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
|
Liu N, Yan X, Lv B, Wu Y, Hu X, Zheng C, Tao S, Deng R, Dou J, Zeng B, Jiang G. A study on the association between gut microbiota, inflammation, and type 2 diabetes. Appl Microbiol Biotechnol 2024; 108:213. [PMID: 38358546 PMCID: PMC10869376 DOI: 10.1007/s00253-024-13041-5] [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: 12/05/2023] [Revised: 01/09/2024] [Accepted: 01/28/2024] [Indexed: 02/16/2024]
Abstract
Type 2 diabetes mellitus (T2DM) was reported to be associated with impaired immune response and alterations in microbial composition and function. However, the underlying mechanism remains elusive. To investigate the association among retinoic acid-inducible gene-I-like receptors (RLRs) signaling pathway, intestinal bacterial microbiome, microbial tryptophan metabolites, inflammation, and a longer course of T2DM, 14 patients with T2DM and 7 healthy controls were enrolled. 16S rRNA amplicon sequencing and untargeted metabolomics were utilized to analyze the stool samples. RNA sequencing (RNA-seq) was carried out on the peripheral blood samples. Additionally, C57BL/6J specific pathogen-free (SPF) mice were used. It was found that the longer course of T2DM could lead to a decrease in the abundance of probiotics in the intestinal microbiome. In addition, the production of microbial tryptophan derivative skatole declined as a consequence of the reduced abundance of related intestinal microbes. Furthermore, low abundances of probiotics, such as Bacteroides and Faecalibacterium, could trigger the inflammatory response by activating the RLRs signaling pathway. The increased level of the member of TNF receptor-associated factors (TRAF) family, nuclear factor kappa-B (NF-κB) activator (TANK), in the animal colon activated nuclear factor kappa B subunit 2 (NFκB2), resulting in inflammatory damage. In summary, it was revealed that the low abundances of probiotics could activate the RLR signaling pathway, which could in turn activate its downstream signaling pathway, NF-κB, highlighting a relationship among gut microbes, inflammation, and a longer course of T2DM. KEY POINTS: Hyperglycemia may suppress tryptophanase activity. The low abundance of Bacteroides combined with the decrease of Dopa decarboxylase (DDC) activity may lead to the decrease of the production of tryptophan microbial derivative skatole, and the low abundance of Bacteroides or reduced skatole may further lead to the increase of blood glucose by downregulating the expression of glucagon-like peptide-1 (GLP1). A low abundance of anti-inflammatory bacteria may induce an inflammatory response by triggering the RLR signaling pathway and then activating its downstream NF-κB signaling pathway in prolonged T2DM.
Collapse
Affiliation(s)
- Nannan Liu
- Laboratory of Diabetes Research Center, College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, No. 11, Bei San Huan Dong Lu, Beijing, 100029, Chaoyang District, China
| | - Xuehua Yan
- College of Traditional Chinese Medicine, Xinjiang Medical University, No.393 Xin Medical Road, Urumqi, 830011, Xinjiang, China
| | - Bohan Lv
- Laboratory of Diabetes Research Center, College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, No. 11, Bei San Huan Dong Lu, Beijing, 100029, Chaoyang District, China
| | - Yanxiang Wu
- Laboratory of Diabetes Research Center, College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, No. 11, Bei San Huan Dong Lu, Beijing, 100029, Chaoyang District, China
| | - Xuehong Hu
- Laboratory of Diabetes Research Center, College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, No. 11, Bei San Huan Dong Lu, Beijing, 100029, Chaoyang District, China
| | - Chunyan Zheng
- Laboratory of Diabetes Research Center, College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, No. 11, Bei San Huan Dong Lu, Beijing, 100029, Chaoyang District, China
| | - Siyu Tao
- Laboratory of Diabetes Research Center, College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, No. 11, Bei San Huan Dong Lu, Beijing, 100029, Chaoyang District, China
| | - Ruxue Deng
- Laboratory of Diabetes Research Center, College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, No. 11, Bei San Huan Dong Lu, Beijing, 100029, Chaoyang District, China
| | - Jinfang Dou
- Laboratory of Diabetes Research Center, College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, No. 11, Bei San Huan Dong Lu, Beijing, 100029, Chaoyang District, China
| | - Binfang Zeng
- College of Traditional Chinese Medicine, Xinjiang Medical University, No.393 Xin Medical Road, Urumqi, 830011, Xinjiang, China
| | - Guangjian Jiang
- Laboratory of Diabetes Research Center, College of Traditional Chinese Medicine, Beijing University of Chinese Medicine, No. 11, Bei San Huan Dong Lu, Beijing, 100029, Chaoyang District, China.
| |
Collapse
|
4
|
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
|
5
|
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
|
6
|
Pant S, Bhati T, Dimri A, Arora R, Siraj F, Raisuddin S, Rastogi S. Chlamydia trachomatis infection regulates the expression of tetraspanins, activin-A, and inhibin-A in tubal ectopic pregnancy. Pathog Dis 2023; 81:ftad018. [PMID: 37480234 DOI: 10.1093/femspd/ftad018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 05/17/2023] [Accepted: 07/20/2023] [Indexed: 07/23/2023] Open
Abstract
Mechanism of Chlamydia trachomatis causing tubal ectopic pregnancy (EP) is not well understood. Tetraspanins (tspans), activin-A, and inhibin-A might play a role in the development of pathological conditions leading to EP. The study aimed to elucidate the expression of tspans, activin-A, and inhibin-A with a role of associated cytokines in C. trachomatis-associated EP and analyze interacting partners of DEGs, with an expression of a few important interacting genes. Fallopian tissue and serum were collected from 100 EP (Group I) and 100 controls (Group II) from SJH, New Delhi, India. Detection of C. trachomatis was done by polymerase chain reaction (PCR) and IgG antibodies were detected by enzyme-linked immunosorbent assay. Expression of tspans, activin-A, inhibin-A, and cytokines was analyzed by real time (RT)-PCR and their interacting genes were assessed by STRING. Expression of few disease-associated interacting genes was studied by RT-PCR. A total of 29% (Group I) were C. trachomatis positive. Tspans and activin-A were significantly upregulated, while inhibin-A was significantly downregulated in Group Ia. ITGA1, TLR-2, ITGB2, and Smad-3 were a few interacting genes. Expression of ITGA1, TLR-2, and Smad-3 was significantly upregulated in C. trachomatis-positive EP. Results suggested dysregulated tspans, activin-A, and inhibin-A might play a role in C. trachomatis-infected tubal EP.
Collapse
Affiliation(s)
- Shipra Pant
- Molecular Microbiology Laboratory, ICMR-National Institute of Pathology, Sriramachari Bhawan, Safdarjung Hospital Campus, Post Box 4909, New Delhi 110029, India
| | - Tanu Bhati
- Molecular Microbiology Laboratory, ICMR-National Institute of Pathology, Sriramachari Bhawan, Safdarjung Hospital Campus, Post Box 4909, New Delhi 110029, India
| | - Astha Dimri
- Molecular Microbiology Laboratory, ICMR-National Institute of Pathology, Sriramachari Bhawan, Safdarjung Hospital Campus, Post Box 4909, New Delhi 110029, India
| | - Renu Arora
- Department of Obstetrics and Gynaecology, Vardhman Mahavir Medical College (VMMC) and Safdarjung Hospital, New Delhi 110029, India
| | - Fouzia Siraj
- Pathology Laboratory, ICMR-National Institute of Pathology, Sriramachari Bhawan, Safdarjung Hospital Campus, Post Box 4909, New Delhi 110029, India
| | - Sheikh Raisuddin
- Department of Medical Elementology and Toxicology, Jamia Hamdard, Hamdard Nagar, New Delhi 10062, India
| | - Sangita Rastogi
- Molecular Microbiology Laboratory, ICMR-National Institute of Pathology, Sriramachari Bhawan, Safdarjung Hospital Campus, Post Box 4909, New Delhi 110029, India
| |
Collapse
|
7
|
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
|
8
|
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
|
9
|
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
|
10
|
Sixt BS. Host cell death during infection with Chlamydia: a double-edged sword. FEMS Microbiol Rev 2021; 45:5902849. [PMID: 32897321 PMCID: PMC7794043 DOI: 10.1093/femsre/fuaa043] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 09/02/2020] [Indexed: 12/12/2022] Open
Abstract
The phylum Chlamydiae constitutes a group of obligate intracellular bacteria that infect a remarkably diverse range of host species. Some representatives are significant pathogens of clinical or veterinary importance. For instance, Chlamydia trachomatis is the leading infectious cause of blindness and the most common bacterial agent of sexually transmitted diseases. Chlamydiae are exceptionally dependent on their eukaryotic host cells as a consequence of their developmental biology. At the same time, host cell death is an integral part of the chlamydial infection cycle. It is therefore not surprising that the bacteria have evolved exquisite and versatile strategies to modulate host cell survival and death programs to their advantage. The recent introduction of tools for genetic modification of Chlamydia spp., in combination with our increasing awareness of the complexity of regulated cell death in eukaryotic cells, and in particular of its connections to cell-intrinsic immunity, has revived the interest in this virulence trait. However, recent advances also challenged long-standing assumptions and highlighted major knowledge gaps. This review summarizes current knowledge in the field and discusses possible directions for future research, which could lead us to a deeper understanding of Chlamydia's virulence strategies and may even inspire novel therapeutic approaches.
Collapse
Affiliation(s)
- Barbara S Sixt
- The Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå Centre for Microbial Research (UCMR), Department of Molecular Biology, Umeå University, SE-901 87 Umeå, Sweden
| |
Collapse
|
11
|
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
|
12
|
Dockterman J, Coers J. Immunopathogenesis of genital Chlamydia infection: insights from mouse models. Pathog Dis 2021; 79:ftab012. [PMID: 33538819 PMCID: PMC8189015 DOI: 10.1093/femspd/ftab012] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Accepted: 02/02/2021] [Indexed: 12/13/2022] Open
Abstract
Chlamydiae are pathogenic intracellular bacteria that cause a wide variety of diseases throughout the globe, affecting the eye, lung, coronary arteries and female genital tract. Rather than by direct cellular toxicity, Chlamydia infection generally causes pathology by inducing fibrosis and scarring that is largely mediated by host inflammation. While a robust immune response is required for clearance of the infection, certain elements of that immune response may also damage infected tissue, leading to, in the case of female genital infection, disease sequelae such as pelvic inflammatory disease, infertility and ectopic pregnancy. It has become increasingly clear that the components of the immune system that destroy bacteria and those that cause pathology only partially overlap. In the ongoing quest for a vaccine that prevents Chlamydia-induced disease, it is important to target mechanisms that can achieve protective immunity while preventing mechanisms that damage tissue. This review focuses on mouse models of genital Chlamydia infection and synthesizes recent studies to generate a comprehensive model for immunity in the murine female genital tract, clarifying the respective contributions of various branches of innate and adaptive immunity to both host protection and pathogenic genital scarring.
Collapse
Affiliation(s)
- Jacob Dockterman
- Department of Immunology, Duke University Medical Center, Durham, NC 22710, USA
| | - Jörn Coers
- Department of Immunology, Duke University Medical Center, Durham, NC 22710, USA
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 22710, USA
| |
Collapse
|
13
|
Holbrook JA, Jarosz-Griffiths HH, Caseley E, Lara-Reyna S, Poulter JA, Williams-Gray CH, Peckham D, McDermott MF. Neurodegenerative Disease and the NLRP3 Inflammasome. Front Pharmacol 2021; 12:643254. [PMID: 33776778 PMCID: PMC7987926 DOI: 10.3389/fphar.2021.643254] [Citation(s) in RCA: 131] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 01/22/2021] [Indexed: 12/13/2022] Open
Abstract
The prevalence of neurodegenerative disease has increased significantly in recent years, and with a rapidly aging global population, this trend is expected to continue. These diseases are characterised by a progressive neuronal loss in the brain or peripheral nervous system, and generally involve protein aggregation, as well as metabolic abnormalities and immune dysregulation. Although the vast majority of neurodegeneration is idiopathic, there are many known genetic and environmental triggers. In the past decade, research exploring low-grade systemic inflammation and its impact on the development and progression of neurodegenerative disease has increased. A particular research focus has been whether systemic inflammation arises only as a secondary effect of disease or is also a cause of pathology. The inflammasomes, and more specifically the NLRP3 inflammasome, a crucial component of the innate immune system, is usually activated in response to infection or tissue damage. Dysregulation of the NLRP3 inflammasome has been implicated in the progression of several neurodegenerative disorders, such as Alzheimer's disease, Parkinson's disease, Huntington's disease, amyotrophic lateral sclerosis, and prion diseases. This review aims to summarise current literature on the role of the NLRP3 inflammasome in the pathogenesis of neurodegenerative diseases, and recent work investigating NLRP3 inflammasome inhibition as a potential future therapy.
Collapse
Affiliation(s)
- Jonathan A. Holbrook
- Department of Clinical Neurosciences, John Van Geest Centre for Brain Repair, University of Cambridge, Cambridge, United Kingdom
| | - Heledd H. Jarosz-Griffiths
- Leeds Institute of Rheumatic and Musculoskeletal Medicine (LIRMM), University of Leeds, Leeds, United Kingdom
- Leeds Institute of Medical Research at St. James’s University Hospital, Leeds, United Kingdom
- Leeds Cystic Fibrosis Trust Strategic Research Centre, University of Leeds, Leeds, United Kingdom
| | - Emily Caseley
- Leeds Institute of Rheumatic and Musculoskeletal Medicine (LIRMM), University of Leeds, Leeds, United Kingdom
- Leeds Institute of Medical Research at St. James’s University Hospital, Leeds, United Kingdom
| | - Samuel Lara-Reyna
- Institute of Microbiology and Infection, University of Birmingham, Birmingham, United Kingdom
| | - James A. Poulter
- Leeds Institute of Rheumatic and Musculoskeletal Medicine (LIRMM), University of Leeds, Leeds, United Kingdom
- Leeds Institute of Medical Research at St. James’s University Hospital, Leeds, United Kingdom
| | - Caroline H. Williams-Gray
- Department of Clinical Neurosciences, John Van Geest Centre for Brain Repair, University of Cambridge, Cambridge, United Kingdom
| | - Daniel Peckham
- Leeds Institute of Medical Research at St. James’s University Hospital, Leeds, United Kingdom
- Leeds Cystic Fibrosis Trust Strategic Research Centre, University of Leeds, Leeds, United Kingdom
- Leeds Centre for Cystic Fibrosis, St James’s University Hospital, Leeds, United Kingdom
| | - Michael F. McDermott
- Leeds Institute of Rheumatic and Musculoskeletal Medicine (LIRMM), University of Leeds, Leeds, United Kingdom
- Leeds Cystic Fibrosis Trust Strategic Research Centre, University of Leeds, Leeds, United Kingdom
| |
Collapse
|
14
|
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
|
15
|
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
|
16
|
Effectiveness of Platelet-Rich Plasma in the Prevention of Chlamydia-Induced Hydrosalpinx in a Murine Model. Reprod Sci 2020; 28:1031-1040. [PMID: 32989630 DOI: 10.1007/s43032-020-00329-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 09/20/2020] [Indexed: 10/23/2022]
Abstract
Chlamydia trachomatis (C. trachomatis) is a major pathogen implicated in the formation of hydrosalpinx in the female reproductive tract. In mice, a related strain of Chlamydia, Chlamydia trachomatis (C. trachomatis) can induce almost 100% bilateral hydrosalpinx. This model was used as a hydrosalpinx induction model to test whether oviduct delivery of platelet-rich plasma (PRP) can attenuate chlamydia induction of hydrosalpinx in a mouse model. Mice were infected intravaginally with Chlamydia muridarum organisms, and 21 days after the infection, PRP was instilled into the lumen of one oviduct, and a sham instillation with phosphate buffer solution was performed on the contralateral oviduct. Mice were then sacrificed at designated time points after infection for oviduct pathologic evaluation including incidence, severity, and histopathologic grade of chronic inflammation. Oviduct instillation of PRP was associated with a 36% reduction in the incidence of hydrosalpinx and a 33% reduction in severity compared with sham. The median grade of chronic inflammation on histopathology was significantly lower with PRP instillation compared with sham and control. No differences were observed in vaginal or rectal shedding of C. muridarum between the test group and the control group. In short, the results suggest that oviduct instillation of PRP can significantly reduce the incidence and severity of C. muridarum-induced hydrosalpinx without affecting chlamydial infection courses in CBA/J mice.
Collapse
|
17
|
Xie L, He C, Chen J, Tang L, Zhou Z, Zhong G. Suppression of Chlamydial Pathogenicity by Nonspecific CD8 + T Lymphocytes. Infect Immun 2020; 88:e00315-20. [PMID: 32747602 PMCID: PMC7504968 DOI: 10.1128/iai.00315-20] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 07/27/2020] [Indexed: 12/13/2022] Open
Abstract
Chlamydia trachomatis, a leading infectious cause of tubal infertility, induces upper genital tract pathology, such as hydrosalpinx, which can be modeled with Chlamydia muridarum infection in mice. Following C. muridarum inoculation, wild-type mice develop robust hydrosalpinx, but OT1 mice fail to do so because their T cell receptors are engineered to recognize a single ovalbumin epitope (OVA457-462). These observations have demonstrated a critical role of Chlamydia-specific T cells in chlamydial pathogenicity. In the current study, we have also found that OT1 mice can actively inhibit chlamydial pathogenicity. First, depletion of CD8+ T cells from OT1 mice led to the induction of significant hydrosalpinx by Chlamydia, indicating that CD8+ T cells are necessary to inhibit chlamydial pathogenicity. Second, adoptive transfer of CD8+ T cells from OT1 mice to CD8 knockout mice significantly reduced chlamydial induction of hydrosalpinx, demonstrating that OT1 CD8+ T cells are sufficient for attenuating chlamydial pathogenicity in CD8 knockout mice. Finally, CD8+ T cells from OT1 mice also significantly inhibited hydrosalpinx development in wild-type mice following an intravaginal inoculation with Chlamydia Since T cells in OT1 mice are engineered to recognize only the OVA457-462 epitope, the above observations have demonstrated a chlamydial antigen-independent immune mechanism for regulating chlamydial pathogenicity. Further characterization of this mechanism may provide information for developing strategies to reduce infertility-causing pathology induced by infections.
Collapse
Affiliation(s)
- Lingxiang Xie
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
- The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Conghui He
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Jianlin Chen
- The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Lingli Tang
- The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Zhiguang Zhou
- The Second Xiangya Hospital of 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
|
18
|
NOD1/NOD2 and RIP2 Regulate Endoplasmic Reticulum Stress-Induced Inflammation during Chlamydia Infection. mBio 2020; 11:mBio.00979-20. [PMID: 32487756 PMCID: PMC7267884 DOI: 10.1128/mbio.00979-20] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Understanding the initiation of the inflammatory response during Chlamydia infection is of public health importance given the impact of this disease on young women in the United States. Many young women are chronically infected with Chlamydia but are asymptomatic and therefore do not seek treatment, leaving them at risk of long-term reproductive harm due to inflammation in response to infection. Our manuscript explores the role of the endoplasmic reticulum stress response pathway initiated by an innate receptor in the development of this inflammation. The inflammatory response to Chlamydia infection is likely to be multifactorial and involve a variety of ligand-dependent and -independent recognition pathways. We previously reported the presence of NOD1/NOD2-dependent endoplasmic reticulum (ER) stress-induced inflammation during Chlamydia muridarum infection in vitro, but the relevance of this finding to an in vivo context is unclear. Here, we examined the ER stress response to in vivo Chlamydia infection. The induction of interleukin 6 (IL-6) production after systemic Chlamydia infection correlated with expression of ER stress response genes. Furthermore, when tauroursodeoxycholate (TUDCA) was used to inhibit the ER stress response, an increased bacterial burden was detected, suggesting that ER stress-driven inflammation can contribute to systemic bacterial clearance. Mice lacking both NOD1 and NOD2 or RIP2 exhibited slightly higher systemic bacterial burdens after infection with Chlamydia. Overall, these data suggest a model where RIP2 and NOD1/NOD2 proteins link ER stress responses with the induction of Chlamydia-specific inflammatory responses.
Collapse
|
19
|
Chlamydia Deficient in Plasmid-Encoded pGP3 Is Prevented from Spreading to Large Intestine. Infect Immun 2020; 88:IAI.00120-20. [PMID: 32205401 DOI: 10.1128/iai.00120-20] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 03/16/2020] [Indexed: 12/11/2022] Open
Abstract
The cryptic plasmid pCM is critical for chlamydial colonization in the gastrointestinal tract. Nevertheless, orally inoculated plasmid-free Chlamydia sp. was still able to colonize the gut. Surprisingly, orally inoculated Chlamydia sp. deficient in only plasmid-encoded pGP3 was no longer able to colonize the gut. A comparison of live organism recoveries from individual gastrointestinal tissues revealed that pGP3-deficient Chlamydia sp. survived significantly better than plasmid-free Chlamydia sp. in small intestinal tissues. However, the small intestinal pGP3-deficient Chlamydia sp. failed to reach the large intestine, explaining the lack of live pGP3-deficient Chlamydia sp. in rectal swabs following an oral inoculation. Interestingly, pGP3-deficient Chlamydia sp. was able to colonize the colon following an intracolon inoculation, suggesting that pGP3-deficient Chlamydia sp. might be prevented from spreading from the small intestine to the large intestine. This hypothesis is supported by the finding that following an intrajejunal inoculation that bypasses the gastric barrier, pGP3-deficient Chlamydia sp. still failed to reach the large intestine, although similarly inoculated plasmid-free Chlamydia sp. was able to do so. Interestingly, when both types of organisms were intrajejunally coinoculated into the same mouse small intestine, plasmid-free Chlamydia sp. was no longer able to spread to the large intestine, suggesting that pGP3-deficient Chlamydia sp. might be able to activate an intestinal resistance for regulating Chlamydia sp. spreading. Thus, the current study has not only provided evidence for reconciling a previously identified conflicting phenotype but also revealed a potential intestinal resistance to chlamydial spreading. Efforts are under way to further define the mechanism of the putative intestinal resistance.
Collapse
|
20
|
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
|
21
|
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
|
22
|
Wang Y, Arthur EW, Liu N, Li X, Xiang W, Maxwell A, Li Z, Zhou Z. iTRAQ-Based Quantitative Proteomics Analysis of HeLa Cells Infected With Chlamydia muridarum TC0668 Mutant and Wild-Type Strains. Front Microbiol 2019; 10:2553. [PMID: 31787950 PMCID: PMC6854023 DOI: 10.3389/fmicb.2019.02553] [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: 08/20/2019] [Accepted: 10/22/2019] [Indexed: 12/15/2022] Open
Abstract
Chlamydia muridarum, an obligate intracellular pathogen, was used to establish a murine model of female upper genital tract infection by Chlamydia trachomatis. TC0668 in C. muridarum is a hypothetical chromosomal virulence protein that is involved in upper genital tract pathogenesis. The infection of mice with the C. muridarum TC0668-mutant (G216*) strain results in less pathological damage in the upper genital tract. In this study, an isobaric tags for relative and absolute quantitation (iTRAQ)-based quantitative proteomics analysis was performed to identify differentially expressed proteins between TC0668 wild-type (TC0668wt) and TC0668 mutant (TC0668mut) strains at 6, 12, 18, and 24 h post-infection (p.i.). Of the 550 proteins differentially expressed at 18 h p.i., 222 and 328 were up-regulated and down-regulated, respectively, inTC0668mut-infected cells. The expression of seven up-regulated proteins (encoded by SRPRB, JAK1, PMM1, HLA-DQB1, THBS1, ITPR1, and BCAP31) and three down-regulated proteins (encoded by MAPKAPK2, TRAFD1, and IFI16) from the iTRAQ analysis were validated using quantitative real-time (qRT)-PCR. The qRT-PCR results were consistent with those of iTRAQ. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses revealed that the differentially expressed proteins primarily participated in inflammatory responses, fibrosis, metabolic processes, and complement coagulation cascades, and were mainly enriched in the phosphatidylinositol 3′-kinase (PI3K)/Akt, nuclear factor kappa-B (NF-κB), and other signaling pathways. Using western-blotting and immunofluorescence detection, significant differences in activation of the PI3K/Akt and NF-κB signaling pathways were observed between the TC0668wt- and TC0668mut-infected cells. Differentially expressed proteins linked with inflammation and fibrosis were used in a protein-protein interaction network analysis. The results suggest that TC0668 may play a pivotal role in C. muridarum-induced genital pathology by inducing inflammatory responses and fibrosis, which may involve the activation of the PI3K/Akt and NF-κB signaling pathways.
Collapse
Affiliation(s)
- Yingzi Wang
- Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hengyang, China.,Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Pathogenic Biology Institute, University of South China, Hengyang, China
| | - Emmanuel Wirekoh Arthur
- Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hengyang, China.,Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Pathogenic Biology Institute, University of South China, Hengyang, China
| | - Na Liu
- Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hengyang, China.,Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Pathogenic Biology Institute, University of South China, Hengyang, China
| | - Xiaofang Li
- Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hengyang, China.,Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Pathogenic Biology Institute, University of South China, Hengyang, China
| | - Wenjing Xiang
- Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hengyang, China.,Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Pathogenic Biology Institute, University of South China, Hengyang, China
| | - Asamoah Maxwell
- Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hengyang, China.,Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Pathogenic Biology Institute, University of South China, Hengyang, China
| | - Zhongyu Li
- Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hengyang, China.,Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Pathogenic Biology Institute, University of South China, Hengyang, China
| | - Zhou Zhou
- Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hengyang, China.,Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Pathogenic Biology Institute, University of South China, Hengyang, China
| |
Collapse
|
23
|
Xu JZ, Kumar R, Gong H, Liu L, Ramos-Solis N, Li Y, Derbigny WA. Toll-Like Receptor 3 Deficiency Leads to Altered Immune Responses to Chlamydia trachomatis Infection in Human Oviduct Epithelial Cells. Infect Immun 2019; 87:e00483-19. [PMID: 31383744 PMCID: PMC6759307 DOI: 10.1128/iai.00483-19] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2019] [Accepted: 07/26/2019] [Indexed: 12/16/2022] Open
Abstract
Reproductive tract pathology caused by Chlamydia trachomatis infection is an important global cause of human infertility. To better understand the mechanisms associated with Chlamydia-induced genital tract pathogenesis in humans, we used CRISPR genome editing to disrupt Toll-like receptor 3 (TLR3) function in the human oviduct epithelial (hOE) cell line OE-E6/E7 in order to investigate the possible role(s) of TLR3 signaling in the immune response to Chlamydia Disruption of TLR3 function in these cells significantly diminished the Chlamydia-induced synthesis of several inflammation biomarkers, including interferon beta (IFN-β), interleukin-6 (IL-6), interleukin-6 receptor alpha (IL-6Rα), soluble interleukin-6 receptor beta (sIL-6Rβ, or gp130), IL-8, IL-20, IL-26, IL-34, soluble tumor necrosis factor receptor 1 (sTNF-R1), tumor necrosis factor ligand superfamily member 13B (TNFSF13B), matrix metalloproteinase 1 (MMP-1), MMP-2, and MMP-3. In contrast, the Chlamydia-induced synthesis of CCL5, IL-29 (IFN-λ1), and IL-28A (IFN-λ2) was significantly increased in TLR3-deficient hOE cells compared to their wild-type counterparts. Our results indicate a role for TLR3 signaling in limiting the genital tract fibrosis, scarring, and chronic inflammation often associated with human chlamydial disease. Interestingly, we saw that Chlamydia infection induced the production of biomarkers associated with persistence, tumor metastasis, and autoimmunity, such as soluble CD163 (sCD163), chitinase-3-like protein 1, osteopontin, and pentraxin-3, in hOE cells; however, their expression levels were significantly dysregulated in TLR3-deficient hOE cells. Finally, we demonstrate using hOE cells that TLR3 deficiency resulted in an increased amount of chlamydial lipopolysaccharide (LPS) within Chlamydia inclusions, which is suggestive that TLR3 deficiency leads to enhanced chlamydial replication and possibly increased genital tract pathogenesis during human infection.
Collapse
Affiliation(s)
- Jerry Z Xu
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Ramesh Kumar
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Haoli Gong
- Xiangya Second Hospital, Central South University, Changsha, Hunan Province, People's Republic of China
| | - Luyao Liu
- Xiangya Second Hospital, Central South University, Changsha, Hunan Province, People's Republic of China
| | - Nicole Ramos-Solis
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Yujing Li
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Wilbert A Derbigny
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| |
Collapse
|
24
|
Caspase-11 Contributes to Oviduct Pathology during Genital Chlamydia Infection in Mice. Infect Immun 2019; 87:IAI.00262-19. [PMID: 31160363 DOI: 10.1128/iai.00262-19] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 05/24/2019] [Indexed: 11/20/2022] Open
Abstract
It has been shown that caspase-1, but not its upstream activator, ASC, contributes to oviduct pathology during mouse genital Chlamydia muridarum infection. We hypothesized that this dichotomy is due to the inadvertent absence of caspase-11 in previously used caspase-1-deficient mice. To address this, we studied the independent contributions of caspase-1 and -11 during genital Chlamydia infection. Our results show that caspase-11 deficiency was sufficient to recapitulate the effect of the combined absence of both caspase-1 and caspase-11 on oviduct pathology. Further, mice that were deficient for both caspase-1 and -11 but that expressed caspase-11 as a transgene (essentially, caspase-1-deficient mice) had no significant difference in oviduct pathology from control mice. Caspase-11-deficient mice showed reduced dilation in both the oviducts and uterus. To determine the mechanism by which caspase-11-deficient mice developed reduced pathology, the chlamydial burden and immune cell infiltration were determined in the oviducts. In the caspase-11-deficient mice, we observed increased chlamydial burdens in the upper genital tract, which correlated with increased CD4 T cell recruitment, suggesting a contribution of caspase-11 in infection control. Additionally, there were significantly fewer neutrophils in the oviducts of caspase-11-deficient mice, supporting the observed decrease in the incidence of oviduct pathology. Therefore, caspase-11 activation contributes to pathogen control and oviduct disease independently of caspase-1 activation.
Collapse
|
25
|
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
|
26
|
Chlamydia trachomatis ct143 stimulates secretion of proinflammatory cytokines via activating the p38/MAPK signal pathway in THP-1 cells. Mol Immunol 2018; 105:233-239. [PMID: 30554084 DOI: 10.1016/j.molimm.2018.12.007] [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: 08/03/2018] [Revised: 11/03/2018] [Accepted: 12/09/2018] [Indexed: 01/22/2023]
Abstract
Chlamydia trachomatis (Ct) infections can cause bacterial sexually-transmitted and preventable blindness. The Ct infections induced excessive cytokines generation which attributed to pathologic changes in host cells. However, the precise mechanisms of Ct-induced cytokines production are still unclear.CT143 protein was identified as a novel Ct specific protein with high immunogenicity. In the present study. The CT143 fusion protein was recombined and purified. The mice immune serum was prepared by immunizing BALB/c mice with the purified fusion protein. The specificity of the antibody was confirmed using Immunoblotting. Indirect immunoflurescence assay (IFA) and Immunoblotting assays were performed to detect the temporal and spatial characteristics of CT143 in Ct infected cells. ELISA was performed to analyze the secretion of proinflammatory cytokines IL-1β, IL-8 and TNF-α by human macrophages under the stimulation of CT143 protein. Finally, the involvement of p38 signaling in CT143-induced cytokine secretion was validated. CT143 protein was located in the inclusion body and represented an Elementary body (EB)-related protein, which may be encoded by the mid- and late-stage expressing genes. CT143 protein could stimulate the secretion of inflammatory cytokines in macrophages which differentiated from THP-1 This induction may be mediated by the activation of p38 signaling. In summary, CT143 protein is involved in inflammatory processes during Ct infection.
Collapse
|
27
|
Zhang Y, Li W, Zou L, Gong Y, Zhang P, Xing S, Yang H. Metabonomic study of the protective effect of Fukeqianjin formula on multi-pathogen induced pelvic inflammatory disease in rats. Chin Med 2018; 13:61. [PMID: 30555525 PMCID: PMC6288860 DOI: 10.1186/s13020-018-0217-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 11/29/2018] [Indexed: 01/02/2023] Open
Abstract
Background Fukeqianjin formula has been effectively used in the treatment of pelvic inflammatory disease (PID) and the related complications in clinic. Although there have been some studies about the underlying mechanism that focus on its anti-inflammatory and immunoregulatory activities. But the mechanism is still not fully understood. The aim of this study was to investigate the alteration of plasma metabolic profiles in PID rats and the regulatory effect of Fukeqianjin formula on potential biomarkers. Methods Pelvic inflammatory model was established by intrauterine inoculation of multiple pathogens combined with mechanical injury of endometrium. Rats were randomly divided into normal group, model group, azithromycin group, high-and low-dose of Fukeqianjin formula treatment group (FF-H, and FF-L, respectively). After 14 days of intragastric administration, the plasm levels of interleukin-1β (IL-1β) and nitric oxide (NO) were measured. To further recognize and identify potential biomarkers and metabolic pathways, an ultra-performance liquid chromatography-quadrupole-Exactive Orbitrap-mass spectrometry (UPLC-Q-Exactive Orbitrap-MS) metabonomic method combined with multivariate analyses including principal component analysis (PCA), partial least squares discriminant analysis (PLS-DA) and orthogonal partial least squares discriminant analysis (OPLS-DA), was employed to analyze the metabolic profiling. Results Compared with normal group, the plasma levels of IL-1β and NO were significantly increased in the PID model group (P < 0. 05), and obviously decreased after high-dose intervention of Fukeqianjin formula (P < 0. 01). The PCA, PLS-DA and OPLS-DA analysis showed that PID rats were clearly separated from normal rats. Compared with the PID model group, the metabolite profiles of Fukeqianjin formula treatment group was gradually restored to normal. Meanwhile, 14 potential metabolite biomarkers, which were mainly related to the metabolic pathways of intervening glycerophospholipid metabolism, linoleic acid metabolism/alpha-linolenic acid metabolism, amino acid metabolism, arachidonic acid metabolism, and unsaturated fatty acids biosynthesis, have been identified. Fukeqianjin formula exerts good regulatory effect on the abnormal metabolism of PID rats. Conclusions Intrauterine inoculation of multiple pathogens combined with mechanical injury of endometrium could significantly disturb the plasma metabolic profiles of rats. Fukeqianjin formula has potential therapeutic effect on multi-pathogen-induced PID by ameliorating metabolism disorders and alleviating the inflammatory response. Electronic supplementary material The online version of this article (10.1186/s13020-018-0217-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yan Zhang
- 1School of Medicine, Chengdu University, No. 2025, Cheng Luo Road, Chengdu, 610106 Sichuan People's Republic of China
| | - Wei Li
- 1School of Medicine, Chengdu University, No. 2025, Cheng Luo Road, Chengdu, 610106 Sichuan People's Republic of China
| | - Liang Zou
- 1School of Medicine, Chengdu University, No. 2025, Cheng Luo Road, Chengdu, 610106 Sichuan People's Republic of China
| | - Yun Gong
- Zhuzhou Qianjin Pharmaceutical Ltd. Co., No. 801 Zhuzhou Avenue, Tianyuan District, Zhuzhou, 412000 Hunan People's Republic of China
| | - Peng Zhang
- Zhuzhou Qianjin Pharmaceutical Ltd. Co., No. 801 Zhuzhou Avenue, Tianyuan District, Zhuzhou, 412000 Hunan People's Republic of China
| | - Shasha Xing
- 3Drug Clinical Trial Center, Affiliated Hospital of Chengdu University, 2nd Ring Road, Jinniu District, Chengdu, 610081 Sichuan People's Republic of China
| | - Hang Yang
- 1School of Medicine, Chengdu University, No. 2025, Cheng Luo Road, Chengdu, 610106 Sichuan People's Republic of China
| |
Collapse
|
28
|
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
|
29
|
Anti-Inflammatory Effect of Feiyangchangweiyan Capsule on Rat Pelvic Inflammatory Disease through JNK/NF- κB Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2018; 2018:8476147. [PMID: 29681986 PMCID: PMC5851019 DOI: 10.1155/2018/8476147] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 01/22/2018] [Accepted: 01/24/2018] [Indexed: 11/18/2022]
Abstract
Objectives In this study, we aimed to illustrate the preventive effect and possible mechanisms of Feiyangchangweiyan capsule (FYCWYC) on rat pelvic inflammatory disease (PID) model. Methods To construct the rat PID model, upper genital tract was infected by multipathogen, and then drugs were orally administered for 8 days. The histological examination, immunohistochemical analysis, and ELISA were carried out. Furthermore, Western blotting was used to analyze the expression of Akt, MAPKs, NF-κB p65, and IκB-α in uterus. Results As the results showed, infiltrations of neutrophils and lymphocytes in uterus were significantly suppressed, and IL-1β, IL-6, CXCL-1, and TNF-α were also reduced in a dose-dependent manner. We also found that FYCWYC inhibited apoptosis induced by infection. Furthermore, FYCWYC could block the infection-induced nuclear translocation of NF-κB. We found that FYCWYC treatment only decreased the phosphorylation of JNK induced by infection and had no effects on Akt and P38. Additional, the effects of SP600125, an inhibitor of phospho-JNK, were similar to the results of FYCWYC. Conclusions Taken together, our results demonstrated that FYCWYC had anti-inflammatory effect in pathogen-induced PID model, and the mechanism might be through inhibiting NF-κB nuclear translocation which is mediated by JNK.
Collapse
|
30
|
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
|
31
|
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
|
32
|
Webster SJ, Goodall JC. New concepts in Chlamydia induced inflammasome responses. Microbes Infect 2017; 20:424-431. [PMID: 29248634 DOI: 10.1016/j.micinf.2017.11.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 10/27/2017] [Accepted: 11/21/2017] [Indexed: 02/07/2023]
Abstract
Since the concept of the inflammasome was introduced by Martinon, Burns and Tschopp in 2002, there has been an exponential increase in our understanding of how inflammasomes (caspase activating molecular platforms) regulate innate inflammatory responses to infectious microorganisms. Advances in understanding inflammasome biology have been developed using a range of bacterial pathogens. Recent studies investigating inflammasome responses during Chlamydia infection have provided interesting mechanistic insights in to inflammasome activation during intracellular bacterial infection. This review highlights new concepts regulating inflammasome activation to bacterial infections including: interferon-regulated loss of compartmentalisation, mechanisms of canonical and non-canonical inflammasome activation and their relevance to Chlamydia infections are discussed.
Collapse
Affiliation(s)
- Steve J Webster
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Hills Road, Cambridge CB2 0QQ, UK.
| | - Jane C Goodall
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Hills Road, Cambridge CB2 0QQ, UK.
| |
Collapse
|
33
|
Sun X, Tian Q, Wang L, Xue M, Zhong G. IL-6-mediated signaling pathways limit Chlamydia muridarum infection and exacerbate its pathogenicity in the mouse genital tract. Microbes Infect 2017; 19:536-545. [PMID: 28864426 PMCID: PMC6034988 DOI: 10.1016/j.micinf.2017.08.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Revised: 07/29/2017] [Accepted: 08/18/2017] [Indexed: 12/19/2022]
Abstract
Chlamydia muridarum induction of mouse hydrosalpinx, depending on both tubal infection and inflammation, has been used for investigating Chlamydia trachomatis pathogenesis. We now report that IL-6 both inhibits C. muridarum infection and exacerbates pathogenicity in the mouse genital tract. When intravaginally inoculated with a high dose of C. muridarum, IL-6-deficient mice developed more extensive genital tract infection with severe hydrosalpinx, suggesting that IL-6 is required for controlling the high dose infection but not essential for C. muridarum-induced pathology. However, at a low dose, IL-6-deficient mice still developed more extensive infection in the genital tract but no longer with significant pathology, suggesting that IL-6 is required for both controlling the low dose infection and exacerbating the low dose infection-induced pathology. The lack of hydrosalpinx in IL-6-deficient mice correlated with significantly reduced inflammatory infiltration in the oviduct tissue and decreased spleen CD4+ and CD8+ T cells that produce TNFα. Thus, IL-6-dependent pathways are important for both limiting chlamydial colonization in the genital tract mucosal tissues regardless of the infection doses and exacerbating chlamydial pathogenicity in the upper genital tract when IL-6-independent pathogenic mechanisms are not yet activated with a low infection dose.
Collapse
Affiliation(s)
- Xin Sun
- Department of Obstetrics and Gynecology, 3rd Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; 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
| | - Qi Tian
- Department of Obstetrics and Gynecology, 3rd Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; 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
| | - Luying Wang
- 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
| | - Min Xue
- Department of Obstetrics and Gynecology, 3rd Xiangya Hospital, Central South University, Changsha, Hunan 410008, 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 78229, USA.
| |
Collapse
|
34
|
Zhang Y, Shao L, Li X, Zhong G. Uterotubal junction prevents chlamydial ascension via innate immunity. PLoS One 2017; 12:e0183189. [PMID: 28797102 PMCID: PMC5552320 DOI: 10.1371/journal.pone.0183189] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Accepted: 07/31/2017] [Indexed: 12/27/2022] Open
Abstract
Ascension to the oviduct is necessary for Chlamydia to induce tubal infertility. Using the Chlamydia muridarum induction of hydrosalpinx mouse model, we have demonstrated a significant role of the uterotubal junction in preventing chlamydial ascending infection. First, delivery of C. muridarum to either side of the uterotubal junction resulted in significant reduction in live organisms from the tissues on the opposite sides. However, the recovery yields remained similar among different sections of the uterine horn. These observations suggest that the uterotubal junction may function as a barrier between the uterine horn and oviduct. Second, deficiency in innate immunity signaling pathways mediated by either MyD88 or STING significantly compromised the uterotubal junction barrier function, permitting C. muridarum to spread freely between uterine horn and oviduct. Finally, transcervical inoculation of C. muridarum led to significantly higher incidence of bilateral hydrosalpinges in the STING-deficient mice while the same inoculation mainly induced unilateral hydrosalpinx in the wild type mice, suggesting that the STING pathway-dependent uterotubal junction plays a significant role in preventing tubal pathology. Thus, we have demonstrated for the first time that the uterotubal junction is a functional barrier for preventing tubal infection by a sexually transmitted agent, providing the first in vivo evidence for detecting chlamydial infection by the STING pathway.
Collapse
Affiliation(s)
- Yuyang Zhang
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, PR China
- Department of Microbiology and Immunology, University of Texas Health Science Center at San Antonio, Texas, United States of America
| | - Lili Shao
- Department of Microbiology and Immunology, University of Texas Health Science Center at San Antonio, Texas, United States of America
| | - Xiaodong Li
- Department of Microbiology and Immunology, University of Texas Health Science Center at San Antonio, Texas, United States of America
| | - Guangming Zhong
- Department of Microbiology and Immunology, University of Texas Health Science Center at San Antonio, Texas, United States of America
- * E-mail:
| |
Collapse
|
35
|
Chlamydia muridarum with Mutations in Chromosomal Genes tc0237 and/or tc0668 Is Deficient in Colonizing the Mouse Gastrointestinal Tract. Infect Immun 2017; 85:IAI.00321-17. [PMID: 28584162 PMCID: PMC5520443 DOI: 10.1128/iai.00321-17] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Accepted: 05/26/2017] [Indexed: 12/20/2022] Open
Abstract
Chlamydiae colonize the gastrointestinal tracts of both animals and humans. However, their medical significance remains unknown. We have previously shown that wild-type Chlamydia muridarum spreads to and establishes stable colonization of the gastrointestinal tract following intravaginal inoculation. In the present study, we found that C. muridarum with mutations in chromosomal genes tc0237 and/or tc0668 was defective in spreading to the mouse gastrointestinal tract, which correlated with its attenuated pathogenicity in the upper genital tract. This correlation was more consistent than that of chlamydial pathogenicity with ascending infection in the genital tract, since attenuated C. muridarum spread significantly less to the gastrointestinal tract but maintained robust ascending infection of the upper genital tract. Transcervical inoculation further confirmed the correlation between C. muridarum spreading to the gastrointestinal tract and its pathogenicity in the upper genital tract. Finally, defective spreading of C. muridarum mutants was due to their inability to colonize the gastrointestinal tract since intragastric inoculation did not rescue the mutants' colonization. Thus, promoting C. muridarum colonization of the gastrointestinal tract may represent a primary function of the TC0237 and TC0668 proteins. Correlation of chlamydial colonization of the gastrointestinal tract with chlamydial pathogenicity in the upper genital tract suggests a potential role for gastrointestinal chlamydiae in genital tract pathogenicity.
Collapse
|
36
|
Chlamydia muridarum Infection of Macrophages Stimulates IL-1 β Secretion and Cell Death via Activation of Caspase-1 in an RIP3-Independent Manner. BIOMED RESEARCH INTERNATIONAL 2017; 2017:1592365. [PMID: 28660207 PMCID: PMC5474261 DOI: 10.1155/2017/1592365] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/18/2017] [Accepted: 05/09/2017] [Indexed: 11/18/2022]
Abstract
Chlamydiae are Gram-negative bacteria, which replicate exclusively in the infected host cells. Infection of the host cells by Chlamydiae stimulates the innate immune system leading to an inflammatory response, which is manifested not only by secretion of proinflammatory cytokines such as IL-1β from monocytes, macrophages, and dendritic cells, but also possibly by cell death mediated by Caspase-1 pyroptosis. RIP3 is a molecular switch that determines the development of necrosis or inflammation. However, the involvement of RIP3 in inflammasome activation by Chlamydia muridarum infection has not been clarified. Here, we assessed the role of RIP3 in synergy with Caspase-1 in the induction of IL-1β production in BMDM after either LPS/ATP or Chlamydia muridarum stimulation. The possibility of pyroptosis and necroptosis interplays and the role of RIP3 in IL-1β production during Chlamydia muridarum infection in BMDM was investigated as well. The data indicated that RIP3 is involved in NLRP3 inflammasome activation in LPS/ATP-stimulated BMDMs but not in Chlamydia muridarum infection. Pyroptosis occurred in BMDM after LPS/ATP stimulation or Chlamydia muridarum infection. Moreover, the results also illuminated the important role of the Caspase-1-mediated pyroptosis process which does not involve RIP3. Taken together, these observations may help shed new light on details in inflammatory signaling pathways activated by Chlamydia muridarum infection.
Collapse
|
37
|
Detection of a microbial metabolite by STING regulates inflammasome activation in response to Chlamydia trachomatis infection. PLoS Pathog 2017; 13:e1006383. [PMID: 28570638 PMCID: PMC5453623 DOI: 10.1371/journal.ppat.1006383] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Accepted: 04/26/2017] [Indexed: 01/25/2023] Open
Abstract
The innate immune system is a critical component of host defence against microbial pathogens, but effective responses require an ability to distinguish between infectious and non-infectious insult to prevent inappropriate inflammation. Using the important obligate intracellular human pathogen Chlamydia trachomatis; an organism that causes significant immunopathology, we sought to determine critical host and pathogen factors that contribute to the induction of inflammasome activation. We assayed inflammasome activation by immunoblotting and ELISA to detect IL-1β processing and LDH release to determine pyroptosis. Using primary murine bone marrow derived macrophages or human monocyte derived dendritic cells, infected with live or attenuated Chlamydia trachomatis we report that the live organism activates both canonical and non-canonical inflammasomes, but only canonical inflammasomes controlled IL-1β processing which preceded pyroptosis. NADPH oxidase deficient macrophages were permissive to Chlamydia trachomatis replication and displayed elevated type-1 interferon and inflammasome activation. Conversely, attenuated, non-replicating Chlamydia trachomatis, primed but did not activate inflammasomes and stimulated reduced type-1 interferon responses. This suggested bacterial replication or metabolism as important factors that determine interferon responses and inflammasome activation. We identified STING but not cGAS as a central mediator of interferon regulated inflammasome activation. Interestingly, exogenous delivery of a Chlamydia trachomatis metabolite and STING ligand—cyclic di-AMP, recovered inflammasome activation to attenuated bacteria in a STING dependent manner thus indicating that a bacterial metabolite is a key factor initiating inflammasome activation through STING, independent of cGAS. These data suggest a potential mechanism of how the innate immune system can distinguish between infectious and non-infectious insult and instigate appropriate immune responses that could be therapeutically targeted. Innate responses to bacterial infection such as Chlamydia trachomatis activate inflammasomes to enable the processing of IL-1β, IL-18 and the induction of an inflammatory form of cell death termed pyroptosis. Inflammasomes are crucial to host defence but require tight regulation in order to prevent inappropriate inflammation and immunopathology. Here, we demonstrate that the pro-inflammatory potential of an attenuated strain of Chlamydia trachomatis, that fails to activate the inflammasome, can be rescued by the addition of a bacterial metabolite. The requirement for this metabolite, highlights a novel mechanism of inflammasome regulation and reveals a crucial role for STING mediated interferon signalling independent of cGAS. These findings further our understanding of how the innate immune system can differentiate between potential infectious and non-infectious threats and mount appropriate immune responses.
Collapse
|
38
|
Shao L, Melero J, Zhang N, Arulanandam B, Baseman J, Liu Q, Zhong G. The cryptic plasmid is more important for Chlamydia muridarum to colonize the mouse gastrointestinal tract than to infect the genital tract. PLoS One 2017; 12:e0177691. [PMID: 28542376 PMCID: PMC5444651 DOI: 10.1371/journal.pone.0177691] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Accepted: 05/02/2017] [Indexed: 02/07/2023] Open
Abstract
Chlamydia has been detected in the gastrointestinal tracts of both animals and humans. However, the mechanism by which Chlamydia colonizes the gut remains unclear. Chlamydia muridarum is known to spread from the genital to the gastrointestinal tracts hematogenously. The C. muridarum plasmid is a key pathogenic determinant in the mouse upper genital tract although plasmid-deficient C. muridarum is still able to colonize the upper genital tract. We now report that plasmid-deficient C. muridarum exhibits significantly delayed/reduced spreading from the mouse genital to the gastrointestinal tracts. C. muridarum with or without plasmid maintained similar levels in the mouse circulatory system following intravenous inoculation but the hematogenous plasmid-deficient C. muridarum was significantly less efficient in colonizing the gastrointestinal tract. Consistently, plasmid-deficient C. muridarum failed to restore normal colonization in the gastrointestinal tract even after intragastric inoculation at a high dose. Thus, we have demonstrated a plasmid-dependent colonization of C. muridarum in the gastrointestinal tract, supporting the concept that C. muridarum may have acquired the plasmid for adaptation to the mouse gastrointestinal tract during oral-fecal transmission. Since the plasmid is more important for C. muridarum to colonize the gastrointestinal tract than to infect the genital tract, the current study has laid a foundation for further defining the host pathways targeted by the plasmid-encoded or -regulated chlamydial effectors.
Collapse
Affiliation(s)
- Lili Shao
- Department of Microbiology and Immunology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
- Department of Dermatovenereology, Tianjin Medical University General Hospital, Tianjin, China
| | - Jose Melero
- Department of Microbiology and Immunology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Nu Zhang
- Department of Microbiology and Immunology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Bernard Arulanandam
- Department of Biology, University of Texas at San Antonio, San Antonio, Texas, United States of America
| | - Joel Baseman
- Department of Microbiology and Immunology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Quanzhong Liu
- Department of Dermatovenereology, Tianjin Medical University General Hospital, Tianjin, China
| | - Guangming Zhong
- Department of Microbiology and Immunology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| |
Collapse
|
39
|
Kerr MC, Gomez GA, Ferguson C, Tanzer MC, Murphy JM, Yap AS, Parton RG, Huston WM, Teasdale RD. Laser-mediated rupture of chlamydial inclusions triggers pathogen egress and host cell necrosis. Nat Commun 2017; 8:14729. [PMID: 28281536 PMCID: PMC5353685 DOI: 10.1038/ncomms14729] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Accepted: 01/25/2017] [Indexed: 12/21/2022] Open
Abstract
Remarkably little is known about how intracellular pathogens exit the host cell in order to infect new hosts. Pathogenic chlamydiae egress by first rupturing their replicative niche (the inclusion) before rapidly lysing the host cell. Here we apply a laser ablation strategy to specifically disrupt the chlamydial inclusion, thereby uncoupling inclusion rupture from the subsequent cell lysis and allowing us to dissect the molecular events involved in each step. Pharmacological inhibition of host cell calpains inhibits inclusion rupture, but not subsequent cell lysis. Further, we demonstrate that inclusion rupture triggers a rapid necrotic cell death pathway independent of BAK, BAX, RIP1 and caspases. Both processes work sequentially to efficiently liberate the pathogen from the host cytoplasm, promoting secondary infection. These results reconcile the pathogen's known capacity to promote host cell survival and induce cell death.
Collapse
Affiliation(s)
- Markus C. Kerr
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Guillermo A. Gomez
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Charles Ferguson
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Maria C. Tanzer
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria 3052, Australia
| | - James M. Murphy
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria 3052, Australia
| | - Alpha S. Yap
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Robert G. Parton
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Wilhelmina M. Huston
- School of Life Sciences, University of Technology Sydney, Ultimo, New South Wales 2007, Australia
| | - Rohan D Teasdale
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland 4072, Australia
| |
Collapse
|
40
|
Shaw JH, Behar AR, Snider TA, Allen NA, Lutter EI. Comparison of Murine Cervicovaginal Infection by Chlamydial Strains: Identification of Extrusions Shed In vivo. Front Cell Infect Microbiol 2017; 7:18. [PMID: 28217555 PMCID: PMC5289954 DOI: 10.3389/fcimb.2017.00018] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Accepted: 01/16/2017] [Indexed: 12/23/2022] Open
Abstract
Chlamydia trachomatis is the leading cause of bacterial sexually transmitted infections (STIs) and preventable blindness. Untreated, asymptomatic infection as well as frequent re-infection are common and may drive pelvic inflammatory disease, ectopic pregnancy, and infertility. In vivo models of chlamydial infection continue to be instrumental in progress toward a vaccine and further elucidating the pathogenesis of this intracellular bacterium, however significant gaps in our understanding remain. Chlamydial host cell exit occurs via two mechanisms, lysis and extrusion, although the latter has yet to be reported in vivo and its biological role is unclear. The objective of this study was to investigate whether chlamydial extrusions are shed in vivo following infection with multiple strains of Chlamydia. We utilized an established C3H/HeJ murine cervicovaginal infection model with C. trachomatis serovars D and L2 and the Chlamydia muridarum strain MoPn to monitor the (i) time course of infection and mode of host cell exit, (ii) mucosal and systemic immune response to infection, and (iii) gross and histopathology following clearance of active infection. The key finding herein is the first identification of chlamydial extrusions shed from host cells in an in vivo model. Extrusions, a recently appreciated mode of host cell exit and potential means of dissemination, had been previously observed solely in vitro. The results of this study demonstrate that chlamydial extrusions exist in vivo and thus warrant further investigation to determine their role in chlamydial pathogenesis.
Collapse
Affiliation(s)
- Jennifer H Shaw
- Department of Integrative Biology, Oklahoma State University Stillwater, OK, USA
| | - Amanda R Behar
- Department of Microbiology and Molecular Genetics, Oklahoma State University Stillwater, OK, USA
| | - Timothy A Snider
- Department of Veterinary Pathobiology, Oklahoma State University Stillwater, OK, USA
| | - Noah A Allen
- Department of Integrative Biology, Oklahoma State University Stillwater, OK, USA
| | - Erika I Lutter
- Department of Microbiology and Molecular Genetics, Oklahoma State University Stillwater, OK, USA
| |
Collapse
|
41
|
Verma V, Dhanda RS, Møller NF, Yadav M. Inflammasomes and Their Role in Innate Immunity of Sexually Transmitted Infections. Front Immunol 2016; 7:540. [PMID: 27994587 PMCID: PMC5136550 DOI: 10.3389/fimmu.2016.00540] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2016] [Accepted: 11/16/2016] [Indexed: 12/15/2022] Open
Abstract
Inflammasomes are multiprotein complexes present in the cytosol as pattern recognition receptors or as sensors of damage-associated molecular patterns. After recognition of microbe-associated molecular patterns or host-derived danger signals, nucleotide oligomerization domain-like receptors oligomerize to form inflammasomes. The activation of inflammasomes results in an alarm, which is raised to alert adjacent cells through the processing and release of a number of other substrates present in the cytosol. A wide array of inflammasomes and their adapter molecules have been identified in the host’s innate immune system in response to various pathogens. Components of specific pathogens activate different inflammasomes, which once activated in response to pathogen-induced infection, induce the activation of caspases, and the release of mature forms of interleukin-1β (IL-1β) and IL-18. Identifying the mechanisms underlying pathogen-induced inflammasome activation is important if we are to develop novel therapeutic strategies to target sexually transmitted infections (STIs) related pathogens. This information is currently lacking in literature. In this review, we have discussed the role of various inflammasomes in sensing different STIs, as well as the beneficial or detrimental effects of inflammasome signaling in host resistance. Additionally, we have discussed both canonical and non-canonical processing of IL-1β induced with respect to particular infections. Overall, these findings transform our understanding of both the basic biology and clinical relevance of inflammasomes.
Collapse
Affiliation(s)
- Vivek Verma
- Dr. B. R. Ambedkar Center for Biomedical Research, University of Delhi , New Delhi , India
| | - Rakesh Singh Dhanda
- Department of Translational and Regenerative Medicine, Post Graduate Institute of Medical Education and Research (PGIMER) , Chandigarh , India
| | | | - Manisha Yadav
- Dr. B. R. Ambedkar Center for Biomedical Research, University of Delhi, New Delhi, India; Department of Clinical Microbiology, Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
42
|
Finethy R, Coers J. Sensing the enemy, containing the threat: cell-autonomous immunity to Chlamydia trachomatis. FEMS Microbiol Rev 2016; 40:875-893. [PMID: 28201690 PMCID: PMC5975928 DOI: 10.1093/femsre/fuw027] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Revised: 03/31/2016] [Accepted: 07/01/2016] [Indexed: 01/01/2023] Open
Abstract
The bacterium Chlamydia trachomatis is the etiological agent of the most common sexually transmitted infection in North America and Europe. Medical complications resulting from genital C. trachomatis infections arise predominantly in women where the initial infections often remain asymptomatic and thus unrecognized. Untreated asymptomatic infections in women can ascend into the upper genital tract and establish persistence, ultimately resulting in extensive scarring of the reproductive organs, pelvic inflammatory disease, infertility and ectopic pregnancies. Previously resolved C. trachomatis infections fail to provide protective immune memory, and no effective vaccine against C. trachomatis is currently available. Critical determinants of the pathogenesis and immunogenicity of genital C. trachomatis infections are cell-autonomous immune responses. Cell-autonomous immunity describes the ability of an individual host cell to launch intrinsic immune circuits that execute the detection, containment and elimination of cell-invading pathogens. As an obligate intracellular pathogen C. trachomatis is constantly under attack by cell-intrinsic host defenses. Accordingly, C. trachomatis evolved to subvert and co-opt cell-autonomous immune pathways. This review will provide a critical summary of our current understanding of cell-autonomous immunity to C. trachomatis and its role in shaping host resistance, inflammation and adaptive immunity to genital C. trachomatis infections.
Collapse
Affiliation(s)
- Ryan Finethy
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC, USA
| | - Jörn Coers
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC, USA
- Department of Immunology, Duke University Medical Center, Durham, NC, USA
| |
Collapse
|
43
|
Zhong G. Chlamydial Plasmid-Dependent Pathogenicity. Trends Microbiol 2016; 25:141-152. [PMID: 27712952 DOI: 10.1016/j.tim.2016.09.006] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Revised: 09/11/2016] [Accepted: 09/19/2016] [Indexed: 10/20/2022]
Abstract
Most Chlamydia species carry a 7.5kb plasmid encoding eight open reading frames conventionally called plasmid glycoproteins 1-8 or pGP1-8. Although the plasmid is not critical for chlamydial growth in vitro, its role in chlamydial pathogenesis is clearly demonstrated in the genital tracts of mice infected with Chlamydia muridarum, a model for investigating the human pathogen Chlamydia trachomatis. Plasmid-free C. trachomatis is also attenuated in both the mouse genital tract and nonhuman primate ocular tissue. Deficiency in pGP3 alone, which is regulated by pGP4, largely reproduced the in vivo but not in vitro phenotypes of the plasmid-free organisms, suggesting that pGP3 is a key in vivo virulence factor. The positive and negative regulations of some chromosomal genes by pGP4 and pGP5, respectively, may allow the plasmid to promote chlamydial adaptation to varied animal tissue environments. The focus of this review is to summarize the progress on the pathogenic functions of the plasmid-encoded open reading frames, which may motivate further investigation of the molecular mechanisms of chlamydial pathogenicity and development of medical utility of the chlamydial plasmid system.
Collapse
Affiliation(s)
- Guangming Zhong
- Department of Microbiology and Immunology, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA.
| |
Collapse
|
44
|
Pettengill MA, Abdul-Sater A, Coutinho-Silva R, Ojcius DM. Danger signals, inflammasomes, and the intricate intracellular lives of chlamydiae. Biomed J 2016; 39:306-315. [PMID: 27884377 PMCID: PMC6138786 DOI: 10.1016/j.bj.2016.07.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Accepted: 07/13/2016] [Indexed: 12/20/2022] Open
Abstract
Chlamydiae are obligate intracellular bacterial pathogens, and as such are sensitive to alterations in the cellular physiology of their hosts. Chlamydial infections often cause pathologic consequences due to prolonged localized inflammation. Considerable advances have been made in the last few years regarding our understanding of how two key inflammation-associated signaling pathways influence the biology of Chlamydia infections: inflammation regulating purinergic signaling pathways significantly impact intracellular chlamydial development, and inflammasome activation modulates both chlamydial growth and infection mediated pro-inflammatory cytokine production. We review here elements of both pathways, presenting the latest developments contributing to our understanding of how chlamydial infections are influenced by inflammasomes and purinergic signaling.
Collapse
Affiliation(s)
| | - Ali Abdul-Sater
- School of Kinesiology and Health Science, York University, Toronto, Ontario, Canada
| | - Robson Coutinho-Silva
- Laboratory of Immunophysiology, Carlos Chagas Filho Biophysics Institute of the Federal University of Rio de Janeiro, Brazil
| | - David M Ojcius
- Laboratory of Immunophysiology, Carlos Chagas Filho Biophysics Institute of the Federal University of Rio de Janeiro, Brazil; Department of Biomedical Sciences, University of the Pacific, Arthur Dugoni School of Dentistry, San Francisco, USA.
| |
Collapse
|
45
|
Deviant Behavior: Tick-Borne Pathogens and Inflammasome Signaling. Vet Sci 2016; 3:vetsci3040027. [PMID: 29056735 PMCID: PMC5606592 DOI: 10.3390/vetsci3040027] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Revised: 09/22/2016] [Accepted: 09/23/2016] [Indexed: 12/11/2022] Open
Abstract
In the face of an assault, host cells mount an immediate response orchestrated by innate immunity. Two of the best described innate immune signaling networks are the Toll- and the Nod-like receptor pathways. Extensive work has been done characterizing both signaling cascades with several recent advances on the forefront of inflammasome biology. In this review, we will discuss how more commonly-studied pathogens differ from tick-transmitted microbes in the context of Nod-like receptor signaling and inflammasome formation. Because pathogens transmitted by ticks have unique characteristics, we offer the opinion that these microbes can be used to uncover novel principles of Nod-like receptor biology.
Collapse
|
46
|
Lupfer C, Anand PK. Integrating Inflammasome Signaling in Sexually Transmitted Infections. Trends Immunol 2016; 37:703-714. [PMID: 27592079 PMCID: PMC5086000 DOI: 10.1016/j.it.2016.08.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Revised: 08/03/2016] [Accepted: 08/05/2016] [Indexed: 02/07/2023]
Abstract
Inflammasomes are cytosolic multiprotein platforms with pivotal roles in infectious diseases. Activation of inflammasomes results in proinflammatory cytokine signaling and pyroptosis. Sexually transmitted infections (STIs) are a major health problem worldwide, yet few studies have probed the impact of inflammasome signaling during these infections. Due to the dearth of appropriate infection models, our current understanding of inflammasomes in STIs is mostly drawn from results obtained in vitro, from distant infection sites, or from related microbial strains that are not sexually transmitted. Understanding how inflammasomes influence the outcome of STIs may lead to the development of novel and effective strategies to control disease and prevent transmission. Here we discuss and highlight the recent progress in this field.
Collapse
Affiliation(s)
- Christopher Lupfer
- Department of Biology, Missouri State University, Springfield, MO 65809, USA
| | - Paras K Anand
- Infectious Diseases and Immunity, Imperial College London, London W12 0NN, UK.
| |
Collapse
|
47
|
Intravenous Inoculation with Chlamydia muridarum Leads to a Long-Lasting Infection Restricted to the Gastrointestinal Tract. Infect Immun 2016; 84:2382-2388. [PMID: 27271744 DOI: 10.1128/iai.00432-16] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Accepted: 06/01/2016] [Indexed: 12/16/2022] Open
Abstract
Chlamydia has been detected in the gastrointestinal tracts of both animals and humans. However, it remains unclear whether the chlamydial organisms can be introduced into the gastrointestinal tract via pathways independent of the oral and anal routes. We have recently shown that Chlamydia muridarum spreads from the genital tract to the gastrointestinal tract potentially via the circulatory system. To test whether hematogenous C. muridarum can spread to and establish a long-lasting colonization in the mouse gastrointestinal tract, we inoculated mice intravenously with a luciferase-expressing C. muridarum strain and monitored its distribution. After tail vein inoculation, most luciferase-generated bioluminescence signals were detected in the mouse abdominal area throughout the experiment. The ex vivo imaging revealed that the abdominal signals came from the gastrointestinal tract tissues. Simultaneous monitoring of chlamydial organisms in individual organs or tissues revealed an initial stage of systemic spreading followed by a long-lasting infection in the gastrointestinal tract. A retro-orbital vein inoculation of the C. muridarum organisms at a lower dose in a different mouse strain also led to colonization of the gastrointestinal tract. We have demonstrated that intravenous C. muridarum inoculation can result in colonization of the gastrointestinal tract, suggesting that the chlamydial organisms may use the sexual behavior-independent circulation pathway to infect the gastrointestinal tract.
Collapse
|
48
|
Zou W, Wen X, Sheng X, Zheng YI, Xiao Z, Luo J, Chen S, Wang Y, Cheng Z, Xiang D, Nie Y. Gas chromatography-mass spectrometric method-based urine metabolomic profile of rats with pelvic inflammatory disease. Exp Ther Med 2016; 11:1653-1660. [PMID: 27168785 PMCID: PMC4840532 DOI: 10.3892/etm.2016.3142] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2015] [Accepted: 02/23/2016] [Indexed: 12/12/2022] Open
Abstract
Pelvic inflammatory disease (PID) can lead to a poor outcome of severe sequelae, and the current methods of clinical diagnosis are not satisfactory. Metabolomics is an effective method for the identification of disease-related metabolite biomarkers to facilitate disease diagnosis. However, to the best of our knowledge, no PID-associated metabolomic study has yet been carried out. The metabolomic changes of rats with PID were investigated in the present study. A PID model was constructed by the multi-pathogenic infection of the upper genital tract in rats. Infiltration of inflammatory cells and elevated expression levels of the cytokines interleukin (IL)-1β and IL-6 in the uterus and fallopian tubes validated the disease model. Gas chromatography-mass spectrometry coupled with derivatization was used to determine the urine metabolomic profile. Principal component analysis and partial least squares-discriminant analysis of the data sets showed a clear separation of metabolic profiles between rats with PID and control rats. Eighteen differentiating metabolites were found, including four amino acids, three fatty acids, nine organic acids, and two sugars, which indicated alterations in sugar metabolism, the citric acid cycle, amino acid metabolism and fatty acid metabolism. These metabolites could be potential biomarkers of PID, and this research may offer a new approach to evaluate the effect of anti-PID drugs in pre-clinical or clinical trials.
Collapse
Affiliation(s)
- Wei Zou
- Key Laboratory of Hunan Province for Traditional Chinese Medicine in Obstetrics and Gynecology Research, The Maternal and Child Health Hospital of Hunan Province, Changsha, Hunan 410008, P.R. China
| | - Xiaoke Wen
- Key Laboratory of Hunan Province for Traditional Chinese Medicine in Obstetrics and Gynecology Research, The Maternal and Child Health Hospital of Hunan Province, Changsha, Hunan 410008, P.R. China
| | - Xiaoqi Sheng
- Technical Research Centre of Clinical Preventive and Treatment for Children, The Maternal and Child Health Hospital of Hunan Province, Changsha, Hunan 410008, P.R. China
| | - Y I Zheng
- Key Laboratory of Hunan Province for Traditional Chinese Medicine in Obstetrics and Gynecology Research, The Maternal and Child Health Hospital of Hunan Province, Changsha, Hunan 410008, P.R. China
| | - Zuoqi Xiao
- Key Laboratory of Hunan Province for Traditional Chinese Medicine in Obstetrics and Gynecology Research, The Maternal and Child Health Hospital of Hunan Province, Changsha, Hunan 410008, P.R. China
| | - Jieying Luo
- College of Pharmacy, Hunan University of Traditional Chinese Medicine, Changsha, Hunan 410007, P.R. China
| | - Shuqiong Chen
- Traditional Chinese Medicine Department of Gynecology, The Maternal and Child Health Hospital of Hunan Province, Changsha, Hunan 410008, P.R. China
| | - Yichao Wang
- Technical Research Centre of Clinical Preventive and Treatment for Children, The Maternal and Child Health Hospital of Hunan Province, Changsha, Hunan 410008, P.R. China
| | - Zeneng Cheng
- School of Pharmaceutical Sciences, Central South University, Changsha, Hunan 410011, P.R. China
| | - Daxiong Xiang
- Clinic Pharmacy Research Laboratory, Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, P.R. China
| | - Yichu Nie
- State Key Laboratory of Respiratory Diseases, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510120, P.R. China
| |
Collapse
|
49
|
Cerny KL, Ribeiro RAC, Jeoung M, Ko C, Bridges PJ. Estrogen Receptor Alpha (ESR1)-Dependent Regulation of the Mouse Oviductal Transcriptome. PLoS One 2016; 11:e0147685. [PMID: 26808832 PMCID: PMC4725743 DOI: 10.1371/journal.pone.0147685] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Accepted: 01/07/2016] [Indexed: 02/06/2023] Open
Abstract
Estrogen receptor-α (ESR1) is an important transcriptional regulator in the mammalian oviduct, however ESR1-dependent regulation of the transcriptome of this organ is not well defined, especially at the genomic level. The objective of this study was therefore to investigate estradiol- and ESR1-dependent regulation of the transcriptome of the oviduct using transgenic mice, both with (ESR1KO) and without (wild-type, WT) a global deletion of ESR1. Oviducts were collected from ESR1KO and WT littermates at 23 days of age, or ESR1KO and WT mice were treated with 5 IU PMSG to stimulate follicular development and the production of ovarian estradiol, and the oviducts collected 48 h later. RNA extracted from whole oviducts was hybridized to Affymetrix Genechip Mouse Genome 430–2.0 arrays (n = 3 arrays per genotype and treatment) or reverse transcribed to cDNA for analysis of the expression of selected mRNAs by real-time PCR. Following microarray analysis, a statistical two-way ANOVA and pairwise comparison (LSD test) revealed 2428 differentially expressed transcripts (DEG’s, P < 0.01). Genotype affected the expression of 2215 genes, treatment (PMSG) affected the expression of 465 genes, and genotype x treatment affected the expression of 438 genes. With the goal of determining estradiol/ESR1-regulated function, gene ontology (GO) and bioinformatic pathway analyses were performed on DEG’s in the oviducts of PMSG-treated ESR1KO versus PMSG-treated WT mice. Significantly enriched GO molecular function categories included binding and catalytic activity. Significantly enriched GO cellular component categories indicated the extracellular region. Significantly enriched GO biological process categories involved a single organism, modulation of a measurable attribute and developmental processes. Bioinformatic analysis revealed ESR1-regulation of the immune response within the oviduct as the primary canonical pathway. In summary, a transcriptomal profile of estradiol- and ESR1-regulated gene expression and related bioinformatic analysis is presented to increase our understanding of how estradiol/ESR1 affects function of the oviduct, and to identify genes that may be proven as important regulators of fertility in the future.
Collapse
Affiliation(s)
- Katheryn L. Cerny
- Department of Animal and Food Sciences, University of Kentucky, Lexington, KY 40546, United States of America
| | - Rosanne A. C. Ribeiro
- Department of Animal and Food Sciences, University of Kentucky, Lexington, KY 40546, United States of America
| | - Myoungkun Jeoung
- Department of Clinical Sciences, University of Kentucky, Lexington, KY 40536, United States of America
| | - CheMyong Ko
- Department of Comparative Biosciences, University of Illinois at Urbana-Champaign, Urbana, IL 61802, United States of America
| | - Phillip J. Bridges
- Department of Animal and Food Sciences, University of Kentucky, Lexington, KY 40546, United States of America
- Department of Clinical Sciences, University of Kentucky, Lexington, KY 40536, United States of America
- * E-mail:
| |
Collapse
|
50
|
Murthy AK, Li W, Ramsey KH. Immunopathogenesis of Chlamydial Infections. Curr Top Microbiol Immunol 2016; 412:183-215. [PMID: 27370346 DOI: 10.1007/82_2016_18] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Chlamydial infections lead to a number of clinically relevant diseases and induce significant morbidity in human populations. It is generally understood that certain components of the host immune response to infection also mediate such disease pathologies. A clear understanding of pathogenic mechanisms will enable us to devise better preventive and/or intervention strategies to mitigate the morbidity caused by these infections. Over the years, numerous studies have been conducted to explore the immunopathogenic mechanisms of Chlamydia-induced diseases of the eye, reproductive tract, respiratory tract, and cardiovascular systems. In this article, we provide an overview of the diseases caused by Chlamydia, animal models used to study disease pathology, and a historical context to the efforts to understand chlamydial pathogenesis. Furthermore, we discuss recent findings regarding pathogenesis, with an emphasis on the role of the adaptive immune response in the development of chlamydial disease sequelae. Finally, we summarize the key insights obtained from studies of chlamydial pathogenesis and avenues that remain to be explored in order to inform the next steps of vaccine development against chlamydial infections.
Collapse
Affiliation(s)
- Ashlesh K Murthy
- Department of Pathology, Midwestern University, 555, 31st Steet, Downers Grove, IL, 60515, USA.
| | - Weidang Li
- Department of Pathology, Midwestern University, 555, 31st Steet, Downers Grove, IL, 60515, USA
| | - Kyle H Ramsey
- Department of Microbiology and Immunology, Midwestern University, Downers Grove, IL, 60515, USA
| |
Collapse
|