNF-α and IL-13. This process is called the second attack which transform tubal repair initiated by the first attack into long-term tubal fibrosis/hydrosalpinx. Elucidating the pathogenic mechanism of Chlamydia infection can provide new ideas for the development of Chlamydia vaccine, which is expected to solve the problems of infertility caused by repeated CT infection in women.
Collapse
Affiliation(s)
- Ying Xu
- Department of Immunology, School of Basic Medical Science, Central South University, Changsha 410078, China.
| | - Jie Wang
- Department of Immunology, School of Basic Medical Science, Central South University, Changsha 410078, China.
| |
Collapse
|
11
|
Cortina ME, Bishop RC, DeVasure BA, Coppens I, Derré I. The inclusion membrane protein IncS is critical for initiation of the Chlamydia intracellular developmental cycle. PLoS Pathog 2022; 18:e1010818. [PMID: 36084160 PMCID: PMC9491573 DOI: 10.1371/journal.ppat.1010818] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Revised: 09/21/2022] [Accepted: 08/22/2022] [Indexed: 11/18/2022] Open
Abstract
All Chlamydia species are obligate intracellular bacteria that undergo a unique biphasic developmental cycle strictly in the lumen of a membrane bound compartment, the inclusion. Chlamydia specific Type III secreted effectors, known as inclusion membrane proteins (Inc), are embedded into the inclusion membrane. Progression through the developmental cycle, in particular early events of conversion from infectious (EB) to replicative (RB) bacteria, is important for intracellular replication, but poorly understood. Here, we identified the inclusion membrane protein IncS as a critical factor for Chlamydia development. We show that a C. trachomatis conditional mutant is impaired in transition from EB to RB in human cells, and C. muridarum mutant bacteria fail to develop in a mouse model of Chlamydia infection. Thus, IncS represents a promising target for therapeutic intervention of the leading cause of sexually transmitted infections of bacterial origin.
Collapse
Affiliation(s)
- María Eugenia Cortina
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of Medicine, Charlottesville, Virginia, United States of America
| | - R. Clayton Bishop
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of Medicine, Charlottesville, Virginia, United States of America
| | - Brittany A. DeVasure
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of Medicine, Charlottesville, Virginia, United States of America
| | - Isabelle Coppens
- Department of Molecular Microbiology and Immunology, Johns Hopkins School of Public Health, Baltimore, Maryland, United States of America
| | - Isabelle Derré
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of Medicine, Charlottesville, Virginia, United States of America
| |
Collapse
|
12
|
Zhou Z, Tian Q, Wang L, Zhong G. Chlamydia Deficient in Plasmid-Encoded Glycoprotein 3 (pGP3) as an Attenuated Live Oral Vaccine. Infect Immun 2022; 90:e0047221. [PMID: 35100010 PMCID: PMC8929356 DOI: 10.1128/iai.00472-21] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 01/18/2022] [Indexed: 11/20/2022] Open
Abstract
Despite the extensive efforts, there is still a lack of a licensed vaccine against Chlamydia trachomatis in humans. The mouse genital tract infection with Chlamydia muridarum has been used to both investigate chlamydial pathogenic mechanisms and evaluate vaccine candidates due to the C. muridarum's ability to induce mouse hydrosalpinx. C. muridarum mutants lacking the entire plasmid or deficient in only the plasmid-encoded pGP3 are highly attenuated in inducing hydrosalpinx. We now report that intravaginal immunization with these mutants as live attenuated vaccines protected mice from hydrosalpinx induced by wild type C. muridarum. However, these mutants still productively infected the mouse genital tract. Further, the mutant-infected mice were only partially protected against the subsequent infection with wild type C. muridarum. Thus, these mutants as vaccines are neither safe nor effective when they are delivered via the genital tract. Interestingly, these mutants were highly deficient in colonizing the gastrointestinal tract. Particularly, the pGP3-deficient mutant failed to shed live organisms from mice following an oral inoculation, suggesting that the pGP3-deficient mutant may be developed into a safe oral vaccine. Indeed, oral inoculation with the pGP3-deficient mutant induced robust transmucosal immunity against both the infection and pathogenicity of wild type C. muridarum in the genital tract. Thus, we have demonstrated that the plasmid-encoded virulence factor pGP3 may be targeted for developing an attenuated live oral vaccine.
Collapse
Affiliation(s)
- Zengzi Zhou
- The 3rd Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, Texas, USA
| | - Qi Tian
- Department of Medical Genetics, Maternal and Child Health Hospital of Hunan Province, Changsha, Hunan, China
| | - Luying Wang
- The 3rd Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Guangming Zhong
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, Texas, USA
| |
Collapse
|
13
|
Evidence for cGAS-STING signaling in the female genital tract resistance to Chlamydia trachomatis infection. Infect Immun 2022; 90:e0067021. [PMID: 34978925 DOI: 10.1128/iai.00670-21] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Sexually transmitted Chlamydia trachomatis can ascend to the upper genital tract due to its resistance to innate immunity in the lower genital tract. C. trachomatis can activate cGAS-STING signaling pathway in cultured cells via either cGAS or STING. The current study was designed to evaluate the role of the cGAS-STING pathway in innate immunity against C. trachomatis in the mouse genital tract. Following intravaginal inoculation, C. trachomatis significantly declined by day 5 following a peak infection on day 3 while the mouse-adapted C. muridarum continued to rise for >1 week, indicating that C. trachomatis is susceptible to the innate immunity in the female mouse genital tract. This conclusion was supported by the observation of a similar shedding course in mice deficient in adaptive immunity. Thus, C. trachomatis can be used to evaluate innate immunity in the female genital tract. It was found that mice deficient in either cGAS or STING significantly increased the yields of live C. trachomatis on day 5, indicating an essential role of the cGAS-STING signaling pathway in innate immunity of the mouse genital tract. Comparison of live C. trachomatis recovered from different genital tissues revealed that the cGAS-STING-dependent immunity against C. trachomatis was restricted to the mouse lower genital tract regardless of whether C. trachomatis was inoculated intravaginally or transcervically. Thus, we have demonstrated an essential role of the cGAS-STING signaling pathway in innate immunity against chlamydial infection, laying a foundation for further illuminating the mechanisms of the innate immunity in the female lower genital tract.
Collapse
|
14
|
Characterization of pathogenic CD8 + T cells in Chlamydia-infected OT1 mice. Infect Immun 2021; 90:e0045321. [PMID: 34724387 DOI: 10.1128/iai.00453-21] [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
Chlamydia trachomatis is a leading infectious cause of infertility in women due to its induction of lasting pathology such as hydrosalpinx. Chlamydia muridarum induces mouse hydrosalpinx because C. muridarum can both invade tubal epithelia directly (as a 1st hit) and induce lymphocytes to promote hydrosalpinx indirectly (as a 2nd hit). In the current study, a critical role of CD8+ T cells in chlamydial induction of hydrosalpinx was validated in both wild type C57BL/6J and OT1 transgenic mice. OT1 mice failed to develop hydrosalpinx partially due to the failure of their lymphocytes to recognize chlamydial antigens. CD8+ T cells from naïve C57BL/6J rescued the recipient OT1 mice to develop hydrosalpinx when naïve CD8+ T cells were transferred at the time of infection with Chlamydia. However, when the transfer was delayed for 2 weeks or longer after the chlamydial infection, naïve CD8+ T cells no longer promoted hydrosalpinx. Nevertheless, Chlamydia-immunized CD8+ T cells still promoted significant hydrosalpinx in the recipient OT1 mice even when the transfer was delayed for 3 weeks. Thus, CD8+ T cells must be primed within 2 weeks after chlamydial infection to be pathogenic but once primed, they can promote hydrosalpinx for >3 weeks. However, Chlamydia-primed CD4+ T cells failed to promote chlamydial induction of pathology in OT1 mice. This study has optimized an OT1 mouse-based model for revealing the pathogenic mechanisms of Chlamydia-specific CD8+ T cells.
Collapse
|
15
|
Horner PJ, Anyalechi GE, Geisler WM. What Can Serology Tell Us About the Burden of Infertility in Women Caused by Chlamydia? J Infect Dis 2021; 224:S80-S85. [PMID: 34396401 DOI: 10.1093/infdis/jiab047] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Chlamydia trachomatis (CT) causes pelvic inflammatory disease, which may result in tubal factor infertility (TFI) in women. Serologic assays may be used to determine the proportion of women with and without TFI who have had previous CT infection and to generate estimates of infertility attributable to chlamydia. Unfortunately, most existing CT serologic assays are challenged by low sensitivity and, sometimes, specificity for prior CT infection; however, they are currently the only available tests available to detect prior CT infection. Modeling methods such as finite mixture modeling may be a useful adjunct to quantitative serologic data to obtain better estimates of CT-related infertility. In this article, we review CT serological assays, including the use of antigens preferentially expressed during upper genital tract infection, and suggest future research directions. These methodologic improvements, coupled with creation of new biomarkers for previous CT infection, should improve our understanding of chlamydia's contribution to female infertility.
Collapse
Affiliation(s)
- Patrick J Horner
- Population Health Sciences, University of Bristol, Bristol, United Kingdom.,National Institute for Health Research, Health Protection Research Unit in Behavioural Science and Evaluation, University of Bristol, Bristol, United Kingdom
| | - Gloria E Anyalechi
- Division of STD Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - William M Geisler
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
16
|
Abstract
Chlamydia in the genital tract is known to spread via the blood circulation system to the large intestinal lumen to achieve long-lasting colonization. However, the precise pathways for genital Chlamydia to access to the large intestinal lumen remain unclear. The spleen was recently reported to be critical for the chlamydial spreading. In the current study, it was found that following intravaginal inoculation with Chlamydia, mice with or without splenectomy both produced infectious Chlamydia in the rectal swabs, indicating that spleen is not essential for genital Chlamydia to spread to the gastrointestinal tract. This conclusion was validated by the observation that intravenously inoculated Chlamydia was also detected in the rectal swabs of mice regardless of splenectomy. Careful comparison of the tissue distribution of live chlamydial organisms following intravenous inoculation revealed redundant pathways for Chlamydia to reach the large intestine lumen. The intravenously inoculated Chlamydia was predominantly recruited to the spleen within 12h and then detected in the stomach lumen by 24h, the intestinal lumen by 48h and rectal swabs by 72h. These observations suggest a potential spleen-to-stomach pathway for hematogenous Chlamydia to reach the large intestine lumen. This conclusion was supported by the observation made in mice under coprophagy-free condition. However, in the absence of spleen, hematogenous Chlamydia was predominantly recruited to the liver and then simultaneously detected in the intestinal tissue and lumen, suggesting a potential liver-to-intestine pathway for Chlamydia to reach the large intestine lumen. Thus, genital/hematogenous Chlamydia may reach the large intestinal lumen via multiple redundant pathways.
Collapse
|
17
|
Gastrointestinal Chlamydia-induced CD8 + T cells promote chlamydial pathogenicity in the female upper genital tract. Infect Immun 2021; 89:e0020521. [PMID: 34227838 DOI: 10.1128/iai.00205-21] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Chlamydia is known to both ascend to the upper genital tract and spread to the gastrointestinal tract following intravaginal inoculation. The gastrointestinal Chlamydia was recently reported to promote chlamydial pathogenicity in the genital tract since mice intravaginally inoculated with an attenuated Chlamydia, which alone failed to develop pathology in the genital tract, were restored to develop hydrosalpinx by intragastric co-inoculation with wild type Chlamydia. Gastrointestinal Chlamydia promoted hydrosalpinx via an indirect mechanism since Chlamydia in the gut did not directly spread to the genital tract lumen. In the current study, we further investigated the role of CD8+ T cells in the promotion of hydrosalpinx by gastrointestinal Chlamydia. First, we confirmed that intragastric co-inoculation with wild type Chlamydia promoted hydrosalpinx in mice that were inoculated with an attenuated Chlamydia in the genital tract one week earlier. Second, the promotion of hydrosalpinx by intragastrically co-inoculated Chlamydia was blocked by depleting CD8+ T cells. Third, adoptive transfer of the gastrointestinal Chlamydia-induced CD8+ T cells was sufficient for promoting hydrosalpinx in mice that were intravaginally inoculated with an attenuated Chlamydia. These observations have demonstrated that CD8+ T cells induced by gastrointestinal Chlamydia are both necessary and sufficient for promoting hydrosalpinx in the genital tract. The study has laid a foundation for further revealing the mechanisms by which Chlamydia-induced T lymphocyte responses (as a 2nd hit) promote hydrosalpinx in mice with genital Chlamydia-triggered tubal injury (as a 1st hit), a continuing effort in testing the two-hit hypothesis as a chlamydial pathogenic mechanism.
Collapse
|
18
|
Hu C, Wu H, Sun Y, Kong J, Shao L, Chen X, Liu Q, Liu Y. GlgA plays an important role in the induction of hydrosalpinx by Chlamydia muridarum. Pathog Dis 2021; 78:5857168. [PMID: 32533831 DOI: 10.1093/femspd/ftaa027] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 06/12/2020] [Indexed: 01/10/2023] Open
Abstract
While glycogen synthase A deficiency can reduce the growth and proliferation of Chlamydia muridarum, the effect of glycogen synthase A on the pathogenic process of C. muridarum remains unclear. To characterize the effect of glycogen synthase A deficiency on the pathogenicity of C. muridarum in the genital tract, BALB/c mice were intravaginally inoculated with wild-type, plasmid-free and glycogen synthase A-deficient C. muridarum, and the genital tract tissue was isolated to assess the severity of hydrosalpinx and the levels of oviduct dilatation at day 60 after infection. The glycogen storage capacity and in vitro infection ability of different C. muridarum strains were analyzed by periodic acid-Schiff staining and quantification of progeny elementary body(EB) formation. The tissue homogenate was used to determine the recovery of different C. muridarum strains. The results show that glycogen synthase A-deficient C. muridarum induced reduction of hydrosalpinx and attenuated the extent of oviduct dilatation in mice, and exhibited reduced growth and proliferation in the mouse lower genital tract. In addition, glycogen synthase A point mutations at different sites reduced the glycogen storage capacity and in vitro infectivity of C. muridarum to different degrees. Glycogen synthase A deficiency also reduced the host inflammatory reaction and ascending infection of C. muridarum.
Collapse
Affiliation(s)
- Chunmin Hu
- Department of Dermatovenereology, Tianjin Medical University General Hospital, 154 Anshan Rd., Tianjin 300052, PR China
| | - Haoqing Wu
- Department of Dermatovenereology, Tianjin Medical University General Hospital, 154 Anshan Rd., Tianjin 300052, PR China
| | - Yina Sun
- Key Laboratory of Hormones and Development (Ministry of Health), Tianjin Key Laboratory of Metabolic Diseases, Tianjin Metabolic Diseases Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, 22 Qixiangtai Rd., Tianjin 300070, China
| | - Jie Kong
- Department of Dermatovenereology, Tianjin Medical University General Hospital, 154 Anshan Rd., Tianjin 300052, PR China
| | - LiLi Shao
- Department of Dermatovenereology, Tianjin Medical University General Hospital, 154 Anshan Rd., Tianjin 300052, PR China
| | - Xiaojun Chen
- Department of Immunology, Tianjin Key Laboratory of Cellular and Molecular Immunology, Key Laboratory of Immune Microenvironment and Disease of the Educational Ministry, Tianjin Medical University, Tianjin 300070, China
| | - Quanzhong Liu
- Department of Dermatovenereology, Tianjin Medical University General Hospital, 154 Anshan Rd., Tianjin 300052, PR China
| | - Yuanjun Liu
- Department of Dermatovenereology, Tianjin Medical University General Hospital, 154 Anshan Rd., Tianjin 300052, PR China
| |
Collapse
|
19
|
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: 20] [Impact Index Per Article: 5.0] [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
|
20
|
He C, Xu Y, Huo Z, Wang J, Jia T, Li XD, Zhong G. Regulation of Chlamydia spreading from the small intestine to the large intestine via an immunological barrier. Immunol Cell Biol 2021; 99:611-621. [PMID: 33565158 DOI: 10.1111/imcb.12446] [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] [Received: 08/18/2020] [Revised: 11/29/2020] [Accepted: 02/08/2021] [Indexed: 12/14/2022]
Abstract
The obligate intracellular bacterium Chlamydia is a genital tract pathogen that can also colonize the gastrointestinal tract for long periods. The long-lasting colonization is dependent on chlamydial spreading from the small intestine to the large intestine. We previously reported that a mutant Chlamydia was able to activate an intestinal barrier for blocking its own spreading to the large intestine. In the current study, we used the mutant Chlamydia colonization model to confirm the intestinal barrier function and further to determine the immunological basis of the barrier with gene-deficient mice. Recombination activating gene 1-/- mice failed to block the mutant Chlamydia spreading, while mice deficient in toll-like receptors, myeloid differentiation primary response 88 or stimulator of interferon genes still blocked the spreading, suggesting that the intestinal barrier function is dependent on lymphocytes that express antigen receptors. Mice deficient in CD4, but not CD8 nor μ chain failed to prevent the chlamydial spreading, indicating a protective role of CD4+ cells in the intestinal barrier. Consistently, adoptive transfer of CD4+ T cells reconstituted the intestinal barrier in CD4-/- mice. More importantly, CD4+ but not CD8+ T cells nor B cells restored the intestinal barrier function in recombination activating gene 1-/- mice. Thus, CD4+ T cells are necessary and sufficient for maintaining the intestinal barrier function, indicating that the spread of an intracellular bacterium from the small intestine to the large intestine is regulated by an immunological barrier. This study has also laid a foundation for further illuminating the mechanisms by which a CD4+ T cell-dependent intestinal barrier regulates bacterial spreading in the gut.
Collapse
Affiliation(s)
- Conghui He
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.,Department of Immunology, Medical College of Hebei North University, Zhangjiakou, Hebei, China
| | - Ying Xu
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.,Department of Immunology, Xiangya School of Medicine, Central South University, Changsha City, Hunan, China
| | - Zhi Huo
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.,Department of Immunology, Xiangya School of Medicine, Central South University, Changsha City, Hunan, China
| | - Jie Wang
- Department of Immunology, Xiangya School of Medicine, Central South University, Changsha City, Hunan, China
| | - Tianjun Jia
- Department of Immunology, Medical College of Hebei North University, Zhangjiakou, Hebei, China
| | - Xiao-Dong Li
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Guangming Zhong
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| |
Collapse
|
21
|
Effects of Immunomodulatory Drug Fingolimod (FTY720) on Chlamydia Dissemination and Pathogenesis. Infect Immun 2020; 88:IAI.00281-20. [PMID: 32868341 DOI: 10.1128/iai.00281-20] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 08/24/2020] [Indexed: 12/21/2022] Open
Abstract
Fingolimod (FTY720), an FDA-approved immunomodulatory drug for treating multiple sclerosis, is an agonist of sphingosine-1-phosphate receptor (S1PR), which has been used as a research tool for inhibiting immune cell trafficking. FTY720 was recently reported to inhibit Chlamydia dissemination. Since genital Chlamydia spreading to the gastrointestinal tract correlated with its pathogenicity in the upper genital tract, we evaluated the effect of FTY720 on chlamydial pathogenicity in the current study. Following an intravaginal inoculation, live chlamydial organisms were detected in mouse rectal swabs. FTY720 treatment significantly delayed live organism shedding in the rectal swabs. However, FTY720 failed to block chlamydial spreading to the gastrointestinal tract. The live chlamydial organisms recovered from rectal swabs reached similar levels between mice with or without FTY720 treatment by day 42 in C57BL/6J and day 28 in CBA/J mice, respectively. Thus, genital Chlamydia is able to launch a 2nd wave of spreading via an FTY720-resistant pathway after the 1st wave of spreading is inhibited by FTY720. As a result, all mice developed significant hydrosalpinx. The FTY720-resistant spreading led to stable colonization of chlamydial organisms in the colon. Consistently, FTY720 did not alter the colonization of intracolonically inoculated Chlamydia Thus, we have demonstrated that, following a delay in chlamydial spreading caused by FTY720, genital Chlamydia is able to both spread to the gastrointestinal tract via an FTY720-resistant pathway and maintain its pathogenicity in the upper genital tract. Further characterization of the FTY720-resistant pathway(s) explored by Chlamydia for spreading to the gastrointestinal tract may promote our understanding of Chlamydia pathogenic mechanisms.
Collapse
|
22
|
Antibodies to Variable Domain 4 Linear Epitopes of the Chlamydia trachomatis Major Outer Membrane Protein Are Not Associated with Chlamydia Resolution or Reinfection in Women. mSphere 2020; 5:5/5/e00654-20. [PMID: 32968007 PMCID: PMC7568647 DOI: 10.1128/msphere.00654-20] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
C. trachomatis infection is the most common bacterial sexually transmitted infection, and infection in women can lead to pelvic inflammatory disease and infertility. No licensed vaccine exists to prevent C. trachomatis infection, and investigations of the natural immune response may inform the design of targeted vaccines for C. trachomatis. Our study fills a gap in knowledge regarding the epitope specificity of antibody responses that are elicited in response to C. trachomatis infection in women. We identified several new B cell epitopes for C. trachomatis antigens and confirmed B cell epitopes that have been identified by other methods. Our finding that women produce antibodies to the VD4-MOMP regardless of infection outcome provides insight into vaccine development, suggesting that vaccines targeting VD4-MOMP may need to elicit higher-titer antibody responses than natural infection imparts or that additional vaccine targets should be pursued in the future. Chlamydia trachomatis is an obligate intracellular bacterium. C. trachomatis infection is the most prevalent bacterial sexually transmitted infection and can lead to pelvic inflammatory disease and infertility in women. There is no licensed vaccine for C. trachomatis prevention, in part due to gaps in our knowledge of C. trachomatis-specific immune responses elicited during human infections. Previous investigations of the antibody response to C. trachomatis have identified immunodominant antigens and antibodies that can neutralize infection in cell culture. However, epitope-specific responses to C. trachomatis are not well characterized, and the impact of these antibodies on infection outcome is unknown. We recently developed a technology called deep sequence-coupled biopanning that uses bacteriophage virus-like particles to display peptides from antigens and affinity select against human serum IgG. Here, we used this technology to map C. trachomatis-specific antibodies in groups of women with defined outcomes following C. trachomatis infection: (i) C. trachomatis negative upon presentation for treatment (“spontaneous resolvers”), (ii) C. trachomatis negative at a 3-month follow-up visit after treatment (“nonreinfected”), and (iii) C. trachomatis positive at a 3-month follow-up after treatment (“reinfected”). This analysis yielded immunodominant epitopes that had been previously described but also identified new epitopes targeted by human antibody responses to C. trachomatis. We focused on human antibody responses to the C. trachomatis variable domain 4 serovar-conserved region of the major outer membrane protein (VD4-MOMP), a previously described immunodominant epitope. All three groups of women produced IgG to the VD4-MOMP, suggesting that detection of serum antibodies to VD4-MOMP in women with urogenital C. trachomatis infection is not associated with protection against reinfection. IMPORTANCEC. trachomatis infection is the most common bacterial sexually transmitted infection, and infection in women can lead to pelvic inflammatory disease and infertility. No licensed vaccine exists to prevent C. trachomatis infection, and investigations of the natural immune response may inform the design of targeted vaccines for C. trachomatis. Our study fills a gap in knowledge regarding the epitope specificity of antibody responses that are elicited in response to C. trachomatis infection in women. We identified several new B cell epitopes for C. trachomatis antigens and confirmed B cell epitopes that have been identified by other methods. Our finding that women produce antibodies to the VD4-MOMP regardless of infection outcome provides insight into vaccine development, suggesting that vaccines targeting VD4-MOMP may need to elicit higher-titer antibody responses than natural infection imparts or that additional vaccine targets should be pursued in the future.
Collapse
|
23
|
Innate Lymphoid Cells Are Required for Endometrial Resistance to Chlamydia trachomatis Infection. Infect Immun 2020; 88:IAI.00152-20. [PMID: 32341118 DOI: 10.1128/iai.00152-20] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 04/21/2020] [Indexed: 12/16/2022] Open
Abstract
In some women, sexually transmitted Chlamydia trachomatis may ascend to infect the endometrium, leading to pelvic inflammatory disease. To identify endometrial innate immune components that interact with Chlamydia, we introduced C. trachomatis into mouse endometrium via transcervical inoculation and compared the infectious yields in mice with and without immunodeficiency. Live C. trachomatis recovered from vaginal swabs or endometrial tissues peaked on day 3 and then declined in all mice with or without deficiency in adaptive immunity, indicating a critical role for innate immunity in endometrial control of C. trachomatis infection. Additional knockout of interleukin 2 receptor common gamma chain (IL-2Rγc) from adaptive immunity-deficient mice significantly compromised the endometrial innate immunity, demonstrating an important role for innate lymphoid cells (ILCs). Consistently, deficiency in IL-7 receptor alone, a common gamma chain-containing receptor required for ILC development, significantly reduced endometrial innate immunity. Furthermore, mice deficient in RORγt or T-bet became more susceptible to endometrial infection with C. trachomatis, suggesting a role for group 3-like ILCs in endometrial innate immunity. Furthermore, genetic deletion of gamma interferon (IFN-γ) but not IL-22 or antibody-mediated depletion of IFN-γ from adaptive immunity-deficient mice significantly compromised the endometrial innate immunity. Finally, depletion of NK1.1+ cells from adaptive immunity-deficient mice both significantly reduced IFN-γ and increased C. trachomatis burden in the endometrial tissue, confirming that mouse ILCs contribute significantly to endometrial innate immunity via an IFN-γ-dependent effector mechanism. It will be worth investigating whether IFN-γ-producing ILCs also improve endometrial resistance to sexually transmitted C. trachomatis infection in women.
Collapse
|
24
|
Gastrointestinal Coinfection Promotes Chlamydial Pathogenicity in the Genital Tract. Infect Immun 2020; 88:IAI.00905-19. [PMID: 31988173 DOI: 10.1128/iai.00905-19] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Accepted: 01/21/2020] [Indexed: 01/11/2023] Open
Abstract
Sexually transmitted Chlamydia, which can cause fibrotic pathology in women's genital tracts, is also frequently detected in the gastrointestinal tract. However, the medical significance of the gastrointestinal Chlamydia remains unclear. A murine Chlamydia readily spreads from the mouse genital tract to the gastrointestinal tract while inducing oviduct fibrotic blockage or hydrosalpinx. We previously proposed a two-hit model in which the mouse gastrointestinal Chlamydia might induce the second hit to promote genital tract pathology, and we are now providing experimental evidence for testing the hypothesis. First, chlamydial mutants that are attenuated in inducing hydrosalpinx in the genital tract also reduce their colonization in the gastrointestinal tract, leading to a better correlation of chlamydial induction of hydrosalpinx with chlamydial colonization in the gastrointestinal tract than in the genital tract. Second, intragastric coinoculation with a wild-type Chlamydia rescued an attenuated Chlamydia mutant to induce hydrosalpinx, while the chlamydial mutant infection in the genital tract alone was unable to induce any significant hydrosalpinx. Finally, the coinoculated gastrointestinal Chlamydia failed to directly spread to the genital tract lumen, suggesting that gastrointestinal Chlamydia may promote genital pathology via an indirect mechanism. Thus, we have demonstrated a significant role of gastrointestinal Chlamydia in promoting pathology in the genital tract possibly via an indirect mechanism. This study provides a novel direction/dimension for further investigating chlamydial pathogenic mechanisms.
Collapse
|
25
|
Gitsels A, Van Lent S, Sanders N, Vanrompay D. Chlamydia: what is on the outside does matter. Crit Rev Microbiol 2020; 46:100-119. [PMID: 32093536 DOI: 10.1080/1040841x.2020.1730300] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
This review summarises major highlights on the structural biology of the chlamydial envelope. Chlamydiae are obligate intracellular bacteria, characterised by a unique biphasic developmental cycle. Depending on the stage of their lifecycle, they appear in the form of elementary or reticulate bodies. Since these particles have distinctive functions, it is not surprising that their envelope differs in lipid as well as in protein content. Vice versa, by identifying surface proteins, specific characteristics of the particles such as rigidity or immunogenicity may be deduced. Detailed information on the bacterial membranes will increase our understanding on the host-pathogen interactions chlamydiae employ to survive and grow and might lead to new strategies to battle chlamydial infections.
Collapse
Affiliation(s)
- Arlieke Gitsels
- Laboratory of Immunology and Animal Biotechnology, Department of Animal Sciences and Aquatic Ecology, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| | - Sarah Van Lent
- Laboratory of Immunology and Animal Biotechnology, Department of Animal Sciences and Aquatic Ecology, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| | - Niek Sanders
- Laboratory of Gene Therapy, Department of Nutrition, Genetics and Ethology, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Daisy Vanrompay
- Laboratory of Immunology and Animal Biotechnology, Department of Animal Sciences and Aquatic Ecology, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| |
Collapse
|
26
|
The Cryptic Plasmid Improves Chlamydia Fitness in Different Regions of the Gastrointestinal Tract. Infect Immun 2020; 88:IAI.00860-19. [PMID: 31871102 DOI: 10.1128/iai.00860-19] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2019] [Accepted: 12/20/2019] [Indexed: 12/19/2022] Open
Abstract
The cryptic plasmid is important for chlamydial colonization in the gastrointestinal tract. We used a combination of intragastric, intrajejunal, and intracolon inoculations to reveal the impact of the plasmid on chlamydial colonization in distinct regions of gastrointestinal tract. Following an intragastric inoculation, the plasmid significantly improved chlamydial colonization. At the tissue level, plasmid-positive Chlamydia produced infectious progenies throughout gastrointestinal tract. However, to our surprise, plasmid-deficient Chlamydia failed to produce infectious progenies in small intestine, although infectious progenies were eventually detected in large intestine, indicating a critical role of the plasmid in chlamydial differentiation into infectious particles in small intestine. The noninfectious status may represent persistent infection, since Chlamydia genomes proliferated in the same tissues. Following an intrajejunal inoculation that bypasses the gastric barrier, plasmid-deficient Chlamydia produced infectious progenies in small intestine but was 530-fold less infectious than plasmid-positive Chlamydia, suggesting that (i) the noninfectious status developed after intragastric inoculation might be induced by a combination of gastric and intestinal effectors and (ii) chlamydial colonization in small intestine was highly dependent on plasmid. Finally, following an intracolon inoculation, the dependence of chlamydial colonization on plasmid increased over time. Thus, we have demonstrated that the plasmid may be able to improve chlamydial fitness in different gut regions via different mechanisms, which has laid a foundation to further reveal the specific mechanisms.
Collapse
|
27
|
Jansen M, van Ess E, Ouburg S, Gerds M, Morré S, Land J. C-reactive protein as a marker of persistent Chlamydia trachomatis infection is not associated with tubal factor infertility-an independent clinical validation study. Hum Reprod Open 2019; 2019:hoz029. [PMID: 31858023 PMCID: PMC6913217 DOI: 10.1093/hropen/hoz029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 05/26/2019] [Indexed: 12/05/2022] Open
Abstract
STUDY QUESTION Does C-reactive protein (CRP), as a marker of persisting low-grade inflammation, identify Chlamydia trachomatis IgG antibody test (CAT)-positive women who are at the highest risk for tubal factor infertility (TFI)? SUMMARY ANSWER No association was found between slightly elevated CRP (seCRP) levels and TFI in our CAT-positive patient population. WHAT IS KNOWN ALREADY In the fertility work-up, CAT is used to estimate the risk for TFI and to select high-risk patients for additional invasive diagnostic procedures (e.g. hysterosalpingography and laparoscopy). However, a high number of false positives exist among CAT-positive patients. In a previous study, it has been suggested that women with TFI may be identified more accurately when combining CAT with CRP, a marker for persistent low-grade inflammation. STUDY DESIGN, SIZE, DURATION Our original retrospective cohort consisted of 887 consecutive female patients who visited the fertility clinic of a tertiary care centre between 2007 and 2015. All CAT-positive women who underwent laparoscopy (as the reference test for evaluation of tubal function) and who had not undergone previous pelvic surgery were included in the study. CRP was determined in spare serum samples, and medical data was obtained by chart review. PARTICIPANTS/MATERIALS, SETTING, METHODS A total of 101 women (11.4%) were CAT-positive, and 64 of these 101 women (7.2%) met all inclusion criteria. CAT was performed with an ELISA. TFI was assessed by laparoscopy and strictly defined as extensive peri-adnexal adhesions and/or distal occlusion of at least one tube. In spare sera, CRP was performed with a high-sensitivity CRP ELISA, and CRP levels between 3 and 10 mg/L were defined as positive. Analyses were corrected for BMI, endometriosis and smoking. MAIN RESULTS AND THE ROLE OF CHANCE There was no statistically significant association between seCRP level and TFI after adjusting for BMI, endometriosis and smoking (odds ratio 1.0; 95% CI 0.3–3.3; n = 64). LIMITATIONS, REASONS FOR CAUTION Our retrospective study had a small sample size due to a low CAT-positivity rate and a conservative clinical policy with regard to invasive diagnostic testing. Additionally, CRP levels were only measured once, while they may change throughout the menstrual cycle and in time. WIDER IMPLICATIONS OF THE FINDINGS Contrary to previous findings, our results show CRP is not suitable as a marker of persistent low-grade inflammation in CAT-positive women. Other inflammatory markers and immunogenetic host factors should be studied on their clinical validity and utility to improve non-invasive risk assessment for TFI in the fertility work-up. STUDY FUNDING/COMPETING INTEREST(S) This work was partially supported by the European EuroTrans-Bio Grant [Reference number 110012 ETB] and the Eurostars grant (E!9372). S.A.M., a full-time employee of Amsterdam University Medical Centres location VUMC (0.56 fte) and the Maastricht University Medical Center (0.44 fte), is the founder (2011) and CEO of TubaScan Ltd, a spin-off company, Dept. of Medical Microbiology and Infection Prevention, Amsterdam UMC, location VUmc, Amsterdam, the Netherlands. S.O. and E.F.v.E. at the time of conducting this research had a partial appointment at TubaScan Ltd.
Collapse
Affiliation(s)
- Me Jansen
- Institute for Public Health Genomics, Department of Genetics and Cell Biology, School for Oncology and Developmental Biology (GROW), Faculty of Health, Medicine and Life Sciences, Maastricht University, 6229 ER, Maastricht, The Netherlands.,Amsterdam UMC, Location VUmc, Department of Medical Microbiology and Infection Control, Laboratory of Immunogenetics,1081 HZ, Amsterdam, The Netherlands.,Amsterdam UMC, Location VUmc, Department of Clinical Genetics, Section Community Genetics, Amsterdam Public Health Research Institute, 1081 HV, Amsterdam, The Netherlands
| | - Ef van Ess
- Amsterdam UMC, Location VUmc, Department of Medical Microbiology and Infection Control, Laboratory of Immunogenetics,1081 HZ, Amsterdam, The Netherlands
| | - S Ouburg
- Amsterdam UMC, Location VUmc, Department of Medical Microbiology and Infection Control, Laboratory of Immunogenetics,1081 HZ, Amsterdam, The Netherlands
| | - Ml Gerds
- Department of Emergency Medicine, Hospital Tjongerschans, 8441 PW Heerenveen, The Netherlands
| | - Sa Morré
- Institute for Public Health Genomics, Department of Genetics and Cell Biology, School for Oncology and Developmental Biology (GROW), Faculty of Health, Medicine and Life Sciences, Maastricht University, 6229 ER, Maastricht, The Netherlands.,Amsterdam UMC, Location VUmc, Department of Medical Microbiology and Infection Control, Laboratory of Immunogenetics,1081 HZ, Amsterdam, The Netherlands
| | - Ja Land
- Institute for Public Health Genomics, Department of Genetics and Cell Biology, School for Oncology and Developmental Biology (GROW), Faculty of Health, Medicine and Life Sciences, Maastricht University, 6229 ER, Maastricht, The Netherlands
| |
Collapse
|
28
|
Howe SE, Shillova N, Konjufca V. Dissemination of Chlamydia from the reproductive tract to the gastro-intestinal tract occurs in stages and relies on Chlamydia transport by host cells. PLoS Pathog 2019; 15:e1008207. [PMID: 31790512 PMCID: PMC6907867 DOI: 10.1371/journal.ppat.1008207] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 12/12/2019] [Accepted: 11/12/2019] [Indexed: 12/19/2022] Open
Abstract
Chlamydia trachomatis is a Gram-negative bacterial pathogen and a major cause of sexually transmitted disease and preventable blindness. In women, infections with C. trachomatis may lead to pelvic inflammatory disease (PID), ectopic pregnancy, chronic pelvic pain, and infertility. In addition to infecting the female reproductive tract (FRT), Chlamydia spp. are routinely found in the gastro-intestinal (GI) tract of animals and humans and can be a reservoir for reinfection of the FRT. Whether Chlamydia disseminates from the FRT to the GI tract via internal routes remains unknown. Using mouse-specific C. muridarum as a model pathogen we show that Chlamydia disseminates from the FRT to the GI tract in a stepwise manner, by first infecting the FRT-draining iliac lymph nodes (ILNs), then the spleen, then the GI tract. Tissue CD11c+ DCs mediate the first step: FRT to ILN Chlamydia transport, which relies on CCR7:CCL21/CCL19 signaling. The second step, Chlamydia transport from ILN to the spleen, also relies on cell transport. However, this step is dependent on cell migration mediated by sphingosine 1-phosphate (S1P) signaling. Finally, spleen to GI tract Chlamydia spread is the third critical step, and is significantly hindered in splenectomized mice. Inhibition of Chlamydia dissemination significantly reduces or precludes the induction of Chlamydia-specific serum IgG antibodies, presence of which is correlated with FRT pathology in women. This study reveals important insights in context of Chlamydia spp. pathogenesis and will inform the development of therapeutic targets and vaccines to combat this pathogen. Chlamydia trachomatis is a bacterial pathogen and a major cause of sexually transmitted disease and preventable blindness worldwide. In women, C. trachomatis may cause PID, ectopic pregnancy, chronic pelvic pain, and infertility. Chlamydia spp. are routinely found in the gastro-intestinal (GI) tract of humans and animals. However, whether and how Chlamydia spreads internally to the GI tract following the female reproductive tract (FRT) infection remains unknown. Using a mouse model of infection here we show that Chlamydia spreads to the GI tract in a stepwise manner, by first infecting the FRT-draining iliac lymph nodes (ILNs), then the spleen and the GI tract. Tissue DCs mediate the first step: FRT to ILN Chlamydia spread, which relies on CCR7:CCL21/CCL19 signaling. The second step, ILN to spleen spread, also relies on cell migration, and is dependent on sphingosine 1-phosphate (S1P) signaling. Finally, spleen to GI tract Chlamydia spread is the third critical step and is significantly hindered in splenectomized mice. Our study reveals important insight in context of Chlamydia pathogenesis. In addition, this work will inform the identification of therapeutic targets and development of vaccines against this pathogen.
Collapse
Affiliation(s)
- Savannah E. Howe
- School of Biological Sciences, Microbiology Program, Southern Illinois University, Carbondale, Illinois, United States of America
| | - Nita Shillova
- School of Biological Sciences, Microbiology Program, Southern Illinois University, Carbondale, Illinois, United States of America
| | - Vjollca Konjufca
- School of Biological Sciences, Microbiology Program, Southern Illinois University, Carbondale, Illinois, United States of America
- * E-mail:
| |
Collapse
|
29
|
Hepler RW, Nahas DD, Lucas B, Kaufhold R, Flynn JA, Galli JD, Swoyer R, Wagner JM, Espeseth AS, Joyce JG, Cook JC, Durr E. Spectroscopic analysis of chlamydial major outer membrane protein in support of structure elucidation. Protein Sci 2019; 27:1923-1941. [PMID: 30144190 PMCID: PMC6201732 DOI: 10.1002/pro.3501] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 07/17/2018] [Accepted: 08/22/2018] [Indexed: 12/26/2022]
Abstract
Chlamydial major outer membrane protein (MOMP) is the major protein constituent of the bacterial pathogen Chlamydia trachomatis. Chlamydia trachomatis Serovars D–K are the leading cause of genital tract infections which can lead to infertility or ectopic pregnancies. A vaccine against Chlamydia is highly desirable but currently not available. MOMP accounts for ~ 60% of the chlamydial protein mass and is considered to be one of the lead vaccine candidates against C. trachomatis. We report on the spectroscopic analysis of C. trachomatis native MOMP Serovars D, E, F, and J as well as C. muridarum MOMP by size exclusion chromatography multi angle light scattering (SEC MALS), circular dichroism (CD) and attenuated total reflectance Fourier transform infrared spectroscopy (ATR‐FTIR). MOMP was purified from the native bacterium grown in either adherent HeLa cells or in different suspension cell lines. Our results confirm that MOMP forms homo‐trimers in detergent micelles. The secondary structure composition of C. trachomatis MOMP was conserved across serovars, but different from composition of C. muridarum MOMP with a 13% (CD) to 18% (ATR‐FTIR) reduction in β‐sheet conformation for C. trachomatis MOMP. When Serovar E MOMP was isolated from suspension cell lines the α‐helix content increased by 7% (CD) to 13% (ATIR‐FTIR). Maintenance of a native‐like tertiary and quaternary structure in subunit vaccines is important for the generation of protective antibodies. This biophysical characterization of MOMP presented here serves, in the absence of functional assays, as a method for monitoring the structural integrity of MOMP.
Collapse
Affiliation(s)
- Robert W Hepler
- Infectious Diseases and Vaccines Discovery, MRL, Merck & Co., Inc., Kenilworth, New Jersey
| | - Debbie D Nahas
- Infectious Diseases and Vaccines Discovery, MRL, Merck & Co., Inc., Kenilworth, New Jersey
| | - Bob Lucas
- Infectious Diseases and Vaccines Discovery, MRL, Merck & Co., Inc., Kenilworth, New Jersey
| | - Robin Kaufhold
- Infectious Diseases and Vaccines Discovery, MRL, Merck & Co., Inc., Kenilworth, New Jersey
| | - Jessica A Flynn
- Infectious Diseases and Vaccines Discovery, MRL, Merck & Co., Inc., Kenilworth, New Jersey
| | - Jennifer D Galli
- Infectious Diseases and Vaccines Discovery, MRL, Merck & Co., Inc., Kenilworth, New Jersey
| | - Ryan Swoyer
- Infectious Diseases and Vaccines Discovery, MRL, Merck & Co., Inc., Kenilworth, New Jersey
| | - James M Wagner
- Vaccine Process Development, MRL, Merck & Co., Inc., Kenilworth, New Jersey
| | - Amy S Espeseth
- Infectious Diseases and Vaccines Discovery, MRL, Merck & Co., Inc., Kenilworth, New Jersey
| | - Joseph G Joyce
- Vaccine Process Development, MRL, Merck & Co., Inc., Kenilworth, New Jersey
| | - James C Cook
- Infectious Diseases and Vaccines Discovery, MRL, Merck & Co., Inc., Kenilworth, New Jersey
| | - Eberhard Durr
- Infectious Diseases and Vaccines Discovery, MRL, Merck & Co., Inc., Kenilworth, New Jersey
| |
Collapse
|
30
|
Chlamydia muridarum Induces Pathology in the Female Upper Genital Tract via Distinct Mechanisms. Infect Immun 2019; 87:IAI.00145-19. [PMID: 31085708 DOI: 10.1128/iai.00145-19] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Accepted: 05/06/2019] [Indexed: 12/23/2022] Open
Abstract
Sexually transmitted infection with Chlamydia trachomatis may lead to fibrotic blockage in women's upper genital tracts, resulting in tubal infertility. Intravaginal inoculation with C. muridarum readily induces fibrotic blockage or hydrosalpinx in mice and is used for investigating C. trachomatis pathogenicity. Using this model in combination with an antibody depletion approach, we confirmed CD4+ T cell-mediated protective immunity and a CD8+ T cell-dependent pathogenic mechanism during chlamydial infection in C57BL/6J mice. However, when mice genetically deficient in CD8+ T cells were evaluated, we found, surprisingly, that these mice were still able to develop robust hydrosalpinx following C. muridarum infection, both contradicting the observation made in C57BL/6J mice and suggesting a pathogenic mechanism that is independent of CD8+ T cells. We further found that depletion of CD4+ T cells from CD8+ T cell-deficient mice significantly reduced chlamydial induction of hydrosalpinx, indicating that CD4+ T cells became pathogenic in mice genetically deficient in CD8+ T cells. Since depletion of CD4+ T cells both promoted chlamydial infection and reduced chlamydial pathogenicity in CD8+ T cell-deficient mice, we propose that in the absence of CD8+ T cells, some CD4+ T cells may remain protective (as in C57BL/6J mice), while others may directly contribute to chlamydial pathogenicity. Thus, chlamydial pathogenicity can be mediated by distinct host mechanisms, depending upon host genetics and infection conditions. The CD8+ T cell-deficient mouse model may be useful for further investigating the mechanisms by which CD4+ T cells promote chlamydial pathogenicity.
Collapse
|
31
|
Lin H, He C, Koprivsek JJ, Chen J, Zhou Z, Arulanandam B, Xu Z, Tang L, Zhong G. Antigen-Specific CD4 + T Cell-Derived Gamma Interferon Is Both Necessary and Sufficient for Clearing Chlamydia from the Small Intestine but Not the Large Intestine. Infect Immun 2019; 87:e00055-19. [PMID: 30962403 PMCID: PMC6529659 DOI: 10.1128/iai.00055-19] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 03/27/2019] [Indexed: 02/07/2023] Open
Abstract
The genital tract pathogen Chlamydia trachomatis is frequently detected in the gastrointestinal tract, but the host immunity that regulates chlamydial colonization in the gut remains unclear. In a Chlamydia muridarum-C57 mouse model, chlamydial organisms are cleared from the genital tract in ∼4 weeks, but the genital organisms can spread to the gastrointestinal tract. We found that the gastrointestinal chlamydial organisms were cleared from the small intestine by day 28, paralleling their infection course in the genital tract, but persisted in the large intestine for long periods. Mice deficient in α/β T cells or CD4+ T cells but not CD8+ T cells showed chlamydial persistence in the small intestine, indicating a critical role for CD4+ T cells in clearing Chlamydia from the small intestine. The CD4+ T cell-dependent clearance is likely mediated by gamma interferon (IFN-γ), since mice deficient in IFN-γ but not interleukin 22 (IL-22) signaling pathways rescued chlamydial colonization in the small intestine. Furthermore, exogenous IFN-γ was sufficient for clearing Chlamydia from the small intestine but not the large intestine. Mice deficient in developing Chlamydia-specific Th1 immunity showed chlamydial persistence in the small intestine. Finally, IFN-γ-producing CD4+ but not CD8+ T cells from immunized donor mice were sufficient for eliminating Chlamydia from the small intestine but not the large intestine of recipient mice. Thus, we have demonstrated a critical role for Th1 immunity in clearing Chlamydia from the small intestine but not the large intestine, indicating that chlamydial colonization in different regions of the gastrointestinal tract is regulated by distinct immune mechanisms.
Collapse
Affiliation(s)
- Hui Lin
- Department of Laboratory Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Microbiology, Immunology, and Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Conghui He
- Department of Microbiology, Immunology, and Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - John J Koprivsek
- Department of Microbiology, Immunology, and Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Jianlin Chen
- Department of Obstetrics & Gynecology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhiguang Zhou
- Department of Metabolism & Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Bernard Arulanandam
- Department of Biology, College of Science, University of Texas at San Antonio, San Antonio, Texas, USA
| | - Zhenming Xu
- Department of Microbiology, Immunology, and Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Lingli Tang
- Department of Laboratory Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Guangming Zhong
- Department of Microbiology, Immunology, and Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| |
Collapse
|
32
|
The Plasmid-Encoded pGP3 Promotes Chlamydia Evasion of Acidic Barriers in Both Stomach and Vagina. Infect Immun 2019; 87:IAI.00844-18. [PMID: 30858342 DOI: 10.1128/iai.00844-18] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2018] [Accepted: 03/06/2019] [Indexed: 02/06/2023] Open
Abstract
Although Chlamydia trachomatis is a human genital tract pathogen, chlamydial organisms have frequently been detected in both vaginal and rectal swab samples of animals and humans. The plasmid-encoded pGP3, a genital tract virulence factor, is essential for Chlamydia muridarum to colonize the mouse gastrointestinal tract. However, intracolon inoculation to bypass the gastric barrier rescued the colonization ability of a pGP3-deficient C. muridarum mutant, suggesting that pGP3 is required for C. muridarum to reach but not to colonize the large intestine. The pGP3-deficient mutant was rapidly cleared in the stomach and was 100-fold more susceptible to gastric killing. In mice genetically deficient in gastrin, a key regulator for gastric acid production, or pharmacologically treated with a proton pump inhibitor, the ability of pGP3-deficient C. muridarum to colonize the gastrointestinal tract was rescued. The pGP3-dependent resistance was further recapitulated in vitro with treatments with HCl, pepsin, or sarkosyl. In the genital tract, deficiency in pGP3 significantly reduced C. muridarum survival in the mouse vagina and increased C. muridarum susceptibility to vaginal killing by ∼8 times. The pGP3-deficient C. muridarum was more susceptible to lactic acid killing, and the pGP3 deficiency also significantly increased C. trachomatis susceptibility to lactic acid. The above-described observations together suggest that Chlamydia may have acquired the plasmid-encoded pGP3 to overcome the gastric barrier during its adaptation to the gastrointestinal tract and the pGP3-dependent resistance may enable chlamydial evasion of the female lower genital tract barrier during sexual transmission.
Collapse
|
33
|
Darville T, Albritton HL, Zhong W, Dong L, O'Connell CM, Poston TB, Quayle AJ, Goonetilleke N, Wiesenfeld HC, Hillier SL, Zheng X. Anti-chlamydia IgG and IgA are insufficient to prevent endometrial chlamydia infection in women, and increased anti-chlamydia IgG is associated with enhanced risk for incident infection. Am J Reprod Immunol 2019; 81:e13103. [PMID: 30784128 DOI: 10.1111/aji.13103] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 02/08/2019] [Accepted: 02/11/2019] [Indexed: 12/23/2022] Open
Abstract
PROBLEM Chlamydia infections in women can ascend to the upper genital tract, and repeated infections are common, placing women at risk for sequelae. The protective role of anti-chlamydia antibodies to surface exposed antigens in ascending and incident infection is unclear. METHOD OF STUDY A whole-bacterial ELISA was used to quantify chlamydia-specific IgG and IgA in serum and cervical secretions of 151 high-risk women followed longitudinally. Correlations were determined between antibody and cervical burden, and causal mediation analysis investigated the effect of antibody on ascension. We examined the relationship of antibody to incident infection using the marginal Cox model. RESULTS Serum and cervical anti-chlamydia IgG and cervical IgA levels correlated inversely with cervical burden. While lower burden was associated with reduced ascension, causal mediation analysis revealed that the indirect effects of antibody mediated through reductions in bacterial burden were insufficient to prevent ascension. Analysis of women uninfected at enrollment revealed that serum and cervical anti-chlamydia IgG were associated with increased risk of incident infection; hazard ratio increased 3.6-fold (95% CI, 1.3-10.3), and 22.6-fold (95% CI, 3.1-165.2) with each unit of serum and cervical IgG, respectively. CONCLUSION Although anti-chlamydia IgG and IgA correlated with reduced cervical chlamydia burden, they failed to prevent ascension and increased levels of anti-chlamydia IgG were associated with increased risk for incident infection.
Collapse
Affiliation(s)
- Toni Darville
- Department of Pediatrics, University of North Carolina, Chapel Hill, North Carolina
| | - Hannah L Albritton
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center, New Orleans, Louisiana
| | - Wujuan Zhong
- Department of Biostatistics, University of North Carolina, Chapel Hill, North Carolina
| | - Li Dong
- Department of Biostatistics, University of North Carolina, Chapel Hill, North Carolina
| | | | - Taylor B Poston
- Department of Pediatrics, University of North Carolina, Chapel Hill, North Carolina
| | - Alison J Quayle
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center, New Orleans, Louisiana
| | - Nilu Goonetilleke
- Departments of Microbiology & Immunology and Medicine, University of North Carolina, Chapel Hill, North Carolina
| | - Harold C Wiesenfeld
- Department of Obstetrics, Gynecology and Reproductive Sciences, The University of Pittsburgh School of Medicine, The Magee-Womens Research Institute, Pittsburgh, Pennsylvania
| | - Sharon L Hillier
- Department of Obstetrics, Gynecology and Reproductive Sciences, The University of Pittsburgh School of Medicine, The Magee-Womens Research Institute, Pittsburgh, Pennsylvania
| | - Xiaojing Zheng
- Department of Pediatrics, University of North Carolina, Chapel Hill, North Carolina
| |
Collapse
|
34
|
van Ess EF, Eck-Hauer A, Land JA, Morré SA, Ouburg S. Combining individual Chlamydia trachomatis IgG antibodies MOMP, TARP, CPAF, OMP2, and HSP60 for tubal factor infertility prediction. Am J Reprod Immunol 2019; 81:e13091. [PMID: 30629310 PMCID: PMC6593993 DOI: 10.1111/aji.13091] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 11/30/2018] [Accepted: 01/08/2019] [Indexed: 02/04/2023] Open
Abstract
PROBLEM Tubal factor infertility (TFI) is a severe complication of genital Chlamydia trachomatis infections. In fertility workup, chlamydia antibody test (CAT) is used to predict TFI. The predictive value for TFI of most commonly used CAT is moderate. METHOD OF STUDY A total of 183 infertile Dutch Caucasian women were included in this study. All underwent tubal patency testing (hysterosalpingography [HSG] or laparoscopy). Cases had TFI, and controls had no TFI (ie normal findings during HSG or laparoscopy). TFI was categorized based on severity (TFI 1-TFI 4). This study investigated the predictive values of major outer membrane protein (MOMP), translocated actin-recruiting phosphoprotein (TARP), chlamydial protease-like activity factor (CPAF), heat shock protein-60 (HSP60) and outer membrane protein 2 (OMP2) for TFI. A predictive algorithm is developed to detect TFI with a high certainty based on combinations of antibody titres. Serum was tested with the Mikrogen recomLine immunoblot and quantified with the recomScan. A greedy algorithm that explores all possible antibody combinations was developed. RESULTS Significant differences in the distributions of antigen titres between cases and controls were observed for CPAF (P = 0.0021), HSP60 (P = 0.0061), MOMP (P = 0.0497) and OMP2 (P = 0.0016). Single antibodies could not discriminate between TFI and controls by themselves. The greedy algorithm performs better in specificity, positive predictive value (PPV), accuracy and clinical utility index than the original Mikrogen algorithm. CPAF combined with HSP60 identified 18.2% of TFI cases with 100% certainty. Most of the TFI 4 cases were identified with cut-offs of CPAF > 10.7 or OMP2 > 3.9. CONCLUSION This proof-of-principle study shows that combinations of antibodies in serum are predictive for TFI. A commercially available test can be adapted to predict TFI with a 100% specificity.
Collapse
Affiliation(s)
- Eleanne F van Ess
- Department of Medical Microbiology & Infection Control, Laboratory of Immunogenetics, Amsterdam UMC, VU University Medical Centre, Amsterdam, The Netherlands
| | - Anat Eck-Hauer
- Department of Medical Microbiology & Infection Control, Laboratory of Immunogenetics, Amsterdam UMC, VU University Medical Centre, Amsterdam, The Netherlands
| | - Jolande A Land
- Department of Genetics and Cell Biology, Faculty of Health, Medicine & Life Sciences, Institute for Public Health Genomics (IPHG), Research Institute GROW, University of Maastricht, Maastricht, The Netherlands
| | - Servaas A Morré
- Department of Medical Microbiology & Infection Control, Laboratory of Immunogenetics, Amsterdam UMC, VU University Medical Centre, Amsterdam, The Netherlands.,Department of Genetics and Cell Biology, Faculty of Health, Medicine & Life Sciences, Institute for Public Health Genomics (IPHG), Research Institute GROW, University of Maastricht, Maastricht, The Netherlands
| | - Sander Ouburg
- Department of Medical Microbiology & Infection Control, Laboratory of Immunogenetics, Amsterdam UMC, VU University Medical Centre, Amsterdam, The Netherlands
| |
Collapse
|
35
|
Huang Y, Sun Y, Qin T, Liu Y. The Structural Integrity of Plasmid-Encoded Pgp3 Is Essential for Induction of Hydrosalpinx by Chlamydia muridarum. Front Cell Infect Microbiol 2019; 9:13. [PMID: 30805313 PMCID: PMC6370636 DOI: 10.3389/fcimb.2019.00013] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 01/16/2019] [Indexed: 11/17/2022] Open
Abstract
Pgp3 consists of globular N- and C-terminal domains connected by a triple-helical coiled-coil middle domain. We demonstrated previously that Pgp3 is required for induction of hydrosalpinx by Chlamydia muridarum. We constructed C. muridarum transformants harboring deletion of the Pgp3 N-terminus (pgp3Δn), C-terminus (pgp3Δc), or middle domain (pgp3Δm). C3H/HeJ and CBA/J mice infected with pgp3Δn or pgp3Δm failed to induce hydrosalpinx in oviduct tissue. However, the pgp3Δc transformant induced mild hydrosalpinx in 20% of C3H/HeJ mice (severity score 0.2 ± 0.6) and in 40% of CBA/J mice (severity score 0.8 ± 1.3). The attenuated pathogenicity of the transformants harboring Pgp3 domain deletions was correlated with impaired in vitro growth and significantly reduced infectivity in the mouse lower genital tract. Moreover, the oviduct tissue of C3H/HeJ and CBA/J mice infected with the Pgp3-domain-deficient transformants displayed less inflammatory cell infiltration. Thus, the structural integrity of plasmid-encoded Pgp3 is essential for induction of hydrosalpinx by C. muridarum.
Collapse
Affiliation(s)
- Yumeng Huang
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin, China
| | - Yina Sun
- Key Laboratory of Hormones and Development (Ministry of Health), Tianjin Key Laboratory of Metabolic Diseases, Tianjin Metabolic Diseases Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China
| | - Tai Qin
- Key Laboratory of Cancer Prevention and Therapy, Department of Pancreatic Cancer, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Yuanjun Liu
- Department of Dermatovenereology, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
36
|
Luan X, Peng B, Li Z, Tang L, Chen C, Chen L, Wu H, Sun Z, Lu C. Vaccination with MIP or Pgp3 induces cross-serovar protection against chlamydial genital tract infection in mice. Immunobiology 2018; 224:223-230. [PMID: 30558842 DOI: 10.1016/j.imbio.2018.11.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 11/15/2018] [Accepted: 11/19/2018] [Indexed: 01/26/2023]
Abstract
Previously we reported that recombinant Chlamydia muridarum macrophage infectivity potentiator (MIP) provided partial protection against C. muridarum genital tract infection in mice. On the other hand, Chlamydia trachomatis plasmid encoded Pgp3could induce the protection against C. muridarum air way infection. This study aimed to evaluate the immunogenicity of MIP and Pgp3 from C. trachomatis serovar D and further investigate whether MIP and Pgp3 provide cross-serovar protection against C. muridarum genital tract infection in mice. Our results showed that vaccination by any regimen, including MIP alone, Pgp3 alone or MIP plus Pgp3, induced specific serum antibody production and Th1-dominant cellular responses in mice. Live chlamydial shedding from the vaginal and inflammatory pathologies in the oviduct markedly reduced. However, MIP + Pgp3 vaccination did not provide better protection than the single immunization. In conclusion, this study demonstrated that both MIP and Pgp3 can induce cross-serovar protective against chlamydial genital tract infection, and provided the guide for the development of optimal multisubunit vaccines against C. trachomatis infection.
Collapse
Affiliation(s)
- Xiuli Luan
- Pathogenic Biology Institute, Hengyang Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, 421001, China; Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang, 421001, China
| | - Bo Peng
- Pathogenic Biology Institute, Hengyang Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, 421001, China; Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang, 421001, China; Department of Pathology, Hengyang Medical College, University of South China, Hengyang, 421001, China
| | - Zhongyu Li
- Pathogenic Biology Institute, Hengyang Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, 421001, China; Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang, 421001, China
| | - Lingli Tang
- Department of Clinic Diagnosis, Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Chaoqun Chen
- Pathogenic Biology Institute, Hengyang Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, 421001, China
| | - Lili Chen
- Pathogenic Biology Institute, Hengyang Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, 421001, China
| | - Haiying Wu
- Pathogenic Biology Institute, Hengyang Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, 421001, China
| | - Zhenjie Sun
- Pathogenic Biology Institute, Hengyang Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, 421001, China
| | - Chunxue Lu
- Pathogenic Biology Institute, Hengyang Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, 421001, China; Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang, 421001, China.
| |
Collapse
|
37
|
Mixed Chlamydia trachomatis Peptide Antigens Provide a Specific and Sensitive Single-Well Colorimetric Enzyme-Linked Immunosorbent Assay for Detection of Human Anti -C. trachomatis Antibodies. mSphere 2018; 3:3/6/e00484-18. [PMID: 30404936 PMCID: PMC6222056 DOI: 10.1128/msphere.00484-18] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
For detection of anti-C. trachomatis antibodies by serological assays, use of classical chlamydial antigens results in high cross-reactivity and poor sensitivity. Previously, we discovered 48 strongly reactive peptide antigens of C. trachomatis-specific B-cell epitopes from 21 immunodominant proteins, and individual testing and combined scoring of 5 to 11 peptide antigens provided highly sensitive and specific detection of anti-C. trachomatis antibodies in chemiluminescent ELISAs. To simplify this method, this study established a single-well labor-saving colorimetric ELISA using a mixture of 12 strongly reactive C. trachomatis peptide antigens (Ctr Mix1) for detection of anti-C. trachomatis antibodies. This Ctr Mix1 ELISA (94% sensitivity and 98% specificity) outperformed 4 commercial ELISAs (49% to 79% sensitivity and 98% specificity). This ELISA can be easily implemented and commercialized, with convenient setup for use in nonspecialized laboratories. Thus, this mixed peptide assay with superior specificity and sensitivity will improve serodiagnosis of C. trachomatis infections. Sensitive and specific detection of anti-Chlamydia trachomatis antibodies in standard enzyme-linked immunosorbent assays (ELISAs) is compromised by cross-reactivity and poor sensitivity of classical C. trachomatis antigens. Previously, we discovered 48 strongly reactive peptide antigens of C. trachomatis-specific B-cell epitopes from 21 immunodominant proteins. By comprehensive individual testing of 11 top-ranked peptide antigens, we found very high sensitivity and specificity for detection of anti-C. trachomatis antibodies in chemiluminescent ELISAs. The current study established a labor-saving colorimetric ELISA by using a mixture of 12 strongly reactive C. trachomatis peptide antigens (Ctr Mix1) in a single well/serum rather than assaying reactivity to each individual peptide. For performance evaluation, we used a simulated population of 212 anti-C. trachomatis antibody-positive and -negative sera from 125 women with NAAT-confirmed active C. trachomatis infection and from 87 healthy women at low risk for C. trachomatis infection. In comparison to a composite reference standard (CRS) for anti-C. trachomatis antibody status, the Ctr Mix1 IgG ELISA achieved 93.9% sensitivity, significantly superior to the 49% to 79% sensitivities of four commercial anti-C. trachomatis IgG ELISAs, and 98% specificity of all tested assays. Compared to the labor-intensive individual peptide testing, this mixed peptide ELISA retained high specificity with only marginal, ∼5% sensitivity loss. By ROC-AUC, likelihood ratio, and predictive value analyses, the Ctr Mix1 ELISA performed satisfactorily at 10% to 75% prevalence range of anti-C. trachomatis antibodies but significantly better than commercial ELISAs. Thus, the labor-saving mixed peptide colorimetric ELISA format provides simultaneously high specificity and sensitivity for detection of anti-C. trachomatis antibodies. IMPORTANCE For detection of anti-C. trachomatis antibodies by serological assays, use of classical chlamydial antigens results in high cross-reactivity and poor sensitivity. Previously, we discovered 48 strongly reactive peptide antigens of C. trachomatis-specific B-cell epitopes from 21 immunodominant proteins, and individual testing and combined scoring of 5 to 11 peptide antigens provided highly sensitive and specific detection of anti-C. trachomatis antibodies in chemiluminescent ELISAs. To simplify this method, this study established a single-well labor-saving colorimetric ELISA using a mixture of 12 strongly reactive C. trachomatis peptide antigens (Ctr Mix1) for detection of anti-C. trachomatis antibodies. This Ctr Mix1 ELISA (94% sensitivity and 98% specificity) outperformed 4 commercial ELISAs (49% to 79% sensitivity and 98% specificity). This ELISA can be easily implemented and commercialized, with convenient setup for use in nonspecialized laboratories. Thus, this mixed peptide assay with superior specificity and sensitivity will improve serodiagnosis of C. trachomatis infections.
Collapse
|
38
|
Performance of Chlamydia trachomatis OmcB Enzyme-Linked Immunosorbent Assay in Serodiagnosis of Chlamydia trachomatis Infection in Women. J Clin Microbiol 2018; 56:JCM.00275-18. [PMID: 29899001 DOI: 10.1128/jcm.00275-18] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 06/04/2018] [Indexed: 11/20/2022] Open
Abstract
Chlamydia trachomatis serological assays with improved sensitivity over commercially available assays are needed to evaluate the burden of C. trachomatis infection and the effectiveness of prevention efforts. We evaluated the performance of a C. trachomatis outer membrane complex protein B (OmcB) enzyme-linked immunosorbent assay (ELISA) in the detection of anti-C. trachomatis antibody responses in C. trachomatis-infected women. OmcB ELISA was less sensitive than our C. trachomatis elementary body (EB) ELISA, but it was highly specific. The magnitude of the antibody response was higher in African-Americans and those with prior C. trachomatis infection. Unlike EB ELISA, the IgG1 response to C. trachomatis OmcB was short-lived and was not maintained by repeat C. trachomatis infection.
Collapse
|
39
|
Zou Y, Lei W, Su S, Bu J, Zhu S, Huang Q, Li Z. Chlamydia trachomatis plasmid-encoded protein Pgp3 inhibits apoptosis via the PI3K-AKT-mediated MDM2-p53 axis. Mol Cell Biochem 2018; 452:167-176. [PMID: 30132214 DOI: 10.1007/s11010-018-3422-9] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 07/26/2017] [Indexed: 11/24/2022]
Abstract
Chlamydia trachomatis, the most common human pathogen that causes trachoma and sexually transmitted disease, has developed various strategies for inhibiting host cell apoptosis. Activation of the PI3K (phosphoinositide 3-kinase)/AKT-mediated MDM2 (murine double minute 2)-p53 pathway plays a prominent role in the apoptosis resistance arising from C. trachomatis infection. However, the precise upstream mechanisms by which C. trachomatis activates this pathway have not been adequately investigated. Here, we reveal that the secreted C. trachomatis plasmid-encoded protein Pgp3 inhibits apoptosis in HeLa cells. This process requires the activation of the PI3K/AKT signaling pathway, thereby leading to phosphorylation and nuclear entry of MDM2, and p53 degradation. PI3 K inhibitor LY294002 and MDM2 inhibitor Nutlin-3a block Pgp3-induced inhibition of HeLa cell apoptosis, suggesting a critical role for the PI3K/AKT pathway and its effect on the MDM2-p53 axis in Pgp3 anti-apoptotic activity.
Collapse
Affiliation(s)
- Yan Zou
- Institute of Pathogenic Biology, 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, 421001, Hunan, People's Republic of China.,Clinical Laboratory, Maternity and Child Health Care Hospital in Xiangtan, Xiangtan, 411100, Hunan, People's Republic of China
| | - Wenbo Lei
- Institute of Pathogenic Biology, 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, 421001, Hunan, People's Republic of China
| | - Shengmei Su
- Institute of Pathogenic Biology, 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, 421001, Hunan, People's Republic of China
| | - Jichang Bu
- Institute of Pathogenic Biology, 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, 421001, Hunan, People's Republic of China
| | - Shunxin Zhu
- Clinical Laboratory, Maternity and Child Health Care Hospital in Xiangtan, Xiangtan, 411100, Hunan, People's Republic of China
| | - Qiulin Huang
- Department of General Surgery, The First Affiliated Hospital of University of South China, Hengyang, 421001, Hunan, People's Republic of China.
| | - Zhongyu Li
- Institute of Pathogenic Biology, 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, 421001, Hunan, People's Republic of China.
| |
Collapse
|
40
|
Comprehensive Molecular Serology of Human Chlamydia trachomatis Infections by Peptide Enzyme-Linked Immunosorbent Assays. mSphere 2018; 3:3/4/e00253-18. [PMID: 30068559 PMCID: PMC6070734 DOI: 10.1128/msphere.00253-18] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
For detection of anti-Chlamydia trachomatis antibodies by serological assays, use of classical whole-organism chlamydial antigens results in high cross-reactivity. These antigens bind mainly antibodies against the major outer membrane protein (OmpA) and bind antibodies against other immunodominant non-OmpA proteins to a lesser extent, resulting in poor assay sensitivity. The specificity of C. trachomatis serology is also compromised by the high prevalence of cross-reactive anti-C. pneumoniae antibodies in human populations. We previously identified 48 highly specific C. trachomatis B cell epitope peptide antigens of 21 immunodominant proteins. This study validated peptide antigen-based novel ELISAs that provide highly specific and sensitive detection of anti-C. trachomatis antibodies. Compared to four commercial ELISAs that achieved only poor sensitivities (51.5% to 64.8%), the combined signals of 5 to 11 peptides provided high sensitivity (86.5% to 91.8%) at the same 98% specificity. Thus, by using multiple peptide antigens of immunodominant proteins, we created simple ELISAs with specificity and sensitivity superior to standard C. trachomatis serodiagnosis. Sensitive species-specific detection of anti-Chlamydia trachomatis antibodies is compromised by cross-reactivity of the C. trachomatis antigens used in standard microimmunofluorescence (MIF) testing and enzyme-linked immunosorbent assays (ELISAs). Previously, we discovered 48 strongly reactive C. trachomatis-specific B cell epitope peptides from 21 immunodominant proteins. Here we comprehensively evaluated the 11 top-ranked C. trachomatis-specific peptide antigens from 8 proteins for use in C. trachomatis serology. Sera from 125 women with nucleic acid amplification test (NAAT)-confirmed active C. trachomatis infection and from 49 healthy women with a low risk of C. trachomatis infection were used as anti-C. trachomatis antibody-positive and -negative sera. Results obtained for detection of IgG1, IgG3, and IgA1 antibodies against the 11 C. trachomatis peptide antigens were compared to results from 4 commercial anti-C. trachomatis IgG ELISAs. Using composite reference standards (CRS) of all assays for anti-C. trachomatis antibody status, commercial ELISAs detected antibodies in antibody-positive women with sensitivities of 51.5% to 64.8%. In contrast, a combination of the results of all 11 peptides detected IgG (IgG1 and IgG3) antibodies with 91.8% sensitivity, and a labor-saving combination of the 5 optimal peptides still detected antibodies in antibody-positive women with 86.5% sensitivity (all at 98% specificity). The superior performance of the combined peptide ELISAs was confirmed by area under the receiver operating characteristic curve (ROC-AUC), likelihood ratio, and predictive value analyses. The higher sensitivity of the peptide assays results from using multiple B cell epitopes of several C. trachomatis immunodominant proteins, including OmpA, compared to exclusively using the OmpA antigens used in commercial ELISAs. Thus, ELISAs with combined use of synthetic peptide antigens for C. trachomatis antibody detection have the advantage of simultaneous high sensitivity and high specificity. IMPORTANCE For detection of anti-Chlamydia trachomatis antibodies by serological assays, use of classical whole-organism chlamydial antigens results in high cross-reactivity. These antigens bind mainly antibodies against the major outer membrane protein (OmpA) and bind antibodies against other immunodominant non-OmpA proteins to a lesser extent, resulting in poor assay sensitivity. The specificity of C. trachomatis serology is also compromised by the high prevalence of cross-reactive anti-C. pneumoniae antibodies in human populations. We previously identified 48 highly specific C. trachomatis B cell epitope peptide antigens of 21 immunodominant proteins. This study validated peptide antigen-based novel ELISAs that provide highly specific and sensitive detection of anti-C. trachomatis antibodies. Compared to four commercial ELISAs that achieved only poor sensitivities (51.5% to 64.8%), the combined signals of 5 to 11 peptides provided high sensitivity (86.5% to 91.8%) at the same 98% specificity. Thus, by using multiple peptide antigens of immunodominant proteins, we created simple ELISAs with specificity and sensitivity superior to standard C. trachomatis serodiagnosis.
Collapse
|
41
|
Woodhall SC, Gorwitz RJ, Migchelsen SJ, Gottlieb SL, Horner PJ, Geisler WM, Winstanley C, Hufnagel K, Waterboer T, Martin DL, Huston WM, Gaydos CA, Deal C, Unemo M, Dunbar JK, Bernstein K. Advancing the public health applications of Chlamydia trachomatis serology. THE LANCET. INFECTIOUS DISEASES 2018; 18:e399-e407. [PMID: 29983342 DOI: 10.1016/s1473-3099(18)30159-2] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Revised: 02/04/2018] [Accepted: 02/09/2018] [Indexed: 12/22/2022]
Abstract
Genital Chlamydia trachomatis infection is the most commonly diagnosed sexually transmitted infection. Trachoma is caused by ocular infection with C trachomatis and is the leading infectious cause of blindness worldwide. New serological assays for C trachomatis could facilitate improved understanding of C trachomatis epidemiology and prevention. C trachomatis serology offers a means of investigating the incidence of chlamydia infection and might be developed as a biomarker of scarring sequelae, such as pelvic inflammatory disease. Therefore, serological assays have potential as epidemiological tools to quantify unmet need, inform service planning, evaluate interventions including screening and treatment, and to assess new vaccine candidates. However, questions about the performance characteristics and interpretation of C trachomatis serological assays remain, which must be addressed to advance development within this field. In this Personal View, we explore the available information about C trachomatis serology and propose several priority actions. These actions involve development of target product profiles to guide assay selection and assessment across multiple applications and populations, establishment of a serum bank to facilitate assay development and evaluation, and development of technical and statistical methods for assay evaluation and analysis of serological findings. The field of C trachomatis serology will benefit from collaboration across the public health community to align technological developments with their potential applications.
Collapse
Affiliation(s)
- Sarah C Woodhall
- Blood Safety, Hepatitis, Sexually Transmitted Infections (STI) and HIV Service, Public Health England, London, UK; National Institute for Health Research Health Protection Research Unit in Evaluation of Interventions, University of Bristol, Bristol, UK; National Institute for Health Research Health Protection Research Unit in Blood Borne and Sexually Transmitted Infections, University College London, London, UK.
| | - Rachel J Gorwitz
- Division of STD Prevention, US Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Stephanie J Migchelsen
- Blood Safety, Hepatitis, Sexually Transmitted Infections (STI) and HIV Service, Public Health England, London, UK; Clinical Research Department, London School of Hygiene & Tropical Medicine, London, UK
| | - Sami L Gottlieb
- Department of Reproductive Health and Research, World Health Organization, Geneva, Switzerland
| | - Patrick J Horner
- National Institute for Health Research Health Protection Research Unit in Evaluation of Interventions, University of Bristol, Bristol, UK; Population Health Sciences, University of Bristol, Bristol, UK
| | - William M Geisler
- Division of Infectious Diseases, University of Alabama at Birmingham, Birmingham, AL, USA
| | | | - Katrin Hufnagel
- Division of Molecular Diagnostics of Oncogenic Infections, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Tim Waterboer
- Division of Molecular Diagnostics of Oncogenic Infections, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Diana L Martin
- Division of Parasitic Diseases and Malaria, US Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Wilhelmina M Huston
- School of Life Sciences, University of Technology Sydney, Sydney, NSW, Australia
| | - Charlotte A Gaydos
- Division of Infectious Diseases, Johns Hopkins University, Baltimore, MD, USA
| | - Carolyn Deal
- Division of Microbiology and Infectious Diseases, National Institute of Health, Bethesda, MD, USA
| | - Magnus Unemo
- WHO Collaborating Centre for Gonorrhoea and Other Sexually Transmitted Infections, Örebro University, Örebro, Sweden
| | - J Kevin Dunbar
- Blood Safety, Hepatitis, Sexually Transmitted Infections (STI) and HIV Service, Public Health England, London, UK; National Institute for Health Research Health Protection Research Unit in Evaluation of Interventions, University of Bristol, Bristol, UK; National Institute for Health Research Health Protection Research Unit in Blood Borne and Sexually Transmitted Infections, University College London, London, UK
| | - Kyle Bernstein
- Division of STD Prevention, US Centers for Disease Control and Prevention, Atlanta, GA, USA
| |
Collapse
|
42
|
Chlamydial plasmid-encoded virulence factor Pgp3 interacts with human cathelicidin peptide LL-37 to modulate immune response. Microbes Infect 2018; 21:50-55. [PMID: 29959096 DOI: 10.1016/j.micinf.2018.06.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Revised: 06/17/2018] [Accepted: 06/19/2018] [Indexed: 11/23/2022]
Abstract
We have previously reported that Chlamydia trachomatis plasmid-encoded Pgp3 is able to neutralize anti-chlamydial activity of human cathelicidin peptide LL-37 by binding to and forming stable complex with LL-37. Besides its microbicidal activity, LL-37 also modulates immune response, including inducing cytokine/chemokine production in fibroblast/epithelial cells and recruitment of inflammatory cells. We now report that LL-37 was significantly induced in the genital tracts of women diagnosed positive for C. trachomatis. Both the LL-37-stimulated IL-6/8 production in human endometrial epithelial cells and the LL-37-induced neutrophil chemotaxis were blocked by Pgp3. Interestingly, although Pgp3 itself alone could not induce cytokines in epithelial cell cells, it did so in neutrophils. Importantly, the Pgp3 proinflammatory activity in neutrophils was significantly enhanced by forming complex with LL-37 although LL-37 alone failed to induce cytokine production in neutrophils. Thus, we have demonstrated that Pgp3 can modulate the proinflammatory activities of LL-37 on epithelial cells by forming stable complex with LL-37 but the Pgp3's own proinflammatory activity on myeloid cells is enhanced by forming the same complex. We hypothesize that Chlamydia may use Pgp3 to both block detrimental inflammation for improving its own fitness in the genital tract epithelial tissue and activate myeloid cell-mediated inflammation for potentially promoting spreading between the hosts, the latter of which may inevitably contribute to the development of inflammatory sequelae such as tubal fibrosis.
Collapse
|
43
|
van Ess EF, Ouburg S, Spaargaren J, Land JA, Morré SA. Performance of the multitarget Mikrogen Chlamydia trachomatis IgG ELISA in the prediction of tubal factor infertility (TFI) in subfertile women: comparison with the Medac MOMP IgG ELISA plus. Pathog Dis 2018; 75:3883981. [PMID: 28854690 DOI: 10.1093/femspd/ftx067] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Accepted: 06/20/2017] [Indexed: 12/31/2022] Open
Abstract
There is a need for more accurate Chlamydia trachomatis (CT) IgG antibody tests for tubal factor infertility (TFI) diagnostics. We evaluated the predictive value for TFI of Medac ELISA plus (MOMP) and multitarget Mikrogen ELISA (MOMP-CPAF-TARP). Based on Medac ELISA plus results, 183 subfertile women underwent either hysterosalpingography or laparoscopy to diagnose TFI. TFI was defined as extensive adhesions and/or distal occlusion of at least one tube. Women not fulfilling the definition of TFI served as controls. Serum was subsequently tested with Mikrogen ELISA and results were compared. 48 patients had TFI, 135 were controls. Mikrogen ELISA tested 125 patients positive/borderline of which 32% had TFI. Medac ELISA plus tested 77 patients positive/borderline of which 29.9% had TFI. Mikrogen tested 40 out of 48 TFI patients positive/borderline, Medac 23 out of 48. Kappa value was 0.34. PPV of Mikrogen ELISA and Medac ELISA plus were respectively 32% (95% CI 26%-39%) and 30% (95% CI 24%-37%), and NPV 86% (95% CI 81%-91%) and 76% (95% CI 70%-82%). Both tests were comparable in the prediction of TFI. However, Mikrogen ELISA had a higher NPV and might be more reliable in identifying patients without TFI. Kappa-value showed limited concordance between both tests.
Collapse
Affiliation(s)
- Eleanne F van Ess
- Laboratory of Immunogenetics, Department of Medical Microbiology and Infection Control, VU University Medical Center, 1007 MB Amsterdam, The Netherlands
| | - Sander Ouburg
- Laboratory of Immunogenetics, Department of Medical Microbiology and Infection Control, VU University Medical Center, 1007 MB Amsterdam, The Netherlands
| | - Joke Spaargaren
- Laboratory of Immunogenetics, Department of Medical Microbiology and Infection Control, VU University Medical Center, 1007 MB Amsterdam, The Netherlands
| | - Jolande A Land
- Department of Obstetrics and Gynaecology, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, The Netherlands
| | - Servaas A Morré
- Laboratory of Immunogenetics, Department of Medical Microbiology and Infection Control, VU University Medical Center, 1007 MB Amsterdam, The Netherlands.,Institute for Public Health Genomics (IPHG), Department of Genetics and Cell Biology, Research Institute GROW, Faculty of Health, Medicine and Life Sciences, University of Maastricht, 6200 MB Maastricht, The Netherlands
| |
Collapse
|
44
|
Zhu C, Lin H, Tang L, Chen J, Wu Y, Zhong G. Oral Chlamydia vaccination induces transmucosal protection in the airway. Vaccine 2018; 36:2061-2068. [PMID: 29550196 DOI: 10.1016/j.vaccine.2018.03.015] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 03/06/2018] [Accepted: 03/07/2018] [Indexed: 11/19/2022]
Abstract
Although Chlamydia has been frequently detected in the gastrointestinal tracts of both humans and animals, it is not associated with any gastrointestinal pathology. We have recently shown that gastrointestinal Chlamydiamuridarum is not only non-pathogenic but also induces protective immunity in the genital tract. We now report that the transmucosal immunity induced by a single oral immunization with C.muridarum protected the mouse airway from a subsequent challenge infection. The oral immunization significantly reduced chlamydial burden in the airway as early as day 3 after intranasal challenge. As a result, the airway chlamydial spreading to extra-airway tissues was completely prevented on day 3 and significantly reduced on day 9. The immunized mice were protected from any significant systemic toxicity caused by the intranasal challenge since there was no significant bodyweight drop in the immunized mice. This robust protection correlated well with Chlamydia-specific antibodies that recognize chlamydial organism surface antigens and T cell responses that are dominated with a Th1 phenotype. The immunized mice developed high ratios of IgG2b/c over IgG1 levels and IFNγ-producing over IL-5- or IL-13-producing lymphocytes. Thus, we have demonstrated that oral vaccination with C. muridarum can induce Th1-dominant transmucosal immunity in the airway. Together with previous studies, we propose that non-pathogenic colonization of Chlamydia in the gastrointestinal tract be explored as an oral delivery system for inducing protection against infections and pathologies in extra-gastrointestinal tissues.
Collapse
Affiliation(s)
- Cuiming Zhu
- Department of Medical Microbiology, Institute of Pathogenic Biology, University of South China, Hengyang, Hunan 421001, China; Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, TX 78229, United States
| | - Hui Lin
- The 2nd Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, TX 78229, United States
| | - Lingli Tang
- The 2nd Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Jianlin Chen
- The 2nd Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Yimou Wu
- Department of Medical Microbiology, Institute of Pathogenic Biology, University of South China, Hengyang, Hunan 421001, China.
| | - Guangming Zhong
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, TX 78229, United States.
| |
Collapse
|
45
|
Sexually Transmitted Infections and Human Immunodeficiency Virus: Are We Applying the Lessons Learned? Sex Transm Dis 2018; 44:141-142. [PMID: 28178110 DOI: 10.1097/olq.0000000000000588] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
46
|
Nonpathogenic Colonization with Chlamydia in the Gastrointestinal Tract as Oral Vaccination for Inducing Transmucosal Protection. Infect Immun 2018; 86:IAI.00630-17. [PMID: 29133348 DOI: 10.1128/iai.00630-17] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 11/07/2017] [Indexed: 02/07/2023] Open
Abstract
Chlamydia has been detected in the gastrointestinal tracts of humans and animals. We now report that gastrointestinal Chlamydia muridarum is able to induce robust transmucosal protection in mice. C. muridarum colonization in the gastrointestinal tract correlated with both a shortened course of C. muridarum genital tract infection and stronger protection against subsequent genital tract challenge infection. Mice preinoculated intragastrically with C. muridarum became highly resistant to subsequent C. muridarum infection in the genital tract, resulting in prevention of pathology in the upper genital tract. The transmucosal protection in the genital tract was rapidly induced, durable, and dependent on major histocompatibility complex (MHC) class II antigen presentation but not MHC class I antigen presentation. Although a deficiency in CD4+ T cells only partially reduced the transmucosal protection, depletion of CD4+ T cells from B cell-deficient mice completely abolished the protection, suggesting a synergistic role of both CD4+ T and B cells in the gastrointestinal C. muridarum-induced transmucosal immunity. However, the same protective immunity did not significantly affect C. muridarum colonization in the gastrointestinal tract. The long-lasting colonization with C. muridarum was restricted to the gastrointestinal tract and was nonpathogenic to either gastrointestinal or extragastrointestinal tissues. Furthermore, gastrointestinal C. muridarum did not alter the gut microbiota or the development of gut mucosal resident memory T cell responses to a nonchlamydial infection. Thus, Chlamydia may be developed into a safe and orally deliverable replicating vaccine for inducing transmucosal protection.
Collapse
|
47
|
Chlamydia trachomatis and chlamydia-like bacteria: new enemies of human pregnancies. Curr Opin Infect Dis 2018; 30:289-296. [PMID: 28306562 DOI: 10.1097/qco.0000000000000369] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE OF REVIEW This review provides an update on the roles of Chlamydia trachomatis and the related Waddlia chondrophila and Parachlamydia acanthamoebae in miscarriage, stillbirths and preterm labour in humans. A broad audience, including microbiologist, infectiologists, obstetricians and gynaecologists, should be aware of the potential threat of these Chlamydiales for human reproduction. RECENT FINDINGS Despite increasing laboratory techniques and possibilities to perform diagnostic tests, the cause of miscarriage is only identified in 50% of the cases. Intracellular bacteria, such as C. trachomatis and Chlamydia-related bacteria, are difficult to detect in routine clinical samples and could represent possible agents of miscarriages. C. trachomatis is considered the world largest sexual transmitted bacterial agent and is associated with adverse pregnancy outcome in human. In the last decade Chlamydia-like organisms, such as W. chondrophila and P. acanthamoebae, have also been associated with adverse pregnancy outcomes in human and/or animals. SUMMARY We review here the current evidences for a pathogenic role in humans, the diagnostic approaches and possible treatment options of C. trachomatis, W. chondrophila and P. acanthamoebae.
Collapse
|
48
|
Zhong G. Chlamydia Spreading from the Genital Tract to the Gastrointestinal Tract - A Two-Hit Hypothesis. Trends Microbiol 2017; 26:611-623. [PMID: 29289422 DOI: 10.1016/j.tim.2017.12.002] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 11/21/2017] [Accepted: 12/06/2017] [Indexed: 12/18/2022]
Abstract
Chlamydia trachomatis, a leading bacterial cause of sexually transmitted infection-induced infertility, is frequently detected in the gastrointestinal tract. Chlamydia muridarum, a model pathogen for investigating C. trachomatis pathogenesis, readily spreads from the mouse genital tract to the gastrointestinal tract, establishing long-lasting colonization. C. muridarum mutants, despite their ability to activate acute oviduct inflammation, are attenuated in inducing tubal fibrosis and are no longer able to colonize the gastrointestinal tract, suggesting that the spread of C. muridarum to the gastrointestinal tract may contribute to its pathogenicity in the upper genital tract. However, gastrointestinal C. muridarum cannot directly autoinoculate the genital tract. Both antigen-specific CD8+ T cells and profibrotic cytokines, such as TNFα and IL-13, are essential for C. muridarum to induce tubal fibrosis; this may be induced by the gastrointestinal C. muridarum, as a second hit, to transmucosally convert tubal repairing - initiated by C. muridarum infection of tubal epithelial cells (serving as the first hit) - into pathogenic fibrosis. Testing the two-hit mouse model should both add new knowledge to the growing list of mechanisms by which gastrointestinal microbes contribute to pathologies in extragastrointestinal tissues and provide information for investigating the potential role of gastrointestinal C. trachomatis in human chlamydial pathogenesis.
Collapse
Affiliation(s)
- Guangming Zhong
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health, Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA.
| |
Collapse
|
49
|
Transcervical Inoculation with Chlamydia trachomatis Induces Infertility in HLA-DR4 Transgenic and Wild-Type Mice. Infect Immun 2017; 86:IAI.00722-17. [PMID: 29038126 DOI: 10.1128/iai.00722-17] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Accepted: 10/10/2017] [Indexed: 12/27/2022] Open
Abstract
Chlamydia trachomatis is the leading cause of infection-induced infertility in women. Attempts to control this epidemic with screening programs and antibiotic therapy have failed. Currently, a vaccine to prevent C. trachomatis infections is not available. In order to develop an animal model for evaluating vaccine antigens that can be applied to humans, we used C. trachomatis serovar D (strain UW-3/Cx) to induce infertility in mice whose major histocompatibility complex class II antigen was replaced with the human leukocyte antigen DR4 (HLA-DR4). Transcervical inoculation of medroxyprogesterone-treated HLA-DR4 transgenic mice with 5 × 105C. trachomatis D inclusion forming units (IFU) induced a significant reduction in fertility, with a mean number of embryos/mouse of 4.4 ± 1.3 compared to 7.8 ± 0.5 for the uninfected control mice (P < 0.05). A similar fertility reduction was elicited in the wild-type (WT) C57BL/6 mice (4.3 ± 1.4 embryos/mouse) compared to the levels of the WT controls (9.1 ± 0.4 embryos/mouse) (P < 0.05). Following infection, WT mice mounted more robust humoral and cellular immune responses than HLA-DR4 mice. As determined by vaginal shedding, HLA-DR4 mice were more susceptible to a transcervical C. trachomatis D infection than WT mice. To assess if HLA-DR4 transgenic and WT mice could be protected by vaccination, 104 IFU of C. trachomatis D was delivered intranasally, and mice were challenged transcervically 6 weeks later with 5 × 105 IFU of C. trachomatis D. As determined by severity and length of vaginal shedding, WT C57BL/6 and HLA-DR4 mice were significantly protected by vaccination. The advantages and limitations of the HLA-DR4 transgenic mouse model for evaluating human C. trachomatis vaccine antigens are discussed.
Collapse
|
50
|
The Genital Tract Virulence Factor pGP3 Is Essential for Chlamydia muridarum Colonization in the Gastrointestinal Tract. Infect Immun 2017; 86:IAI.00429-17. [PMID: 29038127 DOI: 10.1128/iai.00429-17] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Accepted: 10/04/2017] [Indexed: 02/06/2023] Open
Abstract
The cryptic plasmid is essential for Chlamydia muridarum dissemination from the genital tract to the gastrointestinal (GI) tract. Following intravaginal inoculation, a C. muridarum strain deficient in plasmid-encoded pGP3 or pGP4 but not pGP5, pGP7, or pGP8 failed to spread to the mouse gastrointestinal tract, although mice infected with these strains developed productive genital tract infections. pGP3- or pGP4-deficient strains also failed to colonize the gastrointestinal tract when delivered intragastrically. pGP4 regulates pGP3, while pGP3 does not affect pGP4 expression, indicating that pGP3 is critical for C. muridarum colonization of the gastrointestinal tract. Mutants deficient in GlgA, a chromosome-encoded protein regulated by pGP4, also consistently colonized the mouse gastrointestinal tract. Interestingly, C. muridarum colonization of the gastrointestinal tract positively correlated with pathogenicity in the upper genital tract. pGP3-deficient C. muridarum strains did not induce hydrosalpinx or spread to the GI tract even when delivered to the oviduct by intrabursal inoculation. Thus, the current study not only has revealed that pGP3 is a novel chlamydial colonization factor in the gastrointestinal tract but also has laid a foundation for investigating the significance of gastrointestinal Chlamydia.
Collapse
|