1
|
McCullough A, Huang S, Weber MM. Pathogenicity and virulence of Chlamydia trachomatis: Insights into host interactions, immune evasion, and intracellular survival. Virulence 2025; 16:2503423. [PMID: 40353442 PMCID: PMC12090877 DOI: 10.1080/21505594.2025.2503423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 04/22/2025] [Accepted: 05/03/2025] [Indexed: 05/14/2025] Open
Abstract
Chlamydia trachomatis is an obligate intracellular pathogen and the leading cause of bacterial sexually transmitted infections and infectious blindness worldwide. All Chlamydia species share a unique biphasic developmental cycle, alternating between infectious elementary bodies (EBs) and replicative reticulate bodies (RBs). The pathogenesis of C. trachomatis is driven by a sophisticated arsenal of adhesins, conventional type III secretion system effector proteins, and inclusion membrane proteins that subvert host cellular processes to establish infection and promote survival. In this review, we highlight the molecular mechanisms underlying C. trachomatis infection, focusing on key stages of its developmental cycle, including adhesion, invasion, replication, and egress. We delve into its interactions with host cytoskeletal structures, immune signaling pathways, and intracellular trafficking systems, as well as its strategies for immune evasion and persistence. Understanding these mechanisms offers critical insights into C. trachomatis pathogenesis and identifies promising avenues for therapeutic and vaccine development.
Collapse
Affiliation(s)
- Alix McCullough
- Department of Microbiology and Immunology, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Steven Huang
- Department of Microbiology and Immunology, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Mary M. Weber
- Department of Microbiology and Immunology, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| |
Collapse
|
2
|
Kintner J, Callaghan M, Bulawa L, Chu A, Ma Z, Williams DL, Schoborg RV, Kruppa MD, Hall JV. Dectin-1 stimulating β-glucans inhibit Chlamydia infections both in vitro and in vivo. Pathog Dis 2025; 83:ftaf002. [PMID: 39886876 PMCID: PMC11840957 DOI: 10.1093/femspd/ftaf002] [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: 07/29/2024] [Revised: 01/03/2025] [Accepted: 01/27/2025] [Indexed: 02/01/2025] Open
Abstract
Chlamydia trachomatis and Candida albicans are common inhabitants of the female genital tract. Candida albicans can impact the viability and pathogenesis of some bacteria. Previously, we investigated physical interactions between Ch. trachomatis elementary bodies (EBs) and Ca. albicans. This work indicated that EBs bind to Ca. albicans and become noninfectious by 24 h post-binding. Here, we continue our investigation of these interkingdom, polymicrobial interactions. Candida albicans adheres to bacteria or host surfaces via agglutinin-like sequence or heat shock 70 (Ssa) proteins. Chlamydia trachomatis EBs did not bind Ca. albicans Ssa2 deficient strains as efficiently as wild-type or complemented strains, indicating a role for this protein in chlamydial adherence to Candida. Additionally, Ca. albicans β-glucans inhibit chlamydial infection when exposure occurs during EB adsorption onto cervical cells. Laminarin, a β-glucan agonist of the C-type lectin receptor Dectin-1, inhibited chlamydial infection in both cervical epithelial cells and mice when exposure occurred prior to, during, or immediately following EB inoculation. Conversely, a Dectin-1 antagonist laminarin did not inhibit infection in vitro, suggesting that β-glucan inhibition of Ch. trachomatis requires C-type lectin receptor signaling. Overall, our data demonstrate that β-glucans from multiple species, including Ca. albicans, inhibit Chlamydia via stimulation of host-signaling pathways.
Collapse
Affiliation(s)
- Jennifer Kintner
- Department of Biomedical Sciences, East Tennessee State University, Johnson City, TN 37614, USA
- Center for Infectious Disease, Inflammation and Immunity, East Tennessee State University, Johnson City, TN 37614, USA
| | - Morgan Callaghan
- Department of Biomedical Sciences, East Tennessee State University, Johnson City, TN 37614, USA
| | - Lillith Bulawa
- Department of Biomedical Sciences, East Tennessee State University, Johnson City, TN 37614, USA
| | - Angela Chu
- Department of Biomedical Sciences, East Tennessee State University, Johnson City, TN 37614, USA
| | - Zuchao Ma
- Center for Infectious Disease, Inflammation and Immunity, East Tennessee State University, Johnson City, TN 37614, USA
- Department of Surgery, East Tennessee State University, Johnson City, TN 37614, USA
| | - David L Williams
- Center for Infectious Disease, Inflammation and Immunity, East Tennessee State University, Johnson City, TN 37614, USA
- Department of Surgery, East Tennessee State University, Johnson City, TN 37614, USA
| | - Robert V Schoborg
- Center for Infectious Disease, Inflammation and Immunity, East Tennessee State University, Johnson City, TN 37614, USA
- Department of Medical Education at Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA
| | - Michael D Kruppa
- Department of Biomedical Sciences, East Tennessee State University, Johnson City, TN 37614, USA
- Center for Infectious Disease, Inflammation and Immunity, East Tennessee State University, Johnson City, TN 37614, USA
| | - Jennifer V Hall
- Department of Biomedical Sciences, East Tennessee State University, Johnson City, TN 37614, USA
- Center for Infectious Disease, Inflammation and Immunity, East Tennessee State University, Johnson City, TN 37614, USA
| |
Collapse
|
3
|
Tang X, Cui Y, Namarra U, Tian X, Rivas-Giorgi F, Fikrig E. Dual roles for a tick protein disulfide isomerase during the life cycle of the Lyme disease agent. mBio 2024; 15:e0175424. [PMID: 39470213 PMCID: PMC11633212 DOI: 10.1128/mbio.01754-24] [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: 06/09/2024] [Accepted: 09/27/2024] [Indexed: 10/30/2024] Open
Abstract
The protein disulfide isomerase (PDI) family is a group of enzymes that have thiol-disulfide oxidoreductase, disulfide isomerase, and redox-dependent chaperone activities. PDIs facilitate diverse infections in mammalian hosts by directly binding to pathogens, immunomodulation, or enabling microbial invasion of host cells. PDI homologs within pathogens are also potential virulence factors. However, whether PDIs within blood-feeding ticks influence microbial infection remains unknown. In this study, we investigated the role of Ixodes scapularis PDIs, on the Lyme disease agent, Borrelia burgdorferi. I. scapularis has five PDIs (IsPDIs), and IsPDIA6 gene expression is reduced upon B. burgdorferi infection in the tick. IsPDIA6-mediated trypsin inhibitor gene expression contributes to B. burgdorferi colonization within the tick midgut. IsPDIA6 is also secreted into the host during tick feeding, alters cytokine/chemokine expression at the tick bite site, and influences the initial stage of bacterial infection in mice. These data demonstrate that a PDI from a blood-feeding vector plays a role in the life cycle of an extracellular pathogen. IMPORTANCE Vector-borne diseases are a leading cause of death and illness worldwide, and more than 80% of the global population live in areas at risk from at least one major vector-borne disease. In this study, we demonstrate a dual role of a specific Ixodes tick protein disulfide isomerase (PDI) in inhibiting the ability of the Lyme disease agent to colonize ticks and also in enhancing the initial stage of spirochete infection of mice. This study represents a novel conceptual advancement that a PDI from a blood-feeding vector plays important roles in the life cycle of an extracellular pathogen.
Collapse
Affiliation(s)
- Xiaotian Tang
- Zhejiang Key Laboratory of Biology and Ecological Regulation of Crop Pathogens and Insects, Institute of Insect Sciences, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, China
- Section of Infectious Diseases, Department of Internal Medicine, School of Medicine, Yale University, New Haven, Connecticut, USA
| | - Yingjun Cui
- Section of Infectious Diseases, Department of Internal Medicine, School of Medicine, Yale University, New Haven, Connecticut, USA
| | - Ushuu Namarra
- History of Science, Medicine, and Public Health Program, Yale College, New Haven, Connecticut, USA
| | - Xiuqi Tian
- Section of Infectious Diseases, Department of Internal Medicine, School of Medicine, Yale University, New Haven, Connecticut, USA
| | - Freddie Rivas-Giorgi
- Molecular Biochemistry and Biophysics Program, Yale College, New Haven, Connecticut, USA
| | - Erol Fikrig
- Section of Infectious Diseases, Department of Internal Medicine, School of Medicine, Yale University, New Haven, Connecticut, USA
| |
Collapse
|
4
|
Wang J, Wang B, Xiao J, Chen Y, Wang C. Chlamydia psittaci: A zoonotic pathogen causing avian chlamydiosis and psittacosis. Virulence 2024; 15:2428411. [PMID: 39541409 PMCID: PMC11622591 DOI: 10.1080/21505594.2024.2428411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 10/07/2024] [Accepted: 11/06/2024] [Indexed: 11/16/2024] Open
Abstract
Chlamydia psittaci is an obligate intracellular gram-negative bacterium with a unique biphasic developmental cycle. It is a zoonotic pathogen with a wide range of hosts and can cause avian chlamydiosis in birds and psittacosis in humans. The pathogen is transmitted mainly through horizontal transmission between birds. Cross-species transmission sometimes occurs and human-to-human transmission has recently been confirmed. This review provides an updated overview of C. psittaci from the perspective of both avian chlamydiosis and psittacosis. We include the aspects of genotype, host-pathogen interaction, transmission, epidemiology, detection and diagnosis, clinical manifestation, management, and prevention, aiming to provide a basic understanding of C. psittaci and offer fresh insights focused on zoonosis and cross-species transmission.
Collapse
Affiliation(s)
- Jiewen Wang
- Institute of Pathogenic Biology, School of Basic Medicine, Hengyang Medical College, University of South China; Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hengyang, Hunan, China
- Institute of Cell and Genetics, School of Basic Medicine, Hengyang Medical College, University of South China, Hengyang, Hunan, China
| | - Buwei Wang
- Institute of Pathogenic Biology, School of Basic Medicine, Hengyang Medical College, University of South China; Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hengyang, Hunan, China
| | - Jian Xiao
- The Affiliated Nanhua Hospital, Department of laboratory medicine, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Yuqing Chen
- Clinical Microbiology Laboratory, Xiangtan Central Hospital, Xiangtan, Hunan, China
| | - Chuan Wang
- Institute of Pathogenic Biology, School of Basic Medicine, Hengyang Medical College, University of South China; Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hengyang, Hunan, China
| |
Collapse
|
5
|
Reigada I, Kapp K, Kaudela T, García Soria M, Oksanen T, Hanski L. Tracking Chlamydia - Host interactions and antichlamydial activity in Caenorhabditis elegans. Biomed Pharmacother 2024; 177:116956. [PMID: 38901202 DOI: 10.1016/j.biopha.2024.116956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 06/02/2024] [Accepted: 06/15/2024] [Indexed: 06/22/2024] Open
Abstract
The fading efficacy of antibiotics is a growing global health concern due to its life-threatening consequences and increased healthcare costs. Non-genetic mechanisms of antimicrobial resistance, such as those employed by Chlamydia pneumoniae and Chlamydia trachomatis, complicate treatment as these bacteria can enter a non-replicative, persistent state under stress, evading antibiotics and linking to inflammatory conditions. Understanding chlamydial persistence at the molecular level is challenging, and new models for studying Chlamydia-host interactions in vivo are urgently needed. Caenorhabditis elegans offers an alternative given its immune system and numerous orthologues of human genes. This study established C. elegans as an in vivo model for chlamydial infection. Both Chlamydia species reduced the worm's lifespan, their DNA being detectable at three- and six-days post-infection. Azithromycin at its MIC (25 nM) failed to prevent the infection-induced lifespan reduction, indicating a persister phenotype. In contrast, the methanolic extract of Schisandra chinensis berries showed anti-chlamydial activity both in vitro (in THP-1 macrophages) and in vivo, significantly extending the lifespan of infected C. elegans and reducing the bacterial load. Moreover, S. chinensis increased the transcriptional activity of SKN-1 in the worms, but was unable to impact the bacterial load or lifespan in a sek-1 defective C. elegans strain. In summary, this study validated C. elegans as a chlamydial infection model and showcased S. chinensis berries' in vivo anti-chlamydial potential, possibly through SEK/SKN-1 signaling modulation.
Collapse
Affiliation(s)
- Inés Reigada
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki 00014, Finland
| | - Karmen Kapp
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki 00014, Finland
| | - Theresa Kaudela
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki 00014, Finland
| | - María García Soria
- Department of Pharmacy, Faculty of Health Sciences, Universidad San Jorge (San Jorge University), Zaragoza 50830, Spain
| | - Timo Oksanen
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki 00014, Finland
| | - Leena Hanski
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki 00014, Finland.
| |
Collapse
|
6
|
Hart TM, Sonnert ND, Tang X, Chaurasia R, Allen PE, Hunt JR, Read CB, Johnson EE, Arora G, Dai Y, Cui Y, Chuang YM, Yu Q, Rahman MS, Mendes MT, Rolandelli A, Singh P, Tripathi AK, Ben Mamoun C, Caimano MJ, Radolf JD, Lin YP, Fingerle V, Margos G, Pal U, Johnson RM, Pedra JHF, Azad AF, Salje J, Dimopoulos G, Vinetz JM, Carlyon JA, Palm NW, Fikrig E, Ring AM. An atlas of human vector-borne microbe interactions reveals pathogenicity mechanisms. Cell 2024; 187:4113-4127.e13. [PMID: 38876107 PMCID: PMC11959484 DOI: 10.1016/j.cell.2024.05.023] [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: 02/18/2023] [Revised: 01/15/2024] [Accepted: 05/13/2024] [Indexed: 06/16/2024]
Abstract
Vector-borne diseases are a leading cause of death worldwide and pose a substantial unmet medical need. Pathogens binding to host extracellular proteins (the "exoproteome") represents a crucial interface in the etiology of vector-borne disease. Here, we used bacterial selection to elucidate host-microbe interactions in high throughput (BASEHIT)-a technique enabling interrogation of microbial interactions with 3,324 human exoproteins-to profile the interactomes of 82 human-pathogen samples, including 30 strains of arthropod-borne pathogens and 8 strains of related non-vector-borne pathogens. The resulting atlas revealed 1,303 putative interactions, including hundreds of pairings with potential roles in pathogenesis, including cell invasion, tissue colonization, immune evasion, and host sensing. Subsequent functional investigations uncovered that Lyme disease spirochetes recognize epidermal growth factor as an environmental cue of transcriptional regulation and that conserved interactions between intracellular pathogens and thioredoxins facilitate cell invasion. In summary, this interactome atlas provides molecular-level insights into microbial pathogenesis and reveals potential host-directed targets for next-generation therapeutics.
Collapse
Affiliation(s)
- Thomas M Hart
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06510, USA
| | - Nicole D Sonnert
- Department of Immunobiology, Yale School of Medicine, New Haven, CT 06510, USA; Department of Microbial Pathogenesis, Yale School of Medicine, New Haven, CT 06510, USA
| | - Xiaotian Tang
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06510, USA
| | - Reetika Chaurasia
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06510, USA
| | - Paige E Allen
- Department of Microbiology and Immunology, Virginia Commonwealth University School of Medicine, Richmond, VA 23298, USA
| | - Jason R Hunt
- Department of Microbiology and Immunology, Virginia Commonwealth University School of Medicine, Richmond, VA 23298, USA
| | - Curtis B Read
- Department of Microbiology and Immunology, Virginia Commonwealth University School of Medicine, Richmond, VA 23298, USA
| | - Emily E Johnson
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06510, USA; Department of Epidemiology and Microbial Diseases, Yale School of Public Health, New Haven, CT 06510, USA
| | - Gunjan Arora
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06510, USA
| | - Yile Dai
- Department of Immunobiology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Yingjun Cui
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06510, USA
| | - Yu-Min Chuang
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06510, USA
| | - Qian Yu
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06510, USA
| | - M Sayeedur Rahman
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - M Tays Mendes
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Agustin Rolandelli
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Pallavi Singh
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06510, USA
| | - Abhai K Tripathi
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Choukri Ben Mamoun
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06510, USA; Department of Microbial Pathogenesis, Yale School of Medicine, New Haven, CT 06510, USA
| | - Melissa J Caimano
- Department of Medicine, UConn Health, Farmington, CT 06030, USA; Department of Pediatrics, UConn Health, Farmington, CT 06030, USA; Department of Molecular Biology and Biophysics, UConn Health, Farmington, CT 06030, USA
| | - Justin D Radolf
- Department of Medicine, UConn Health, Farmington, CT 06030, USA; Department of Pediatrics, UConn Health, Farmington, CT 06030, USA; Department of Molecular Biology and Biophysics, UConn Health, Farmington, CT 06030, USA; Department of Genetics and Genome Sciences, UConn Health, Farmington, CT 06030, USA; Department of Immunology, UConn Health, Farmington, CT 06030, USA
| | - Yi-Pin Lin
- Division of Infectious Diseases, Wadsworth Center, New York State Department of Health, Albany, NY 12201, USA
| | - Volker Fingerle
- Bavarian Health and Food Safety Authority, Oberschleißheim, Munich 85764, Bavaria, Germany
| | - Gabriele Margos
- Bavarian Health and Food Safety Authority, Oberschleißheim, Munich 85764, Bavaria, Germany
| | - Utpal Pal
- Department of Veterinary Medicine, University of Maryland, College Park, MD 20742, USA
| | - Raymond M Johnson
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06510, USA; Department of Microbial Pathogenesis, Yale School of Medicine, New Haven, CT 06510, USA
| | - Joao H F Pedra
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Abdu F Azad
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Jeanne Salje
- Department of Pathology, University of Cambridge, Cambridge CB2 1TN, UK; Department of Biochemistry, University of Cambridge, Cambridge CB2 1TN, UK
| | - George Dimopoulos
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Joseph M Vinetz
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06510, USA; Laboratorio ICEMR-Amazonia, Laboratorios de Investigación Y Desarrollo, Facultad de Ciencias Y Filosofia, Universidad Peruana Cayetano Heredia, Lima 15102, Peru; Instituto de Medicina Tropical Alexander Von Humboldt, Universidad Peruana Cayetano Heredia, Lima 15102, Peru
| | - Jason A Carlyon
- Department of Microbiology and Immunology, Virginia Commonwealth University School of Medicine, Richmond, VA 23298, USA.
| | - Noah W Palm
- Department of Immunobiology, Yale School of Medicine, New Haven, CT 06510, USA.
| | - Erol Fikrig
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06510, USA.
| | - Aaron M Ring
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA 98102, USA.
| |
Collapse
|
7
|
Kanaan H, Chapalain A, Chokr A, Doublet P, Gilbert C. Legionella pneumophila cell surface RtxA release by LapD/LapG and its role in virulence. BMC Microbiol 2024; 24:266. [PMID: 39026145 PMCID: PMC11264772 DOI: 10.1186/s12866-024-03395-1] [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: 07/06/2023] [Accepted: 06/20/2024] [Indexed: 07/20/2024] Open
Abstract
BACKGROUND Legionella pneumophila is a Gram-negative intracellular bacillus and is the causative agent of a severe form of pneumonia called Legionnaires' disease which accounts for 2-9% of cases of community acquired pneumonia. It produces an extremely large protein belonging to the RTX (Repeats in ToXin) family, called RtxA, and we previously reported that RtxA is transported by a dedicated type 1 secretion system (T1SS) to the cell surface. RTX proteins have been shown to participate in the virulence or biofilm formation of various bacteria, the most studied models being the pore forming hemolysin A (HlyA) of Escherichia coli and the biofilm associated protein LapA of P. fluorescens. LapA localization depends on the enzymatic release by LapD/LapG complex activity. This study aimed to elucidate the dual localization (cell surface associated or released state) of L. pneumophila RTX protein (RtxA) and whether this released versus sequestered state of RtxA plays a role in L. pneumophila virulence. RESULTS The hereby work reveals that, in vitro, LapG periplasmic protease cleaves RtxA N-terminus in the middle of a di-alanine motif (position 108-109). Consistently, a strain lacking LapG protease maintains RtxA on the cell surface, whereas a strain lacking the c-di-GMP receptor LapD does not exhibit cell surface RtxA because of its continuous cleavage and release, as in the LapA-D-G model of Pseudomonas fluorescens. Interestingly, our data point out a key role of RtxA in enhancing the infection process of amoeba cells, regardless of its location (embedded or released); therefore, this may be the result of a secondary role of this surface protein. CONCLUSIONS This is the first experimental identification of the cleavage site within the RTX protein family. The primary role of RtxA in Legionella is still questionable as in many other bacterial species, hence it sounds reasonable to propose a major function in biofilm formation, promoting cell aggregation when RtxA is embedded in the outer membrane and facilitating biofilm dispersion in case of RtxA release. The role of RtxA in enhancing the infection process may be a result of its action on host cells (i.e., PDI interaction or pore-formation), and independently of its status (embedded or released).
Collapse
Affiliation(s)
- Hussein Kanaan
- Centre International de Recherche en Infectiologie (CIRI), Université Lyon 1, INSERM U1111, CNRS UMR5308, ENS, Lyon Bât. Rosalind Franklin, 50 avenue Tony Garnier, Lyon, 69007, France
- Research Laboratory of Microbiology (RLM), Department of Life and Earth Sciences, Faculty of Sciences I, Lebanese University, Hadat Campus, Beirut, Lebanon
| | - Annelise Chapalain
- Centre International de Recherche en Infectiologie (CIRI), Université Lyon 1, INSERM U1111, CNRS UMR5308, ENS, Lyon Bât. Rosalind Franklin, 50 avenue Tony Garnier, Lyon, 69007, France
| | - Ali Chokr
- Research Laboratory of Microbiology (RLM), Department of Life and Earth Sciences, Faculty of Sciences I, Lebanese University, Hadat Campus, Beirut, Lebanon
| | - Patricia Doublet
- Centre International de Recherche en Infectiologie (CIRI), Université Lyon 1, INSERM U1111, CNRS UMR5308, ENS, Lyon Bât. Rosalind Franklin, 50 avenue Tony Garnier, Lyon, 69007, France
| | - Christophe Gilbert
- Centre International de Recherche en Infectiologie (CIRI), Université Lyon 1, INSERM U1111, CNRS UMR5308, ENS, Lyon Bât. Rosalind Franklin, 50 avenue Tony Garnier, Lyon, 69007, France.
| |
Collapse
|
8
|
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
|
9
|
Stengel D, Jörgensen AM, Polidori I, Kapitza P, Ricci F, Bernkop-Schnürch A. The power of sulfhydryl groups: Thiolated lipid-based nanoparticles enhance cellular uptake of nucleic acids. J Colloid Interface Sci 2023; 654:1136-1145. [PMID: 39491903 DOI: 10.1016/j.jcis.2023.10.039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 09/29/2023] [Accepted: 10/09/2023] [Indexed: 11/05/2024]
Abstract
AIM The aim of the study was to evaluate the effect of thiolation of lipid-based nanoparticles (LNPs) on cellular uptake of nucleic acids. METHODS A thiolated surfactant was synthesized by binding palmitic acid covalently to cysteine. Green fluorescent protein (GFP) encoding plasmid DNA (pDNA) was used as model nucleic acid and incorporated via hydrophobic ion-pairing with a cationic cholesterol derivate (DcCholesterol) in LNPs that were prepared by solvent injection method using the thiolated surfactant for surface decoration. LNPs were characterized regarding size, polydispersity index, zeta potential and stability in biorelevant media. The endosomal escape properties of LNPs were evaluated by erythrocytes interaction studies. Cell viability and transfection efficiency on HEK293 cells were investigated. RESULTS The structure of the thiolated surfactant was confirmed by 1H NMR and FT-IR. LNPs containing the nucleic acid-DcCholesterol complex with and without the thiolated surfactant were developed and displayed sizes in the range from 173 nm to 233 nm with a narrow size distribution (PDI < 0.3) and a negative zeta potential. LNPs showed no significant increase in size after 4 h of incubation in artificial body fluids. Erythrocytes interaction studies revealed enhanced endosomal escape properties for thiolated LNPs compared to non-thiolated LNPs. LNPs showed in concentrations lower than 0.74 mg/mL cell viability ≥ 80 %. Transfection studies on HEK cells with thiolated LNPs compared to non-thiolated LNP showed a 4.6-fold higher expression of GFP. CONCLUSION Surface thiolation of LNPs represents a promising tool for enhancing intracellular nucleic acid delivery of LNPs.
Collapse
Affiliation(s)
- Daniel Stengel
- Department of Pharmaceutical Technology, University of Innsbruck, Institute of Pharmacy, Center for Chemistry and Biomedicine, 6020 Innsbruck, Austria
| | - Arne M Jörgensen
- Department of Pharmaceutical Technology, University of Innsbruck, Institute of Pharmacy, Center for Chemistry and Biomedicine, 6020 Innsbruck, Austria
| | - Ilaria Polidori
- Department of Pharmaceutical Technology, University of Innsbruck, Institute of Pharmacy, Center for Chemistry and Biomedicine, 6020 Innsbruck, Austria
| | - Paul Kapitza
- Department of Pharmaceutical Chemistry, University of Innsbruck, Institute of Pharmacy, Center for Chemistry and Biomedicine, 6020 Innsbruck, Austria
| | - Fabrizio Ricci
- Thiomatrix Forschungs- und Beratungs GmbH, Trientlgasse 65, 6020 Innsbruck, Austria
| | - Andreas Bernkop-Schnürch
- Department of Pharmaceutical Technology, University of Innsbruck, Institute of Pharmacy, Center for Chemistry and Biomedicine, 6020 Innsbruck, Austria.
| |
Collapse
|
10
|
Leidecker M, Bertling A, Hussain M, Bischoff M, Eble JA, Fender AC, Jurk K, Rumpf C, Herrmann M, Kehrel BE, Niemann S. Protein Disulfide Isomerase and Extracellular Adherence Protein Cooperatively Potentiate Staphylococcal Invasion into Endothelial Cells. Microbiol Spectr 2023; 11:e0388622. [PMID: 36995240 PMCID: PMC10269700 DOI: 10.1128/spectrum.03886-22] [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: 09/26/2022] [Accepted: 03/09/2023] [Indexed: 03/31/2023] Open
Abstract
Invasion of host cells is an important feature of Staphylococcus aureus. The main internalization pathway involves binding of the bacteria to host cells, e.g., endothelial cells, via a fibronectin (Fn) bridge between S. aureus Fn binding proteins and α5β1-integrin, followed by phagocytosis. The secreted extracellular adherence protein (Eap) has been shown to promote this cellular uptake pathway of not only S. aureus, but also of bacteria otherwise poorly taken up by host cells, such as Staphylococcus carnosus. The exact mechanisms are still unknown. Previously, we demonstrated that Eap induces platelet activation by stimulation of the protein disulfide isomerase (PDI), a catalyst of thiol-disulfide exchange reactions. Here, we show that Eap promotes PDI activity on the surface of endothelial cells, and that this contributes critically to Eap-driven staphylococcal invasion. PDI-stimulated β1-integrin activation followed by increased Fn binding to host cells likely accounts for the Eap-enhanced uptake of S. aureus into non-professional phagocytes. Additionally, Eap supports the binding of S. carnosus to Fn-α5β1 integrin, thereby allowing its uptake into endothelial cells. To our knowledge, this is the first demonstration that PDI is crucial for the uptake of bacteria into host cells. We describe a hitherto unknown function of Eap-the promotion of an enzymatic activity with subsequent enhancement of bacterial uptake-and thus broaden mechanistic insights into its importance as a driver of bacterial pathogenicity. IMPORTANCE Staphylococcus aureus can invade and persist in non-professional phagocytes, thereby escaping host defense mechanisms and antibiotic treatment. The intracellular lifestyle of S. aureus contributes to the development of infection, e.g., in infective endocarditis or chronic osteomyelitis. The extracellular adherence protein secreted by S. aureus promotes its own internalization as well as that of bacteria that are otherwise poorly taken up by host cells, such as Staphylococcus carnosus. In our study, we demonstrate that staphylococcal uptake by endothelial cells requires catalytic disulfide exchange activity by the cell-surface protein disulfide isomerase, and that this critical enzymatic function is enhanced by Eap. The therapeutic application of PDI inhibitors has previously been investigated in the context of thrombosis and hypercoagulability. Our results add another intriguing possibility: therapeutically targeting PDI, i.e., as a candidate approach to modulate the initiation and/or course of S. aureus infectious diseases.
Collapse
Affiliation(s)
- Marleen Leidecker
- Institute of Medical Microbiology, University Hospital of Münster, Münster, Germany
| | - Anne Bertling
- Department of Anaesthesiology and Intensive Care, Experimental and Clinical Haemostasis, University Hospital of Münster, Münster, Germany
| | - Muzaffar Hussain
- Institute of Medical Microbiology, University Hospital of Münster, Münster, Germany
| | - Markus Bischoff
- Institute of Medical Microbiology and Hygiene, Saarland University, Homburg, Germany
| | - Johannes A. Eble
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Münster, Germany
| | - Anke C. Fender
- Department of Anaesthesiology and Intensive Care, Experimental and Clinical Haemostasis, University Hospital of Münster, Münster, Germany
- Institute of Pharmacology, University Hospital Essen, Essen, Germany
| | - Kerstin Jurk
- Department of Anaesthesiology and Intensive Care, Experimental and Clinical Haemostasis, University Hospital of Münster, Münster, Germany
- Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Christine Rumpf
- Institute of Medical Microbiology, University Hospital of Münster, Münster, Germany
| | - Mathias Herrmann
- Institute of Medical Microbiology, University Hospital of Münster, Münster, Germany
| | - Beate E. Kehrel
- Department of Anaesthesiology and Intensive Care, Experimental and Clinical Haemostasis, University Hospital of Münster, Münster, Germany
| | - Silke Niemann
- Institute of Medical Microbiology, University Hospital of Münster, Münster, Germany
| |
Collapse
|
11
|
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
|
12
|
Green RS, Izac JR, Naimi WA, O'Bier N, Breitschwerdt EB, Marconi RT, Carlyon JA. Ehrlichia chaffeensis EplA Interaction With Host Cell Protein Disulfide Isomerase Promotes Infection. Front Cell Infect Microbiol 2020; 10:500. [PMID: 33072622 PMCID: PMC7538545 DOI: 10.3389/fcimb.2020.00500] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 08/11/2020] [Indexed: 12/19/2022] Open
Abstract
Ehrlichia chaffeensis is an obligate intracellular bacterium that invades monocytes to cause the emerging and potentially severe disease, monocytic ehrlichiosis. Ehrlichial invasion of host cells, a process that is essential for the bacterium's survival and pathogenesis, is incompletely understood. In this study, we identified ECH_0377, henceforth designated as EplA (E. chaffeensis PDI ligand A) as an E. chaffeensis adhesin that interacts with host cell protein disulfide isomerase (PDI) to mediate bacterial entry into host cells. EplA is an outer membrane protein that E. chaffeensis expresses during growth in THP-1 monocytic cells. Canine sera confirmed to be positive for exposure to Ehrlichia spp. recognized recombinant EplA, indicating that it is expressed during infection in vivo. EplA antiserum inhibited the bacterium's ability to infect monocytic cells. The EplA-PDI interaction was confirmed via co-immunoprecipitation. Treating host cell surfaces with antibodies that inhibit PDI and/or thioredoxin-1 thiol reductase activity impaired E. chaffeensis infection. Chemical reduction of host cell surfaces, but not bacterial surfaces with tris(2-carboxyethyl)phosphine (TCEP) restored ehrlichial infectivity in the presence of the PDI-neutralizing antibody. Antisera specific for EplA C-terminal residues 95-104 (EplA95−104) or outer membrane protein A amino acids 53-68 (OmpA53−68) reduced E. chaffeensis infection of THP-1 cells. Notably, TCEP rescued ehrlichial infectivity of bacteria that had been treated with anti-EplA95−104, but not anti-EcOmpA53−68. These results demonstrate that EplA contributes to E. chaffeensis infection of monocytic cells by engaging PDI and exploiting the enzyme's reduction of host cell surface disulfide bonds in an EplA C-terminus-dependent manner and identify EplA95−104 and EcOmpA53−68 as novel ehrlichial receptor binding domains.
Collapse
Affiliation(s)
- Ryan S Green
- Department of Microbiology and Immunology, Virginia Commonwealth University Medical Center, School of Medicine, Richmond, VA, United States
| | - Jerilyn R Izac
- Department of Microbiology and Immunology, Virginia Commonwealth University Medical Center, School of Medicine, Richmond, VA, United States
| | - Waheeda A Naimi
- Department of Microbiology and Immunology, Virginia Commonwealth University Medical Center, School of Medicine, Richmond, VA, United States
| | - Nathaniel O'Bier
- Department of Microbiology and Immunology, Virginia Commonwealth University Medical Center, School of Medicine, Richmond, VA, United States
| | - Edward B Breitschwerdt
- Department of Clinical Sciences and the Intracellular Pathogens Research Laboratory, Comparative Medicine Institute, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States
| | - Richard T Marconi
- Department of Microbiology and Immunology, Virginia Commonwealth University Medical Center, School of Medicine, Richmond, VA, United States
| | - Jason A Carlyon
- Department of Microbiology and Immunology, Virginia Commonwealth University Medical Center, School of Medicine, Richmond, VA, United States
| |
Collapse
|
13
|
Green RS, Naimi WA, Oliver LD, O'Bier N, Cho J, Conrad DH, Martin RK, Marconi RT, Carlyon JA. Binding of Host Cell Surface Protein Disulfide Isomerase by Anaplasma phagocytophilum Asp14 Enables Pathogen Infection. mBio 2020; 11:e03141-19. [PMID: 31992623 PMCID: PMC6989111 DOI: 10.1128/mbio.03141-19] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 12/09/2019] [Indexed: 11/20/2022] Open
Abstract
Diverse intracellular pathogens rely on eukaryotic cell surface disulfide reductases to invade host cells. Pharmacologic inhibition of these enzymes is cytotoxic, making it impractical for treatment. Identifying and mechanistically dissecting microbial proteins that co-opt surface reductases could reveal novel targets for disrupting this common infection strategy. Anaplasma phagocytophilum invades neutrophils by an incompletely defined mechanism to cause the potentially fatal disease granulocytic anaplasmosis. The bacterium's adhesin, Asp14, contributes to invasion by virtue of its C terminus engaging an unknown receptor. Yeast-two hybrid analysis identified protein disulfide isomerase (PDI) as an Asp14 binding partner. Coimmunoprecipitation confirmed the interaction and validated it to be Asp14 C terminus dependent. PDI knockdown and antibody-mediated inhibition of PDI reductase activity impaired A. phagocytophilum infection of but not binding to host cells. Infection during PDI inhibition was rescued when the bacterial but not host cell surface disulfide bonds were chemically reduced with tris(2-carboxyethyl)phosphine-HCl (TCEP). TCEP also restored bacterial infectivity in the presence of an Asp14 C terminus blocking antibody that otherwise inhibits infection. A. phagocytophilum failed to productively infect myeloid-specific-PDI conditional-knockout mice, marking the first demonstration of in vivo microbial dependency on PDI for infection. Mutational analyses identified the Asp14 C-terminal residues that are critical for binding PDI. Thus, Asp14 binds and brings PDI proximal to A. phagocytophilum surface disulfide bonds that it reduces, which enables cellular and in vivo infection.IMPORTANCEAnaplasma phagocytophilum infects neutrophils to cause granulocytic anaplasmosis, an emerging potentially fatal disease and the second-most common tick-borne illness in the United States. Treatment options are limited, and no vaccine exists. Due to the bacterium's obligatory intracellular lifestyle, A. phagocytophilum survival and pathogenesis are predicated on its ability to enter host cells. Understanding its invasion mechanism will yield new targets for preventing bacterial entry and, hence, disease. We report a novel entry pathway in which the A. phagocytophilum outer membrane protein Asp14 binds host cell surface protein disulfide isomerase via specific C-terminal residues to promote reduction of bacterial surface disulfide bonds, which is critical for cellular invasion and productive infection in vivo Targeting the Asp14 C terminus could be used to prevent/treat granulocytic anaplasmosis. Our findings have broad implications, as a thematically similar approach could be applied to block infection by other intracellular microbes that exploit cell surface reductases.
Collapse
Affiliation(s)
- Ryan S Green
- Department of Microbiology and Immunology, School of Medicine, Virginia Commonwealth University Medical Center, Richmond, Virginia, USA
| | - Waheeda A Naimi
- Department of Microbiology and Immunology, School of Medicine, Virginia Commonwealth University Medical Center, Richmond, Virginia, USA
| | - Lee D Oliver
- Department of Microbiology and Immunology, School of Medicine, Virginia Commonwealth University Medical Center, Richmond, Virginia, USA
| | - Nathaniel O'Bier
- Department of Microbiology and Immunology, School of Medicine, Virginia Commonwealth University Medical Center, Richmond, Virginia, USA
| | - Jaehyung Cho
- Department of Pharmacology, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Daniel H Conrad
- Department of Microbiology and Immunology, School of Medicine, Virginia Commonwealth University Medical Center, Richmond, Virginia, USA
| | - Rebecca K Martin
- Department of Microbiology and Immunology, School of Medicine, Virginia Commonwealth University Medical Center, Richmond, Virginia, USA
| | - Richard T Marconi
- Department of Microbiology and Immunology, School of Medicine, Virginia Commonwealth University Medical Center, Richmond, Virginia, USA
| | - Jason A Carlyon
- Department of Microbiology and Immunology, School of Medicine, Virginia Commonwealth University Medical Center, Richmond, Virginia, USA
| |
Collapse
|
14
|
Saez D, Dushime R, Wu H, Ramos Cordova LB, Shukla K, Brown-Harding H, Furdui CM, Tsang AW. Sulforaphane promotes chlamydial infection by suppressing mitochondrial protein oxidation and activation of complement C3. Protein Sci 2020; 28:216-227. [PMID: 30367535 DOI: 10.1002/pro.3536] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 10/22/2018] [Accepted: 10/22/2018] [Indexed: 12/30/2022]
Abstract
Sulforaphane (SFN), a phytochemical found in broccoli and other cruciferous vegetables, is a potent antioxidant and anti-inflammatory agent with reported effects in cancer chemoprevention and suppression of infection with intracellular pathogens. Here we report on the impact of SFN on infection with Chlamydia trachomatis (Ct), a common sexually transmitted pathogen responsible for 131 million new cases annually worldwide. Astoundingly, we find that SFN as well as broccoli sprouts extract (BSE) promote Ct infection of human host cells. Both the number and size of Ct inclusions were increased when host cells were pretreated with SFN or BSE. The initial investigations presented here point to both the antioxidant and thiol alkylating properties of SFN as regulators of Ct infection. SFN decreased mitochondrial protein sulfenylation and promoted Ct development, which were both reversed by treatment with mitochondria-targeted paraquat (MitoPQ). Inhibition of the complement component 3 (complement C3) by SFN was also identified as a mechanism by which SFN promotes Ct infections. Mass spectrometry analysis found alkylation of cysteine 1010 (Cys1010) in complement C3 by SFN. The studies reported here raise awareness of the Ct infection promoting activity of SFN, and also identify potential mechanisms underlying this activity.
Collapse
Affiliation(s)
- Daniel Saez
- Department of Internal Medicine, Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, 27157, North Carolina
| | - Rosine Dushime
- Department of Internal Medicine, Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, 27157, North Carolina
| | - Hanzhi Wu
- Department of Internal Medicine, Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, 27157, North Carolina
| | - Lourdes B Ramos Cordova
- Department of Internal Medicine, Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, 27157, North Carolina
| | - Kirtikar Shukla
- Department of Internal Medicine, Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, 27157, North Carolina
| | | | - Cristina M Furdui
- Department of Internal Medicine, Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, 27157, North Carolina
| | - Allen W Tsang
- Department of Internal Medicine, Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, 27157, North Carolina
| |
Collapse
|
15
|
Gitsels A, Sanders N, Vanrompay D. Chlamydial Infection From Outside to Inside. Front Microbiol 2019; 10:2329. [PMID: 31649655 PMCID: PMC6795091 DOI: 10.3389/fmicb.2019.02329] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 09/24/2019] [Indexed: 12/16/2022] Open
Abstract
Chlamydia are obligate intracellular bacteria, characterized by a unique biphasic developmental cycle. Specific interactions with the host cell are crucial for the bacteria’s survival and amplification because of the reduced chlamydial genome. At the start of infection, pathogen-host interactions are set in place in order for Chlamydia to enter the host cell and reach the nutrient-rich peri-Golgi region. Once intracellular localization is established, interactions with organelles and pathways of the host cell enable the necessary hijacking of host-derived nutrients. Detailed information on the aforementioned processes will increase our understanding on the intracellular pathogenesis of chlamydiae and hence might lead to new strategies to battle chlamydial infection. This review summarizes how chlamydiae generate their intracellular niche in the host cell, acquire host-derived nutrients in order to enable their growth and finally exit the host cell in order to infect new cells. Moreover, the evolution in the development of molecular genetic tools, necessary for studying the chlamydial infection biology in more depth, is discussed in great detail.
Collapse
Affiliation(s)
- Arlieke Gitsels
- Laboratory for Immunology and Animal Biotechnology, Department of Animal Sciences and Aquatic Ecology, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| | - Niek Sanders
- Laboratory of Gene Therapy, Department of Nutrition, Genetics and Ethology, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Daisy Vanrompay
- Laboratory for Immunology and Animal Biotechnology, Department of Animal Sciences and Aquatic Ecology, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| |
Collapse
|
16
|
Berry A, Hall JV. The complexity of interactions between female sex hormones and Chlamydia trachomatis infections. CURRENT CLINICAL MICROBIOLOGY REPORTS 2019; 6:67-75. [PMID: 31890462 PMCID: PMC6936955 DOI: 10.1007/s40588-019-00116-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
PURPOSE OF REVIEW This review focuses specifically on the mechanisms by which female sex hormones, estrogen and progesterone, affect Chlamydia trachomatis infections in vivo and in vitro. RECENT FINDINGS Recent data support previous work indicating that estrogen enhances chlamydial development via multiple mechanisms. Progesterone negatively impacts Chlamydia infections also through multiple mechanisms, particularly by altering the immune response. Conflicting data exist regarding the effect of synthetic hormones, such as those found in hormonal contraceptives, on chlamydial infections. SUMMARY Numerous studies over the years have indicated that female sex hormones affect C. trachomatis infection. However, we still do not have a clear understanding of how these hormones alter Chlamydia disease transmission and progression. The studies reviewed here indicate that there are many variables that determine the outcome of Chlamydia/hormone interactions, including: 1) the specific hormone, 2) hormone concentration, 3) cell type or area of the genital tract, 4) hormone responsiveness of cell lines, and 5) animal models.
Collapse
Affiliation(s)
- Amy Berry
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN
- Center for Infectious Disease, Inflammation and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN
| | - Jennifer V. Hall
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN
- Center for Infectious Disease, Inflammation and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN
| |
Collapse
|
17
|
Abstract
The evolutionary separated Gram-negative Chlamydiales show a biphasic life cycle and replicate exclusively within eukaryotic host cells. Members of the genus Chlamydia are responsible for many acute and chronic diseases in humans, and Chlamydia-related bacteria are emerging pathogens. We revisit past efforts to detect cell wall material in Chlamydia and Chlamydia-related bacteria in the context of recent breakthroughs in elucidating the underlying cellular and molecular mechanisms of the chlamydial cell wall biosynthesis. In this review, we also discuss the role of cell wall biosynthesis in chlamydial FtsZ-independent cell division and immune modulation. In the past, penicillin susceptibility of an invisible wall was referred to as the "chlamydial anomaly." In light of new mechanistic insights, chlamydiae may now emerge as model systems to understand how a minimal and modified cell wall biosynthetic machine supports bacterial cell division and how cell wall-targeting beta-lactam antibiotics can also act bacteriostatically rather than bactericidal. On the heels of these discussions, we also delve into the effects of other cell wall antibiotics in individual chlamydial lineages.
Collapse
|
18
|
Dickinson MS, Anderson LN, Webb-Robertson BJM, Hansen JR, Smith RD, Wright AT, Hybiske K. Proximity-dependent proteomics of the Chlamydia trachomatis inclusion membrane reveals functional interactions with endoplasmic reticulum exit sites. PLoS Pathog 2019; 15:e1007698. [PMID: 30943267 PMCID: PMC6464245 DOI: 10.1371/journal.ppat.1007698] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 04/15/2019] [Accepted: 03/12/2019] [Indexed: 11/18/2022] Open
Abstract
Chlamydia trachomatis is the most common cause of bacterial sexually transmitted infection, responsible for millions of infections each year. Despite this high prevalence, the elucidation of the molecular mechanisms of Chlamydia pathogenesis has been difficult due to limitations in genetic tools and its intracellular developmental cycle. Within a host epithelial cell, chlamydiae replicate within a vacuole called the inclusion. Many Chlamydia-host interactions are thought to be mediated by the Inc family of type III secreted proteins that are anchored in the inclusion membrane, but their array of host targets are largely unknown. To investigate how the inclusion membrane proteome changes over the course of an infected cell, we have adapted the APEX2 system of proximity-dependent biotinylation. APEX2 is capable of specifically labeling proteins within a 20 nm radius in living cells. We transformed C. trachomatis to express the enzyme APEX2 fused to known inclusion membrane proteins, allowing biotinylation and purification of inclusion-associated proteins. Using quantitative mass spectrometry against APEX2 labeled samples, we identified over 400 proteins associated with the inclusion membrane at early, middle, and late stages of epithelial cell infection. This system was sensitive enough to detect inclusion interacting proteins early in the developmental cycle, at 8 hours post infection, a previously intractable time point. Mass spectrometry analysis revealed a novel, early association between C. trachomatis inclusions and endoplasmic reticulum exit sites (ERES), functional regions of the ER where COPII-coated vesicles originate. Pharmacological and genetic disruption of ERES function severely restricted early chlamydial growth and the development of infectious progeny. APEX2 is therefore a powerful in situ approach for identifying critical protein interactions on the membranes of pathogen-containing vacuoles. Furthermore, the data derived from proteomic mapping of Chlamydia inclusions has illuminated an important functional role for ERES in promoting chlamydial developmental growth.
Collapse
Affiliation(s)
- Mary S. Dickinson
- Department of Global Health, Graduate Program in Pathobiology, University of Washington, Seattle, WA, United States of America
- Department of Medicine, Division of Allergy and Infectious Diseases, Center for Emerging and Reemerging Infectious Disease (CERID), University of Washington, Seattle, WA, United States of America
| | - Lindsey N. Anderson
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, United States of America
| | | | - Joshua R. Hansen
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, United States of America
| | - Richard D. Smith
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, United States of America
| | - Aaron T. Wright
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, United States of America
- The Gene and Linda Voiland College of Chemical Engineering and Bioengineering, Washington State University, Pullman, WA, United States of America
| | - Kevin Hybiske
- Department of Global Health, Graduate Program in Pathobiology, University of Washington, Seattle, WA, United States of America
- Department of Medicine, Division of Allergy and Infectious Diseases, Center for Emerging and Reemerging Infectious Disease (CERID), University of Washington, Seattle, WA, United States of America
| |
Collapse
|
19
|
Christensen S, McMahon RM, Martin JL, Huston WM. Life inside and out: making and breaking protein disulfide bonds in Chlamydia. Crit Rev Microbiol 2019; 45:33-50. [PMID: 30663449 DOI: 10.1080/1040841x.2018.1538933] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Disulphide bonds are widely used among all domains of life to provide structural stability to proteins and to regulate enzyme activity. Chlamydia spp. are obligate intracellular bacteria that are especially dependent on the formation and degradation of protein disulphide bonds. Members of the genus Chlamydia have a unique biphasic developmental cycle alternating between two distinct cell types; the extracellular infectious elementary body (EB) and the intracellular replicating reticulate body. The proteins in the envelope of the EB are heavily cross-linked with disulphides and this is known to be critical for this infectious phase. In this review, we provide a comprehensive summary of what is known about the redox state of chlamydial envelope proteins throughout the developmental cycle. We focus especially on the factors responsible for degradation and formation of disulphide bonds in Chlamydia and how this system compares with redox regulation in other organisms. Focussing on the unique biology of Chlamydia enables us to provide important insights into how specialized suites of disulphide bond (Dsb) proteins cater for specific bacterial environments and lifecycles.
Collapse
Affiliation(s)
- Signe Christensen
- a Division of Chemistry and Structural Biology , Institute for Molecular Bioscience, University of Queensland , St. Lucia , QLD , Australia.,b Griffith Institute for Drug Discovery, Griffith University , Nathan , QLD , Australia
| | - Róisín M McMahon
- b Griffith Institute for Drug Discovery, Griffith University , Nathan , QLD , Australia
| | - Jennifer L Martin
- b Griffith Institute for Drug Discovery, Griffith University , Nathan , QLD , Australia
| | - Wilhelmina M Huston
- c School of Life Sciences , University of Technology Sydney , Ultimo , NSW , Australia
| |
Collapse
|
20
|
Park JS, Helble JD, Lazarus JE, Yang G, Blondel CJ, Doench JG, Starnbach MN, Waldor MK. A FACS-Based Genome-wide CRISPR Screen Reveals a Requirement for COPI in Chlamydia trachomatis Invasion. iScience 2018; 11:71-84. [PMID: 30590252 PMCID: PMC6308251 DOI: 10.1016/j.isci.2018.12.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 09/20/2018] [Accepted: 12/10/2018] [Indexed: 12/26/2022] Open
Abstract
The invasion of Chlamydia trachomatis, an obligate intracellular bacterium, into epithelial cells is driven by a complex interplay of host and bacterial factors. To comprehensively define the host genes required for pathogen invasion, we undertook a fluorescence-activated cell sorting (FACS)-based CRISPR screen in human cells. A genome-wide loss-of-function library was infected with fluorescent C. trachomatis and then sorted to enrich for invasion-deficient mutants. The screen identified heparan sulfate, a known pathogen receptor, as well as coatomer complex I (COPI). We found that COPI, through a previously unappreciated role, promotes heparan sulfate cell surface presentation, thereby facilitating C. trachomatis attachment. The heparan sulfate defect does not fully account for the resistance of COPI mutants. COPI also promotes the activity of the pathogen's type III secretion system. Together, our findings establish the requirement for COPI in C. trachomatis invasion and the utility of FACS-based CRISPR screening for the elucidation of host factors required for pathogen invasion. FACS-based CRISPR screen to identify host factors required for C. trachomatis invasion Candidate genes comprise heparan sulfate biosynthesis, actin remodeling, and COPI COPI regulates heparan sulfate cell surface presentation and C. trachomatis attachment COPI is also required for efficient C. trachomatis T3SS translocation
Collapse
Affiliation(s)
- Joseph S Park
- Howard Hughes Medical Institute, Boston, MA 02215, USA; Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115, USA; Division of Infectious Diseases, Brigham & Women's Hospital, Boston 02115, MA, USA; Boston University School of Medicine, Boston, MA 02120, USA
| | - Jennifer D Helble
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Jacob E Lazarus
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115, USA; Division of Infectious Diseases, Brigham & Women's Hospital, Boston 02115, MA, USA; Division of Infectious Diseases, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Guanhua Yang
- Division of Infectious Diseases, Brigham & Women's Hospital, Boston 02115, MA, USA
| | - Carlos J Blondel
- Division of Infectious Diseases, Brigham & Women's Hospital, Boston 02115, MA, USA
| | - John G Doench
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Michael N Starnbach
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Matthew K Waldor
- Howard Hughes Medical Institute, Boston, MA 02215, USA; Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115, USA; Division of Infectious Diseases, Brigham & Women's Hospital, Boston 02115, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
| |
Collapse
|
21
|
Wang X, Hybiske K, Stephens RS. Direct visualization of the expression and localization of chlamydial effector proteins within infected host cells. Pathog Dis 2018; 76:4830102. [PMID: 29390129 DOI: 10.1093/femspd/fty011] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 01/25/2018] [Indexed: 11/14/2022] Open
Abstract
Chlamydia secrete into host cells a diverse array of effector proteins, but progress in characterizing the spatiotemporal localization of these proteins has been hindered by a paucity of genetic approaches in Chlamydia and also by the challenge of studying these proteins within the live cellular environment. We adapted a split-green fluorescent protein (GFP) system for use in Chlamydia to label chlamydial effector proteins and track their localization in host cells under native environment. The efficacy of this system was demonstrated by detecting several known Chlamydia proteins including IncA, CT005 and CT694. We further used this approach to detect two chlamydial deubiquitinases (CT867 and CT868) within live cells during the infection. CT868 localized only to the inclusion membrane at early and late developmental stages. CT867 localized to the chlamydial inclusion membrane at an early developmental stage and was concomitantly localized to the host plasma membrane at a late stage during the infection. These data suggest that chlamydial deubiquitinase play important roles for chlamydial pathogenesis by targeting proteins at both the plasma membrane and the chlamydial inclusion membrane. The split-GFP technology was demonstrated to be a robust and efficient approach to identify the secretion and cellular localization of important chlamydial virulence factors.
Collapse
Affiliation(s)
- Xiaogang Wang
- Program in Infectious Diseases, School of Public Health, University of California, 51 Koshland Hall, Berkeley, CA 94720, USA
| | - Kevin Hybiske
- Division of Allergy and Infectious Diseases, Department of Medicine, University of Washington, 750 Republican St Seattle, WA 98109, USA
| | - Richard S Stephens
- Program in Infectious Diseases, School of Public Health, University of California, 51 Koshland Hall, Berkeley, CA 94720, USA
| |
Collapse
|
22
|
Wang X, Hybiske K, Stephens RS. Orchestration of the mammalian host cell glucose transporter proteins-1 and 3 by Chlamydia contributes to intracellular growth and infectivity. Pathog Dis 2018; 75:4411801. [PMID: 29040458 DOI: 10.1093/femspd/ftx108] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 10/05/2017] [Indexed: 01/03/2023] Open
Abstract
Chlamydia are gram-negative obligate intracellular bacteria that replicate within a discrete cellular vacuole, called an inclusion. Although it is known that Chlamydia require essential nutrients from host cells to support their intracellular growth, the molecular mechanisms for acquiring these macromolecules remain uncharacterized. In the present study, it was found that the expression of mammalian cell glucose transporter proteins 1 (GLUT1) and glucose transporter proteins 3 (GLUT3) were up-regulated during chlamydial infection. Up-regulation was dependent on bacterial protein synthesis and Chlamydia-induced MAPK kinase activation. GLUT1, but not GLUT3, was observed in close proximity to the inclusion membrane throughout the chlamydial developmental cycle. The proximity of GLUT1 to the inclusion was dependent on a brefeldin A-sensitive pathway. Knockdown of GLUT1 and GLUT3 with specific siRNA significantly impaired chlamydial development and infectivity. It was discovered that the GLUT1 protein was stabilized during infection by inhibition of host-dependent ubiquitination of GLUT1, and this effect was associated with the chlamydial deubiquitinase effector protein CT868. This report demonstrates that Chlamydia exploits host-derived transporter proteins altering their expression, turnover and localization. Consequently, host cell transporter proteins are manipulated during infection as a transport system to fulfill the carbon source requirements for Chlamydia.
Collapse
Affiliation(s)
- Xiaogang Wang
- Program in Infectious Diseases, School of Public Health, University of California, Berkeley, 51 Koshland Hall, CA 94720, USA.,Division of Infectious Diseases, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, 181 Longwood Ave, Boston, MA 02115, USA
| | - Kevin Hybiske
- Division of Allergy and Infectious Diseases, Department of Medicine, University of Washington, 750 Republican St, Seattle, WA 98109, USA
| | - Richard S Stephens
- Program in Infectious Diseases, School of Public Health, University of California, Berkeley, 51 Koshland Hall, CA 94720, USA
| |
Collapse
|
23
|
Chlamydia exploits filopodial capture and a macropinocytosis-like pathway for host cell entry. PLoS Pathog 2018; 14:e1007051. [PMID: 29727463 PMCID: PMC5955597 DOI: 10.1371/journal.ppat.1007051] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Revised: 05/16/2018] [Accepted: 04/21/2018] [Indexed: 01/08/2023] Open
Abstract
Pathogens hijack host endocytic pathways to force their own entry into eukaryotic target cells. Many bacteria either exploit receptor-mediated zippering or inject virulence proteins directly to trigger membrane reorganisation and cytoskeletal rearrangements. By contrast, extracellular C. trachomatis elementary bodies (EBs) apparently employ facets of both the zipper and trigger mechanisms and are only ~400 nm in diameter. Our cryo-electron tomography of C. trachomatis entry revealed an unexpectedly diverse array of host structures in association with invading EBs, suggesting internalisation may progress by multiple, potentially redundant routes or several sequential events within a single pathway. Here we performed quantitative analysis of actin organisation at chlamydial entry foci, highlighting filopodial capture and phagocytic cups as dominant and conserved morphological structures early during internalisation. We applied inhibitor-based screening and employed reporters to systematically assay and visualise the spatio-temporal contribution of diverse endocytic signalling mediators to C. trachomatis entry. In addition to the recognised roles of the Rac1 GTPase and its associated nucleation-promoting factor (NPF) WAVE, our data revealed an additional unrecognised pathway sharing key hallmarks of macropinocytosis: i) amiloride sensitivity, ii) fluid-phase uptake, iii) recruitment and activity of the NPF N-WASP, and iv) the localised generation of phosphoinositide-3-phosphate (PI3P) species. Given their central role in macropinocytosis and affinity for PI3P, we assessed the role of SNX-PX-BAR family proteins. Strikingly, SNX9 was specifically and transiently enriched at C. trachomatis entry foci. SNX9-/- cells exhibited a 20% defect in EB entry, which was enhanced to 60% when the cells were infected without sedimentation-induced EB adhesion, consistent with a defect in initial EB-host interaction. Correspondingly, filopodial capture of C. trachomatis EBs was specifically attenuated in SNX9-/- cells, implicating SNX9 as a central host mediator of filopodial capture early during chlamydial entry. Our findings identify an unanticipated complexity of signalling underpinning cell entry by this major human pathogen, and suggest intriguing parallels with viral entry mechanisms. Chlamydia trachomatis remains the leading bacterial agent of sexually transmitted disease worldwide and causes a form of blindness called trachoma in Developing nations, which is recognised by the World Health Organisation as a neglected tropical disease. Despite this burden, we know comparatively little about how it causes disease at a molecular level. Chlamydia must live inside human cells to survive, and here we study the mechanism of how it enters cells, which is critical to the lifecycle. We study how the bacterium exploits signalling pathways inside the cell to its own advantage to deform the cell membrane by reorganising the underlying cell skeleton, and identify new factors involved in this process. Our findings suggest intriguing similarities with how some viruses enter cells. A better understanding of these processes may help to develop future vaccines and new treatments.
Collapse
|
24
|
Kókai D, Mosolygó T, Virók DP, Endrész V, Burián K. N-acetyl-cysteine increases the replication of Chlamydia pneumoniae and prolongs the clearance of the pathogen from mice. J Med Microbiol 2018. [PMID: 29521616 DOI: 10.1099/jmm.0.000716] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Purpose. Within the community, 10 % of acquired pneumonia is caused by Chlamydia pneumoniae. N-acetyl-cysteine (NAC) is one of the most commonly used mucolytics in respiratory diseases, but its effect on C. pneumoniae infection has not yet been investigated. In this study, our aim was to investigate whether NAC influences the replication of C. pneumoniae. After determining that NAC does have an effect on C. pneumoniae replication, the effect of an alternative drug called Ambroxol (Ax) was investigated.Methodology. The in vitro effect of NAC and Ax was studied on C. pneumoniae-infected A549 and McCoy cells. Furthermore, the influence of NAC and Ax was examined in mice infected intranasally with C. pneumoniae.Results. NAC treatment resulted in approximately sixfold more efficient C. pneumoniae growth in tissue culture compared to the untreated control cells, and this effect was shown to be based on the increased binding of the bacterium to the host cells. The C. pneumoniae-infected mice to which NAC was given had prolonged and more severe infections than the control mice. Ax decreased C. pneumoniae replication in vitro, which was partially associated with the increased expression of indolamine 2,3-dioxygenase. In animals, using the adapted usual human dose, Ax did not alter the number of recoverable C. pneumoniae.Conclusion. Based on our results, it might be recommended that a mucolytic agent other than NAC, such as Ax, be used in respiratory diseases suspected to be caused by C. pneumoniae.
Collapse
Affiliation(s)
- Dávid Kókai
- Department of Medical Microbiology and Immunobiology, University of Szeged, Dóm tér 10, 6720 Szeged, Hungary
| | - Tímea Mosolygó
- Department of Medical Microbiology and Immunobiology, University of Szeged, Dóm tér 10, 6720 Szeged, Hungary
| | - Dezső P Virók
- Department of Medical Microbiology and Immunobiology, University of Szeged, Dóm tér 10, 6720 Szeged, Hungary
| | - Valéria Endrész
- Department of Medical Microbiology and Immunobiology, University of Szeged, Dóm tér 10, 6720 Szeged, Hungary
| | - Katalin Burián
- Department of Medical Microbiology and Immunobiology, University of Szeged, Dóm tér 10, 6720 Szeged, Hungary
| |
Collapse
|
25
|
McKuen MJ, Mueller KE, Bae YS, Fields KA. Fluorescence-Reported Allelic Exchange Mutagenesis Reveals a Role for Chlamydia trachomatis TmeA in Invasion That Is Independent of Host AHNAK. Infect Immun 2017; 85:e00640-17. [PMID: 28970272 PMCID: PMC5695130 DOI: 10.1128/iai.00640-17] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Accepted: 09/21/2017] [Indexed: 01/05/2023] Open
Abstract
Development of approaches to genetically manipulate Chlamydia is fostering important advances in understanding pathogenesis. Fluorescence-reported allelic exchange mutagenesis (FRAEM) now enables the complete deletion of specific genes in C. trachomatis L2. We have leveraged this technology to delete the coding sequences for a known type III effector. The evidence provided here indicates that CT694/CTL0063 is a virulence protein involved in chlamydial invasion. Based on our findings, we designate the gene product corresponding to ct694-ctl0063translocated membrane-associated effector A (TmeA). Deletion of tmeA did not impact development of intracellular chlamydiae. However, the absence of TmeA manifested as a decrease in infectivity in both tissue culture and murine infection models. The in vitro defect was reflected by impaired invasion of host cells. TmeA binds human AHNAK, and we demonstrate here that AHNAK is transiently recruited by invading chlamydiae. TmeA, however, is not required for endogenous AHNAK recruitment. TmeA also impairs AHNAK-dependent actin bundling activity. This TmeA-mediated effect likely does not explain impaired invasion displayed by the tmeA strain of Chlamydia, since AHNAK-deficient cells revealed no invasion phenotype. Overall, our data indicate the efficacy of FRAEM and reveal a role of TmeA during chlamydial invasion that manifests independently of effects on AHNAK.
Collapse
Affiliation(s)
- M J McKuen
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - K E Mueller
- Department of Microbiology, Immunology & Molecular Genetics, University of Kentucky College of Medicine, Lexington, Kentucky, USA
| | - Y S Bae
- Department of Life Science, Ewha Womans University, Seoul, South Korea
| | - K A Fields
- Department of Microbiology, Immunology & Molecular Genetics, University of Kentucky College of Medicine, Lexington, Kentucky, USA
| |
Collapse
|
26
|
Guo H, Xian JA, Wang AL. Analysis of digital gene expression profiling in hemocytes of white shrimp Litopenaeus vannamei under nitrite stress. FISH & SHELLFISH IMMUNOLOGY 2016; 56:1-11. [PMID: 27377029 DOI: 10.1016/j.fsi.2016.06.059] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Revised: 06/30/2016] [Accepted: 06/30/2016] [Indexed: 06/06/2023]
Abstract
Accumulation of nitrite in water is highly toxic to aquatic animals. To understand immune responses in shrimp under such environmental stress, a digital gene expression (DGE) technology was applied to detect the gene expression profile of the Litopenaeus vannamei hemocytes in response to nitrite for 48 h. A total of 1922 differently expressed unigenes were generated. Of these transcripts, 1269 and 653 genes were up- or down-regulated respectively. Functional categorization and pathways of the differentially expressed genes revealed that immune defense, xenobiotics biodegradation and metabolism, amino acid and nucleobase metabolic process, apoptosis were the differentially regulated processes occurring during nitrite stress. We selected 19 differential expression transcripts (DETs) to validate the sequencing results by real time quantitative PCR (qPCR). The Pearson's correlation coefficient (R) of the 19 DETs was 0.843, which confirmed the consistency and accuracy between these two approaches. Subsequently, we screened 10 genes to examine the changes in the time course of gene expression in more detail. The results indicated that expressions of ATP-binding cassette transporter (ABC transporter), caspase10, QM protein, C type lectin 4 (CTL4), protein disulfide isomerase (PDI), serine protease inhibitor 8 (SPI8), transglutaminase (TGase), chitinase1, inhibitors of apoptosis proteins (IAP) and cytochrome P450 enzyme (CYP450) were induced to participate in the anti-stress defense against nitrite. These results will provide a reference for follow-up study of molecular toxicology and valuable gene information for better understanding of immune response in L. vannamei under environmental stress.
Collapse
Affiliation(s)
- Hui Guo
- Key Laboratory of Marine Ecology and Aquaculture Environment of Zhanjiang, College of Fisheries, Guangdong Ocean University, Zhanjiang, 524025, People's Republic of China.
| | - Jian-An Xian
- Institute of Tropical Bioscience and Biotechnology, Chinese Academy of Tropical Agricultural Sciences, Haikou, 571101, People's Republic of China
| | - An-Li Wang
- Key Laboratory of Ecology and Environmental Science of Guangdong Higher Education Institutes, Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, School of Life Science, South China Normal University, Guangzhou, 510631, People's Republic of China
| |
Collapse
|
27
|
Radomski N, Einenkel R, Müller A, Knittler MR. Chlamydia-host cell interaction not only from a bird's eye view: some lessons fromChlamydia psittaci. FEBS Lett 2016; 590:3920-3940. [DOI: 10.1002/1873-3468.12295] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2016] [Revised: 07/06/2016] [Accepted: 07/07/2016] [Indexed: 12/13/2022]
Affiliation(s)
- Nadine Radomski
- Friedrich-Loeffler-Institut; Institute of Immunology; Isle of Riems Germany
| | - Rebekka Einenkel
- Friedrich-Loeffler-Institut; Institute of Immunology; Isle of Riems Germany
| | - Anne Müller
- Friedrich-Loeffler-Institut; Institute of Immunology; Isle of Riems Germany
| | - Michael R Knittler
- Friedrich-Loeffler-Institut; Institute of Immunology; Isle of Riems Germany
| |
Collapse
|
28
|
Kummer A, Nishanth G, Koschel J, Klawonn F, Schlüter D, Jänsch L. Listeriosis downregulates hepatic cytochrome P450 enzymes in sublethal murine infection. Proteomics Clin Appl 2016; 10:1025-1035. [PMID: 27273978 DOI: 10.1002/prca.201600030] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Revised: 04/11/2016] [Accepted: 06/01/2016] [Indexed: 11/08/2022]
Abstract
PURPOSE Listeria monocytogenes (Lm) can cross the intestinal barrier in humans and then disseminates into different organs. Invasion of the liver occurs even in sublethal infections, however, knowledge of affected physiological processes is scarce. This study employed a sublethal murine infection model to investigate liver responses systematically by proteomics. EXPERIMENTAL DESIGN Liver samples from three stages of the sublethal infection covering the initial invasion, the peak of infection, and the clearance phase (1, 3, 9 days postinoculation) were analyzed in comparison to samples from noninfected mice. Apart from flow cytometry and RT-PCRs for immune status control, liver responses were analyzed by quantitative peptide sequencing (HPLC-Orbitrap Fusion) using 4-plex iTRAQ-labeling. RESULTS Accurate MS characterized about 3600 proteins and statistics revealed 15% of the hepatic proteome as regulated. Immunological data as well as protein regulation dynamics strongly indicate stage-specific hepatic responses in sublethal infections. Most notably, this study detected a comprehensive deregulation of drug metabolizing enzymes at all stages, including 25 components of the cytochrome P450 system. CONCLUSIONS AND CLINICAL RELEVANCE Sublethal Lm infection deregulates hepatic drug metabolizing pathways. This finding indicates the need to monitor drug administration along Lm infections, especially in all patients needing constant medication.
Collapse
Affiliation(s)
- Anne Kummer
- Cellular Proteomics, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Gopala Nishanth
- Otto-von-Guericke University, Magdeburg, Germany.,Organ-specific Immune Regulation, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | | | - Frank Klawonn
- Cellular Proteomics, Helmholtz Centre for Infection Research, Braunschweig, Germany.,Department of Computer Science, Ostfalia University of Applied Sciences, Wolfenbüttel, Germany
| | - Dirk Schlüter
- Otto-von-Guericke University, Magdeburg, Germany.,Organ-specific Immune Regulation, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Lothar Jänsch
- Cellular Proteomics, Helmholtz Centre for Infection Research, Braunschweig, Germany.
| |
Collapse
|
29
|
Fechtner T, Galle JN, Hegemann JH. The novel chlamydial adhesin CPn0473 mediates the lipid raft-dependent uptake of Chlamydia pneumoniae. Cell Microbiol 2016; 18:1094-105. [PMID: 26780295 PMCID: PMC5067637 DOI: 10.1111/cmi.12569] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Revised: 12/21/2015] [Accepted: 01/04/2016] [Indexed: 01/23/2023]
Abstract
Chlamydiae are Gram‐negative, obligate intracellular pathogens that pose a serious threat to public health worldwide. Chlamydial surface molecules are essential for host cell invasion. The first interaction with the host cell is thereby accomplished by the Outer membrane complex protein B (OmcB) binding to heparan sulfate moieties on the host cell surface, followed by the interaction of the chlamydial polymorphic membrane proteins (Pmps) with host cell receptors. Specifically, the interaction of the Pmp21 adhesin and invasin with its human interaction partner, the epidermal growth factor receptor, results in receptor activation, down‐stream signalling and finally internalization of the bacteria. Blocking both, the OmcB and Pmp21 adhesion pathways, did not completely abolish infection, suggesting the presence of additional factors relevant for host cell invasion. Here, we show that the novel surface protein CPn0473 of Chlamydia pneumoniae contributes to the binding and invasion of infectious chlamydial particles. CPn0473 is expressed late in the infection cycle and located on the infectious chlamydial cell surface. Soluble recombinant CPn0473 as well as rCPn0473‐coupled fluorescent latex beads adhere to human epithelial HEp‐2 cells. Interestingly, in classical infection blocking experiments pretreatment of HEp‐2 cells with rCPn0473 does not attenuate adhesion but promotes dose‐dependently internalization by C. pneumoniae suggesting an unusual mode of action for this adhesin. This CPn0473‐dependent promotion of infection by C. pneumoniae depends on two different domains within the protein and requires intact lipid rafts. Thus, inhibition of the interaction of CPn0473 with the host cell could provide a way to reduce the virulence of C. pneumoniae.
Collapse
Affiliation(s)
- Tim Fechtner
- Lehrstuhl für Funktionelle Genomforschung der Mikroorganismen, Heinrich-Heine-Universität Düsseldorf, Universitätsstraße 1, 40225, Düsseldorf, Germany
| | - Jan N Galle
- Lehrstuhl für Funktionelle Genomforschung der Mikroorganismen, Heinrich-Heine-Universität Düsseldorf, Universitätsstraße 1, 40225, Düsseldorf, Germany
| | - Johannes H Hegemann
- Lehrstuhl für Funktionelle Genomforschung der Mikroorganismen, Heinrich-Heine-Universität Düsseldorf, Universitätsstraße 1, 40225, Düsseldorf, Germany
| |
Collapse
|
30
|
An ERp57-mediated disulphide exchange promotes the interaction between Burkholderia cenocepacia and epithelial respiratory cells. Sci Rep 2016; 6:21140. [PMID: 26879174 PMCID: PMC4754759 DOI: 10.1038/srep21140] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Accepted: 01/19/2016] [Indexed: 01/29/2023] Open
Abstract
Previous studies have demonstrated that extracellular glutathione reduces the ability of the Cystic Fibrosis pathogen Burkholderia cenocepacia to infect primary or immortalized epithelial respiratory cells. We report here that the adhesion and invasion ability of B. cenocepacia is limited also by thiol-oxidizing and disulphide-reducing agents and by protein disulfide isomerase (PDI) inhibitors. PDI inhibitors also reduce the proinflammatory response elicited by cells in response to Burkholderia. These findings indicate that a membrane-associated PDI catalyzes thiol/disulphide exchange reactions which favor bacterial infection. The combined use of selective PDI inhibitors, RNA silencing and specific antibodies identified ERp57 as a major PDI involved in the interaction between B. cenocepacia and epithelial cells. This study contributes to the elucidation of the Burkholderia pathogenic mechanisms by showing that this microorganism exploits a membrane-associated host protein to infect epithelial cells and identifies ERp57 as a putative pharmacological target for the treatment of Burkholderia lung infections.
Collapse
|
31
|
Abstract
The lifestyle of Chlamydiae is unique: the bacteria alternate between two morphologically distinct forms, an infectious non-replicative elementary body (EB), and a replicative, non-infectious reticulate body (RB). This review focuses on recent advances in understanding the structure and function of the infectious form of the best-studied member of the phylum, the human pathogen Chlamydia trachomatis. Once considered as an inert particle of little functional capacity, the EB is now perceived as a sophisticated entity that encounters at least three different environments during each infectious cycle. We review current knowledge on its composition and morphology, and emerging metabolic activities. These features confer resistance to the extracellular environment, the ability to penetrate a host cell and ultimately enable the EB to establish a niche enabling bacterial survival and growth. The bacterial and host molecules involved in these processes are beginning to emerge.
Collapse
|
32
|
Poston TB, Darville T. Chlamydia trachomatis: Protective Adaptive Responses and Prospects for a Vaccine. Curr Top Microbiol Immunol 2016; 412:217-237. [PMID: 27033698 DOI: 10.1007/82_2016_6] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Chlamydia trachomatis is the most common cause of sexually transmitted bacterial infection globally. These infections translate to a significant public health burden, particularly women's healthcare costs due to serious disease sequelae such as pelvic inflammatory disease (PID), tubal factor infertility, chronic pelvic pain, and ectopic pregnancy. There is no evidence that natural immunity can provide complete, long-term protection necessary to prevent chronic pathology, making human vaccine development critical. Vaccine design will require careful consideration of protective versus pathological host-response mechanisms in concert with elucidation of optimal antigens and adjuvants. Evidence suggests that a Th1 response, facilitated by IFN-γ-producing CD4 T cells, will be instrumental in generating long-term, sterilizing immunity. Although the role of antibodies is not completely understood, they have exhibited a protective effect by enhancing chlamydial clearance. Future work will require investigation of broadly neutralizing antibodies and antibody-augmented cellular immunity to successfully design a vaccine that potently elicits both arms of the immune response. Sterilizing immunity is the ultimate goal. However, vaccine-induced partial immunity that prevents upper genital tract infection and inflammation would be cost-effective compared to current screening and treatment strategies. In this chapter, we examine evidence from animal and human studies demonstrating protective adaptive immune responses to Chlamydia and discuss future challenges and prospects for vaccine development.
Collapse
Affiliation(s)
- Taylor B Poston
- Department of Pediatrics, University of North Carolina, Chapel Hill, NC, USA
| | - Toni Darville
- Department of Pediatrics, University of North Carolina, Chapel Hill, NC, USA.
| |
Collapse
|
33
|
Nans A, Ford C, Hayward RD. Host-pathogen reorganisation during host cell entry by Chlamydia trachomatis. Microbes Infect 2015; 17:727-31. [PMID: 26320027 PMCID: PMC4670903 DOI: 10.1016/j.micinf.2015.08.004] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 08/20/2015] [Indexed: 12/12/2022]
Abstract
Chlamydia trachomatis is obligate intracellular bacterial pathogen that remains a significant public health burden worldwide. A critical early event during infection is chlamydial entry into non-phagocytic host epithelial cells. Like other Gram-negative bacteria, C. trachomatis uses a type III secretion system (T3SS) to deliver virulence effector proteins into host cells. These effectors trigger bacterial uptake and promote bacterial survival and replication within the host cell. In this review, we highlight recent cryo-electron tomography that has provided striking insights into the initial interactions between Chlamydia and its host. We describe the polarised structure of extracellular C. trachomatis elementary bodies (EBs), and the supramolecular organisation of T3SS complexes on the EB surface, in addition to the changes in host and pathogen architecture that accompany bacterial internalisation and EB encapsulation into early intracellular vacuoles. Finally, we consider the implications for further understanding the mechanism of C. trachomatis entry and how this might relate to those of other bacteria and viruses.
Collapse
Affiliation(s)
- Andrea Nans
- Institute of Structural and Molecular Biology, University College London & Birkbeck, Malet Street, London WC1E 7HX, UK
| | - Charlotte Ford
- Institute of Structural and Molecular Biology, University College London & Birkbeck, Malet Street, London WC1E 7HX, UK
| | - Richard D Hayward
- Institute of Structural and Molecular Biology, University College London & Birkbeck, Malet Street, London WC1E 7HX, UK.
| |
Collapse
|
34
|
Ferrell JC, Fields KA. A working model for the type III secretion mechanism in Chlamydia. Microbes Infect 2015; 18:84-92. [PMID: 26515030 DOI: 10.1016/j.micinf.2015.10.006] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Revised: 10/19/2015] [Accepted: 10/19/2015] [Indexed: 01/09/2023]
Abstract
It has been appreciated for almost 20 years that members of the Chlamydiales possess a virulence-associated type III secretion mechanism. Given the obligate intracellular nature of these bacteria, defining exactly how type III secretion functions to promote pathogenesis has been challenging. We present a working model herein that is based on current evidence.
Collapse
Affiliation(s)
- Joshua C Ferrell
- Department of Microbiology, Immunology & Molecular Genetics, University of Kentucky College of Medicine, Lexington, KY 40536, USA
| | - Kenneth A Fields
- Department of Microbiology, Immunology & Molecular Genetics, University of Kentucky College of Medicine, Lexington, KY 40536, USA.
| |
Collapse
|
35
|
Karunakaran K, Subbarayal P, Vollmuth N, Rudel T. Chlamydia-infected cells shed Gp96 to prevent chlamydial re-infection. Mol Microbiol 2015; 98:694-711. [PMID: 26235316 DOI: 10.1111/mmi.13151] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/29/2015] [Indexed: 12/29/2022]
Abstract
Chlamydia trachomatis is an obligate intracellular human pathogen with a biphasic developmental life cycle. The infectious elementary bodies (EBs) enter a host cell where they transform into reticulate bodies (RBs) that use cellular metabolites to multiply. Re-infection of an infected cell during the replicative phase of chlamydial development may prevent formation of infectious EBs, interrupting the infectious cycle. Here, we report that Glucose Regulated Protein 96 (Gp96), a chaperone for cell surface receptors, binds to and facilitates adherence and entry of C. trachomatis. Gp96 expression was increased early in infection in a MAP kinase-dependent way, thereby increasing chlamydial adherence and invasion. Gp96 co-precipitated with Protein Disulphide Isomerase (PDI), known to be involved in chlamydial host cell entry. During the replicative phase, Gp96 was depleted from infected cells and shed into the supernatant by activation of metalloproteinase TACE (ADAM17). Loss of Gp96 also reduced the activity of PDI on the cell surface. Reduced surface display of Gp96 prevented chlamydial re-infection in a TACE-dependent manner in cell lines but also in primary cells derived from human fimbriae, the natural site of chlamydial infection. Our data suggest a role of infection-induced Gp96 shedding in the protection of the chlamydial replicative niche.
Collapse
Affiliation(s)
- Karthika Karunakaran
- Department of Microbiology, Biocenter, University of Wuerzburg, Am Hubland, Wuerzburg, D-97074, Germany
| | - Prema Subbarayal
- Department of Microbiology, Biocenter, University of Wuerzburg, Am Hubland, Wuerzburg, D-97074, Germany
| | - Nadine Vollmuth
- Department of Microbiology, Biocenter, University of Wuerzburg, Am Hubland, Wuerzburg, D-97074, Germany
| | - Thomas Rudel
- Department of Microbiology, Biocenter, University of Wuerzburg, Am Hubland, Wuerzburg, D-97074, Germany
| |
Collapse
|
36
|
Meng Y, Zhang Q, Zhang M, Gu B, Huang G, Wang Q, Shan W. The protein disulfide isomerase 1 of Phytophthora parasitica (PpPDI1) is associated with the haustoria-like structures and contributes to plant infection. FRONTIERS IN PLANT SCIENCE 2015; 6:632. [PMID: 26347756 PMCID: PMC4539480 DOI: 10.3389/fpls.2015.00632] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Accepted: 07/30/2015] [Indexed: 05/23/2023]
Abstract
Protein disulfide isomerase (PDI) is a ubiquitous and multifunction enzyme belonging to the thioredoxin (TRX) superfamily, which can reduce, oxidize, and catalyze dithiol-disulfide exchange reactions. Other than performing housekeeping functions in helping to maintain proteins in a more stable conformation, there is some evidence to indicate that PDI is involved in pathogen infection processes. In a high-throughput screening for necrosis-inducing factors by Agrobacterium tumefaciens-mediated transient expression assay, a typical PDI gene from Phytophthora parasitica (PpPDI1) was identified and confirmed to induce strong cell death in Nicotiana benthamiana leaves. PpPDI1 is conserved in eukaryotes but predicted to be a secreted protein. Deletion mutant analyses showed that the first CGHC motif in the active domain of PpPDI1 is essential for inducing cell death. Using P. parasitica transformation method, the silencing efficiency was found to be very low, suggesting that PpPDI1 is essential for the pathogen. Translational fusion to the enhanced green fluorescent protein (EGFP) in stable P. parasitica transformants showed that PpPDI1 is associated with haustoria-like structures during pathogen infection. Furthermore, the PpPDI1-EGFP-expressing transformants increase the number of haustoria-like structures and exhibit enhanced virulence to N. benthamiana. These results indicate that PpPDI1 might be a virulence factor of P. parasitica and contributes to plant infection.
Collapse
Affiliation(s)
- Yuling Meng
- College of Plant Protection, Northwest A&F UniversityYangling, China
- State Key Laboratory of Crop Stress Biology for Arid Areas, Northwest A&F UniversityYangling, China
| | - Qiang Zhang
- College of Plant Protection, Northwest A&F UniversityYangling, China
- State Key Laboratory of Crop Stress Biology for Arid Areas, Northwest A&F UniversityYangling, China
| | - Meixiang Zhang
- College of Life Sciences, Northwest A&F UniversityYangling, China
| | - Biao Gu
- College of Plant Protection, Northwest A&F UniversityYangling, China
- State Key Laboratory of Crop Stress Biology for Arid Areas, Northwest A&F UniversityYangling, China
| | - Guiyan Huang
- State Key Laboratory of Crop Stress Biology for Arid Areas, Northwest A&F UniversityYangling, China
- College of Life Sciences, Northwest A&F UniversityYangling, China
| | - Qinhu Wang
- State Key Laboratory of Crop Stress Biology for Arid Areas, Northwest A&F UniversityYangling, China
- College of Life Sciences, Northwest A&F UniversityYangling, China
| | - Weixing Shan
- College of Plant Protection, Northwest A&F UniversityYangling, China
- State Key Laboratory of Crop Stress Biology for Arid Areas, Northwest A&F UniversityYangling, China
| |
Collapse
|
37
|
Jacquez P, Avila G, Boone K, Altiyev A, Puschhof J, Sauter R, Arigi E, Ruiz B, Peng X, Almeida I, Sherman M, Xiao C, Sun J. The Disulfide Bond Cys255-Cys279 in the Immunoglobulin-Like Domain of Anthrax Toxin Receptor 2 Is Required for Membrane Insertion of Anthrax Protective Antigen Pore. PLoS One 2015; 10:e0130832. [PMID: 26107617 PMCID: PMC4479931 DOI: 10.1371/journal.pone.0130832] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2015] [Accepted: 05/25/2015] [Indexed: 11/19/2022] Open
Abstract
Anthrax toxin receptors act as molecular clamps or switches that control anthrax toxin entry, pH-dependent pore formation, and translocation of enzymatic moieties across the endosomal membranes. We previously reported that reduction of the disulfide bonds in the immunoglobulin-like (Ig) domain of the anthrax toxin receptor 2 (ANTXR2) inhibited the function of the protective antigen (PA) pore. In the present study, the disulfide linkage in the Ig domain was identified as Cys255-Cys279 and Cys230-Cys315. Specific disulfide bond deletion mutants were achieved by replacing Cys residues with Ala residues. Deletion of the disulfide bond C255-C279, but not C230-C315, inhibited the PA pore-induced release of the fluorescence dyes from the liposomes, suggesting that C255-C279 is essential for PA pore function. Furthermore, we found that deletion of C255-C279 did not affect PA prepore-to-pore conversion, but inhibited PA pore membrane insertion by trapping the PA membrane-inserting loops in proteinaceous hydrophobic pockets. Fluorescence spectra of Trp59, a residue adjacent to the PA-binding motif in von Willebrand factor A (VWA) domain of ANTXR2, showed that deletion of C255-C279 resulted in a significant conformational change on the receptor ectodomain. The disulfide deletion-induced conformational change on the VWA domain was further confirmed by single-particle 3D reconstruction of the negatively stained PA-receptor heptameric complexes. Together, the biochemical and structural data obtained in this study provides a mechanistic insight into the role of the receptor disulfide bond C255-C279 in anthrax toxin action. Manipulation of the redox states of the receptor, specifically targeting to C255-C279, may become a novel strategy to treat anthrax.
Collapse
Affiliation(s)
- Pedro Jacquez
- Department of Biological Sciences & Border Biomedical Research Center, University of Texas at El Paso, 500 West University Avenue, El Paso, Texas, 79968, United States of America
| | - Gustavo Avila
- Department of Chemistry, University of Texas at El Paso, 500 West University Avenue, El Paso, Texas, 79968, United States of America
| | - Kyle Boone
- Bioinformatics Program of University of Texas at El Paso, 500 West University Avenue, El Paso, Texas, 79968, United States of America
| | - Agamyrat Altiyev
- Bioinformatics Program of University of Texas at El Paso, 500 West University Avenue, El Paso, Texas, 79968, United States of America
| | - Jens Puschhof
- Department of Biological Sciences & Border Biomedical Research Center, University of Texas at El Paso, 500 West University Avenue, El Paso, Texas, 79968, United States of America
| | - Roland Sauter
- Department of Biological Sciences & Border Biomedical Research Center, University of Texas at El Paso, 500 West University Avenue, El Paso, Texas, 79968, United States of America
| | - Emma Arigi
- Department of Biological Sciences & Border Biomedical Research Center, University of Texas at El Paso, 500 West University Avenue, El Paso, Texas, 79968, United States of America
| | - Blanca Ruiz
- Department of Biological Sciences & Border Biomedical Research Center, University of Texas at El Paso, 500 West University Avenue, El Paso, Texas, 79968, United States of America
| | - Xiuli Peng
- China National Key Laboratory of Agricultural Microbiology, Huazhong Agriculture University, Wuhan, 430070, P. R. China
| | - Igor Almeida
- Department of Biological Sciences & Border Biomedical Research Center, University of Texas at El Paso, 500 West University Avenue, El Paso, Texas, 79968, United States of America
| | - Michael Sherman
- Department of Biochemistry and Molecular Biology, Sealy Center for Structural Biology and Molecular Biophysics, University of Texas Medical Branch, Galveston, Texas, 77555, United States of America
| | - Chuan Xiao
- Department of Chemistry, University of Texas at El Paso, 500 West University Avenue, El Paso, Texas, 79968, United States of America
- * E-mail: (CX); (JS)
| | - Jianjun Sun
- Department of Biological Sciences & Border Biomedical Research Center, University of Texas at El Paso, 500 West University Avenue, El Paso, Texas, 79968, United States of America
- * E-mail: (CX); (JS)
| |
Collapse
|
38
|
Quintero CA, Tudela JG, Damiani MT. Rho GTPases as pathogen targets: Focus on curable sexually transmitted infections. Small GTPases 2015; 6:108-18. [PMID: 26023809 DOI: 10.4161/21541248.2014.991233] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Pathogens have evolved highly specialized mechanisms to infect hosts. Several microorganisms modulate the eukaryotic cell surface to facilitate their engulfment. Once internalized, they hijack the molecular machinery of the infected cell for their own benefit. At different stages of phagocytosis, particularly during invasion, certain pathogens manipulate pathways governed by small GTPases. In this review, we focus on the role of Rho proteins on curable, sexually transmitted infections caused by Chlamydia trachomatis, Neisseria gonorrhoeae, Trichomonas vaginalis and Treponema pallidum. Despite the high, worldwide frequencies of these sexually-transmitted diseases, very little is known about the strategies developed by these microorganisms to usurp key eukaryotic proteins that control intracellular signaling and actin dynamics. Improved knowledge of these molecular mechanisms will contribute to the elucidation of how these clinically important pathogens manipulate intracellular processes and parasitize their hosts.
Collapse
Affiliation(s)
- Cristián A Quintero
- a Laboratory of Phagocytosis and Intracellular Trafficking; IHEM-CONICET; School of Medicine; University of Cuyo ; Mendoza , Argentina
| | | | | |
Collapse
|
39
|
Subbarayal P, Karunakaran K, Winkler AC, Rother M, Gonzalez E, Meyer TF, Rudel T. EphrinA2 receptor (EphA2) is an invasion and intracellular signaling receptor for Chlamydia trachomatis. PLoS Pathog 2015; 11:e1004846. [PMID: 25906164 PMCID: PMC4408118 DOI: 10.1371/journal.ppat.1004846] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2014] [Accepted: 04/01/2015] [Indexed: 12/02/2022] Open
Abstract
The obligate intracellular bacterium Chlamydia trachomatis invades into host cells to replicate inside a membrane-bound vacuole called inclusion. Multiple different host proteins are recruited to the inclusion and are functionally modulated to support chlamydial development. Invaded and replicating Chlamydia induces a long-lasting activation of the PI3 kinase signaling pathway that is required for efficient replication. We identified the cell surface tyrosine kinase EphrinA2 receptor (EphA2) as a chlamydial adherence and invasion receptor that induces PI3 kinase (PI3K) activation, promoting chlamydial replication. Interfering with binding of C. trachomatis serovar L2 (Ctr) to EphA2, downregulation of EphA2 expression or inhibition of EphA2 activity significantly reduced Ctr infection. Ctr interacts with and activates EphA2 on the cell surface resulting in Ctr and receptor internalization. During chlamydial replication, EphA2 remains active accumulating around the inclusion and interacts with the p85 regulatory subunit of PI3K to support the activation of the PI3K/Akt signaling pathway that is required for normal chlamydial development. Overexpression of full length EphA2, but not the mutant form lacking the intracellular cytoplasmic domain, enhanced PI3K activation and Ctr infection. Despite the depletion of EphA2 from the cell surface, Ctr infection induces upregulation of EphA2 through the activation of the ERK pathway, which keeps the infected cell in an apoptosis-resistant state. The significance of EphA2 as an entry and intracellular signaling receptor was also observed with the urogenital C. trachomatis-serovar D. Our findings provide the first evidence for a host cell surface receptor that is exploited for invasion as well as for receptor-mediated intracellular signaling to facilitate chlamydial replication. In addition, the engagement of a cell surface receptor at the inclusion membrane is a new mechanism by which Chlamydia subverts the host cell and induces apoptosis resistance. Chlamydia trachomatis are major human pathogens causing ocular and sexually transmitted diseases with hundreds of millions of cases per year. Chlamydia replicate inside the host cell in a membrane bound vacuole called inclusion. The current concept on how Chlamydia communicates with the host cell during its replication is based on the identification of the host protein that interacts with Chlamydia. Here, we describe that C. trachomatis-serovar L2 and D use EphA2, a member of the largest class of human receptor tyrosine kinases, as an adherence and entry receptor that is endocytosed together with the bacteria. Cell surface EphA2 receptor is adopted by Chlamydia to function also at the inclusion to support growth and replication and to keep the infected cell in an apoptosis resistant state. Thus, we show that EphA2 is an undiscovered important surface and intracellular signaling receptor that is crucial for chlamydial infection and development.
Collapse
Affiliation(s)
- Prema Subbarayal
- Department of Microbiology, Biocenter, University of Wuerzburg, Wuerzburg, Germany
| | - Karthika Karunakaran
- Department of Microbiology, Biocenter, University of Wuerzburg, Wuerzburg, Germany
| | - Ann-Cathrin Winkler
- Department of Microbiology, Biocenter, University of Wuerzburg, Wuerzburg, Germany
| | - Marion Rother
- Department of Molecular Biology, Max Planck Institute for Infection Biology, Berlin, Germany; Steinbeis Innovation gGmbH, Center for Systems Biomedicine, Stuttgart, Germany
| | - Erik Gonzalez
- Department of Molecular Biology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Thomas F Meyer
- Department of Molecular Biology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Thomas Rudel
- Department of Microbiology, Biocenter, University of Wuerzburg, Wuerzburg, Germany
| |
Collapse
|
40
|
|
41
|
Ishida K, Matsuo J, Yamamoto Y, Yamaguchi H. Chlamydia pneumoniae effector chlamydial outer protein N sequesters fructose bisphosphate aldolase A, providing a benefit to bacterial growth. BMC Microbiol 2014; 14:330. [PMID: 25528659 PMCID: PMC4302594 DOI: 10.1186/s12866-014-0330-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Accepted: 12/16/2014] [Indexed: 01/13/2023] Open
Abstract
Background Pathogenic chlamydiae are obligate intracellular pathogens and have adapted successfully to human cells, causing sexually transmitted diseases or pneumonia. Chlamydial outer protein N (CopN) is likely a critical effector protein secreted by the type III secretion system in chlamydiae, which manipulates host cells. However, the mechanisms of its action remain to be clarified. In this work, we aimed to identify previously unidentified CopN effector target in host cells. Results We first performed a pull-down assay with recombinant glutathione S-transferase (GST) fusion CopN proteins (GST–CpCopN: Chlamydia pneumoniae TW183, GST–CtCopN: Chlamydia trachomatis D/UW-3/CX) as “bait” and soluble lysates obtained from human immortal epithelial HEp-2 cells as “prey”, followed by SDS-PAGE with mass spectroscopy (MS). We found that a host cell protein specifically bound to GST–CpCopN, but not GST–CtCopN. MS revealed the host protein to be fructose bisphosphate aldolase A (aldolase A), which plays a key role in glycolytic metabolism. We also confirmed the role of aldolase A in chlamydia-infected HEp-2 cells by using two distinct experiments for gene knockdown with an siRNA specific to aldolase A transcripts, and for assessment of glycolytic enzyme gene expression levels. As a result, both the numbers of chlamydial inclusion-forming units and RpoD transcripts were increased in the chlamydia-infected aldolase A knockdown cells, as compared with the wild-type HEp-2 cells. Meanwhile, chlamydial infection tended to enhance expression of aldolase A. Conclusions We discovered that one of the C. pneumoniae CopN targets is the glycolytic enzyme aldolase A. Sequestering aldolase A may be beneficial to bacterial growth in infected host cells.
Collapse
Affiliation(s)
- Kasumi Ishida
- Department of Medical Laboratory Science, Faculty of Health Sciences, Hokkaido University, Sapporo, Hokkaido, 060-0812, Japan. .,Research Fellow of Japan Society for the Promotion of Science, Tokyo, 102-0083, Japan.
| | - Junji Matsuo
- Department of Medical Laboratory Science, Faculty of Health Sciences, Hokkaido University, Sapporo, Hokkaido, 060-0812, Japan.
| | - Yoshimasa Yamamoto
- Department of Biomedical Informatics, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan. .,Japan Science and Technology Agency/Japan International Cooperation Agency, Science and Technology Research Partnership for Sustainable Development (JST/JICA, SATREPS), Osaka, Japan. .,Osaka Prefectural Institute of Public Health, Higashinari-ku, Osaka, 537-0025, Japan.
| | - Hiroyuki Yamaguchi
- Department of Medical Laboratory Science, Faculty of Health Sciences, Hokkaido University, Sapporo, Hokkaido, 060-0812, Japan.
| |
Collapse
|
42
|
Knittler MR, Sachse K. Chlamydia psittaci: update on an underestimated zoonotic agent. Pathog Dis 2014; 73:1-15. [PMID: 25853998 DOI: 10.1093/femspd/ftu007] [Citation(s) in RCA: 149] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/24/2014] [Indexed: 12/16/2022] Open
Abstract
Chlamydia (C.) psittaci is an economically relevant pathogen in poultry and pet birds, where it causes psittacosis/ornithosis, and also a human pathogen causing atypical pneumonia after zoonotic transmission. Despite its well-documented prevalence, the agent has received less attention by researchers than other Chlamydia spp. in the last decades. In the present paper, we review recently published data on C. psittaci infection and attempt to single out characteristic features distinguishing it from related chlamydial agents. It is remarkable that C. psittaci is particularly efficient in disseminating in the host organism causing systemic disease, which occasionally can take a fulminant course. At the cellular level, the pathogen's broad host cell spectrum (from epithelial cells to macrophages), its rapid entry and fast replication, proficient use of intracellular transport routes to mitochondria and the Golgi apparatus, the pronounced physical association of chlamydial inclusions with energy-providing cell compartments, as well as the subversive regulation of host cell survival during productive and persistent states facilitate the characteristic efficient growth and successful host-to-host spread of C. psittaci. At the molecular level, the pathogen was shown to upregulate essential chlamydial genes when facing the host immune response. We hypothesize that this capacity, in concert with expression of specific effectors of the type III secretion system and efficient suppression of selected host defense signals, contributes to successful establishment of the infection in the host. Concerning the immunology of host-pathogen interactions, C. psittaci has been shown to distinguish itself by coping more efficiently than other chlamydiae with pro-inflammatory mediators during early host response, which can, to some extent, explain the effective evasion and adaptation strategies of this bacterium. We conclude that thorough analysis of the large number of whole-genome sequences already available will be essential to identify genetic markers of the species-specific features and trigger more in-depth studies in cellular and animal models to address such vital topics as treatment and vaccination.
Collapse
Affiliation(s)
- Michael R Knittler
- Institute of Immunology, Friedrich-Loeffler-Institut, Isle of Riems, 07743 Jena, Germany
| | - Konrad Sachse
- Institute of Molecular Pathogenesis, Friedrich-Loeffler-Institut, 07743 Jena, Germany
| |
Collapse
|
43
|
Pan S, Chen HH, Correia C, Dai H, Witt TA, Kleppe LS, Burnett JC, Simari RD. Cell surface protein disulfide isomerase regulates natriuretic peptide generation of cyclic guanosine monophosphate. PLoS One 2014; 9:e112986. [PMID: 25419565 PMCID: PMC4242536 DOI: 10.1371/journal.pone.0112986] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2013] [Accepted: 10/21/2014] [Indexed: 12/21/2022] Open
Abstract
Rationale The family of natriuretic peptides (NPs), including atrial natriuretic peptide (ANP), B-type natriuretic peptide (BNP), and C-type natriuretic peptide (CNP), exert important and diverse actions for cardiovascular and renal homeostasis. The autocrine and paracrine functions of the NPs are primarily mediated through the cellular membrane bound guanylyl cyclase-linked receptors GC-A (NPR-A) and GC-B (NPR-B). As the ligands and receptors each contain disulfide bonds, a regulatory role for the cell surface protein disulfide isomerase (PDI) was investigated. Objective We utilized complementary in vitro and in vivo models to determine the potential role of PDI in regulating the ability of the NPs to generate its second messenger, cyclic guanosine monophosphate. Methods and Results Inhibition of PDI attenuated the ability of ANP, BNP and CNP to generate cGMP in human mesangial cells (HMCs), human umbilical vein endothelial cells (HUVECs), and human aortic smooth muscle cells (HASMCs), each of which were shown to express PDI. In LLC-PK1 cells, where PDI expression was undetectable by immunoblotting, PDI inhibition had a minimal effect on cGMP generation. Addition of PDI to cultured LLC-PK1 cells increased intracellular cGMP generation mediated by ANP. Inhibition of PDI in vivo attenuated NP-mediated generation of cGMP by ANP. Surface Plasmon Resonance demonstrated modest and differential binding of the natriuretic peptides with immobilized PDI in a cell free system. However, PDI was shown to co-localize on the surface of cells with GC-A and GC-B by co-immunoprecpitation and immunohistochemistry. Conclusion These data demonstrate for the first time that cell surface PDI expression and function regulate the capacity of natriuretic peptides to generate cGMP through interaction with their receptors.
Collapse
Affiliation(s)
- Shuchong Pan
- Division of Cardiovascular Diseases, Mayo Clinic, Rochester, MN, United States of America
| | - Horng H. Chen
- Division of Cardiovascular Diseases, Mayo Clinic, Rochester, MN, United States of America
| | - Cristina Correia
- Division of Oncology Research, Mayo Clinic, Rochester, MN, United States of America
| | - Haiming Dai
- Division of Oncology Research, Mayo Clinic, Rochester, MN, United States of America
| | - Tyra A. Witt
- Division of Cardiovascular Diseases, Mayo Clinic, Rochester, MN, United States of America
| | - Laurel S. Kleppe
- Division of Cardiovascular Diseases, Mayo Clinic, Rochester, MN, United States of America
| | - John C. Burnett
- Division of Cardiovascular Diseases, Mayo Clinic, Rochester, MN, United States of America
| | - Robert D. Simari
- Division of Cardiovascular Diseases, Mayo Clinic, Rochester, MN, United States of America
- * E-mail:
| |
Collapse
|
44
|
Abstract
A defining characteristic of Chlamydia spp. is their developmental cycle characterized by outer membrane transformations of cysteine bonds among cysteine-rich outer membrane proteins. The reduction-oxidation states of host cell compartments were monitored during the developmental cycle using live fluorescence microscopy. Organelle redox states were studied using redox-sensitive green fluorescent protein (roGFP1) expressed in CF15 epithelial cells and targeted to the cytosol, mitochondria, and endoplasmic reticulum (ER). The redox properties of chlamydiae and the inclusion were monitored using roGFP expressed by Chlamydia trachomatis following transformation. Despite the large morphological changes associated with chlamydial infection, redox potentials of the cytosol (Ψcyto [average, −320 mV]), mitochondria (Ψmito [average, −345 mV]), and the ER (ΨER [average, −258 mV]) and their characteristic redox regulatory abilities remained unchanged until the cells died, at which point Ψcyto and Ψmito became more oxidized and ΨER became more reduced. The redox status of the chamydial cytoplasm was measured following transformation and expression of the roGFP biosensor in C. trachomatis throughout the developmental cycle. The periplasmic and outer membrane redox states were assessed by the level of cysteine cross-linking of cysteine-rich envelope proteins. In both cases, the chlamydiae were highly reduced early in the developmental cycle and became oxidized late in the developmental cycle. The production of a late-developmental-stage oxidoreductase/isomerase, DsbJ, may play a key role in the regulation of the oxidoreductive developmental-stage-specific process. Infectious Chlamydia organisms have highly oxidized and cysteine cross-linked membrane proteins that confer environmental stability when outside their host cells. Once these organisms infect a new host cell, the proteins become reduced and remain reduced during the active growth stage. These proteins become oxidized at the end of their growth cycle, wherein infectious organisms are produced and released to the environment. How chlamydiae mediate and regulate this key step in their pathogenesis is unknown. Using biosensors specifically targeted to different compartments within the infected host cell and for the chlamydial organisms themselves, the oxidoreductive states of these compartments were measured during the course of infection. We found that the host cell redox states are not changed by infection with C. trachomatis, whereas the state of the chlamydial organisms remains reduced during infection until the late developmental stages, wherein the organisms’ cytosol and periplasm become oxidized and they acquire environmental resistance and infectivity.
Collapse
|
45
|
Feng F, Chen L, Lian C, Xia H, Zhou Y, Yao Q, Chen K. Comparative proteomic analysis reveals the suppressive effects of dietary high glucose on the midgut growth of silkworm. J Proteomics 2014; 108:124-32. [PMID: 24878427 DOI: 10.1016/j.jprot.2014.05.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2014] [Revised: 05/16/2014] [Accepted: 05/18/2014] [Indexed: 10/25/2022]
Abstract
UNLABELLED The silkworm, Bombyx mori, is an important model of lepidoptera insect, and it has been used for several models of human diseases. In human being, long-term high-sugar diet can induce the occurrence of diabetes and other related diseases. Interestingly, our experiments revealed the high glucose diet also has a suppressive effect on the development of silkworms. To investigate the molecular mechanism by which high-glucose diet inhibited the midgut growth in silkworms, we employed comparative proteomic analysis to globally identify proteins differentially expressed in normal and high-glucose diet group silkworms. In all, 28 differently proteins were suppressed and 5 proteins induced in high-glucose diet group. Gene ontology analysis showed that most of these differently proteins are mainly involved in metabolic process, catalytic and cellular process. A development related protein, imaginal disk growth factor (IDGF), was further confirmed by western blot exclusively expressing in the normal diet group silkworms. Taken together, our data suggests that IDGF plays a critical role in impairing the development of silkworms by a high-glucose diet. BIOLOGICAL SIGNIFICANCE Glucose has been thought to play essential roles in growth and development of silkworm. In this paper, we certified firstly that high-glucose diet can suppress the growth of silkworm, and comparative proteomic was employed to reveal the inhibition mechanism. Moreover, an important regulation related protein (IDGF) was found to involve in this inhibition process. These results will help us get a deeper understanding of the relationship between diet and healthy. Furthermore, IDGF may be the critical protein for reducing the blood sugar in silkworm, and it may be used for screening human hypoglycemic drug. The work has not been submitted elsewhere for publication, in whole or in part, and all the authors have approved the manuscript.
Collapse
Affiliation(s)
- Fan Feng
- School of Food and Biological Engineering, Jiangsu University, 301# Xuefu Road, Zhenjiang, Jiangsu Province 212013, PR China; Institute of Life Sciences, Jiangsu University, 301# Xuefu Road, Zhenjiang, Jiangsu Province 212013, PR China
| | - Liang Chen
- Institute of Life Sciences, Jiangsu University, 301# Xuefu Road, Zhenjiang, Jiangsu Province 212013, PR China
| | - Chaoqun Lian
- School of Food and Biological Engineering, Jiangsu University, 301# Xuefu Road, Zhenjiang, Jiangsu Province 212013, PR China
| | - Hengchuan Xia
- Institute of Life Sciences, Jiangsu University, 301# Xuefu Road, Zhenjiang, Jiangsu Province 212013, PR China
| | - Yang Zhou
- Institute of Life Sciences, Jiangsu University, 301# Xuefu Road, Zhenjiang, Jiangsu Province 212013, PR China
| | - Qin Yao
- Institute of Life Sciences, Jiangsu University, 301# Xuefu Road, Zhenjiang, Jiangsu Province 212013, PR China
| | - Keping Chen
- School of Food and Biological Engineering, Jiangsu University, 301# Xuefu Road, Zhenjiang, Jiangsu Province 212013, PR China; Institute of Life Sciences, Jiangsu University, 301# Xuefu Road, Zhenjiang, Jiangsu Province 212013, PR China.
| |
Collapse
|
46
|
Nunes A, Gomes JP. Evolution, phylogeny, and molecular epidemiology of Chlamydia. INFECTION GENETICS AND EVOLUTION 2014; 23:49-64. [PMID: 24509351 DOI: 10.1016/j.meegid.2014.01.029] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2013] [Revised: 01/16/2014] [Accepted: 01/19/2014] [Indexed: 10/25/2022]
Abstract
The Chlamydiaceae are a family of obligate intracellular bacteria characterized by a unique biphasic developmental cycle. It encompasses the single genus Chlamydia, which involves nine species that affect a wide range of vertebral hosts, causing infections with serious impact on human health (mainly due to Chlamydia trachomatis infections) and on farming and veterinary industries. It is believed that Chlamydiales originated ∼700mya, whereas C. trachomatis likely split from the other Chlamydiaceae during the last 6mya. This corresponds to the emergence of modern human lineages, with the first descriptions of chlamydial infections as ancient as four millennia. Chlamydiaceae have undergone a massive genome reduction, on behalf of the deletional bias "use it or lose it", stabilizing at 1-1.2Mb and keeping a striking genome synteny. Their phylogeny reveals species segregation according to biological properties, with huge differences in terms of host range, tissue tropism, and disease outcomes. Genome differences rely on the occurrence of mutations in the >700 orthologous genes, as well as on events of recombination, gene loss, inversion, and paralogous expansion, affecting both a hypervariable region named the plasticity zone, and genes essentially encoding polymorphic and transmembrane head membrane proteins, type III secretion effectors and some metabolic pathways. Procedures for molecular typing are still not consensual but have allowed the knowledge of molecular epidemiology patterns for some species as well as the identification of outbreaks and emergence of successful clones for C. trachomatis. This manuscript intends to provide a comprehensive review on the evolution, phylogeny, and molecular epidemiology of Chlamydia.
Collapse
Affiliation(s)
- Alexandra Nunes
- Reference Laboratory of Bacterial Sexually Transmitted Infections and Bioinformatics Unit, Department of Infectious Diseases, National Institute of Health, Av. Padre Cruz, 1649-016 Lisbon, Portugal
| | - João P Gomes
- Reference Laboratory of Bacterial Sexually Transmitted Infections and Bioinformatics Unit, Department of Infectious Diseases, National Institute of Health, Av. Padre Cruz, 1649-016 Lisbon, Portugal.
| |
Collapse
|
47
|
Zhou Z, Liu H, Gu G, Wang G, Wu W, Zhang C, Ren J. Immunoproteomic to identify antigens in the intestinal mucosa of Crohn's disease patients. PLoS One 2013; 8:e81662. [PMID: 24358121 PMCID: PMC3864798 DOI: 10.1371/journal.pone.0081662] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2013] [Accepted: 10/15/2013] [Indexed: 12/29/2022] Open
Abstract
Incidences of Crohn disease (CD) have increased significantly in the last decade. Immunoproteomics are a promising method to identify biomarkers of different diseases. In the present study, we used immunoproteomics to study proteins of intestinal mucosal lesions and neighboring normal intestinal mucosa of 8 CD patients. Reactive proteins were validated by Western blotting. Approximately 50 protein spots localized in the 4 to 7 pI range were detected on two-dimensional electrophoresis gels, and 6 differentially expressed protein spots between 10 and 100 kDa were identified. Reactive proteins were identified as prohibitin, calreticulin, apolipoprotein A-I, intelectin-1, protein disulfide isomerase, and glutathione s-transferase Pi. Western blotting was conducted on the intestinal mucosa of another 4 CD patients to validate the reactive proteins. We found that intestinal mucosal lesions had high levels of prohibitin expression. Glutathione s-transferase expression was detected in 100% of the intestinal mucosa examined. Thus, we report 6 autoantigens of CD, including 3 new and 3 previously reported autoantigens. Intelectin-1, protein disulfide isomerase, and glutathione-s-transferases may be used as biomarkers for CD pathogenesis.
Collapse
Affiliation(s)
- Zheng Zhou
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, Anhui, Hefei, P.R. China
| | - Haiyan Liu
- Department of Critical Care, First Affiliated Hospital of Anhui Medical University, Anhui, Hefei, P.R. China
| | - Guosheng Gu
- Research Institute of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, P.R. China
| | - Gefei Wang
- Research Institute of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, P.R. China
| | - Wenyong Wu
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, Anhui, Hefei, P.R. China
| | - Changle Zhang
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, Anhui, Hefei, P.R. China
| | - Jianan Ren
- Research Institute of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, P.R. China
- * E-mail:
| |
Collapse
|
48
|
Bastidas RJ, Elwell CA, Engel JN, Valdivia RH. Chlamydial intracellular survival strategies. Cold Spring Harb Perspect Med 2013; 3:a010256. [PMID: 23637308 DOI: 10.1101/cshperspect.a010256] [Citation(s) in RCA: 164] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Chlamydia trachomatis is the most common sexually transmitted bacterial pathogen and the causative agent of blinding trachoma. Although Chlamydia is protected from humoral immune responses by residing within remodeled intracellular vacuoles, it still must contend with multilayered intracellular innate immune defenses deployed by its host while scavenging for nutrients. Here we provide an overview of Chlamydia biology and highlight recent findings detailing how this vacuole-bound pathogen manipulates host-cellular functions to invade host cells and maintain a replicative niche.
Collapse
Affiliation(s)
- Robert J Bastidas
- Department of Molecular Genetics and Microbiology, Center for Microbial Pathogenesis, Duke University Medical Center, Durham, NC 27710, USA
| | | | | | | |
Collapse
|
49
|
Schust DJ, Ibana JA, Buckner LR, Ficarra M, Sugimoto J, Amedee AM, Quayle AJ. Potential mechanisms for increased HIV-1 transmission across the endocervical epithelium during C. trachomatis infection. Curr HIV Res 2012; 10:218-27. [PMID: 22384841 DOI: 10.2174/157016212800618093] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2011] [Revised: 12/18/2011] [Accepted: 12/28/2011] [Indexed: 11/22/2022]
Abstract
Among the now pandemic sexually transmitted infections (STIs), Chlamydia trachomatis (C. trachomatis) is the predominant bacterial pathogen and human immunodeficiency virus type 1 (HIV-1) is the most lethal of the viral pathogens. The female genital tract is the primary site for heterosexual transmission of both C. trachomatis and HIV-1. Infection with C. trachomatis, and with a variety of other STIs, increases the risk for transmission of HIV-1, although the mechanisms for this finding remain unclear. We have used in vitro modeling to assess the mechanisms by which infection with genital C. trachomatis serovars might increase the transmission of HIV-1 across the female genital tract. C. trachomatis infection of an immortalized endocervical epithelial cell line (A2EN) increases the cell surface expression of the HIV-1 alternative primary receptor, galactosyl ceramide (GalCer), and of the HIV-1 co-receptors, CXCR4 and CCR5. C. trachomatis infection also increases the binding of HIV-1 to A2EN cells, and, subsequently, increases levels of virus in co-cultures of HIV-exposed A2EN and susceptible MT4-R5 T cells. Finally, in vivo endocervical cell sampling reveals a dramatic increase in the number of CD4+, CXCR4 and/or CCR5 positive T cell targets in the endocervix of C. trachomatis positive women when compared to those who are C. trachomatis negative. This combination of in vitro and in vivo results suggests several mechanisms for increased transmission of HIV-1 across the endocervices of C. trachomatis-infected women.
Collapse
Affiliation(s)
- Danny J Schust
- Department of Obstetrics, Gynecology and Women's Health, University of Missouri School of Medicine, Columbia, MO, USA.
| | | | | | | | | | | | | |
Collapse
|
50
|
WASS MN, STANWAY R, BLAGBOROUGH AM, LAL K, PRIETO JH, RAINE D, STERNBERG MJE, TALMAN AM, TOMLEY F, YATES J, SINDEN RE. Proteomic analysis of Plasmodium in the mosquito: progress and pitfalls. Parasitology 2012; 139:1131-45. [PMID: 22336136 PMCID: PMC3417538 DOI: 10.1017/s0031182012000133] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2011] [Revised: 01/10/2012] [Accepted: 01/11/2012] [Indexed: 11/06/2022]
Abstract
Here we discuss proteomic analyses of whole cell preparations of the mosquito stages of malaria parasite development (i.e. gametocytes, microgamete, ookinete, oocyst and sporozoite) of Plasmodium berghei. We also include critiques of the proteomes of two cell fractions from the purified ookinete, namely the micronemes and cell surface. Whereas we summarise key biological interpretations of the data, we also try to identify key methodological constraints we have met, only some of which we were able to resolve. Recognising the need to translate the potential of current genome sequencing into functional understanding, we report our efforts to develop more powerful combinations of methods for the in silico prediction of protein function and location. We have applied this analysis to the proteome of the male gamete, a cell whose very simple structural organisation facilitated interpretation of data. Some of the in silico predictions made have now been supported by ongoing protein tagging and genetic knockout studies. We hope this discussion may assist future studies.
Collapse
Affiliation(s)
- M. N. WASS
- The Centre for Bioinformatics, Department of Life Sciences, Imperial College, London SW7 2AZ
| | - R. STANWAY
- The Malaria Centre, Department of Life Sciences, Imperial College, London SW7 2AZ
- University of Bern, Institute of Cell Biology, Baltzerstrasse 4, CH-3012 Bern
| | - A. M. BLAGBOROUGH
- The Malaria Centre, Department of Life Sciences, Imperial College, London SW7 2AZ
| | - K. LAL
- The Malaria Centre, Department of Life Sciences, Imperial College, London SW7 2AZ
| | - J. H. PRIETO
- The Scripps Research Institute, 10550 North Torrey Pines Rd., Department of Chemical Physiology, SR11 , La Jolla, CA 92037
| | - D. RAINE
- The Malaria Centre, Department of Life Sciences, Imperial College, London SW7 2AZ
| | - M. J. E. STERNBERG
- The Centre for Bioinformatics, Department of Life Sciences, Imperial College, London SW7 2AZ
| | - A. M. TALMAN
- The Malaria Centre, Department of Life Sciences, Imperial College, London SW7 2AZ
- Department of Microbial Pathogenesis, Yale University School of Medicine, 295 Congress Avenue, New Haven, CT 06519
| | - F. TOMLEY
- Pathology & Infectious Diseases, The Royal Veterinary College, Hawkshead Lane, North Mymms, Hatfield, Hertfordshire AL9 7TA
| | - J. YATES
- The Scripps Research Institute, 10550 North Torrey Pines Rd., Department of Chemical Physiology, SR11 , La Jolla, CA 92037
| | - R. E. SINDEN
- The Malaria Centre, Department of Life Sciences, Imperial College, London SW7 2AZ
| |
Collapse
|