1
|
Yang S, Zeng J, Yu J, Sun R, Tuo Y, Bai H. Insights into Chlamydia Development and Host Cells Response. Microorganisms 2024; 12:1302. [PMID: 39065071 PMCID: PMC11279054 DOI: 10.3390/microorganisms12071302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 06/15/2024] [Accepted: 06/23/2024] [Indexed: 07/28/2024] Open
Abstract
Chlamydia infections commonly afflict both humans and animals, resulting in significant morbidity and imposing a substantial socioeconomic burden worldwide. As an obligate intracellular pathogen, Chlamydia interacts with other cell organelles to obtain necessary nutrients and establishes an intracellular niche for the development of a biphasic intracellular cycle. Eventually, the host cells undergo lysis or extrusion, releasing infectious elementary bodies and facilitating the spread of infection. This review provides insights into Chlamydia development and host cell responses, summarizing the latest research on the biphasic developmental cycle, nutrient acquisition, intracellular metabolism, host cell fates following Chlamydia invasion, prevalent diseases associated with Chlamydia infection, treatment options, and vaccine prevention strategies. A comprehensive understanding of these mechanisms will contribute to a deeper comprehension of the intricate equilibrium between Chlamydia within host cells and the progression of human disease.
Collapse
Affiliation(s)
| | | | | | | | | | - Hong Bai
- Tianjin Key Laboratory of Cellular and Molecular Immunology (The Educational Ministry of China), Tianjin Institute of Immunology, Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China; (S.Y.); (J.Z.); (J.Y.); (R.S.); (Y.T.)
| |
Collapse
|
2
|
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
|
3
|
Ardizzone CM, Taylor CM, Toh E, Lillis RA, Elnaggar JH, Lammons JW, Mott PD, Duffy EL, Shen L, Quayle AJ. Association of Chlamydia trachomatis burden with the vaginal microbiota, bacterial vaginosis, and metronidazole treatment. Front Cell Infect Microbiol 2023; 13:1289449. [PMID: 38149008 PMCID: PMC10750252 DOI: 10.3389/fcimb.2023.1289449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 11/21/2023] [Indexed: 12/28/2023] Open
Abstract
Bacterial vaginosis (BV), a dysbiosis of the vaginal microbiota, is a common coinfection with Chlamydia trachomatis (Ct), and BV-associated bacteria (BVAB) and their products have been implicated in aiding Ct evade natural immunity. Here, we determined if a non-optimal vaginal microbiota was associated with a higher genital Ct burden and if metronidazole, a standard treatment for BV, would reduce Ct burden or aid in natural clearance of Ct infection. Cervicovaginal samples were collected from women at enrollment and, if testing positive for Ct infection, at a follow-up visit approximately one week later. Cervical Ct burden was assessed by inclusion forming units (IFU) and Ct genome copy number (GCN), and 16S rRNA gene sequencing was used to determine the composition of the vaginal microbiota. We observed a six-log spectrum of IFU and an eight-log spectrum of GCN in our study participants at their enrollment visit, but BV, as indicated by Amsel's criteria, Nugent scoring, or VALENCIA community state typing, did not predict infectious and total Ct burden, although IFU : GCN increased with Amsel and Nugent scores and in BV-like community state types. Ct burden was, however, associated with the abundance of bacterial species in the vaginal microbiota, negatively with Lactobacillus crispatus and positively with Prevotella bivia. Women diagnosed with BV were treated with metronidazole, and Ct burden was significantly reduced in those who resolved BV with treatment. A subset of women naturally cleared Ct infection in the interim, typified by low Ct burden at enrollment and resolution of BV. Abundance of many BVAB decreased, and Lactobacillus increased, in response to metronidazole treatment, but no changes in abundances of specific vaginal bacteria were unique to women who spontaneously cleared Ct infection.
Collapse
Affiliation(s)
- Caleb M. Ardizzone
- 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
| | - Evelyn Toh
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Rebecca A. Lillis
- Department of Medicine, Section of Infectious Diseases, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Jacob H. Elnaggar
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - John W. Lammons
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Patricia Dehon Mott
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Emily L. Duffy
- Department of Medicine, Section of Infectious Diseases, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Li Shen
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center, 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
|
4
|
Graspeuntner S, Koethke K, Scholz C, Semmler L, Lupatsii M, Kirchhoff L, Herrmann J, Rox K, Wittstein K, Käding N, Hanker LC, Stadler M, Brönstrup M, Müller R, Shima K, Rupp J. Sorangicin A Is Active against Chlamydia in Cell Culture, Explanted Fallopian Tubes, and Topical In Vivo Treatment. Antibiotics (Basel) 2023; 12:antibiotics12050795. [PMID: 37237698 DOI: 10.3390/antibiotics12050795] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 04/17/2023] [Accepted: 04/20/2023] [Indexed: 05/28/2023] Open
Abstract
Current treatment of Chlamydia trachomatis using doxycycline and azithromycin introduces detrimental side effects on the host's microbiota. As a potential alternative treatment, the myxobacterial natural product sorangicin A (SorA) blocks the bacterial RNA polymerase. In this study we analyzed the effectiveness of SorA against C. trachomatis in cell culture, and explanted fallopian tubes and systemic and local treatment in mice, providing also pharmacokinetic data on SorA. Potential side effects of SorA on the vaginal and gut microbiome were assessed in mice and against human-derived Lactobacillus species. SorA showed minimal inhibitory concentrations of 80 ng/mL (normoxia) to 120 ng/mL (hypoxia) against C. trachomatis in vitro and was eradicating C. trachomatis at a concentration of 1 µg/mL from fallopian tubes. In vivo, SorA reduced chlamydial shedding by more than 100-fold within the first days of infection by topical application corresponding with vaginal detection of SorA only upon topical treatment, but not after systemic application. SorA changed gut microbial composition during intraperitoneal application only and did neither alter the vaginal microbiota in mice nor affect growth of human-derived lactobacilli. Additional dose escalations and/or pharmaceutical modifications will be needed to optimize application of SorA and to reach sufficient anti-chlamydial activity in vivo.
Collapse
Affiliation(s)
- Simon Graspeuntner
- Department of Infectious Diseases and Microbiology, University of Luebeck, 23538 Luebeck, Germany
- German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, 23538 Lübeck, Germany
| | - Katharina Koethke
- Department of Infectious Diseases and Microbiology, University of Luebeck, 23538 Luebeck, Germany
| | - Celeste Scholz
- Department of Infectious Diseases and Microbiology, University of Luebeck, 23538 Luebeck, Germany
| | - Lea Semmler
- Department of Infectious Diseases and Microbiology, University of Luebeck, 23538 Luebeck, Germany
| | - Mariia Lupatsii
- Department of Infectious Diseases and Microbiology, University of Luebeck, 23538 Luebeck, Germany
| | - Laura Kirchhoff
- Department of Infectious Diseases and Microbiology, University of Luebeck, 23538 Luebeck, Germany
| | - Jennifer Herrmann
- Helmholtz Centre for Infection Research (HZI), Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), and Department of Pharmacy, Saarland University, 66123 Saarbrücken, Germany
- German Center for Infection Research (DZIF), Partner Site Hannover-Braunschweig, 38124 Braunschweig, Germany
| | - Katharina Rox
- German Center for Infection Research (DZIF), Partner Site Hannover-Braunschweig, 38124 Braunschweig, Germany
- Department of Chemical Biology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
| | - Kathrin Wittstein
- German Center for Infection Research (DZIF), Partner Site Hannover-Braunschweig, 38124 Braunschweig, Germany
- Department of Microbial Drugs, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
| | - Nadja Käding
- Department of Infectious Diseases and Microbiology, University of Luebeck, 23538 Luebeck, Germany
- German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, 23538 Lübeck, Germany
| | - Lars C Hanker
- Department of Obstetrics and Gynecology, University Hospital of Schleswig Holstein, 23538 Luebeck, Germany
| | - Marc Stadler
- German Center for Infection Research (DZIF), Partner Site Hannover-Braunschweig, 38124 Braunschweig, Germany
- Department of Microbial Drugs, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
| | - Mark Brönstrup
- German Center for Infection Research (DZIF), Partner Site Hannover-Braunschweig, 38124 Braunschweig, Germany
- Department of Chemical Biology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
| | - Rolf Müller
- Helmholtz Centre for Infection Research (HZI), Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), and Department of Pharmacy, Saarland University, 66123 Saarbrücken, Germany
- German Center for Infection Research (DZIF), Partner Site Hannover-Braunschweig, 38124 Braunschweig, Germany
| | - Kensuke Shima
- Department of Infectious Diseases and Microbiology, University of Luebeck, 23538 Luebeck, Germany
| | - Jan Rupp
- Department of Infectious Diseases and Microbiology, University of Luebeck, 23538 Luebeck, Germany
- German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, 23538 Lübeck, Germany
| |
Collapse
|
5
|
Banerjee A, Sun Y, Muramatsu MK, Toh E, Nelson DE. A Member of an Ancient Family of Bacterial Amino Acids Transporters Contributes to Chlamydia Nutritional Virulence and Immune Evasion. Infect Immun 2023; 91:e0048322. [PMID: 36847502 PMCID: PMC10068747 DOI: 10.1128/iai.00483-22] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 01/31/2023] [Indexed: 03/01/2023] Open
Abstract
Many obligate intracellular bacteria, including members of the genus Chlamydia, cannot synthesize a variety of amino acids de novo and acquire these from host cells via largely unknown mechanisms. Previously, we determined that a missense mutation in ctl0225, a conserved Chlamydia open reading frame of unknown function, mediated sensitivity to interferon gamma. Here, we show evidence that CTL0225 is a member of the SnatA family of neutral amino acid transporters that contributes to the import of several amino acids into Chlamydia cells. Further, we show that CTL0225 orthologs from two other distantly related obligate intracellular pathogens (Coxiella burnetii and Buchnera aphidicola) are sufficient to import valine into Escherichia coli. We also show that chlamydia infection and interferon exposure have opposing effects on amino acid metabolism, potentially explaining the relationship between CTL0225 and interferon sensitivity. Overall, we show that phylogenetically diverse intracellular pathogens use an ancient family of amino acid transporters to acquire host amino acids and provide another example of how nutritional virulence and immune evasion can be linked in obligate intracellular pathogens.
Collapse
Affiliation(s)
- Arkaprabha Banerjee
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Yuan Sun
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Matthew K. Muramatsu
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Evelyn Toh
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - David E. Nelson
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| |
Collapse
|
6
|
Intracellular lifestyle of Chlamydia trachomatis and host-pathogen interactions. Nat Rev Microbiol 2023:10.1038/s41579-023-00860-y. [PMID: 36788308 DOI: 10.1038/s41579-023-00860-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/23/2023] [Indexed: 02/16/2023]
Abstract
In recent years, substantial progress has been made in the understanding of the intracellular lifestyle of Chlamydia trachomatis and how the bacteria establish themselves in the human host. As an obligate intracellular pathogenic bacterium with a strongly reduced coding capacity, C. trachomatis depends on the provision of nutrients from the host cell. In this Review, we summarize the current understanding of how C. trachomatis establishes its intracellular replication niche, how its metabolism functions in the host cell, how it can defend itself against the cell autonomous and innate immune response and how it overcomes adverse situations through the transition to a persistent state. In particular, we focus on those processes for which a mechanistic understanding has been achieved.
Collapse
|
7
|
N'Gadjaga MD, Perrinet S, Connor MG, Bertolin G, Millot GA, Subtil A. Chlamydia trachomatis development requires both host glycolysis and oxidative phosphorylation but has only minor effects on these pathways. J Biol Chem 2022; 298:102338. [PMID: 35931114 PMCID: PMC9449673 DOI: 10.1016/j.jbc.2022.102338] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 07/27/2022] [Accepted: 07/28/2022] [Indexed: 11/17/2022] Open
Abstract
The obligate intracellular bacteria Chlamydia trachomatis obtain all nutrients from the cytoplasm of their epithelial host cells and stimulate glucose uptake by these cells. They even hijack host ATP, exerting a strong metabolic pressure on their host at the peak of the proliferative stage of their developmental cycle. However, it is largely unknown whether infection modulates the metabolism of the host cell. Also, the reliance of the bacteria on host metabolism might change during their progression through their biphasic developmental cycle. Herein, using primary epithelial cells and 2 cell lines of nontumoral origin, we showed that between the 2 main ATP-producing pathways of the host, oxidative phosphorylation (OxPhos) remained stable and glycolysis was slightly increased. Inhibition of either pathway strongly reduced bacterial proliferation, implicating that optimal bacterial growth required both pathways to function at full capacity. While we found C. trachomatis displayed some degree of energetic autonomy in the synthesis of proteins expressed at the onset of infection, functional host glycolysis was necessary for the establishment of early inclusions, whereas OxPhos contributed less. These observations correlated with the relative contributions of the pathways in maintaining ATP levels in epithelial cells, with glycolysis contributing the most. Altogether, this work highlights the dependence of C. trachomatis on both host glycolysis and OxPhos for efficient bacterial replication. However, ATP consumption appears at equilibrium with the normal production capacity of the host and the bacteria, so that no major shift between these pathways is required to meet bacterial needs.
Collapse
Affiliation(s)
- Maimouna D N'Gadjaga
- Institut Pasteur, CNRS UMR3691, Cellular Biology of Microbial Infection, Université Paris Cité, Paris, France; Sorbonne Université, Collège Doctoral, Paris, France
| | - Stéphanie Perrinet
- Institut Pasteur, CNRS UMR3691, Cellular Biology of Microbial Infection, Université Paris Cité, Paris, France
| | - Michael G Connor
- Institut Pasteur, Chromatin and Infection, Université Paris Cité, Paris, France
| | - Giulia Bertolin
- CNRS, IGDR (Institute of Genetics and Development of Rennes), UMR 6290, Univ Rennes, Rennes, France
| | - Gaël A Millot
- Institut Pasteur, Hub Bioinformatique et Biostatistique-DBC, Université Paris Cité, Paris, France
| | - Agathe Subtil
- Institut Pasteur, CNRS UMR3691, Cellular Biology of Microbial Infection, Université Paris Cité, Paris, France.
| |
Collapse
|
8
|
Zhao X, Jiang Y, Xu M, Hu J, Feng N, Deng H, Lu C, Huang T. Indoleamine 2,3-dioxygenase 1 regulates breast cancer tamoxifen resistance through interleukin-6/signal transducer and activator of transcription 3. Toxicol Appl Pharmacol 2022; 440:115921. [DOI: 10.1016/j.taap.2022.115921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 02/01/2022] [Accepted: 02/08/2022] [Indexed: 11/25/2022]
|
9
|
Li H, Zang Y, Wang C, Li H, Fan A, Han C, Xue F. The Interaction Between Microorganisms, Metabolites, and Immune System in the Female Genital Tract Microenvironment. Front Cell Infect Microbiol 2020; 10:609488. [PMID: 33425785 PMCID: PMC7785791 DOI: 10.3389/fcimb.2020.609488] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 11/18/2020] [Indexed: 12/24/2022] Open
Abstract
The female reproductive tract microenvironment includes microorganisms, metabolites, and immune components, and the balance of the interactions among them plays an important role in maintaining female reproductive tract homeostasis and health. When any one of the reproductive tract microorganisms, metabolites, or immunity is out of balance, it will affect the other two, leading to the occurrence and development of diseases and the appearance of corresponding symptoms and signs, such as infertility, miscarriage, premature delivery, and gynecological tumors caused by infectious diseases of the reproductive tract. Nutrients in the female reproductive tract provide symbiotic and pathogenic microorganisms with a source of nutrients for their own reproduction and utilization. At the same time, this interaction with the host forms a variety of metabolites. Changes in metabolites in the host reproductive tract are related not only to the interaction between the host and microbiota under dysbiosis but also to changes in host immunity or the environment, all of which will participate in the pathogenesis of diseases and lead to disease-related phenotypes. Microorganisms and their metabolites can also interact with host immunity, activate host immunity, and change the host immune status and are closely related to persistent genital pathogen infections, aggravation of infectious diseases, severe pregnancy outcomes, and even gynecological cancers. Therefore, studying the interaction between microorganisms, metabolites, and immunity in the reproductive tract cannot only reveal the pathogenic mechanisms that lead to inflammation of the reproductive tract, adverse pregnancy outcomes and tumorigenesis but also provide a basis for further research on the diagnosis and treatment of targets.
Collapse
Affiliation(s)
- Huanrong Li
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China.,Department of Gynecology and Obstetrics, Tianjin Key Laboratory of Female Reproductive Health and Eugenic, Tianjin Medical University General Hospital, Tianjin, China
| | - Yuqin Zang
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China.,Department of Gynecology and Obstetrics, Tianjin Key Laboratory of Female Reproductive Health and Eugenic, Tianjin Medical University General Hospital, Tianjin, China
| | - Chen Wang
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China.,Department of Gynecology and Obstetrics, Tianjin Key Laboratory of Female Reproductive Health and Eugenic, Tianjin Medical University General Hospital, Tianjin, China
| | - Huiyang Li
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China.,Department of Gynecology and Obstetrics, Tianjin Key Laboratory of Female Reproductive Health and Eugenic, Tianjin Medical University General Hospital, Tianjin, China
| | - Aiping Fan
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China.,Department of Gynecology and Obstetrics, Tianjin Key Laboratory of Female Reproductive Health and Eugenic, Tianjin Medical University General Hospital, Tianjin, China
| | - Cha Han
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China.,Department of Gynecology and Obstetrics, Tianjin Key Laboratory of Female Reproductive Health and Eugenic, Tianjin Medical University General Hospital, Tianjin, China
| | - Fengxia Xue
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China.,Department of Gynecology and Obstetrics, Tianjin Key Laboratory of Female Reproductive Health and Eugenic, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
10
|
Mechanisms controlling bacterial infection in myeloid cells under hypoxic conditions. Cell Mol Life Sci 2020; 78:1887-1907. [PMID: 33125509 PMCID: PMC7966188 DOI: 10.1007/s00018-020-03684-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Revised: 09/08/2020] [Accepted: 10/09/2020] [Indexed: 12/12/2022]
Abstract
Various factors of the tissue microenvironment such as the oxygen concentration influence the host-pathogen interaction. During the past decade, hypoxia-driven signaling via hypoxia-inducible factors (HIF) has emerged as an important factor that affects both the pathogen and the host. In this chapter, we will review the current knowledge of this complex interplay, with a particular emphasis given to the impact of hypoxia and HIF on the inflammatory and antimicrobial activity of myeloid cells, the bacterial responses to hypoxia and the containment of bacterial infections under oxygen-limited conditions. We will also summarize how low oxygen concentrations influence the metabolism of neutrophils, macrophages and dendritic cells. Finally, we will discuss the consequences of hypoxia and HIFα activation for the invading pathogen, with a focus on Pseudomonas aeruginosa, Mycobacterium tuberculosis, Coxiella burnetii, Salmonella enterica and Staphylococcus aureus. This includes a description of the mechanisms and microbial factors, which the pathogens use to sense and react to hypoxic conditions.
Collapse
|
11
|
Kaul NC, Mohapatra SR, Adam I, Tucher C, Tretter T, Opitz CA, Lorenz HM, Tykocinski LO. Hypoxia decreases the T helper cell-suppressive capacity of synovial fibroblasts by downregulating IDO1-mediated tryptophan metabolism. Rheumatology (Oxford) 2020; 59:1148-1158. [PMID: 31846032 DOI: 10.1093/rheumatology/kez587] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 10/07/2019] [Indexed: 11/14/2022] Open
Abstract
OBJECTIVE The development of RA is linked to local infiltration of immune cells and to changes in the phenotype of synovial fibroblasts. Synovial fibroblasts possess the capacity to suppress T cell responses through indoleamine 2, 3-dioxygenase 1 (IDO1)-mediated tryptophan metabolism. However, synovial fibroblasts from RA patients are restricted in this immune-modulatory function. Moreover, hypoxic conditions are detected within synovial tissues of RA patients, with oxygen tensions of only 3.2% O2. This study aims at investigating the effects of hypoxia on the interaction between T cells and synovial fibroblasts, particularly on the T cell-suppressive capacities of synovial fibroblasts. METHODS Synovial fibroblasts were cultured with Th cells under normoxic and hypoxic conditions (3% O2). Th cell proliferation was detected by flow cytometry. Tryptophan and kynurenine amounts were measured by HPLC. IDO1 expression and signal transducer and activator of transcription 1 (STAT1) phosphorylation were quantified by real-time PCR or western blot, and cytokine secretion by ELISA. RESULTS Hypoxic conditions strongly diminished the Th cell-suppressive capacities of both OA synovial fibroblasts and RA synovial fibroblasts. Accordingly, IDO1 mRNA and protein expression, STAT1 phosphorylation and tryptophan metabolism were greatly reduced in OA synovial fibroblasts by hypoxia. MMP-3, IL-6, IL-10 and IFNγ secretion were significantly decreased under hypoxia in synovial fibroblast-Th cell co-cultures, while IL-17A levels were elevated. Supplementation with IFNγ, a well-known inducer of IDO1 expression, could rescue neither IDO1 expression nor Th cell suppression under hypoxic conditions. CONCLUSION Hypoxia strongly affected the crosstalk between synovial fibroblasts and Th cells. By reducing the efficiency of synovial fibroblasts to restrict Th cell proliferation and by increasing the expression of IL-17A, hypoxia might have implications on the pathophysiology of RA.
Collapse
Affiliation(s)
- Nathalie-Christin Kaul
- Division of Rheumatology, Department of Internal Medicine V, Heidelberg University Hospital
| | - Soumya R Mohapatra
- Brain Cancer Metabolism, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Isabell Adam
- Brain Cancer Metabolism, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Christine Tucher
- Division of Rheumatology, Department of Internal Medicine V, Heidelberg University Hospital
| | - Theresa Tretter
- Division of Rheumatology, Department of Internal Medicine V, Heidelberg University Hospital
| | - Christiane A Opitz
- Brain Cancer Metabolism, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Hanns-Martin Lorenz
- Division of Rheumatology, Department of Internal Medicine V, Heidelberg University Hospital
| | - Lars-Oliver Tykocinski
- Division of Rheumatology, Department of Internal Medicine V, Heidelberg University Hospital
| |
Collapse
|
12
|
Bishop RC, Boretto M, Rutkowski MR, Vankelecom H, Derré I. Murine Endometrial Organoids to Model Chlamydia Infection. Front Cell Infect Microbiol 2020; 10:416. [PMID: 32923409 PMCID: PMC7456808 DOI: 10.3389/fcimb.2020.00416] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 07/07/2020] [Indexed: 01/07/2023] Open
Abstract
The obligate intracellular bacterium Chlamydia trachomatis is the leading cause of bacterial sexually transmitted infections. Once internalized in host cells, C. trachomatis undergoes a biphasic developmental cycle within a membrane-bound compartment, known as the inclusion. Successful establishment of the intracellular niche relies on bacterial Type III effector proteins, such as Inc proteins. In vitro and in vivo systems have contributed to elucidating the intracellular lifestyle of C. trachomatis, but additional models combining the archetypal environment of infection with the advantages of in vitro systems are needed. Organoids are three-dimensional structures that recapitulate the microanatomy of an organ's epithelial layer, bridging the gap between in vitro and in vivo systems. Organoids are emerging as relevant model systems to study interactions between bacterial pathogens and their hosts. Here, we took advantage of recently developed murine endometrial organoids (EMOs) and present a C. trachomatis-murine EMO infection model system. Confocal microscopy of EMOs infected with fluorescent protein-expressing bacteria revealed that inclusions are formed within the cytosol of epithelial cells. Moreover, infection with a C. trachomatis strain that allows for the tracking of RB to EB transition indicated that the bacteria undergo a full developmental cycle, which was confirmed by harvesting infectious bacteria from infected EMOs. Finally, the inducible gene expression and cellular localization of a Chlamydia Inc protein within infected EMOs further demonstrated that this model is compatible with the study of Type III secreted effectors. Altogether, we describe a novel and relevant system for the study of Chlamydia-host interactions.
Collapse
Affiliation(s)
- R Clayton Bishop
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA, United States
| | - Matteo Boretto
- Unit of Stem Cell Research, Cluster of Stem Cell and Developmental Biology, Department of Development and Regenerations, University of Leuven, Leuven, Belgium
| | - Melanie R Rutkowski
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA, United States
| | - Hugo Vankelecom
- Unit of Stem Cell Research, Cluster of Stem Cell and Developmental Biology, Department of Development and Regenerations, University of Leuven, Leuven, Belgium
| | - Isabelle Derré
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA, United States
| |
Collapse
|
13
|
Comparison of In Vitro Chlamydia muridarum Infection Under Aerobic and Anaerobic Conditions. Curr Microbiol 2020; 77:1580-1589. [PMID: 32253468 DOI: 10.1007/s00284-020-01966-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 03/24/2020] [Indexed: 10/24/2022]
Abstract
Although Chlamydia infects host body regions that are hypoxic to anoxic, standard Chlamydiae culture conditions are in CO2 enriched (5%) atmospheric oxygen (21%). Because of its success in causing disease in principally anaerobic body sites, e.g., vaginal tract, we hypothesize that Chlamydia has an anaerobic life cycle that plays a role in its maintenance in the host. Using a model system developed for the anaerobic culture of mammalian cells, we assessed the anoxic infectious cycle of C. muridarum in anaerobically cultured HeLa 229 cells. In the absence of oxygen, C. muridarum is capable of going through their life cycle, although its cycle is slowed (2 days post-infection anaerobic vs. 1 day aerobic). Interestingly, in addition to a slower rate of replication, there is a reduction in Chlamydia inclusion number and size as compared to aerobic controls. Anaerobic infected host cell physiology also changed with IL-6 and IL-8 production significantly lower (p ≤ 0.05) compared to aerobic infected host cells (day 4 post-infection). These findings demonstrate that Chlamydia are capable of replicating in the absence of oxygen.
Collapse
|
14
|
Maffei B, Laverrière M, Wu Y, Triboulet S, Perrinet S, Duchateau M, Matondo M, Hollis RL, Gourley C, Rupp J, Keillor JW, Subtil A. Infection-driven activation of transglutaminase 2 boosts glucose uptake and hexosamine biosynthesis in epithelial cells. EMBO J 2020; 39:e102166. [PMID: 32134139 DOI: 10.15252/embj.2019102166] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 01/21/2020] [Accepted: 01/31/2020] [Indexed: 12/16/2022] Open
Abstract
Transglutaminase 2 (TG2) is a ubiquitously expressed enzyme with transamidating activity. We report here that both expression and activity of TG2 are enhanced in mammalian epithelial cells infected with the obligate intracellular bacteria Chlamydia trachomatis. Genetic or pharmacological inhibition of TG2 impairs bacterial development. We show that TG2 increases glucose import by up-regulating the transcription of the glucose transporter genes GLUT-1 and GLUT-3. Furthermore, TG2 activation drives one specific glucose-dependent pathway in the host, i.e., hexosamine biosynthesis. Mechanistically, we identify the glucosamine:fructose-6-phosphate amidotransferase (GFPT) among the substrates of TG2. GFPT modification by TG2 increases its enzymatic activity, resulting in higher levels of UDP-N-acetylglucosamine biosynthesis and protein O-GlcNAcylation. The correlation between TG2 transamidating activity and O-GlcNAcylation is disrupted in infected cells because host hexosamine biosynthesis is being exploited by the bacteria, in particular to assist their division. In conclusion, our work establishes TG2 as a key player in controlling glucose-derived metabolic pathways in mammalian cells, themselves hijacked by C. trachomatis to sustain their own metabolic needs.
Collapse
Affiliation(s)
- Benoit Maffei
- Unité de Biologie cellulaire de l'infection microbienne, CNRS UMR3691, Institut Pasteur, Paris, France.,Collège Doctoral, Sorbonne Université, Paris, France
| | - Marc Laverrière
- Unité de Biologie cellulaire de l'infection microbienne, CNRS UMR3691, Institut Pasteur, Paris, France
| | - Yongzheng Wu
- Unité de Biologie cellulaire de l'infection microbienne, CNRS UMR3691, Institut Pasteur, Paris, France
| | - Sébastien Triboulet
- Unité de Biologie cellulaire de l'infection microbienne, CNRS UMR3691, Institut Pasteur, Paris, France
| | - Stéphanie Perrinet
- Unité de Biologie cellulaire de l'infection microbienne, CNRS UMR3691, Institut Pasteur, Paris, France
| | - Magalie Duchateau
- Plateforme Protéomique, Unité de Spectrométrie de Masse pour la Biologie, USR 2000 CNRS, Institut Pasteur, Paris, France
| | - Mariette Matondo
- Plateforme Protéomique, Unité de Spectrométrie de Masse pour la Biologie, USR 2000 CNRS, Institut Pasteur, Paris, France
| | - Robert L Hollis
- Nicola Murray Centre for Ovarian Cancer Research, Cancer Research UK Edinburgh Centre, MRC IGMM, University of Edinburgh, Edinburgh, UK
| | - Charlie Gourley
- Nicola Murray Centre for Ovarian Cancer Research, Cancer Research UK Edinburgh Centre, MRC IGMM, University of Edinburgh, Edinburgh, UK
| | - Jan Rupp
- Department of Infectious Diseases and Microbiology, University of Lübeck, Lübeck, Germany
| | - Jeffrey W Keillor
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, ON, Canada
| | - Agathe Subtil
- Unité de Biologie cellulaire de l'infection microbienne, CNRS UMR3691, Institut Pasteur, Paris, France
| |
Collapse
|
15
|
Cheok YY, Lee CYQ, Cheong HC, Looi CY, Wong WF. Chronic Inflammatory Diseases at Secondary Sites Ensuing Urogenital or Pulmonary Chlamydia Infections. Microorganisms 2020; 8:microorganisms8010127. [PMID: 31963395 PMCID: PMC7022716 DOI: 10.3390/microorganisms8010127] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Revised: 01/02/2020] [Accepted: 01/08/2020] [Indexed: 12/11/2022] Open
Abstract
Chlamydia trachomatis and C. pneumoniae are members of the Chlamydiaceae family of obligate intracellular bacteria. The former causes diseases predominantly at the mucosal epithelial layer of the urogenital or eye, leading to pelvic inflammatory diseases or blindness; while the latter is a major causative agent for pulmonary infection. On top of these well-described diseases at the respective primary infection sites, Chlamydia are notoriously known to migrate and cause pathologies at remote sites of a host. One such example is the sexually acquired reactive arthritis that often occurs at few weeks after genital C. trachomatis infection. C. pneumoniae, on the other hand, has been implicated in an extensive list of chronic inflammatory diseases which include atherosclerosis, multiple sclerosis, Alzheimer’s disease, asthma, and primary biliary cirrhosis. This review summarizes the Chlamydia infection associated diseases at the secondary sites of infection, and describes the potential mechanisms involved in the disease migration and pathogenesis.
Collapse
Affiliation(s)
- Yi Ying Cheok
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, Kuala Lumpur 50603, Malaysia; (Y.Y.C.); (C.Y.Q.L.); (H.C.C.)
| | - Chalystha Yie Qin Lee
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, Kuala Lumpur 50603, Malaysia; (Y.Y.C.); (C.Y.Q.L.); (H.C.C.)
| | - Heng Choon Cheong
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, Kuala Lumpur 50603, Malaysia; (Y.Y.C.); (C.Y.Q.L.); (H.C.C.)
| | - Chung Yeng Looi
- School of Biosciences, Faculty of Health and Medical Sciences, Taylor’s University, Subang Jaya 47500, Malaysia;
| | - Won Fen Wong
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, Kuala Lumpur 50603, Malaysia; (Y.Y.C.); (C.Y.Q.L.); (H.C.C.)
- Correspondence: ; Tel.: +603-7967-6672
| |
Collapse
|
16
|
Loeper N, Graspeuntner S, Ledig S, Kaufhold I, Hoellen F, Schiefer A, Henrichfreise B, Pfarr K, Hoerauf A, Shima K, Rupp J. Elaborations on Corallopyronin A as a Novel Treatment Strategy Against Genital Chlamydial Infections. Front Microbiol 2019; 10:943. [PMID: 31134007 PMCID: PMC6514060 DOI: 10.3389/fmicb.2019.00943] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 04/15/2019] [Indexed: 11/13/2022] Open
Abstract
Ascending Chlamydia trachomatis infection causes functional damage to the fallopian tubes, which may lead to ectopic pregnancy and infertility in women. Treatment failures using the standard regimens of doxycycline and azithromycin have been observed. We tested the polyketide-derived α-pyrone antibiotic Corallopyronin A (CorA) that inhibits the bacterial DNA dependent RNA polymerase and has strong activity against various extracellular and some intracellular bacteria. Extensive testing in cell culture infection models and in an ex vivo human fallopian tube model under different oxygen concentrations was performed to assess the anti-chlamydial efficacy of CorA at physiological conditions. CorA showed high efficacy against C. trachomatis (MICN/H: 0.5 μg/mL for serovar D and L2), C. muridarum (MICN/H: 0.5 μg/mL), and C. pneumoniae (MICN/H: 1 μg/mL) under normoxic (N) and hypoxic (H) conditions. Recoverable inclusion forming units were significantly lower already at 0.25 μg/mL for all tested chlamydiae. CorA at a concentration of 1 μg/mL was also effective against already established C. trachomatis and C. pneumoniae infections (up to 24 h.p.i.) in epithelial cells, while efficacy against C. muridarum was limited to earlier time points. A preliminary study using a C. muridarum genital infection model revealed corresponding limitations in the efficacy. Importantly, in an ex vivo human fallopian tube model, the growth of C. trachomatis was significantly inhibited by CorA at concentrations of 1–2 μg/mL under normoxic and hypoxic conditions. The overall high efficacies of CorA against C. trachomatis in cell culture and an ex vivo human fallopian tube model under physiological oxygen concentrations qualifies this drug as a candidate that should be further investigated.
Collapse
Affiliation(s)
- Nathalie Loeper
- Department of Infectious Diseases and Microbiology, University of Lübeck, Lübeck, Germany
| | - Simon Graspeuntner
- Department of Infectious Diseases and Microbiology, University of Lübeck, Lübeck, Germany
| | - Svea Ledig
- Department of Infectious Diseases and Microbiology, University of Lübeck, Lübeck, Germany
| | - Inga Kaufhold
- Department of Infectious Diseases and Microbiology, University of Lübeck, Lübeck, Germany
| | - Friederike Hoellen
- Department of Obstetrics and Gynecology, University Hospital of Schleswig-Holstein, University of Lübeck, Lübeck, Germany
| | - Andrea Schiefer
- Institute of Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Bonn, Germany.,German Center for Infection Research (DZIF), Partner Sites Bonn-Cologne/Hamburg-Lübeck-Borstel-Riems, Lübeck, Germany
| | - Beate Henrichfreise
- Institute of Pharmaceutical Microbiology, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Kenneth Pfarr
- Institute of Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Bonn, Germany.,German Center for Infection Research (DZIF), Partner Sites Bonn-Cologne/Hamburg-Lübeck-Borstel-Riems, Lübeck, Germany
| | - Achim Hoerauf
- Institute of Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Bonn, Germany.,German Center for Infection Research (DZIF), Partner Sites Bonn-Cologne/Hamburg-Lübeck-Borstel-Riems, Lübeck, Germany
| | - Kensuke Shima
- Department of Infectious Diseases and Microbiology, University of Lübeck, Lübeck, Germany
| | - Jan Rupp
- Department of Infectious Diseases and Microbiology, University of Lübeck, Lübeck, Germany.,German Center for Infection Research (DZIF), Partner Sites Bonn-Cologne/Hamburg-Lübeck-Borstel-Riems, Lübeck, Germany
| |
Collapse
|
17
|
Matsuo J, Sakai K, Okubo T, Yamaguchi H. Chlamydia pneumoniaeenhances Interleukin 8 (IL-8) production with reduced azithromycin sensitivity under hypoxia. APMIS 2019; 127:131-138. [DOI: 10.1111/apm.12924] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Accepted: 12/19/2018] [Indexed: 01/21/2023]
Affiliation(s)
- Junji Matsuo
- Department of Medical Laboratory Science; Faculty of Health Sciences; Hokkaido University; Sapporo Japan
| | - Kohei Sakai
- Department of Medical Laboratory Science; Faculty of Health Sciences; Hokkaido University; Sapporo Japan
| | - Torahiko Okubo
- Department of Medical Laboratory Science; Faculty of Health Sciences; Hokkaido University; Sapporo Japan
| | - Hiroyuki Yamaguchi
- Department of Medical Laboratory Science; Faculty of Health Sciences; Hokkaido University; Sapporo Japan
| |
Collapse
|
18
|
Lim YJ, Foo TC, Yeung AWS, Tu X, Ma Y, Hawkins CL, Witting PK, Jameson GNL, Terentis AC, Thomas SR. Human Indoleamine 2,3-Dioxygenase 1 Is an Efficient Mammalian Nitrite Reductase. Biochemistry 2019; 58:974-986. [PMID: 30585477 DOI: 10.1021/acs.biochem.8b01231] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The heme enzyme indoleamine 2,3-dioxygenase-1 (IDO1) catalyzes the first reaction of l-tryptophan oxidation along the kynurenine pathway. IDO1 is a central immunoregulatory enzyme with important implications for inflammation, infectious disease, autoimmune disorders, and cancer. Here we demonstrate that IDO1 is a mammalian nitrite reductase capable of chemically reducing nitrite to nitric oxide (NO) under hypoxia. Ultraviolet-visible absorption and resonance Raman spectroscopy showed that incubation of dithionite-reduced, ferrous-IDO1 protein (FeII-IDO1) with nitrite under anaerobic conditions resulted in the time-dependent formation of an FeII-nitrosyl IDO1 species, which was inhibited by substrate l-tryptophan, dependent on the concentration of nitrite or IDO1, and independent of the concentration of the reductant, dithionite. The bimolecular rate constant for IDO1 nitrite reductase activity was determined as 5.4 M-1 s-1 (pH 7.4, 23 °C), which was comparable to that measured for myoglobin (3.6 M-1 s-1; pH 7.4, 23 °C), an efficient and biologically important mammalian heme-based nitrite reductase. IDO1 nitrite reductase activity was pH-dependent but differed with myoglobin in that it showed a reduced proton dependency at pH >7. Electron paramagnetic resonance studies measuring NO production showed that the conventional IDO1 dioxygenase reducing cofactors, ascorbate and methylene blue, enhanced IDO1's nitrite reductase activity and the time- and IDO1 concentration-dependent release of NO in a manner inhibited by l-tryptophan or the IDO inhibitor 1-methyl-l-tryptophan. These data identify IDO1 as an efficient mammalian nitrite reductase that is capable of generating NO under anaerobic conditions. IDO1's nitrite reductase activity may have important implications for the enzyme's biological actions when expressed within hypoxic tissues.
Collapse
Affiliation(s)
| | - Timothy C Foo
- Department of Chemistry and Biochemistry , Florida Atlantic University , Boca Raton , Florida 33431 , United States
| | | | | | | | - Clare L Hawkins
- Department of Biomedical Sciences , University of Copenhagen , Copenhagen N DK-2200 , Denmark
| | - Paul K Witting
- Discipline of Pathology, Charles Perkins Centre, Faculty of Medicine and Health , University of Sydney , Sydney , NSW 2006 , Australia
| | - Guy N L Jameson
- School of Chemistry, Bio21 Molecular Science and Biotechnology Institute , The University of Melbourne , Parkville , VIC 3010 , Australia
| | - Andrew C Terentis
- Department of Chemistry and Biochemistry , Florida Atlantic University , Boca Raton , Florida 33431 , United States
| | | |
Collapse
|
19
|
Panzetta ME, Valdivia RH, Saka HA. Chlamydia Persistence: A Survival Strategy to Evade Antimicrobial Effects in-vitro and in-vivo. Front Microbiol 2018; 9:3101. [PMID: 30619180 PMCID: PMC6299033 DOI: 10.3389/fmicb.2018.03101] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 11/30/2018] [Indexed: 11/13/2022] Open
Abstract
The Chlamydiaceae comprise a group of highly adapted bacterial pathogens sharing a unique intracellular lifestyle. Three Chlamydia species are pathogenic to humans: Chlamydia trachomatis, Chlamydia pneumoniae, and Chlamydia psittaci. C. trachomatis is the leading bacterial cause of sexually-transmitted infections and infectious blindness worldwide. Chlamydia pneumoniae is a major cause of community-acquired atypical pneumonia. C. psittaci primarily affects psittacine birds and can be transmitted to humans causing psittacosis, a potentially fatal form of pneumonia. As opposed to other bacterial pathogens, the spread of clinically relevant antimicrobial resistance genes does not seem to be a major problem for the treatment of Chlamydia infections. However, when exposed to stressing conditions, like those arising from exposure to antimicrobial stimuli, these bacteria undergo a temporary interruption in their replication cycle and enter a viable but non-cultivable state known as persistence. When the stressing conditions are removed, Chlamydia resumes replication and generation of infectious particles. This review gives an overview of the different survival strategies used by Chlamydia to evade the deleterious effects of penicillin and IFNγ, with a focus on the different models used to study Chlamydia persistence, their contribution to elucidating the molecular basis of this complex phenomenon and their potential implications for studies in animal models of infection.
Collapse
Affiliation(s)
- Maria Emilia Panzetta
- CIBICI-CONICET, Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Raphael H. Valdivia
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, United States
| | - Hector Alex Saka
- CIBICI-CONICET, Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| |
Collapse
|
20
|
Shima K, Kaeding N, Ogunsulire IM, Kaufhold I, Klinger M, Rupp J. Interferon-γ interferes with host cell metabolism during intracellular Chlamydia trachomatis infection. Cytokine 2018; 112:95-101. [PMID: 29885991 DOI: 10.1016/j.cyto.2018.05.039] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Revised: 05/18/2018] [Accepted: 05/31/2018] [Indexed: 12/11/2022]
Abstract
Interferon-γ (IFN-γ) is a central mediator of host immune responses including T-cell differentiation and activation of macrophages for the control of bacterial pathogens. Anti-bacterial mechanisms of IFN-γ against the obligate intracellular bacteria Chlamydiatrachomatis in epithelial cells have been intensively investigated in the past, focusing on cellular tryptophan depletion by an IFN-γ induced expression of the indoleamine 2, 3-deoxygenase (IDO). In this study, we could show that IFN-γ treatment caused a significant reduction of the host cell glycolysis that was accompanied by a reduction of glucose transporter-1 (GLUT1) and hypoxia inducible factor-1α (HIF-1α) expression. Furthermore, C. trachomatis induced enhancement of glycolytic and mitochondrial activation were significantly suppressed by IFN-γ treatment. We could further show that glucose starvation, as observed under IFN-γ treatment, was associated with an attenuated antimicrobial efficacy of doxycycline (DOX) against C. trachomatis. In conclusions, anti-chlamydial activity of IFN-γ goes beyond tryptophan depletion including interference with cellular energy metabolism resulting reduced progeny, but also impaired antimicrobial susceptibility of C. trachomatis.
Collapse
Affiliation(s)
- Kensuke Shima
- Department of Infectious Diseases and Microbiology, University of Luebeck, Luebeck, Germany.
| | - Nadja Kaeding
- Department of Infectious Diseases and Microbiology, University of Luebeck, Luebeck, Germany
| | | | - Inga Kaufhold
- Department of Infectious Diseases and Microbiology, University of Luebeck, Luebeck, Germany
| | | | - Jan Rupp
- Department of Infectious Diseases and Microbiology, University of Luebeck, Luebeck, Germany; German Center for Infection Research (DZIF), Partner Site Hamburg-Luebeck-Borstel, Germany
| |
Collapse
|
21
|
Duan KM, Ma JH, Wang SY, Huang Z, Zhou Y, Yu H. The role of tryptophan metabolism in postpartum depression. Metab Brain Dis 2018; 33:647-660. [PMID: 29307018 DOI: 10.1007/s11011-017-0178-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Accepted: 12/26/2017] [Indexed: 01/01/2023]
Abstract
The Postpartum depression (PPD) is the most common postpartum psychiatric disorder, afflicting approximately 10%-20% of new mothers. Clinical symptoms of the PPD include depressive disorder, agitation, insomnia, anxiety and confusion, resulting in an increase in suicidal tendencies, thereby having significant impacts on the puerpera, newborn and their family. A growing body of data indicate a role for alterations in tryptophan metabolism in the PPD. The metabolism of tryptophan produces an array of crucial factors that can differentially regulate key physiological processes linked to the PPD. Importantly, an increase in stress hormones and immune-inflammatory activity drives tryptophan to the production of neuroregulatory kynurenine pathway products and away from the serotonin and melatonin pathways. This links the PPD to other disorders of depressed mood, which are classically associated with decreased serotonin and melatonin, coupled to increases in kynurenine pathway products. Several kynurenine pathway products, such as kynurenic acid and quinolinic acid, can have neuroregulatory effects, with consequences pathological underpinnings of the PPD. The current article reviews the role of alterations in tryptophan metabolism in the PPD.
Collapse
Affiliation(s)
- Kai-Ming Duan
- Department of Anesthesiology, Third Xiangya Hospital of Central South University, Changsha, 410013, People's Republic of China
| | - Jia-Hui Ma
- Department of Anesthesiology, Third Xiangya Hospital of Central South University, Changsha, 410013, People's Republic of China
| | - Sai-Ying Wang
- Department of Anesthesiology, Third Xiangya Hospital of Central South University, Changsha, 410013, People's Republic of China.
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, 410008, People's Republic of China.
- Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, 410078, People's Republic of China.
- Hunan Province Cooperation Innovation Center for Molecular Target New Drug Study, Hengyang, 421001, People's Republic of China.
| | - ZhengDong Huang
- Department of Anesthesiology, Third Xiangya Hospital of Central South University, Changsha, 410013, People's Republic of China
| | - YingYong Zhou
- Department of Anesthesiology, Third Xiangya Hospital of Central South University, Changsha, 410013, People's Republic of China
| | - HeYa Yu
- Department of Anesthesiology, Third Xiangya Hospital of Central South University, Changsha, 410013, People's Republic of China
| |
Collapse
|
22
|
Loeper N, Graspeuntner S, Rupp J. Microbiota changes impact on sexually transmitted infections and the development of pelvic inflammatory disease. Microbes Infect 2018; 20:505-511. [PMID: 29452257 DOI: 10.1016/j.micinf.2018.02.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 02/01/2018] [Accepted: 02/02/2018] [Indexed: 02/06/2023]
Abstract
The integrity of the human urogenital microbiome is crucial for women's health and well-being. An imbalance of the urogenital microbiota increases the risk for sexually transmitted infections. In this review, we discuss the microbiota composition of the female urogenital tract and its role in protecting from sexually transmitted infections and the emergence of pelvic inflammatory disease.
Collapse
Affiliation(s)
- Nathalie Loeper
- Department of Infectious Diseases and Microbiology, University of Luebeck, 23538 Luebeck, Germany
| | - Simon Graspeuntner
- Department of Infectious Diseases and Microbiology, University of Luebeck, 23538 Luebeck, Germany
| | - Jan Rupp
- Department of Infectious Diseases and Microbiology, University of Luebeck, 23538 Luebeck, Germany.
| |
Collapse
|
23
|
Shima K, Coopmeiners J, Graspeuntner S, Dalhoff K, Rupp J. Impact of micro-environmental changes on respiratory tract infections with intracellular bacteria. FEBS Lett 2016; 590:3887-3904. [PMID: 27509029 DOI: 10.1002/1873-3468.12353] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 07/29/2016] [Accepted: 08/05/2016] [Indexed: 12/26/2022]
Abstract
Community-acquired pneumonia is caused by intra- and extracellular bacteria, with some of these bacteria also being linked to the pathogenesis of chronic lung diseases, including asthma and chronic obstructive pulmonary disease. Chlamydia pneumoniae is an obligate intracellular pathogen that is highly sensitive to micro-environmental conditions controlling both pathogen growth and host immune responses. The availability of nutrients, as well as changes in oxygen, pH and interferon-γ levels, have been shown to directly influence the chlamydial life cycle and clearance. Although the lung has been traditionally regarded as a sterile environment, sequencing approaches have enabled the identification of a large number of bacteria in healthy and diseased lungs. The influence of the lung microbiota on respiratory infections has not been extensively studied so far and data on chlamydial infections are currently unavailable. In the present study, we speculate on how lung microbiota might interfere with acute and chronic infections by focusing exemplarily on the obligate intracellular C. pneumoniae. Furthermore, we consider changes in the gut microbiota as an additional player in the control of lung infections, especially in view the increasing evidence suggesting the involvement of the gut microbiota in various immunological processes throughout the human body.
Collapse
Affiliation(s)
- Kensuke Shima
- Department of Infectious Diseases and Microbiology, University of Lübeck, Germany
| | - Jonas Coopmeiners
- Department of Infectious Diseases and Microbiology, University of Lübeck, Germany
| | - Simon Graspeuntner
- Department of Infectious Diseases and Microbiology, University of Lübeck, Germany
| | - Klaus Dalhoff
- Medical Clinic III, University-Hospital Schleswig-Holstein/Campus Lübeck, Germany
| | - Jan Rupp
- Department of Infectious Diseases and Microbiology, University of Lübeck, Germany
| |
Collapse
|
24
|
Schefold JC, Fritschi N, Fusch G, Bahonjic A, Doehner W, von Haehling S, Pschowski R, Storm C, Schroeder T. Influence of core body temperature on Tryptophan metabolism, kynurenines, and estimated IDO activity in critically ill patients receiving target temperature management following cardiac arrest. Resuscitation 2016; 107:107-14. [PMID: 27565863 DOI: 10.1016/j.resuscitation.2016.07.239] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Revised: 07/22/2016] [Accepted: 07/26/2016] [Indexed: 12/21/2022]
Abstract
BACKGROUND/AIMS Temperature control improves neurological prognosis in comatose cardiac arrest (CA) survivors. Previous reports demonstrate that most affected patients show signs of significant systemic inflammation. In an effort to better characterize potential temperature-related effects on key inflammatory pathways, we investigate the course of Tryptophan (Trp) levels, Tryptophan catabolites (including kynurenines) and indoleamine-2,3-dioxygenase (IDO)-activity in post CA patients. MATERIAL/METHODS In an observational blinded endpoint analysis, a total of n=270 serial samples from 20 post CA patients (63.1±16.6 yrs., 45% shockable rhythm, mean time to return of spontaneous circulation (ROSC) 26.6±16.0min) treated with target temperature management (TTM) were analyzed. Core body temperatures, course of Trp, Trp catabolites (incl. kynurenines), and estimated IDO-activity were followed up for a maximum of 7 days after ROSC. Patients were followed up until hospital discharge or death and functional outcome was recorded. RESULTS Over the 7-day observational interval, marked changes in Trp serum levels and IDO-activity were noted. In general, Trp serum levels but not IDO-activity seemed to parallel with the course of core body temperature. In explorative analyses, a correlation of Trp (rho=0.271 (95%-CI: 0.16-0.38, p<0.0001) and IDO-activity (rho=-0.155, 95%-CI: -0.27 to -0.037, p=0.01) with core body temperature was observed. Linear mixed effect models revealed a positive significant association of core body temperature with Trp serum levels (Likelihood ratio test χ(2)=6.35, p=0.012). In patients with good (vs. unfavorable) outcome, a tendency toward higher Trp serum levels, lower IDO-activity, and lower Kynurenic acid levels was noted. CONCLUSIONS We observed significant changes in Trp catabolism and IDO-activity that appeared temperature associated in post CA patients. Under hypothermia, decreased serum levels of Trp and increased IDO-activity were noted. We speculate from our data that IDO-induction during hypothermia contributes to the previously described increased susceptibility to infection or sepsis under reduced temperatures.
Collapse
Affiliation(s)
- Joerg C Schefold
- Department of Intensive Care Medicine, Inselspital, Bern University Hospital, Bern, Switzerland; Department of Nephrology and Intensive Care Medicine, Charite University Medicine, Berlin, Germany
| | - Nora Fritschi
- Department of Intensive Care Medicine, Inselspital, Bern University Hospital, Bern, Switzerland
| | - Gerhard Fusch
- Department of Pediatrics, McMaster University, Hamilton, Canada
| | - Aldin Bahonjic
- Department of Pediatrics, McMaster University, Hamilton, Canada
| | - Wolfram Doehner
- Centre for Stroke Research Berlin, Charité Medical School, Berlin, Germany
| | - Stephan von Haehling
- University of Göttingen Medical School, Department of Cardiology and Pneumology, Göttingen, Germany
| | - Rene Pschowski
- Department of Nephrology and Intensive Care Medicine, Charite University Medicine, Berlin, Germany; Department of Gastroenterology, Charite University Medicine, Berlin, Germany
| | - Christian Storm
- Department of Nephrology and Intensive Care Medicine, Charite University Medicine, Berlin, Germany
| | - Tim Schroeder
- Department of Nephrology and Intensive Care Medicine, Charite University Medicine, Berlin, Germany
| |
Collapse
|
25
|
Negative Impact of Hypoxia on Tryptophan 2,3-Dioxygenase Function. Mediators Inflamm 2016; 2016:1638916. [PMID: 27563172 PMCID: PMC4985583 DOI: 10.1155/2016/1638916] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Revised: 06/14/2016] [Accepted: 06/26/2016] [Indexed: 02/01/2023] Open
Abstract
Tryptophan is an essential amino acid for hosts and pathogens. The liver enzyme tryptophan 2,3-dioxygenase (TDO) provokes, by its ability to degrade tryptophan to N-formylkynurenine, the precursor of the immune-relevant kynurenines, direct and indirect antimicrobial and immunoregulatory states. Up to now these TDO-mediated broad-spectrum effector functions have never been observed under hypoxia in vitro, although physiologic oxygen concentrations in liver tissue are low, especially in case of infection. Here we analysed recombinant expressed human TDO and ex vivo murine TDO functions under different oxygen conditions and show that TDO-induced restrictions of clinically relevant pathogens (bacteria, parasites) and of T cell proliferation are abrogated under hypoxic conditions. We pinpointed the loss of TDO efficiency to the reduction of TDO activity, since cell survival and TDO protein levels were unaffected. In conclusion, the potent antimicrobial as well as immunoregulatory effects of TDO were substantially impaired under hypoxic conditions that pathophysiologically occur in vivo. This might be detrimental for the appropriate host immune response towards relevant pathogens.
Collapse
|
26
|
Kaufmann E, Spohr C, Battenfeld S, De Paepe D, Holzhauser T, Balks E, Homolka S, Reiling N, Gilleron M, Bastian M. BCG Vaccination Induces Robust CD4+ T Cell Responses to Mycobacterium tuberculosis Complex–Specific Lipopeptides in Guinea Pigs. THE JOURNAL OF IMMUNOLOGY 2016; 196:2723-32. [DOI: 10.4049/jimmunol.1502307] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Accepted: 01/12/2016] [Indexed: 01/11/2023]
|
27
|
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
|
28
|
Jennewein J, Matuszak J, Walter S, Felmy B, Gendera K, Schatz V, Nowottny M, Liebsch G, Hensel M, Hardt WD, Gerlach RG, Jantsch J. Low-oxygen tensions found in Salmonella-infected gut tissue boost Salmonella replication in macrophages by impairing antimicrobial activity and augmenting Salmonella virulence. Cell Microbiol 2015; 17:1833-47. [PMID: 26104016 DOI: 10.1111/cmi.12476] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Revised: 05/17/2015] [Accepted: 06/18/2015] [Indexed: 12/28/2022]
Abstract
In Salmonella infection, the Salmonella pathogenicity island-2 (SPI-2)-encoded type three secretion system (T3SS2) is of key importance for systemic disease and survival in host cells. For instance, in the streptomycin-pretreated mouse model SPI-2-dependent Salmonella replication in lamina propria CD11c(-)CXCR1(-) monocytic phagocytes/macrophages (MΦ) is required for the development of colitis. In addition, containment of intracellular Salmonella in the gut critically depends on the antimicrobial effects of the phagocyte NADPH oxidase (PHOX), and possibly type 2 nitric oxide synthase (NOS2). For both antimicrobial enzyme complexes, oxygen is an essential substrate. However, the amount of available oxygen upon enteroinvasive Salmonella infection in the gut tissue and its impact on Salmonella-MΦ interactions was unknown. Therefore, we measured the gut tissue oxygen levels in a model of Salmonella enterocolitis using luminescence two-dimensional in vivo oxygen imaging. We found that gut tissue oxygen levels dropped from ∼78 Torr (∼11% O2) to values of ∼16 Torr (∼2% O2) during infection. Because in vivo virulence of Salmonella depends on the Salmonella survival in MΦ, Salmonella-MΦ interaction was analysed under such low oxygen values. These experiments revealed an increased intracellular replication and survival of wild-type and t3ss2 non-expressing Salmonella. These findings were paralleled by blunted nitric oxide and reactive oxygen species (ROS) production and reduced Salmonella ROS perception. In addition, hypoxia enhanced SPI-2 transcription and translocation of SPI-2-encoded virulence protein. Neither pharmacological blockade of PHOX and NOS2 nor impairment of T3SS2 virulence function alone mimicked the effect of hypoxia on Salmonella replication under normoxic conditions. However, if t3ss2 non-expressing Salmonella were used, hypoxia did not further enhance Salmonella recovery in a PHOX and NOS2-deficient situation. Hence, these data suggest that hypoxia-induced impairment of antimicrobial activity and Salmonella virulence cooperate to allow for enhanced Salmonella replication in MΦ.
Collapse
Affiliation(s)
- Jonas Jennewein
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Institut für Klinische Mikrobiologie und Hygiene, Universitätsklinikum Erlangen und Friedrich-Alexander Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany.,Institut für Klinische Mikrobiologie und Hygiene, Universitätsklinikum Regensburg und Universität Regensburg, Regensburg, Germany
| | - Jasmin Matuszak
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Institut für Klinische Mikrobiologie und Hygiene, Universitätsklinikum Erlangen und Friedrich-Alexander Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Steffi Walter
- Robert Koch-Institut, Bereich Wernigerode, Wernigerode, Germany
| | - Boas Felmy
- Institut für Mikrobiologie, ETH Zürich, Zürich, Switzerland
| | - Kathrin Gendera
- Robert Koch-Institut, Bereich Wernigerode, Wernigerode, Germany
| | - Valentin Schatz
- Institut für Klinische Mikrobiologie und Hygiene, Universitätsklinikum Regensburg und Universität Regensburg, Regensburg, Germany
| | - Monika Nowottny
- Institut für Klinische Mikrobiologie und Hygiene, Universitätsklinikum Regensburg und Universität Regensburg, Regensburg, Germany
| | | | - Michael Hensel
- Abteilung Mikrobiologie, Fachbereich Biologie / Chemie, Universität Osnabrück, Osnabrück, Germany
| | | | - Roman G Gerlach
- Robert Koch-Institut, Bereich Wernigerode, Wernigerode, Germany
| | - Jonathan Jantsch
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Institut für Klinische Mikrobiologie und Hygiene, Universitätsklinikum Erlangen und Friedrich-Alexander Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany.,Institut für Klinische Mikrobiologie und Hygiene, Universitätsklinikum Regensburg und Universität Regensburg, Regensburg, Germany
| |
Collapse
|
29
|
Abstract
IDO1 (indoleamine 2,3-dioxygenase 1) is a member of a unique class of mammalian haem dioxygenases that catalyse the oxidative catabolism of the least-abundant essential amino acid, L-Trp (L-tryptophan), along the kynurenine pathway. Significant increases in knowledge have been recently gained with respect to understanding the fundamental biochemistry of IDO1 including its catalytic reaction mechanism, the scope of enzyme reactions it catalyses, the biochemical mechanisms controlling IDO1 expression and enzyme activity, and the discovery of enzyme inhibitors. Major advances in understanding the roles of IDO1 in physiology and disease have also been realised. IDO1 is recognised as a prominent immune regulatory enzyme capable of modulating immune cell activation status and phenotype via several molecular mechanisms including enzyme-dependent deprivation of L-Trp and its conversion into the aryl hydrocarbon receptor ligand kynurenine and other bioactive kynurenine pathway metabolites, or non-enzymatic cell signalling actions involving tyrosine phosphorylation of IDO1. Through these different modes of biochemical signalling, IDO1 regulates certain physiological functions (e.g. pregnancy) and modulates the pathogenesis and severity of diverse conditions including chronic inflammation, infectious disease, allergic and autoimmune disorders, transplantation, neuropathology and cancer. In the present review, we detail the current understanding of IDO1’s catalytic actions and the biochemical mechanisms regulating IDO1 expression and activity. We also discuss the biological functions of IDO1 with a focus on the enzyme's immune-modulatory function, its medical implications in diverse pathological settings and its utility as a therapeutic target.
Collapse
|
30
|
Bhanothu V, Lakshmi V, Theophilus JP, Rozati R, Badhini P, Vijayalaxmi B. Investigation of Toll-Like Receptor-2 (2258G/A) and Interferon Gamma (+874T/A) Gene Polymorphisms among Infertile Women with Female Genital Tuberculosis. PLoS One 2015; 10:e0130273. [PMID: 26114934 PMCID: PMC4483232 DOI: 10.1371/journal.pone.0130273] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Accepted: 05/19/2015] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Toll-like receptor 2 (TLR2) and interferon-gamma (IFN-γ) coordinate with a diverse array of cellular programs through the transcriptional regulation of immunologically relevant genes and play an important role in immune system, reproductive physiology and basic pathology. Alterations in the functions of TLR2 2258G (guanine)/ A, IFN-γ (+874T/A) and signalling molecules that result from polymorphisms are often associated with susceptibility or resistance, which may, in turn, establish the innate host response to various infectious diseases. Presently, we proposed to investigate the risk of common single nucleotide polymorphism (SNP) of TLR2 and IFN-γ genes, for their effect on infertility in women with female genital tuberculosis (FGTB) and healthy women as controls. METHODOLOGY/PRINCIPAL FINDINGS Genotyping of TLR2 and IFN-γ gene polymorphisms was performed by amplification refractory mutation system multi-gene/multi-primer polymerase chain reaction followed by restriction fragment length polymorphism in 175 FGTB patients and 100 healthy control women (HCW). The TLR2 polymorphism [adenine (A) allele] was observed in 57.7 and 58.0% of FGTB patients and HCW, respectively. The IFN-γ (+874T/A) polymorphism (A allele) was significant in 74.3 and 71.0% of FGTB patients and HCW, respectively, while the odds ratios for the AA and TA genotypes for predisposition of FGTB were found to be 0.304 and 1.650 in HCW, respectively. The SNP of TLR2 was not associated with FGTB but the SNP of IFN-γ was found to be associated with mycobacteria infections and to induce infertility. CONCLUSIONS/SIGNIFICANCE At present, we hypothesize that infertile women with FGTB and HCW without tuberculosis (TB) have identical frequency of TLR variants, which may be adequate in the production of IFN-γ in response to Mycobacterium tuberculosis infections. Thus, the study appears to be the first of its kind reporting a mutation in the IFN-γ gene [+874 T (thymine) to A] responsible for susceptibility to TB infections and further inducing infertility.
Collapse
Affiliation(s)
- Venkanna Bhanothu
- Department of Zoology, University College of Science, Osmania University, Hyderabad, Telangana State, India
| | - Vemu Lakshmi
- Department of Microbiology, Nizam’s Institute of Medical Sciences, Hyderabad, Telangana State, India
| | - Jane P. Theophilus
- Department of Zoology, University College of Science, Osmania University, Hyderabad, Telangana State, India
| | - Roya Rozati
- Department of Obstetrics and Gynecology, Owaisi Hospital & Research Centre, Hyderabad, Telangana State, India
| | - Prabhakar Badhini
- Department of Genetics, University College of Science, Osmania University, Hyderabad, Telangana State, India
| | - Boda Vijayalaxmi
- Department of Obstetrics and Gynecology, Owaisi Hospital & Research Centre, Hyderabad, Telangana State, India
| |
Collapse
|
31
|
Kolawole AO, Hixon BP, Dameron LS, Chrisman IM, Smirnov VV. Catalytic activity of human indoleamine 2,3-dioxygenase (hIDO1) at low oxygen. Arch Biochem Biophys 2015; 570:47-57. [PMID: 25712221 PMCID: PMC4412315 DOI: 10.1016/j.abb.2015.02.014] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Revised: 02/12/2015] [Accepted: 02/15/2015] [Indexed: 11/30/2022]
Abstract
A cytokine-inducible extrahepatic human indoleamine 2,3-dioxygenase (hIDO1) catalyzes the first step of the kynurenine pathway. Immunosuppressive activity of hIDO1 in tumor cells weakens host T-cell immunity, contributing to the progression of cancer. Here we report on enzyme kinetics and catalytic mechanism of hIDO1, studied at varied levels of dioxygen (O2) and L-tryptophan (L-Trp). Using a cytochrome b5-based activating system, we measured the initial rates of O2 decay with a Clark-type oxygen electrode at physiologically-relevant levels of both substrates. Kinetics was also studied in the presence of two substrate analogs: 1-methyl-L-tryptophan and norharmane. Quantitative analysis supports a steady-state rather than a rapid equilibrium kinetic mechanism, where the rates of individual pathways, leading to a ternary complex, are significantly different, and the overall rate of catalysis depends on contributions of both routes. One path, where O2 binds to ferrous hIDO1 first, is faster than the second route, which starts with the binding of L-Trp. However, L-Trp complexation with free ferrous hIDO1 is more rapid than that of O2. As the level of L-Trp increases, the slower route becomes a significant contributor to the overall rate, resulting in observed substrate inhibition.
Collapse
Affiliation(s)
- Ayodele O Kolawole
- Department of Chemistry and Biochemistry, Center for Biomolecular Structure and Dynamics, University of Montana, Missoula, MT 59812, United States
| | - Brian P Hixon
- Department of Chemistry and Biochemistry, Center for Biomolecular Structure and Dynamics, University of Montana, Missoula, MT 59812, United States
| | - Laura S Dameron
- Department of Chemistry and Biochemistry, Center for Biomolecular Structure and Dynamics, University of Montana, Missoula, MT 59812, United States
| | - Ian M Chrisman
- Department of Chemistry and Biochemistry, Center for Biomolecular Structure and Dynamics, University of Montana, Missoula, MT 59812, United States
| | - Valeriy V Smirnov
- Department of Chemistry and Biochemistry, Center for Biomolecular Structure and Dynamics, University of Montana, Missoula, MT 59812, United States.
| |
Collapse
|
32
|
Inman RD. Reactive arthritis. Rheumatology (Oxford) 2015. [DOI: 10.1016/b978-0-323-09138-1.00112-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
|
33
|
Jantsch J, Schödel J. Hypoxia and hypoxia-inducible factors in myeloid cell-driven host defense and tissue homeostasis. Immunobiology 2014; 220:305-14. [PMID: 25439732 DOI: 10.1016/j.imbio.2014.09.009] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Revised: 08/01/2014] [Accepted: 09/05/2014] [Indexed: 02/08/2023]
Abstract
The impact of tissue oxygenation and hypoxia on immune cells has been recognized as a major determinant of host defense and tissue homeostasis. In this review, we will summarize the available data on tissue oxygenation in inflamed and infected tissue and the effect of low tissue oxygenation on myeloid cell function. Furthermore, we will highlight effects of the master regulators of the cellular hypoxic response, hypoxia-inducible transcription factors (HIF), in myeloid cells in antimicrobial defense and tissue homeostasis.
Collapse
Affiliation(s)
- Jonathan Jantsch
- Institut für Klinische Mikrobiologie und Hygiene, Universitätsklinikum Regensburg, Germany; Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen und Friedrich-Alexander Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany.
| | - Johannes Schödel
- Medizinische Klinik 4, Universitätsklinikum Erlangen und Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany; Translational Research Center, Universitätsklinikum Erlangen und Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany.
| |
Collapse
|
34
|
Häcker G, Heuer D, Ojcius DM. Is the hoopla over CPAF justified? Pathog Dis 2014; 72:1-2. [PMID: 25113036 DOI: 10.1111/2049-632x.12211] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Accepted: 08/04/2014] [Indexed: 01/19/2023] Open
Affiliation(s)
- Georg Häcker
- Institute of Medical Microbiology and Hygiene, University Medical Centre Freiburg, Freiburg, Germany.
| | | | | |
Collapse
|
35
|
Stavropoulos PG, Soura E, Kanelleas A, Katsambas A, Antoniou C. Reactive arthritis. J Eur Acad Dermatol Venereol 2014; 29:415-24. [PMID: 25199646 DOI: 10.1111/jdv.12741] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Accepted: 08/08/2014] [Indexed: 01/20/2023]
Abstract
Reactive arthritis (ReA) is an immune-mediated seronegative arthritis that belongs to the group of spondyloarthropathies and develops after a gastrointestinal or genitourinary system infection. The condition is considered to be characterized by a triad of symptoms (conjunctivitis, arthritis and urethritis) although a constellation of other manifestations may also be present. ReA is characterized by psoriasiform dermatological manifestations that may resemble those of pustular psoriasis and, similar to guttate psoriasis, is a post-infectious entity. Also, the articular manifestations of the disorder are similar to those of psoriatic arthritis and both conditions show a correlation with HLA-B27. These facts have led several authors to suggest that there is a connection between ReA and psoriasis, listing ReA among the disorders related to psoriasis. However, the pathogenetic mechanism behind the condition is complex and poorly understood. Bacterial antigenicity, the type of host response (i.e. Th1/Th2 imbalance) and various genetic factors (i.e. HLA-B27 etc.) play an important role in the development of the disorder. It is unknown whether all the aforementioned factors are part of a mechanism that could be similar to, or share basic aspects with known psoriasis pathogenesis mechanisms.
Collapse
Affiliation(s)
- P G Stavropoulos
- 1st Department of Dermatology/University Clinic, 'Andreas Syggros' Hospital, Athens, Greece
| | | | | | | | | |
Collapse
|
36
|
Aiyar A, Quayle AJ, Buckner LR, Sherchand SP, Chang TL, Zea AH, Martin DH, Belland RJ. Influence of the tryptophan-indole-IFNγ axis on human genital Chlamydia trachomatis infection: role of vaginal co-infections. Front Cell Infect Microbiol 2014; 4:72. [PMID: 24918090 PMCID: PMC4042155 DOI: 10.3389/fcimb.2014.00072] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Accepted: 05/15/2014] [Indexed: 11/13/2022] Open
Abstract
The natural history of genital Chlamydia trachomatis infections can vary widely; infections can spontaneously resolve but can also last from months to years, potentially progressing to cause significant pathology. The host and bacterial factors underlying this wide variation are not completely understood, but emphasize the bacterium's capacity to evade/adapt to the genital immune response, and/or exploit local environmental conditions to survive this immune response. IFNγ is considered to be a primary host protective cytokine against endocervical C.trachomatis infections. IFNγ acts by inducing the host enzyme indoleamine 2,3-dioxgenase, which catabolizes tryptophan, thereby depriving the bacterium of this essential amino acid. In vitro studies have revealed that tryptophan deprivation causes Chlamydia to enter a viable but non-infectious growth pattern that is termed a persistent growth form, characterized by a unique morphology and gene expression pattern. Provision of tryptophan can reactivate the bacterium to the normal developmental cycle. There is a significant difference in the capacity of ocular and genital C. trachomatis serovars to counter tryptophan deprivation. The latter uniquely encode a functional tryptophan synthase to synthesize tryptophan via indole salvage, should indole be available in the infection microenvironment. In vitro studies have confirmed the capacity of indole to mitigate the effects of IFNγ; it has been suggested that a perturbed vaginal microbiome may provide a source of indole in vivo. Consistent with this hypothesis, the microbiome associated with bacterial vaginosis includes species that encode a tryptophanase to produce indole. In this review, we discuss the natural history of genital chlamydial infections, morphological and molecular changes imposed by IFNγ on Chlamydia, and finally, the microenvironmental conditions associated with vaginal co-infections that can ameliorate the effects of IFNγ on C. trachomatis.
Collapse
Affiliation(s)
- Ashok Aiyar
- Department of Microbiology, Immunology, and Parasitology, 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
| | - Lyndsey R Buckner
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center New Orleans, LA, USA
| | - Shardulendra P Sherchand
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center New Orleans, LA, USA
| | - Theresa L Chang
- Department of Microbiology and Molecular Genetics, Public Health Research Institute Center, New Jersey Medical School-Rutgers, The State University of New Jersey Newark, NJ, USA
| | - Arnold H Zea
- Department of Microbiology, Immunology, and Parasitology, 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
| | - Robert J Belland
- Department of Microbiology, Immunology, and Biochemistry, University of Tennessee Health Sciences Center Memphis, TN, USA
| |
Collapse
|
37
|
Jerchel S, Kaufhold I, Schuchardt L, Shima K, Rupp J. Host immune responses after hypoxic reactivation of IFN-γ induced persistent Chlamydia trachomatis infection. Front Cell Infect Microbiol 2014; 4:43. [PMID: 24783060 PMCID: PMC3997002 DOI: 10.3389/fcimb.2014.00043] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Accepted: 03/25/2014] [Indexed: 01/05/2023] Open
Abstract
Genital tract infections with Chlamydia trachomatis (C. trachomatis) are the most frequent sexually transmitted disease worldwide. Severe clinical sequelae such as pelvic inflammatory disease (PID), tubal occlusion, and tubal infertility are linked to inflammatory processes of chronically infected tissues. The oxygen concentrations in the female urogenital tract are physiologically low and further diminished (0.5–5% O2, hypoxia) during an ongoing inflammation. However, little is known about the effect of a low oxygen environment on genital C. trachomatis infections. In this study, we investigated the host immune responses during reactivation of IFN-γ induced persistent C. trachomatis infection under hypoxia. For this purpose, the activation of the MAP-kinases p44/42 and p38 as well as the induction of the pro-inflammatory cytokines IL-1β, IL-6, IL-8, and MCP-1 were analyzed. Upon hypoxic reactivation of IFN-γ induced persistent C. trachomatis infection, the phosphorylation of the p44/42 but not of the p38 MAP-kinase was significantly diminished compared to IFN-γ induced chlamydial persistence under normoxic condition. In addition, significantly reduced IL-6 and IL-8 mRNA expression levels were observed for reactivated Chlamydiae under hypoxia compared to a persistent chlamydial infection under normoxia. Our findings indicate that hypoxia not only reactivates IFN-γ induced persistent C. trachomatis infections resulting in increased bacterial growth and progeny but also dampens inflammatory host immune signaling responses that are normally observed in a normoxic environment.
Collapse
Affiliation(s)
- Stefan Jerchel
- Institute of Medical Microbiology and Hygiene, University of Lübeck Lübeck, Germany
| | - Inga Kaufhold
- Institute of Medical Microbiology and Hygiene, University of Lübeck Lübeck, Germany
| | - Larissa Schuchardt
- Institute of Medical Microbiology and Hygiene, University of Lübeck Lübeck, Germany
| | - Kensuke Shima
- Institute of Medical Microbiology and Hygiene, University of Lübeck Lübeck, Germany
| | - Jan Rupp
- Institute of Medical Microbiology and Hygiene, University of Lübeck Lübeck, Germany ; Medical Clinic III/Infectious Diseases, University Hospital of Schleswig-Holstein Lübeck, Germany
| |
Collapse
|
38
|
Käding N, Szaszák M, Rupp J. Imaging of Chlamydia and host cell metabolism. Future Microbiol 2014; 9:509-21. [DOI: 10.2217/fmb.14.13] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
ABSTRACT: Chlamydial infections cause a wide range of acute and chronic diseases. Chlamydia trachomatis is the most common sexually transmitted bacterium while Chlamydia pneumoniae causes infections of the upper and lower respiratory tract. Chlamydia are obligate, intracellular bacteria with a biphasic developmental cycle that involves unique metabolic changes. Aside from entering an actively replicating state, Chlamydia may also implement persistent infections depending on different microenvironmental factors. In addition, changes in local oxygen availability and the composition of surrounding host microbiota are suggested to affect chlamydial growth and metabolism. Both bacteria and host cells endure characteristic metabolic changes during infection. Technical developments in recent years enable us to separately characterize chlamydial and host cell metabolism in living cells. This article focuses on novel approaches to analyze chlamydial metabolism such as NAD(P)H fluorescence lifetime imaging by two-photon microscopy. In addition, we provide an overview regarding promising future possibilities to further elucidate host–pathogen metabolic interactions.
Collapse
Affiliation(s)
- Nadja Käding
- Institute of Medical Microbiology & Hygiene, University of Lübeck, Ratzeburger Allee 160, 23538 Lübeck, Germany
| | - Márta Szaszák
- Institute of Medical Microbiology & Hygiene, University of Lübeck, Ratzeburger Allee 160, 23538 Lübeck, Germany
| | - Jan Rupp
- Institute of Medical Microbiology & Hygiene, University of Lübeck, Ratzeburger Allee 160, 23538 Lübeck, Germany
- Medical Clinic III/University Hospital Schleswig-Holstein, Campus Lübeck, Ratzeburger Allee 160, 23538 Lübeck, Germany
| |
Collapse
|
39
|
Cytomegalovirus infection impairs immunosuppressive and antimicrobial effector functions of human multipotent mesenchymal stromal cells. Mediators Inflamm 2014; 2014:898630. [PMID: 24782599 PMCID: PMC3981523 DOI: 10.1155/2014/898630] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Accepted: 02/10/2014] [Indexed: 12/31/2022] Open
Abstract
Human mesenchymal stromal cells (MSC) possess immunosuppressive and antimicrobial effects that are partly mediated by the tryptophan-catabolizing enzyme indoleamine-2,3-dioxygenase (IDO). Therefore MSC represent a promising novel cellular immunosuppressant which has the potential to control steroid-refractory acute graft versus host disease (GvHD). In addition, MSC are capable of reducing the risk of infection in patients after haematopoietic stem cell transplantation (HST). Recent data indicate that signals from the microenvironment including those from microbes may modulate MSC effector functions. As Cytomegalovirus (CMV) represents a prominent pathogen in immunocompromised hosts, especially in patients following HST, we investigated the impact of CMV infection on MSC-mediated effects on the immune system. We demonstrate that CMV-infected MSC lose their cytokine-induced immunosuppressive capacity and are no longer able to restrict microbial growth. IDO expression is substantially impaired following CMV infection of MSC and this interaction critically depends on intact virus and the number of MSC as well as the viral load. Since overt CMV infection may undermine the clinical efficacy of MSC in the treatment of GvHD in transplant patients, we recommend that patients scheduled for MSC therapy should undergo thorough evaluation for an active CMV infection and receive CMV-directed antiviral therapy prior to the administration of MSC.
Collapse
|
40
|
Omsland A, Sixt BS, Horn M, Hackstadt T. Chlamydial metabolism revisited: interspecies metabolic variability and developmental stage-specific physiologic activities. FEMS Microbiol Rev 2014; 38:779-801. [PMID: 24484402 DOI: 10.1111/1574-6976.12059] [Citation(s) in RCA: 91] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2013] [Revised: 01/08/2014] [Accepted: 01/13/2014] [Indexed: 01/07/2023] Open
Abstract
Chlamydiae are a group of obligate intracellular bacteria comprising important human and animal pathogens as well as symbionts of ubiquitous protists. They are characterized by a developmental cycle including two main morphologically and physiologically distinct stages, the replicating reticulate body and the infectious nondividing elementary body. In this review, we reconstruct the history of studies that have led to our current perception of chlamydial physiology, focusing on their energy and central carbon metabolism. We then compare the metabolic capabilities of pathogenic and environmental chlamydiae highlighting interspecies variability among the metabolically more flexible environmental strains. We discuss recent findings suggesting that chlamydiae may not live as energy parasites throughout the developmental cycle and that elementary bodies are not metabolically inert but exhibit metabolic activity under appropriate axenic conditions. The observed host-free metabolic activity of elementary bodies may reflect adequate recapitulation of the intracellular environment, but there is evidence that this activity is biologically relevant and required for extracellular survival and maintenance of infectivity. The recent discoveries call for a reconsideration of chlamydial metabolism and future in-depth analyses to better understand how species- and stage-specific differences in chlamydial physiology may affect virulence, tissue tropism, and host adaptation.
Collapse
Affiliation(s)
- Anders Omsland
- Host-Parasite Interactions Section, Laboratory of Intracellular Parasites, NIAID, NIH, Hamilton, MT, USA
| | | | | | | |
Collapse
|
41
|
Iannitti RG, Casagrande A, De Luca A, Cunha C, Sorci G, Riuzzi F, Borghi M, Galosi C, Massi-Benedetti C, Oury TD, Cariani L, Russo M, Porcaro L, Colombo C, Majo F, Lucidi V, Fiscarelli E, Ricciotti G, Lass-Flörl C, Ratclif L, Esposito A, De Benedictis FM, Donato R, Carvalho A, Romani L. Hypoxia promotes danger-mediated inflammation via receptor for advanced glycation end products in cystic fibrosis. Am J Respir Crit Care Med 2014; 188:1338-50. [PMID: 24127697 DOI: 10.1164/rccm.201305-0986oc] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
RATIONALE Hypoxia regulates the inflammatory-antiinflammatory balance by the receptor for advanced glycation end products (RAGE), a versatile sensor of damage-associated molecular patterns. The multiligand nature of RAGE places this receptor in the midst of chronic inflammatory diseases. OBJECTIVES To characterize the impact of the hypoxia-RAGE pathway on pathogenic airway inflammation preventing effective pathogen clearance in cystic fibrosis (CF) and elucidate the potential role of this danger signal in pathogenesis and therapy of lung inflammation. METHODS We used in vivo and in vitro models to study the impact of hypoxia on RAGE expression and activity in human and murine CF, the nature of the RAGE ligand, and the impact of RAGE on lung inflammation and antimicrobial resistance in fungal and bacterial pneumonia. MEASUREMENTS AND MAIN RESULTS Sustained expression of RAGE and its ligand S100B was observed in murine lung and human epithelial cells and exerted a proximal role in promoting inflammation in murine and human CF, as revealed by functional studies and analysis of the genetic variability of AGER in patients with CF. Both hypoxia and infections contributed to the sustained activation of the S100B-RAGE pathway, being RAGE up-regulated by hypoxia and S100B by infection by Toll-like receptors. Inhibiting the RAGE pathway in vivo with soluble (s) RAGE reduced pathogen load and inflammation in experimental CF, whereas sRAGE production was defective in patients with CF. CONCLUSIONS A causal link between hyperactivation of RAGE and inflammation in CF has been observed, such that targeting pathogenic inflammation alleviated inflammation in CF and measurement of sRAGE levels could be a useful biomarker for RAGE-dependent inflammation in patients with CF.
Collapse
Affiliation(s)
- Rossana G Iannitti
- 1 Department of Experimental Medicine and Biochemical Sciences, University of Perugia, Perugia, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Lima CBC, Santos SAD, Andrade Júnior DRD. Hypoxic stress, hepatocytes and CACO-2 viability and susceptibility to Shigella flexneri invasion. Rev Inst Med Trop Sao Paulo 2014; 55:341-6. [PMID: 24037289 PMCID: PMC4105072 DOI: 10.1590/s0036-46652013000500008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2012] [Accepted: 03/06/2013] [Indexed: 12/18/2022] Open
Abstract
SUMMARY Inflammation due to Shigella flexneri can cause damage to the colonic mucosa and cell death by necrosis and apoptosis. This bacteria can reach the bloodstream in this way, and the liver through portal veins. Hypoxia is a condition present in many human diseases, and it may induce bacterial translocation from intestinal lumen. We studied the ability of S. flexneri to invade rat hepatocytes and Caco-2 cells both in normoxic and hypoxic microenvironments, as well as morphological and physiological alterations in these cells after infection under hypoxia. We used the primary culture of rat hepatocytes as a model of study. We analyzed the following parameters in normoxic and hypoxic conditions: morphology, cell viability, bacterial recovery and lactate dehydrogenase (LDH) released. The results showed that there were fewer bacteria within the Caco-2 cells than in hepatocytes in normoxic and hypoxic conditions. We observed that the higher the multiplicity of infection (MOI) the greater the bacterial recovery in hepatocytes. The hypoxic condition decreased the bacterial recovery in hepatocytes. The cytotoxicity evaluated by LDH released by cells was significantly higher in cells submitted to hypoxia than normoxia. Caco-2 cells in normoxia released 63% more LDH than hepatocytes. LDH increased 164% when hepatocytes were submitted to hypoxia and just 21% when Caco-2 cells were in the same condition. The apoptosis evaluated by Tunel was significantly higher in cells submitted to hypoxia than normoxia. When comparing hypoxic cells, we obtained more apoptotic hepatocytes than apoptotic Caco-2 cells. Concluding our results contribute to a better knowledge of interactions between studied cells and Shigella flexneri. These data may be useful in the future to define strategies to combat this virulent pathogen.
Collapse
Affiliation(s)
- Camila Bárbara Cantalupo Lima
- Laboratory of Bacteriology (LIM 54), Department of Infectious Diseases, Faculty of Medicine, University of Sao Paulo, Sao PauloSP, Brazil, , ,
| | | | | |
Collapse
|
43
|
De Luca A, Carvalho A, Cunha C, Iannitti RG, Pitzurra L, Giovannini G, Mencacci A, Bartolommei L, Moretti S, Massi-Benedetti C, Fuchs D, De Bernardis F, Puccetti P, Romani L. IL-22 and IDO1 affect immunity and tolerance to murine and human vaginal candidiasis. PLoS Pathog 2013; 9:e1003486. [PMID: 23853597 PMCID: PMC3708875 DOI: 10.1371/journal.ppat.1003486] [Citation(s) in RCA: 101] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2012] [Accepted: 05/25/2013] [Indexed: 01/14/2023] Open
Abstract
The ability to tolerate Candida albicans, a human commensal of the gastrointestinal tract and vagina, implicates that host defense mechanisms of resistance and tolerance cooperate to limit fungal burden and inflammation at the different body sites. We evaluated resistance and tolerance to the fungus in experimental and human vulvovaginal candidiasis (VVC) as well as in recurrent VVC (RVVC). Resistance and tolerance mechanisms were both activated in murine VVC, involving IL-22 and IL-10-producing regulatory T cells, respectively, with a major contribution by the enzyme indoleamine 2,3-dioxygenase 1 (IDO1). IDO1 was responsible for the production of tolerogenic kynurenines, such that replacement therapy with kynurenines restored immunoprotection to VVC. In humans, two functional genetic variants in IL22 and IDO1 genes were found to be associated with heightened resistance to RVVC, and they correlated with increased local expression of IL-22, IDO1 and kynurenines. Thus, IL-22 and IDO1 are crucial in balancing resistance with tolerance to Candida, their deficiencies are risk factors for RVVC, and targeting tolerance via therapeutic kynurenines may benefit patients with RVVC.
Collapse
MESH Headings
- Animals
- Candida albicans/drug effects
- Candida albicans/immunology
- Candida albicans/isolation & purification
- Candidiasis, Vulvovaginal/genetics
- Candidiasis, Vulvovaginal/immunology
- Candidiasis, Vulvovaginal/metabolism
- Candidiasis, Vulvovaginal/microbiology
- Female
- Genetic Association Studies
- Genetic Variation
- Humans
- Immune Tolerance/drug effects
- Immunity, Mucosal/drug effects
- Immunologic Factors/metabolism
- Immunologic Factors/therapeutic use
- Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics
- Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism
- Interleukin-10/biosynthesis
- Interleukins/biosynthesis
- Interleukins/genetics
- Kynurenine/metabolism
- Kynurenine/therapeutic use
- Mice
- Mice, Inbred C57BL
- Mice, SCID
- Recurrence
- Severe Combined Immunodeficiency/drug therapy
- Severe Combined Immunodeficiency/immunology
- Severe Combined Immunodeficiency/physiopathology
- Specific Pathogen-Free Organisms
- T-Lymphocytes, Regulatory/drug effects
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- Interleukin-22
Collapse
Affiliation(s)
- Antonella De Luca
- Department of Experimental Medicine and Biochemical Sciences, University of Perugia, Perugia, Italy
- Istituto Superiore di Sanità, Roma, Italy, Italy
| | - Agostinho Carvalho
- Department of Experimental Medicine and Biochemical Sciences, University of Perugia, Perugia, Italy
| | - Cristina Cunha
- Department of Experimental Medicine and Biochemical Sciences, University of Perugia, Perugia, Italy
| | - Rossana G. Iannitti
- Department of Experimental Medicine and Biochemical Sciences, University of Perugia, Perugia, Italy
| | - Lucia Pitzurra
- Department of Experimental Medicine and Biochemical Sciences, University of Perugia, Perugia, Italy
- Microbiology, S. Maria della Misericordia Medical Center, Perugia, Italy
| | - Gloria Giovannini
- Department of Experimental Medicine and Biochemical Sciences, University of Perugia, Perugia, Italy
| | - Antonella Mencacci
- Department of Experimental Medicine and Biochemical Sciences, University of Perugia, Perugia, Italy
- Microbiology, S. Maria della Misericordia Medical Center, Perugia, Italy
| | - Lorenzo Bartolommei
- Department of Experimental Medicine and Biochemical Sciences, University of Perugia, Perugia, Italy
| | - Silvia Moretti
- Department of Experimental Medicine and Biochemical Sciences, University of Perugia, Perugia, Italy
| | - Cristina Massi-Benedetti
- Department of Experimental Medicine and Biochemical Sciences, University of Perugia, Perugia, Italy
| | | | | | - Paolo Puccetti
- Department of Experimental Medicine and Biochemical Sciences, University of Perugia, Perugia, Italy
| | - Luigina Romani
- Department of Experimental Medicine and Biochemical Sciences, University of Perugia, Perugia, Italy
| |
Collapse
|
44
|
Host metabolism promotes growth of Chlamydia pneumoniae in a low oxygen environment. Int J Med Microbiol 2013; 303:239-46. [DOI: 10.1016/j.ijmm.2013.03.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Revised: 03/22/2013] [Accepted: 03/30/2013] [Indexed: 12/14/2022] Open
|
45
|
Schmidt SK, Ebel S, Keil E, Woite C, Ernst JF, Benzin AE, Rupp J, Däubener W. Regulation of IDO activity by oxygen supply: inhibitory effects on antimicrobial and immunoregulatory functions. PLoS One 2013; 8:e63301. [PMID: 23675474 PMCID: PMC3652816 DOI: 10.1371/journal.pone.0063301] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2012] [Accepted: 04/02/2013] [Indexed: 01/06/2023] Open
Abstract
Tryptophan is an essential amino acid for human beings as well as for some microorganisms. In human cells the interferon-γ (IFN-γ) inducible enzyme indoleamine 2,3-dioxygenase (IDO) reduces local tryptophan levels and is therefore able to mediate broad-spectrum effector functions: IDO activity restricts the growth of various clinically relevant pathogens such as bacteria, parasites and viruses. On the other hand, it has been observed that IDO has immunoregulatory functions as it efficiently controls the activation and survival of T-cells. Although these important effects have been analysed in much detail, they have been observed in vitro using cells cultured in the presence of 20% O2 (normoxia). Such high oxygen concentrations are not present in vivo especially within infected and inflamed tissues. We therefore analysed IDO-mediated effects under lower oxygen concentrations in vitro and observed that the function of IDO is substantially impaired in tumour cells as well as in native cells. Hypoxia led to reduced IDO expression and as a result to reduced production of kynurenine, the downstream product of tryptophan degradation. Consequently, effector functions of IDO were abrogated under hypoxic conditions: in different human cell lines such as tumour cells (glioblastoma, HeLa) but also in native cells (human foreskin fibroblasts; HFF) IDO lost the capacity to inhibit the growth of bacteria (Staphylococcus aureus), parasites (Toxoplasma gondii) or viruses (herpes simplex virus type 1). Additionally, IDO could no longer efficiently control the proliferation of T-cells that have been co-cultured with IDO expressing HFF cells in vitro. In conclusion, the potent antimicrobial as well as immunoregulatory functions of IDO were substantially impaired under hypoxic conditions that pathophysiologically occurs in vivo.
Collapse
Affiliation(s)
- Silvia K. Schmidt
- Institute of Medical Microbiology and Hospital Hygiene, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Sebastian Ebel
- Institute of Medical Microbiology and Hospital Hygiene, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Eric Keil
- Institute of Medical Microbiology and Hospital Hygiene, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Claudia Woite
- Institute of Medical Microbiology and Hospital Hygiene, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Joachim F. Ernst
- Institute for Molecular Mycology, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Anika E. Benzin
- Institute of Medical Microbiology and Hygiene, University of Lübeck, Lübeck, Germany
| | - Jan Rupp
- Institute of Medical Microbiology and Hygiene, University of Lübeck, Lübeck, Germany
- Medical Clinic III/UK-SH, Campus Lübeck, Lübeck, Germany
| | - Walter Däubener
- Institute of Medical Microbiology and Hospital Hygiene, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
- * E-mail:
| |
Collapse
|
46
|
Activities of first-choice antimicrobials against gamma interferon-treated Chlamydia trachomatis differ in hypoxia. Antimicrob Agents Chemother 2013; 57:2828-30. [PMID: 23478971 DOI: 10.1128/aac.02211-12] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Gamma interferon (IFN-γ)-mediated host responses play a central role in resolving genital Chlamydia trachomatis infections but may also result in persistence of the pathogen, which shows reduced susceptibility to antimicrobials. The antichlamydial function of IFN-γ is oxygen dependent, and the efficacy of antimicrobials against C. trachomatis is reduced in a low-oxygen environment. In this study, we show that the antichlamydial efficacies of azithromycin and doxycycline differ in IFN-γ-treated cells under hypoxia.
Collapse
|
47
|
Jerchel S, Knebel G, König P, Bohlmann MK, Rupp J. A human fallopian tube model for investigation of C. trachomatis infections. J Vis Exp 2012:4036. [PMID: 22907315 PMCID: PMC3486760 DOI: 10.3791/4036] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Genital tract infections with Chlamydia trachomatis (C. trachomatis) are the most frequent transmitted sexually disease in women worldwide. Inefficient clearance or persistence of the pathogens may lead to ascending infections of the upper genital tract and are supposed to cause chronic inflammatory damage to infected tissues 1,2. As a consequence, severe clinical sequelae like pelvic inflammatory disease (PID), tubal occlusion and infertility may occur 3,4. Most of the research with C. trachomatis has been conducted in epithelial cell lines (e.g. HEp-2 cells and HeLa-229) or in mice. However, as with cell- culture based models, they do neither reflect the physiology of native tissue nor the pathophysiology of C. trachomatis genital tract infections in vivo5. Further limitations are given by the fact that central signaling cascades (e.g. IFN-γ mediated JAK/STAT signaling pathway) that control intracellular chlamydial growth fundamentally differ between mice and humans 6,7. We and others therefore established a whole organ fallopian tube model to investigate direct interactions between C. trachomatis and human fallopian tube cells ex vivo8,9. For this purpose, human fallopian tubes from women undergoing hysterectomy were collected and infected with C. trachomatis serovar D. Within 24 h post infection, specimen where analyzed using scanning electron microscopy (SEM) and transmission electron microscopy (TEM) to detect Chlamydia trachomatis mediated epithelial damage as well as C. trachomatis inclusion formation in the fallopian tissue.
Collapse
Affiliation(s)
- Stefan Jerchel
- Institute of Medical Microbiology and Hygiene, University of Lübeck
| | | | | | | | | |
Collapse
|
48
|
Shao R, Wang X, Wang W, Stener-Victorin E, Mallard C, Brännström M, Billig H. From mice to women and back again: causalities and clues for Chlamydia-induced tubal ectopic pregnancy. Fertil Steril 2012; 98:1175-85. [PMID: 22884019 DOI: 10.1016/j.fertnstert.2012.07.1113] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2012] [Revised: 07/12/2012] [Accepted: 07/12/2012] [Indexed: 01/12/2023]
Abstract
OBJECTIVE To provide an overview of knockout mouse models that have pathological tubal phenotypes after Chlamydia muridarum infection, discuss factors and pathological processes that contribute to inflammation, summarize data on tubal transport and progression of tubal implantation from studies in humans and animal models, and highlight research questions in the field. DESIGN A search of the relevant literature using PubMed and other online tools. SETTING University-based preclinical and clinical research laboratories. PATIENT(S) Women with tubal ectopic pregnancy after Chlamydia trachomatis infection. INTERVENTION(S) None. MAIN OUTCOME MEASURE(S) Critical review of the literature. RESULT(S) Chlamydia trachomatis infection poses a major threat to human reproduction. Biological and epidemiological evidence suggests that progression of Chlamydia infection causes intense and persistent inflammation, injury, and scarring in the fallopian tube, leading to a substantially increased risk of ectopic pregnancy and infertility. The main targets of Chlamydia infection are epithelial cells lining the mucosal surface, which play a central role in host immune responses and pathophysiology. Tubal phenotypes at the cellular level in mutant mice appear to reflect alterations in the balance between inflammatory mediator and factor deficiency. While studies in mice infected with Chlamydia muridarum have provided insight into potential inflammatory mediators linked to fallopian tube pathology, it is unclear how inflammation induced by Chlamydia infection prevents or retards normal tubal transport and causes embryo implantation in the fallopian tube. CONCLUSION(S) Given the similarities in the tubal physiology of humans and rodents, knockout mouse models can be used to study certain aspects of tubal functions, such as gamete transport and early embryo implantation. Elucidation of the exact molecular mechanisms of immune and inflammatory responses caused by Chlamydia infection in human fallopian tubal cells in vitro and understanding how Chlamydia infection affects tubal transport and implantation in animal studies in vivo may explain how Chlamydia trachomatis infection drives inflammation and develops the tubal pathology in women with tubal ectopic pregnancy.
Collapse
Affiliation(s)
- Ruijin Shao
- Department of Physiology/Endocrinology, Institute of Neuroscience and Physiology, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden.
| | | | | | | | | | | | | |
Collapse
|
49
|
Nickel D, Busch M, Mayer D, Hagemann B, Knoll V, Stenger S. Hypoxia Triggers the Expression of Human β Defensin 2 and Antimicrobial Activity againstMycobacterium tuberculosisin Human Macrophages. THE JOURNAL OF IMMUNOLOGY 2012; 188:4001-7. [DOI: 10.4049/jimmunol.1100976] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
50
|
Abstract
Reactive arthritis (ReA), an inflammatory arthritic condition that is commonly associated with Chlamydia infections, represents a significant health burden, yet is poorly understood. The enigma of this disease is reflected in its problematic name and in its ill-defined pathogenesis. The existence of persistent pathogens in the arthritic joint is acknowledged, but their relevance remains elusive. Progress is being made in understanding the underlying mechanisms of ReA, whereby an imbalance between type 1 and type 2 immune responses seems to be critical in determining susceptibility to disease. Such an imbalance occurs prior to the initiation of an adaptive immune response, suggesting that innate cellular and molecular mechanisms in ReA should be prioritized as fruitful areas for investigation.
Collapse
|