1
|
Rodrigues R, Sousa C, Barros A, Vale N. Chlamydia trachomatis: From Urogenital Infections to the Pathway of Infertility. Genes (Basel) 2025; 16:205. [PMID: 40004534 PMCID: PMC11855039 DOI: 10.3390/genes16020205] [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: 01/10/2025] [Revised: 02/02/2025] [Accepted: 02/05/2025] [Indexed: 02/27/2025] Open
Abstract
Chlamydia trachomatis (CT) is a major cause of sexually transmitted infections (STIs) worldwide, with significant implications for reproductive health. The bacterium's genome contains highly polymorphic regions, influencing both the type and severity of infections. These genetic variations, particularly those occurring in the major outer membrane protein (MOMP) gene, are critical for classifying the bacterium into distinct serovars and enable CT to adapt to diverse host environments, contributing to its immune evasion, persistence, and pathogenicity. Persistent or untreated urogenital infections can lead to chronic inflammation, tissue damage, and pelvic inflammatory disease, ultimately increasing the risk of ectopic pregnancy, spontaneous abortion, and infertility. This review consolidates current knowledge on the genetic diversity of CT, its potential role in modulating infection outcomes, and its immune evasion mechanisms. By integrating scientific evidence linking chlamydial infections to infertility, we underscore the urgent need for targeted research to address this critical public health challenge.
Collapse
Affiliation(s)
- Rafaela Rodrigues
- PerMed Research Group, RISE-Health, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal; (R.R.); (C.S.); (A.B.)
- Molecular Diagnostics Laboratory, Unilabs Portugal, Centro Empresarial Lionesa Porto, Rua Lionesa, Leça do Balio, 4465-671 Matosinhos, Portugal
| | - Carlos Sousa
- PerMed Research Group, RISE-Health, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal; (R.R.); (C.S.); (A.B.)
- Molecular Diagnostics Laboratory, Unilabs Portugal, Centro Empresarial Lionesa Porto, Rua Lionesa, Leça do Balio, 4465-671 Matosinhos, Portugal
| | - Alberto Barros
- PerMed Research Group, RISE-Health, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal; (R.R.); (C.S.); (A.B.)
- RISE-Health, Department of Pathology, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
- Centre for Reproductive Genetics Alberto Barros, 4100-012 Porto, Portugal
| | - Nuno Vale
- PerMed Research Group, RISE-Health, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal; (R.R.); (C.S.); (A.B.)
- RISE-Health, Department of Community Medicine, Health Information and Decision (MEDCIDS), Faculty of Medicine, University of Porto, Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal
| |
Collapse
|
2
|
Reot L, Adapen C, Cannou C, Nunez N, Lakoum S, Pimienta C, Lacroix L, Binois O, Frydman N, Nugeyre MT, Le Grand R, Menu E. Seminal plasma inhibits Chlamydia trachomatis infection in vitro, and may have consequences on mucosal immunity. Sci Rep 2024; 14:21050. [PMID: 39251689 PMCID: PMC11384662 DOI: 10.1038/s41598-024-71499-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 08/28/2024] [Indexed: 09/11/2024] Open
Abstract
Seminal plasma (SP) is the main vector of C. trachomatis (CT) during heterosexual transmission from male to female. It has immunomodulatory properties and impacts the susceptibility to HIV-1 infection, but its role has not been explored during CT infection. In the female reproductive tract (FRT), CT infection induces cytokine production and neutrophil recruitment. The role of neutrophils during CT infection is partially described, they could be at the origin of the pathology observed during CT infection. During this study, we developed an experimental in vitro model to characterize the impact of CT infection and SP on endocervical epithelial cell immune response in the FRT. We also studied the impact of the epithelial cell response on neutrophil phenotype and functions. We showed that the production by epithelial cells of pro-inflammatory cytokines increased during CT infection. Moreover, the pool of SP as well as individuals SP inhibited CT infection in a dose-dependent manner. The pool of SP inhibited cytokine production in a dose-dependent manner. The pool of SP altered gene expression profiles of infected cells. The culture supernatants of cells infected or not with CT, in presence or not of the pool of SP, had an impact on neutrophil phenotype and functions: they affected markers of neutrophil maturation, activation and adhesion capacity, as well as the survival, ROS production and phagocytosis ability. This study proposes a novel approach to study the impact of the environment on the phenotype and functions of neutrophils in the FRT. It highlights the impact of the factors of the FRT environment, in particular SP and CT infection, on the mucosal inflammation and the need to take into account the SP component while studying sexually transmitted infections during heterosexual transmission from male to female.
Collapse
Affiliation(s)
- Louis Reot
- Center for Immunology of Viral, Auto-Immune, Hematological and Bacterial Diseases [IMVA-HB/Infectious Disease Models and Innovative Therapies (IDMIT)], Commissariat à l'Energie Atomique et aux Energies Alternatives (CEA), Inserm, Université Paris-Saclay, Fontenay-aux-Roses, France
| | - Cindy Adapen
- Center for Immunology of Viral, Auto-Immune, Hematological and Bacterial Diseases [IMVA-HB/Infectious Disease Models and Innovative Therapies (IDMIT)], Commissariat à l'Energie Atomique et aux Energies Alternatives (CEA), Inserm, Université Paris-Saclay, Fontenay-aux-Roses, France
| | - Claude Cannou
- Center for Immunology of Viral, Auto-Immune, Hematological and Bacterial Diseases [IMVA-HB/Infectious Disease Models and Innovative Therapies (IDMIT)], Commissariat à l'Energie Atomique et aux Energies Alternatives (CEA), Inserm, Université Paris-Saclay, Fontenay-aux-Roses, France
- Mucosal Immunity and Sexually Transmitted Infection Control (MISTIC) Group, Department of Virology, Institut Pasteur, Université Paris Cité, Paris, France
| | | | | | - Camille Pimienta
- Center for Immunology of Viral, Auto-Immune, Hematological and Bacterial Diseases [IMVA-HB/Infectious Disease Models and Innovative Therapies (IDMIT)], Commissariat à l'Energie Atomique et aux Energies Alternatives (CEA), Inserm, Université Paris-Saclay, Fontenay-aux-Roses, France
| | - Laetitia Lacroix
- Center for Immunology of Viral, Auto-Immune, Hematological and Bacterial Diseases [IMVA-HB/Infectious Disease Models and Innovative Therapies (IDMIT)], Commissariat à l'Energie Atomique et aux Energies Alternatives (CEA), Inserm, Université Paris-Saclay, Fontenay-aux-Roses, France
| | - Olivier Binois
- Service de Biologie de la Reproduction CECOS, Assistance Publique Hôpitaux de Paris, Hôpital Antoine Béclère, Université Paris-Saclay, Clamart, France
| | - Nelly Frydman
- Service de Biologie de la Reproduction CECOS, Assistance Publique Hôpitaux de Paris, Hôpital Antoine Béclère, Université Paris-Saclay, Clamart, France
| | - Marie-Thérèse Nugeyre
- Center for Immunology of Viral, Auto-Immune, Hematological and Bacterial Diseases [IMVA-HB/Infectious Disease Models and Innovative Therapies (IDMIT)], Commissariat à l'Energie Atomique et aux Energies Alternatives (CEA), Inserm, Université Paris-Saclay, Fontenay-aux-Roses, France
- Mucosal Immunity and Sexually Transmitted Infection Control (MISTIC) Group, Department of Virology, Institut Pasteur, Université Paris Cité, Paris, France
| | - Roger Le Grand
- Center for Immunology of Viral, Auto-Immune, Hematological and Bacterial Diseases [IMVA-HB/Infectious Disease Models and Innovative Therapies (IDMIT)], Commissariat à l'Energie Atomique et aux Energies Alternatives (CEA), Inserm, Université Paris-Saclay, Fontenay-aux-Roses, France
| | - Elisabeth Menu
- Center for Immunology of Viral, Auto-Immune, Hematological and Bacterial Diseases [IMVA-HB/Infectious Disease Models and Innovative Therapies (IDMIT)], Commissariat à l'Energie Atomique et aux Energies Alternatives (CEA), Inserm, Université Paris-Saclay, Fontenay-aux-Roses, France.
- Mucosal Immunity and Sexually Transmitted Infection Control (MISTIC) Group, Department of Virology, Institut Pasteur, Université Paris Cité, Paris, France.
| |
Collapse
|
3
|
Walker FC, Derré I. Contributions of diverse models of the female reproductive tract to the study of Chlamydia trachomatis-host interactions. Curr Opin Microbiol 2024; 77:102416. [PMID: 38103413 PMCID: PMC10922760 DOI: 10.1016/j.mib.2023.102416] [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: 09/21/2023] [Revised: 11/20/2023] [Accepted: 11/21/2023] [Indexed: 12/19/2023]
Abstract
Chlamydia trachomatis is a common cause of sexually transmitted infections in humans with devastating sequelae. Understanding of disease on all scales, from molecular details to the immunology underlying pathology, is essential for identifying new ways of preventing and treating chlamydia. Infection models of various complexity are essential to understand all aspects of chlamydia pathogenesis. Cell culture systems allow for research into molecular details of infection, including characterization of the unique biphasic Chlamydia developmental cycle and the role of type-III-secreted effectors in modifying the host environment to allow for infection. Multicell type and organoid culture provide means to investigate how cells other than the infected cells contribute to the control of infection. Emerging comprehensive three-dimensional biomimetic systems may fill an important gap in current models to provide information on complex phenotypes that cannot be modeled in simpler in vitro models.
Collapse
Affiliation(s)
- Forrest C Walker
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA, United States of America
| | - Isabelle Derré
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA, United States of America.
| |
Collapse
|
4
|
Wang X, Wu H, Fang C, Li Z. Insights into innate immune cell evasion by Chlamydia trachomatis. Front Immunol 2024; 15:1289644. [PMID: 38333214 PMCID: PMC10850350 DOI: 10.3389/fimmu.2024.1289644] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 01/11/2024] [Indexed: 02/10/2024] Open
Abstract
Chlamydia trachomatis, is a kind of obligate intracellular pathogen. The removal of C. trachomatis relies primarily on specific cellular immunity. It is currently considered that CD4+ Th1 cytokine responses are the major protective immunity against C. trachomatis infection and reinfection rather than CD8+ T cells. The non-specific immunity (innate immunity) also plays an important role in the infection process. To survive inside the cells, the first process that C. trachomatis faces is the innate immune response. As the "sentry" of the body, mast cells attempt to engulf and remove C. trachomatis. Dendritic cells present antigen of C. trachomatis to the "commanders" (T cells) through MHC-I and MHC-II. IFN-γ produced by activated T cells and natural killer cells (NK) further activates macrophages. They form the body's "combat troops" and produce immunity against C. trachomatis in the tissues and blood. In addition, the role of eosinophils, basophils, innate lymphoid cells (ILCs), natural killer T (NKT) cells, γδT cells and B-1 cells should not be underestimated in the infection of C. trachomatis. The protective role of innate immunity is insufficient, and sexually transmitted diseases (STDs) caused by C. trachomatis infections tend to be insidious and recalcitrant. As a consequence, C. trachomatis has developed a unique evasion mechanism that triggers inflammatory immunopathology and acts as a bridge to protective to pathological adaptive immunity. This review focuses on the recent advances in how C. trachomatis evades various innate immune cells, which contributes to vaccine development and our understanding of the pathophysiologic consequences of C. trachomatis infection.
Collapse
Affiliation(s)
| | | | | | - Zhongyu Li
- Institute of Pathogenic Biology, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, School of Nursing, Hengyang Medical College, University of South China, Hengyang, China
| |
Collapse
|
5
|
Dolat L, Carpenter VK, Chen YS, Suzuki M, Smith EP, Kuddar O, Valdivia RH. Chlamydia repurposes the actin-binding protein EPS8 to disassemble epithelial tight junctions and promote infection. Cell Host Microbe 2022; 30:1685-1700.e10. [PMID: 36395759 PMCID: PMC9793342 DOI: 10.1016/j.chom.2022.10.013] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 08/08/2022] [Accepted: 10/21/2022] [Indexed: 11/17/2022]
Abstract
Invasive microbial pathogens often disrupt epithelial barriers, yet the mechanisms used to dismantle tight junctions are poorly understood. Here, we show that the obligate pathogen Chlamydia trachomatis uses the effector protein TepP to transiently disassemble tight junctions early during infection. TepP alters the tyrosine phosphorylation status of host proteins involved in cytoskeletal regulation, including the filamentous actin-binding protein EPS8. We determined that TepP and EPS8 are necessary and sufficient to remodel tight junctions and that the ensuing disruption of epithelial barrier function promotes secondary invasion events. The genetic deletion of EPS8 renders epithelial cells and endometrial organoids resistant to TepP-mediated tight junction remodeling. Finally, TepP and EPS8 promote infection in murine models of infections, with TepP mutants displaying defects in ascension to the upper genital tract. These findings reveal a non-canonical function of EPS8 in the disassembly of epithelial junctions and an important role for Chlamydia pathogenesis.
Collapse
Affiliation(s)
- Lee Dolat
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Victoria K Carpenter
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Yi-Shan Chen
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Michitaka Suzuki
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Erin P Smith
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Ozge Kuddar
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Raphael H Valdivia
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA.
| |
Collapse
|
6
|
Chlamydia trachomatis suppresses host cell store-operated Ca 2+ entry and inhibits NFAT/calcineurin signaling. Sci Rep 2022; 12:21406. [PMID: 36496532 PMCID: PMC9741641 DOI: 10.1038/s41598-022-25786-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 12/05/2022] [Indexed: 12/13/2022] Open
Abstract
The obligate intracellular bacterium, Chlamydia trachomatis, replicates within a parasitophorous vacuole termed an inclusion. During development, host proteins critical for regulating intracellular calcium (Ca2+) homeostasis interact with the inclusion membrane. The inclusion membrane protein, MrcA, interacts with the inositol-trisphosphate receptor (IP3R), an ER cationic channel that conducts Ca2+. Stromal interaction molecule 1 (STIM1), an ER transmembrane protein important for regulating store-operated Ca2+ entry (SOCE), localizes to the inclusion membrane via an uncharacterized interaction. We therefore examined Ca2+ mobilization in C. trachomatis infected cells. Utilizing a variety of Ca2+ indicators to assess changes in cytosolic Ca2+ concentration, we demonstrate that C. trachomatis impairs host cell SOCE. Ca2+ regulates many cellular signaling pathways. We find that the SOCE-dependent NFAT/calcineurin signaling pathway is impaired in C. trachomatis infected HeLa cells and likely has major implications on host cell physiology as it relates to C. trachomatis pathogenesis.
Collapse
|
7
|
Edwards VL, McComb E, Gleghorn JP, Forney L, Bavoil PM, Ravel J. Three-dimensional models of the cervicovaginal epithelia to study host-microbiome interactions and sexually transmitted infections. Pathog Dis 2022; 80:6655985. [PMID: 35927516 PMCID: PMC9419571 DOI: 10.1093/femspd/ftac026] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 06/14/2022] [Accepted: 08/02/2022] [Indexed: 02/03/2023] Open
Abstract
2D cell culture systems have historically provided controlled, reproducible means to analyze host-pathogen interactions observed in the human reproductive tract. Although inexpensive, straightforward, and requiring a very short time commitment, these models recapitulate neither the functionality of multilayered cell types nor the associated microbiome that occurs in a human. Animal models have commonly been used to recreate the complexity of human infections. However, extensive modifications of animal models are required to recreate interactions that resemble those in the human reproductive tract. 3D cell culture models have emerged as alternative means of reproducing vital elements of human infections at a fraction of the cost of animal models and on a scale that allows for replicative experiments. Here, we describe a new 3D model that utilizes transwells with epithelial cells seeded apically and a basolateral extracellular matrix (ECM)-like layer. The model produced tissues with morphologic and physiological resemblance to human cervical and vaginal epithelia, including mucus levels produced by cervical cells. Infection by Chlamydia trachomatis and Neisseria gonorrhoeae was demonstrated, as well as the growth of bacterial species observed in the human vaginal microbiota. This enabled controlled mechanistic analyses of the interactions between host cells, the vaginal microbiota, and STI pathogens. Affordable and semi high-throughput 3D models of the cervicovaginal epithelia that are physiologically relevant by sustaining vaginal bacterial colonization, and facilitate studies of chlamydial and gonococcal infections.
Collapse
Affiliation(s)
- Vonetta L Edwards
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, United States,Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, United States
| | | | - Jason P Gleghorn
- Department of Biomedical Engineering, University of Delaware, Newark, DE, United States
| | - Larry Forney
- Department of Biological Sciences, University of Idaho, Moscow, ID, United States
| | - Patrik M Bavoil
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, United States,Department of Microbial Pathogenesis, University of Maryland, Baltimore, MD, United States
| | - Jacques Ravel
- Corresponding author: Institute for Genome Sciences and Department of Microbiology and Immunology, University of Maryland School of Medicine, Health Science Research Facility (HSRDF), 670 W. Baltimore Street, Baltimore, MD 21201, United States. Tel: +1 410-706-5674; E-mail:
| |
Collapse
|
8
|
Park D, Reddy AP, Wilmarth PA, Jensen JT, Han L. Mucus secretions from a conditionally reprogrammed primary endocervical cell culture. F&S SCIENCE 2022; 3:159-165. [PMID: 35560013 PMCID: PMC9947459 DOI: 10.1016/j.xfss.2022.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 03/27/2022] [Accepted: 03/29/2022] [Indexed: 11/27/2022]
Abstract
OBJECTIVE To determine if the secretions collected from a conditionally reprogrammed primary endocervical cell culture are suitable surrogates for mucus studies. DESIGN Experimental. SETTING University research center. ANIMAL(S) Female rhesus macaque (n = 2). INTERVENTION(S) None. MAIN OUTCOME MEASURE(S) Quantitative proteomic analysis using tandem mass tag mass spectrometry liquid chromatography/tandem mass spectrometry. RESULT(S) We identified 3,047 proteins, common proteins present in both primary endocervical cell cultures and the mucus of rhesus macaques. We found a 71% overlap in the top 500 most prevalent proteins in the samples. Cell culture secretions contained many essential mucus proteins, including MUC5B, the primary mucin of the endocervix. CONCLUSION(S) Similarities in secreted proteins suggest that conditionally reprogrammed primary endocervical cells could be used to study mucus secretion in vitro.
Collapse
Affiliation(s)
- Daye Park
- Augusta University/University of Georgia Medical Partnership, Athens, Georgia.
| | - Ashok P. Reddy
- Proteomic Shared Resource, Oregon Health & Science University, Portland, OR, USA
| | - Phillip A. Wilmarth
- Proteomic Shared Resource, Oregon Health & Science University, Portland, OR, USA
| | - Jeffrey T. Jensen
- Oregon Health & Science University, Department of Obstetrics and Gynecology, Portland, OR, USA,Oregon National Primate Research Center, Division of Reproductive and Developmental Sciences, Portland, OR, USA
| | - Leo Han
- Oregon Health & Science University, Department of Obstetrics and Gynecology, Portland, OR, USA,Oregon National Primate Research Center, Division of Reproductive and Developmental Sciences, Portland, OR, USA
| |
Collapse
|
9
|
Barnes AB, Keener RM, Schott BH, Wang L, Valdivia RH, Ko DC. Human genetic diversity regulating the TLR10/TLR1/TLR6 locus confers increased cytokines in response to Chlamydia trachomatis. HGG ADVANCES 2022; 3:100071. [PMID: 35047856 PMCID: PMC8756536 DOI: 10.1016/j.xhgg.2021.100071] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 11/15/2021] [Indexed: 12/25/2022] Open
Abstract
Human genetic diversity can have profound effects on health outcomes upon exposure to infectious agents. For infections with Chlamydia trachomatis (C. trachomatis), the wide range of genital and ocular disease manifestations are likely influenced by human genetic differences that regulate interactions between C. trachomatis and host cells. We leveraged this diversity in cellular responses to demonstrate the importance of variation at the Toll-like receptor 1 (TLR1), TLR6, and TLR10 locus to cytokine production in response to C. trachomatis. We determined that a single-nucleotide polymorphism (SNP) (rs1057807), located in a region that forms a loop with the TLR6 promoter, is associated with increased expression of TLR1, TLR6, and TLR10 and secreted levels of ten C. trachomatis-induced cytokines. Production of these C. trachomatis-induced cytokines is primarily dependent on MyD88 and TLR6 based on experiments using inhibitors, blocking antibodies, RNAi, and protein overexpression. Population genetic analyses further demonstrated that the mean IL-6 response of cells from two European populations were higher than the mean response of cells from three African populations and that this difference was partially attributable to variation in rs1057807 allele frequency. In contrast, a SNP associated with a different pro-inflammatory cytokine (rs2869462 associated with the chemokine CXCL10) exhibited an opposite response, underscoring the complexity of how different genetic variants contribute to an individual's immune response. This multidisciplinary study has identified a long-range chromatin interaction and genetic variation that regulates TLR6 to broaden our understanding of how human genetic variation affects the C. trachomatis-induced immune response.
Collapse
Affiliation(s)
- Alyson B. Barnes
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, Durham, NC 27710, USA
| | - Rachel M. Keener
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, Durham, NC 27710, USA
- University Program in Genetics and Genomics, Duke University, Durham, NC 27710, USA
| | - Benjamin H. Schott
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, Durham, NC 27710, USA
- University Program in Genetics and Genomics, Duke University, Durham, NC 27710, USA
| | - Liuyang Wang
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, Durham, NC 27710, USA
| | - Raphael H. Valdivia
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, Durham, NC 27710, USA
| | - Dennis C. Ko
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, Durham, NC 27710, USA
- University Program in Genetics and Genomics, Duke University, Durham, NC 27710, USA
- Division of Infectious Diseases, Department of Medicine, School of Medicine, Duke University, Durham, NC 27710, USA
| |
Collapse
|
10
|
Lock JY, Caboni M, Strandwitz P, Morrissette M, DiBona K, Joughin BA, Lewis K, Carrier RL. An in vitro intestinal model captures immunomodulatory properties of the microbiota in inflammation. Gut Microbes 2022; 14:2039002. [PMID: 35316142 PMCID: PMC8942420 DOI: 10.1080/19490976.2022.2039002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 01/24/2022] [Indexed: 02/04/2023] Open
Abstract
Considerable effort has been put forth to understand mechanisms by which the microbiota modulates and responds to inflammation. Here, we explored whether oxidation metabolites produced by the host during inflammation, sodium nitrate and trimethylamine oxide, impact the composition of a human stool bacterial population in a gut simulator. We then assessed whether an immune-competent in vitro intestinal model responded differently to spent medium from bacteria exposed to these cues compared to spent medium from a control bacterial population. The host-derived oxidation products were found to decrease levels of Bacteroidaceae and overall microbiota metabolic potential, while increasing levels of proinflammatory Enterobacteriaceae and lipopolysaccharide in bacterial cultures, reflecting shifts that occur in vivo in inflammation. Spent microbiota media induced elevated intracellular mucin levels and reduced intestinal monolayer integrity as reflected in transepithelial electrical resistance relative to fresh medium controls. However, multiplexed cytokine analysis revealed markedly different cytokine signatures from intestinal cultures exposed to spent medium with added oxidation products relative to spent control medium, while cytokine signatures of cultures exposed to fresh media were similar regardless of addition of host-derived cues. Further, the presence of immune cells in the intestinal model was required for this differentiation of cytokine signatures. This study indicates that simple in vitro immune-competent intestinal models can capture bacterial-mammalian cross-talk in response to host-derived oxidation products and supports utility of these systems for mechanistic studies of interactions between the gut microbiome and host in inflammation.
Collapse
Affiliation(s)
- Jaclyn Y. Lock
- Department of Bioengineering, Northeastern University, Boston, Massachusetts, USA
| | - Mariaelena Caboni
- Antimicrobial Discovery Center, Department of Biology, Northeastern University, Boston, Massachusetts, USA
| | - Philip Strandwitz
- Antimicrobial Discovery Center, Department of Biology, Northeastern University, Boston, Massachusetts, USA
| | - Madeleine Morrissette
- Antimicrobial Discovery Center, Department of Biology, Northeastern University, Boston, Massachusetts, USA
| | - Kevin DiBona
- Department of Biochemistry, Northeastern University, Boston, Massachusetts, USA
| | - Brian A. Joughin
- The Koch Institute for Integrative Cancer Research at Mit and the Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massacusetts, USA
| | - Kim Lewis
- Antimicrobial Discovery Center, Department of Biology, Northeastern University, Boston, Massachusetts, USA
| | - Rebecca L. Carrier
- Department of Bioengineering, Northeastern University, Boston, Massachusetts, USA
- Department of Chemical Engineering, Northeastern University, Boston, Massachusetts, USA
- Department of Biology, Northeastern University, Boston, Massachusetts, USA
| |
Collapse
|
11
|
Mott PD, Taylor CM, Lillis RA, Ardizzone CM, Albritton HL, Luo M, Calabresi KG, Martin DH, Myers L, Quayle AJ. Differences in the Genital Microbiota in Women Who Naturally Clear Chlamydia trachomatis Infection Compared to Women Who Do Not Clear; A Pilot Study. Front Cell Infect Microbiol 2021; 11:615770. [PMID: 33912473 PMCID: PMC8072278 DOI: 10.3389/fcimb.2021.615770] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 03/03/2021] [Indexed: 02/06/2023] Open
Abstract
In vitro studies indicate IFNγ is central to Chlamydia trachomatis (Ct) eradication, but its function may be compromised by anaerobes typically associated with bacterial vaginosis (BV), a frequent co-morbidity in women with Ct. Here we investigated the associations between natural clearance of cervical Ct infection, the vaginal microbiome, and the requirements for IFNγ by evaluating the vaginal microbial and cytokine composition of Ct treatment visit samples from women who cleared Ct infection in the interim between their Ct screening and Ct treatment visit. The pilot cohort was young, predominantly African American, and characterized by a high rate of BV that was treated with metronidazole at the Ct screening visit. The rate of natural Ct clearance was 23.6% by the Ct treatment visit (median 9 days). 16S rRNA gene sequencing revealed that metronidazole-treated women who had a Lactobacillus spp.-dominant vaginal microbiota (CST 2 or 3) at the Ct treatment visit, were more prevalent in the Ct clearing population than the non-clearing population (86% v. 50%). L. iners (CST2) was the major Lactobacillus spp. present in Ct clearers, and 33% still remained anaerobe-dominant (CST1). Vaginal IFNγ levels were not significantly different in Ct clearers and non-clearers and were several logs lower than that required for killing Ct in vitro. An expanded panel of IFNγ-induced and proinflammatory cytokines and chemokines also did not reveal differences between Ct clearers and non-clearers, but, rather, suggested signatures better associated with specific CSTs. Taken together, these findings suggest that BV-associated bacteria may impede Ct clearance, but a Lactobacillus spp.-dominant microbiome is not an absolute requirement to clear. Further, IFNγ may be required at lower concentrations than in vitro modeling indicates, suggesting it may act together with other factors in vivo. Data also revealed that the vaginal bacteria-driven inflammation add complexity to the genital cytokine milieu, but changes in this microbiota may contribute to, or provide cytokine biomarkers, for a shift to Ct clearance.
Collapse
Affiliation(s)
- Patricia Dehon Mott
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Christopher M. Taylor
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Rebecca A. Lillis
- Division of Infectious Diseases, Department of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Caleb M. Ardizzone
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Hannah L. Albritton
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Meng Luo
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Kaitlyn G. Calabresi
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - David H. Martin
- Division of Infectious Diseases, Department of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Leann Myers
- Department of Biostatistics and Data Science, Tulane University School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, United States
| | - Alison J. Quayle
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| |
Collapse
|
12
|
Primary ectocervical epithelial cells display lower permissivity to Chlamydia trachomatis than HeLa cells and a globally higher pro-inflammatory profile. Sci Rep 2021; 11:5848. [PMID: 33712643 PMCID: PMC7955086 DOI: 10.1038/s41598-021-85123-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 02/24/2021] [Indexed: 12/24/2022] Open
Abstract
The tumoral origin and extensive passaging of HeLa cells, a most commonly used cervical epithelial cell line, raise concerns on their suitability to study the cell responses to infection. The present study was designed to isolate primary epithelial cells from human ectocervix explants and characterize their susceptibility to C. trachomatis infection. We achieved a high purity of isolation, assessed by the expression of E-cadherin and cytokeratin 14. The infectious progeny in these primary epithelial cells was lower than in HeLa cells. We showed that the difference in culture medium, and the addition of serum in HeLa cultures, accounted for a large part of these differences. However, all things considered the primary ectocervical epithelial cells remained less permissive than HeLa cells to C. trachomatis serovar L2 or D development. Finally, the basal level of transcription of genes coding for pro-inflammatory cytokines was globally higher in primary epithelial cells than in HeLa cells. Transcription of several pro-inflammatory genes was further induced by infection with C. trachomatis serovar L2 or serovar D. In conclusion, primary epithelial cells have a strong capacity to mount an inflammatory response to Chlamydia infection. Our simplified purification protocol from human explants should facilitate future studies to understand the contribution of this response to limiting the spread of the pathogen to the upper female genital tract.
Collapse
|
13
|
Schott BH, Antonia AL, Wang L, Pittman KJ, Sixt BS, Barnes AB, Valdivia RH, Ko DC. Modeling of variables in cellular infection reveals CXCL10 levels are regulated by human genetic variation and the Chlamydia-encoded CPAF protease. Sci Rep 2020; 10:18269. [PMID: 33106516 PMCID: PMC7588472 DOI: 10.1038/s41598-020-75129-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 10/12/2020] [Indexed: 01/01/2023] Open
Abstract
Susceptibility to infectious diseases is determined by a complex interaction between host and pathogen. For infections with the obligate intracellular bacterium Chlamydia trachomatis, variation in immune activation and disease presentation are regulated by both host genetic diversity and pathogen immune evasion. Previously, we discovered a single nucleotide polymorphism (rs2869462) associated with absolute abundance of CXCL10, a pro-inflammatory T-cell chemokine. Here, we report that levels of CXCL10 change during C. trachomatis infection of cultured cells in a manner dependent on both host and pathogen. Linear modeling of cellular traits associated with CXCL10 levels identified a strong, negative correlation with bacterial burden, suggesting that C. trachomatis actively suppresses CXCL10. We identified the pathogen-encoded factor responsible for this suppression as the chlamydial protease- or proteasome-like activity factor, CPAF. Further, we applied our modeling approach to other host cytokines in response to C. trachomatis and found evidence that RANTES, another T-cell chemoattractant, is actively suppressed by Chlamydia. However, this observed suppression of RANTES is not mediated by CPAF. Overall, our results demonstrate that CPAF suppresses CXCL10 to evade the host cytokine response and that modeling of cellular infection parameters can reveal previously unrecognized facets of host-pathogen interactions.
Collapse
Affiliation(s)
- Benjamin H Schott
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, 0049 CARL Building Box 3053, 213 Research Drive, Durham, NC, 27710, USA
- Duke University Program in Genetics and Genomics, Duke University, Durham, NC, 27710, USA
| | - Alejandro L Antonia
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, 0049 CARL Building Box 3053, 213 Research Drive, Durham, NC, 27710, USA
| | - Liuyang Wang
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, 0049 CARL Building Box 3053, 213 Research Drive, Durham, NC, 27710, USA
| | - Kelly J Pittman
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, 0049 CARL Building Box 3053, 213 Research Drive, Durham, NC, 27710, USA
| | - Barbara S Sixt
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, 0049 CARL Building Box 3053, 213 Research Drive, Durham, NC, 27710, USA
- Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå Centre for Microbial Research, Department of Molecular Biology, Umeå University, Umeå, Sweden
| | - Alyson B Barnes
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, 0049 CARL Building Box 3053, 213 Research Drive, Durham, NC, 27710, USA
| | - Raphael H Valdivia
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, 0049 CARL Building Box 3053, 213 Research Drive, Durham, NC, 27710, USA
| | - Dennis C Ko
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, 0049 CARL Building Box 3053, 213 Research Drive, Durham, NC, 27710, USA.
- Duke University Program in Genetics and Genomics, Duke University, Durham, NC, 27710, USA.
- Division of Infectious Diseases, Department of Medicine, School of Medicine, Duke University, Durham, NC, 27710, USA.
| |
Collapse
|
14
|
McQueen BE, Kiatthanapaiboon A, Fulcher ML, Lam M, Patton K, Powell E, Kollipara A, Madden V, Suchland RJ, Wyrick P, O'Connell CM, Reidel B, Kesimer M, Randell SH, Darville T, Nagarajan UM. Human Fallopian Tube Epithelial Cell Culture Model To Study Host Responses to Chlamydia trachomatis Infection. Infect Immun 2020; 88:e00105-20. [PMID: 32601108 PMCID: PMC7440757 DOI: 10.1128/iai.00105-20] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 06/23/2020] [Indexed: 12/20/2022] Open
Abstract
Chlamydia trachomatis infection of the human fallopian tubes can lead to damaging inflammation and scarring, ultimately resulting in infertility. To study the human cellular responses to chlamydial infection, researchers have frequently used transformed cell lines that can have limited translational relevance. We developed a primary human fallopian tube epithelial cell model based on a method previously established for culture of primary human bronchial epithelial cells. After protease digestion and physical dissociation of excised fallopian tubes, epithelial cell precursors were expanded in growth factor-containing medium. Expanded cells were cryopreserved to generate a biobank of cells from multiple donors and cultured at an air-liquid interface. Culture conditions stimulated cellular differentiation into polarized mucin-secreting and multiciliated cells, recapitulating the architecture of human fallopian tube epithelium. The polarized and differentiated cells were infected with a clinical isolate of C. trachomatis, and inclusions containing chlamydial developmental forms were visualized by fluorescence and electron microscopy. Apical secretions from infected cells contained increased amounts of proteins associated with chlamydial growth and replication, including transferrin receptor protein 1, the amino acid transporters SLC3A2 and SLC1A5, and the T-cell chemoattractants CXCL10, CXCL11, and RANTES. Flow cytometry revealed that chlamydial infection induced cell surface expression of T-cell homing and activation proteins, including ICAM-1, VCAM-1, HLA class I and II, and interferon gamma receptor. This human fallopian tube epithelial cell culture model is an important tool with translational potential for studying cellular responses to Chlamydia and other sexually transmitted pathogens.
Collapse
Affiliation(s)
- Bryan E McQueen
- Department of Pediatrics, University of North Carolina, Chapel Hill, North Carolina, USA
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Amy Kiatthanapaiboon
- Department of Pediatrics, University of North Carolina, Chapel Hill, North Carolina, USA
| | - M Leslie Fulcher
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Mariam Lam
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Kate Patton
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Emily Powell
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Avinash Kollipara
- Department of Pediatrics, University of North Carolina, Chapel Hill, North Carolina, USA
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Victoria Madden
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Robert J Suchland
- University of Washington, Division of Allergy and Infectious Diseases, Department of Medicine, Seattle, Washington, USA
| | - Priscilla Wyrick
- Department of Pediatrics, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Catherine M O'Connell
- Department of Pediatrics, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Boris Reidel
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Mehmet Kesimer
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Scott H Randell
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Toni Darville
- Department of Pediatrics, University of North Carolina, Chapel Hill, North Carolina, USA
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Uma M Nagarajan
- Department of Pediatrics, University of North Carolina, Chapel Hill, North Carolina, USA
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, North Carolina, USA
| |
Collapse
|
15
|
Li Y, Chen H, Li S, Li Y, Liu G, Bai J, Luo H, Lan X, He Z. LncSSBP1 Functions as a Negative Regulator of IL-6 Through Interaction With hnRNPK in Bronchial Epithelial Cells Infected With Talaromyces marneffei. Front Immunol 2020; 10:2977. [PMID: 31998294 PMCID: PMC6966331 DOI: 10.3389/fimmu.2019.02977] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Accepted: 12/04/2019] [Indexed: 12/12/2022] Open
Abstract
Talaromyces marneffei (TM) is an important opportunistic pathogenic fungus capable of causing disseminated lethal infection. In our previous study, we identified host lncRNAs and mRNAs that are dysregulated in TM-infected bronchial epithelial cells. In this report, we verified that IL-6, a key factor in acute inflammatory response, is down-regulated in TM pathogenesis. To elucidate the mechanism of IL-6 regulation, we analyzed the coding/non-coding network, and identified lncSSBP1, a novel lncRNA that is up-regulated by TM. Our results demonstrate that overexpression of lncSSBP1 decreases IL-6 mRNA expression, whereas knockdown of lncSSBP1 enhances IL-6 mRNA expression. Though lncSSBP1 is primarily localized to the nucleus, bioinformatics analysis suggests that it is unlikely to function as competing endogenous RNA or to interact with IL-6 transcription factors. Instead, RNA pull down and RNA immunoprecipitation assays showed that lncSSBP1 binds specifically to heterogenous nuclear ribonucleoprotein K (hnRNPK), which is involved in IL-6 mRNA processing. Our findings suggest that lncSSBP1 may affect IL-6 mRNA expression during TM infection through interaction with hnRNPk in bronchial epithelial cells. Our results suggest a novel pathway by which TM may suppress the immune response to its advantage.
Collapse
Affiliation(s)
- Yinghua Li
- Department of Pulmonary and Critical Care Medicine, Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Huan Chen
- Department of Pulmonary and Critical Care Medicine, Sixth Affiliated Hospital of Guangxi Medical University, Yulin, China
| | - Shuyi Li
- Guangxi Colleges and Universities Key Laboratory of Preclinical Medicine Research, Guangxi Medical University, Nanning, China
| | - Yu Li
- Department of Pulmonary and Critical Care Medicine, Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Guangnan Liu
- Department of Pulmonary and Critical Care Medicine, Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Jing Bai
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Honglin Luo
- School of Basic Medicine, Guangxi Medical University, Nanning, China
| | - Xiuwan Lan
- Guangxi Colleges and Universities Key Laboratory of Preclinical Medicine Research, Guangxi Medical University, Nanning, China
| | - Zhiyi He
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
16
|
Faris R, Andersen SE, McCullough A, Gourronc F, Klingelhutz AJ, Weber MM. Chlamydia trachomatis Serovars Drive Differential Production of Proinflammatory Cytokines and Chemokines Depending on the Type of Cell Infected. Front Cell Infect Microbiol 2019; 9:399. [PMID: 32039039 PMCID: PMC6988789 DOI: 10.3389/fcimb.2019.00399] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 11/07/2019] [Indexed: 12/22/2022] Open
Abstract
Chlamydia trachomatis serovars A-C infect conjunctival epithelial cells and untreated infection can lead to blindness. D-K infect genital tract epithelial cells resulting in pelvic inflammatory disease, ectopic pregnancy, and sterility while L1-L3 infect epithelial cells and macrophages, causing an invasive infection. Despite some strains of Chlamydia sharing high nucleotide sequence similarity, the bacterial and host factors that govern tissue and cellular tropism remain largely unknown. Following introduction of C. trachomatis via intercourse, epithelial cells of the vagina, foreskin, and ectocervix are exposed to large numbers of the pathogen, yet their response to infection and the dynamics of chlamydial growth in these cells has not been well-characterized compared to growth in more permissive cell types that harbor C. trachomatis. We compared intracellular replication and inclusion development of representative C. trachomatis serovars in immortalized human conjunctival epithelial, urogenital epithelial, PMA stimulated THP-1 (macrophages), and HeLa cells. We demonstrate that urogenital epithelial cells of the vagina, ectocervix, and foreskin restrict replication of serovar A while promoting robust replication and inclusion development of serovar D and L2. Macrophages restrict serovars D and A while L2 proliferates in these cells. Furthermore, we show that GM-CSF, RANTES, GROα, IL-1α, IL-1β, IP-10, IL-8, and IL-18 are produced in a cell-type and serovar-specific manner. Collectively we have established a series of human cell lines that represent some of the first cell types to encounter C. trachomatis following exposure and show that differential production of key cytokines early during infection could regulate serovar-host cell specificity.
Collapse
Affiliation(s)
- Robert Faris
- Department of Microbiology and Immunology, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Shelby E Andersen
- Department of Microbiology and Immunology, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Alix McCullough
- Department of Microbiology and Immunology, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Françoise Gourronc
- Department of Microbiology and Immunology, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Aloysius J Klingelhutz
- Department of Microbiology and Immunology, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Mary M Weber
- Department of Microbiology and Immunology, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| |
Collapse
|
17
|
Flanagan TW, Sebastian MN, Battaglia DM, Foster TP, Cormier SA, Nichols CD. 5-HT 2 receptor activation alleviates airway inflammation and structural remodeling in a chronic mouse asthma model. Life Sci 2019; 236:116790. [PMID: 31626791 DOI: 10.1016/j.lfs.2019.116790] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 08/09/2019] [Accepted: 08/24/2019] [Indexed: 12/20/2022]
Abstract
AIMS Although the bulk of research into the biology of serotonin 5-HT2A receptors has focused on its role in the CNS, selective activation of these receptors in peripheral tissues can produce profound anti-inflammatory effects. We previously demonstrated that the small molecule 5-HT2 receptor agonist (R)-2,5-dimethoxy-4-iodoamphetamine [(R)-DOI] inhibits TNF-α-mediated proinflammatory signaling cascades and inflammation via 5-HT2A receptor activation and prevents the development of, and inflammation associated with, acute allergic asthma in a mouse ovalbumin (OVA) model. Here, we investigated the ability of (R)-DOI to reverse inflammation and symptoms associated with established asthma in a newly developed model of chronic asthma. METHODS An 18-week ovalbumin challenge period was performed to generate persistent, chronic asthma in BALB/c mice. Four once daily intranasal treatments of (R)-DOI were administered one week after allergen cessation, with respiratory parameters being measured by whole-body plethysmography (WBP). Cytokine and chemokine levels were measured by quantitative real-time polymerase chain reaction (qRT-PCR) in homogenized lung tissue, bronchoalveolar (BALF) fluid was analyzed for chemokine modulation by multiplex assays, and Periodic Acid-Schiff and Masson's Trichrome staining was performed to determine goblet cell infiltration and overall changes to lung morphology. KEY FINDINGS 5-HT2 activation via (R)-DOI attenuates elevated airway hyperresponsiveness to methacholine, reduces pulmonary inflammation and mucus production, and reduces airway structural remodeling and collagen deposition by nearly 70%. SIGNIFICANCE Overall, these data provide support for the therapeutic potential of (R)-DOI and 5-HT2 receptor activation for the treatment of asthma, and identifies (R)-DOI as a novel therapeutic compound against pulmonary fibrosis.
Collapse
Affiliation(s)
- Thomas W Flanagan
- Department of Pharmacology and Experimental Therapeutics, Louisiana State University Health Sciences Center, New Orleans, LA, 70112, USA
| | - Melaine N Sebastian
- Department of Pharmacology and Experimental Therapeutics, Louisiana State University Health Sciences Center, New Orleans, LA, 70112, USA
| | - Diana M Battaglia
- Department of Microbiology, Immunology, And Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA, 70112, USA
| | - Timothy P Foster
- Department of Microbiology, Immunology, And Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA, 70112, USA
| | - Stephania A Cormier
- Department of Biological Sciences, Louisiana State University, 202 Life Sciences Building, Baton Rouge, LA, 70803, USA
| | - Charles D Nichols
- Department of Pharmacology and Experimental Therapeutics, Louisiana State University Health Sciences Center, New Orleans, LA, 70112, USA.
| |
Collapse
|
18
|
Mwatelah R, McKinnon LR, Baxter C, Abdool Karim Q, Abdool Karim SS. Mechanisms of sexually transmitted infection-induced inflammation in women: implications for HIV risk. J Int AIDS Soc 2019; 22 Suppl 6:e25346. [PMID: 31468677 PMCID: PMC6715949 DOI: 10.1002/jia2.25346] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Accepted: 06/20/2019] [Indexed: 12/16/2022] Open
Abstract
INTRODUCTION Globally, sexually transmitted infections (STI) affect >300 million people annually, and are a major cause of sexual and reproductive health complications in women. In this commentary, we describe how STIs interact with the immune and non-immune cells, both within and below the cervicovaginal mucosal barrier, to cause inflammation, which in turn has been associated with increased HIV acquisition risk. DISCUSSION STIs have a major impact on the female genital mucosa, which is an important biological and physical barrier that forms the first line of defence against invading microorganisms such as HIV. Pattern recognition of STI pathogens, by receptors expressed either on the cell surface or inside the cell, typically triggers inflammation at the mucosal barrier. The types of mucosal responses vary by STI, and can be asymptomatic or culminate in the formation of discharge, ulcers and/or warts. While the aim of this response is to clear the invading microbes, in many cases these responses are either evaded or cause pathology that impairs barrier integrity and increases HIV access to target cells in the sub-mucosa. In addition, innate responses to STIs can result in an increased number of immune cells, including those that are the primary targets of HIV, and may contribute to the association between STIs and increased susceptibility to HIV acquisition. Many of these cells are mediators of adaptive immunity, including tissue-resident cells that may also display innate-like functions. Bacterial vaginosis (BV) is another common cause of inflammation, and evidence for multiple interactions between BV, STIs and HIV suggest that susceptibility to these conditions should be considered in concert. CONCLUSIONS STIs and other microbes can induce inflammation in the genital tract, perturbing the normal robust function of the mucosal barrier against HIV. While the impact of STIs on the mucosal immune system and HIV acquisition is often under-appreciated, understanding their interactions of the infections with the immune responses play an important role in improving treatment and reducing the risk of HIV acquisition. The frequent sub-clinical inflammation associated with STIs underscores the need for better STI diagnostics to reverse the immunological consequences of infection.
Collapse
Affiliation(s)
- Ruth Mwatelah
- Department of Medical Microbiology and Infectious DiseasesUniversity of ManitobaWinnipegCanada
| | - Lyle R McKinnon
- Department of Medical Microbiology and Infectious DiseasesUniversity of ManitobaWinnipegCanada
- Centre for the AIDS Programme of Research in South Africa (CAPRISA)University of KwaZulu‐NatalDurbanSouth Africa
| | - Cheryl Baxter
- Centre for the AIDS Programme of Research in South Africa (CAPRISA)University of KwaZulu‐NatalDurbanSouth Africa
| | - Quarraisha Abdool Karim
- Centre for the AIDS Programme of Research in South Africa (CAPRISA)University of KwaZulu‐NatalDurbanSouth Africa
- Department of EpidemiologyColumbia UniversityNew YorkNYUSA
| | - Salim S Abdool Karim
- Centre for the AIDS Programme of Research in South Africa (CAPRISA)University of KwaZulu‐NatalDurbanSouth Africa
- Department of EpidemiologyColumbia UniversityNew YorkNYUSA
| |
Collapse
|
19
|
Dolat L, Valdivia RH. A renewed tool kit to explore Chlamydia pathogenesis: from molecular genetics to new infection models. F1000Res 2019; 8. [PMID: 31249676 PMCID: PMC6589931 DOI: 10.12688/f1000research.18832.1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/12/2019] [Indexed: 12/21/2022] Open
Abstract
Chlamydia trachomatis is the most prevalent sexually transmitted bacterial pathogen and the leading cause of preventable blindness in the developing world.
C. trachomatis invades the epithelium of the conjunctiva and genital tract and replicates within an intracellular membrane-bound compartment termed the inclusion. To invade and replicate in mammalian cells,
Chlamydia remodels epithelial surfaces by reorganizing the cytoskeleton and cell–cell adhesions, reprograms membrane trafficking, and modulates cell signaling to dampen innate immune responses. If the infection ascends to the upper female genital tract, it can result in pelvic inflammatory disease and tissue scarring.
C. trachomatis infections are associated with infertility, ectopic pregnancies, the fibrotic disorder endometriosis, and potentially cancers of the cervix and uterus. Unfortunately, the molecular mechanisms by which this clinically important human pathogen subverts host cellular functions and causes disease have remained relatively poorly understood because of the dearth of molecular genetic tools to study
Chlamydiae and limitations of both
in vivo and
in vitro infection models. In this review, we discuss recent advances in the experimental molecular tool kit available to dissect
C. trachomatis infections with a special focus on
Chlamydia-induced epithelial barrier disruption by regulating the structure, function, and dynamics of epithelial cell–cell junctions.
Collapse
Affiliation(s)
- Lee Dolat
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, USA
| | - Raphael H Valdivia
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, USA
| |
Collapse
|
20
|
Ziklo N, Huston WM, Taing K, Timms P. High expression of IDO1 and TGF-β1 during recurrence and post infection clearance with Chlamydia trachomatis, are independent of host IFN-γ response. BMC Infect Dis 2019; 19:218. [PMID: 30832593 PMCID: PMC6398247 DOI: 10.1186/s12879-019-3843-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 02/21/2019] [Indexed: 11/11/2022] Open
Abstract
Background Chlamydia trachomatis infections in women continue to be a major public health concern due to their high prevalence and consequent reproductive morbidities. While antibiotics are usually efficient to clear the Chlamydia, repeat infections are common and may contribute to pathological outcomes. Interferon-gamma (IFN-γ)-mediated immunity has been suggested to be protective against reinfection, and represent an important anti-chlamydial agent, primarily via the induction of indoleamine-2,3 dioxygenase 1 (IDO1) enzyme. IDO1 catalyzes the degradation of tryptophan, which can eliminate C. trachomatis infection in vitro. Here, we sought to measure IDO1 expression levels and related immune markers during different C. trachomatis infection statuses (repeated vs single infection vs post antibiotic treatment), in vitro and in vivo. Methods In this study, we measured the expression levels of IDO1 and immune regulatory markers, transforming growth factor β1 (TGF-β1) and forkhead box P3 (FoxP3), in vaginal swab samples of C. trachomatis-infected women, with either single or repeated infection. In addition, we used an in vitro co-culture model of endometrial carcinoma cell-line and peripheral blood mononuclear cells (PBMCs) to measure the same immune markers. Results We found that in women with repeated C. trachomatis infections vaginal IDO1 and TGF-β1 expression levels were significantly increased. Whereas, women who cleared their infection post antibiotic treatment, had increased levels of IDO1 and TGF-β1, as well as FoxP3. Similarly, using the in vitro model, we found significant upregulation of IDO1 and TGF-β1 levels in the co-culture infected with C. trachomatis. Furthermore, we found that in PBMCs infected with C. trachomatis there was a significant upregulation in IDO1 levels, which was independent of IFN-γ. In fact, C. trachomatis infection in PBMCs failed to induce IFN-γ levels in comparison to the uninfected culture. Conclusions Our data provide evidence for a regulatory immune response comprised of IDO1, TGF-β1 and FoxP3 in women post antibiotic treatment. In this study, we demonstrated a significant increase in IDO1 expression levels in response to C. trachomatis infection, both in vivo and in vitro, without elevated IFN-γ levels. This study implicates IDO1 and TGF-β1 as part of the immune response to repeated C. trachomatis infections, independently of IFN-γ. Electronic supplementary material The online version of this article (10.1186/s12879-019-3843-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Noa Ziklo
- Faculty of Science, Health, Education & Engineering, University of the Sunshine Coast, Sippy Downs, Sunshine Coast, QLD, Australia.
| | - Wilhelmina M Huston
- School of Life Sciences, Faculty of Science, University of Technology, Sydney, Australia
| | - Kuong Taing
- Sunshine Coast Sexual Health and HIV Service (Clinic 87), Nambour, Sunshine Coast, QLD, Australia
| | - Peter Timms
- Faculty of Science, Health, Education & Engineering, University of the Sunshine Coast, Sippy Downs, Sunshine Coast, QLD, Australia
| |
Collapse
|
21
|
Kuratli J, Pesch T, Marti H, Leonard CA, Blenn C, Torgerson P, Borel N. Water Filtered Infrared A and Visible Light (wIRA/VIS) Irradiation Reduces Chlamydia trachomatis Infectivity Independent of Targeted Cytokine Inhibition. Front Microbiol 2018; 9:2757. [PMID: 30524392 PMCID: PMC6262300 DOI: 10.3389/fmicb.2018.02757] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Accepted: 10/29/2018] [Indexed: 11/13/2022] Open
Abstract
Chlamydia trachomatis is the major cause of infectious blindness and represents the most common bacterial sexually transmitted infection worldwide. Considering the potential side effects of antibiotic therapy and increasing threat of antibiotic resistance, alternative therapeutic strategies are needed. Previous studies showed that water filtered infrared A alone (wIRA) or in combination with visible light (wIRA/VIS) reduced C. trachomatis infectivity. Furthermore, wIRA/VIS irradiation led to secretion of pro-inflammatory cytokines similar to that observed upon C. trachomatis infection. We confirmed the results of previous studies, namely that cytokine secretion (IL-6, IL-8, and RANTES/CCL5) upon wIRA/VIS treatment, and the subsequent reduction of chlamydial infectivity, are independent of the addition of cycloheximide, a host protein synthesis inhibitor. Reproducible cytokine release upon irradiation indicated that cytokines might be involved in the anti-chlamydial mechanism of wIRA/VIS. This hypothesis was tested by inhibiting IL-6, IL-8, and RANTES secretion in C. trachomatis or mock-infected cells by gene silencing or pharmaceutical inhibition. Celastrol, a substance derived from Trypterygium wilfordii, used in traditional Chinese medicine and known for anti-cancer and anti-inflammatory effects, was used for IL-6 and IL-8 inhibition, while Maraviroc, a competitive CCR5 antagonist and anti-HIV drug, served as a RANTES/CCL5 inhibitor. HeLa cell cytotoxicity and impact on chlamydial morphology, size and inclusion number was evaluated upon increasing inhibitor concentration, and concentrations of 0.1 and 1 μM Celastrol and 10 and 20 μM Maraviroc were subsequently selected for irradiation experiments. Celastrol at any concentration reduced chlamydial infectivity, an effect only observed for 20 μM Maraviroc. Triple dose irradiation (24, 36, 40 hpi) significantly reduced chlamydial infectivity regardless of IL-6, IL-8, or RANTES/CCL5 gene silencing, Celastrol or Maraviroc treatment. Neither gene silencing nor pharmaceutical cytokine inhibition provoked the chlamydial stress response. The anti-chlamydial effect of wIRA/VIS is independent of cytokine inhibition under all conditions evaluated. Thus, factors other than host cell cytokines must be involved in the working mechanism of wIRA/VIS. This study gives a first insight into the working mechanism of wIRA/VIS in relation to an integral component of the host immune system and supports the potential of wIRA/VIS as a promising new tool for treatment in trachoma.
Collapse
Affiliation(s)
- Jasmin Kuratli
- Institute of Veterinary Pathology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Theresa Pesch
- Institute of Veterinary Pathology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Hanna Marti
- Institute of Veterinary Pathology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Cory Ann Leonard
- Institute of Veterinary Pathology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Christian Blenn
- Institute of Veterinary Pathology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Paul Torgerson
- Section of Veterinary Epidemiology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Nicole Borel
- Institute of Veterinary Pathology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| |
Collapse
|
22
|
Embryotoxic cytokines—Potential roles in embryo loss and fetal programming. J Reprod Immunol 2018; 125:80-88. [DOI: 10.1016/j.jri.2017.12.003] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 12/13/2017] [Accepted: 12/21/2017] [Indexed: 12/22/2022]
|
23
|
The Effector TepP Mediates Recruitment and Activation of Phosphoinositide 3-Kinase on Early Chlamydia trachomatis Vacuoles. mSphere 2017; 2:mSphere00207-17. [PMID: 28744480 PMCID: PMC5518268 DOI: 10.1128/msphere.00207-17] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Accepted: 06/27/2017] [Indexed: 11/20/2022] Open
Abstract
Chlamydia trachomatis delivers multiple type 3 secreted effector proteins to host epithelial cells to manipulate cytoskeletal functions, membrane dynamics, and signaling pathways. TepP is the most abundant effector protein secreted early in infection, but its molecular function is poorly understood. In this report, we provide evidence that TepP is important for bacterial replication in cervical epithelial cells, activation of type I IFN genes, and recruitment of class I phosphoinositide 3-kinases (PI3K) and signaling adaptor protein CrkL to nascent pathogen-containing vacuoles (inclusions). We also show that TepP is a target of tyrosine phosphorylation by Src kinases but that these modifications do not appear to influence the recruitment of PI3K or CrkL. The translocation of TepP correlated with an increase in the intracellular pools of phosphoinositide-(3,4,5)-triphosphate but not the activation of the prosurvival kinase Akt, suggesting that TepP-mediated activation of PI3K is spatially restricted to early inclusions. Furthermore, we linked PI3K activity to the dampening of transcription of type I interferon (IFN)-induced genes early in infection. Overall, these findings indicate that TepP can modulate cell signaling and, potentially, membrane trafficking events by spatially restricted activation of PI3K. IMPORTANCE This article shows that Chlamydia recruits PI3K, an enzyme important for host cell survival and internal membrane functions, to the pathogens inside cells by secreting a scaffolding protein called TepP. TepP enhances Chlamydia replication and dampens the activation of immune responses.
Collapse
|
24
|
Leonard CA, Schoborg RV, Borel N. Productive and Penicillin-Stressed Chlamydia pecorum Infection Induces Nuclear Factor Kappa B Activation and Interleukin-6 Secretion In Vitro. Front Cell Infect Microbiol 2017; 7:180. [PMID: 28553623 PMCID: PMC5425588 DOI: 10.3389/fcimb.2017.00180] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 04/25/2017] [Indexed: 11/13/2022] Open
Abstract
Nuclear factor kappa B (NFκB) is an inflammatory transcription factor that plays an important role in the host immune response to infection. The potential for chlamydiae to activate NFκB has been an area of interest, however most work has focused on chlamydiae impacting human health. Given that inflammation characteristic of chlamydial infection may be associated with severe disease outcomes or contribute to poor overall fitness in farmed animals, we evaluated the ability of porcine chlamydiae to induce NFκB activation in vitro. C. pecorum infection induced both NFκB nuclear translocation and activation at 2 hours post infection (hpi), an effect strongly enhanced by suppression of host de novo protein synthesis. C. suis and C. trachomatis showed less capacity for NFκB activation compared to C. pecorum, suggesting a species-specific variation in NFκB activation. At 24 hpi, C. pecorum induced significant NFκB activation, an effect not abolished by penicillin (beta lactam)-induced chlamydial stress. C. pecorum-dependent secretion of interleukin 6 was also detected in the culture supernatant of infected cells at 24 hpi, and this effect, too, was unchanged by penicillin-induced chlamydial stress. Taken together, these results suggest that NFκB participates in the early inflammatory response to C. pecorum and that stressed chlamydiae can promote inflammation.
Collapse
Affiliation(s)
- Cory A Leonard
- Department of Pathobiology, Institute of Veterinary Pathology, University of ZurichZurich, Switzerland
| | - Robert V Schoborg
- Department of Biomedical Sciences, Center for Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State UniversityJohnson City, TN, USA
| | - Nicole Borel
- Department of Pathobiology, Institute of Veterinary Pathology, University of ZurichZurich, Switzerland
| |
Collapse
|
25
|
Refaat B, Ashshi AM, Batwa SA, Ahmad J, Idris S, Kutbi SY, Malibary FA, Kamfar FF. The prevalence of Chlamydia trachomatis and Mycoplasma genitalium tubal infections and their effects on the expression of IL-6 and leukaemia inhibitory factor in Fallopian tubes with and without an ectopic pregnancy. Innate Immun 2016; 22:534-545. [PMID: 27511901 DOI: 10.1177/1753425916662326] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Accepted: 07/12/2016] [Indexed: 11/16/2022] Open
Abstract
This was a prospective case-control study that measured the prevalence of Chlamydia trachomatis (CT), Neisseria gonorrhoeae (NG) and Mycoplasma genitalium (MG) by an IVD CE multiplex PCR kit in fresh Fallopian tubes (FT) obtained from 96 ectopic pregnancies (EP) and 61 controls in the midluteal phase of the cycle. We later measured the expression profile of IL-6, leukaemia inhibitory factor (LIF) and their signalling molecules, in respect to the type and number of infections, by immunohistochemistry, ELISA and quantitative RT-PCR. The frequencies of CT, and MG mono- and co-infections were significantly higher in EP. IL-6, LIF, their receptors and intracellular mediators were significantly up-regulated at the gene and protein levels in positive compared with negative FTs within each group (P < 0.05). EP tubal samples with co-infections showed the highest significant expression of the candidate cytokines by all techniques (P < 0.05). CT and MG are frequent in EP and up-regulate the tubal expression of IL-6, LIF and their signalling molecules. Both cytokines could be involved in the tubal immune response against bacterial infections, as well as the pathogenesis of EP. Further studies are needed to explore the roles of IL-6 family in infection-induced tubal inflammation and EP.
Collapse
Affiliation(s)
- Bassem Refaat
- Laboratory Medicine Department, Faculty of Applied Medical Sciences, Umm Al-Qura University, Al Abdeyah, Makkah, KSA
| | - Ahmed Mohamed Ashshi
- Laboratory Medicine Department, Faculty of Applied Medical Sciences, Umm Al-Qura University, Al Abdeyah, Makkah, KSA
| | - Sarah Abdullah Batwa
- Obstertics and Gynaecology Department, Maternity and Children Hospital, Al-Aziziyah, Ministry of Health, Jeddah, KSA
| | - Jawwad Ahmad
- Laboratory Medicine Department, Faculty of Applied Medical Sciences, Umm Al-Qura University, Al Abdeyah, Makkah, KSA
| | - Shakir Idris
- Laboratory Medicine Department, Faculty of Applied Medical Sciences, Umm Al-Qura University, Al Abdeyah, Makkah, KSA
| | - Seham Yahia Kutbi
- Obstertics and Gynaecology Department, Maternity and Children Hospital, Al-Aziziyah, Ministry of Health, Jeddah, KSA
| | - Faizah Ahmed Malibary
- Obstertics and Gynaecology Department, Maternity and Children Hospital, Al-Aziziyah, Ministry of Health, Jeddah, KSA
| | - Fadi Fayez Kamfar
- Pathology Department, Clinical Laboratories, Maternity and Children Hospital, Ministry of Health, Makkah, KSA
| |
Collapse
|
26
|
Derrick T, Last AR, Burr SE, Roberts CH, Nabicassa M, Cassama E, Bailey RL, Mabey DCW, Burton MJ, Holland MJ. Inverse relationship between microRNA-155 and -184 expression with increasing conjunctival inflammation during ocular Chlamydia trachomatis infection. BMC Infect Dis 2016; 16:60. [PMID: 26842862 PMCID: PMC4739388 DOI: 10.1186/s12879-016-1367-8] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Accepted: 01/22/2016] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Trachoma, a preventable blinding eye disease, is initiated by ocular infection with Chlamydia trachomatis (Ct). We previously showed that microRNAs (miR) -147b and miR-1285 were up-regulated in inflammatory trachomatous scarring. During the initial stage of disease, follicular trachoma with current Ct infection, the differential expression of miR has not yet been investigated. METHODS Conjunctival samples were collected from 163 children aged 1-9 years old living in a trachoma-endemic region of Guinea Bissau, West Africa. Small RNA sequencing (RNAseq) was carried out on samples from five children with follicular trachoma and current Ct infection and five children with healthy conjunctivae and no Ct infection. Small RNAseq was also carried out on human epithelial cell lines infected with ocular Ct strains A2497 and isogenic plasmid-free A2497 in vitro. Results were validated by quantitative PCR (qPCR) in 163 clinical samples. RESULTS Differential expression of RNAseq data identified 12 miR with changes in relative expression during follicular trachoma, of which 9 were confirmed as differentially expressed by qPCR (miR-155, miR-150, miR-142, miR-181b, miR-181a, miR-342, miR-132, miR-4728 and miR-184). MiR-155 and miR-184 expression had a direct relationship with the degree of clinical inflammation. MiR-155 was up-regulated (OR = 2.533 ((95 % CI = 1.291-4.971); P = 0.0069) and miR-184 was down-regulated (OR = 0.416 ((95 % CI = 0.300-0.578); P = 1.61*10(-7)) as the severity of clinical inflammation increased. Differential miR expression was not detected in HEp-2 or HCjE epithelial cells 48 h post infection with Ct in vitro. HCjE cells, a conjunctival epithelial cell line, had a markedly different miR background expression compared to HEp-2 cells. CONCLUSIONS In follicular trachoma, expression of miR-155 and miR-184 is correlated with the severity of inflammation. This likely reflects host regulation of the immune response and a prolonged period of wound healing following the clearance of Ct. Prolonged healing may be associated with subsequent development of scarring trachoma.
Collapse
Affiliation(s)
- Tamsyn Derrick
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom.
| | - Anna R Last
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom.
| | - Sarah E Burr
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom.
- Disease Control and Elimination Theme, Medical Research Council Unit The Gambia, Fajara, The Gambia.
| | - Chrissy H Roberts
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom.
| | - Meno Nabicassa
- Programa Nacional de Saúde de Visão, Ministério de Saúde Publica, Bissau, Guinea Bissau.
| | - Eunice Cassama
- Programa Nacional de Saúde de Visão, Ministério de Saúde Publica, Bissau, Guinea Bissau.
| | - Robin L Bailey
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom.
| | - David C W Mabey
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom.
| | - Matthew J Burton
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom.
| | - Martin J Holland
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom.
| |
Collapse
|
27
|
Buckner LR, Amedee AM, Albritton HL, Kozlowski PA, Lacour N, McGowin CL, Schust DJ, Quayle AJ. Chlamydia trachomatis Infection of Endocervical Epithelial Cells Enhances Early HIV Transmission Events. PLoS One 2016; 11:e0146663. [PMID: 26730599 PMCID: PMC4701475 DOI: 10.1371/journal.pone.0146663] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Accepted: 12/21/2015] [Indexed: 01/20/2023] Open
Abstract
Chlamydia trachomatis causes a predominantly asymptomatic, but generally inflammatory, genital infection that is associated with an increased risk for HIV acquisition. Endocervical epithelial cells provide the major niche for this obligate intracellular bacterium in women, and the endocervix is also a tissue in which HIV transmission can occur. The mechanism by which CT infection enhances HIV susceptibility at this site, however, is not well understood. Utilizing the A2EN immortalized endocervical epithelial cell line grown on cell culture inserts, we evaluated the direct role that CT-infected epithelial cells play in facilitating HIV transmission events. We determined that CT infection significantly enhanced the apical-to-basolateral migration of cell-associated, but not cell-free, HIVBaL, a CCR5-tropic strain of virus, across the endocervical epithelial barrier. We also established that basolateral supernatants from CT-infected A2EN cells significantly enhanced HIV replication in peripheral mononuclear cells and a CCR5+ T cell line. These results suggest that CT infection of endocervical epithelial cells could facilitate both HIV crossing the mucosal barrier and subsequent infection or replication in underlying target cells. Our studies provide a mechanism by which this common STI could potentially promote the establishment of founder virus populations and the maintenance of local HIV reservoirs in the endocervix. Development of an HIV/STI co-infection model also provides a tool to further explore the role of other sexually transmitted infections in enhancing HIV acquisition.
Collapse
Affiliation(s)
- Lyndsey R. Buckner
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, Louisiana 70112, United States of America
| | - Angela M. Amedee
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, Louisiana 70112, United States of America
| | - Hannah L. Albritton
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, Louisiana 70112, United States of America
| | - Pamela A. Kozlowski
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, Louisiana 70112, United States of America
| | - Nedra Lacour
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, Louisiana 70112, United States of America
| | - Chris L. McGowin
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, Louisiana 70112, United States of America
- Department of Medicine, Section of Infectious Diseases, Louisiana State University Health Sciences Center, New Orleans, Louisiana, 70112, United States of America
| | - Danny J. Schust
- Department of Obstetrics, Gynecology and Women’s Health, University of Missouri, Columbia, MO 65201, United States of America
| | - Alison J. Quayle
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, Louisiana 70112, United States of America
| |
Collapse
|
28
|
Menon S, Alexander K, Timms P, Allan JA, Huston WM. CXCL10, CXCL11, HLA-A and IL-1β are induced in peripheral blood mononuclear cells from women with Chlamydia trachomatis related infertility. Pathog Dis 2015; 74:ftv099. [PMID: 26512034 DOI: 10.1093/femspd/ftv099] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/18/2015] [Indexed: 12/16/2022] Open
Abstract
Chlamydia trachomatis infections can result in the development of serious sequelae such as pelvic inflammatory disease and tubal infertility. In this study, peripheral blood mononuclear cells from women who were undergoing or had recently undergone IVF treatment were cultured ex vivo with C. trachomatis to identify the immune responses associated with women who had serological evidence of a history of Chlamydia infection. Cytokines secreted into the supernatant from the cultures were measured using ELISA, and the level of IL-1β was found to be significantly higher in Chlamydia positive women than Chlamydia negative women. qRT-PCR analysis of the expression of 88 immune-related genes showed trends towards an upregulation of CXCL10, CXCL11 and HLA-A in Chlamydia positive women compared with Chlamydia negative women. These findings support that some women launch a more marked proinflammatory response upon infection with C. trachomatis and this may be associated with why C. trachomatis induces infertility in some infected women.
Collapse
Affiliation(s)
- Shruti Menon
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD 4059, Australia
| | - Kimberly Alexander
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD 4059, Australia
| | - Peter Timms
- Faculty of Science, Health, Education and Engineering, University of the Sunshine Coast, QLD 4558, Australia
| | - John A Allan
- Wesley and St Andrews Research Institute, The Wesley Hospital, Auchenflower, QLD 4066, Australia UC Health Clinical School, The Wesley Hospital, Auchenflower, QLD 4066, Australia
| | - Wilhelmina M Huston
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD 4059, Australia Wesley and St Andrews Research Institute, The Wesley Hospital, Auchenflower, QLD 4066, Australia
| |
Collapse
|
29
|
Derrick T, Roberts CH, Last AR, Burr SE, Holland MJ. Trachoma and Ocular Chlamydial Infection in the Era of Genomics. Mediators Inflamm 2015; 2015:791847. [PMID: 26424969 PMCID: PMC4573990 DOI: 10.1155/2015/791847] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Accepted: 08/05/2015] [Indexed: 12/19/2022] Open
Abstract
Trachoma is a blinding disease usually caused by infection with Chlamydia trachomatis (Ct) serovars A, B, and C in the upper tarsal conjunctiva. Individuals in endemic regions are repeatedly infected with Ct throughout childhood. A proportion of individuals experience prolonged or severe inflammatory episodes that are known to be significant risk factors for ocular scarring in later life. Continued scarring often leads to trichiasis and in-turning of the eyelashes, which causes pain and can eventually cause blindness. The mechanisms driving the chronic immunopathology in the conjunctiva, which largely progresses in the absence of detectable Ct infection in adults, are likely to be multifactorial. Socioeconomic status, education, and behavior have been identified as contributing to the risk of scarring and inflammation. We focus on the contribution of host and pathogen genetic variation, bacterial ecology of the conjunctiva, and host epigenetic imprinting including small RNA regulation by both host and pathogen in the development of ocular pathology. Each of these factors or processes contributes to pathogenic outcomes in other inflammatory diseases and we outline their potential role in trachoma.
Collapse
Affiliation(s)
- Tamsyn Derrick
- Department of Clinical Research, Faculty of Infectious Tropical Diseases, London School of Hygiene and Tropical Medicine, London WC1E 7HT, UK
| | - Chrissy h. Roberts
- Department of Clinical Research, Faculty of Infectious Tropical Diseases, London School of Hygiene and Tropical Medicine, London WC1E 7HT, UK
| | - Anna R. Last
- Department of Clinical Research, Faculty of Infectious Tropical Diseases, London School of Hygiene and Tropical Medicine, London WC1E 7HT, UK
| | - Sarah E. Burr
- Department of Clinical Research, Faculty of Infectious Tropical Diseases, London School of Hygiene and Tropical Medicine, London WC1E 7HT, UK
| | - Martin J. Holland
- Department of Clinical Research, Faculty of Infectious Tropical Diseases, London School of Hygiene and Tropical Medicine, London WC1E 7HT, UK
| |
Collapse
|
30
|
Research Progress on Chlamydia trachomatis Infection and Related Cytokines. INFECTION INTERNATIONAL 2015. [DOI: 10.1515/ii-2017-0109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
AbstractChlamydia trachomatis(Ct) infection can induce host cells to produce numerous cytokines. Cytokines play important roles in inflammatory response. Although inflammation can protect the body, persistent inflammation can lead to pathological changes and tissue damages. Further research should determine whether cytokine production directly affects development and outcomes of inflammation. This study summarizes Ct infection and related cytokines.
Collapse
|
31
|
Lekovich J, Witkin SS, Doulaveris G, Orfanelli T, Shulman B, Pereira N, Rosenwaks Z, Spandorfer SD. Elevated serum interleukin-1β levels and interleukin-1β-to-interleukin-1 receptor antagonist ratio 1 week after embryo transfer are associated with ectopic pregnancy. Fertil Steril 2015; 104:1190-4. [PMID: 26279136 DOI: 10.1016/j.fertnstert.2015.07.1145] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Revised: 07/02/2015] [Accepted: 07/08/2015] [Indexed: 11/16/2022]
Abstract
OBJECTIVE To investigate whether interleukin-1β (IL-1β) and interleukin-1 receptor antagonist (IL-1RA) serum levels in the early luteal phase differ in IVF cycles that result in an ectopic pregnancy (EP) when compared with other outcomes. DESIGN Retrospective cohort. SETTING Not applicable. PATIENT(S) A total of 307 women whose serum samples were available, with the following IVF outcomes: 103 live births, 80 negative pregnancy tests, 52 biochemical pregnancies, 47 EPs, and 25 miscarriages. INTERVENTION(S) Serum samples were obtained on cycle days 24 and 28 (cycle day 14 = day of egg retrieval). Levels of IL-1β and IL-1RA were determined by quantitative ELISA performed by blinded personnel. MAIN OUTCOME MEASURE(S) IL-1β and IL-1RA levels, IL-1β-to-IL-1RA ratio versus cycle outcome. RESULT(S) The IL-1β levels were predictive of an EP. At cycle days 24 and 28 the mean IL-1β levels were higher in patients with an EP (127.1 pg/mL and 166.9 pg/mL, respectively) than in women with any other IVF outcome (15.8-55.3 pg/mL and 14.8-75.5 pg/mL, respectively). At cycle day 24 the IL-1β-to-IL-1RA ratio was 0.18 in the ectopic group versus 0.01-0.09 in the other groups. CONCLUSION(S) Elevated IL-1β levels and IL-1β-to-IL-1RA ratio as early as 4 days before the first pregnancy test are associated with an EP. If confirmed by prospective studies, clinical application of these findings could potentially improve EP detection.
Collapse
Affiliation(s)
- Jovana Lekovich
- The Ronald O. Perelman and Claudia Cohen Center for Reproductive Medicine, Weill Cornell Medical College, New York, New York.
| | - Steven S Witkin
- Division of Immunology and Infectious Diseases, Department of Obstetrics and Gynecology, Weill Cornell Medical College, New York, New York
| | - Georgios Doulaveris
- Division of Immunology and Infectious Diseases, Department of Obstetrics and Gynecology, Weill Cornell Medical College, New York, New York
| | - Theofano Orfanelli
- Division of Immunology and Infectious Diseases, Department of Obstetrics and Gynecology, Weill Cornell Medical College, New York, New York
| | - Brittney Shulman
- The Ronald O. Perelman and Claudia Cohen Center for Reproductive Medicine, Weill Cornell Medical College, New York, New York
| | - Nigel Pereira
- The Ronald O. Perelman and Claudia Cohen Center for Reproductive Medicine, Weill Cornell Medical College, New York, New York
| | - Zev Rosenwaks
- The Ronald O. Perelman and Claudia Cohen Center for Reproductive Medicine, Weill Cornell Medical College, New York, New York
| | - Steven D Spandorfer
- The Ronald O. Perelman and Claudia Cohen Center for Reproductive Medicine, Weill Cornell Medical College, New York, New York
| |
Collapse
|
32
|
Zhao X, Zhu D, Ye J, Li X, Wang Z, Zhang L, Xu W. The potential protective role of the combination of IL-22 and TNF-α against genital tract Chlamydia trachomatis infection. Cytokine 2015; 73:66-73. [PMID: 25734538 DOI: 10.1016/j.cyto.2015.01.027] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Revised: 01/20/2015] [Accepted: 01/23/2015] [Indexed: 02/07/2023]
Abstract
Th22 cells are a novel class of lymphocytes characterized by the secretion of both IL-22 and TNF-α. In summary, Th22 cells have little or no direct impact on other immune cells, but exert selective effects on epithelia. It is not known, however, whether Th22 cells play a role in genital mucosal immunity. Here, we demonstrate that IL-22 and TNF-α synergistically induce several immunomodulatory molecules, such as the antimicrobial peptide mBD-2 (murine β-defensin 2) and the antimicrobial chemokines CXCL-9, -10, and -11 in primary murine oviduct epithelial cells (MOECs). The induction of innate immunity is relevant in an in vitro infection model, in which MOECs stimulated with Th22 cell supernatants or recombinant IL-22 and TNF-α effectively inhibit the growth of Chlamydia trachomatis and maintain the survival of the epithelia compared with IL-22 or TNF-α alone. In summary, we demonstrate that the Th22 cell cytokines IL-22 and TNF-α play important roles in genital tract infection. The potential for Th22 cell cytokines to modulate innate immune mediators may lead to the development of new topical agents to treat and/or prevent immune-mediated sexually transmitted diseases (STDs). In summary, we demonstrate that IL-22 and TNF-α represent a potent, synergistic cytokine combination for inducing genital mucosal immunity.
Collapse
Affiliation(s)
- Xiumin Zhao
- Department of Obstetrics and Gynecology, Taizhou First People's Hospital, Taizhou, Zhejiang 318020, PR China
| | - Danyang Zhu
- Department of Obstetrics and Gynecology, Taizhou First People's Hospital, Taizhou, Zhejiang 318020, PR China
| | - Jiangbin Ye
- First Affiliated Hospital, Wengzhou Medical University, Wengzhou, Zhejiang 325035, PR China
| | - Xingqun Li
- First Affiliated Hospital, Wengzhou Medical University, Wengzhou, Zhejiang 325035, PR China
| | - Zhibin Wang
- Department of Microbiology and Immunology, Wengzhou Medical University, Wengzhou, Zhejiang 325035, PR China
| | - Lifang Zhang
- Department of Microbiology and Immunology, Wengzhou Medical University, Wengzhou, Zhejiang 325035, PR China
| | - Wen Xu
- Department of Microbiology and Immunology, Wengzhou Medical University, Wengzhou, Zhejiang 325035, PR China.
| |
Collapse
|
33
|
Healy LL, Cronin JG, Sheldon IM. Polarized Epithelial Cells Secrete Interleukin 6 Apically in the Bovine Endometrium. Biol Reprod 2015; 92:151. [PMID: 25740541 DOI: 10.1095/biolreprod.115.127936] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Accepted: 03/02/2015] [Indexed: 11/01/2022] Open
Abstract
Endometrial epithelial cells are the first line of defense against pathogenic bacteria infecting the uterus. Innate immune responses by these polarized epithelial cells to bacteria and tissue damage are characterized by release of the chemokine (C-X-C motif) ligand 8 (CXCL8) to attract immune cells from the circulation to the site of infection, where they are regulated by the cytokine interleukin (IL) 6. The present study tested the hypothesis that IL6 is predominantly secreted apically from polarized bovine endometrial epithelial cells in response to stimuli associated with bacterial infection and tissue damage. In postpartum animals, concentrations of IL6, but not of CXCL8, were higher in uterine mucus than in peripheral blood. In vitro, polarized endometrial epithelial cells only secreted IL6 apically when treated with bacteria, the pathogen-associated molecule lipopolysaccharide, or the damage-associated molecule IL1alpha, whereas CXCL8 accumulated apically and basolaterally. Furthermore, IL6 accumulated apically irrespective of whether lipopolysaccharide was applied to the apical or basolateral surface of epithelial cells. Secretion of IL6 from epithelial cells was dependent on the trans-Golgi network but was not affected by exogenous ovarian steroids or by coculture with stromal cells. However, a confluent epithelium was essential to protect underlying stromal cells against noxious challenges, including bacteria, lipopolysaccharide, IL1alpha, and a cytolysin. In summary, when a confluent endometrial epithelial cell barrier is faced with infection and damage, chemokines attract immune cells to the uterine lumen, but IL6 is solely secreted apically to ensure immune cells are only exposed to IL6 once they reach the lumen.
Collapse
Affiliation(s)
- Laura L Healy
- Institute of Life Science, College of Medicine, Swansea University, Singleton Park, Swansea, United Kingdom
| | - James G Cronin
- Institute of Life Science, College of Medicine, Swansea University, Singleton Park, Swansea, United Kingdom
| | - I Martin Sheldon
- Institute of Life Science, College of Medicine, Swansea University, Singleton Park, Swansea, United Kingdom
| |
Collapse
|
34
|
Hua Z, Frohlich KM, Zhang Y, Feng X, Zhang J, Shen L. Andrographolide inhibits intracellular Chlamydia trachomatis multiplication and reduces secretion of proinflammatory mediators produced by human epithelial cells. Pathog Dis 2014; 73:1-11. [PMID: 25854005 DOI: 10.1093/femspd/ftu022] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/15/2014] [Indexed: 11/12/2022] Open
Abstract
Chlamydia trachomatis is the most common sexually transmitted bacterial disease worldwide. Untreated C. trachomatis infections may cause inflammation and ultimately damage tissues. Here, we evaluated the ability of Andrographolide (Andro), a natural diterpenoid lactone component of Andrographis paniculata, to inhibit C. trachomatis infection in cultured human cervical epithelial cells. We found that Andro exposure inhibited C. trachomatis growth in a dose- and time-dependent manner. The greatest inhibitory effect was observed when exponentially growing C. trachomatis was exposed to Andro. Electron micrographs demonstrated the accumulation of unusual, structurally deficient chlamydial organisms, correlated with a decrease in levels of OmcB expressed at the late stage of infection. Additionally, Andro significantly reduced the secretion of interleukin6, CXCL8 and interferon-γ-induced protein10 produced by host cells infected with C. trachomatis. These results indicate the efficacy of Andro to perturb C. trachomatis transition from the metabolically active reticulate body to the infectious elementary body and concurrently reduce the production of a proinflammatory mediator by epithelial cells in vitro. Further dissection of Andro's anti-Chlamydia action may provide identification of novel therapeutic targets.
Collapse
Affiliation(s)
- Ziyu Hua
- Department of Neonatology, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China, 400014
| | - Kyla M Frohlich
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Yan Zhang
- Department of Neonatology, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China, 400014
| | | | - Jiaxing Zhang
- Department of Neonatology, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China, 400014
| | - Li Shen
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| |
Collapse
|
35
|
Transcriptional profiling of human epithelial cells infected with plasmid-bearing and plasmid-deficient Chlamydia trachomatis. Infect Immun 2014; 83:534-43. [PMID: 25404022 DOI: 10.1128/iai.02764-14] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Chlamydia trachomatis is an obligate intracellular epitheliotropic bacterial pathogen of humans. Infection of the eye can result in trachoma, the leading cause of preventable blindness in the world. The pathophysiology of blinding trachoma is driven by multiple episodes of reinfection of conjunctival epithelial cells, producing an intense chronic inflammatory response resulting in submucosal tissue remodeling and scarring. Recent reports have shown that infection with trachoma organisms lacking the cryptic chlamydial plasmid is highly attenuated in macaque eyes, a relevant experimental model of human trachoma infection. To better understand the molecular basis of plasmid-mediated infection attenuation and the potential modulation of host immunity, we conducted transcriptional profiling of human epithelial cells infected with C. trachomatis plasmid-bearing (A2497) and plasmid-deficient (A2497P(-)) organisms. Infection of human epithelial cells with either strain increased the expression of host genes coding for proinflammatory (granulocyte-macrophage colony-stimulating factor [GM-CSF], macrophage colony-stimulating factor [MCSF], interleukin-6 [IL-6], IL-8, IL-1α, CXCL1, CXCL2, CXCL3, intercellular adhesion molecule 1 [ICAM1]), chemoattraction (CCL20, CCL5, CXCL10), immune suppression (PD-L1, NFKB1B, TNFAIP3, CGB), apoptosis (CASP9, FAS, IL-24), and cell growth and fibrosis (EGR1 and IL-20) proteins. Statistically significant increases in the levels of expression of many of these genes were found in A2497-infected cells compared to the levels of expression in A2497P(-)-infected cells. Our findings suggest that the chlamydial plasmid plays a focal role in the host cell inflammatory response to infection and immune avoidance. These results provide new insights into the role of the chlamydial plasmid as a chlamydial virulence factor and its contributions to trachoma pathogenesis.
Collapse
|
36
|
Marti H, Koschwanez M, Pesch T, Blenn C, Borel N. Water-filtered infrared a irradiation in combination with visible light inhibits acute chlamydial infection. PLoS One 2014; 9:e102239. [PMID: 25019934 PMCID: PMC4096919 DOI: 10.1371/journal.pone.0102239] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2014] [Accepted: 06/17/2014] [Indexed: 11/19/2022] Open
Abstract
New therapeutic strategies are needed to overcome drawbacks in treatment of infections with intracellular bacteria. Chlamydiaceae are Gram-negative bacteria implicated in acute and chronic diseases such as abortion in animals and trachoma in humans. Water-filtered infrared A (wIRA) is short wavelength infrared radiation with a spectrum ranging from 780 to 1400 nm. In clinical settings, wIRA alone and in combination with visible light (VIS) has proven its efficacy in acute and chronic wound healing processes. This is the first study to demonstrate that wIRA irradiation combined with VIS (wIRA/VIS) diminishes recovery of infectious elementary bodies (EBs) of both intra- and extracellular Chlamydia (C.) in two different cell lines (Vero, HeLa) regardless of the chlamydial strain (C. pecorum, C. trachomatis serovar E) as shown by indirect immunofluorescence and titration by subpassage. Moreover, a single exposure to wIRA/VIS at 40 hours post infection (hpi) led to a significant reduction of C. pecorum inclusion frequency in Vero cells and C. trachomatis in HeLa cells, respectively. A triple dose of irradiation (24, 36, 40 hpi) during the course of C. trachomatis infection further reduced chlamydial inclusion frequency in HeLa cells without inducing the chlamydial persistence/stress response, as ascertained by electron microscopy. Irradiation of host cells (HeLa, Vero) neither affected cell viability nor induced any molecular markers of cytotoxicity as investigated by Alamar blue assay and Western blot analysis. Chlamydial infection, irradiation, and the combination of both showed a similar release pattern of a subset of pro-inflammatory cytokines (MIF/GIF, Serpin E1, RANTES, IL-6, IL-8) and chemokines (IL-16, IP-10, ENA-78, MIG, MIP-1α/β) from host cells. Initial investigation into the mechanism indicated possible thermal effects on Chlamydia due to irradiation. In summary, we demonstrate a non-chemical reduction of chlamydial infection using the combination of water-filtered infrared A and visible light.
Collapse
Affiliation(s)
- Hanna Marti
- Institute of Veterinary Pathology, University of Zurich-Vetsuisse, Zurich, Switzerland
| | - Maria Koschwanez
- Institute of Veterinary Pathology, University of Zurich-Vetsuisse, Zurich, Switzerland
| | - Theresa Pesch
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, Zurich, Switzerland
| | - Christian Blenn
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, Zurich, Switzerland
| | - Nicole Borel
- Institute of Veterinary Pathology, University of Zurich-Vetsuisse, Zurich, Switzerland
| |
Collapse
|
37
|
Lewis ME, Belland RJ, AbdelRahman YM, Beatty WL, Aiyar AA, Zea AH, Greene SJ, Marrero L, Buckner LR, Tate DJ, McGowin CL, Kozlowski PA, O'Brien M, Lillis RA, Martin DH, Quayle AJ. Morphologic and molecular evaluation of Chlamydia trachomatis growth in human endocervix reveals distinct growth patterns. Front Cell Infect Microbiol 2014; 4:71. [PMID: 24959423 PMCID: PMC4050528 DOI: 10.3389/fcimb.2014.00071] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2014] [Accepted: 05/15/2014] [Indexed: 11/13/2022] Open
Abstract
In vitro models of Chlamydia trachomatis growth have long been studied to predict growth in vivo. Alternative or persistent growth modes in vitro have been shown to occur under the influence of numerous stressors but have not been studied in vivo. Here, we report the development of methods for sampling human infections from the endocervix in a manner that permits a multifaceted analysis of the bacteria, host and the endocervical environment. Our approach permits evaluating total bacterial load, transcriptional patterns, morphology by immunofluorescence and electron microscopy, and levels of cytokines and nutrients in the infection microenvironment. By applying this approach to two pilot patients with disparate infections, we have determined that their contrasting growth patterns correlate with strikingly distinct transcriptional biomarkers, and are associated with differences in local levels of IFNγ. Our multifaceted approach will be useful to dissect infections in the human host and be useful in identifying patients at risk for chronic disease. Importantly, the molecular and morphological analyses described here indicate that persistent growth forms can be isolated from the human endocervix when the infection microenvironment resembles the in vitro model of IFNγ-induced persistence.
Collapse
Affiliation(s)
- Maria E Lewis
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center New Orleans, LA, USA
| | - Robert J Belland
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Sciences Center Memphis, TN, USA
| | - Yasser M AbdelRahman
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Sciences Center Memphis, TN, USA ; Department of Microbiology and Immunology, Faculty of Pharmacy, Cairo University Cairo, Egypt
| | - Wandy L Beatty
- Department of Molecular Microbiology, Washington University School of Medicine St. Louis, MO, USA
| | - Ashok A Aiyar
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center New Orleans, LA, USA
| | - Arnold H Zea
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center New Orleans, LA, USA
| | - Sheila J Greene
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center New Orleans, LA, USA
| | - Luis Marrero
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center New Orleans, LA, USA
| | - Lyndsey R Buckner
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center New Orleans, LA, USA
| | - David J Tate
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center New Orleans, LA, USA
| | - Chris L McGowin
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center New Orleans, LA, USA
| | - Pamela A Kozlowski
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center New Orleans, LA, USA
| | - Michelle O'Brien
- Section of Infectious Diseases, Department of Medicine, Louisiana State University Health Sciences Center New Orleans, LA, USA
| | - Rebecca A Lillis
- Section of Infectious Diseases, Department of Medicine, Louisiana State University Health Sciences Center New Orleans, LA, USA
| | - David H Martin
- Section of Infectious Diseases, Department of Medicine, Louisiana State University Health Sciences Center New Orleans, LA, USA
| | - Alison J Quayle
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center New Orleans, LA, USA
| |
Collapse
|
38
|
Frohlich KM, Hua Z, Quayle AJ, Wang J, Lewis ME, Chou CW, Luo M, Buckner LR, Shen L. Membrane vesicle production by Chlamydia trachomatis as an adaptive response. Front Cell Infect Microbiol 2014; 4:73. [PMID: 24959424 PMCID: PMC4050530 DOI: 10.3389/fcimb.2014.00073] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2014] [Accepted: 05/19/2014] [Indexed: 01/08/2023] Open
Abstract
Bacteria have evolved specific adaptive responses to cope with changing environments. These adaptations include stress response phenotypes with dynamic modifications of the bacterial cell envelope and generation of membrane vesicles (MVs). The obligate intracellular bacterium, Chlamydia trachomatis, typically has a biphasic lifestyle, but can enter into an altered growth state typified by morphologically aberrant chlamydial forms, termed persistent growth forms, when induced by stress in vitro. How C. trachomatis can adapt to a persistent growth state in host epithelial cells in vivo is not well understood, but is an important question, since it extends the host-bacterial relationship in vitro and has thus been indicated as a survival mechanism in chronic chlamydial infections. Here, we review recent findings on the mechanistic aspects of bacterial adaptation to stress with a focus on how C. trachomatis remodels its envelope, produces MVs, and the potential important consequences of MV production with respect to host-pathogen interactions. Emerging data suggest that the generation of MVs may be an important mechanism for C. trachomatis intracellular survival of stress, and thus may aid in the establishment of a chronic infection in human genital epithelial cells.
Collapse
Affiliation(s)
- Kyla M Frohlich
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center New Orleans, LA, USA
| | - Ziyu Hua
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center New Orleans, LA, USA ; Department of Neonatology, Ministry of Education Key Laboratory of Child Development and Disorder, The Children's Hospital, Chongqing Medical University Chongqing, China
| | - Alison J Quayle
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center New Orleans, LA, USA
| | - Jin Wang
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center New Orleans, LA, USA
| | - Maria E Lewis
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center New Orleans, LA, USA
| | - Chau-wen Chou
- Department of Chemistry, University of Georgia Athens, GA, USA
| | - Miao Luo
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center New Orleans, LA, USA
| | - Lyndsey R Buckner
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center New Orleans, LA, USA
| | - Li Shen
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center New Orleans, LA, USA
| |
Collapse
|
39
|
Jang JE, Kim HM, Kim H, Jeon DY, Park CH, Kwon SY, Chung JW, Khang G. Inflammatory Responses to Hydroxyapatite/Poly(lactic-co-glycolic acid) Scaffolds with Variation of Compositions. POLYMER-KOREA 2014. [DOI: 10.7317/pk.2014.38.2.156] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|