1
|
Benjamin SV, Jégouzo SAF, Lieng C, Daniels C, Coispeau M, Lau RJ, Kim S, Metaxa Y, Philpott J, Li T, Dai C, Wang X, Newby ML, Pier GB, Crispin M, Clements A, Taylor ME, Drickamer K. A human lectin array for characterizing host-pathogen interactions. J Biol Chem 2024; 300:107869. [PMID: 39384043 PMCID: PMC11566865 DOI: 10.1016/j.jbc.2024.107869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 09/21/2024] [Accepted: 10/01/2024] [Indexed: 10/11/2024] Open
Abstract
A human lectin array has been developed to probe the interactions of innate immune receptors with pathogenic and commensal microorganisms. Following the successful introduction of a lectin array containing all of the cow C-type carbohydrate-recognition domains (CRDs), a human array described here contains the C-type CRDs as well as CRDs from other classes of sugar-binding receptors, including galectins, siglecs, R-type CRDs, ficolins, intelectins, and chitinase-like lectins. The array is constructed with CRDs modified with single-site biotin tags, ensuring that the sugar-binding sites in CRDs are displayed on a streptavidin-coated surface in a defined orientation and are accessible to the surfaces of microbes. A common approach used for expression and display of CRDs from all of the different structural categories of glycan-binding receptors allows comparisons across lectin families. In addition to previously documented protocols for binding of fluorescently labeled bacteria, methods have been developed for detecting unlabeled bacteria bound to the array by counter-staining with DNA-binding dye. Screening has also been undertaken with viral glycoproteins and bacterial and fungal polysaccharides. The array provides an unbiased screen for sugar ligands that interact with receptors and many show binding not anticipated from earlier studies. For example, some of the galectins bind with high affinity to bacterial glycans that lack lactose or N-acetyllactosamine. The results demonstrate the utility of the human lectin array for providing a unique overview of the interactions of multiple classes of glycan-binding proteins in the innate immune system with different types of microorganisms.
Collapse
Affiliation(s)
- Stefi V Benjamin
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | - Sabine A F Jégouzo
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | - Chloe Lieng
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | - Connor Daniels
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | - Marine Coispeau
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | - Rikin J Lau
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | - Suyeon Kim
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | - Yasmine Metaxa
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | - James Philpott
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | - Tiannuo Li
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | - Chao Dai
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | - Xin Wang
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | - Maddy L Newby
- School of Biological Sciences, University of Southampton, United Kingdom
| | - Gerald B Pier
- Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Max Crispin
- School of Biological Sciences, University of Southampton, United Kingdom
| | - Abigail Clements
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | - Maureen E Taylor
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | - Kurt Drickamer
- Department of Life Sciences, Imperial College London, London, United Kingdom.
| |
Collapse
|
2
|
Jin J, Yuan Y, Xian W, Tang Z, Fu J, Liu X. The ever-increasing necessity of mass spectrometry in dissecting protein post-translational modifications catalyzed by bacterial effectors. Mol Microbiol 2023. [PMID: 37127430 DOI: 10.1111/mmi.15071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 04/20/2023] [Accepted: 04/21/2023] [Indexed: 05/03/2023]
Abstract
Protein post-translational modifications (PTMs), such as ADP-ribosylation and phosphorylation, regulate multiple fundamental biological processes in cells. During bacterial infection, effector proteins are delivered into host cells through dedicated bacterial secretion systems and can modulate important cellular pathways by covalently modifying their host targets. These strategies enable intruding bacteria to subvert various host processes, thereby promoting their own survival and proliferation. Despite rapid expansion of our understanding of effector-mediated PTMs in host cells, analytical measurements of these molecular events still pose significant challenges in the study of host-pathogen interactions. Nevertheless, with major technical breakthroughs in the last two decades, mass spectrometry (MS) has evolved to be a valuable tool for detecting protein PTMs and mapping modification sites. Additionally, large-scale PTM profiling, facilitated by different enrichment strategies prior to MS analysis, allows high-throughput screening of host enzymatic substrates of bacterial effectors. In this review, we summarize the advances in the studies of two representative PTMs (i.e., ADP-ribosylation and phosphorylation) catalyzed by bacterial effectors during infection. Importantly, we will discuss the ever-increasing role of MS in understanding these molecular events and how the latest MS-based tools can aid in future studies of this booming area of pathogenic bacteria-host interactions.
Collapse
Affiliation(s)
- Jie Jin
- Department of Microbiology and Infectious Disease Center, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Yi Yuan
- Department of Microbiology and Infectious Disease Center, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Wei Xian
- Department of Microbiology and Infectious Disease Center, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Zhiheng Tang
- Department of Microbiology and Infectious Disease Center, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Jiaqi Fu
- Department of Respiratory Medicine, Infectious Diseases and Pathogen Biology Center, Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, State Key Laboratory for Zoonotic Diseases, The First Hospital of Jilin University, Changchun, China
| | - Xiaoyun Liu
- Department of Microbiology and Infectious Disease Center, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
- NHC Key Laboratory of Medical Immunology, Peking University, Beijing, China
| |
Collapse
|
3
|
Popov G, Fiebig-Comyn A, Syriste L, Little DJ, Skarina T, Stogios PJ, Birstonas S, Coombes BK, Savchenko A. Distinct Molecular Features of NleG Type 3 Secreted Effectors Allow for Different Roles during Citrobacter rodentium Infection in Mice. Infect Immun 2023; 91:e0050522. [PMID: 36511702 PMCID: PMC9872709 DOI: 10.1128/iai.00505-22] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 11/19/2022] [Indexed: 12/15/2022] Open
Abstract
The NleGs are the largest family of type 3 secreted effectors in attaching and effacing (A/E) pathogens, such as enterohemorrhagic Escherichia coli (EHEC), enteropathogenic E. coli, and Citrobacter rodentium. NleG effectors contain a conserved C-terminal U-box domain acting as a ubiquitin protein ligase and target host proteins via a variable N-terminal portion. The specific roles of these effectors during infection remain uncertain. Here, we demonstrate that the three NleG effectors-NleG1Cr, NleG7Cr, and NleG8Cr-encoded by C. rodentium DBS100 play distinct roles during infection in mice. Using individual nleGCr knockout strains, we show that NleG7Cr contributes to bacterial survival during enteric infection while NleG1Cr promotes the expression of diarrheal symptoms and NleG8Cr contributes to accelerated lethality in susceptible mice. Furthermore, the NleG8Cr effector contains a C-terminal PDZ domain binding motif that enables interaction with the host protein GOPC. Both the PDZ domain binding motif and the ability to engage with host ubiquitination machinery via the intact U-box domain proved to be necessary for NleG8Cr function, contributing to the observed phenotype during infection. We also establish that the PTZ binding motif in the EHEC NleG8 (NleG8Ec) effector, which shares 60% identity with NleG8Cr, is engaged in interactions with human GOPC. The crystal structure of the NleG8Ec C-terminal peptide in complex with the GOPC PDZ domain, determined to 1.85 Å, revealed a conserved interaction mode similar to that observed between GOPC and eukaryotic PDZ domain binding motifs. Despite these common features, nleG8Ec does not complement the ΔnleG8Cr phenotype during infection, revealing functional diversification between these NleG effectors.
Collapse
Affiliation(s)
- Georgy Popov
- Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Aline Fiebig-Comyn
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| | - Lukas Syriste
- Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Dustin J. Little
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, Ontario, Canada
| | - Tatiana Skarina
- Department of Chemical Engineering and Applied Chemistry, Toronto University, Toronto, Ontario, Canada
| | - Peter J. Stogios
- Department of Chemical Engineering and Applied Chemistry, Toronto University, Toronto, Ontario, Canada
| | - Sarah Birstonas
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, Ontario, Canada
| | - Brian K. Coombes
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| | - Alexei Savchenko
- Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, Alberta, Canada
- Department of Chemical Engineering and Applied Chemistry, Toronto University, Toronto, Ontario, Canada
| |
Collapse
|
4
|
Abstract
PURPOSE OF REVIEW This review updates recent findings about Escherichia coli O157:H7 virulence factors and its bovine reservoir. This Shiga toxin (Stx)-producing E. coli belongs to the Enterohemorrhagic E. coli (EHEC) pathotype causing hemorrhagic colitis. Its low infectious dose makes it an efficient, severe, foodborne pathogen. Although EHEC remains in the intestine, Stx can translocate systemically and is cytotoxic to microvascular endothelial cells, especially in the kidney and brain. Disease can progress to life-threatening hemolytic uremic syndrome (HUS) with hemolytic anemia, acute kidney failure, and thrombocytopenia. Young children, the immunocompromised, and the elderly are at the highest risk for HUS. Healthy ruminants are the major reservoir of EHEC and cattle are the primary source of human exposure. RECENT FINDINGS Advances in understanding E. coli O157:H7 pathogenesis include molecular mechanisms of virulence, bacterial adherence, type three secretion effectors, intestinal microbiome, inflammation, and reservoir maintenance. SUMMARY Many aspects of E. coli O157:H7 disease remain unclear and include the role of the human and bovine intestinal microbiomes in infection. Therapeutic strategies involve controlling inflammatory responses and/or intestinal barrier function. Finally, elimination/reduction of E. coli O157:H7 in cattle using CRISPR-engineered conjugative bacterial plasmids and/or on-farm management likely hold solutions to reduce infections and increase food safety/security.
Collapse
|
5
|
Sanchez-Garrido J, Ruano-Gallego D, Choudhary JS, Frankel G. The type III secretion system effector network hypothesis. Trends Microbiol 2022; 30:524-533. [PMID: 34840074 DOI: 10.1016/j.tim.2021.10.007] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 10/26/2021] [Accepted: 10/28/2021] [Indexed: 11/18/2022]
Abstract
Type III secretion system (T3SS) effectors are key virulence factors that underpin the infection strategy of many clinically important Gram-negative pathogens, including Salmonella enterica, Shigella spp., enteropathogenic and enterohemorrhagic Escherichia coli and their murine equivalent, Citrobacter rodentium. The cellular processes or proteins targeted by the effectors can be common to multiple pathogens or pathogen-specific. The main approach to understanding T3SS-mediated pathogenesis has been to determine the contribution of one effector at a time, with the aim of piecing together individual functions and unveiling infection mechanisms. However, in contrast to this prevailing approach, simultaneous deletion of multiple effectors revealed that they function as an interconnected network in vivo, uncovering effector codependency and context-dependent effector essentiality. This paradigm shift in T3SS biology is at the heart of this opinion article.
Collapse
Affiliation(s)
- Julia Sanchez-Garrido
- Centre for Molecular Microbiology and Infection, Department of Life Sciences, Imperial College, London, UK.
| | - David Ruano-Gallego
- Department of Molecular Evolution, Centro de Astrobiología, Instituto Nacional de Técnica Aeroespacial-Consejo Superior de Investigaciones Científicas (INTA-CSIC), Madrid, Spain.
| | - Jyoti S Choudhary
- Functional Proteomics Group, Chester Beatty Laboratories, Institute of Cancer Research, London, UK
| | - Gad Frankel
- Centre for Molecular Microbiology and Infection, Department of Life Sciences, Imperial College, London, UK
| |
Collapse
|
6
|
Jiang L, Yang W, Jiang X, Yao T, Wang L, Yang B. Virulence-related O islands in enterohemorrhagic Escherichia coli O157:H7. Gut Microbes 2022; 13:1992237. [PMID: 34711138 PMCID: PMC8565820 DOI: 10.1080/19490976.2021.1992237] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Enterohemorrhagic Escherichia coli (EHEC) O157:H7 is a principally foodborne pathogen linked to serious diseases, including bloody diarrhea, hemorrhagic colitis, and hemolytic uremic syndrome. Comparative genomics analysis revealed that EHEC O157 contains 177 unique genomic islands, termed O islands, compared with the nonpathogenic E. coli K-12 laboratory strain. These O islands contribute largely to the pathogenicity of EHEC O157:H7 by providing numerous virulence factors, effectors, virulence regulatory proteins, and virulence regulatory sRNAs. The present review aimed to provide a comprehensive understanding of the research progress on the function of O islands, especially focusing on virulence-related O islands.
Collapse
Affiliation(s)
- Lingyan Jiang
- TEDA Institute of Biological Sciences and Biotechnology, Nankai University, TEDA, Tianjin, P. R. China
| | - Wen Yang
- TEDA Institute of Biological Sciences and Biotechnology, Nankai University, TEDA, Tianjin, P. R. China
| | - Xinlei Jiang
- School of Environmental and Municipal Engineering, Tianjin Chengjian University, Tianjin, P. R. China
| | - Ting Yao
- TEDA Institute of Biological Sciences and Biotechnology, Nankai University, TEDA, Tianjin, P. R. China
| | - Lu Wang
- TEDA Institute of Biological Sciences and Biotechnology, Nankai University, TEDA, Tianjin, P. R. China
| | - Bin Yang
- TEDA Institute of Biological Sciences and Biotechnology, Nankai University, TEDA, Tianjin, P. R. China,CONTACT Bin Yang TEDA Institute of Biological Sciences and Biotechnology, Nankai University, TEDA, Tianjin300457, P. R. China
| |
Collapse
|
7
|
Type III secretion system effector subnetworks elicit distinct host immune responses to infection. Curr Opin Microbiol 2021; 64:19-26. [PMID: 34537517 DOI: 10.1016/j.mib.2021.08.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 08/26/2021] [Accepted: 08/27/2021] [Indexed: 01/18/2023]
Abstract
Citrobacter rodentium, a natural mouse pathogen which colonises the colon of immuno-competent mice, provides a robust model for interrogating host-pathogen-microbiota interactions in vivo. This model has been key to providing new insights into local host responses to enteric infection, including changes in intestinal epithelial cell immunometabolism and mucosal immunity. C. rodentium injects 31 bacterial effectors into epithelial cells via a type III secretion system (T3SS). Recently, these effectors were shown to be able to form multiple intracellular subnetworks which can withstand significant contractions whilst maintaining virulence. Here we highlight recent advances in understanding gut mucosal responses to infection and effector biology, as well as potential uses for artificial intelligence (AI) in understanding infectious disease and speculate on the role of T3SS effector networks in host adaption.
Collapse
|
8
|
Mikolčević P, Hloušek-Kasun A, Ahel I, Mikoč A. ADP-ribosylation systems in bacteria and viruses. Comput Struct Biotechnol J 2021; 19:2366-2383. [PMID: 34025930 PMCID: PMC8120803 DOI: 10.1016/j.csbj.2021.04.023] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 04/07/2021] [Accepted: 04/07/2021] [Indexed: 12/30/2022] Open
Abstract
ADP-ribosylation is an ancient posttranslational modification present in all kingdoms of life. The system likely originated in bacteria where it functions in inter- and intra-species conflict, stress response and pathogenicity. It was repeatedly adopted via lateral transfer by eukaryotes, including humans, where it has a pivotal role in epigenetics, DNA-damage repair, apoptosis, and other crucial pathways including the immune response to pathogenic bacteria and viruses. In other words, the same ammunition used by pathogens is adapted by eukaryotes to fight back. While we know quite a lot about the eukaryotic system, expanding rather patchy knowledge on bacterial and viral ADP-ribosylation would give us not only a better understanding of the system as a whole but a fighting advantage in this constant arms race. By writing this review we hope to put into focus the available information and give a perspective on how this system works and can be exploited in the search for therapeutic targets in the future. The relevance of the subject is especially highlighted by the current situation of being amid the world pandemic caused by a virus harbouring and dependent on a representative of such a system.
Collapse
Affiliation(s)
- Petra Mikolčević
- Division of Molecular Biology, Ruđer Bošković Institute, Zagreb, Croatia
| | | | - Ivan Ahel
- Sir William Dunn School of Pathology, University of Oxford, UK
| | - Andreja Mikoč
- Division of Molecular Biology, Ruđer Bošković Institute, Zagreb, Croatia
| |
Collapse
|
9
|
Ruano-Gallego D, Sanchez-Garrido J, Kozik Z, Núñez-Berrueco E, Cepeda-Molero M, Mullineaux-Sanders C, Naemi Baghshomali Y, Slater SL, Wagner N, Glegola-Madejska I, Roumeliotis TI, Pupko T, Fernández LÁ, Rodríguez-Patón A, Choudhary JS, Frankel G. Type III secretion system effectors form robust and flexible intracellular virulence networks. Science 2021; 371:eabc9531. [PMID: 33707240 DOI: 10.1126/science.abc9531] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 12/15/2020] [Accepted: 01/15/2021] [Indexed: 12/14/2022]
Abstract
Infections with many Gram-negative pathogens, including Escherichia coli, Salmonella, Shigella, and Yersinia, rely on type III secretion system (T3SS) effectors. We hypothesized that while hijacking processes within mammalian cells, the effectors operate as a robust network that can tolerate substantial contractions. This was tested in vivo using the mouse pathogen Citrobacter rodentium (encoding 31 effectors). Sequential gene deletions showed that effector essentiality for infection was context dependent and that the network could tolerate 60% contraction while maintaining pathogenicity. Despite inducing very different colonic cytokine profiles (e.g., interleukin-22, interleukin-17, interferon-γ, or granulocyte-macrophage colony-stimulating factor), different networks induced protective immunity. Using data from >100 distinct mutant combinations, we built and trained a machine learning model able to predict colonization outcomes, which were confirmed experimentally. Furthermore, reproducing the human-restricted enteropathogenic E. coli effector repertoire in C. rodentium was not sufficient for efficient colonization, which implicates effector networks in host adaptation. These results unveil the extreme robustness of both T3SS effector networks and host responses.
Collapse
Affiliation(s)
- David Ruano-Gallego
- Centre for Molecular Microbiology and Infection, Department of Life Sciences, Imperial College, London, UK
| | - Julia Sanchez-Garrido
- Centre for Molecular Microbiology and Infection, Department of Life Sciences, Imperial College, London, UK
| | - Zuzanna Kozik
- Functional Proteomics Group, Chester Beatty Laboratories, Institute of Cancer Research, London, UK
| | - Elena Núñez-Berrueco
- Laboratorio de Inteligencia Artificial, Departamento de Inteligencia Artificial, Universidad Politécnica de Madrid, Campus de Montegancedo, Boadilla del Monte, Madrid, Spain
| | - Massiel Cepeda-Molero
- Centre for Molecular Microbiology and Infection, Department of Life Sciences, Imperial College, London, UK
| | | | - Yasaman Naemi Baghshomali
- Centre for Molecular Microbiology and Infection, Department of Life Sciences, Imperial College, London, UK
| | - Sabrina L Slater
- Centre for Molecular Microbiology and Infection, Department of Life Sciences, Imperial College, London, UK
| | - Naama Wagner
- The Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Izabela Glegola-Madejska
- Centre for Molecular Microbiology and Infection, Department of Life Sciences, Imperial College, London, UK
| | - Theodoros I Roumeliotis
- Functional Proteomics Group, Chester Beatty Laboratories, Institute of Cancer Research, London, UK
| | - Tal Pupko
- The Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Luis Ángel Fernández
- Centro Nacional de Biotecnología (CNB-CSIC), Department of Microbial Biotechnology, Madrid, Spain
| | - Alfonso Rodríguez-Patón
- Laboratorio de Inteligencia Artificial, Departamento de Inteligencia Artificial, Universidad Politécnica de Madrid, Campus de Montegancedo, Boadilla del Monte, Madrid, Spain
| | - Jyoti S Choudhary
- Functional Proteomics Group, Chester Beatty Laboratories, Institute of Cancer Research, London, UK.
| | - Gad Frankel
- Centre for Molecular Microbiology and Infection, Department of Life Sciences, Imperial College, London, UK.
| |
Collapse
|
10
|
Dhanda AS, Yang D, Kooner A, Guttman JA. Distribution of PDLIM1 at actin-rich structures generated by invasive and adherent bacterial pathogens. Anat Rec (Hoboken) 2020; 304:919-938. [PMID: 33022122 DOI: 10.1002/ar.24523] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 07/06/2020] [Accepted: 07/28/2020] [Indexed: 12/15/2022]
Abstract
The enteric bacterial pathogens Listeria monocytogenes (Listeria) and enteropathogenic Escherichia coli (EPEC) remodel the eukaryotic actin cytoskeleton during their disease processes. Listeria generate slender actin-rich comet/rocket tails to move intracellularly, and later, finger-like membrane protrusions to spread amongst host cells. EPEC remain extracellular, but generate similar actin-rich membranous protrusions (termed pedestals) to move atop the host epithelia. These structures are crucial for disease as diarrheal (and systemic) infections are significantly abrogated during infections with mutant strains that are unable to generate the structures. The current repertoire of host components enriched within these structures is vast and diverse. In this protein catalog, we and others have found that host actin crosslinkers, such as palladin and α-actinin-1, are routinely exploited. To expand on this list, we set out to investigate the distribution of PDLIM1, a scaffolding protein and binding partner of palladin and α-actinin-1, during bacterial infections. We show that PDLIM1 localizes to the site of initial Listeria entry into cells. Following this, PDLIM1 localizes to actin filament clouds surrounding immotile bacteria, and then colocalizes with actin once the comet/rocket tails are generated. Unlike palladin or α-actinin-1, PDLIM1 is maintained within the actin-rich core of membrane protrusions. Conversely, α-actinin-1, but not PDLIM1 (or palladin), is enriched at the membrane invagination that internalizes the Listeria-containing membrane protrusion. We also show that PDLIM1 is a component of the EPEC pedestal core and that its recruitment is dependent on the bacterial effector Tir. Our findings highlight PDLIM1 as another protein present within pathogen-induced actin-rich structures.
Collapse
Affiliation(s)
- Aaron S Dhanda
- Department of Biological Sciences, Centre for Cell Biology, Development, and Disease, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Diana Yang
- Department of Biological Sciences, Centre for Cell Biology, Development, and Disease, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Avneen Kooner
- Department of Biological Sciences, Centre for Cell Biology, Development, and Disease, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Julian A Guttman
- Department of Biological Sciences, Centre for Cell Biology, Development, and Disease, Simon Fraser University, Burnaby, British Columbia, Canada
| |
Collapse
|
11
|
Goddard PJ, Sanchez-Garrido J, Slater SL, Kalyan M, Ruano-Gallego D, Marchès O, Fernández LÁ, Frankel G, Shenoy AR. Enteropathogenic Escherichia coli Stimulates Effector-Driven Rapid Caspase-4 Activation in Human Macrophages. Cell Rep 2020; 27:1008-1017.e6. [PMID: 31018119 PMCID: PMC6486487 DOI: 10.1016/j.celrep.2019.03.100] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 02/20/2019] [Accepted: 03/27/2019] [Indexed: 12/20/2022] Open
Abstract
Microbial infections can stimulate the assembly of inflammasomes, which activate caspase-1. The gastrointestinal pathogen enteropathogenic Escherichia coli (EPEC) causes localized actin polymerization in host cells. Actin polymerization requires the binding of the bacterial adhesin intimin to Tir, which is delivered to host cells via a type 3 secretion system (T3SS). We show that EPEC induces T3SS-dependent rapid non-canonical NLRP3 inflammasome activation in human macrophages. Notably, caspase-4 activation by EPEC triggers pyroptosis and cytokine processing through the NLRP3-caspase-1 inflammasome. Mechanistically, caspase-4 activation requires the detection of LPS and EPEC-induced actin polymerization, either via Tir tyrosine phosphorylation and the phosphotyrosine-binding adaptor NCK or Tir and the NCK-mimicking effector TccP. An engineered E. coli K12 could reconstitute Tir-intimin signaling, which is necessary and sufficient for inflammasome activation, ruling out the involvement of other virulence factors. Our studies reveal a crosstalk between caspase-4 and caspase-1 that is cooperatively stimulated by LPS and effector-driven actin polymerization. EPEC bacteria expressing virulence genes induce rapid human macrophage pyroptosis Bacterial LPS sensing by caspase-4 activates NLRP3-caspase-1 inflammasomes Actin polymerization driven by Tir-intimin signaling promotes pyroptosis Caspase-1 mediates cytokine processing and gasdermin D cleavage, leading to pyroptosis
Collapse
Affiliation(s)
- Philippa J Goddard
- Department of Life Sciences, Medical Research Council Centre for Molecular Bacteriology & Infection, Imperial College London, London, UK; Department of Medicine, Medical Research Council Centre for Molecular Bacteriology & Infection, Imperial College London, London, UK
| | - Julia Sanchez-Garrido
- Department of Medicine, Medical Research Council Centre for Molecular Bacteriology & Infection, Imperial College London, London, UK
| | - Sabrina L Slater
- Department of Life Sciences, Medical Research Council Centre for Molecular Bacteriology & Infection, Imperial College London, London, UK
| | - Mohini Kalyan
- Department of Medicine, Medical Research Council Centre for Molecular Bacteriology & Infection, Imperial College London, London, UK
| | - David Ruano-Gallego
- Department of Microbial Biotechnology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Olivier Marchès
- Department of Life Sciences, Medical Research Council Centre for Molecular Bacteriology & Infection, Imperial College London, London, UK
| | - Luis Ángel Fernández
- Department of Microbial Biotechnology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Gad Frankel
- Department of Life Sciences, Medical Research Council Centre for Molecular Bacteriology & Infection, Imperial College London, London, UK
| | - Avinash R Shenoy
- Department of Medicine, Medical Research Council Centre for Molecular Bacteriology & Infection, Imperial College London, London, UK.
| |
Collapse
|
12
|
Sanchez‐Garrido J, Slater SL, Clements A, Shenoy AR, Frankel G. Vying for the control of inflammasomes: The cytosolic frontier of enteric bacterial pathogen-host interactions. Cell Microbiol 2020; 22:e13184. [PMID: 32185892 PMCID: PMC7154749 DOI: 10.1111/cmi.13184] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 01/13/2020] [Accepted: 01/30/2020] [Indexed: 12/13/2022]
Abstract
Enteric pathogen-host interactions occur at multiple interfaces, including the intestinal epithelium and deeper organs of the immune system. Microbial ligands and activities are detected by host sensors that elicit a range of immune responses. Membrane-bound toll-like receptors and cytosolic inflammasome pathways are key signal transducers that trigger the production of pro-inflammatory molecules, such as cytokines and chemokines, and regulate cell death in response to infection. In recent years, the inflammasomes have emerged as a key frontier in the tussle between bacterial pathogens and the host. Inflammasomes are complexes that activate caspase-1 and are regulated by related caspases, such as caspase-11, -4, -5 and -8. Importantly, enteric bacterial pathogens can actively engage or evade inflammasome signalling systems. Extracellular, vacuolar and cytosolic bacteria have developed divergent strategies to subvert inflammasomes. While some pathogens take advantage of inflammasome activation (e.g. Listeria monocytogenes, Helicobacter pylori), others (e.g. E. coli, Salmonella, Shigella, Yersinia sp.) deploy a range of virulence factors, mainly type 3 secretion system effectors, that subvert or inhibit inflammasomes. In this review we focus on inflammasome pathways and their immune functions, and discuss how enteric bacterial pathogens interact with them. These studies have not only shed light on inflammasome-mediated immunity, but also the exciting area of mammalian cytosolic immune surveillance.
Collapse
Affiliation(s)
| | | | | | - Avinash R. Shenoy
- Department of Infectious Disease, MRC Centre for Molecular Bacteriology & InfectionImperial College LondonLondonUK
| | - Gad Frankel
- Department of Life SciencesImperial College LondonLondonUK
| |
Collapse
|
13
|
Jégouzo SAF, Nelson C, Hardwick T, Wong STA, Lau NKK, Neoh GKE, Castellanos-Rueda R, Huang Z, Mignot B, Hirdaramani A, Howitt A, Frewin K, Shen Z, Fox RJ, Wong R, Ando M, Emony L, Zhu H, Holder A, Werling D, Krishnan N, Robertson BD, Clements A, Taylor ME, Drickamer K. Mammalian lectin arrays for screening host-microbe interactions. J Biol Chem 2020; 295:4541-4555. [PMID: 32094229 PMCID: PMC7135977 DOI: 10.1074/jbc.ra120.012783] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 02/14/2020] [Indexed: 12/22/2022] Open
Abstract
Many members of the C-type lectin family of glycan-binding receptors have been ascribed roles in the recognition of microorganisms and serve as key receptors in the innate immune response to pathogens. Other mammalian receptors have become targets through which pathogens enter target cells. These receptor roles have often been documented with binding studies involving individual pairs of receptors and microorganisms. To provide a systematic overview of interactions between microbes and the large complement of C-type lectins, here we developed a lectin array and suitable protocols for labeling of microbes that could be used to probe this array. The array contains C-type lectins from cow, chosen as a model organism of agricultural interest for which the relevant pathogen–receptor interactions have not been previously investigated in detail. Screening with yeast cells and various strains of both Gram-positive and -negative bacteria revealed distinct binding patterns, which in some cases could be explained by binding to lipopolysaccharides or capsular polysaccharides, but in other cases they suggested the presence of novel glycan targets on many of the microorganisms. These results are consistent with interactions previously ascribed to the receptors, but they also highlight binding to additional sugar targets that have not previously been recognized. Our findings indicate that mammalian lectin arrays represent unique discovery tools for identifying both novel ligands and new receptor functions.
Collapse
Affiliation(s)
- Sabine A F Jégouzo
- Department of Life Sciences, Imperial College London, London SW7 2AZ, United Kingdom
| | - Conor Nelson
- Department of Life Sciences, Imperial College London, London SW7 2AZ, United Kingdom
| | - Thomas Hardwick
- Department of Life Sciences, Imperial College London, London SW7 2AZ, United Kingdom
| | - S T Angel Wong
- Department of Life Sciences, Imperial College London, London SW7 2AZ, United Kingdom
| | - Noel Kuan Kiat Lau
- Department of Life Sciences, Imperial College London, London SW7 2AZ, United Kingdom
| | - Gaik Kin Emily Neoh
- Department of Life Sciences, Imperial College London, London SW7 2AZ, United Kingdom
| | | | - Zhiyao Huang
- Department of Life Sciences, Imperial College London, London SW7 2AZ, United Kingdom
| | - Benjamin Mignot
- Department of Life Sciences, Imperial College London, London SW7 2AZ, United Kingdom
| | - Aanya Hirdaramani
- Department of Life Sciences, Imperial College London, London SW7 2AZ, United Kingdom
| | - Annie Howitt
- Department of Life Sciences, Imperial College London, London SW7 2AZ, United Kingdom
| | - Kathryn Frewin
- Department of Life Sciences, Imperial College London, London SW7 2AZ, United Kingdom
| | - Zheng Shen
- Department of Life Sciences, Imperial College London, London SW7 2AZ, United Kingdom
| | - Rhys J Fox
- Department of Life Sciences, Imperial College London, London SW7 2AZ, United Kingdom
| | - Rachel Wong
- Department of Life Sciences, Imperial College London, London SW7 2AZ, United Kingdom
| | - Momoko Ando
- Department of Life Sciences, Imperial College London, London SW7 2AZ, United Kingdom
| | - Lauren Emony
- Department of Life Sciences, Imperial College London, London SW7 2AZ, United Kingdom
| | - Henderson Zhu
- Department of Life Sciences, Imperial College London, London SW7 2AZ, United Kingdom
| | - Angela Holder
- Department of Pathobiology and Population Sciences, Royal Veterinary College, North Mymms, Hatfield, Hertfordshire AL9 7TA, United Kingdom
| | - Dirk Werling
- Department of Pathobiology and Population Sciences, Royal Veterinary College, North Mymms, Hatfield, Hertfordshire AL9 7TA, United Kingdom
| | - Nitya Krishnan
- Department of Infectious Disease and MRC Centre for Molecular Bacteriology and Infection, Imperial College London, London SW7 2AZ, United Kingdom
| | - Brian D Robertson
- Department of Infectious Disease and MRC Centre for Molecular Bacteriology and Infection, Imperial College London, London SW7 2AZ, United Kingdom
| | - Abigail Clements
- Department of Life Sciences and MRC Centre for Molecular Bacteriology and Infection, Imperial College London, London SW7 2AZ, United Kingdom
| | - Maureen E Taylor
- Department of Life Sciences, Imperial College London, London SW7 2AZ, United Kingdom
| | - Kurt Drickamer
- Department of Life Sciences, Imperial College London, London SW7 2AZ, United Kingdom
| |
Collapse
|
14
|
Enteropathogenic Escherichia coli (EPEC) Recruitment of PAR Polarity Protein Atypical PKCζ to Pedestals and Cell-Cell Contacts Precedes Disruption of Tight Junctions in Intestinal Epithelial Cells. Int J Mol Sci 2020; 21:ijms21020527. [PMID: 31947656 PMCID: PMC7014222 DOI: 10.3390/ijms21020527] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 01/09/2020] [Accepted: 01/10/2020] [Indexed: 12/26/2022] Open
Abstract
Enteropathogenic Escherichia coli (EPEC) uses a type three secretion system to inject effector proteins into host intestinal epithelial cells, causing diarrhea. EPEC induces the formation of pedestals underlying attached bacteria, disrupts tight junction (TJ) structure and function, and alters apico-basal polarity by redistributing the polarity proteins Crb3 and Pals1, although the mechanisms are unknown. Here we investigate the temporal relationship of PAR polarity complex and TJ disruption following EPEC infection. EPEC recruits active aPKCζ, a PAR polarity protein, to actin within pedestals and at the plasma membrane prior to disrupting TJ. The EPEC effector EspF binds the endocytic protein sorting nexin 9 (SNX9). This interaction impacts actin pedestal organization, recruitment of active aPKCζ to actin at cell–cell borders, endocytosis of JAM-A S285 and occludin, and TJ barrier function. Collectively, data presented herein support the hypothesis that EPEC-induced perturbation of TJ is a downstream effect of disruption of the PAR complex and that EspF binding to SNX9 contributes to this phenotype. aPKCζ phosphorylates polarity and TJ proteins and participates in actin dynamics. Therefore, the early recruitment of aPKCζ to EPEC pedestals and increased interaction with actin at the membrane may destabilize polarity complexes ultimately resulting in perturbation of TJ.
Collapse
|
15
|
Yáñez JM, Yoshida GM, Parra Á, Correa K, Barría A, Bassini LN, Christensen KA, López ME, Carvalheiro R, Lhorente JP, Pulgar R. Comparative Genomic Analysis of Three Salmonid Species Identifies Functional Candidate Genes Involved in Resistance to the Intracellular Bacterium Piscirickettsia salmonis. Front Genet 2019; 10:665. [PMID: 31428125 PMCID: PMC6690157 DOI: 10.3389/fgene.2019.00665] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2019] [Accepted: 06/25/2019] [Indexed: 12/23/2022] Open
Abstract
Piscirickettsia salmonis is the etiologic agent of salmon rickettsial syndrome (SRS) and is responsible for considerable economic losses in salmon aquaculture. The bacterium affects coho salmon (CS; Oncorhynchus kisutch), Atlantic salmon (AS; Salmo salar), and rainbow trout (RT; Oncorhynchus mykiss) in several countries, including Norway, Canada, Scotland, Ireland, and Chile. We used Bayesian genome-wide association study analyses to investigate the genetic architecture of resistance to P. salmonis in farmed populations of these species. Resistance to SRS was defined as the number of days to death and as binary survival (BS). A total of 828 CS, 2130 RT, and 2601 AS individuals were phenotyped and then genotyped using double-digest restriction site-associated DNA sequencing and 57K and 50K Affymetrix® Axiom® single nucleotide polymorphism (SNP) panels, respectively. Both traits of SRS resistance in CS and RT appeared to be under oligogenic control. In AS, there was evidence of polygenic control of SRS resistance. To identify candidate genes associated with resistance, we applied a comparative genomics approach in which we systematically explored the complete set of genes adjacent to SNPs, which explained more than 1% of the genetic variance of resistance in each salmonid species (533 genes in total). Thus, genes were classified based on the following criteria: i) shared function of their protein domains among species, ii) shared orthology among species, iii) proximity to the SNP explaining the highest proportion of the genetic variance, and iv) presence in more than one genomic region explaining more than 1% of the genetic variance within species. Our results allowed us to identify 120 candidate genes belonging to at least one of the four criteria described above. Of these, 21 of them were part of at least two of the criteria defined above and are suggested to be strong functional candidates influencing P. salmonis resistance. These genes are related to diverse biological processes, such as kinase activity, GTP hydrolysis, helicase activity, lipid metabolism, cytoskeletal dynamics, inflammation, and innate immune response, which seem essential in the host response against P. salmonis infection. These results provide fundamental knowledge on the potential functional genes underpinning resistance against P. salmonis in three salmonid species.
Collapse
Affiliation(s)
- José M. Yáñez
- Facultad de Ciencias Veterinarias y Pecuarias, Universidad de Chile, Santiago, Chile
- Núcleo Milenio INVASAL, Concepción, Chile
| | - Grazyella M. Yoshida
- Facultad de Ciencias Veterinarias y Pecuarias, Universidad de Chile, Santiago, Chile
| | - Ángel Parra
- Facultad de Ciencias Veterinarias y Pecuarias, Universidad de Chile, Santiago, Chile
- Instituto de Nutrición y Tecnología de los Alimentos, Universidad de Chile, Santiago, Chile
- Doctorado en Acuicultura. Programa Cooperativo Universidad de Chile, Universidad Católica del Norte, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
- Facultad de Ciencias del Mar, Universidad Católica del Norte, Coquimbo, Chile
| | | | - Agustín Barría
- Facultad de Ciencias Veterinarias y Pecuarias, Universidad de Chile, Santiago, Chile
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh Easter Bush, Midlothian, United Kingdom
| | - Liane N. Bassini
- Escuela de Medicina Veterinaria, Facultad de Ciencias de la Vida, Universidad Andres Bello, Santiago, Chile
| | | | - Maria E. López
- Facultad de Ciencias Veterinarias y Pecuarias, Universidad de Chile, Santiago, Chile
- Department of Animal Breeding and Genetics, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Roberto Carvalheiro
- School of Agricultural and Veterinarian Sciences, São Paulo State University (Unesp), Jaboticabal, Brazil
- National Council for Scientific and Technological Development (CNPq), Brasília, Brazil
| | | | - Rodrigo Pulgar
- Instituto de Nutrición y Tecnología de los Alimentos, Universidad de Chile, Santiago, Chile
| |
Collapse
|
16
|
Cheng RA, Wiedmann M. The ADP-Ribosylating Toxins of Salmonella. Toxins (Basel) 2019; 11:E416. [PMID: 31315299 PMCID: PMC6669713 DOI: 10.3390/toxins11070416] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 07/10/2019] [Accepted: 07/10/2019] [Indexed: 12/22/2022] Open
Abstract
A number of pathogenic bacteria utilize toxins to mediate disease in a susceptible host. The foodborne pathogen Salmonella is one of the most important and well-studied bacterial pathogens. Recently, whole genome sequence characterizations revealed the presence of multiple novel ADP-ribosylating toxins encoded by a variety of Salmonella serovars. In this review, we discuss both the classical (SpvB) and novel (typhoid toxin, ArtAB, and SboC/SeoC) ADP-ribosylating toxins of Salmonella, including the structure and function of these toxins and our current understanding of their contributions to virulence.
Collapse
Affiliation(s)
- Rachel A Cheng
- Department of Food Science, Cornell University, Ithaca, NY 14853, USA.
| | - Martin Wiedmann
- Department of Food Science, Cornell University, Ithaca, NY 14853, USA
| |
Collapse
|
17
|
Abstract
ADP-ribosylation (ADPr) is an ancient reversible modification of cellular macromolecules controlling major biological processes as diverse as DNA damage repair, transcriptional regulation, intracellular transport, immune and stress responses, cell survival and proliferation. Furthermore, enzymatic reactions of ADPr are central in the pathogenesis of many human diseases, including infectious conditions. By providing a review of ADPr signalling in bacterial systems, we highlight the relevance of this chemical modification in the pathogenesis of human diseases depending on host-pathogen interactions. The post-antibiotic era has raised the need to find alternative approaches to antibiotic administration, as major pathogens becoming resistant to antibiotics. An in-depth understanding of ADPr reactions provides the rationale for designing novel antimicrobial strategies for treatment of infectious diseases. In addition, the understanding of mechanisms of ADPr by bacterial virulence factors offers important hints to improve our knowledge on cellular processes regulated by eukaryotic homologous enzymes, which are often involved in the pathogenesis of human diseases.
Collapse
|
18
|
Dudkiewicz M, Pawłowski K. A novel conserved family of Macro-like domains-putative new players in ADP-ribosylation signaling. PeerJ 2019; 7:e6863. [PMID: 31106069 PMCID: PMC6500376 DOI: 10.7717/peerj.6863] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 03/28/2019] [Indexed: 12/30/2022] Open
Abstract
The presence of many completely uncharacterized proteins, even in well-studied organisms such as humans, seriously hampers a full understanding of the functioning of living cells. One such example is the human protein C12ORF4, which belongs to the DUF2362 family, present in many eukaryotic lineages and conserved in metazoans. The only functional information available on C12ORF4 (Chromosome 12 Open Reading Frame 4) is its involvement in mast cell degranulation and its being a genetic cause of autosomal intellectual disability. Bioinformatics analysis of the DUF2362 family provides strong evidence that it is a novel member of the Macro clan/superfamily. Sequence similarity analysis versus other representatives of the Macro superfamily of ADP-ribose-binding proteins and mapping sequence conservation on predicted three-dimensional structure provides hypotheses regarding the molecular function for members of the DUF2362 family. For example, the available functional data suggest a possible role for C12ORF4 in ADP-ribosylation signaling in asthma and related inflammatory diseases. This novel family appears to be a likely novel ADP-ribosylation “reader” and “eraser,” a previously unnoticed putative new player in cell signaling by this emerging post-translational modification.
Collapse
Affiliation(s)
- Małgorzata Dudkiewicz
- Department of Experimental Design and Bioinformatics, Faculty of Agriculture and Biology, Warsaw University of Life Sciences, Warszawa, Poland
| | - Krzysztof Pawłowski
- Department of Experimental Design and Bioinformatics, Faculty of Agriculture and Biology, Warsaw University of Life Sciences, Warszawa, Poland.,Department of Translational Mecicine, Clinical Sciences, Lund University, Lund, Sweden
| |
Collapse
|
19
|
Velle KB, Campellone KG. Enteropathogenic E. coli relies on collaboration between the formin mDia1 and the Arp2/3 complex for actin pedestal biogenesis and maintenance. PLoS Pathog 2018; 14:e1007485. [PMID: 30550556 PMCID: PMC6310289 DOI: 10.1371/journal.ppat.1007485] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 12/28/2018] [Accepted: 11/23/2018] [Indexed: 12/16/2022] Open
Abstract
Enteropathogenic and enterohemorrhagic E. coli (EPEC and EHEC) are closely related extracellular pathogens that reorganize host cell actin into “pedestals” beneath the tightly adherent bacteria. This pedestal-forming activity is both a critical step in pathogenesis, and it makes EPEC and EHEC useful models for studying the actin rearrangements that underlie membrane protrusions. To generate pedestals, EPEC relies on the tyrosine phosphorylated bacterial effector protein Tir to bind host adaptor proteins that recruit N-WASP, a nucleation-promoting factor that activates the Arp2/3 complex to drive actin polymerization. In contrast, EHEC depends on the effector EspFU to multimerize N-WASP and promote Arp2/3 activation. Although these core pathways of pedestal assembly are well-characterized, the contributions of additional actin nucleation factors are unknown. We investigated potential cooperation between the Arp2/3 complex and other classes of nucleators using chemical inhibitors, siRNAs, and knockout cell lines. We found that inhibition of formins impairs actin pedestal assembly, motility, and cellular colonization for bacteria using the EPEC, but not the EHEC, pathway of actin polymerization. We also identified mDia1 as the formin contributing to EPEC pedestal assembly, as its expression level positively correlates with the efficiency of pedestal formation, and it localizes to the base of pedestals both during their initiation and once they have reached steady state. Collectively, our data suggest that mDia1 enhances EPEC pedestal biogenesis and maintenance by generating seed filaments to be used by the N-WASP-Arp2/3-dependent actin nucleation machinery and by sustaining Src-mediated phosphorylation of Tir. Microbial pathogens that rearrange the host actin cytoskeleton have made valuable contributions to our understanding of cell signaling and movement. The assembly and organization of the actin cytoskeleton is driven by proteins called nucleators, which can be manipulated by bacteria including enteropathogenic Escherichia coli (EPEC), a frequent cause of pediatric diarrhea in developing countries. After ingestion, EPEC adhere tightly to cells of the intestine and hijack the underlying cytoskeleton to create protrusions called actin pedestals. While mechanisms of pedestal assembly involving a nucleator called the Arp2/3 complex have been defined for EPEC, the contribution of additional host nucleators has not been determined. We assessed the roles of several actin nucleators in EPEC pedestals and found that in addition to Arp2/3 complex-mediated nucleation, the formin mDia1 is a key contributor to actin assembly. These findings highlight the importance of nucleator collaboration in pathogenesis, and also advance our understanding of the molecular and cellular basis of EPEC infection, which is ultimately important for the discovery of new drug targets.
Collapse
Affiliation(s)
- Katrina B. Velle
- Department of Molecular and Cell Biology, Institute for Systems Genomics, University of Connecticut, Storrs, Connecticut, United States of America
| | - Kenneth G. Campellone
- Department of Molecular and Cell Biology, Institute for Systems Genomics, University of Connecticut, Storrs, Connecticut, United States of America
- * E-mail:
| |
Collapse
|
20
|
The Pseudomonas aeruginosa T6SS-VgrG1b spike is topped by a PAAR protein eliciting DNA damage to bacterial competitors. Proc Natl Acad Sci U S A 2018; 115:12519-12524. [PMID: 30455305 DOI: 10.1073/pnas.1814181115] [Citation(s) in RCA: 99] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The type VI secretion system (T6SS) is a supramolecular complex involved in the delivery of potent toxins during bacterial competition. Pseudomonas aeruginosa possesses three T6SS gene clusters and several hcp and vgrG gene islands, the latter encoding the spike at the T6SS tip. The vgrG1b cluster encompasses seven genes whose organization and sequences are highly conserved in P. aeruginosa genomes, except for two genes that we called tse7 and tsi7 We show that Tse7 is a Tox-GHH2 domain nuclease which is distinct from other T6SS nucleases identified thus far. Expression of this toxin induces the SOS response, causes growth arrest and ultimately results in DNA degradation. The cytotoxic domain of Tse7 lies at its C terminus, while the N terminus is a predicted PAAR domain. We find that Tse7 sits on the tip of the VgrG1b spike and that specific residues at the PAAR-VgrG1b interface are essential for VgrG1b-dependent delivery of Tse7 into bacterial prey. We also show that the delivery of Tse7 is dependent on the H1-T6SS cluster, and injection of the nuclease into bacterial competitors is deployed for interbacterial competition. Tsi7, the cognate immunity protein, protects the producer from the deleterious effect of Tse7 through a direct protein-protein interaction so specific that toxin/immunity pairs are effective only if they originate from the same P. aeruginosa isolate. Overall, our study highlights the diversity of T6SS effectors, the exquisite fitting of toxins on the tip of the T6SS, and the specificity in Tsi7-dependent protection, suggesting a role in interstrain competition.
Collapse
|
21
|
Pollard DJ, Berger CN, So EC, Yu L, Hadavizadeh K, Jennings P, Tate EW, Choudhary JS, Frankel G. Broad-Spectrum Regulation of Nonreceptor Tyrosine Kinases by the Bacterial ADP-Ribosyltransferase EspJ. mBio 2018; 9:e00170-18. [PMID: 29636436 PMCID: PMC5893879 DOI: 10.1128/mbio.00170-18] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Accepted: 03/08/2018] [Indexed: 12/11/2022] Open
Abstract
Tyrosine phosphorylation is key for signal transduction from exogenous stimuli, including the defense against pathogens. Conversely, pathogens can subvert protein phosphorylation to control host immune responses and facilitate invasion and dissemination. The bacterial effectors EspJ and SeoC are injected into host cells through a type III secretion system by enteropathogenic and enterohemorrhagic Escherichia coli (EPEC and EHEC, respectively), Citrobacter rodentium, and Salmonella enterica, where they inhibit Src kinase by coupled amidation and ADP-ribosylation. C. rodentium, which is used to model EPEC and EHEC infections in humans, is a mouse pathogen triggering colonic crypt hyperplasia (CCH) and colitis. Enumeration of bacterial shedding and CCH confirmed that EspJ affects neither tolerance nor resistance to infection. However, comparison of the proteomes of intestinal epithelial cells isolated from mice infected with wild-type C. rodentium or C. rodentium encoding catalytically inactive EspJ revealed that EspJ-induced ADP-ribosylation regulates multiple nonreceptor tyrosine kinases in vivo Investigation of the substrate repertoire of EspJ revealed that in HeLa and A549 cells, Src and Csk were significantly targeted; in polarized Caco2 cells, EspJ targeted Src and Csk and the Src family kinase (SFK) Yes1, while in differentiated Thp1 cells, EspJ modified Csk, the SFKs Hck and Lyn, the Tec family kinases Tec and Btk, and the adapter tyrosine kinase Syk. Furthermore, Abl (HeLa and Caco2) and Lyn (Caco2) were enriched specifically in the EspJ-containing samples. Biochemical assays revealed that EspJ, the only bacterial ADP-ribosyltransferase that targets mammalian kinases, controls immune responses and the Src/Csk signaling axis.IMPORTANCE Enteropathogenic and enterohemorrhagic Escherichia coli (EPEC and EHEC, respectively) strains cause significant mortality and morbidity worldwide. Citrobacter rodentium is a mouse pathogen used to model EPEC and EHEC pathogenesis in vivo Diarrheal disease is triggered following injection of bacterial effectors, via a type III secretion system (T3SS), into intestinal epithelial cells (IECs). While insights into the role of the effectors were historically obtained from pathological, immunologic, or cell culture phenotypes, subtle roles of individual effectors in vivo are often masked. The aim of this study was to elucidate the role and specificity of the ADP-ribosyltransferase effector EspJ. For the first time, we show that the in vivo processes affected by a T3SS effector can be studied by comparing the proteomes of IECs extracted from mice infected with wild-type C. rodentium or an espJ catalytic mutant. We show that EspJ, the only bacterial ADP-ribosyltransferase that targets mammalian kinases, regulates the host immune response in vivo.
Collapse
Affiliation(s)
- Dominic J Pollard
- Centre for Molecular Microbiology and Infection, Department of Life Sciences, Imperial College, London United Kingdom, London, United Kingdom
| | - Cedric N Berger
- Centre for Molecular Microbiology and Infection, Department of Life Sciences, Imperial College, London United Kingdom, London, United Kingdom
| | - Ernest C So
- Centre for Molecular Microbiology and Infection, Department of Life Sciences, Imperial College, London United Kingdom, London, United Kingdom
| | - Lu Yu
- Functional Proteomics Group, Chester Beatty Laboratories, Institute of Cancer Research, London, United Kingdom
| | - Kate Hadavizadeh
- Department of Chemistry, Imperial College, London United Kingdom, London, United Kingdom
| | | | - Edward W Tate
- Department of Chemistry, Imperial College, London United Kingdom, London, United Kingdom
| | - Jyoti S Choudhary
- Functional Proteomics Group, Chester Beatty Laboratories, Institute of Cancer Research, London, United Kingdom
| | - Gad Frankel
- Centre for Molecular Microbiology and Infection, Department of Life Sciences, Imperial College, London United Kingdom, London, United Kingdom
| |
Collapse
|
22
|
Pinaud L, Sansonetti PJ, Phalipon A. Host Cell Targeting by Enteropathogenic Bacteria T3SS Effectors. Trends Microbiol 2018; 26:266-283. [DOI: 10.1016/j.tim.2018.01.010] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Revised: 01/29/2018] [Accepted: 01/29/2018] [Indexed: 12/23/2022]
|
23
|
El Qaidi S, Wu M, Zhu C, Hardwidge PR. Salmonella, E. coli, and Citrobacter Type III Secretion System Effector Proteins that Alter Host Innate Immunity. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1111:205-218. [PMID: 30411307 DOI: 10.1007/5584_2018_289] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Bacteria deliver virulence proteins termed 'effectors' to counteract host innate immunity. Protein-protein interactions within the host cell ultimately subvert the generation of an inflammatory response to the infecting pathogen. Here we briefly describe a subset of T3SS effectors produced by enterohemorrhagic Escherichia coli (EHEC), enteropathogenic E. coli (EPEC), Citrobacter rodentium, and Salmonella enterica that inhibit innate immune pathways. These effectors are interesting for structural and mechanistic reasons, as well as for their potential utility in being engineered to treat human autoimmune disorders associated with perturbations in NF-κB signaling.
Collapse
Affiliation(s)
- Samir El Qaidi
- College of Veterinary Medicine, Kansas State University, Manhattan, KS, USA
| | - Miaomiao Wu
- College of Veterinary Medicine, Kansas State University, Manhattan, KS, USA
| | - Congrui Zhu
- College of Veterinary Medicine, Kansas State University, Manhattan, KS, USA
| | - Philip R Hardwidge
- College of Veterinary Medicine, Kansas State University, Manhattan, KS, USA.
| |
Collapse
|
24
|
Shenoy AR, Furniss RCD, Goddard PJ, Clements A. Modulation of Host Cell Processes by T3SS Effectors. Curr Top Microbiol Immunol 2018; 416:73-115. [PMID: 30178263 DOI: 10.1007/82_2018_106] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Two of the enteric Escherichia coli pathotypes-enteropathogenic E. coli (EPEC) and enterohaemorrhagic E. coli (EHEC)-have a conserved type 3 secretion system which is essential for virulence. The T3SS is used to translocate between 25 and 50 bacterial proteins directly into the host cytosol where they manipulate a variety of host cell processes to establish a successful infection. In this chapter, we discuss effectors from EPEC/EHEC in the context of the host proteins and processes that they target-the actin cytoskeleton, small guanosine triphosphatases and innate immune signalling pathways that regulate inflammation and cell death. Many of these translocated proteins have been extensively characterised, which has helped obtain insights into the mechanisms of pathogenesis of these bacteria and also understand the host pathways they target in more detail. With increasing knowledge of the positive and negative regulation of host signalling pathways by different effectors, a future challenge is to investigate how the specific effector repertoire of each strain cooperates over the course of an infection.
Collapse
Affiliation(s)
- Avinash R Shenoy
- MRC Centre for Molecular Bacteriology and Infection, Imperial College London, Armstrong Road, SW7 2AZ, London, UK
| | - R Christopher D Furniss
- MRC Centre for Molecular Bacteriology and Infection, Imperial College London, Armstrong Road, SW7 2AZ, London, UK
| | - Philippa J Goddard
- MRC Centre for Molecular Bacteriology and Infection, Imperial College London, Armstrong Road, SW7 2AZ, London, UK
| | - Abigail Clements
- MRC Centre for Molecular Bacteriology and Infection, Imperial College London, Armstrong Road, SW7 2AZ, London, UK.
| |
Collapse
|
25
|
Scott NE, Hartland EL. Post-translational Mechanisms of Host Subversion by Bacterial Effectors. Trends Mol Med 2017; 23:1088-1102. [PMID: 29150361 DOI: 10.1016/j.molmed.2017.10.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2017] [Revised: 10/19/2017] [Accepted: 10/19/2017] [Indexed: 12/19/2022]
Abstract
Bacterial effector proteins are a specialized class of secreted proteins that are translocated directly into the host cytoplasm by bacterial pathogens. Effector proteins have diverse activities and targets, and many mediate post-translational modifications of host proteins. Effector proteins offer potential in novel biotechnological and medical applications as enzymes that may modify human proteins. Here, we discuss the mechanisms used by effectors to subvert the human host through blocking, blunting, or subverting immune mechanisms. This capacity allows bacteria to control host cell function to support pathogen survival, replication and dissemination to other hosts. In addition, we highlight that knowledge of effector protein activity may be used to develop chemical inhibitors as a new approach to treat bacterial infections.
Collapse
Affiliation(s)
- Nichollas E Scott
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne 3000, Australia
| | - Elizabeth L Hartland
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton 3168, Australia; Department of Molecular and Translational Science, Monash University, Clayton 3168, Australia.
| |
Collapse
|
26
|
Pearson JS, Giogha C, Wong Fok Lung T, Hartland EL. The Genetics of EnteropathogenicEscherichia coliVirulence. Annu Rev Genet 2016; 50:493-513. [DOI: 10.1146/annurev-genet-120215-035138] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Jaclyn S. Pearson
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria 3010, Australia; , , ,
| | - Cristina Giogha
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria 3010, Australia; , , ,
| | - Tania Wong Fok Lung
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria 3010, Australia; , , ,
| | - Elizabeth L. Hartland
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria 3010, Australia; , , ,
| |
Collapse
|
27
|
The Type III Secretion System Effector SeoC of Salmonella enterica subsp. salamae and S. enterica subsp. arizonae ADP-Ribosylates Src and Inhibits Opsonophagocytosis. Infect Immun 2016; 84:3618-3628. [PMID: 27736780 PMCID: PMC5116738 DOI: 10.1128/iai.00704-16] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Accepted: 10/02/2016] [Indexed: 01/23/2023] Open
Abstract
Salmonella species utilize type III secretion systems (T3SSs) to translocate effectors into the cytosol of mammalian host cells, subverting cell signaling and facilitating the onset of gastroenteritis. In this study, we compared a draft genome assembly of Salmonella enterica subsp. salamae strain 3588/07 against the genomes of S. enterica subsp. enterica serovar Typhimurium strain LT2 and Salmonella bongori strain 12419. S. enterica subsp. salamae encodes the Salmonella pathogenicity island 1 (SPI-1), SPI-2, and the locus of enterocyte effacement (LEE) T3SSs. Though several key S Typhimurium effector genes are missing (e.g., avrA, sopB, and sseL), S. enterica subsp. salamae invades HeLa cells and contains homologues of S. bongori sboK and sboC, which we named seoC SboC and SeoC are homologues of EspJ from enteropathogenic and enterohemorrhagic Escherichia coli (EPEC and EHEC, respectively), which inhibit Src kinase-dependent phagocytosis by ADP-ribosylation. By screening 73 clinical and environmental Salmonella isolates, we identified EspJ homologues in S. bongori, S. enterica subsp. salamae, and Salmonella enterica subsp. arizonae The β-lactamase TEM-1 reporter system showed that SeoC is translocated by the SPI-1 T3SS. All the Salmonella SeoC/SboC homologues ADP-ribosylate Src E310 in vitro Ectopic expression of SeoC/SboC inhibited phagocytosis of IgG-opsonized beads into Cos-7 cells stably expressing green fluorescent protein (GFP)-FcγRIIa. Concurrently, S. enterica subsp. salamae infection of J774.A1 macrophages inhibited phagocytosis of beads, in a seoC-dependent manner. These results show that S. bongori, S. enterica subsp. salamae, and S. enterica subsp. arizonae share features of the infection strategy of extracellular pathogens EPEC and EHEC and shed light on the complexities of the T3SS effector repertoires of Enterobacteriaceae.
Collapse
|
28
|
Humphreys D, Singh V, Koronakis V. Inhibition of WAVE Regulatory Complex Activation by a Bacterial Virulence Effector Counteracts Pathogen Phagocytosis. Cell Rep 2016; 17:697-707. [PMID: 27732847 PMCID: PMC5081413 DOI: 10.1016/j.celrep.2016.09.039] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Revised: 06/24/2016] [Accepted: 09/14/2016] [Indexed: 02/05/2023] Open
Abstract
To establish pathogenicity, bacteria must evade phagocytosis directed by remodeling of the actin cytoskeleton. We show that macrophages facilitate pathogen phagocytosis through actin polymerization mediated by the WAVE regulatory complex (WRC), small GTPases Arf and Rac1, and the Arf1 activator ARNO. To establish extracellular infections, enteropathogenic (EPEC) and enterohaemorrhagic (EHEC) Escherichia coli hijack the actin cytoskeleton by injecting virulence effectors into the host cell. Here, we find that the virulence effector EspG counteracts WRC-dependent phagocytosis, enabling EPEC and EHEC to remain extracellular. By reconstituting membrane-associated actin polymerization, we find that EspG disabled WRC activation through two mechanisms: EspG interaction with Arf6 blocked signaling to ARNO while EspG binding of Arf1 impeded collaboration with Rac1, thereby inhibiting WRC recruitment and activation. Investigating the mode of EspG interference revealed sites in Arf1 required for WRC activation and a mechanism facilitating pathogen evasion of innate host defenses.
Collapse
Affiliation(s)
- Daniel Humphreys
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QP, UK.
| | - Vikash Singh
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QP, UK
| | - Vassilis Koronakis
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QP, UK.
| |
Collapse
|
29
|
Mavridou DAI, Gonzalez D, Clements A, Foster KR. The pUltra plasmid series: A robust and flexible tool for fluorescent labeling of Enterobacteria. Plasmid 2016; 87-88:65-71. [PMID: 27693407 DOI: 10.1016/j.plasmid.2016.09.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Revised: 09/20/2016] [Accepted: 09/28/2016] [Indexed: 10/20/2022]
Abstract
Fluorescent labeling has been an invaluable tool for the study of living organisms and bacterial species are no exception to this. Here we present and characterize the pUltra plasmids which express constitutively a fluorescent protein gene (GFP, RFP, YFP or CFP) from a strong synthetic promoter and are suitable for the fluorescent labeling of a broad range of Enterobacteria. The amount of expressed fluorophore from these genetic constructs is such, that the contours of the cells can be delineated on the basis of the fluorescent signal only. In addition, labeling through the pUltra plasmids can be used successfully for fluorescence and confocal microscopy while unambiguous distinction of cells labeled with different colors can be carried out efficiently by microscopy or flow cytometry. We compare the labeling provided by the pUltra plasmids with that of another plasmid series encoding fluorescent proteins and we show that the pUltra constructs are vastly superior in signal intensity and discrimination power without having any detectable growth rate effects for the bacterial population. We also use the pUltra plasmids to produce mixtures of differentially labeled pathogenic Escherichia, Shigella and Salmonella species which we test during infection of mammalian cells. We find that even inside the host cell, different strains can be distinguished effortlessly based on their fluorescence. We, therefore, conclude that the pUltra plasmids are a powerful labeling tool especially useful for complex biological experiments such as the visualization of ecosystems of different bacterial species or of enteric pathogens in contact with their hosts.
Collapse
Affiliation(s)
- Despoina A I Mavridou
- MRC Centre for Molecular Bacteriology and Infection, Department of Life Sciences, Faculty of Natural Sciences, Imperial College London, Kensington, London SW7 2DD, UK.
| | - Diego Gonzalez
- Department of Zoology, University of Oxford, Oxford OX1 3PS, UK
| | - Abigail Clements
- MRC Centre for Molecular Bacteriology and Infection, Department of Life Sciences, Faculty of Natural Sciences, Imperial College London, Kensington, London SW7 2DD, UK
| | - Kevin R Foster
- Department of Zoology, University of Oxford, Oxford OX1 3PS, UK
| |
Collapse
|
30
|
Law RJ, Law HT, Scurll JM, Scholz R, Santos AS, Shames SR, Deng W, Croxen MA, Li Y, de Hoog CL, van der Heijden J, Foster LJ, Guttman JA, Finlay BB. Quantitative Mass Spectrometry Identifies Novel Host Binding Partners for Pathogenic Escherichia coli Type III Secretion System Effectors. J Proteome Res 2016; 15:1613-22. [DOI: 10.1021/acs.jproteome.6b00074] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
| | - Hong T. Law
- Department
of Biological Sciences, Simon Fraser University, Burnaby, British Columbia, Canada V5A 1S6
| | | | | | | | | | | | | | | | | | | | | | - Julian A. Guttman
- Department
of Biological Sciences, Simon Fraser University, Burnaby, British Columbia, Canada V5A 1S6
| | | |
Collapse
|
31
|
Van Nhieu GT, Romero S. Common Themes in Cytoskeletal Remodeling by Intracellular Bacterial Effectors. Handb Exp Pharmacol 2016; 235:207-235. [PMID: 27807696 DOI: 10.1007/164_2016_42] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Bacterial pathogens interact with various types of tissues to promote infection. Because it controls the formation of membrane extensions, adhesive processes, or the junction integrity, the actin cytoskeleton is a key target of pathogens during infection. We will highlight common and specific functions of the actin cytoskeleton during bacterial infections, by first reviewing the mechanisms of intracellular motility of invasive Shigella, Listeria, and Rickettsia. Through the models of EPEC/EHEC, Shigella, Salmonella, and Chlamydia spp., we will illustrate various strategies of diversion of actin cytoskeletal processes used by these bacteria to colonize or breach epithelial/endothelial barriers.
Collapse
Affiliation(s)
- Guy Tran Van Nhieu
- Equipe Communication Intercellulaire et Infections Microbiennes, Centre de Recherche Interdisciplinaire en Biologie (CIRB), Collège de France, 75005, Paris, France. .,Institut National de la Santé et de la Recherche Médicale U1050, 75005, Paris, France. .,Centre National de la Recherche Scientifique UMR7241, 75005, Paris, France. .,MEMOLIFE Laboratory of Excellence and Paris Science Lettre, 75005, Paris, France.
| | - Stéphane Romero
- Equipe Communication Intercellulaire et Infections Microbiennes, Centre de Recherche Interdisciplinaire en Biologie (CIRB), Collège de France, 75005, Paris, France.,Institut National de la Santé et de la Recherche Médicale U1050, 75005, Paris, France.,Centre National de la Recherche Scientifique UMR7241, 75005, Paris, France.,MEMOLIFE Laboratory of Excellence and Paris Science Lettre, 75005, Paris, France
| |
Collapse
|
32
|
Jank T, Belyi Y, Aktories K. Bacterial glycosyltransferase toxins. Cell Microbiol 2015; 17:1752-65. [DOI: 10.1111/cmi.12533] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Revised: 10/05/2015] [Accepted: 10/06/2015] [Indexed: 12/28/2022]
Affiliation(s)
- Thomas Jank
- Institute for Experimental and Clinical Pharmacology and Toxicology; Albert-Ludwigs University of Freiburg; Freiburg Germany
| | - Yury Belyi
- Gamaleya Research Institute; Moscow 123098 Russia
- Freiburg Institute for Advanced Studies (FRIAS); Albert-Ludwigs University of Freiburg; Freiburg Germany
| | - Klaus Aktories
- Institute for Experimental and Clinical Pharmacology and Toxicology; Albert-Ludwigs University of Freiburg; Freiburg Germany
- Freiburg Institute for Advanced Studies (FRIAS); Albert-Ludwigs University of Freiburg; Freiburg Germany
| |
Collapse
|
33
|
RD-1 encoded EspJ protein gets phosphorylated prior to affect the growth and intracellular survival of mycobacteria. Sci Rep 2015; 5:12717. [PMID: 26228622 PMCID: PMC4521147 DOI: 10.1038/srep12717] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Accepted: 07/02/2015] [Indexed: 01/12/2023] Open
Abstract
Mycobacterium tuberculosis (MTB) synchronizes a number of processes and controls a series of events to subvert host defense mechanisms for the sake of residing inside macrophages. Besides these, MTB also possesses a wide range of signal enzyme systems, including eleven serine threonine protein kinases (STPKs). The present study describes STPK modulated modification in one of the hypothetical proteins of the RD1 region; EspJ (ESX-1 secretion associated protein), which is predicted to be involved in virulence of MTB. We have employed knock-out MTB, and M. bovis BCG as a surrogate strain to elaborate the consequence of the phosphorylation of EspJ. The molecular and mass spectrometric analyses in this study, confirmed EspJ as one of the substrates of STPKs. The ectopic expression of phosphoablative mutants of espJ in M. bovis BCG also articulated the effect of phosphorylation on the growth and in survival of mycobacteria. Importantly, the level of phosphorylation of EspJ also differed between pathogenic H37 Rv (Rv) and non pathogenic H37 Ra (Ra) strains of MTB. This further suggested that to a certain extent, the STPKs mediated phosphorylation may be accountable, in determining the growth and in intra-cellular survival of mycobacteria.
Collapse
|
34
|
Tir Triggers Expression of CXCL1 in Enterocytes and Neutrophil Recruitment during Citrobacter rodentium Infection. Infect Immun 2015; 83:3342-54. [PMID: 26077760 DOI: 10.1128/iai.00291-15] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 06/09/2015] [Indexed: 12/28/2022] Open
Abstract
The hallmarks of enteropathogenic Escherichia coli (EPEC) infection are formation of attaching and effacing (A/E) lesions on mucosal surfaces and actin-rich pedestals on cultured cells, both of which are dependent on the type III secretion system effector Tir. Following translocation into cultured cells and clustering by intimin, Tir Y474 is phosphorylated, leading to recruitment of Nck, activation of N-WASP, and actin polymerization via the Arp2/3 complex. A secondary, weak, actin polymerization pathway is triggered via an NPY motif (Y454). Importantly, Y454 and Y474 play no role in A/E lesion formation on mucosal surfaces following infection with the EPEC-like mouse pathogen Citrobacter rodentium. In this study, we investigated the roles of Tir segments located upstream of Y451 and downstream of Y471 in C. rodentium colonization and A/E lesion formation. We also tested the role that Tir residues Y451 and Y471 play in host immune responses to C. rodentium infection. We found that deletion of amino acids 382 to 462 or 478 to 547 had no impact on the ability of Tir to mediate A/E lesion formation, although deletion of amino acids 478 to 547 affected Tir translocation. Examination of enterocytes isolated from infected mice revealed that a C. rodentium strain expressing Tir_Y451A/Y471A recruited significantly fewer neutrophils to the colon and triggered less colonic hyperplasia on day 14 postinfection than the wild-type strain. Consistently, enterocytes isolated from mice infected with C. rodentium expressing Tir_Y451A/Y471A expressed significantly less CXCL1. These result show that Tir-induced actin remodeling plays a direct role in modulation of immune responses to C. rodentium infection.
Collapse
|