1
|
Raab JE, Harju TB, Toperzer JD, Duncan-Lowey JK, Goldberg MB, Russo BC. A translocation-competent pore is required for Shigella flexneri to escape from the double membrane vacuole during intercellular spread. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.11.623084. [PMID: 39605318 PMCID: PMC11601285 DOI: 10.1101/2024.11.11.623084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Type 3 secretion systems (T3SSs) enable bacterial virulence by translocating virulence proteins (effectors) into host cells. Shigella flexneri require T3SS to invade and to spread between cells in the colon. In order to spread, S. flexneri forms membrane protrusions that push into the adjacent host cell. These protrusions are resolved into double membrane vacuoles (DMVs) that the bacteria quickly escape. The mechanisms required for escape from the DMV are poorly understood, but the T3SS translocon pore protein IpaC is essential. Here, we show IpaC forms a pore that is competent for translocation of T3SS effectors as bacteria spread between cells. To do so, we used a genetic approach to test mutations of IpaC that disrupt its ability to translocate and to form pores. We show that during spread, IpaC is efficiently inserted into the plasma membrane, the membrane-embedded IpaC forms pore complexes, and the IpaC-dependent pores translocate effectors that are necessary for S. flexneri to escape the DMV. We further show that T3SS activation is regulated through a distinct mechanism at spread compared to at invasion; activation of T3SS secretion does not require pore formation during spread. Thus, we show that a distinct regulation of the T3SS during S. flexneri intercellular spread enables the placement of effectors both around S. flexneri and across membranes of the DMV. Altogether, this study provides new insights into how S. flexneri escapes the DMV. IMPORTANCE The type 3 secretion system (T3SS) is required for virulence in many bacterial pathogens that infect humans. The T3SS forms a pore through which virulence proteins are delivered into host cells to enable bacterial infection. Our work investigates the Shigella translocon pore protein IpaC, which is essential not only for bacteria to invade cells, but also for bacteria to spread between cells. An ability to spread between cells is essential for pathogenesis, thus understanding the mechanisms that enable spread is important for understanding how S. flexneri infection causes illness. We show that IpaC delivers virulence factors across the host membrane for S. flexneri to efficiently spread. This study furthers our understanding of the mechanisms involved in T3SS secretion and of translocon pore function during S. flexneri intercellular spread.
Collapse
|
2
|
Raab JE, Hamilton DJ, Harju TB, Huynh TN, Russo BC. Pushing boundaries: mechanisms enabling bacterial pathogens to spread between cells. Infect Immun 2024; 92:e0052423. [PMID: 38661369 PMCID: PMC11385730 DOI: 10.1128/iai.00524-23] [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] [Indexed: 04/26/2024] Open
Abstract
For multiple intracellular bacterial pathogens, the ability to spread directly into adjacent epithelial cells is an essential step for disease in humans. For pathogens such as Shigella, Listeria, Rickettsia, and Burkholderia, this intercellular movement frequently requires the pathogens to manipulate the host actin cytoskeleton and deform the plasma membrane into structures known as protrusions, which extend into neighboring cells. The protrusion is then typically resolved into a double-membrane vacuole (DMV) from which the pathogen quickly escapes into the cytosol, where additional rounds of intercellular spread occur. Significant progress over the last few years has begun to define the mechanisms by which intracellular bacterial pathogens spread. This review highlights the interactions of bacterial and host factors that drive mechanisms required for intercellular spread with a focus on how protrusion structures form and resolve.
Collapse
Affiliation(s)
- Julie E. Raab
- Department of Immunology and Microbiology, School of Medicine, University of Colorado—Anschutz Medical Campus, Denver, Colorado, USA
| | - Desmond J. Hamilton
- Department of Immunology and Microbiology, School of Medicine, University of Colorado—Anschutz Medical Campus, Denver, Colorado, USA
| | - Tucker B. Harju
- Department of Immunology and Microbiology, School of Medicine, University of Colorado—Anschutz Medical Campus, Denver, Colorado, USA
| | - Thao N. Huynh
- Department of Immunology and Microbiology, School of Medicine, University of Colorado—Anschutz Medical Campus, Denver, Colorado, USA
| | - Brian C. Russo
- Department of Immunology and Microbiology, School of Medicine, University of Colorado—Anschutz Medical Campus, Denver, Colorado, USA
| |
Collapse
|
3
|
Anandachar MS, Roy S, Sinha S, Boadi A, Katkar GD, Ghosh P. Diverse gut pathogens exploit the host engulfment pathway via a conserved mechanism. J Biol Chem 2023; 299:105390. [PMID: 37890785 PMCID: PMC10696401 DOI: 10.1016/j.jbc.2023.105390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 09/22/2023] [Accepted: 10/15/2023] [Indexed: 10/29/2023] Open
Abstract
Macrophages clear infections by engulfing and digesting pathogens within phagolysosomes. Pathogens escape this fate by engaging in a molecular arms race; they use WxxxE motif-containing "effector" proteins to subvert the host cells they invade and seek refuge within protective vacuoles. Here, we define the host component of the molecular arms race as an evolutionarily conserved polar "hot spot" on the PH domain of ELMO1 (Engulfment and Cell Motility protein 1), which is targeted by diverse WxxxE effectors. Using homology modeling and site-directed mutagenesis, we show that a lysine triad within the "patch" directly binds all WxxxE effectors tested: SifA (Salmonella), IpgB1 and IpgB2 (Shigella), and Map (enteropathogenic Escherichia coli). Using an integrated SifA-host protein-protein interaction network, in silico network perturbation, and functional studies, we show that the major consequences of preventing SifA-ELMO1 interaction are reduced Rac1 activity and microbial invasion. That multiple effectors of diverse structure, function, and sequence bind the same hot spot on ELMO1 suggests that the WxxxE effector(s)-ELMO1 interface is a convergence point of intrusion detection and/or host vulnerability. We conclude that the interface may represent the fault line in coevolved molecular adaptations between pathogens and the host, and its disruption may serve as a therapeutic strategy.
Collapse
Affiliation(s)
- Mahitha Shree Anandachar
- Department of Cellular and Molecular Medicine, University of California San Diego, San Diego, California, USA; Department of Pathology, University of California San Diego, San Diego, California, USA
| | - Suchismita Roy
- Department of Cellular and Molecular Medicine, University of California San Diego, San Diego, California, USA
| | - Saptarshi Sinha
- Department of Cellular and Molecular Medicine, University of California San Diego, San Diego, California, USA
| | - Agyekum Boadi
- Department of Cellular and Molecular Medicine, University of California San Diego, San Diego, California, USA
| | - Gajanan D Katkar
- Department of Cellular and Molecular Medicine, University of California San Diego, San Diego, California, USA.
| | - Pradipta Ghosh
- Department of Cellular and Molecular Medicine, University of California San Diego, San Diego, California, USA; Department of Medicine, University of California San Diego, San Diego, California, USA.
| |
Collapse
|
4
|
Achi SC, Karimilangi S, Lie D, Sayed IM, Das S. The WxxxE proteins in microbial pathogenesis. Crit Rev Microbiol 2023; 49:197-213. [PMID: 35287539 PMCID: PMC9737147 DOI: 10.1080/1040841x.2022.2046546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Revised: 02/10/2022] [Accepted: 02/19/2022] [Indexed: 12/14/2022]
Abstract
Effector proteins secreted by pathogens modulate various host cellular processes and help in bacterial pathogenesis. Some of these proteins, injected by enteric pathogens via Type Three Secretion System (T3SS) were grouped together based on a conserved signature motif (WxxxE) present in them. The presence of WxxxE motif is not limited to effectors released by enteric pathogens or the T3SS but has been detected in non-enteric pathogens, plant pathogens and in association with Type II and Type IV secretion systems. WxxxE effectors are involved in actin organization, inflammation regulation, vacuole or tubule formation, endolysosomal signalling regulation, tight junction disruption, and apoptosis. The WxxxE sequence has also been identified in TIR [Toll/interleukin-1 (IL-1) receptor] domains of bacteria and host. In the present review, we have focussed on the established and predicted functions of WxxxE effectors secreted by several pathogens, including enteric, non-enteric, and plant pathogens.
Collapse
Affiliation(s)
| | - Sareh Karimilangi
- Department of Pathology, University of California San Diego, La Jolla, CA, USA
| | - Dominique Lie
- Department of Pathology, University of California San Diego, La Jolla, CA, USA
| | - Ibrahim M. Sayed
- Department of Pathology, University of California San Diego, La Jolla, CA, USA
| | - Soumita Das
- Department of Pathology, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
5
|
Hartland EL, Ghosal D, Giogha C. Manipulation of epithelial cell architecture by the bacterial pathogens Listeria and Shigella. Curr Opin Cell Biol 2022; 79:102131. [PMID: 36215855 DOI: 10.1016/j.ceb.2022.102131] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Revised: 08/22/2022] [Accepted: 09/06/2022] [Indexed: 01/31/2023]
Abstract
Subversion of the host cell cytoskeleton is a virulence attribute common to many bacterial pathogens. On mucosal surfaces, bacteria have evolved distinct ways of interacting with the polarised epithelium and manipulating host cell structure to propagate infection. For example, Shigella and Listeria induce cytoskeletal changes to induce their own uptake into enterocytes in order to replicate within an intracellular environment and then spread from cell-to-cell by harnessing the host actin cytoskeleton. In this review, we highlight some recent studies that advance our understanding of the role of the host cell cytoskeleton in the mechanical and molecular processes of pathogen invasion, cell-to-cell spread and the impact of infection on epithelial intercellular tension and innate mucosal defence.
Collapse
Affiliation(s)
- Elizabeth L Hartland
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Victoria, Australia; Department of Molecular and Translational Science, Monash University, Clayton, Victoria, Australia.
| | - Debnath Ghosal
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne, VIC, Australia
| | - Cristina Giogha
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Victoria, Australia; Department of Molecular and Translational Science, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
6
|
Abstract
The technology of recombineering, in vivo genetic engineering, was initially developed in Escherichia coli and uses bacteriophage-encoded homologous recombination proteins to efficiently recombine DNA at short homologies (35 to 50 nt). Because the technology is homology driven, genomic DNA can be modified precisely and independently of restriction site location. Recombineering uses linear DNA substrates that are introduced into the cell by electroporation; these can be PCR products, synthetic double-strand DNA (dsDNA), or single-strand DNA (ssDNA). Here we describe the applications, challenges, and factors affecting ssDNA and dsDNA recombineering in a variety of non-model bacteria, both Gram-negative and -positive, and recent breakthroughs in the field. We list different microbes in which the widely used phage λ Red and Rac RecET recombination systems have been used for in vivo genetic engineering. New homologous ssDNA and dsDNA recombineering systems isolated from non-model bacteria are also described. The Basic Protocol outlines a method for ssDNA recombineering in the non-model species of Shewanella. The Alternate Protocol describes the use of CRISPR/Cas as a counter-selection system in conjunction with recombineering to enhance recovery of recombinants. We provide additional background information, pertinent considerations for experimental design, and parameters critical for success. The design of ssDNA oligonucleotides (oligos) and various internet-based tools for oligo selection from genome sequences are also described, as is the use of oligo-mediated recombination. This simple form of genome editing uses only ssDNA oligo(s) and does not require an exogenous recombination system. The information presented here should help researchers identify a recombineering system suitable for their microbe(s) of interest. If no system has been characterized for a specific microbe, researchers can find guidance in developing a recombineering system from scratch. We provide a flowchart of decision-making paths for strategically applying annealase-dependent or oligo-mediated recombination in non-model and undomesticated bacteria. © 2022 Wiley Periodicals LLC. This article has been contributed to by U.S. Government employees and their work is in the public domain in the USA. Basic Protocol: ssDNA recombineering in Shewanella species Alternate Protocol: ssDNA recombineering coupled to CRISPR/Cas9 in Shewanella species.
Collapse
Affiliation(s)
- Anna Corts
- Cultivarium, 490 Arsenal Way, Ste 110, Watertown, Massachusetts 02472
| | - Lynn C. Thomason
- Molecular Control and Genetics Section, RNA Biology Laboratory, National Cancer Institute at Frederick, National Institutes of Health, Frederick, Maryland 21702
| | - Nina Costantino
- Molecular Control and Genetics Section, RNA Biology Laboratory, National Cancer Institute at Frederick, National Institutes of Health, Frederick, Maryland 21702
| | - Donald L. Court
- Emeritus, Molecular Control and Genetics Section, RNA Biology Laboratory, National Cancer Institute at Frederick, National Institutes of Health, Frederick, Maryland 21702
| |
Collapse
|
7
|
The type three secretion system effector protein IpgB1 promotes Shigella flexneri cell-to-cell spread through double-membrane vacuole escape. PLoS Pathog 2022; 18:e1010380. [PMID: 35202448 PMCID: PMC8903249 DOI: 10.1371/journal.ppat.1010380] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 03/08/2022] [Accepted: 02/18/2022] [Indexed: 12/11/2022] Open
Abstract
S. flexneri is an important human pathogen that causes bacillary dysentery. During infection, S. flexneri invades colonic epithelial cells, hijacks the host cell cytoskeleton to move in the cytosol of infected cells, and spreads from cell to cell through formation of membrane protrusions that project into adjacent cells and resolve into double membrane vacuoles (DMVs). S. flexneri cell-to-cell spread requires the integrity of the bacterial type three secretion system (T3SS). However, the exact role of the T3SS effector proteins in the dissemination process remains poorly understood. Here, we investigated the role of the T3SS effector protein IpgB1 in S. flexneri dissemination. IpgB1 was previously characterized as a guanine nucleotide exchange factor (GEF) that contributes to invasion. In addition to the invasion defect, we showed that the ipgB1 mutant formed smaller infection foci in HT-29 cells. Complementation of this phenotype required the GEF activity of IpgB1. Using live confocal microscopy, we showed that the ipgB1 mutant is specifically impaired in DMV escape. Depletion of Rac1, the host cell target of IpgB1 during invasion, as well as pharmacological inhibition of Rac1 signaling, reduced cell-to-cell spread and DMV escape. In a targeted siRNA screen, we uncovered that RhoA depletion restored ipgB1 cell-to-cell spread and DMV escape, revealing a critical role for the IpgB1-Rac1 axis in antagonizing RhoA-mediated restriction of DMV escape. Using an infant rabbit model of shigellosis, we showed that the ipgB1 mutant formed fewer and smaller infection foci in the colon of infected animals, which correlated with attenuated symptoms of disease, including epithelial fenestration and bloody diarrhea. Our results demonstrate that, in addition to its role during invasion, IpgB1 modulates Rho family small GTPase signaling to promote cell-to-cell spread, DMV escape, and S. flexneri pathogenesis.
Collapse
|
8
|
Parapini S, Paone S, Erba E, Cavicchini L, Pourshaban M, Celani F, Contini A, D’Alessandro S, Olivieri A. In Vitro Antimalarial Activity of Inhibitors of the Human GTPase Rac1. Antimicrob Agents Chemother 2022; 66:e0149821. [PMID: 34723630 PMCID: PMC8765435 DOI: 10.1128/aac.01498-21] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 10/19/2021] [Indexed: 11/20/2022] Open
Abstract
Malaria accounts for millions of cases and thousands of deaths every year. In the absence of an effective vaccine, drugs are still the most important tool in the fight against the disease. Plasmodium parasites developed resistance to all classes of known antimalarial drugs. Thus, the search for antimalarial drugs with novel mechanisms of action is compelling. The human GTPase Rac1 plays a role in parasite invasion of the host cell in many intracellular pathogens. Also, in Plasmodium falciparum, the involvement of Rac1 during both the invasion process and parasite intracellular development was suggested. The aim of this work is to test a panel of Rac1 inhibitors as potential antimalarial drugs. Fourteen commercially available or newly synthesized inhibitors of Rac1 were tested for antimalarial activity. Among these, EHop-016 was the most effective against P. falciparum in vitro, with nanomolar 50% inhibitory concentrations (IC50s) (138.8 ± 16.0 nM on the chloroquine-sensitive D10 strain and 321.5 ± 28.5 nM on the chloroquine-resistant W2 strain) and a selectivity index of 37.8. EHop-016 did not inhibit parasite invasion of red blood cells but affected parasite growth inside them. Among the tested Rac1 inhibitors, EHop-016 showed promising activity that raises attention to this class of molecules as potential antimalarials and deserves further investigation.
Collapse
Affiliation(s)
- Silvia Parapini
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, Milan, Italy
| | - Silvio Paone
- Dipartimento di Sanità Pubblica e Malattie Infettive, Sapienza Università di Roma, Rome, Italy
- Dipartimento di Malattie Infettive, Istituto Superiore di Sanità, Rome, Italy
| | - Emanuela Erba
- Dipartimento di Scienze Farmaceutiche, Università degli Studi di Milano, Milan, Italy
| | - Loredana Cavicchini
- Dipartimento di Scienze Biomediche, Chirurgiche e Odontoiatriche, Università degli Studi di Milano, Milan, Italy
| | | | - Francesco Celani
- Dipartimento di Malattie Infettive, Istituto Superiore di Sanità, Rome, Italy
| | - Alessandro Contini
- Dipartimento di Scienze Farmaceutiche, Università degli Studi di Milano, Milan, Italy
| | - Sarah D’Alessandro
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - Anna Olivieri
- Dipartimento di Malattie Infettive, Istituto Superiore di Sanità, Rome, Italy
| |
Collapse
|
9
|
Hajra D, Nair AV, Chakravortty D. An elegant nano-injection machinery for sabotaging the host: Role of Type III secretion system in virulence of different human and animal pathogenic bacteria. Phys Life Rev 2021; 38:25-54. [PMID: 34090822 DOI: 10.1016/j.plrev.2021.05.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Accepted: 05/23/2021] [Indexed: 01/22/2023]
Abstract
Various Gram-negative bacteria possess a specialized membrane-bound protein secretion system known as the Type III secretion system (T3SS), which transports the bacterial effector proteins into the host cytosol thereby helping in bacterial pathogenesis. The T3SS has a special needle-like translocon that can sense the contact with the host cell membrane and translocate effectors. The export apparatus of T3SS recognizes these effector proteins bound to chaperones and translocates them into the host cell. Once in the host cell cytoplasm, these effector proteins result in modulation of the host system and promote bacterial localization and infection. Using molecular biology, bioinformatics, genetic techniques, electron microscopic studies, and mathematical modeling, the structure and function of the T3SS and the corresponding effector proteins in various bacteria have been studied. The strategies used by different human pathogenic bacteria to modulate the host system and thereby enhance their virulence mechanism using T3SS have also been well studied. Here we review the history, evolution, and general structure of the T3SS, highlighting the details of its comparison with the flagellar export machinery. Also, this article provides mechanistic details about the common role of T3SS in subversion and manipulation of host cellular processes. Additionally, this review describes specific T3SS apparatus and the role of their specific effectors in bacterial pathogenesis by considering several human and animal pathogenic bacteria.
Collapse
Affiliation(s)
- Dipasree Hajra
- Department of Microbiology & Cell Biology, Indian Institute of Science, India
| | - Abhilash Vijay Nair
- Department of Microbiology & Cell Biology, Indian Institute of Science, India
| | | |
Collapse
|
10
|
Kühn S, Bergqvist J, Gil M, Valenzuela C, Barrio L, Lebreton S, Zurzolo C, Enninga J. Actin Assembly around the Shigella-Containing Vacuole Promotes Successful Infection. Cell Rep 2021; 31:107638. [PMID: 32402280 PMCID: PMC7225751 DOI: 10.1016/j.celrep.2020.107638] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 03/10/2020] [Accepted: 04/21/2020] [Indexed: 12/13/2022] Open
Abstract
The enteroinvasive bacterium Shigella flexneri forces its uptake into non-phagocytic host cells through the translocation of T3SS effectors that subvert the actin cytoskeleton. Here, we report de novo actin polymerization after cellular entry around the bacterium-containing vacuole (BCV) leading to the formation of a dynamic actin cocoon. This cocoon is thicker than any described cellular actin structure and functions as a gatekeeper for the cytosolic access of the pathogen. Host CDC42, TOCA-1, N-WASP, WIP, the Arp2/3 complex, cortactin, coronin, and cofilin are recruited to the actin cocoon. They are subverted by T3SS effectors, such as IpgD, IpgB1, and IcsB. IcsB immobilizes components of the actin polymerization machinery at the BCV dependent on its fatty acyltransferase activity. This represents a unique microbial subversion strategy through localized entrapment of host actin regulators causing massive actin assembly. We propose that the cocoon promotes subsequent invasion steps for successful Shigella infection. A thick actin cocoon forms de novo around the Shigella-containing vacuole upon entry The effector IcsB entraps host actin regulators at the vacuole by lipidation Cdc42, N-WASP, and the Arp2/3 complex are major actin cocoon regulators Cocoon formation promotes subsequent Shigella niche formation and dissemination
Collapse
Affiliation(s)
- Sonja Kühn
- Institut Pasteur, Department of Cell Biology and Infection, Dynamics of Host-Pathogen Interactions Unit, 25 Rue du Dr. Roux, 75015 Paris, France; CNRS UMR3691, 25 Rue du Dr. Roux, 75015 Paris, France
| | - John Bergqvist
- Institut Pasteur, Department of Cell Biology and Infection, Dynamics of Host-Pathogen Interactions Unit, 25 Rue du Dr. Roux, 75015 Paris, France; CNRS UMR3691, 25 Rue du Dr. Roux, 75015 Paris, France
| | - Magdalena Gil
- Institut Pasteur, Department of Cell Biology and Infection, Dynamics of Host-Pathogen Interactions Unit, 25 Rue du Dr. Roux, 75015 Paris, France; CNRS UMR3691, 25 Rue du Dr. Roux, 75015 Paris, France
| | - Camila Valenzuela
- Institut Pasteur, Department of Cell Biology and Infection, Dynamics of Host-Pathogen Interactions Unit, 25 Rue du Dr. Roux, 75015 Paris, France; CNRS UMR3691, 25 Rue du Dr. Roux, 75015 Paris, France
| | - Laura Barrio
- Institut Pasteur, Department of Cell Biology and Infection, Dynamics of Host-Pathogen Interactions Unit, 25 Rue du Dr. Roux, 75015 Paris, France; CNRS UMR3691, 25 Rue du Dr. Roux, 75015 Paris, France
| | - Stéphanie Lebreton
- Institut Pasteur, Department of Cell Biology and Infection, Membrane Trafficking and Pathogenesis Unit, 28 Rue du Dr. Roux, 75015 Paris, France
| | - Chiara Zurzolo
- Institut Pasteur, Department of Cell Biology and Infection, Membrane Trafficking and Pathogenesis Unit, 28 Rue du Dr. Roux, 75015 Paris, France
| | - Jost Enninga
- Institut Pasteur, Department of Cell Biology and Infection, Dynamics of Host-Pathogen Interactions Unit, 25 Rue du Dr. Roux, 75015 Paris, France; CNRS UMR3691, 25 Rue du Dr. Roux, 75015 Paris, France.
| |
Collapse
|
11
|
Chang YY, Enninga J, Stévenin V. New methods to decrypt emerging macropinosome functions during the host-pathogen crosstalk. Cell Microbiol 2021; 23:e13342. [PMID: 33848057 PMCID: PMC8365644 DOI: 10.1111/cmi.13342] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 03/30/2021] [Accepted: 03/31/2021] [Indexed: 12/18/2022]
Abstract
Large volumes of liquid and other materials from the extracellular environment are internalised by eukaryotic cells via an endocytic process called macropinocytosis. It is now recognised that this fundamental and evolutionarily conserved pathway is hijacked by numerous intracellular pathogens as an entry portal to the host cell interior. Yet, an increasing number of additional cellular functions of macropinosomes in pathologic processes have been reported beyond this role for fluid internalisation. It emerges that the identity of macropinosomes can vary hugely and change rapidly during their lifetime. A deeper understanding of this important multi-faceted compartment is based on novel methods for their investigation. These methods are either imaging-based for the tracking of macropinosome dynamics, or they provide the means to extract macropinosomes at high purity for comprehensive proteomic analyses. Here, we portray these new approaches for the investigation of macropinosomes. We document how these method developments have provided insights for a new understanding of the intracellular lifestyle of the bacterial pathogens Shigella and Salmonella. We suggest that a systematic complete characterisation of macropinosome subversion with these approaches during other infection processes and pathologies will be highly beneficial for our understanding of the underlying cellular and molecular processes.
Collapse
Affiliation(s)
- Yuen-Yan Chang
- Institut Pasteur, Dynamics of Host-Pathogen Interactions Unit and CNRS UMR 3691, Paris, France.,Division of Molecular and Cellular Biology, National Institute of Child Health and Human Development, NIH, Bethesda, Maryland, USA
| | - Jost Enninga
- Institut Pasteur, Dynamics of Host-Pathogen Interactions Unit and CNRS UMR 3691, Paris, France
| | - Virginie Stévenin
- Institut Pasteur, Dynamics of Host-Pathogen Interactions Unit and CNRS UMR 3691, Paris, France.,Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, Netherlands.,Université Paris Diderot, Ecole doctorale BioSPC, Paris, France
| |
Collapse
|
12
|
Paone S, D'Alessandro S, Parapini S, Celani F, Tirelli V, Pourshaban M, Olivieri A. Characterization of the erythrocyte GTPase Rac1 in relation to Plasmodium falciparum invasion. Sci Rep 2020; 10:22054. [PMID: 33328606 PMCID: PMC7744522 DOI: 10.1038/s41598-020-79052-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 11/30/2020] [Indexed: 12/01/2022] Open
Abstract
Malaria is still a devastating disease with 228 million cases globally and 405,000 lethal outcomes in 2018, mainly in children under five years of age. The threat of emerging malaria strains resistant to currently available drugs has made the search for novel drug targets compelling. The process by which Plasmodium falciparum parasites invade the host cell has been widely studied, but only a few erythrocyte proteins involved in this process have been identified so far. The erythrocyte protein Rac1 is a GTPase that plays an important role in host cell invasion by many intracellular pathogens. Here we show that Rac1 is recruited in proximity to the site of parasite entry during P. falciparum invasion process and that subsequently localizes to the parasitophorous vacuole membrane. We also suggest that this GTPase may be involved in erythrocyte invasion by P. falciparum, by testing the effect of specific Rac1 inhibitory compounds. Finally, we suggest a secondary role of the erythrocyte GTPase also in parasite intracellular development. We here characterize a new erythrocyte protein potentially involved in P. falciparum invasion of the host cell and propose the human GTPase Rac1 as a novel and promising antimalarial drug target.
Collapse
Affiliation(s)
- Silvio Paone
- Dipartimento di Malattie Infettive, Istituto Superiore di Sanità, Rome, Italy.,Dipartimento di Sanità Pubblica e Malattie Infettive, Sapienza University of Rome, Rome, Italy
| | - Sarah D'Alessandro
- Dipartimento di Scienze Biomediche, Chirurgiche e Odontoiatriche, University of Milan, Milan, Italy
| | - Silvia Parapini
- Dipartimento di Scienze Biomediche Per La Salute, University of Milan, Milan, Italy
| | - Francesco Celani
- Dipartimento di Malattie Infettive, Istituto Superiore di Sanità, Rome, Italy
| | - Valentina Tirelli
- Dipartimento di Malattie Infettive, Istituto Superiore di Sanità, Rome, Italy
| | | | - Anna Olivieri
- Dipartimento di Malattie Infettive, Istituto Superiore di Sanità, Rome, Italy.
| |
Collapse
|
13
|
Menanteau-Ledouble S, Schachner O, Lawrence ML, El-Matbouli M. Effects of siRNA silencing on the susceptibility of the fish cell line CHSE-214 to Yersinia ruckeri. Vet Res 2020; 51:45. [PMID: 32197655 PMCID: PMC7083013 DOI: 10.1186/s13567-020-00760-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Accepted: 01/12/2020] [Indexed: 01/08/2023] Open
Abstract
Yersinia ruckeri is a facultative intracellular enterobacterium mostly known as the causative agent of enteric redmouth disease in salmonid fish. In the present study, we applied RNA inhibition to silence twenty pre-selected genes on the genome of a fish cell line (CHSE-214) followed by a gentamicin assay to quantify the effect of silencing on the cells’ susceptibility to infection and found that silencing of 18 out of 20 genes significantly reduced the number of Y. ruckeri recovered. These findings improve our understanding of the infection process by Y. ruckeri and of the interactions between this bacterial pathogen and host cells.
Collapse
Affiliation(s)
| | - Oskar Schachner
- Clinical Division of Fish Medicine, University of Veterinary Medicine, Vienna, Austria
| | - Mark L Lawrence
- Feed the Future Fish Innovation Lab for Fish, Mississippi State, MS, USA
| | - Mansour El-Matbouli
- Clinical Division of Fish Medicine, University of Veterinary Medicine, Vienna, Austria
| |
Collapse
|
14
|
|
15
|
Wu M, Chang Y, Hu H, Mu R, Zhang Y, Qin X, Duan X, Li W, Tu H, Zhang W, Wang G, Han Q, Li A, Zhou T, Iwai K, Zhang X, Li H. LUBAC controls chromosome alignment by targeting CENP-E to attached kinetochores. Nat Commun 2019; 10:273. [PMID: 30655516 PMCID: PMC6336796 DOI: 10.1038/s41467-018-08043-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Accepted: 12/07/2018] [Indexed: 11/30/2022] Open
Abstract
Faithful chromosome segregation requires proper chromosome congression at prometaphase and dynamic maintenance of the aligned chromosomes at metaphase. Chromosome missegregation can result in aneuploidy, birth defects and cancer. The kinetochore-bound KMN network and the kinesin motor CENP-E are critical for kinetochore-microtubule attachment and chromosome stability. The linear ubiquitin chain assembly complex (LUBAC) attaches linear ubiquitin chains to substrates, with well-established roles in immune response. Here, we identify LUBAC as a key player of chromosome alignment during mitosis. LUBAC catalyzes linear ubiquitination of the kinetochore motor CENP-E, which is specifically required for the localization of CENP-E at attached kinetochores, but not unattached ones. KNL1 acts as a receptor of linear ubiquitin chains to anchor CENP-E at attached kinetochores in prometaphase and metaphase. Thus, linear ubiquitination promotes chromosome congression and dynamic chromosome alignment by coupling the dynamic kinetochore microtubule receptor CENP-E to the static one, the KMN network. During cell division, faithful chromosome segregation requires proper chromosome congression and dynamic maintenance of the aligned chromosomes. Here, the authors find that LUBAC promotes dynamic chromosome congression and alignment by targeting kinetochore motor CENP-E to the KMN network.
Collapse
Affiliation(s)
- Min Wu
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, 100850, Beijing, China
| | - Yan Chang
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, 100850, Beijing, China
| | - Huaibin Hu
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, 100850, Beijing, China
| | - Rui Mu
- Department of Radiation Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China
| | - Yucheng Zhang
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, 100850, Beijing, China
| | - Xuanhe Qin
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, 100850, Beijing, China
| | - Xiaotao Duan
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, 100850, Beijing, China
| | - Weihua Li
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, 100850, Beijing, China
| | - Haiqing Tu
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, 100850, Beijing, China
| | - Weina Zhang
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, 100850, Beijing, China
| | - Guang Wang
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, 100850, Beijing, China
| | - Qiuying Han
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, 100850, Beijing, China
| | - Ailing Li
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, 100850, Beijing, China
| | - Tao Zhou
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, 100850, Beijing, China
| | - Kazuhiro Iwai
- Department of Molecular and Cellular Physiology, Graduate School of Medicine, Kyoto University, Yoshida-konoe-cho, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Xuemin Zhang
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, 100850, Beijing, China.
| | - Huiyan Li
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, 100850, Beijing, China. .,School of Basic Medical Sciences, Fudan University, 200032, Shanghai, China.
| |
Collapse
|
16
|
Lapaquette P, Fritah S, Lhocine N, Andrieux A, Nigro G, Mounier J, Sansonetti P, Dejean A. Shigella entry unveils a calcium/calpain-dependent mechanism for inhibiting sumoylation. eLife 2017; 6:27444. [PMID: 29231810 PMCID: PMC5745084 DOI: 10.7554/elife.27444] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Accepted: 12/11/2017] [Indexed: 12/28/2022] Open
Abstract
Disruption of the sumoylation/desumoylation equilibrium is associated with several disease states such as cancer and infections, however the mechanisms regulating the global SUMO balance remain poorly defined. Here, we show that infection by Shigella flexneri, the causative agent of human bacillary dysentery, switches off host sumoylation during epithelial cell infection in vitro and in vivo and that this effect is mainly mediated by a calcium/calpain-induced cleavage of the SUMO E1 enzyme SAE2, thus leading to sumoylation inhibition. Furthermore, we describe a mechanism by which Shigella promotes its own invasion by altering the sumoylation state of RhoGDIα, a master negative regulator of RhoGTPase activity and actin polymerization. Together, our data suggest that SUMO modification is essential to restrain pathogenic bacterial entry by limiting cytoskeletal rearrangement induced by bacterial effectors. Moreover, these findings identify calcium-activated calpains as powerful modulators of cellular sumoylation levels with potentially broad implications in several physiological and pathological situations.
Collapse
Affiliation(s)
- Pierre Lapaquette
- Nuclear Organization and Oncogenesis Unit, Institut Pasteur, Paris, France.,INSERM, U993, Paris, France
| | - Sabrina Fritah
- Nuclear Organization and Oncogenesis Unit, Institut Pasteur, Paris, France.,INSERM, U993, Paris, France
| | - Nouara Lhocine
- Unité de Pathogénie Microbienne Moléculaire, Institut Pasteur, Paris, France.,INSERM, U786, Paris, France
| | - Alexandra Andrieux
- Nuclear Organization and Oncogenesis Unit, Institut Pasteur, Paris, France.,INSERM, U993, Paris, France
| | - Giulia Nigro
- Unité de Pathogénie Microbienne Moléculaire, Institut Pasteur, Paris, France.,INSERM, U786, Paris, France
| | - Joëlle Mounier
- Unité de Pathogénie Microbienne Moléculaire, Institut Pasteur, Paris, France.,INSERM, U786, Paris, France
| | - Philippe Sansonetti
- Unité de Pathogénie Microbienne Moléculaire, Institut Pasteur, Paris, France.,INSERM, U786, Paris, France
| | - Anne Dejean
- Nuclear Organization and Oncogenesis Unit, Institut Pasteur, Paris, France.,INSERM, U993, Paris, France
| |
Collapse
|
17
|
A systematic exploration of the interactions between bacterial effector proteins and host cell membranes. Nat Commun 2017; 8:532. [PMID: 28912547 PMCID: PMC5599653 DOI: 10.1038/s41467-017-00700-7] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 07/20/2017] [Indexed: 01/08/2023] Open
Abstract
Membrane-bound organelles serve as platforms for the assembly of multi-protein complexes that function as hubs of signal transduction in eukaryotic cells. Microbial pathogens have evolved virulence factors that reprogram these host signaling responses, but the underlying molecular mechanisms are poorly understood. Here we test the ability of ~200 type III and type IV effector proteins from six Gram-negative bacterial species to interact with the eukaryotic plasma membrane and intracellular organelles. We show that over 30% of the effectors localize to yeast and mammalian cell membranes, including a subset of previously uncharacterized Legionella effectors that appear to be able to regulate yeast vacuolar fusion. A combined genetic, cellular, and biochemical approach supports that some of the tested bacterial effectors can bind to membrane phospholipids and may regulate membrane trafficking. Finally, we show that the type III effector IpgB1 from Shigella flexneri may bind to acidic phospholipids and regulate actin filament dynamics.Microbial pathogens secrete effector proteins into host cells to affect cellular functions. Here, the authors use a yeast-based screen to study around 200 effectors from six bacterial species, showing that over 30% of them interact with the eukaryotic plasma membrane or intracellular organelles.
Collapse
|
18
|
Aktories K, Schwan C, Lang AE. ADP-Ribosylation and Cross-Linking of Actin by Bacterial Protein Toxins. Handb Exp Pharmacol 2017; 235:179-206. [PMID: 27316913 DOI: 10.1007/164_2016_26] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Actin and the actin cytoskeleton play fundamental roles in host-pathogen interactions. Proper function of the actin cytoskeleton is crucial for innate and acquired immune defense. Bacterial toxins attack the actin cytoskeleton by targeting regulators of actin. Moreover, actin is directly modified by various bacterial protein toxins and effectors, which cause ADP-ribosylation or cross-linking of actin. Modification of actin can result in inhibition or stimulation of actin polymerization. Toxins, acting directly on actin, are reviewed.
Collapse
Affiliation(s)
- Klaus Aktories
- Institute for Experimental and Clinical Pharmacology and Toxicology, Albert-Ludwigs-Universität Freiburg, Freiburg, 79104, Germany. .,Freiburg Institute of Advanced Studies (FRIAS), Albert-Ludwigs-Universität Freiburg, Freiburg, 79104, Germany.
| | - Carsten Schwan
- Institute for Experimental and Clinical Pharmacology and Toxicology, Albert-Ludwigs-Universität Freiburg, Freiburg, 79104, Germany
| | - Alexander E Lang
- Institute for Experimental and Clinical Pharmacology and Toxicology, Albert-Ludwigs-Universität Freiburg, Freiburg, 79104, Germany
| |
Collapse
|
19
|
Mattock E, Blocker AJ. How Do the Virulence Factors of Shigella Work Together to Cause Disease? Front Cell Infect Microbiol 2017; 7:64. [PMID: 28393050 PMCID: PMC5364150 DOI: 10.3389/fcimb.2017.00064] [Citation(s) in RCA: 147] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Accepted: 02/21/2017] [Indexed: 01/01/2023] Open
Abstract
Shigella is the major cause of bacillary dysentery world-wide. It is divided into four species, named S. flexneri, S. sonnei, S. dysenteriae, and S. boydii, which are distinct genomically and in their ability to cause disease. Shigellosis, the clinical presentation of Shigella infection, is characterized by watery diarrhea, abdominal cramps, and fever. Shigella's ability to cause disease has been attributed to virulence factors, which are encoded on chromosomal pathogenicity islands and the virulence plasmid. However, information on these virulence factors is not often brought together to create a detailed picture of infection, and how this translates into shigellosis symptoms. Firstly, Shigella secretes virulence factors that induce severe inflammation and mediate enterotoxic effects on the colon, producing the classic watery diarrhea seen early in infection. Secondly, Shigella injects virulence effectors into epithelial cells via its Type III Secretion System to subvert the host cell structure and function. This allows invasion of epithelial cells, establishing a replicative niche, and causes erratic destruction of the colonic epithelium. Thirdly, Shigella produces effectors to down-regulate inflammation and the innate immune response. This promotes infection and limits the adaptive immune response, causing the host to remain partially susceptible to re-infection. Combinations of these virulence factors may contribute to the different symptoms and infection capabilities of the diverse Shigella species, in addition to distinct transmission patterns. Further investigation of the dominant species causing disease, using whole-genome sequencing and genotyping, will allow comparison and identification of crucial virulence factors and may contribute to the production of a pan-Shigella vaccine.
Collapse
Affiliation(s)
- Emily Mattock
- Faculty of Biomedical Sciences, Schools of Cellular and Molecular Medicine and Biochemistry, University of Bristol Bristol, UK
| | - Ariel J Blocker
- Faculty of Biomedical Sciences, Schools of Cellular and Molecular Medicine and Biochemistry, University of Bristol Bristol, UK
| |
Collapse
|
20
|
Manipulation of host membranes by the bacterial pathogens Listeria, Francisella, Shigella and Yersinia. Semin Cell Dev Biol 2016; 60:155-167. [PMID: 27448494 PMCID: PMC7082150 DOI: 10.1016/j.semcdb.2016.07.019] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Revised: 07/15/2016] [Accepted: 07/18/2016] [Indexed: 01/07/2023]
Abstract
Bacterial pathogens display an impressive arsenal of molecular mechanisms that allow survival in diverse host niches. Subversion of plasma membrane and cytoskeletal functions are common themes associated to infection by both extracellular and intracellular pathogens. Moreover, intracellular pathogens modify the structure/stability of their membrane-bound compartments and escape degradation from phagocytic or autophagic pathways. Here, we review the manipulation of host membranes by Listeria monocytogenes, Francisella tularensis, Shigella flexneri and Yersinia spp. These four bacterial model pathogens exemplify generalized strategies as well as specific features observed during bacterial infection processes.
Collapse
|
21
|
Weiner A, Mellouk N, Lopez-Montero N, Chang YY, Souque C, Schmitt C, Enninga J. Macropinosomes are Key Players in Early Shigella Invasion and Vacuolar Escape in Epithelial Cells. PLoS Pathog 2016; 12:e1005602. [PMID: 27182929 PMCID: PMC4868309 DOI: 10.1371/journal.ppat.1005602] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Accepted: 04/05/2016] [Indexed: 01/30/2023] Open
Abstract
Intracellular pathogens include all viruses, many bacteria and parasites capable of invading and surviving within host cells. Key to survival is the subversion of host cell pathways by the pathogen for the purpose of propagation and evading the immune system. The intracellular bacterium Shigella flexneri, the causative agent of bacillary dysentery, invades host cells in a vacuole that is subsequently ruptured to allow growth of the pathogen within the host cytoplasm. S. flexneri invasion has been classically described as a macropinocytosis-like process, however the underlying details and the role of macropinosomes in the intracellular bacterial lifestyle have remained elusive. We applied dynamic imaging and advanced large volume correlative light electron microscopy (CLEM) to study the highly transient events of S. flexneri's early invasion into host epithelial cells and elucidate some of its fundamental features. First, we demonstrate a clear distinction between two compartments formed during the first step of invasion: the bacterial containing vacuole and surrounding macropinosomes, often considered identical. Next, we report a functional link between macropinosomes and the process of vacuolar rupture, demonstrating that rupture timing is dependent on the availability of macropinosomes as well as the activity of the small GTPase Rab11 recruited directly to macropinosomes. We go on to reveal that the bacterial containing vacuole and macropinosomes come into direct contact at the onset of vacuolar rupture. Finally, we demonstrate that S. flexneri does not subvert pre-existing host endocytic vesicles during the invasion steps leading to vacuolar rupture, and propose that macropinosomes are the major compartment involved in these events. These results provide the basis for a new model of the early steps of S. flexneri epithelial cell invasion, establishing a different view of the enigmatic process of cytoplasmic access by invasive bacterial pathogens.
Collapse
Affiliation(s)
- Allon Weiner
- Institut Pasteur, Dynamics of Host-Pathogen interactions Unit, Paris, France
- * E-mail: (AW); (JE)
| | - Nora Mellouk
- Institut Pasteur, Dynamics of Host-Pathogen interactions Unit, Paris, France
| | | | - Yuen-Yan Chang
- Institut Pasteur, Dynamics of Host-Pathogen interactions Unit, Paris, France
| | - Célia Souque
- Institut Pasteur, Dynamics of Host-Pathogen interactions Unit, Paris, France
| | | | - Jost Enninga
- Institut Pasteur, Dynamics of Host-Pathogen interactions Unit, Paris, France
- * E-mail: (AW); (JE)
| |
Collapse
|
22
|
Stradal TEB, Costa SCP. Type III Secreted Virulence Factors Manipulating Signaling to Actin Dynamics. Curr Top Microbiol Immunol 2016; 399:175-199. [PMID: 27744505 DOI: 10.1007/82_2016_35] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
A key aspect of bacterial pathogenesis is the colonization and persistence within the host and, later on, its dissemination to new niches. During evolution, bacteria developed a myriad of virulence mechanisms to usurp the host's sophisticated defense mechanisms in order to establish their colonization niche. Elucidation of the highly dynamic and complex interactions between host and pathogens remains an important field of study. Here, we highlight the conserved manipulation of the actin cytoskeleton by some Gram-negative gastrointestinal pathogens, addressing the role of type III secreted bacterial GEFs at the different steps of pathogenesis. As a final topic, we review cytoskeleton dynamics induced by EPEC/EHEC strains for pedestal formation.
Collapse
Affiliation(s)
- Theresia E B Stradal
- Department of Cell Biology, Helmholtz Centre for Infection Research, Inhoffenstr. 7, 38124, Baunschweig, Germany.
| | - Sonia C P Costa
- Department of Cell Biology, Helmholtz Centre for Infection Research, Inhoffenstr. 7, 38124, Baunschweig, Germany
| |
Collapse
|
23
|
Reeves AZ, Spears WE, Du J, Tan KY, Wagers AJ, Lesser CF. Engineering Escherichia coli into a protein delivery system for mammalian cells. ACS Synth Biol 2015; 4:644-54. [PMID: 25853840 PMCID: PMC4487226 DOI: 10.1021/acssynbio.5b00002] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Many Gram-negative pathogens encode type 3 secretion systems, sophisticated nanomachines that deliver proteins directly into the cytoplasm of mammalian cells. These systems present attractive opportunities for therapeutic protein delivery applications; however, their utility has been limited by their inherent pathogenicity. Here, we report the reengineering of a laboratory strain of Escherichia coli with a tunable type 3 secretion system that can efficiently deliver heterologous proteins into mammalian cells, thereby circumventing the need for virulence attenuation. We first introduced a 31 kB region of Shigella flexneri DNA that encodes all of the information needed to form the secretion nanomachine onto a plasmid that can be directly propagated within E. coli or integrated into the E. coli chromosome. To provide flexible control over type 3 secretion and protein delivery, we generated plasmids expressing master regulators of the type 3 system from either constitutive or inducible promoters. We then constructed a Gateway-compatible plasmid library of type 3 secretion sequences to enable rapid screening and identification of sequences that do not perturb function when fused to heterologous protein substrates and optimized their delivery into mammalian cells. Combining these elements, we found that coordinated expression of the type 3 secretion system and modified target protein substrates produces a nonpathogenic strain that expresses, secretes, and delivers heterologous proteins into mammalian cells. This reengineered system thus provides a highly flexible protein delivery platform with potential for future therapeutic applications.
Collapse
Affiliation(s)
- Analise Z. Reeves
- Department
of Medicine, Division of Infectious Diseases, Massachusetts General Hospital, Cambridge, Massachusetts 02139, United States
- Department
of Microbiology and Immunobiology, Harvard Medical School, Boston, Massachusetts 02138, United States
| | - William E. Spears
- Department
of Medicine, Division of Infectious Diseases, Massachusetts General Hospital, Cambridge, Massachusetts 02139, United States
| | - Juan Du
- Department
of Medicine, Division of Infectious Diseases, Massachusetts General Hospital, Cambridge, Massachusetts 02139, United States
- Department
of Microbiology and Immunobiology, Harvard Medical School, Boston, Massachusetts 02138, United States
| | - Kah Yong Tan
- Howard
Hughes Medical Institute and Department of Stem Cell and Regenerative
Biology, Harvard University, Cambridge, Massachusetts 02138, United States
- Harvard Stem Cell Institute, Cambridge, Massachusetts 02138, United States
- Joslin Diabetes Center, Boston, Massachusetts 02215, United States
| | - Amy J. Wagers
- Howard
Hughes Medical Institute and Department of Stem Cell and Regenerative
Biology, Harvard University, Cambridge, Massachusetts 02138, United States
- Harvard Stem Cell Institute, Cambridge, Massachusetts 02138, United States
- Joslin Diabetes Center, Boston, Massachusetts 02215, United States
| | - Cammie F. Lesser
- Department
of Medicine, Division of Infectious Diseases, Massachusetts General Hospital, Cambridge, Massachusetts 02139, United States
- Department
of Microbiology and Immunobiology, Harvard Medical School, Boston, Massachusetts 02138, United States
- Harvard Stem Cell Institute, Cambridge, Massachusetts 02138, United States
| |
Collapse
|
24
|
Kuehl CJ, Dragoi AM, Agaisse H. The Shigella flexneri type 3 secretion system is required for tyrosine kinase-dependent protrusion resolution, and vacuole escape during bacterial dissemination. PLoS One 2014; 9:e112738. [PMID: 25405985 PMCID: PMC4236203 DOI: 10.1371/journal.pone.0112738] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Accepted: 10/14/2014] [Indexed: 01/29/2023] Open
Abstract
Shigella flexneri is a human pathogen that triggers its own entry into intestinal cells and escapes primary vacuoles to gain access to the cytosolic compartment. As cytosolic and motile bacteria encounter the cell cortex, they spread from cell to cell through formation of membrane protrusions that resolve into secondary vacuoles in adjacent cells. Here, we examined the roles of the Type 3 Secretion System (T3SS) in S. flexneri dissemination in HT-29 intestinal cells infected with the serotype 2a strain 2457T. We generated a 2457T strain defective in the expression of MxiG, a central component of the T3SS needle apparatus. As expected, the ΔmxiG strain was severely affected in its ability to invade HT-29 cells, and expression of mxiG under the control of an arabinose inducible expression system (ΔmxiG/pmxiG) restored full infectivity. In this experimental system, removal of the inducer after the invasion steps (ΔmxiG/pmxiG (Ara withdrawal)) led to normal actin-based motility in the cytosol of HT-29 cells. However, the time spent in protrusions until vacuole formation was significantly increased. Moreover, the number of formed protrusions that failed to resolve into vacuoles was also increased. Accordingly, the ΔmxiG/pmxiG (Ara withdrawal) strain failed to trigger tyrosine phosphorylation in membrane protrusions, a signaling event that is required for the resolution of protrusions into vacuoles. Finally, the ΔmxiG/pmxiG (Ara withdrawal) strain failed to escape from the formed secondary vacuoles, as previously reported in non-intestinal cells. Thus, the T3SS system displays multiple roles in S. flexneri dissemination in intestinal cells, including the tyrosine kinase signaling-dependent resolution of membrane protrusions into secondary vacuoles, and the escape from the formed secondary vacuoles.
Collapse
Affiliation(s)
- Carole J. Kuehl
- Department of Microbial Pathogenesis, Yale School of Medicine, Boyer Center for Molecular Medicine, New Haven, Connecticut, United States of America
| | - Ana-Maria Dragoi
- Department of Microbial Pathogenesis, Yale School of Medicine, Boyer Center for Molecular Medicine, New Haven, Connecticut, United States of America
| | - Hervé Agaisse
- Department of Microbial Pathogenesis, Yale School of Medicine, Boyer Center for Molecular Medicine, New Haven, Connecticut, United States of America
| |
Collapse
|
25
|
Molecular mechanisms of host cytoskeletal rearrangements by Shigella invasins. Int J Mol Sci 2014; 15:18253-66. [PMID: 25310650 PMCID: PMC4227214 DOI: 10.3390/ijms151018253] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Revised: 09/23/2014] [Accepted: 09/25/2014] [Indexed: 11/16/2022] Open
Abstract
Pathogen-induced reorganization of the host cell cytoskeleton is a common strategy utilized in host cell invasion by many facultative intracellular bacteria, such as Shigella, Listeria, enteroinvasive E. coli and Salmonella. Shigella is an enteroinvasive intracellular pathogen that preferentially infects human epithelial cells and causes bacillary dysentery. Invasion of Shigella into intestinal epithelial cells requires extensive remodeling of the actin cytoskeleton with the aid of pathogenic effector proteins injected into the host cell by the activity of the type III secretion system. These so-called Shigella invasins, including IpaA, IpaC, IpgB1, IpgB2 and IpgD, modulate the actin-regulatory system in a concerted manner to guarantee efficient entry of the bacteria into host cells.
Collapse
|
26
|
Popoff MR. Bacterial factors exploit eukaryotic Rho GTPase signaling cascades to promote invasion and proliferation within their host. Small GTPases 2014; 5:28209. [PMID: 25203748 DOI: 10.4161/sgtp.28209] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Actin cytoskeleton is a main target of many bacterial pathogens. Among the multiple regulation steps of the actin cytoskeleton, bacterial factors interact preferentially with RhoGTPases. Pathogens secrete either toxins which diffuse in the surrounding environment, or directly inject virulence factors into target cells. Bacterial toxins, which interfere with RhoGTPases, and to some extent with RasGTPases, catalyze a covalent modification (ADPribosylation, glucosylation, deamidation, adenylation, proteolysis) blocking these molecules in their active or inactive state, resulting in alteration of epithelial and/or endothelial barriers, which contributes to dissemination of bacteria in the host. Injected bacterial virulence factors preferentially manipulate the RhoGTPase signaling cascade by mimicry of eukaryotic regulatory proteins leading to local actin cytoskeleton rearrangement, which mediates bacterial entry into host cells or in contrast escape to phagocytosis and immune defense. Invasive bacteria can also manipulate RhoGTPase signaling through recognition and stimulation of cell surface receptor(s). Changes in RhoGTPase activation state is sensed by the innate immunity pathways and allows the host cell to adapt an appropriate defense response.
Collapse
Affiliation(s)
- Michel R Popoff
- Unité des Bactéries anaérobies et Toxines; Institut Pasteur; Paris, France
| |
Collapse
|
27
|
Costa SCP, Lesser CF. A multifunctional region of the Shigella type 3 effector IpgB1 is important for secretion from bacteria and membrane targeting in eukaryotic cells. PLoS One 2014; 9:e93461. [PMID: 24718571 PMCID: PMC3981709 DOI: 10.1371/journal.pone.0093461] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2014] [Accepted: 03/05/2014] [Indexed: 11/29/2022] Open
Abstract
Type 3 secretion systems are complex nanomachines used by many Gram–negative bacteria to deliver tens of proteins (effectors) directly into host cells. Once delivered into host cells, effectors often target to specific cellular loci where they usurp host cell processes to their advantage. Here, using the yeast model system, we identify the membrane localization domain (MLD) of IpgB1, a stretch of 20 amino acids enriched for hydrophobic residues essential for the targeting of this effector to the plasma membrane. Embedded within these residues are ten that define the IpgB1 chaperone-binding domain for Spa15. As observed with dedicated class IA chaperones that mask hydrophobic MLDs, Spa15, a class IB chaperone, promotes IpgB1 stability by binding this hydrophobic region. However, despite being stable, an IpgB1 allele that lacks the MLD is not recognized as a secreted substrate. Similarly, deletion of the chaperone binding domains of IpgB1 and three additional Spa15-dependent effectors result in alleles that are no longer recognized as secreted substrates despite the presence of intact N-terminal secretion signal sequences. This is in contrast with MLD-containing effectors that bind class IA dedicated chaperones, as deletion of the MLD of these effectors alleviates the chaperone requirement for secretion. These observations indicate that at least for substrates of class IB chaperones, the chaperone-effector complex plays a major role in defining type 3 secreted proteins and highlight how a single region of an effector can play important roles both within prokaryotic and eukaryotic cells.
Collapse
Affiliation(s)
- Sonia C. P. Costa
- Department of Medicine (Microbiology and Immunobiology), Division of Infectious Diseases, Massachusetts General Hospital and Harvard Medical School, Cambridge, Massachusetts, United States of America
| | - Cammie F. Lesser
- Department of Medicine (Microbiology and Immunobiology), Division of Infectious Diseases, Massachusetts General Hospital and Harvard Medical School, Cambridge, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
28
|
Edouard S, Raoult D. Use of the plaque assay for testing the antibiotic susceptibility of intracellular bacteria. Future Microbiol 2014; 8:1301-16. [PMID: 24059920 DOI: 10.2217/fmb.13.98] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The plaque assay was first described for titration of bacterial inoculums and clonal isolation, and was later adapted for testing antibiotics susceptibility and to study virulence factors and motility of bacteria. Over time, the sensitivity and reproducibility of the technique has been improved. Usually, the number of plaques is counted; however, the recent development of informatics tools has stimulated interest in the quantification of plaque size. Owing to this new approach, the plaque assay has been used to characterize the host cell response when infected cells are treated with antimicrobial agents. It was found that statins prevented cell lesions following rickettsial infection; in other studies, some antibiotics were found to cause apoptosis of host cells, suggesting a toxic activity. Here, we present an overview of the plaque assay as it has been used to investigate intracellular bacteria.
Collapse
Affiliation(s)
- Sophie Edouard
- Aix Marseille Université, URMITE, UM63, CNRS 7278, IRD198, Inserm 1, 95, 13005 Marseille, France
| | | |
Collapse
|
29
|
Schmutz C, Ahrné E, Kasper CA, Tschon T, Sorg I, Dreier RF, Schmidt A, Arrieumerlou C. Systems-level overview of host protein phosphorylation during Shigella flexneri infection revealed by phosphoproteomics. Mol Cell Proteomics 2013; 12:2952-68. [PMID: 23828894 PMCID: PMC3790303 DOI: 10.1074/mcp.m113.029918] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2013] [Revised: 06/14/2013] [Indexed: 01/01/2023] Open
Abstract
The enteroinvasive bacterium Shigella flexneri invades the intestinal epithelium of humans. During infection, several injected effector proteins promote bacterial internalization, and interfere with multiple host cell responses. To obtain a systems-level overview of host signaling during infection, we analyzed the global dynamics of protein phosphorylation by liquid chromatography-tandem MS and identified several hundred of proteins undergoing a phosphorylation change during the first hours of infection. Functional bioinformatic analysis revealed that they were mostly related to the cytoskeleton, transcription, signal transduction, and cell cycle. Fuzzy c-means clustering identified six temporal profiles of phosphorylation and a functional module composed of ATM-phosphorylated proteins related to genotoxic stress. Pathway enrichment analysis defined mTOR as the most overrepresented pathway. We showed that mTOR complex 1 and 2 were required for S6 kinase and AKT activation, respectively. Comparison with a published phosphoproteome of Salmonella typhimurium-infected cells revealed a large subset of coregulated phosphoproteins. Finally, we showed that S. flexneri effector OspF affected the phosphorylation of several hundred proteins, thereby demonstrating the wide-reaching impact of a single bacterial effector on the host signaling network.
Collapse
Affiliation(s)
- Christoph Schmutz
- From the ‡Biozentrum, University of Basel, Klingelbergstrasse 50/70, 4056 Basel, Switzerland
| | - Erik Ahrné
- From the ‡Biozentrum, University of Basel, Klingelbergstrasse 50/70, 4056 Basel, Switzerland
| | - Christoph A. Kasper
- From the ‡Biozentrum, University of Basel, Klingelbergstrasse 50/70, 4056 Basel, Switzerland
| | - Therese Tschon
- From the ‡Biozentrum, University of Basel, Klingelbergstrasse 50/70, 4056 Basel, Switzerland
| | - Isabel Sorg
- From the ‡Biozentrum, University of Basel, Klingelbergstrasse 50/70, 4056 Basel, Switzerland
| | - Roland F. Dreier
- From the ‡Biozentrum, University of Basel, Klingelbergstrasse 50/70, 4056 Basel, Switzerland
| | - Alexander Schmidt
- From the ‡Biozentrum, University of Basel, Klingelbergstrasse 50/70, 4056 Basel, Switzerland
| | - Cécile Arrieumerlou
- From the ‡Biozentrum, University of Basel, Klingelbergstrasse 50/70, 4056 Basel, Switzerland
| |
Collapse
|
30
|
The Shigella type three secretion system effector OspG directly and specifically binds to host ubiquitin for activation. PLoS One 2013; 8:e57558. [PMID: 23469023 PMCID: PMC3585378 DOI: 10.1371/journal.pone.0057558] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2012] [Accepted: 01/23/2013] [Indexed: 11/19/2022] Open
Abstract
The genus Shigella infects human gut epithelial cells to cause diarrhea and gastrointestinal disorders. Like many other Gram-negative bacterial pathogens, the virulence of Shigella spp. relies on a conserved type three secretion system that delivers a handful of effector proteins into host cells to manipulate various host cell physiology. However, many of the Shigella type III effectors remain functionally uncharacterized. Here we observe that OspG, one of the Shigella effectors, interacted with ubiquitin conjugates and poly-ubiquitin chains of either K48 or K63 linkage in eukaryotic host cells. Purified OspG protein formed a stable complex with ubiquitin but showed no interactions with other ubiquitin-like proteins. OspG binding to ubiquitin required the carboxyl terminal helical region in OspG and the canonical I44-centered hydrophobic surface in ubiquitin. OspG and OspG-homologous effectors, NleH1/2 from enteropathogenic E coli (EPEC), contain sub-domains I-VII of eukaryotic serine/threonine kinase. GST-tagged OspG and NleH1/2 could undergo autophosphorylation, the former of which was significantly stimulated by ubiquitin binding. Ubiquitin binding was also required for OspG functioning in attenuating host NF-κB signaling. Our data illustrate a new mechanism that bacterial pathogen like Shigella exploits ubiquitin binding to activate its secreted virulence effector for its functioning in host eukaryotic cells.
Collapse
|
31
|
Actin cytoskeleton manipulation by effector proteins secreted by diarrheagenic Escherichia coli pathotypes. BIOMED RESEARCH INTERNATIONAL 2012; 2013:374395. [PMID: 23509714 PMCID: PMC3591105 DOI: 10.1155/2013/374395] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/24/2012] [Accepted: 10/22/2012] [Indexed: 11/18/2022]
Abstract
The actin cytoskeleton is a dynamic structure necessary for cell and tissue organization, including the maintenance of epithelial barriers. Disruption of the epithelial barrier coincides with alterations of the actin cytoskeleton in several disease states. These disruptions primarily affect the paracellular space, which is normally regulated by tight junctions. Thereby, the actin cytoskeleton is a common and recurring target of bacterial virulence factors. In order to manipulate the actin cytoskeleton, bacteria secrete and inject toxins and effectors to hijack the host cell machinery, which interferes with host-cell pathways and with a number of actin binding proteins. An interesting model to study actin manipulation by bacterial effectors is Escherichia coli since due to its genome plasticity it has acquired diverse genetic mobile elements, which allow having different E. coli varieties in one bacterial species. These E. coli pathotypes, including intracellular and extracellular bacteria, interact with epithelial cells, and their interactions depend on a specific combination of virulence factors. In this paper we focus on E. coli effectors that mimic host cell proteins to manipulate the actin cytoskeleton. The study of bacterial effector-cytoskeleton interaction will contribute not only to the comprehension of the molecular causes of infectious diseases but also to increase our knowledge of cell biology.
Collapse
|
32
|
Sasakawa C. [Pathogenesis of Shigella: the study of bacteria-host interplay at the intestinal mucosal barriers]. Nihon Saikingaku Zasshi 2012; 67:257-68. [PMID: 23269180 DOI: 10.3412/jsb.67.257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Shigella are capable of invading and colonizing the intestinal epithelium, which results in strong inflammatory colitis. Shigella deliver a subset of effectors via the type III secretion system during infection into host cells. The delivered effectors mimic and usurp the host cellular functions, and modulate host cell signaling and immune response, thus playing pivotal roles in promoting the bacterial infection and circumventing host defense systems. This article overviews the characteristics of pathogenesis of Shigella, and highlights current topics related to the roles of the effectors in promoting bacterial infection.
Collapse
Affiliation(s)
- Chihiro Sasakawa
- Nippon Institute for Biological Science, 9-2221-1 Shin-machi, Ome, Tokyo 198-0024, Japan
| |
Collapse
|
33
|
Hierarchies of host factor dynamics at the entry site of Shigella flexneri during host cell invasion. Infect Immun 2012; 80:2548-57. [PMID: 22526677 DOI: 10.1128/iai.06391-11] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Shigella flexneri, the causative agent of bacillary dysentery, induces massive cytoskeletal rearrangement, resulting in its entry into nonphagocytic epithelial cells. The bacterium-engulfing membrane ruffles are formed by polymerizing actin, a process activated through injected bacterial effectors that target host small GTPases and tyrosine kinases. Once inside the host cell, S. flexneri escapes from the endocytic vacuole within minutes to move intra- and intercellularly. We quantified the fluorescence signals from fluorescently tagged host factors that are recruited to the site of pathogen entry and vacuolar escape. Quantitative time lapse fluorescence imaging revealed simultaneous recruitment of polymerizing actin, small GTPases of the Rho family, and tyrosine kinases. In contrast, we found that actin surrounding the vacuole containing bacteria dispersed first from the disassembling membranes, whereas other host factors remained colocalized with the membrane remnants. Furthermore, we found that the disassembly of the membrane remnants took place rapidly, within minutes after bacterial release into the cytoplasm. Superresolution visualization of galectin 3 through photoactivated localization microscopy characterized these remnants as small, specular, patchy structures between 30 and 300 nm in diameter. Using our experimental setup to track the time course of infection, we identified the S. flexneri effector IpgB1 as an accelerator of the infection pace, specifically targeting the entry step, but not vacuolar progression or escape. Together, our studies show that bacterial entry into host cells follows precise kinetics and that this time course can be targeted by the pathogen.
Collapse
|
34
|
Abstract
Small molecular weight GTPases are master regulators of eukaryotic signalling, making them prime targets for bacterial virulence factors. Here, we review the recent advances made in understanding how bacterial type III secreted effector proteins directly activate GTPase signalling cascades. Specifically we focus on the SopE/WxxxE family of effectors that functionally mimic guanine nucleotide exchange factors (GEFs): the endogenous activators of Rho-family GTPases. Recent structural and biochemical studies have provided keen insight into both the signalling potency and substrate specificity of bacterial GEFs. Additionally, these bacterial GEFs display fascinating cell biological properties that provide insight into both host cell physiology and infectious disease strategies.
Collapse
Affiliation(s)
- Robert C Orchard
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | | |
Collapse
|
35
|
Raymond B, Crepin VF, Collins JW, Frankel G. The WxxxE effector EspT triggers expression of immune mediators in an Erk/JNK and NF-κB-dependent manner. Cell Microbiol 2011; 13:1881-93. [PMID: 21848814 PMCID: PMC4977063 DOI: 10.1111/j.1462-5822.2011.01666.x] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Enteropathogenic Escherichia coli (EPEC), enterohaemorrhagic E. coli (EHEC) and Citrobacter rodentium colonize their respective hosts while forming attaching and effacing lesions. Their infection strategy relies on translocation of a battery of type III secretion system effectors, including Map, EspM and EspT, which belong to the WxxxE/SopE family of guanine nucleotide exchange factors. Using the C. rodentium mouse model we found that EspT triggers expression of KC and TNFα in vivo. Indeed, a growing body of evidence suggests that, in addition to subversion of actin dynamics, the SopE and the WxxxE effectors activate signalling pathways involved in immune responses. In this study we found that EspT induces expression of the pro-inflammatory mediators cyclooxygenase-2 (COX-2) an enzyme involved in production of prostaglandin E(2) (PGE2), interleukin (Il)-8 and Il-1β in U937 human macrophages by activating the nuclear factor kappa-B (NF-κB), the extracellular signal-regulated kinases 1 and 2 (Erk1/2) and c-Jun N-terminal kinase (JNK) pathways. Since EspT modulates the activation of Cdc42 and Rac1, which mediates bacterial invasion into epithelial cells, we investigated the involvement of these Rho GTPases and bacterial invasion on pro-inflammatory responses and found that (i) Rac1, but not Cdc42, is involved in EspT-induced Il-8 and Il-1β secretion and (ii) cytochalasin D inhibits EspT-induced EPEC invasion into U937 but not Il-8 or Il-1β secretion. These results suggest that while EPEC translocates a number of effectors (i.e. NleC, NleD, NleE, NleH) that inhibit inflammation, a subset of strains, which encode EspT, employ an infection strategy that also involves upregulation of immune mediators.
Collapse
Affiliation(s)
- Benoit Raymond
- Centre for Molecular Microbiology and Infection, Division of Cell and Molecular Biology, Imperial College, London SW7 2AZ, UK
| | - Valerie F. Crepin
- Centre for Molecular Microbiology and Infection, Division of Cell and Molecular Biology, Imperial College, London SW7 2AZ, UK
| | - James W. Collins
- Centre for Molecular Microbiology and Infection, Division of Cell and Molecular Biology, Imperial College, London SW7 2AZ, UK
| | - Gad Frankel
- Centre for Molecular Microbiology and Infection, Division of Cell and Molecular Biology, Imperial College, London SW7 2AZ, UK
| |
Collapse
|
36
|
Flannagan RS, Jaumouillé V, Huynh KK, Plumb JD, Downey GP, Valvano MA, Grinstein S. Burkholderia cenocepacia disrupts host cell actin cytoskeleton by inactivating Rac and Cdc42. Cell Microbiol 2011; 14:239-54. [PMID: 22023324 DOI: 10.1111/j.1462-5822.2011.01715.x] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Burkholderia cenocepacia, a member of the Burkholderia cepacia complex, is an opportunistic pathogen that causes devastating infections in patients with cystic fibrosis. The ability of B. cenocepacia to survive within host cells could contribute significantly to its virulence in immunocompromised patients. In this study, we explored the mechanisms that enable B. cenocepacia to survive inside macrophages. We found that B. cenocepacia disrupts the actin cytoskeleton of infected macrophages, drastically altering their morphology. Submembranous actin undergoes depolymerization, leading to cell retraction. The bacteria perturb actin architecture by inactivating Rho family GTPases, particularly Rac1 and Cdc42. GTPase inactivation follows internalization of viable B. cenocepacia and compromises phagocyte function: macropinocytosis and phagocytosis are markedly inhibited, likely impairing the microbicidal and antigen-presenting capability of infected macrophages. The type VI secretion system is essential for the bacteria to elicit these changes. This is the first report demonstrating inactivation of Rho family GTPases by a member of the B. cepacia complex.
Collapse
Affiliation(s)
- Ronald S Flannagan
- Program in Cell Biology, Hospital for Sick Children, Toronto, Ontario, Canada M5G 1X8
| | | | | | | | | | | | | |
Collapse
|
37
|
Abstract
Bacterial pathogens interact with host membranes to trigger a wide range of cellular processes during the course of infection. These processes include alterations to the dynamics between the plasma membrane and the actin cytoskeleton, and subversion of the membrane-associated pathways involved in vesicle trafficking. Such changes facilitate the entry and replication of the pathogen, and prevent its phagocytosis and degradation. In this Review, we describe the manipulation of host membranes by numerous bacterial effectors that target phosphoinositide metabolism, GTPase signalling and autophagy.
Collapse
|
38
|
Wölke S, Ackermann N, Heesemann J. The Yersinia enterocolitica type 3 secretion system (T3SS) as toolbox for studying the cell biological effects of bacterial Rho GTPase modulating T3SS effector proteins. Cell Microbiol 2011; 13:1339-57. [PMID: 21718421 DOI: 10.1111/j.1462-5822.2011.01623.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The bacterial effector proteins IpgB(1) and IpgB(2) of Shigella and Map of Escherichia coli activate the Rho GTPases Rac1, RhoA and Cdc42, respectively, whereas YopE and YopT of Yersinia inhibit these Rho family GTPases. We established a Yersinia toolbox which allows to study the cellular effects of these effectors in different combinations in the context of Yersinia type 3 secretion system (Ysc)-T3SS-mediated injection into HeLa cells. For this purpose hybrid proteins were constructed by fusion of YopE with the effector protein of interest. As expected, injected hybrid proteins induced membrane ruffles and Yersinia uptake for IpgB(1) , stress fibres for IpgB(2) and microspikes for Map. By co-infection experiments we could demonstrate (i) IpgB(2) -mediated and ROCK-dependent inhibition of IpgB(1) -mediated Rac1 effects, (ii) YopT-mediated suppression of IpgB(1) -induced Yersinia invasion and (iii) failure of YopE-mediated suppression of IpgB(1) -induced Yersinia invasion, presumably due to preferential inhibition of RhoG by YopE GAP function. By infecting polarized MDCK cells we could demonstrate that Map or IpgB(1) but not IpgB(2) affects cell monolayer integrity. In summary, the Yersinia toolbox is suitable to study cellular effects of effector proteins of diverse bacterial species separately or in combination in the context of bacterial T3SS-mediated injection.
Collapse
Affiliation(s)
- Stefan Wölke
- Max von Pettenkofer Institut, LMU Munich, Pettenkofer Straße 9A, 80336 Munich, Germany
| | | | | |
Collapse
|
39
|
Abstract
Many bacterial pathogens produce protein toxins to outmanoeuvre the immune system of the host. Some of these proteins target regulatory GTPases such as those belonging to the RHO family, which control the actin cytoskeleton of the host cell. In this Review, I discuss a diversity of mechanisms that are used by bacterial effectors and toxins to modulate the activity of host GTPases, with a focus on covalent modifications such as ADP-ribosylation, glucosylation, adenylylation, proteolysis, deamidation and transglutamination.
Collapse
Affiliation(s)
- Klaus Aktories
- Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Albert-Ludwigs-Universität Freiburg, Albertstr. 25, 79104 Freiburg, Germany.
| |
Collapse
|
40
|
Ki KH, Park DY, Lee SH, Kim NY, Choi BM, Noh GJ. The optimal concentration of siRNA for gene silencing in primary cultured astrocytes and microglial cells of rats. Korean J Anesthesiol 2010; 59:403-10. [PMID: 21253378 PMCID: PMC3022134 DOI: 10.4097/kjae.2010.59.6.403] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2010] [Revised: 06/28/2010] [Accepted: 07/15/2010] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Small interfering RNAs (siRNAs) have been used to knockdown specific gene expression in various cells. Astrocytes and microglial cells play a key role in fundamental central nervous system functions and in chronic neuroinflammation. The aims of this study were to determine the optimal concentration of siRNA demonstrating efficient transfection and inhibition of gene expression via RNA interference (RNAi) and lower cytotoxicity, in primary cultured astrocytes and microglial cells of rats. METHODS Astrocytes and microglial cells were isolated from the cerebral cortices of 2-day-old rats. Both the cells were transfected using transfection reagent (Lipofectamine™ 2000), and fluorescein-labeled double-stranded RNA (dsRNA) or siRNA targeting green fluorescent protein. Transfection efficiency and cytotoxicity of dsRNA, and the degrees of RNAi induced by siRNA in these cells, were evaluated at various concentrations of RNA. RESULTS Transfection efficiencies of dsRNA in both astrocytes and microglial cells were significantly higher (P < 0.05) at the concentrations of 20, 40, and 80 nM than at the concentrations of 0, 5, and 10 nM. There were no significant cytotoxicities within the applied concentrations of dsRNA (0-80 nM). The degrees of RNAi induced by siRNA were significantly higher (P < 0.05) at the concentrations of 5, 10, 20, 40, 80 nM, and 20, 40, 80 nM in astrocytes and microglial cells, respectively, compared with the control (0 nM). CONCLUSIONS The siRNA concentration of 20 nM may be appropriate to induce RNAi in both astrocytes and microglial cells, while demonstrating low cytotoxicity, high transfection efficiency, and effective RNAi.
Collapse
Affiliation(s)
- Kyeong Ho Ki
- Department of Clinical Pharmacology and Therapeutics, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | | | | | | | | | | |
Collapse
|
41
|
Lazarev VN, Borisenko GG, Shkarupeta MM, Demina IA, Serebryakova MV, Galyamina MA, Levitskiy SA, Govorun VM. The role of intracellular glutathione in the progression of Chlamydia trachomatis infection. Free Radic Biol Med 2010; 49:1947-55. [PMID: 20888409 DOI: 10.1016/j.freeradbiomed.2010.09.024] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2010] [Revised: 08/17/2010] [Accepted: 09/23/2010] [Indexed: 12/17/2022]
Abstract
The productive internalization in the host cell of Chlamydia trachomatis elementary bodies and their infectivity depends on the degree of reduction of disulfide bonds in the outer envelope of the elementary body. We have hypothesized that the reducing agent may be intracellular glutathione (GSH). Three approaches were used to modulate the intracellular GSH concentration: (1) treatment of cells with buthionine sulfoximine, which causes irreversible inhibition of GSH biosynthesis; (2) hydrogen peroxide-induced oxidation of GSH by intracellular glutathione peroxidases; and (3) treatment of cells with N-acetyl-l-cysteine (NAC), a precursor of glutathione. In the first two cases, we observed a four- to sixfold inhibition of C. trachomatis infection, whereas in NAC-treated cells we detected an increase in the size of chlamydial inclusions. Using a proteomics approach, we showed that the inhibition of chlamydial infection does not combine with alterations in protein expression patterns after cell treatment. These results suggest that GSH plays a key role in the reduction of disulfide bonds in the C. trachomatis outer envelope at an initial stage of the infection.
Collapse
|
42
|
Wilson BA, Ho M. Recent insights into Pasteurella multocida toxin and other G-protein-modulating bacterial toxins. Future Microbiol 2010; 5:1185-201. [PMID: 20722598 DOI: 10.2217/fmb.10.91] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Over the past few decades, our understanding of the bacterial protein toxins that modulate G proteins has advanced tremendously through extensive biochemical and structural analyses. This article provides an updated survey of the various toxins that target G proteins, ending with a focus on recent mechanistic insights in our understanding of the deamidating toxin family. The dermonecrotic toxin from Pasteurella multocida (PMT) was recently added to the list of toxins that disrupt G-protein signal transduction through selective deamidation of their targets. The C3 deamidase domain of PMT has no sequence similarity to the deamidase domains of the dermonecrotic toxins from Escherichia coli (cytotoxic necrotizing factor [CNF]1-3), Yersinia (CNFY) and Bordetella (dermonecrotic toxin). The structure of PMT-C3 belongs to a family of transglutaminase-like proteins, with active site Cys-His-Asp catalytic triads distinct from E. coli CNF1.
Collapse
Affiliation(s)
- Brenda A Wilson
- Department of Microbiology and Institute for Genomic Biology, University of Illinois at Urbana-Champaign, 601 S. Goodwin Ave, B128 CLSL, Urbana, IL 61801, USA.
| | | |
Collapse
|
43
|
Lillington JED, Lovett JE, Johnson S, Roversi P, Timmel CR, Lea SM. Shigella flexneri Spa15 crystal structure verified in solution by double electron electron resonance. J Mol Biol 2010; 405:427-35. [PMID: 21075116 PMCID: PMC3021122 DOI: 10.1016/j.jmb.2010.10.053] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2010] [Revised: 10/25/2010] [Accepted: 10/27/2010] [Indexed: 12/20/2022]
Abstract
Shigella flexneri Spa15 is a chaperone of the type 3 secretion system, which binds a number of effectors to ensure their stabilization prior to secretion. One of these effectors is IpgB1, a mimic of the human Ras-like Rho guanosine triphosphatase RhoG. In this study, Spa15 alone and in complex with IpgB1 has been studied by double electron electron resonance, an experiment that gives distance information showing the spacial separation of attached spin labels. This distance is explained by determining the crystal structure of the spin-labeled Spa15 where labels are seen to be buried in hydrophobic pockets. The double electron electron resonance experiment on the Spa15 complex with IpgB1 shows that IpgB1 does not bind Spa15 in the same way as is seen in the homologous Salmonella sp. chaperone:effector complex InvB:SipA.
Collapse
|
44
|
Ashida H, Ogawa M, Kim M, Suzuki S, Sanada T, Punginelli C, Mimuro H, Sasakawa C. Shigella deploy multiple countermeasures against host innate immune responses. Curr Opin Microbiol 2010; 14:16-23. [PMID: 20934372 DOI: 10.1016/j.mib.2010.08.014] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2010] [Accepted: 08/31/2010] [Indexed: 11/28/2022]
Abstract
Although the intestinal epithelium is equipped with multiple defense systems that sense bacterial components, transmit alarms to the immune system, clear the bacteria, and renew the injured epithelial lining, mucosal bacterial pathogens are capable of efficiently colonizing the intestinal epithelium, because they have evolved systems that modulate the inflammatory and immune responses of the host and exploit the harmful environments as replicative niches. In this review we highlight current topics concerning Shigella's tactics that interfere with the innate immune systems.
Collapse
Affiliation(s)
- Hiroshi Ashida
- Department of Microbiology and Immunology, Institute of Medical Science, University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Dunn JD, Valdivia RH. Uncivil engineers: Chlamydia, Salmonella and Shigella alter cytoskeleton architecture to invade epithelial cells. Future Microbiol 2010; 5:1219-32. [DOI: 10.2217/fmb.10.77] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The obligate intracellular bacterial pathogen Chlamydia trachomatis is a major cause of blindness and sexually transmitted diseases. Like the enteric pathogens Salmonella and Shigella, Chlamydia injects effector proteins into epithelial cells to initiate extensive remodeling of the actin cytoskeleton at the bacterial attachment site, which culminates in the engulfment of the bacterium by plasma membrane extensions. Numerous Salmonella and Shigella effectors promote this remodeling by activating Rho GTPases and tyrosine kinase signaling cascades and by directly manipulating actin dynamics. Recent studies indicate that similar host-cell alterations occur during Chlamydia invasion, but few effectors are known. The identification of additional Chlamydia effectors and the elucidation of their modes of function are critical steps towards an understanding of how this clinically important pathogen breaches epithelial surfaces and causes infection.
Collapse
Affiliation(s)
- Joe Dan Dunn
- Department of Molecular Genetics & Microbiology & Center for Microbial Pathogenesis Duke University Medical Center, 272 Jones Building, Box 3580, Durham, NC 27710, USA
| | | |
Collapse
|
46
|
Klink BU, Barden S, Heidler TV, Borchers C, Ladwein M, Stradal TEB, Rottner K, Heinz DW. Structure of Shigella IpgB2 in complex with human RhoA: implications for the mechanism of bacterial guanine nucleotide exchange factor mimicry. J Biol Chem 2010; 285:17197-208. [PMID: 20363740 PMCID: PMC2878080 DOI: 10.1074/jbc.m110.107953] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2010] [Revised: 03/11/2010] [Indexed: 11/06/2022] Open
Abstract
A common theme in bacterial pathogenesis is the manipulation of eukaryotic cells by targeting the cytoskeleton. This is in most cases achieved either by modifying actin, or indirectly via activation of key regulators controlling actin dynamics such as Rho-GTPases. A novel group of bacterial virulence factors termed the WXXXE family has emerged as guanine nucleotide exchange factors (GEFs) for these GTPases. The precise mechanism of nucleotide exchange, however, has remained unclear. Here we report the structure of the WXXXE-protein IpgB2 from Shigella flexneri and its complex with human RhoA. We unambiguously identify IpgB2 as a bacterial RhoA-GEF and dissect the molecular mechanism of GDP release, an essential prerequisite for GTP binding. Our observations uncover that IpgB2 induces conformational changes on RhoA mimicking DbI- but not DOCK family GEFs. We also show that dissociation of the GDP.Mg(2+) complex is preceded by the displacement of the metal ion to the alpha-phosphate of the nucleotide, diminishing its affinity to the GTPase. These data refine our understanding of the mode of action not only of WXXXE GEFs but also of mammalian GEFs of the DH/PH family.
Collapse
Affiliation(s)
| | | | | | - Christina Borchers
- Signaling and Motility Group, Helmholtz Zentrum für Infektionsforschung, D-38124 Braunschweig and
| | | | - Theresia E. B. Stradal
- Signaling and Motility Group, Helmholtz Zentrum für Infektionsforschung, D-38124 Braunschweig and
- the Institute of General Zoology and Genetics, University of Münster, D-48149 Münster, Germany
| | | | | |
Collapse
|
47
|
|
48
|
Müller AJ, Hoffmann C, Hardt WD. Caspase-1 activation via Rho GTPases: a common theme in mucosal infections? PLoS Pathog 2010; 6:e1000795. [PMID: 20195519 PMCID: PMC2829067 DOI: 10.1371/journal.ppat.1000795] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Affiliation(s)
| | - Claudia Hoffmann
- Institute of Microbiology, D-BIOL, ETH Zürich, Zürich, Switzerland
| | | |
Collapse
|
49
|
Pieper R, Zhang Q, Parmar PP, Huang ST, Clark DJ, Alami H, Donohue-Rolfe A, Fleischmann RD, Peterson SN, Tzipori S. The Shigella dysenteriae serotype 1 proteome, profiled in the host intestinal environment, reveals major metabolic modifications and increased expression of invasive proteins. Proteomics 2010; 9:5029-45. [PMID: 19813213 DOI: 10.1002/pmic.200900196] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Shigella dysenteriae serotype 1 (SD1) causes the most severe form of epidemic bacillary dysentery. We present the first comprehensive proteome analysis of this pathogen, profiling proteins from bacteria cultured in vitro and bacterial isolates from the large bowel of infected gnotobiotic piglets (in vivo). Overall, 1061 distinct gene products were identified. Differential display analysis revealed that SD1 cells switched to an anaerobic energy metabolism in vivo. High in vivo abundances of amino acid decarboxylases (GadB and AdiA) which enhance pH homeostasis in the cytoplasm and protein disaggregation chaperones (HdeA, HdeB and ClpB) were indicative of a coordinated bacterial survival response to acid stress. Several type III secretion system effectors were increased in abundance in vivo, including OspF, IpaC and IpaD. These proteins are implicated in invasion of colonocytes and subversion of the host immune response in S. flexneri. These observations likely reflect an adaptive response of SD1 to the hostile host environment. Seven proteins, among them the type III secretion system effectors OspC2 and IpaB, were detected as antigens in Western blots using piglet antisera. The outer membrane protein OmpA, the heat shock protein HtpG and OspC2 represent novel SD1 subunit vaccine candidates and drug targets.
Collapse
Affiliation(s)
- Rembert Pieper
- Pathogen Functional Genomics Resource Center, J. Craig Venter Institute, 9704 Medical Center Drive, Rockville, MD 20850, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Abstract
Subversion of Rho family small GTPases, which control actin dynamics, is a common infection strategy used by bacterial pathogens. In particular, Salmonella enterica serovar Typhimurium, Shigella flexneri, enteropathogenic Escherichia coli (EPEC), and enterohemorrhagic Escherichia coli (EHEC) translocate type III secretion system (T3SS) effector proteins to modulate the Rho GTPases RhoA, Cdc42, and Rac1, which trigger formation of stress fibers, filopodia, and lamellipodia/ruffles, respectively. The Salmonella effector SopE is a guanine nucleotide exchange factor (GEF) that activates Rac1 and Cdc42, which induce "the trigger mechanism of cell entry." Based on a conserved Trp-xxx-Glu motif, the T3SS effector proteins IpgB1 and IpgB2 of Shigella, SifA and SifB of Salmonella, and Map of EPEC and EHEC were grouped together into a WxxxE family; recent studies identified the T3SS EPEC and EHEC effectors EspM and EspT as new family members. Recent structural and functional studies have shown that representatives of the WxxxE effectors share with SopE a 3-D fold and GEF activity. In this minireview, we summarize contemporary findings related to the SopE and WxxxE GEFs in the context of their role in subverting general host cell signaling pathways and infection.
Collapse
|