1
|
Walocha R, Kim M, Wong-Ng J, Gobaa S, Sauvonnet N. Organoids and organ-on-chip technology for investigating host-microorganism interactions. Microbes Infect 2024; 26:105319. [PMID: 38447861 DOI: 10.1016/j.micinf.2024.105319] [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: 10/03/2023] [Revised: 02/26/2024] [Accepted: 03/01/2024] [Indexed: 03/08/2024]
Abstract
Recent advances in organoid and organ-on-chip (OoC) technologies offer an unprecedented level of tissue mimicry. These models can recapitulate the diversity of cellular composition, 3D organization, and mechanical stimulation. These approaches are intensively used to understand complex diseases. This review focuses on the latest advances in this field to study host-microorganism interactions.
Collapse
Affiliation(s)
- Remigiusz Walocha
- Tissue Homeostasis Group, Biomaterials and Microfluidics Core Facility, Institut Pasteur, Université Paris Cité, Paris, France; Biomaterials and Microfluidics Core Facility, Institut Pasteur, Université Paris Cité, Paris, France
| | - MinHee Kim
- Biomaterials and Microfluidics Core Facility, Institut Pasteur, Université Paris Cité, Paris, France
| | - Jérôme Wong-Ng
- Biomaterials and Microfluidics Core Facility, Institut Pasteur, Université Paris Cité, Paris, France
| | - Samy Gobaa
- Biomaterials and Microfluidics Core Facility, Institut Pasteur, Université Paris Cité, Paris, France
| | - Nathalie Sauvonnet
- Tissue Homeostasis Group, Biomaterials and Microfluidics Core Facility, Institut Pasteur, Université Paris Cité, Paris, France; Biomaterials and Microfluidics Core Facility, Institut Pasteur, Université Paris Cité, Paris, France.
| |
Collapse
|
2
|
De Gaetano GV, Lentini G, Coppolino F, Famà A, Pietrocola G, Beninati C. Engagement of α 3β 1 and α 2β 1 integrins by hypervirulent Streptococcus agalactiae in invasion of polarized enterocytes. Front Microbiol 2024; 15:1367898. [PMID: 38511003 PMCID: PMC10951081 DOI: 10.3389/fmicb.2024.1367898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 02/19/2024] [Indexed: 03/22/2024] Open
Abstract
The gut represents an important site of colonization of the commensal bacterium Streptococcus agalactiae (group B Streptococcus or GBS), which can also behave as a deadly pathogen in neonates and adults. Invasion of the intestinal epithelial barrier is likely a crucial step in the pathogenesis of neonatal infections caused by GBS belonging to clonal complex 17 (CC17). We have previously shown that the prototypical CC17 BM110 strain invades polarized enterocyte-like cells through their lateral surfaces using an endocytic pathway. By analyzing the cellular distribution of putative GBS receptors in human enterocyte-like Caco-2 cells, we find here that the alpha 3 (α3) and alpha 2 (α2) integrin subunits are selectively expressed on lateral enterocyte surfaces at equatorial and parabasal levels along the vertical axis of polarized cells, in an area corresponding to GBS entry sites. The α3β1 and α2β1 integrins were not readily accessible in fully differentiated Caco-2 monolayers but could be exposed to specific antibodies after weakening of intercellular junctions in calcium-free media. Under these conditions, anti-α3β1 and anti-α2β1 antibodies significantly reduced GBS adhesion to and invasion of enterocytes. After endocytosis, α3β1 and α2β1 integrins localized to areas of actin remodeling around GBS containing vacuoles. Taken together, these data indicate that GBS can invade enterocytes by binding to α3β1 and α2β1 integrins on the lateral membrane of polarized enterocytes, resulting in cytoskeletal remodeling and bacterial internalization. Blocking integrins might represent a viable strategy to prevent GBS invasion of gut epithelial tissues.
Collapse
Affiliation(s)
| | - Germana Lentini
- Department of Human Pathology, University of Messina, Messina, Italy
| | - Francesco Coppolino
- Department of Biomedical, Dental and Imaging Sciences, University of Messina, Messina, Italy
| | - Agata Famà
- Department of Human Pathology, University of Messina, Messina, Italy
| | - Giampiero Pietrocola
- Department of Molecular Medicine, Biochemistry Section, University of Pavia, Pavia, Italy
| | - Concetta Beninati
- Department of Human Pathology, University of Messina, Messina, Italy
- Scylla Biotech Srl, Messina, Italy
| |
Collapse
|
3
|
Gabor CE, Hazen TH, Delaine-Elias BC, Rasko DA, Barry EM. Genomic, transcriptomic, and phenotypic differences among archetype Shigella flexneri strains of serotypes 2a, 3a, and 6. mSphere 2023; 8:e0040823. [PMID: 37830809 PMCID: PMC10732043 DOI: 10.1128/msphere.00408-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 08/30/2023] [Indexed: 10/14/2023] Open
Abstract
IMPORTANCE Given the genomic diversity between S. flexneri serotypes and the paucity of data to support serotype-specific phenotypic differences, we applied in silico and in vitro functional analyses of archetype strains of 2457T (Sf2a), J17B (Sf3a), and CH060 (Sf6). These archetype strains represent the three leading S. flexneri serotypes recommended for inclusion in multivalent vaccines. Characterizing the genomic and phenotypic variation among these clinically prevalent serotypes is an important step toward understanding serotype-specific host-pathogen interactions to optimize the efficacy of multivalent vaccines and therapeutics. This study underpins the importance for further large-scale serotype-targeted analyses.
Collapse
Affiliation(s)
- Caitlin E. Gabor
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Tracy H. Hazen
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - BreOnna C. Delaine-Elias
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - David A. Rasko
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Center for Pathogen Research, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Eileen M. Barry
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
4
|
Matanza XM, Clements A. Pathogenicity and virulence of Shigella sonnei: A highly drug-resistant pathogen of increasing prevalence. Virulence 2023; 14:2280838. [PMID: 37994877 PMCID: PMC10732612 DOI: 10.1080/21505594.2023.2280838] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 11/01/2023] [Indexed: 11/24/2023] Open
Abstract
Shigella spp. are the causative agent of shigellosis (or bacillary dysentery), a diarrhoeal disease characterized for the bacterial invasion of gut epithelial cells. Among the 4 species included in the genus, Shigella flexneri is principally responsible for the disease in the developing world while Shigella sonnei is the main causative agent in high-income countries. Remarkably, as more countries improve their socioeconomic conditions, we observe an increase in the relative prevalence of S. sonnei. To date, the reasons behind this change in aetiology depending on economic growth are not understood. S. flexneri has been widely used as a model to study the pathogenesis of the genus, but as more research data are collected, important discrepancies with S. sonnei have come to light. In comparison to S. flexneri, S. sonnei can be differentiated in numerous aspects; it presents a characteristic O-antigen identical to that of one serogroup of the environmental bacterium Plesiomonas shigelloides, a group 4 capsule, antibacterial mechanisms to outcompete and displace gut commensal bacteria, and a poorer adaptation to an intracellular lifestyle. In addition, the World Health Organization (WHO) have recognized the significant threat posed by antibiotic-resistant strains of S. sonnei, demanding new approaches. This review gathers knowledge on what is known about S. sonnei within the context of other Shigella spp. and aims to open the door for future research on understanding the increasing spread of this pathogen.
Collapse
Affiliation(s)
- Xosé M. Matanza
- Centre for Bacterial Resistance Biology, Department of Life Sciences, Imperial College London, London, UK
| | - Abigail Clements
- Centre for Bacterial Resistance Biology, Department of Life Sciences, Imperial College London, London, UK
| |
Collapse
|
5
|
Boero E, Vezzani G, Micoli F, Pizza M, Rossi O. Functional assays to evaluate antibody-mediated responses against Shigella: a review. Front Cell Infect Microbiol 2023; 13:1171213. [PMID: 37260708 PMCID: PMC10227456 DOI: 10.3389/fcimb.2023.1171213] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 04/27/2023] [Indexed: 06/02/2023] Open
Abstract
Shigella is a major global pathogen and the etiological agent of shigellosis, a diarrheal disease that primarily affects low- and middle-income countries. Shigellosis is characterized by a complex, multistep pathogenesis during which bacteria use multiple invasion proteins to manipulate and invade the intestinal epithelium. Antibodies, especially against the O-antigen and some invasion proteins, play a protective role as titres against specific antigens inversely correlate with disease severity; however, the context of antibody action during pathogenesis remains to be elucidated, especially with Shigella being mostly an intracellular pathogen. In the absence of a correlate of protection, functional assays rebuilding salient moments of Shigella pathogenesis can improve our understanding of the role of protective antibodies in blocking infection and disease. In vitro assays are important tools to build correlates of protection. Only recently animal models to recapitulate human pathogenesis, often not in full, have been established. This review aims to discuss in vitro assays to evaluate the functionality of anti-Shigella antibodies in polyclonal sera in light of the multistep and multifaced Shigella infection process. Indeed, measurement of antibody level alone may limit the evaluation of full vaccine potential. Serum bactericidal assay (SBA), and other functional assays such as opsonophagocytic killing assays (OPKA), and adhesion/invasion inhibition assays (AIA), are instead physiologically relevant and may provide important information regarding the role played by these effector mechanisms in protective immunity. Ultimately, the review aims at providing scientists in the field with new points of view regarding the significance of functional assays of choice which may be more representative of immune-mediated protection mechanisms.
Collapse
Affiliation(s)
- Elena Boero
- GSK Vaccines Institute for Global Health (GVGH) S.r.l., Siena, Italy
| | - Giacomo Vezzani
- GSK Vaccines Institute for Global Health (GVGH) S.r.l., Siena, Italy
| | - Francesca Micoli
- GSK Vaccines Institute for Global Health (GVGH) S.r.l., Siena, Italy
| | - Mariagrazia Pizza
- GSK Vaccines Institute for Global Health (GVGH) S.r.l., Siena, Italy
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | - Omar Rossi
- GSK Vaccines Institute for Global Health (GVGH) S.r.l., Siena, Italy
| |
Collapse
|
6
|
Friebel J, Schinnerling K, Weigt K, Heldt C, Fromm A, Bojarski C, Siegmund B, Epple HJ, Kikhney J, Moter A, Schneider T, Schulzke JD, Moos V, Schumann M. Uptake of Tropheryma whipplei by Intestinal Epithelia. Int J Mol Sci 2023; 24:ijms24076197. [PMID: 37047170 PMCID: PMC10094206 DOI: 10.3390/ijms24076197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 03/18/2023] [Accepted: 03/21/2023] [Indexed: 03/29/2023] Open
Abstract
Background: Tropheryma whipplei (TW) can cause different pathologies, e.g., Whipple’s disease and transient gastroenteritis. The mechanism by which the bacteria pass the intestinal epithelial barrier, and the mechanism of TW-induced gastroenteritis are currently unknown. Methods: Using ex vivo disease models comprising human duodenal mucosa exposed to TW in Ussing chambers, various intestinal epithelial cell (IEC) cultures exposed to TW and a macrophage/IEC coculture model served to characterize endocytic uptake mechanisms and barrier function. Results: TW exposed ex vivo to human small intestinal mucosae is capable of autonomously entering IECs, thereby invading the mucosa. Using dominant-negative mutants, TW uptake was shown to be dynamin- and caveolin-dependent but independent of clathrin-mediated endocytosis. Complementary inhibitor experiments suggested a role for the activation of the Ras/Rac1 pathway and actin polymerization. TW-invaded IECs underwent apoptosis, thereby causing an epithelial barrier defect, and were subsequently subject to phagocytosis by macrophages. Conclusions: TW enters epithelia via an actin-, dynamin-, caveolin-, and Ras-Rac1-dependent endocytosis mechanism and consecutively causes IEC apoptosis primarily in IECs invaded by multiple TW bacteria. This results in a barrier leak. Moreover, we propose that TW-packed IECs can be subject to phagocytic uptake by macrophages, thereby opening a potential entry point of TW into intestinal macrophages.
Collapse
Affiliation(s)
- Julian Friebel
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité, 12203 Berlin, Germany
- Department of Gastroenterology, Infectiology and Rheumatology, Campus Benjamin Franklin, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 12203 Berlin, Germany
- Berlin Institute of Health at Charité—Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Katina Schinnerling
- Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago 8370146, Chile
| | - Kathleen Weigt
- Department of Gastroenterology, Infectiology and Rheumatology, Campus Benjamin Franklin, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 12203 Berlin, Germany
| | - Claudia Heldt
- Department of Gastroenterology, Infectiology and Rheumatology, Campus Benjamin Franklin, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 12203 Berlin, Germany
| | - Anja Fromm
- Institute of Clinical Physiology, Campus Benjamin Franklin, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 12203 Berlin, Germany
| | - Christian Bojarski
- Department of Gastroenterology, Infectiology and Rheumatology, Campus Benjamin Franklin, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 12203 Berlin, Germany
| | - Britta Siegmund
- Department of Gastroenterology, Infectiology and Rheumatology, Campus Benjamin Franklin, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 12203 Berlin, Germany
| | - Hans-Jörg Epple
- Department of Gastroenterology, Infectiology and Rheumatology, Campus Benjamin Franklin, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 12203 Berlin, Germany
| | - Judith Kikhney
- Institute for Microbiology, Infectious Diseases, and Immunology, Biofilmcenter, Campus Benjamin Franklin, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 12203 Berlin, Germany
- MoKi Analytics GmbH, 12207 Berlin, Germany
| | - Annette Moter
- Institute for Microbiology, Infectious Diseases, and Immunology, Biofilmcenter, Campus Benjamin Franklin, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 12203 Berlin, Germany
- German Konsiliarlabor for Tropheryma whipplei, 10117 Berlin, Germany
- Moter Diagnostics, 12207 Berlin, Germany
| | - Thomas Schneider
- Department of Gastroenterology, Infectiology and Rheumatology, Campus Benjamin Franklin, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 12203 Berlin, Germany
| | - Jörg D. Schulzke
- Department of Gastroenterology, Infectiology and Rheumatology, Campus Benjamin Franklin, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 12203 Berlin, Germany
- Institute of Clinical Physiology, Campus Benjamin Franklin, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 12203 Berlin, Germany
| | - Verena Moos
- Department of Gastroenterology, Infectiology and Rheumatology, Campus Benjamin Franklin, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 12203 Berlin, Germany
| | - Michael Schumann
- Department of Gastroenterology, Infectiology and Rheumatology, Campus Benjamin Franklin, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 12203 Berlin, Germany
- Berlin Institute of Health at Charité—Universitätsmedizin Berlin, 10117 Berlin, Germany
- Correspondence: ; Tel.: +49-30-450-513536
| |
Collapse
|
7
|
Boquet-Pujadas A, Feaugas T, Petracchini A, Grassart A, Mary H, Manich M, Gobaa S, Olivo-Marin JC, Sauvonnet N, Labruyère E. 4D live imaging and computational modeling of a functional gut-on-a-chip evaluate how peristalsis facilitates enteric pathogen invasion. SCIENCE ADVANCES 2022; 8:eabo5767. [PMID: 36269830 PMCID: PMC9586479 DOI: 10.1126/sciadv.abo5767] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 09/02/2022] [Indexed: 05/31/2023]
Abstract
Physical forces are essential to biological function, but their impact at the tissue level is not fully understood. The gut is under continuous mechanical stress because of peristalsis. To assess the influence of mechanical cues on enteropathogen invasion, we combine computational imaging with a mechanically active gut-on-a-chip. After infecting the device with either of two microbes, we image their behavior in real time while mapping the mechanical stress within the tissue. This is achieved by reconstructing three-dimensional videos of the ongoing invasion and leveraging on-manifold inverse problems together with viscoelastic rheology. Our results show that peristalsis accelerates the destruction and invasion of intestinal tissue by Entamoeba histolytica and colonization by Shigella flexneri. Local tension facilitates parasite penetration and activates virulence genes in the bacteria. Overall, our work highlights the fundamental role of physical cues during host-pathogen interactions and introduces a framework that opens the door to study mechanobiology on deformable tissues.
Collapse
Affiliation(s)
- Aleix Boquet-Pujadas
- Bioimage Analysis Unit, Institut Pasteur, Université Paris Cité, Paris, France
- Intracellular Trafficking and Tissue Homeostasis Group, Institut Pasteur, Université Paris Cité, Paris, France
- Biomedical Imaging Group, École polytechnique fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Thomas Feaugas
- Intracellular Trafficking and Tissue Homeostasis Group, Institut Pasteur, Paris, France
| | - Alba Petracchini
- Bioimage Analysis Unit, Institut Pasteur, Université Paris Cité, Paris, France
- Intracellular Trafficking and Tissue Homeostasis Group, Institut Pasteur, Université Paris Cité, Paris, France
| | - Alexandre Grassart
- Intracellular Trafficking and Tissue Homeostasis Group, Institut Pasteur, Paris, France
- Unit of Bioengineering and Microbiology, Center for Microbes, Development and Health (CMDH), Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
| | - Héloïse Mary
- Biomaterials and Microfluidics Core Facility, Institut Pasteur, Université Paris Cité, Paris, France
| | - Maria Manich
- Bioimage Analysis Unit, Institut Pasteur, Université Paris Cité, Paris, France
- Intracellular Trafficking and Tissue Homeostasis Group, Institut Pasteur, Université Paris Cité, Paris, France
| | - Samy Gobaa
- Biomaterials and Microfluidics Core Facility, Institut Pasteur, Université Paris Cité, Paris, France
| | - Jean-Christophe Olivo-Marin
- Bioimage Analysis Unit, Institut Pasteur, Université Paris Cité, Paris, France
- Intracellular Trafficking and Tissue Homeostasis Group, Institut Pasteur, Université Paris Cité, Paris, France
| | - Nathalie Sauvonnet
- Intracellular Trafficking and Tissue Homeostasis Group, Institut Pasteur, Paris, France
| | - Elisabeth Labruyère
- Bioimage Analysis Unit, Institut Pasteur, Université Paris Cité, Paris, France
- Intracellular Trafficking and Tissue Homeostasis Group, Institut Pasteur, Université Paris Cité, Paris, France
| |
Collapse
|
8
|
Delfini M, Stakenborg N, Viola MF, Boeckxstaens G. Macrophages in the gut: Masters in multitasking. Immunity 2022; 55:1530-1548. [PMID: 36103851 DOI: 10.1016/j.immuni.2022.08.005] [Citation(s) in RCA: 71] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 07/17/2022] [Accepted: 08/09/2022] [Indexed: 11/05/2022]
Abstract
The gastrointestinal tract has the important task of absorbing nutrients, a complex process that requires an intact barrier allowing the passage of nutrients but that simultaneously protects the host against invading microorganisms. To maintain and regulate intestinal homeostasis, the gut is equipped with one of the largest populations of macrophages in the body. Here, we will discuss our current understanding of intestinal macrophage heterogeneity and describe their main functions in the different anatomical niches of the gut during steady state. In addition, their role in inflammatory conditions such as infection, inflammatory bowel disease, and postoperative ileus are discussed, highlighting the roles of macrophages in immune defense. To conclude, we describe the interaction between macrophages and the enteric nervous system during development and adulthood and highlight their contribution to neurodegeneration in the context of aging and diabetes.
Collapse
Affiliation(s)
- Marcello Delfini
- Translational Research Center for GI Disorders (TARGID), Department of Chronic Diseases, Metabolism and Ageing, KU Leuven-University of Leuven, Leuven, Belgium
| | - Nathalie Stakenborg
- Translational Research Center for GI Disorders (TARGID), Department of Chronic Diseases, Metabolism and Ageing, KU Leuven-University of Leuven, Leuven, Belgium
| | - Maria Francesca Viola
- Translational Research Center for GI Disorders (TARGID), Department of Chronic Diseases, Metabolism and Ageing, KU Leuven-University of Leuven, Leuven, Belgium
| | - Guy Boeckxstaens
- Translational Research Center for GI Disorders (TARGID), Department of Chronic Diseases, Metabolism and Ageing, KU Leuven-University of Leuven, Leuven, Belgium.
| |
Collapse
|
9
|
Griffiths G, Gruenberg J, Marsh M, Wohlmann J, Jones AT, Parton RG. Nanoparticle entry into cells; the cell biology weak link. Adv Drug Deliv Rev 2022; 188:114403. [PMID: 35777667 DOI: 10.1016/j.addr.2022.114403] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 06/22/2022] [Accepted: 06/23/2022] [Indexed: 12/22/2022]
Abstract
Nanoparticles (NP) are attractive options for the therapeutic delivery of active pharmaceutical drugs, proteins and nucleic acids into cells, tissues and organs. Research into the development and application of NP most often starts with a diverse group of scientists, including chemists, bioengineers and material and pharmaceutical scientists, who design, fabricate and characterize NP in vitro (Stage 1). The next step (Stage 2) generally investigates cell toxicity as well as the processes by which NP bind, are internalized and deliver their cargo to appropriate model tissue culture cells. Subsequently, in Stage 3, selected NP are tested in animal systems, mostly mouse. Whereas the chemistry-based development and analysis in Stage 1 is increasingly sophisticated, the investigations in Stage 2 are not what could be regarded as 'state-of-the-art' for the cell biology field and the quality of research into NP interactions with cells is often sub-standard. In this review we describe our current understanding of the mechanisms by which particles gain entry into mammalian cells via endocytosis. We summarize the most important areas for concern, highlight some of the most common mis-conceptions, and identify areas where NP scientists could engage with trained cell biologists. Our survey of the different mechanisms of uptake into cells makes us suspect that claims for roles for caveolae, as well as macropinocytosis, in NP uptake into cells have been exaggerated, whereas phagocytosis has been under-appreciated.
Collapse
Affiliation(s)
- Gareth Griffiths
- Department Biosciences, University of Oslo, Blindernveien 31, PO Box 1041, 0316 Oslo, Norway.
| | - Jean Gruenberg
- Department of Biochemistry, University of Geneva, 30 quai E. Ansermet, 1211-Geneva-4, Switzerland
| | - Mark Marsh
- Laboratory for Molecular Cell Biology, University College London, Gower Street, London, WC1E 6BT, UK
| | - Jens Wohlmann
- Department Biosciences, University of Oslo, Blindernveien 31, PO Box 1041, 0316 Oslo, Norway
| | - Arwyn T Jones
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Redwood Building, Cardiff, Wales CF103NB, UK
| | - Robert G Parton
- Institute for Molecular Bioscience and Centre for Microscopy and Microanalysis, The University of Queensland, Qld 4072, Australia
| |
Collapse
|
10
|
Abstract
Shigella flexneri is an intracellular human pathogen that invades colonic cells and causes bloody diarrhea. S. flexneri evolved from commensal Escherichia coli, and genome comparisons reveal that S. flexneri has lost approximately 20% of its genes through the process of pathoadaptation, including a disproportionate number of genes associated with the turnover of the nucleotide-based second messenger cyclic di-GMP (c-di-GMP); however, the remaining c-di-GMP turnover enzymes are highly conserved. c-di-GMP regulates many behavioral changes in other bacteria in response to changing environmental conditions, including biofilm formation, but this signaling system has not been examined in S. flexneri. In this study, we expressed VCA0956, a constitutively active c-di-GMP synthesizing diguanylate cyclase (DGC) from Vibrio cholerae, in S. flexneri to determine if virulence phenotypes were regulated by c-di-GMP. We found that expressing VCA0956 in S. flexneri increased c-di-GMP levels, and this corresponds with increased biofilm formation and reduced acid resistance, host cell invasion, and plaque size. We examined the impact of VCA0956 expression on the S. flexneri transcriptome and found that genes related to acid resistance were repressed, and this corresponded with decreased survival to acid shock. We also found that individual S. flexneri DGC mutants exhibit reduced biofilm formation and reduced host cell invasion and plaque size, as well as increased resistance to acid shock. This study highlights the importance of c-di-GMP signaling in regulating S. flexneri virulence phenotypes. IMPORTANCE The intracellular human pathogen Shigella causes dysentery, resulting in as many as one million deaths per year. Currently, there is no approved vaccine for the prevention of shigellosis, and the incidence of antimicrobial resistance among Shigella species is on the rise. Here, we explored how the widely conserved c-di-GMP bacterial signaling system alters Shigella behaviors associated with pathogenesis. We found that expressing or removing enzymes associated with c-di-GMP synthesis results in changes in Shigella's ability to form biofilms, invade host cells, form lesions in host cell monolayers, and resist acid stress.
Collapse
|
11
|
SepA Enhances Shigella Invasion of Epithelial Cells by Degrading Alpha-1 Antitrypsin and Producing a Neutrophil Chemoattractant. mBio 2021; 12:e0283321. [PMID: 34724811 PMCID: PMC8561385 DOI: 10.1128/mbio.02833-21] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Shigella spp. are highly adapted pathogens that cause bacillary dysentery in human and nonhuman primates. An unusual feature of Shigella pathogenesis is that this organism invades the colonic epithelia from the basolateral pole. Therefore, it has evolved the ability to disrupt the intestinal epithelial barrier to reach the basolateral surface. We have shown previously that the secreted serine protease A (SepA), which belongs to the family of serine protease autotransporters of Enterobacteriaceae, is responsible for the initial destabilization of the intestinal epithelial barrier that facilitates Shigella invasion. However, the mechanisms used by SepA to regulate this process remain unknown. To investigate the protein targets cleaved by SepA in the intestinal epithelium, we incubated a sample of homogenized human colon with purified SepA or with a catalytically inactive mutant of this protease. We discovered that SepA targets an array of 18 different proteins, including alpha-1 antitrypsin (AAT), a major circulating serine proteinase inhibitor in humans. In contrast to other serine proteases, SepA cleaved AAT without forming an inhibiting complex, which resulted in the generation of a neutrophil chemoattractant. We demonstrated that the products of the AAT-SepA reaction induce a mild but significant increase in neutrophil transepithelial migration in vitro. Moreover, the presence of AAT during Shigella infection stimulated neutrophil migration and dramatically enhanced the number of bacteria invading the intestinal epithelium in a SepA-dependent manner. We conclude that by cleaving AAT, SepA releases a chemoattractant that promotes neutrophil migration, which in turn disrupts the intestinal epithelial barrier to enable Shigella invasion. IMPORTANCE Shigella is the second leading cause of diarrheal death globally. In this study, we identified the host protein targets of SepA, Shigella's major protein secreted in culture. We demonstrated that by cleaving AAT, a serine protease inhibitor important to protect surrounding tissue at inflammatory sites, SepA releases a neutrophil chemoattractant that enhances Shigella invasion. Moreover, SepA degraded AAT without becoming inhibited by the cleaved product, and SepA catalytic activity was enhanced at higher concentrations of AAT. Activation of SepA by an excess of AAT may be physiologically relevant at the early stages of Shigella infection, when the amount of synthesized SepA is very low compared to the concentration of AAT in the intestinal lumen. This observation may also help to explain the adeptness of Shigella infectivity at low dose, despite the requirement of reaching the basolateral side to invade and colonize the colonic epithelium.
Collapse
|
12
|
De Gaetano GV, Lentini G, Galbo R, Coppolino F, Famà A, Teti G, Beninati C. Invasion and trafficking of hypervirulent group B streptococci in polarized enterocytes. PLoS One 2021; 16:e0253242. [PMID: 34129624 PMCID: PMC8205152 DOI: 10.1371/journal.pone.0253242] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 06/01/2021] [Indexed: 11/29/2022] Open
Abstract
Streptococcus agalactiae (group B streptococcus or GBS) is a commensal bacterium that can frequently behave as a pathogen, particularly in the neonatal period and in the elderly. The gut is a primary site of GBS colonization and a potential port of entry during neonatal infections caused by hypervirulent clonal complex 17 (CC17) strains. Here we studied the interactions between the prototypical CC17 BM110 strain and polarized enterocytes using the Caco-2 cell line. GBS could adhere to and invade these cells through their apical or basolateral surfaces. Basolateral invasion was considerably more efficient than apical invasion and predominated under conditions resulting in weakening of cell-to-cell junctions. Bacterial internalization occurred by a mechanism involving caveolae- and lipid raft-dependent endocytosis and actin re-organization, but not clathrin-dependent endocytosis. In the first steps of Caco-2 invasion, GBS colocalized with the early endocytic marker EEA-1, to later reside in acidic vacuoles. Taken together, these data suggest that CC17 GBS selectively adheres to the lateral surface of enterocytes from which it enters through caveolar lipid rafts using a classical, actin-dependent endocytic pathway. These data may be useful to develop alternative preventive strategies aimed at blocking GBS invasion of the intestinal barrier.
Collapse
Affiliation(s)
| | - Germana Lentini
- Department of Human Pathology, University of Messina, Messina, Italy
| | - Roberta Galbo
- Department of Chemical, Biological and Pharmaceutical Sciences, University of Messina, Messina, Italy
| | | | - Agata Famà
- Department of Human Pathology, University of Messina, Messina, Italy
| | | | - Concetta Beninati
- Department of Human Pathology, University of Messina, Messina, Italy
- Scylla Biotech Srl, Messina, Italy
- * E-mail:
| |
Collapse
|
13
|
Jiang L, Luo Y, Cao X, Liu W, Song G, Zhang Z. LuxS quorum sensing system mediating Lactobacillus plantarum probiotic characteristics. Arch Microbiol 2021; 203:4141-4148. [PMID: 34057545 DOI: 10.1007/s00203-021-02404-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 05/21/2021] [Accepted: 05/22/2021] [Indexed: 11/25/2022]
Abstract
Lactobacillus plantarum is one of common probiotics in fermented foods. Quorum sensing (QS) is a common communication way within bacteria. It is not clear whether the probiotic properties of L. plantarum mediated by QS. Here, Lb. plantarum YM-4-3 was examined for resistance of pH, bile, antimicrobial and luxS gene expression pattern. The study found that: (1) the supernatant of YM-4-3 had bacteriostatic effect to Escherichia coli O157:H7, Listeria monocytogenes and Staphylococcus aureus; (2) Lb. plantarum YM-4-3 shown tolerance property to the strongest acid culture that pH value of 3; (3) the bile tolerance of Lb. plantarum YM-4-3 was significant difference with the growth stage, the early exponential phase of the growth culture can tolerate bile of 0.4% (w/v), while the stationary growth stage can only tolerate bile of 0.2%; (4) Lb. plantarum YM-4-3 luxS gene was contrary expression along with the growth. (5) Compared with the wild-type strain, the adhesion ability of Lb. plantarum YM-4-3 ΔluxS was decreased obviously. These results showed that AI-2 LuxS quorum sensing system mediating Lb. plantarum acid, bile tolerance, antimicrobial and adhesion of probiotics.
Collapse
Affiliation(s)
- Liming Jiang
- Key Laboratory of Applied Marine Biotechnology, Ministry of Education, Ningbo University, Ningbo, 315211, People's Republic of China.,Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, 650500, People's Republic of China
| | - Yiyong Luo
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, 650500, People's Republic of China.
| | - Xuebin Cao
- Key Laboratory of Applied Marine Biotechnology, Ministry of Education, Ningbo University, Ningbo, 315211, People's Republic of China.,Yantai Jinghai Marine Fisheries Co., Ltd, Yantai, People's Republic of China
| | - Wen Liu
- Department of Rheumatology Immunology, The First People's Hospital of Hefei, 230061, Hefei, People's Republic of China
| | - Gang Song
- Longhaixingang Science and Technology Co, Ltd, 121000, Jinzhou, People's Republic of China
| | - Zhizhen Zhang
- Lanzhou Inspection and Quarantine Co, Ltd, 730070, Lanzhou, People's Republic of China
| |
Collapse
|
14
|
Baddal B, Marrazzo P. Refining Host-Pathogen Interactions: Organ-on-Chip Side of the Coin. Pathogens 2021; 10:203. [PMID: 33668558 PMCID: PMC7918822 DOI: 10.3390/pathogens10020203] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 02/09/2021] [Accepted: 02/11/2021] [Indexed: 02/07/2023] Open
Abstract
Bioinspired organ-level in vitro platforms that recapitulate human organ physiology and organ-specific responses have emerged as effective technologies for infectious disease research, drug discovery, and personalized medicine. A major challenge in tissue engineering for infectious diseases has been the reconstruction of the dynamic 3D microenvironment reflecting the architectural and functional complexity of the human body in order to more accurately model the initiation and progression of host-microbe interactions. By bridging the gap between in vitro experimental models and human pathophysiology and providing alternatives for animal models, organ-on-chip microfluidic devices have so far been implemented in multiple research areas, contributing to major advances in the field. Given the emergence of the recent pandemic, plug-and-play organ chips may hold the key for tackling an unmet clinical need in the development of effective therapeutic strategies. In this review, latest studies harnessing organ-on-chip platforms to unravel host-pathogen interactions are presented to highlight the prospects for the microfluidic technology in infectious diseases research.
Collapse
Affiliation(s)
- Buket Baddal
- Department of Medical Microbiology and Clinical Microbiology, Faculty of Medicine, Near East University, Nicosia 99138, Cyprus
| | - Pasquale Marrazzo
- Department of Experimental, Diagnostic and Specialty Medicine, Alma Mater Studiorum University of Bologna, 40126 Bologna, Italy;
| |
Collapse
|
15
|
Ranganathan S, Smith EM, Foulke-Abel JD, Barry EM. Research in a time of enteroids and organoids: how the human gut model has transformed the study of enteric bacterial pathogens. Gut Microbes 2020; 12:1795492. [PMID: 32795243 PMCID: PMC7524385 DOI: 10.1080/19490976.2020.1795389] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 06/29/2020] [Accepted: 07/01/2020] [Indexed: 02/03/2023] Open
Abstract
Enteric bacterial pathogens cause significant morbidity and mortality globally. Studies in tissue culture and animal models shaped our initial understanding of these host-pathogen interactions. However, intrinsic shortcomings in these models limit their application, especially in translational applications like drug screening and vaccine development. Human intestinal enteroid and organoid models overcome some limitations of existing models and advance the study of enteric pathogens. In this review, we detail the use of human enteroids and organoids to investigate the pathogenesis of invasive bacteria Shigella, Listeria, and Salmonella, and noninvasive bacteria pathogenic Escherichia coli, Clostridium difficile, and Vibrio cholerae. We highlight how these studies confirm previously identified mechanisms and, importantly, reveal novel ones. We also discuss the challenges for model advancement, including platform engineering to integrate environmental conditions, innate immune cells and the resident microbiome, and the potential for pre-clinical testing of recently developed antimicrobial drugs and vaccines.
Collapse
Affiliation(s)
- Sridevi Ranganathan
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Emily M. Smith
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Jennifer D. Foulke-Abel
- Department of Medicine, Division of Gastroenterology and Hepatology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Eileen M. Barry
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
16
|
Yang S, Li Y, Wang B, Yang N, Huang X, Chen Q, Geng S, Zhou Y, Shi H, Wang L, Brugman S, Savelkoul H, Liu G. Acute porcine epidemic diarrhea virus infection reshapes the intestinal microbiota. Virology 2020; 548:200-212. [PMID: 32763491 PMCID: PMC7353907 DOI: 10.1016/j.virol.2020.07.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 06/30/2020] [Accepted: 07/01/2020] [Indexed: 02/07/2023]
Abstract
The intestinal microbiota is crucial to intestinal homeostasis. Porcine epidemic diarrhea virus (PEDV) is high pathogenic to intestines, causing diarrhea, even death in piglets. To investigate the detailed relationship between PEDV infection and intestinal microbiota, the composition and distribution of intestinal microbiota from pigs were first analyzed using 16S rRNA sequencing technology. The results demonstrated that the composition and distribution of microbes in different intestinal segments were quite similar between 1-week-old and 2-week-old piglets but different from 4-week-old (weaned) piglets. Then piglets at different ages were inoculated with PEDV. The results showed that the 1-week-old piglets exhibited the most severe pathogenicity comparing to the other age groups. Further investigations indicated that Lactobacillus, Escherichia coli, and Lactococcus in the intestinal microbiota of piglets were significantly changed by PEDV infection. These results strengthen our understanding of viruses influencing intestinal microbes and remind us of the potential association between PEDV and intestinal microbes.
Collapse
Affiliation(s)
- Shanshan Yang
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, 730046, China; Cell Biology and Immunology Group, Wageningen University and Research, Wageningen, the Netherlands
| | - Yang Li
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, 730046, China
| | - Bin Wang
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, 730046, China
| | - Ning Yang
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, 730046, China
| | - Xin Huang
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, 730046, China
| | - Qingbo Chen
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, 730046, China
| | - Shuxian Geng
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, 730046, China
| | - Yawei Zhou
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, 730046, China
| | - Han Shi
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, 730046, China
| | - Leyi Wang
- Department of Veterinary Clinical Medicine and the Veterinary Diagnostic Laboratory, College of Veterinary Medicine, University of Illinois, Urbana, IL, 61802, USA
| | - Sylvia Brugman
- Cell Biology and Immunology Group, Wageningen University and Research, Wageningen, the Netherlands
| | - Huub Savelkoul
- Cell Biology and Immunology Group, Wageningen University and Research, Wageningen, the Netherlands
| | - Guangliang Liu
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, 730046, China.
| |
Collapse
|
17
|
Nigro G, Arena ET, Sachse M, Moya-Nilges M, Marteyn BS, Sansonetti PJ, Campbell-Valois FX. Mapping of Shigella flexneri's tissue distribution and type III secretion apparatus activity during infection of the large intestine of guinea pigs. Pathog Dis 2020; 77:5580288. [PMID: 31578543 PMCID: PMC6920510 DOI: 10.1093/femspd/ftz054] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 09/30/2019] [Indexed: 12/12/2022] Open
Abstract
Shigella spp. are bacterial pathogens that invade the human colonic mucosa using a type III secretion apparatus (T3SA), a proteinaceous device activated upon contact with host cells. Active T3SAs translocate proteins that carve the intracellular niche of Shigella spp. Nevertheless, the activation state of the T3SA has not been addressed in vivo. Here, we used a green fluorescent protein transcription-based secretion activity reporter (TSAR) to provide a spatio-temporal description of S. flexneri T3SAs activity in the colon of Guinea pigs. First, we observed that early mucus release is triggered in the vicinity of luminal bacteria with inactive T3SA. Subsequent mucosal invasion showed bacteria with active T3SA associated with the brush border, eventually penetrating into epithelial cells. From 2 to 8 h post-challenge, the infection foci expanded, and these intracellular bacteria displayed homogeneously high-secreting activity, while extracellular foci within the lamina propria featured bacteria with low secretion activity. We also found evidence that within lamina propria macrophages, bacteria reside in vacuoles instead of accessing the cytosol. Finally, bacteria were cleared from tissues between 8 and 24 h post-challenge, highlighting the hit-and-run colonization strategy of Shigella. This study demonstrates how genetically encoded reporters can contribute to deciphering pathogenesis in vivo.
Collapse
Affiliation(s)
- Giulia Nigro
- Unité de Pathogénie Microbienne Moléculaire, Institut Pasteur, INSERM U1202, 24-28 rue du Docteur-Roux, 75015 Paris, France
| | - Ellen T Arena
- Unité de Pathogénie Microbienne Moléculaire, Institut Pasteur, INSERM U1202, 24-28 rue du Docteur-Roux, 75015 Paris, France.,Laboratory for Optical and Computational Instrumentation, University of Wisconsin-Madison, Laboratory for Optical and Computational Instrumentation, 271 Animal Sciences, 1675 Observatory Drive, Madison, WI 53706, USA
| | - Martin Sachse
- Ultrastructural Bioimaging unit, Institut Pasteur, 24-28 rue du Docteur-Roux, 75015 Paris, France
| | - Maryse Moya-Nilges
- Ultrastructural Bioimaging unit, Institut Pasteur, 24-28 rue du Docteur-Roux, 75015 Paris, France
| | - Benoit S Marteyn
- Unité de Pathogénie Microbienne Moléculaire, Institut Pasteur, INSERM U1202, 24-28 rue du Docteur-Roux, 75015 Paris, France.,Architecture et Réactivité de l'ARN, Université de Strasbourg, CNRS UPR9002, 2 Allée Konrad Roentgen, 67084 Strasbourg, France.,Unité Pathogenèse des Infections Vasculaires, Institut Pasteur, 24-28 rue du Docteur-Roux, 75015 Paris, France
| | - Philippe J Sansonetti
- Unité de Pathogénie Microbienne Moléculaire, Institut Pasteur, INSERM U1202, 24-28 rue du Docteur-Roux, 75015 Paris, France.,Chaire de Microbiologie et Maladies Infectieuses, Collège de France, 11 Place Marcelin Berthelot, 75231 Paris, France
| | - F-X Campbell-Valois
- Unité de Pathogénie Microbienne Moléculaire, Institut Pasteur, INSERM U1202, 24-28 rue du Docteur-Roux, 75015 Paris, France.,The Host-Microbe Interactions Laboratory, Department of Chemistry and Biomolecular Sciences, University of Ottawa, 150 Louis-Pasteur private, Ottawa, ON, K1N 6N5, Canada.,Department of Biochemistry, Microbiology and Immunology, University of Ottawa, 451 Smyth Rd, Ottawa, ON, K1N 6N5, Canada
| |
Collapse
|
18
|
Rey C, Chang YY, Latour-Lambert P, Varet H, Proux C, Legendre R, Coppée JY, Enninga J. Transcytosis subversion by M cell-to-enterocyte spread promotes Shigella flexneri and Listeria monocytogenes intracellular bacterial dissemination. PLoS Pathog 2020; 16:e1008446. [PMID: 32282860 PMCID: PMC7179946 DOI: 10.1371/journal.ppat.1008446] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 04/23/2020] [Accepted: 02/29/2020] [Indexed: 11/19/2022] Open
Abstract
Microfold (M) cell host-pathogen interaction studies would benefit from the visual analysis of dynamic cellular and microbial interplays. We adapted a human in vitro M cell model to physiological bacterial infections, expression of fluorescent localization reporters and long-term three-dimensional time-lapse microscopy. This approach allows following key steps of M cell infection dynamics at subcellular resolution, from the apical onset to basolateral epithelial dissemination. We focused on the intracellular pathogen Shigella flexneri, classically reported to transcytose through M cells to initiate bacillary dysentery in humans, while eliciting poorly protective immune responses. Our workflow was critical to reveal that S. flexneri develops a bimodal lifestyle within M cells leading to rapid transcytosis or delayed vacuolar rupture, followed by direct actin motility-based propagation to neighboring enterocytes. Moreover, we show that Listeria monocytogenes, another intracellular pathogen sharing a tropism for M cells, disseminates in a similar manner and evades M cell transcytosis completely. We established that actin-based M cell-to-enterocyte spread is the major dissemination pathway for both pathogens and avoids their exposure to basolateral compartments in our system. Our results challenge the notion that intracellular pathogens are readily transcytosed by M cells to inductive immune compartments in vivo, providing a potential mechanism for their ability to evade adaptive immunity. Microfold (M) epithelial cells are important for the onset of infections and induction of immune responses in many mucosal diseases. We extended a human in vitro M cell model to apical infections, expression of fluorescent host and microbial reporters and real-time fluorescence microscopy. Focusing on the human intracellular pathogen S. flexneri, responsible for bacillary dysentery, this workflow allowed us to uncover that the bacterium can subvert the immunological sampling function of M cells by promoting a cytosolic lifestyle and spreading directly to neighboring enterocytes. This mechanism was shared with the etiologic agent of listeriosis, the intracellular pathogen L. monocytogenes and allowed both pathogens to avoid exposure to underlying immune compartments. These results may provide a mechanism for the ability of intracellular pathogens to evade adaptive immunity in vivo, emphasizing the importance of advanced studies of M cell host-pathogen interactions to understand early steps of mucosal invasion and their consequences on immunity.
Collapse
Affiliation(s)
- Camille Rey
- Institut Pasteur, Dynamics of Host-Pathogen Interactions Unit, Paris, France, and Centre National de le la Recherche Scientifique (CNRS) UMR3691, Paris, France
| | - Yuen-Yan Chang
- Institut Pasteur, Dynamics of Host-Pathogen Interactions Unit, Paris, France, and Centre National de le la Recherche Scientifique (CNRS) UMR3691, Paris, France
| | - Patricia Latour-Lambert
- Institut Pasteur, Dynamics of Host-Pathogen Interactions Unit, Paris, France, and Centre National de le la Recherche Scientifique (CNRS) UMR3691, Paris, France
| | - Hugo Varet
- Institut Pasteur, Transcriptome and Epigenome Platform, Paris, France
- Institut Pasteur, Hub Bioinformatique et Biostatistique, Département de Biologie Computationnelle (USR 3756 IP CNRS), Paris, France
| | - Caroline Proux
- Institut Pasteur, Transcriptome and Epigenome Platform, Paris, France
| | - Rachel Legendre
- Institut Pasteur, Transcriptome and Epigenome Platform, Paris, France
- Institut Pasteur, Hub Bioinformatique et Biostatistique, Département de Biologie Computationnelle (USR 3756 IP CNRS), Paris, France
| | - Jean-Yves Coppée
- Institut Pasteur, Transcriptome and Epigenome Platform, Paris, France
| | - Jost Enninga
- Institut Pasteur, Dynamics of Host-Pathogen Interactions Unit, Paris, France, and Centre National de le la Recherche Scientifique (CNRS) UMR3691, Paris, France
- * E-mail:
| |
Collapse
|
19
|
Watson JL, Sanchez-Garrido J, Goddard PJ, Torraca V, Mostowy S, Shenoy AR, Clements A. Shigella sonnei O-Antigen Inhibits Internalization, Vacuole Escape, and Inflammasome Activation. mBio 2019; 10:e02654-19. [PMID: 31848280 PMCID: PMC6918081 DOI: 10.1128/mbio.02654-19] [Citation(s) in RCA: 14] [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: 10/15/2019] [Accepted: 11/11/2019] [Indexed: 12/04/2022] Open
Abstract
Two Shigella species, Shigella flexneri and Shigella sonnei, cause approximately 90% of bacterial dysentery worldwide. While S. flexneri is the dominant species in low-income countries, S. sonnei causes the majority of infections in middle- and high-income countries. S. flexneri is a prototypic cytosolic bacterium; once intracellular, it rapidly escapes the phagocytic vacuole and causes pyroptosis of macrophages, which is important for pathogenesis and bacterial spread. In contrast, little is known about the invasion, vacuole escape, and induction of pyroptosis during S. sonnei infection of macrophages. We demonstrate here that S. sonnei causes substantially less pyroptosis in human primary monocyte-derived macrophages and THP1 cells. This is due to reduced bacterial uptake and lower relative vacuole escape, which results in fewer cytosolic S. sonnei and hence reduced activation of caspase-1 inflammasomes. Mechanistically, the O-antigen (O-Ag), which in S. sonnei is contained in both the lipopolysaccharide and the capsule, was responsible for reduced uptake and the type 3 secretion system (T3SS) was required for vacuole escape. Our findings suggest that S. sonnei has adapted to an extracellular lifestyle by incorporating multiple layers of O-Ag onto its surface compared to other Shigella species.IMPORTANCE Diarrheal disease remains the second leading cause of death in children under five. Shigella remains a significant cause of diarrheal disease with two species, S. flexneri and S. sonnei, causing the majority of infections. S. flexneri are well known to cause cell death in macrophages, which contributes to the inflammatory nature of Shigella diarrhea. Here, we demonstrate that S. sonnei causes less cell death than S. flexneri due to a reduced number of bacteria present in the cell cytosol. We identify the O-Ag polysaccharide which, uniquely among Shigella spp., is present in two forms on the bacterial cell surface as the bacterial factor responsible. Our data indicate that S. sonnei differs from S. flexneri in key aspects of infection and that more attention should be given to characterization of S. sonnei infection.
Collapse
Affiliation(s)
- Jayne L Watson
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | - Julia Sanchez-Garrido
- Department of Infectious Disease, MRC Centre for Molecular Bacteriology and Infection, Imperial College London, London, United Kingdom
| | - Philippa J Goddard
- Department of Life Sciences, Imperial College London, London, United Kingdom
- Department of Infectious Disease, MRC Centre for Molecular Bacteriology and Infection, Imperial College London, London, United Kingdom
| | - Vincenzo Torraca
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Serge Mostowy
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Avinash R Shenoy
- Department of Infectious Disease, MRC Centre for Molecular Bacteriology and Infection, Imperial College London, London, United Kingdom
| | - Abigail Clements
- Department of Life Sciences, Imperial College London, London, United Kingdom
| |
Collapse
|
20
|
Tran ENH, Day CJ, Poole J, Jennings MP, Morona R. Specific blood group antibodies inhibit Shigella flexneri interaction with human cells in the absence of spinoculation. Biochem Biophys Res Commun 2019; 521:131-136. [PMID: 31630794 DOI: 10.1016/j.bbrc.2019.10.091] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 10/10/2019] [Indexed: 10/25/2022]
Abstract
The classical models of investigating Shigella flexneri adherence and invasion of tissue culture cells involve either bacterial centrifugation (spinoculation) or the use of AfaE adhesin to overcome the low infection rate observed in vitro. However clinically, S. flexneri clearly adheres and invades the human colon in the absence of 'spinoculation'. Additionally, certain S. flexneri tissue cell based assays (e.g. plaque assays and infection of T84 epithelial cells on Transwells®), do not require spinoculation. In the absence of spinoculation, we recently showed that glycan-glycan interactions play an important role in S. flexneri interaction with host cells, and that in particular the S. flexneri 2a lipopolysaccharide O antigen glycan has a high affinity for the blood group A glycan. During the investigation of the effect of blood group A antibodies on S. flexneri interaction with cells, we discovered that Panc-1 cells exhibited a high rate of infection in the absence of spinoculation. Select blood group A antibodies inhibited invasion of Panc-1 cells, and adherence to T84 cells. The use of Panc-1 cells represents a simplified model to study S. flexneri pathogenesis and does not require either spinoculation or exogenous adhesins.
Collapse
Affiliation(s)
- Elizabeth Ngoc Hoa Tran
- School of Biological Sciences, Department of Molecular & Biomedical Science, Research Centre for Infectious Diseases, University of Adelaide, Adelaide, 5005, Australia
| | - Christopher J Day
- Institute for Glycomics, Griffith University Gold Coast Campus, Queensland, 4222, Australia
| | - Jessica Poole
- Institute for Glycomics, Griffith University Gold Coast Campus, Queensland, 4222, Australia
| | - Michael P Jennings
- Institute for Glycomics, Griffith University Gold Coast Campus, Queensland, 4222, Australia
| | - Renato Morona
- School of Biological Sciences, Department of Molecular & Biomedical Science, Research Centre for Infectious Diseases, University of Adelaide, Adelaide, 5005, Australia.
| |
Collapse
|
21
|
Wang Y, Li L, Xue S, Han Y, Yang K. Characteristics and formation mechanism of intestinal bacteria particles emitted from aerated wastewater treatment tanks. WATER RESEARCH 2019; 163:114862. [PMID: 31336209 DOI: 10.1016/j.watres.2019.114862] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 07/01/2019] [Accepted: 07/13/2019] [Indexed: 06/10/2023]
Abstract
Aeration tanks in municipal wastewater treatment plants (WWTPs) are regarded as sources of bioaerosols, often containing particles and microbes. In this study, intestinal bacteria were investigated from biochemical reaction tanks (BRTs) of six municipal WWTPs. It was observed that 86 CFU/m3 of intestinal bacteria (in average) occurred in the BRTs installed surface aerator, which was higher than those adopted submerged aeration (67 CFU/m3 in average). 62.72% of fine particles were observed in the BRTs supplied oxygen by submerged aerator, while 75.73% of coarse particles emitted during surface aeration. Pseudomonas sp., Serratia sp. and Acinetobacter sp. were identified as pathogenic bacteria presented in the intestinal bacteria population and most of them existed initially in water or sludge, particularly in water surface. The emission level and particle size distribution were significantly correlated with aeration mode adopted by the WWTPs. The bioaerosols particles emitted from surface aeration process was higher than that from submerged aeration process. Meanwhile, the BRTs with submerged aerators released more fine particles, which can get into the alveoli and represented the potential challenge to human health. Canonical correspondence analysis results exhibited that population of intestinal bacteria had a positive correlation with aeration rate and water quality. As the intestinal bacteria in the bioaerosols emitted from the WWTPs may pose a potential risk to onsite operators, aeration tanks in WWTPs should be paid more attention as a source of intestinal bacterial emissions.
Collapse
Affiliation(s)
- Yanjie Wang
- State Key Laboratory of Environmental Aquatic Chemistry, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, PR China; School of Public Health, Zhengzhou University, Zhengzhou, 450001, PR China; Lancaster Environment Centre, Lancaster University, Lancaster, LA1 4YQ, UK.
| | - Lin Li
- State Key Laboratory of Environmental Aquatic Chemistry, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, PR China; National Engineering Laboratory for VOCs Pollution Control Material & Technology, University of Chinese Academy of Sciences, Beijing, 101408, PR China.
| | - Song Xue
- State Key Laboratory of Environmental Aquatic Chemistry, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, PR China; Fujian Provincial Colleges and University Engineering Research Center of Solid Waste Resource Utilization, Longyan University, Longyan, PR China.
| | - Yunping Han
- State Key Laboratory of Environmental Aquatic Chemistry, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, PR China; National Engineering Laboratory for VOCs Pollution Control Material & Technology, University of Chinese Academy of Sciences, Beijing, 101408, PR China.
| | - Kaixiong Yang
- State Key Laboratory of Environmental Aquatic Chemistry, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, PR China; National Engineering Laboratory for VOCs Pollution Control Material & Technology, University of Chinese Academy of Sciences, Beijing, 101408, PR China.
| |
Collapse
|
22
|
Bioengineered Human Organ-on-Chip Reveals Intestinal Microenvironment and Mechanical Forces Impacting Shigella Infection. Cell Host Microbe 2019; 26:435-444.e4. [PMID: 31492657 DOI: 10.1016/j.chom.2019.08.007] [Citation(s) in RCA: 115] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 07/11/2019] [Accepted: 08/09/2019] [Indexed: 12/20/2022]
Abstract
Intestinal epithelial cells are constantly exposed to pathogens and mechanical forces. However, the impact of mechanical forces on infections leading to diarrheal diseases remains largely unknown. Here, we addressed whether flow and peristalsis impact the infectivity of the human pathogen Shigella within a 3D colonic epithelium using Intestine-Chip technology. Strikingly, infection is significantly increased and minimal bacterial loads are sufficient to invade enterocytes from the apical side and trigger loss of barrier integrity, thereby shifting the paradigm about early stage Shigella invasion. Shigella quickly colonizes epithelial crypt-like invaginations and demonstrates the essential role of the microenvironment. Furthermore, by modulating the mechanical forces of the microenvironment, we find that peristalsis impacts Shigella invasion. Collectively, our results reveal that Shigella leverages the intestinal microenvironment by taking advantage of the microarchitecture and mechanical forces to efficiently invade the intestine. This approach will enable molecular and mechanistic interrogation of human-restricted enteric pathogens.
Collapse
|
23
|
Ndungo E, Pasetti MF. Functional antibodies as immunological endpoints to evaluate protective immunity against Shigella. Hum Vaccin Immunother 2019; 16:197-205. [PMID: 31287754 PMCID: PMC7670857 DOI: 10.1080/21645515.2019.1640427] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
The development, clinical advancement and licensure of vaccines, and monitoring of vaccine effectiveness could be expedited and simplified by the ability to measure immunological endpoints that can predict a favorable clinical outcome. Antigen-specific and functional antibodies have been described in the context of naturally acquired immunity and vaccination against Shigella, and their presence in serum has been associated with reduced risk of disease in human subjects. The relevance of these antibodies as correlates of protective immunity, their mechanistic contribution to protection (e.g. target antigens, interference with pathogenesis, and participation in microbial clearance), and factors that influence their magnitude and makeup (e.g. host age, health condition, and environment) are important considerations that need to be explored. In addition to facilitating vaccine evaluation, immunological correlates of protection could be useful for identifying groups at risk and advancing immune therapies. Herein we discuss the precedent and value of functional antibodies as immunological endpoints to predict vaccine efficacy and the relevance of functional antibody activity to evaluate protective immunity against shigellosis.
Collapse
Affiliation(s)
- Esther Ndungo
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Marcela F Pasetti
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
24
|
Liao C, Fang K, Xiao J, Zhang W, Zhang B, Yuan W, Lu W, Xu D. Critical determinants of human neutrophil peptide 1 for enhancing host epithelial adhesion of Shigella flexneri. Cell Microbiol 2019; 21:e13069. [PMID: 31218775 DOI: 10.1111/cmi.13069] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 05/17/2019] [Accepted: 06/04/2019] [Indexed: 11/27/2022]
Abstract
Human neutrophil peptides (HNPs), also known as human myeloid α-defensins degranulated by infiltrating neutrophils at bacterial infection loci, exhibit broad antomicrobial activities against bacteria, fungi, and viruses. We have made a surprising recent finding that Shigella, a highly contagious, yet poorly adhesive enteric pathogen, exploits human α-defensins including HNP1 to enhance its adhesion to and invasion of host epithelial cells. However, the critical molecular determinants responsible for HNP1-enhanced Shigella adhesion and invasion have yet to be investigated. Using cultured epithelial cells and polarised Caco2 cells as an in vitro infection model, we demonstrated that HNP1 promoted Shigella infection in a structure- and sequence-dependent manner, with two bulky hydrophobic residues, Trp26 and Phe28 important for HNP1 self-assembly, being most critical. The functional importance of hydrophobicity for HNP1-enhanced Shigella infection was further verified by substitutions for Trp26 of a series of unnatural amino acids with straight aliphatic side chains of different lengths. Dissection of the Shigella infection process revealed that bacteria-rather than host cells-bound HNP1 contributed most to the enhancement. Further, mutagenesis analysis of bacterial surface components, while precluding the involvement of lipopolysaccharides (LPS) in the interaction with HNP1, identified outer membrane proteins and the Type 3 secretion apparatus as putative binding targets of HNP1 involved in enhanced Shigella adhesion and invasion. Our findings provide molecular and mechanistic insights into the mode of action of HNP1 in promoting Shigella infection, thus showcasing another example of how innate immune factors may serve as a double-edged sword in health and disease.
Collapse
Affiliation(s)
- Chongbing Liao
- Center for Translational Medicine Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, China.,Key Laboratory of Biomedical Information Engineering of the Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China.,Institute of Human Virology and Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Kun Fang
- Department of Internal Medicine, Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jiu Xiao
- Key Laboratory of Biomedical Information Engineering of the Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Wei Zhang
- Center for Translational Medicine Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Bing Zhang
- Center for Translational Medicine Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Weirong Yuan
- Institute of Human Virology and Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Wuyuan Lu
- Institute of Human Virology and Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Dan Xu
- Center for Translational Medicine Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, China.,Key Laboratory of Biomedical Information Engineering of the Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China.,Institute of Human Virology and Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
25
|
Liu G, Pilla G, Tang CM. Shigella host: Pathogen interactions: Keeping bacteria in the loop. Cell Microbiol 2019; 21:e13062. [PMID: 31134722 DOI: 10.1111/cmi.13062] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 05/05/2019] [Accepted: 05/20/2019] [Indexed: 12/15/2022]
Abstract
Shigella spp. are Gram-negative enteric pathogens and the leading cause of bacterial dysentery worldwide. Since the discovery more than three decades ago that the large virulence plasmid of Shigella is essential for pathogenesis, our understanding of how the bacterium orchestrates inflammation and tissue destruction at the mucosal surface has been informed by studies employing the rabbit ileal loop model. Here, we outline how Phillippe Sansonetti, together with his co-workers and collaborators, exploited this model to provide a holistic view of how Shigella survives in the intestinal tract, traverses the intestinal epithelial barrier, and manipulates the host immune system to cause disease.
Collapse
Affiliation(s)
- Guangyu Liu
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Giulia Pilla
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Christoph M Tang
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| |
Collapse
|
26
|
Brunner K, Samassa F, Sansonetti PJ, Phalipon A. Shigella-mediated immunosuppression in the human gut: subversion extends from innate to adaptive immune responses. Hum Vaccin Immunother 2019; 15:1317-1325. [PMID: 30964713 PMCID: PMC6663138 DOI: 10.1080/21645515.2019.1594132] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 02/13/2019] [Accepted: 02/27/2019] [Indexed: 02/07/2023] Open
Abstract
The enteropathogen, Shigella, is highly virulent and remarkably adjusted to the intestinal environment of its almost exclusive human host. Key for Shigella pathogenicity is the injection of virulence effectors into the host cell via its type three secretion system (T3SS), initiating disease onset and progression by the vast diversity of the secreted T3SS effectors and their respective cellular targets. The multifaceted modulation of host signaling pathways exerted by Shigella T3SS effectors, which include the subversion of host innate immune defenses and the promotion of intracellular bacterial survival and dissemination, have been extensively reviewed in the recent past. This review focuses on the human species specificity of Shigella by discussing some possible evasion mechanisms towards the human, but not non-human or rodent gut innate defense barrier, leading to the lack of a relevant animal infection model. In addition, subversion mechanisms of the adaptive immune response are highlighted summarizing research advances of the recent years. In particular, the new paradigm of Shigella pathogenicity constituted of invasion-independent T3SS effector-mediated targeting of activated, human lymphocytes is discussed. Along with consequences on vaccine development, these findings offer new directions for future research endeavors towards a better understanding of immunity to Shigella infection.
Collapse
Affiliation(s)
- Katja Brunner
- Molecular Microbial Pathogenesis Unit, Department of Cellular Biology of Infection, Institut Pasteur, Paris, France
- INSERM U1202, Paris, France
| | - Fatoumata Samassa
- Molecular Microbial Pathogenesis Unit, Department of Cellular Biology of Infection, Institut Pasteur, Paris, France
- INSERM U1202, Paris, France
| | - Philippe J. Sansonetti
- Molecular Microbial Pathogenesis Unit, Department of Cellular Biology of Infection, Institut Pasteur, Paris, France
- INSERM U1202, Paris, France
- Chaire de Microbiologie et Maladies Infectieuses, Collège de France, Paris, France
| | - Armelle Phalipon
- Molecular Microbial Pathogenesis Unit, Department of Cellular Biology of Infection, Institut Pasteur, Paris, France
- INSERM U1202, Paris, France
| |
Collapse
|
27
|
Koestler BJ, Ward CM, Fisher CR, Rajan A, Maresso AW, Payne SM. Human Intestinal Enteroids as a Model System of Shigella Pathogenesis. Infect Immun 2019; 87:e00733-18. [PMID: 30642906 PMCID: PMC6434139 DOI: 10.1128/iai.00733-18] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Accepted: 01/06/2019] [Indexed: 01/01/2023] Open
Abstract
The enteric bacterium and intracellular human pathogen Shigella causes hundreds of millions of cases of the diarrheal disease shigellosis per year worldwide. Shigella is acquired by ingestion of contaminated food or water; upon reaching the colon, the bacteria invade colonic epithelial cells, replicate intracellularly, spread to adjacent cells, and provoke an intense inflammatory response. There is no animal model that faithfully recapitulates human disease; thus, cultured cells have been used to model Shigella pathogenesis. However, the use of transformed cells in culture does not provide the same environment to the bacteria as the normal human intestinal epithelium. Recent advances in tissue culture now enable the cultivation of human intestinal enteroids (HIEs), which are derived from human intestinal stem cells, grown ex vivo, and then differentiated into "mini-intestines." Here, we demonstrate that HIEs can be used to model Shigella pathogenesis. We show that Shigella flexneri invades polarized HIE monolayers preferentially via the basolateral surface. After S. flexneri invades HIE monolayers, S. flexneri replicates within HIE cells and forms actin tails. S. flexneri also increases the expression of HIE proinflammatory signals and the amino acid transporter SLC7A5. Finally, we demonstrate that disruption of HIE tight junctions enables S. flexneri invasion via the apical surface.
Collapse
Affiliation(s)
- Benjamin J Koestler
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, Texas, USA
- Institute for Cellular and Molecular Biology, The University of Texas at Austin, Austin, Texas, USA
| | - Cara M Ward
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, Texas, USA
- Institute for Cellular and Molecular Biology, The University of Texas at Austin, Austin, Texas, USA
| | - C R Fisher
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, Texas, USA
- Institute for Cellular and Molecular Biology, The University of Texas at Austin, Austin, Texas, USA
| | - Anubama Rajan
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | - Anthony W Maresso
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | - Shelley M Payne
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, Texas, USA
- Institute for Cellular and Molecular Biology, The University of Texas at Austin, Austin, Texas, USA
| |
Collapse
|
28
|
Evaluating Shigella flexneri Pathogenesis in the Human Enteroid Model. Infect Immun 2019; 87:IAI.00740-18. [PMID: 30642900 PMCID: PMC6434113 DOI: 10.1128/iai.00740-18] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Accepted: 01/06/2019] [Indexed: 01/02/2023] Open
Abstract
The enteric pathogen Shigella is one of the leading causes of moderate-to-severe diarrhea and death in young children in developing countries. Transformed cell lines and animal models have been widely used to study Shigella pathogenesis. The enteric pathogen Shigella is one of the leading causes of moderate-to-severe diarrhea and death in young children in developing countries. Transformed cell lines and animal models have been widely used to study Shigella pathogenesis. In addition to altered physiology, transformed cell lines are composed of a single cell type that does not sufficiently represent the complex multicellular environment of the human colon. Most available animal models do not accurately mimic human disease. The human intestinal enteroid model, derived from LGR5+ stem cell-containing intestinal crypts from healthy subjects, represents a technological leap in human gastrointestinal system modeling and provides a more physiologically relevant system that includes multiple cell types and features of the human intestine. We established the utility of this model for studying basic aspects of Shigella pathogenesis and host responses. In this study, we show that Shigellaflexneri is capable of infecting and replicating intracellularly in human enteroids derived from different segments of the intestine. Apical invasion by S. flexneri is very limited but increases ∼10-fold when enteroids are differentiated to include M cells. Invasion via the basolateral surface was at least 2-log10 units more efficient than apical infection. Increased secretion of interleukin-8 and higher expression levels of the mucin glycoprotein Muc2 were observed in the enteroids following S. flexneri infection. The human enteroid model promises to bridge some of the gaps between traditional cell culture, animal models, and human infection.
Collapse
|
29
|
Tawk C, Nigro G, Rodrigues Lopes I, Aguilar C, Lisowski C, Mano M, Sansonetti P, Vogel J, Eulalio A. Stress-induced host membrane remodeling protects from infection by non-motile bacterial pathogens. EMBO J 2018; 37:embj.201798529. [PMID: 30389666 PMCID: PMC6276891 DOI: 10.15252/embj.201798529] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 10/05/2018] [Accepted: 10/10/2018] [Indexed: 01/01/2023] Open
Abstract
While mucosal inflammation is a major source of stress during enteropathogen infection, it remains to be fully elucidated how the host benefits from this environment to clear the pathogen. Here, we show that host stress induced by different stimuli mimicking inflammatory conditions strongly reduces the binding of Shigella flexneri to epithelial cells. Mechanistically, stress activates acid sphingomyelinase leading to host membrane remodeling. Consequently, knockdown or pharmacological inhibition of the acid sphingomyelinase blunts the stress-dependent inhibition of Shigella binding to host cells. Interestingly, stress caused by intracellular Shigella replication also results in remodeling of the host cell membrane, in vitro and in vivo, which precludes re-infection by this and other non-motile pathogens. In contrast, Salmonella Typhimurium overcomes the shortage of permissive entry sites by gathering effectively at the remaining platforms through its flagellar motility. Overall, our findings reveal host membrane remodeling as a novel stress-responsive cell-autonomous defense mechanism that protects epithelial cells from infection by non-motile bacterial pathogens.
Collapse
Affiliation(s)
- Caroline Tawk
- Host RNA Metabolism Group, Institute for Molecular Infection Biology (IMIB), University of Würzburg, Würzburg, Germany.,RNA Biology Group, Institute for Molecular Infection Biology (IMIB), University of Würzburg, Würzburg, Germany
| | - Giulia Nigro
- Molecular Microbial Pathogenesis Laboratory, Institut Pasteur, Paris, France
| | - Ines Rodrigues Lopes
- Functional Genomics and RNA-based Therapeutics, UC-BIOTECH, Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal.,RNA & Infection Group, UC-BIOTECH, Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal
| | - Carmen Aguilar
- Host RNA Metabolism Group, Institute for Molecular Infection Biology (IMIB), University of Würzburg, Würzburg, Germany
| | - Clivia Lisowski
- Host RNA Metabolism Group, Institute for Molecular Infection Biology (IMIB), University of Würzburg, Würzburg, Germany
| | - Miguel Mano
- Functional Genomics and RNA-based Therapeutics, UC-BIOTECH, Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal
| | - Philippe Sansonetti
- Molecular Microbial Pathogenesis Laboratory, Institut Pasteur, Paris, France
| | - Jörg Vogel
- RNA Biology Group, Institute for Molecular Infection Biology (IMIB), University of Würzburg, Würzburg, Germany.,Helmholtz Institute for RNA-Based Infection Research (HIRI), Würzburg, Germany
| | - Ana Eulalio
- Host RNA Metabolism Group, Institute for Molecular Infection Biology (IMIB), University of Würzburg, Würzburg, Germany .,RNA & Infection Group, UC-BIOTECH, Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal
| |
Collapse
|
30
|
Xu D, Liao C, Zhang B, Tolbert WD, He W, Dai Z, Zhang W, Yuan W, Pazgier M, Liu J, Yu J, Sansonetti PJ, Bevins CL, Shao Y, Lu W. Human Enteric α-Defensin 5 Promotes Shigella Infection by Enhancing Bacterial Adhesion and Invasion. Immunity 2018; 48:1233-1244.e6. [PMID: 29858013 PMCID: PMC6051418 DOI: 10.1016/j.immuni.2018.04.014] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Revised: 02/18/2018] [Accepted: 04/13/2018] [Indexed: 01/07/2023]
Abstract
Shigella is a Gram-negative bacterium that causes bacillary dysentery worldwide. It invades the intestinal epithelium to elicit intense inflammation and tissue damage, yet the underlying mechanisms of its host selectivity and low infectious inoculum remain perplexing. Here, we report that Shigella co-opts human α-defensin 5 (HD5), a host defense peptide important for intestinal homeostasis and innate immunity, to enhance its adhesion to and invasion of mucosal tissues. HD5 promoted Shigella infection in vitro in a structure-dependent manner. Shigella, commonly devoid of an effective host-adhesion apparatus, preferentially targeted HD5 to augment its ability to colonize the intestinal epithelium through interactions with multiple bacterial membrane proteins. HD5 exacerbated infectivity and Shigella-induced pathology in a culture of human colorectal tissues and three animal models. Our findings illuminate how Shigella exploits innate immunity by turning HD5 into a virulence factor for infection, unveiling a mechanism of action for this highly proficient human pathogen.
Collapse
Affiliation(s)
- Dan Xu
- Key Laboratory of Biomedical Information Engineering of the Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an, China,Center for Translational Medicine, Frontier Institute of Science and Technology, Xi’an Jiaotong University,Institute of Human Virology and Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Chongbing Liao
- Key Laboratory of Biomedical Information Engineering of the Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an, China,Center for Translational Medicine, Frontier Institute of Science and Technology, Xi’an Jiaotong University
| | - Bing Zhang
- Key Laboratory of Biomedical Information Engineering of the Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an, China
| | - W. David Tolbert
- Institute of Human Virology and Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Wangxiao He
- Key Laboratory of Biomedical Information Engineering of the Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an, China,Center for Translational Medicine, Frontier Institute of Science and Technology, Xi’an Jiaotong University,Institute of Human Virology and Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Zhijun Dai
- The Second Affiliated Hospital, Xi’an Jiaotong University School of Medicine
| | - Wei Zhang
- Key Laboratory of Biomedical Information Engineering of the Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an, China
| | - Weirong Yuan
- Institute of Human Virology and Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Marzena Pazgier
- Institute of Human Virology and Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Jiankang Liu
- Key Laboratory of Biomedical Information Engineering of the Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an, China
| | - Jun Yu
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, Scotland, UK
| | | | - Charles L. Bevins
- Department of Microbiology and Immunology, University of California, School of Medicine, Davis, California, USA
| | - Yongping Shao
- Key Laboratory of Biomedical Information Engineering of the Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an, China,Center for Translational Medicine, Frontier Institute of Science and Technology, Xi’an Jiaotong University,Correspondence to: (lead contact) or
| | - Wuyuan Lu
- Key Laboratory of Biomedical Information Engineering of the Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an, China,Center for Translational Medicine, Frontier Institute of Science and Technology, Xi’an Jiaotong University,Institute of Human Virology and Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland, USA,Correspondence to: (lead contact) or
| |
Collapse
|
31
|
Abstract
The human gut microbiome performs prodigious physiological functions such as production of microbial metabolites, modulation of nutrient digestion and drug metabolism, control of immune system, and prevention of infection. Paradoxically, gut microbiome can also negatively orchestrate the host responses in diseases or chronic disorders, suggesting that the regulated and balanced host-gut microbiome crosstalk is a salient prerequisite in gastrointestinal physiology. To understand the pathophysiological role of host-microbiome crosstalk, it is critical to recreate in vivo relevant models of the host-gut microbiome ecosystem in human. However, controlling the multi-species microbial communities and their uncontrolled growth has remained a notable technical challenge. Furthermore, conventional two-dimensional (2D) or 3D culture systems do not recapitulate multicellular microarchitectures, mechanical dynamics, and tissue-specific functions. Here, we review recent advances and current pitfalls of in vitro and ex vivo models that display human GI functions. We also discuss how the disruptive technologies such as 3D organoids or a human organ-on-a-chip microphysiological system can contribute to better emulate host-gut microbiome crosstalks in health and disease. Finally, the medical and pharmaceutical significance of the gut microbiome-based personalized interventions is underlined as a future perspective.
Collapse
|
32
|
Lin Z, Cai X, Chen M, Ye L, Wu Y, Wang X, Lv Z, Shang Y, Qu D. Virulence and Stress Responses of Shigella flexneri Regulated by PhoP/PhoQ. Front Microbiol 2018; 8:2689. [PMID: 29379483 PMCID: PMC5775216 DOI: 10.3389/fmicb.2017.02689] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Accepted: 12/26/2017] [Indexed: 12/31/2022] Open
Abstract
The two-component signal transduction system PhoP/PhoQ is an important regulator for stress responses and virulence in most Gram-negative bacteria, but characterization of PhoP/PhoQ in Shigella has not been thoroughly investigated. In the present study, we found that deletion of phoPQ (ΔphoPQ) from Shigella flexneri 2a 301 (Sf301) resulted in a significant decline (reduced by more than 15-fold) in invasion of HeLa cells and Caco-2 cells, and less inflammation (− or +) compared to Sf301 (+++) in the guinea pig Sereny test. In low Mg2+ (10 μM) medium or pH 5 medium, the ΔphoPQ strain exhibited a growth deficiency compared to Sf301. The ΔphoPQ strain was more sensitive than Sf301 to polymyxin B, an important antimicrobial agent for treating multi-resistant Gram-negative infections. By comparing the transcriptional profiles of ΔphoPQ and Sf301 using DNA microarrays, 117 differentially expressed genes (DEGs) were identified, which were involved in Mg2+ transport, lipopolysaccharide modification, acid resistance, bacterial virulence, respiratory, and energy metabolism. Based on the reported PhoP box motif [(T/G) GTTTA-5nt-(T/G) GTTTA], we screened 38 suspected PhoP target operons in S. flexneri, and 11 of them (phoPQ, mgtA, slyB, yoaE, yrbL, icsA, yhiWX, rstA, hdeAB, pagP, and shf–rfbU-virK-msbB2) were demonstrated to be PhoP-regulated genes based on electrophoretic mobility shift assays and β-galactosidase assays. One of these PhoP-regulated genes, icsA, is a well-known virulence factor in S. flexneri. In conclusion, our data suggest that the PhoP/PhoQ system modulates S. flexneri virulence (in an icsA-dependent manner) and stress responses of Mg2+, pH and antibacterial peptides.
Collapse
Affiliation(s)
- Zhiwei Lin
- Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, School of Basic Medical Science and Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai, China
| | - Xia Cai
- Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, School of Basic Medical Science and Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai, China
| | - Mingliang Chen
- Shanghai Municipal Center for Disease Control and Prevention, Shanghai, China
| | - Lina Ye
- Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, School of Basic Medical Science and Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai, China
| | - Yang Wu
- Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, School of Basic Medical Science and Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai, China
| | - Xiaofei Wang
- Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, School of Basic Medical Science and Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai, China
| | - Zhihui Lv
- Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, School of Basic Medical Science and Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai, China
| | - Yongpeng Shang
- Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, School of Basic Medical Science and Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai, China
| | - Di Qu
- Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, School of Basic Medical Science and Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai, China
| |
Collapse
|
33
|
Belotserkovsky I, Sansonetti PJ. Shigella and Enteroinvasive Escherichia Coli. Curr Top Microbiol Immunol 2018; 416:1-26. [PMID: 30218158 DOI: 10.1007/82_2018_104] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Shigella and enteroinvasive Escherichia coli (EIEC) are gram-negative bacteria responsible for bacillary dysentery (shigellosis) in humans, which is characterized by invasion and inflammatory destruction of the human colonic epithelium. Different EIEC and Shigella subgroups rose independently from commensal E. coli through patho-adaptive evolution that included loss of functional genes interfering with the virulence and/or with the intracellular lifestyle of the bacteria, as well as acquisition of genetic elements harboring virulence genes. Among the latter is the large virulence plasmid encoding for a type three secretion system (T3SS), which enables translocation of virulence proteins (effectors) from the bacterium directly into the host cell cytoplasm. These effectors enable the pathogen to subvert epithelial cell functions, promoting its own uptake, replication in the host cytosol, and dissemination to adjacent cells while concomitantly inhibiting pro-inflammatory cell death. Furthermore, T3SS effectors are directly involved in Shigella manipulation of immune cells causing their dysfunction and promoting cell death. In the current chapter, we first describe the evolution of the enteroinvasive pathovars and then summarize the overall knowledge concerning the pathogenesis of these bacteria, with a particular focus on Shigella flexneri. Subversion of host cell functions in the human gut, both epithelial and immune cells, by different virulence factors is especially highlighted.
Collapse
Affiliation(s)
- Ilia Belotserkovsky
- Unité de Pathogénie Microbienne Moléculaire, Institut Pasteur, 28 Rue Du Dr Roux, 75724 Cedex 15, Paris, France.
| | - Philippe J Sansonetti
- Microbiologie et Maladies Infectieuses, Collège de France, 11 Place Marcelin Berthelot, 75005, Paris, France
| |
Collapse
|
34
|
Organoids, organs-on-chips and other systems, and microbiota. Emerg Top Life Sci 2017; 1:385-400. [PMID: 33525777 PMCID: PMC7289039 DOI: 10.1042/etls20170047] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Revised: 10/11/2017] [Accepted: 10/16/2017] [Indexed: 12/15/2022]
Abstract
The human gut microbiome is considered an organ in its entirety and has been the subject of extensive research due to its role in physiology, metabolism, digestion, and immune regulation. Disequilibria of the normal microbiome have been associated with the development of several gastrointestinal diseases, but the exact underlying interactions are not well understood. Conventional in vivo and in vitro modelling systems fail to faithfully recapitulate the complexity of the human host–gut microbiome, emphasising the requirement for novel systems that provide a platform to study human host–gut microbiome interactions with a more holistic representation of the human in vivo microenvironment. In this review, we outline the progression and applications of new and old modelling systems with particular focus on their ability to model and to study host–microbiome cross-talk.
Collapse
|
35
|
Sun CH, Wacquier B, Aguilar DI, Carayol N, Denis K, Boucherie S, Valencia-Gallardo C, Simsek C, Erneux C, Lehman A, Enninga J, Arbibe L, Sansonetti P, Dupont G, Combettes L, Tran Van Nhieu G. The Shigella type III effector IpgD recodes Ca 2+ signals during invasion of epithelial cells. EMBO J 2017; 36:2567-2580. [PMID: 28701483 PMCID: PMC5579377 DOI: 10.15252/embj.201696272] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 06/03/2017] [Accepted: 06/13/2017] [Indexed: 01/29/2023] Open
Abstract
The role of second messengers in the diversion of cellular processes by pathogens remains poorly studied despite their importance. Among these, Ca2+ virtually regulates all known cell processes, including cytoskeletal reorganization, inflammation, or cell death pathways. Under physiological conditions, cytosolic Ca2+ increases are transient and oscillatory, defining the so-called Ca2+ code that links cell responses to specific Ca2+ oscillatory patterns. During cell invasion, Shigella induces atypical local and global Ca2+ signals. Here, we show that by hydrolyzing phosphatidylinositol-(4,5)bisphosphate, the Shigella type III effector IpgD dampens inositol-(1,4,5)trisphosphate (InsP3) levels. By modifying InsP3 dynamics and diffusion, IpgD favors the elicitation of long-lasting local Ca2+ signals at Shigella invasion sites and converts Shigella-induced global oscillatory responses into erratic responses with atypical dynamics and amplitude. Furthermore, IpgD eventually inhibits InsP3-dependent responses during prolonged infection kinetics. IpgD thus acts as a pathogen regulator of the Ca2+ code implicated in a versatility of cell functions. Consistent with this function, IpgD prevents the Ca2+-dependent activation of calpain, thereby preserving the integrity of cell adhesion structures during the early stages of infection.
Collapse
Affiliation(s)
- Chun Hui Sun
- Equipe Communication Intercellulaire et Infections Microbiennes, Centre de Recherche Interdisciplinaire en Biologie (CIRB), Collège de France, Paris, France
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1050, Paris, France
- Centre National de la Recherche Scientifique (CNRS) UMR7241, Paris, France
- MEMOLIFE Laboratory of excellence and Paris Sciences et Lettres, Paris, France
- Université Paris Sud, Orsay, France
- Inserm, UMRS1174, Orsay, France
| | - Benjamin Wacquier
- Unité de Chronobiologie Théorique, Université Libre de Bruxelles, Brussels, Belgium
| | - Daniel I Aguilar
- Equipe Communication Intercellulaire et Infections Microbiennes, Centre de Recherche Interdisciplinaire en Biologie (CIRB), Collège de France, Paris, France
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1050, Paris, France
- Centre National de la Recherche Scientifique (CNRS) UMR7241, Paris, France
- MEMOLIFE Laboratory of excellence and Paris Sciences et Lettres, Paris, France
| | - Nathalie Carayol
- Equipe Communication Intercellulaire et Infections Microbiennes, Centre de Recherche Interdisciplinaire en Biologie (CIRB), Collège de France, Paris, France
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1050, Paris, France
- Centre National de la Recherche Scientifique (CNRS) UMR7241, Paris, France
- MEMOLIFE Laboratory of excellence and Paris Sciences et Lettres, Paris, France
| | - Kevin Denis
- Equipe Communication Intercellulaire et Infections Microbiennes, Centre de Recherche Interdisciplinaire en Biologie (CIRB), Collège de France, Paris, France
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1050, Paris, France
- Centre National de la Recherche Scientifique (CNRS) UMR7241, Paris, France
- MEMOLIFE Laboratory of excellence and Paris Sciences et Lettres, Paris, France
| | | | - Cesar Valencia-Gallardo
- Equipe Communication Intercellulaire et Infections Microbiennes, Centre de Recherche Interdisciplinaire en Biologie (CIRB), Collège de France, Paris, France
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1050, Paris, France
- Centre National de la Recherche Scientifique (CNRS) UMR7241, Paris, France
- MEMOLIFE Laboratory of excellence and Paris Sciences et Lettres, Paris, France
| | - Ceren Simsek
- Equipe Communication Intercellulaire et Infections Microbiennes, Centre de Recherche Interdisciplinaire en Biologie (CIRB), Collège de France, Paris, France
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1050, Paris, France
- Centre National de la Recherche Scientifique (CNRS) UMR7241, Paris, France
- MEMOLIFE Laboratory of excellence and Paris Sciences et Lettres, Paris, France
| | - Christophe Erneux
- Interdisciplinary Research Institute (IRIBHM), Université Libre de Bruxelles, Brussels, Belgium
| | - Alexandre Lehman
- Interdisciplinary Research Institute (IRIBHM), Université Libre de Bruxelles, Brussels, Belgium
| | - Jost Enninga
- Département de Biologie Cellulaire et Infection, Institut Pasteur, Unité de la Dynamique des Interactions Hôte-Pathogène, Paris, France
| | - Laurence Arbibe
- Equipe Plasticité du Génome et Infection, INSERM UMR_S1151 - CNRS UMR8253, Institut Necker Enfants Malades (INEM), Université Paris Descartes, Paris, France
| | - Philippe Sansonetti
- Département de Biologie Cellulaire et Infection, Unité de Pathogénie Microbienne Moléculaire, Paris, France
- Unité Inserm 1202, Institut Pasteur, Paris, France
- Collège de France, Paris, France
| | - Geneviève Dupont
- Unité de Chronobiologie Théorique, Université Libre de Bruxelles, Brussels, Belgium
| | | | - Guy Tran Van Nhieu
- Equipe Communication Intercellulaire et Infections Microbiennes, Centre de Recherche Interdisciplinaire en Biologie (CIRB), Collège de France, Paris, France
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1050, Paris, France
- Centre National de la Recherche Scientifique (CNRS) UMR7241, Paris, France
- MEMOLIFE Laboratory of excellence and Paris Sciences et Lettres, Paris, France
| |
Collapse
|
36
|
Maldonado-Contreras A, Birtley JR, Boll E, Zhao Y, Mumy KL, Toscano J, Ayehunie S, Reinecker HC, Stern LJ, McCormick BA. Shigella depends on SepA to destabilize the intestinal epithelial integrity via cofilin activation. Gut Microbes 2017; 8:544-560. [PMID: 28598765 PMCID: PMC5730386 DOI: 10.1080/19490976.2017.1339006] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Shigella is unique among enteric pathogens, as it invades colonic epithelia through the basolateral pole. Therefore, it has evolved the ability to breach the intestinal epithelial barrier to deploy an arsenal of effector proteins, which permits bacterial invasion and leads to a severe inflammatory response. However, the mechanisms used by Shigella to regulate epithelial barrier permeability remain unknown. To address this question, we used both an intestinal polarized model and a human ex-vivo model to further characterize the early events of host-bacteria interactions. Our results showed that secreted Serine Protease A (SepA), which belongs to the serine protease autotransporter of Enterobacteriaceae family, is responsible for critically disrupting the intestinal epithelial barrier. Such disruption facilitates bacterial transit to the basolateral pole of the epithelium, ultimately fostering the hallmarks of the disease pathology. SepA was found to cause a decrease in active LIM Kinase 1 (LIMK1) levels, a negative inhibitor of actin-remodeling proteins, namely cofilin. Correspondingly, we observed increased activation of cofilin, a major actin-polymerization factor known to control opening of tight junctions at the epithelial barrier. Furthermore, we resolved the crystal structure of SepA to elucidate its role on actin-dynamics and barrier disruption. The serine protease activity of SepA was found to be required for the regulatory effects on LIMK1 and cofilin, resulting in the disruption of the epithelial barrier during infection. Altogether, we demonstrate that SepA is indispensable for barrier disruption, ultimately facilitating Shigella transit to the basolateral pole where it effectively invades the epithelium.
Collapse
Affiliation(s)
- Ana Maldonado-Contreras
- Department of Microbiology and Physiological Systems, University of Massachusetts, Medical School, Worcester, MA, USA,CONTACT Beth A. McCormick ; Ana Maldonado-Contreras 55 Lake Ave N, Worcester, MA, 01655
| | - James R. Birtley
- Department of Pathology, University of Massachusetts, Medical School, Worcester, MA, USA
| | - Erik Boll
- Statens Serum Institut, Copenhagen, Denmark
| | - Yun Zhao
- Gastrointestinal Unit and Center for the Study of Inflammatory Bowel Disease, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Karen L. Mumy
- Naval Medical Research Unit Dayton, Wright-Patterson Air Force Base, Dayton, OH, USA
| | - Juan Toscano
- Department of Microbiology and Physiological Systems, University of Massachusetts, Medical School, Worcester, MA, USA
| | | | - Hans-Christian Reinecker
- Gastrointestinal Unit and Center for the Study of Inflammatory Bowel Disease, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Lawrence J. Stern
- Department of Pathology, University of Massachusetts, Medical School, Worcester, MA, USA
| | - Beth A. McCormick
- Department of Microbiology and Physiological Systems, University of Massachusetts, Medical School, Worcester, MA, USA,CONTACT Beth A. McCormick ; Ana Maldonado-Contreras 55 Lake Ave N, Worcester, MA, 01655
| |
Collapse
|
37
|
Wiedemann A, Mijouin L, Ayoub MA, Barilleau E, Canepa S, Teixeira-Gomes AP, Le Vern Y, Rosselin M, Reiter E, Velge P. Identification of the epidermal growth factor receptor as the receptor for Salmonella Rck-dependent invasion. FASEB J 2016; 30:4180-4191. [PMID: 27609774 DOI: 10.1096/fj.201600701r] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Accepted: 08/22/2016] [Indexed: 11/11/2022]
Abstract
The Salmonella Rck outer membrane protein binds to the cell surface, which leads to bacterial internalization via a Zipper mechanism. This invasion process requires induction of cellular signals, including phosphorylation of tyrosine proteins, and activation of c-Src and PI3K, which arises as a result of an interaction with a host cell surface receptor. In this study, epidermal growth factor receptor (EGFR) was identified as the cell signaling receptor required for Rck-mediated adhesion and internalization. First, Rck-mediated adhesion and internalization were shown to be altered when EGFR expression and activity were modulated. Then, immunoprecipitations were performed to demonstrate the Rck-EGFR interaction. Furthermore, surface plasmon resonance biosensor and homogeneous time-resolved fluorescence technologies were used to demonstrate the direct interaction of Rck with the extracellular domain of human EGFR. Finally, our study strongly suggests a noncompetitive binding of Rck and EGF to EGFR. Overall, these results demonstrate that Rck is able to bind to EGFR and thereby establish a tight adherence to provide a signaling cascade, which leads to internalization of Rck-expressing bacteria.-Wiedemann, A., Mijouin, L., Ayoub, M. A., Barilleau, E., Canepa, S., Teixeira-Gomes, A. P., Le Vern, Y., Rosselin, M., Reiter, E., Velge, P. Identification of the epidermal growth factor receptor as the receptor for Salmonella Rck-dependent invasion.
Collapse
Affiliation(s)
- Agnès Wiedemann
- Institut National de la Recherche Agronomique, Unités Mixtes de Recherche 1282, Infectiologie et Santé Publique, Nouzilly, France; .,Université François Rabelais de Tours, Unités Mixtes de Recherche 1282, Infectiologie et Santé Publique, Tours, France
| | - Lily Mijouin
- Institut National de la Recherche Agronomique, Unités Mixtes de Recherche 1282, Infectiologie et Santé Publique, Nouzilly, France.,Université François Rabelais de Tours, Unités Mixtes de Recherche 1282, Infectiologie et Santé Publique, Tours, France
| | - Mohammed Akli Ayoub
- Institut National de la Recherche Agronomique, Unités Mixtes de Recherche 85, Unité Physiologie de la Reproduction et des Comportements, Nouzilly, France.,Centre National de la Recherche Scientifique, Unités Mixtes de Recherche 7247, Nouzilly, France.,L'Institut Français du Cheval et de L'Équitation, Nouzilly, France.,Le Studium Loire Valley Institute for Advanced Studies, Orléans, France; and
| | - Emilie Barilleau
- Institut National de la Recherche Agronomique, Unités Mixtes de Recherche 1282, Infectiologie et Santé Publique, Nouzilly, France.,Université François Rabelais de Tours, Unités Mixtes de Recherche 1282, Infectiologie et Santé Publique, Tours, France
| | - Sylvie Canepa
- Centre National de la Recherche Scientifique, Unités Mixtes de Recherche 7247, Nouzilly, France.,L'Institut Français du Cheval et de L'Équitation, Nouzilly, France.,Institut National de la Recherche Agronomique, Unités Mixtes de Recherche 85, Plate-forme d'Analyse Intégrative des Biomolécules, Nouzilly, France
| | - Ana Paula Teixeira-Gomes
- Institut National de la Recherche Agronomique, Unités Mixtes de Recherche 1282, Infectiologie et Santé Publique, Nouzilly, France.,Université François Rabelais de Tours, Unités Mixtes de Recherche 1282, Infectiologie et Santé Publique, Tours, France.,Institut National de la Recherche Agronomique, Unités Mixtes de Recherche 85, Plate-forme d'Analyse Intégrative des Biomolécules, Nouzilly, France
| | - Yves Le Vern
- Institut National de la Recherche Agronomique, Unités Mixtes de Recherche 1282, Infectiologie et Santé Publique, Nouzilly, France.,Université François Rabelais de Tours, Unités Mixtes de Recherche 1282, Infectiologie et Santé Publique, Tours, France.,Institut National de la Recherche Agronomique, Unités Mixtes de Recherche 85, Plate-forme d'Analyse Intégrative des Biomolécules, Nouzilly, France
| | - Manon Rosselin
- Institut National de la Recherche Agronomique, Unités Mixtes de Recherche 1282, Infectiologie et Santé Publique, Nouzilly, France.,Université François Rabelais de Tours, Unités Mixtes de Recherche 1282, Infectiologie et Santé Publique, Tours, France
| | - Eric Reiter
- Institut National de la Recherche Agronomique, Unités Mixtes de Recherche 85, Unité Physiologie de la Reproduction et des Comportements, Nouzilly, France.,Centre National de la Recherche Scientifique, Unités Mixtes de Recherche 7247, Nouzilly, France.,L'Institut Français du Cheval et de L'Équitation, Nouzilly, France
| | - Philippe Velge
- Institut National de la Recherche Agronomique, Unités Mixtes de Recherche 1282, Infectiologie et Santé Publique, Nouzilly, France.,Université François Rabelais de Tours, Unités Mixtes de Recherche 1282, Infectiologie et Santé Publique, Tours, France
| |
Collapse
|
38
|
Wang X, Jiang F, Zheng J, Chen L, Dong J, Sun L, Zhu Y, Liu B, Yang J, Yang G, Jin Q. The outer membrane phospholipase A is essential for membrane integrity and type III secretion in Shigella flexneri. Open Biol 2016; 6:rsob.160073. [PMID: 27655730 PMCID: PMC5043575 DOI: 10.1098/rsob.160073] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2016] [Accepted: 08/31/2016] [Indexed: 12/11/2022] Open
Abstract
Outer membrane phospholipase A (OMPLA) is an enzyme located in the outer membrane of Gram-negative bacteria. OMPLA exhibits broad substrate specificity, and some of its substrates are located in the cellular envelope. Generally, the enzymatic activity can only be induced by perturbation of the cell envelope integrity through diverse methods. Although OMPLA has been thoroughly studied as a membrane protein in Escherichia coli and is constitutively expressed in many other bacterial pathogens, little is known regarding the functions of OMPLA during the process of bacterial infection. In this study, the proteomic and transcriptomic data indicated that OMPLA in Shigella flexneri, termed PldA, both stabilizes the bacterial membrane and is involved in bacterial infection under ordinary culture conditions. A series of physiological assays substantiated the disorganization of the bacterial outer membrane and the periplasmic space in the ΔpldA mutant strain. Furthermore, the ΔpldA mutant strain showed decreased levels of type III secretion system expression, contributing to the reduced internalization efficiency in host cells. The results of this study support that PldA, which is widespread across Gram-negative bacteria, is an important factor for the bacterial life cycle, particularly in human pathogens.
Collapse
Affiliation(s)
- Xia Wang
- MOH Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, People's Republic of China
| | - Feng Jiang
- MOH Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, People's Republic of China
| | - Jianhua Zheng
- MOH Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, People's Republic of China
| | - Lihong Chen
- MOH Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, People's Republic of China
| | - Jie Dong
- MOH Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, People's Republic of China
| | - Lilian Sun
- MOH Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, People's Republic of China
| | - Yafang Zhu
- MOH Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, People's Republic of China
| | - Bo Liu
- MOH Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, People's Republic of China
| | - Jian Yang
- MOH Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, People's Republic of China
| | - Guowei Yang
- MOH Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, People's Republic of China
| | - Qi Jin
- MOH Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, People's Republic of China
| |
Collapse
|
39
|
Ra EA, Lee TA, Won Kim S, Park A, Choi HJ, Jang I, Kang S, Hee Cheon J, Cho JW, Eun Lee J, Lee S, Park B. TRIM31 promotes Atg5/Atg7-independent autophagy in intestinal cells. Nat Commun 2016; 7:11726. [PMID: 27216961 PMCID: PMC4890305 DOI: 10.1038/ncomms11726] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Accepted: 04/26/2016] [Indexed: 01/08/2023] Open
Abstract
Autophagy is responsible for the bulk degradation of cytosolic constituents and plays an essential role in the intestinal epithelium by controlling beneficial host-bacterial relationships. Atg5 and Atg7 are thought to be critical for autophagy. However, Atg5- or Atg7-deficient cells still form autophagosomes and autolysosomes, and are capable of removing proteins or bacteria. Here, we report that human TRIM31 (tripartite motif), an intestine-specific protein localized in mitochondria, is essential for promoting lipopolysaccharide-induced Atg5/Atg7-independent autophagy. TRIM31 directly interacts with phosphatidylethanolamine in a palmitoylation-dependent manner, leading to induction of autolysosome formation. Depletion of endogenous TRIM31 significantly increases the number of intestinal epithelial cells containing invasive bacteria. Crohn's disease patients display TRIM31 downregulation. Human cytomegalovirus-infected intestinal cells show a decrease in TRIM31 expression as well as a significant increase in bacterial load, reversible by the introduction of wild-type TRIM31. We provide insight into an alternative autophagy pathway that protects against intestinal pathogenic bacterial infection.
Collapse
Affiliation(s)
- Eun A. Ra
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, South Korea
| | - Taeyun A. Lee
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, South Korea
| | - Seung Won Kim
- Department of Internal Medicine and Institute of Gastroenterology, Yonsei University College of Medicine, Seoul 03722, South Korea
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul 03722, South Korea
| | - Areum Park
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, South Korea
| | - Hyun jin Choi
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, South Korea
| | - Insook Jang
- Department of Integrated OMICS for Biomedical Science, Yonsei University, Seoul 03722, South Korea
| | - Sujin Kang
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, South Korea
| | - Jae Hee Cheon
- Department of Internal Medicine and Institute of Gastroenterology, Yonsei University College of Medicine, Seoul 03722, South Korea
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul 03722, South Korea
| | - Jin Won Cho
- Department of Integrated OMICS for Biomedical Science, Yonsei University, Seoul 03722, South Korea
| | - Ji Eun Lee
- Department of Health Science and Technology, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul 06351, South Korea
- Samsung Genome Institute (SGI), Samsung Medical Center, Seoul 06351, South Korea
| | - Sungwook Lee
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, South Korea
| | - Boyoun Park
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, South Korea
| |
Collapse
|
40
|
Abstract
The brush border on the apical surface of enterocytes is a highly specialized structure well-adapted for efficient digestion and nutrient transport, whilst at the same time providing a protective barrier for the intestinal mucosa. The brush border is constituted of a densely ordered array of microvilli, protrusions of the plasma membrane, which are supported by actin-based microfilaments and interacting proteins and anchored in an apical network of actomyosin and intermediate filaments, the so-called terminal web. The highly dynamic, specialized apical domain is both an essential partner for the gut microbiota and an efficient signalling platform that enables adaptation to physiological stimuli from the external and internal milieu. Nevertheless, genetic alterations or various pathological stresses, such as infection, inflammation, and mechanical or nutritional alterations, can jeopardize this equilibrium and compromise intestinal functions. Long-time neglected, the intestinal brush-border shall be enlightening again as the central actor of the complex but essential intestinal homeostasis. Here, we review the processes and components involved in brush border organization and discuss pathological mechanisms that can induce brush border defects and their physiological consequences.
Collapse
|
41
|
Cultured enterocytes internalise bacteria across their basolateral surface for, pathogen-inhibitable, trafficking to the apical compartment. Sci Rep 2015; 5:17359. [PMID: 26612456 PMCID: PMC4661573 DOI: 10.1038/srep17359] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Accepted: 10/23/2015] [Indexed: 01/13/2023] Open
Abstract
In vitro- and in vivo-polarised absorptive epithelia (enterocytes) are considered to be non-phagocytic towards bacteria with invasive pathogenic strains relying on virulence factors to 'force' entry. Here, we report a serendipitous discovery that questions these beliefs. Thus, we uncover in well-established models of human small (Caco-2; TC-7) and large (T84) intestinal enterocytes a polarization-dependent mechanism that can transfer millions of bacteria from the basal to apical compartment. Antibiotic-protection assays, confocal imaging and drug inhibitor data are consistent with a transcellular route in which internalized, basolateral-membrane enclosed bacteria are trafficked to and across the apical surface. Basal-to-apical transport of non-pathogenic bacteria (and inert beads) challenged the idea of pathogens relying on virulence factors to force entry. Indeed, studies with Salmonella demonstrated that it's entry-forcing virulence factor (SPI-I) was not required to enter via the basolateral surface but to promote another virulence-associated event (intra-enterocyte accumulation).
Collapse
|
42
|
Abstract
Shigella species are the causative agents of bacillary dysentery in humans, an invasive disease in which the bacteria enter the cells of the epithelial layer of the large intestine, causing extensive tissue damage and inflammation. They rely on a plasmid-encoded type III secretion system (TTSS) to cause disease; this system and its regulation have been investigated intensively at the molecular level for decades. The lessons learned have not only deepened our knowledge of Shigella biology but also informed in important ways our understanding of the mechanisms used by other pathogenic bacteria to cause disease and to control virulence gene expression. In addition, the Shigella story has played a central role in the development of our appreciation of the contribution of horizontal DNA transfer to pathogen evolution.A 30-kilobase-pair "Entry Region" of the 230-kb virulence plasmid lies at the heart of the Shigella pathogenesis system. Here are located the virB and mxiE regulatory genes and most of the structural genes involved in the expression of the TTSS and its effector proteins. Expression of the virulence genes occurs in response to an array of environmental signals, including temperature, osmolarity, and pH.At the top of the regulatory hierarchy and lying on the plasmid outside the Entry Region isvirF, encoding an AraC-like transcription factor.Virulence gene expression is also controlled by chromosomal genes,such as those encoding the nucleoid-associated proteins H-NS, IHF, and Fis, the two-component regulators OmpR/EnvZ and CpxR/CpxA, the anaerobic regulator Fnr, the iron-responsive regulator Fur, and the topoisomerases of the cell that modulate DNA supercoiling. Small regulatory RNAs,the RNA chaperone Hfq,and translational modulation also affect the expression of the virulence phenotypetranscriptionally and/orposttranscriptionally.
Collapse
|
43
|
Shi R, Yang X, Chen L, Chang HT, Liu HY, Zhao J, Wang XW, Wang CQ. Pathogenicity of Shigella in chickens. PLoS One 2014; 9:e100264. [PMID: 24949637 PMCID: PMC4064985 DOI: 10.1371/journal.pone.0100264] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2013] [Accepted: 05/25/2014] [Indexed: 12/29/2022] Open
Abstract
Shigellosis in chickens was first reported in 2004. This study aimed to determine the pathogenicity of Shigella in chickens and the possibility of cross-infection between humans and chickens. The pathogenicity of Shigella in chickens was examined via infection of three-day-old SPF chickens with Shigella strain ZD02 isolated from a human patient. The virulence and invasiveness were examined by infection of the chicken intestines and primary chicken intestinal epithelial cells. The results showed Shigella can cause death via intraperitoneal injection in SPF chickens, but only induce depression via crop injection. Immunohistochemistry and transmission electron microscopy revealed the Shigella can invade the intestinal epithelia. Immunohistochemistry of the primary chicken intestinal epithelial cells infected with Shigella showed the bacteria were internalized into the epithelial cells. Electron microscopy also confirmed that Shigella invaded primary chicken intestinal epithelia and was encapsulated by phagosome-like membranes. Our data demonstrate that Shigella can invade primary chicken intestinal epithelial cells in vitro and chicken intestinal mucosa in vivo, resulting in pathogenicity and even death. The findings suggest Shigella isolated from human or chicken share similar pathogenicity as well as the possibility of human-poultry cross-infection, which is of public health significance.
Collapse
Affiliation(s)
- Run Shi
- Collage of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou, People's Republic of China
| | - Xia Yang
- Collage of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou, People's Republic of China
| | - Lu Chen
- Collage of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou, People's Republic of China
| | - Hong-tao Chang
- Collage of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou, People's Republic of China
| | - Hong-ying Liu
- Collage of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou, People's Republic of China
| | - Jun Zhao
- Collage of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou, People's Republic of China
| | - Xin-wei Wang
- Collage of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou, People's Republic of China
| | - Chuan-qing Wang
- Collage of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou, People's Republic of China
| |
Collapse
|
44
|
Meghraoui A, Schiavolin L, Allaoui A. Single amino acid substitutions on the needle tip protein IpaD increased Shigella virulence. Microbes Infect 2014; 16:532-9. [PMID: 24726700 DOI: 10.1016/j.micinf.2014.03.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2014] [Revised: 03/22/2014] [Accepted: 03/31/2014] [Indexed: 11/19/2022]
Abstract
Infection of colonic epithelial cells by Shigella is associated with the type III secretion system, which serves as a molecular syringe to inject effectors into host cells. This system includes an extracellular needle used as a conduit for secreted proteins. Two of these proteins, IpaB and IpaD, dock at the needle tip to control secretion and are also involved in the insertion of a translocation pore into host cell membrane allowing effector delivery. To better understand the function of IpaD, we substituted thirteen residues conserved among homologous proteins in other bacterial species. Generated variants were tested for their ability to surface expose IpaB and IpaD, to control secretion, to insert the translocation pore, and to invade host cells. In addition to a first group of seven ipaD variants that behaved similarly to the wild-type strain, we identified a second group with mutations V314D and I319D that deregulated secretion of all effectors, but remained fully invasive. Moreover, we identified a third group with mutations Y153A, T161D, Q165L and Y276A, that exhibited increased levels of translocators secretion, pore formation, and cell entry. Altogether, our results offer a better understanding of the role of IpaD in the control of Shigella virulence.
Collapse
Affiliation(s)
- Alaeddine Meghraoui
- Laboratoire de Bactériologie Moléculaire, Faculté de Médecine, Université Libre de Bruxelles, Route de Lennik, 808, 1070 Bruxelles, Belgium
| | - Lionel Schiavolin
- Laboratoire de Bactériologie Moléculaire, Faculté de Médecine, Université Libre de Bruxelles, Route de Lennik, 808, 1070 Bruxelles, Belgium
| | - Abdelmounaaïm Allaoui
- Laboratoire de Bactériologie Moléculaire, Faculté de Médecine, Université Libre de Bruxelles, Route de Lennik, 808, 1070 Bruxelles, Belgium.
| |
Collapse
|
45
|
Pathogenesis of human enterovirulent bacteria: lessons from cultured, fully differentiated human colon cancer cell lines. Microbiol Mol Biol Rev 2014; 77:380-439. [PMID: 24006470 DOI: 10.1128/mmbr.00064-12] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Hosts are protected from attack by potentially harmful enteric microorganisms, viruses, and parasites by the polarized fully differentiated epithelial cells that make up the epithelium, providing a physical and functional barrier. Enterovirulent bacteria interact with the epithelial polarized cells lining the intestinal barrier, and some invade the cells. A better understanding of the cross talk between enterovirulent bacteria and the polarized intestinal cells has resulted in the identification of essential enterovirulent bacterial structures and virulence gene products playing pivotal roles in pathogenesis. Cultured animal cell lines and cultured human nonintestinal, undifferentiated epithelial cells have been extensively used for understanding the mechanisms by which some human enterovirulent bacteria induce intestinal disorders. Human colon carcinoma cell lines which are able to express in culture the functional and structural characteristics of mature enterocytes and goblet cells have been established, mimicking structurally and functionally an intestinal epithelial barrier. Moreover, Caco-2-derived M-like cells have been established, mimicking the bacterial capture property of M cells of Peyer's patches. This review intends to analyze the cellular and molecular mechanisms of pathogenesis of human enterovirulent bacteria observed in infected cultured human colon carcinoma enterocyte-like HT-29 subpopulations, enterocyte-like Caco-2 and clone cells, the colonic T84 cell line, HT-29 mucus-secreting cell subpopulations, and Caco-2-derived M-like cells, including cell association, cell entry, intracellular lifestyle, structural lesions at the brush border, functional lesions in enterocytes and goblet cells, functional and structural lesions at the junctional domain, and host cellular defense responses.
Collapse
|
46
|
Lima CBC, Santos SAD, Andrade Júnior DRD. Hypoxic stress, hepatocytes and CACO-2 viability and susceptibility to Shigella flexneri invasion. Rev Inst Med Trop Sao Paulo 2014; 55:341-6. [PMID: 24037289 PMCID: PMC4105072 DOI: 10.1590/s0036-46652013000500008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2012] [Accepted: 03/06/2013] [Indexed: 12/18/2022] Open
Abstract
SUMMARY Inflammation due to Shigella flexneri can cause damage to the colonic mucosa and cell death by necrosis and apoptosis. This bacteria can reach the bloodstream in this way, and the liver through portal veins. Hypoxia is a condition present in many human diseases, and it may induce bacterial translocation from intestinal lumen. We studied the ability of S. flexneri to invade rat hepatocytes and Caco-2 cells both in normoxic and hypoxic microenvironments, as well as morphological and physiological alterations in these cells after infection under hypoxia. We used the primary culture of rat hepatocytes as a model of study. We analyzed the following parameters in normoxic and hypoxic conditions: morphology, cell viability, bacterial recovery and lactate dehydrogenase (LDH) released. The results showed that there were fewer bacteria within the Caco-2 cells than in hepatocytes in normoxic and hypoxic conditions. We observed that the higher the multiplicity of infection (MOI) the greater the bacterial recovery in hepatocytes. The hypoxic condition decreased the bacterial recovery in hepatocytes. The cytotoxicity evaluated by LDH released by cells was significantly higher in cells submitted to hypoxia than normoxia. Caco-2 cells in normoxia released 63% more LDH than hepatocytes. LDH increased 164% when hepatocytes were submitted to hypoxia and just 21% when Caco-2 cells were in the same condition. The apoptosis evaluated by Tunel was significantly higher in cells submitted to hypoxia than normoxia. When comparing hypoxic cells, we obtained more apoptotic hepatocytes than apoptotic Caco-2 cells. Concluding our results contribute to a better knowledge of interactions between studied cells and Shigella flexneri. These data may be useful in the future to define strategies to combat this virulent pathogen.
Collapse
Affiliation(s)
- Camila Bárbara Cantalupo Lima
- Laboratory of Bacteriology (LIM 54), Department of Infectious Diseases, Faculty of Medicine, University of Sao Paulo, Sao PauloSP, Brazil, , ,
| | | | | |
Collapse
|
47
|
Barreau F, Hugot JP. Intestinal barrier dysfunction triggered by invasive bacteria. Curr Opin Microbiol 2014; 17:91-8. [PMID: 24440560 DOI: 10.1016/j.mib.2013.12.003] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2013] [Revised: 12/16/2013] [Accepted: 12/17/2013] [Indexed: 12/31/2022]
Abstract
The ability to control uptake across the mucosa and to protect the gut from harmful substances present in the lumen is defined as intestinal barrier function. Two routes are usually distinguished for transepithelial transport. The paracellular route allows the passage of ions and small molecules and is mainly regulated by tight junctions (TJ). The transcellular route concerns large molecules or small particles (including bacteria) and is mediated by cell endocytosis and intracellular vesicular traffic. Enteropathogenic bacteria increase the transcellular permeability, especially in the follicle-associated epithelium. They also modulate TJ opening via the redistribution of TJ proteins and the activation of the myosin light chain kinase (MLCK). This review focuses on the molecular mechanisms involved in the bacteria-induced barrier defect and briefly discusses their consequences in human diseases.
Collapse
Affiliation(s)
- F Barreau
- Université Paris-Diderot Sorbonne Paris-Cité, UMR 843, F-75018 Paris, France; INSERM, UMR 843, F-75018 Paris, France; Labex inflamex, F-75018 Paris, France; INSERM, UMR 1043, Centre de Physiopathologie de Toulouse, Université de Toulouse, France.
| | - J P Hugot
- Université Paris-Diderot Sorbonne Paris-Cité, UMR 843, F-75018 Paris, France; INSERM, UMR 843, F-75018 Paris, France; Labex inflamex, F-75018 Paris, France; Assistance Publique Hôpitaux de Paris, Hôpital Robert Debré, F-75019 Paris, France.
| |
Collapse
|
48
|
Ho SM, Johnson A, Tarapore P, Janakiram V, Zhang X, Leung YK. Environmental epigenetics and its implication on disease risk and health outcomes. ILAR J 2014; 53:289-305. [PMID: 23744968 DOI: 10.1093/ilar.53.3-4.289] [Citation(s) in RCA: 161] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
This review focuses on how environmental factors through epigenetics modify disease risk and health outcomes. Major epigenetic events, such as histone modifications, DNA methylation, and microRNA expression, are described. The function of dose, duration, composition, and window of exposure in remodeling the individual's epigenetic terrain and disease susceptibility are addressed. The ideas of lifelong editing of early-life epigenetic memories, transgenerational effects through germline transmission, and the potential role of hydroxylmethylation of cytosine in developmental reprogramming are discussed. Finally, the epigenetic effects of several major classes of environmental factors are reviewed in the context of pathogenesis of disease. These include endocrine disruptors, tobacco smoke, polycyclic aromatic hydrocarbons, infectious pathogens, particulate matter, diesel exhaust particles, dust mites, fungi, heavy metals, and other indoor and outdoor pollutants. We conclude that the summation of epigenetic modifications induced by multiple environmental exposures, accumulated over time, represented as broad or narrow, acute or chronic, developmental or lifelong, may provide a more precise assessment of risk and consequences. Future investigations may focus on their use as readouts or biomarkers of the totality of past exposure for the prediction of future disease risk and the prescription of effective countermeasures.
Collapse
Affiliation(s)
- Shuk-Mei Ho
- Division of Environmental Genetics and Molecular Toxicology, Department of Environmental Health, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA.
| | | | | | | | | | | |
Collapse
|
49
|
Fiorentino M, Levine MM, Sztein MB, Fasano A. Effect of wild-type Shigella species and attenuated Shigella vaccine candidates on small intestinal barrier function, antigen trafficking, and cytokine release. PLoS One 2014; 9:e85211. [PMID: 24416363 PMCID: PMC3887025 DOI: 10.1371/journal.pone.0085211] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Accepted: 11/27/2013] [Indexed: 01/22/2023] Open
Abstract
Bacterial dysentery due to Shigella species is a major cause of morbidity and mortality worldwide. The pathogenesis of Shigella is based on the bacteria's ability to invade and replicate within the colonic epithelium, resulting in severe intestinal inflammatory response and epithelial destruction. Although the mechanisms of pathogenesis of Shigella in the colon have been extensively studied, little is known on the effect of wild-type Shigella on the small intestine and the role of the host response in the development of the disease. Moreover, to the best of our knowledge no studies have described the effects of apically administered Shigella flexneri 2a and S. dysenteriae 1 vaccine strains on human small intestinal enterocytes. The aim of this study was to assess the coordinated functional and immunological human epithelial responses evoked by strains of Shigella and candidate vaccines on small intestinal enterocytes. To model the interactions of Shigella with the intestinal mucosa, we apically exposed monolayers of human intestinal Caco2 cells to increasing bacterial inocula. We monitored changes in paracellular permeability, examined the organization of tight-junctions and the pro-inflammatory response of epithelial cells. Shigella infection of Caco2 monolayers caused severe mucosal damage, apparent as a drastic increase in paracellular permeability and disruption of tight junctions at the cell-cell boundary. Secretion of pro-inflammatory IL-8 was independent of epithelial barrier dysfunction. Shigella vaccine strains elicited a pro-inflammatory response without affecting the intestinal barrier integrity. Our data show that wild-type Shigella infection causes a severe alteration of the barrier function of a small intestinal cell monolayer (a proxy for mucosa) and might contribute (along with enterotoxins) to the induction of watery diarrhea. Diarrhea may be a mechanism by which the host attempts to eliminate harmful bacteria and transport them from the small to the large intestine where they invade colonocytes inducing a strong inflammatory response.
Collapse
Affiliation(s)
- Maria Fiorentino
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital, Charlestown, Massachusetts, United States of America
| | - Myron M. Levine
- Center for Vaccine Development and the Departments of Pediatrics and Medicine, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Marcelo B. Sztein
- Center for Vaccine Development and the Departments of Pediatrics and Medicine, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Alessio Fasano
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital, Charlestown, Massachusetts, United States of America
| |
Collapse
|
50
|
Jain P, Li J, Porubsky P, Neuenswander B, Egan SM, Aubé J, Rogers S. 3-Substituted Biquinolinium Inhibitors of AraC Family Transcriptional Activator VirF from S. flexneri Obtained Through In Situ Chemical Ionization of 3,4-Disubstituted Dihydroquinolines. RSC Adv 2014; 4:39809-39816. [PMID: 25258678 DOI: 10.1039/c4ra08384a] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
During a structure-activity relationship optimization campaign to develop an inhibitor of AraC family transcriptional activators, we discovered an unexpected transformation of a previously reported inhibitor that occurs under the assay conditions. Once placed in the assay media, the 3, 4-disubstituted dihydroquinoline core of the active analogue rapidly undergoes a decomposition reaction to a quaternary 3-substituted biquinolinium. Further examination established an SAR for this chemotype while also demonstrating its resilience to irreversible binding of biologically relevant nucleophiles.
Collapse
Affiliation(s)
- Prashi Jain
- Center for Chemical Methodologies and Library Development, The University of Kansas, 2034 Becker Drive, Lawrence, Kansas, 66047, USA
| | - Jiaqin Li
- Department of Molecular Biosciences, University of Kansas, Lawrence, KS, USA
| | - Patrick Porubsky
- Center for Chemical Methodologies and Library Development, The University of Kansas, 2034 Becker Drive, Lawrence, Kansas, 66047, USA
| | - Benjamin Neuenswander
- Specialized Chemistry Center, The University of Kansas, 2034 Becker Drive, Lawrence, Kansas 66047, USA
| | - Susan M Egan
- Department of Molecular Biosciences, University of Kansas, Lawrence, KS, USA
| | - Jeffrey Aubé
- Center for Chemical Methodologies and Library Development, The University of Kansas, 2034 Becker Drive, Lawrence, Kansas, 66047, USA ; Specialized Chemistry Center, The University of Kansas, 2034 Becker Drive, Lawrence, Kansas 66047, USA ; University of Kansas Center of Biomedical Research Excellence, Center for Cancer Experimental Therapeutics, 2034 Becker Drive, Lawrence, KS, USA ; Department of Medicinal Chemistry, University of Kansas, Lawrence, KS, USA
| | - Steven Rogers
- University of Kansas Center of Biomedical Research Excellence, Center for Cancer Experimental Therapeutics, 2034 Becker Drive, Lawrence, KS, USA
| |
Collapse
|