1
|
Freeman MJ, Eral NJ, Sauer JD. Listeria monocytogenes requires phosphotransferase systems to facilitate intracellular growth and virulence. PLoS Pathog 2025; 21:e1012492. [PMID: 40233105 PMCID: PMC12052390 DOI: 10.1371/journal.ppat.1012492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 05/05/2025] [Accepted: 03/27/2025] [Indexed: 04/17/2025] Open
Abstract
The metabolism of bacterial pathogens is exquisitely evolved to support virulence in the nutrient-limiting host. Many bacterial pathogens utilize bipartite metabolism to support intracellular growth by splitting carbon utilization between two carbon sources and dividing flux to distinct metabolic needs. For example, previous studies suggest that the professional cytosolic pathogen Listeria monocytogenes (L. monocytogenes) utilizes glycerol and hexose phosphates (e.g., Glucose-6-Phosphate) as catabolic and anabolic carbon sources in the host cytosol, respectively. However, the role of this putative bipartite metabolism in L. monocytogenes virulence has not been fully assessed. Here, we demonstrate that when L. monocytogenes is unable to consume either glycerol (ΔglpD/ΔgolD), hexose phosphates (ΔuhpT), or both (ΔglpD/ΔgolD/ΔuhpT), it is still able to grow in the host cytosol and is 10- to 100-fold attenuated in vivo suggesting that L. monocytogenes consumes alternative carbon source(s) in the host. An in vitro metabolic screen using BioLog's phenotypic microarrays unexpectedly demonstrated that WT and PrfA* (G145S) L. monocytogenes, a strain with constitutive virulence gene expression, use phosphotransferase system (PTS) mediated carbon sources. These findings contrast with the existing metabolic model that cytosolic L. monocytogenes expressing PrfA does not use PTS mediated carbon sources. We next demonstrate that two independent and universal phosphocarrier proteins (PtsI [EI] and PtsH [HPr]), essential for the function of all PTS, are critical for intracellular growth and virulence in vivo. Constitutive virulence gene expression using a PrfA* (G145S) allele in ΔglpD/ΔgolD/ΔuhpT and ΔptsI failed to rescue in vivo virulence defects suggesting phenotypes are due to metabolic disruption and not virulence gene regulation. Finally, in vivo attenuation of ΔptsI and ΔptsH was additive to ΔglpD/ΔgolD/ΔuhpT, suggesting that hexose phosphates and glycerol and PTS mediated carbon source are relevant metabolites. Taken together, these studies indicate that PTS are critical virulence factors for the cytosolic growth and virulence of L. monocytogenes.
Collapse
Affiliation(s)
- Matthew J. Freeman
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Noah J. Eral
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - John-Demian Sauer
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| |
Collapse
|
2
|
Holbert S, Barilleau E, Yan J, Trotereau J, Koczerka M, Charton M, Le Vern Y, Pichon J, Grassl GA, Velge P, Wiedemann A. The Salmonella virulence protein PagN contributes to the advent of a hyper-replicating cytosolic bacterial population. Virulence 2024; 15:2357670. [PMID: 38804638 PMCID: PMC11135831 DOI: 10.1080/21505594.2024.2357670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 04/08/2024] [Indexed: 05/29/2024] Open
Abstract
Salmonella enterica subspecies enterica serovar Typhimurium is an intracellular pathogen that invades and colonizes the intestinal epithelium. Following bacterial invasion, Salmonella is enclosed within a membrane-bound vacuole known as a Salmonella-containing vacuole (SCV). However, a subset of Salmonella has the capability to prematurely rupture the SCV and escape, resulting in Salmonella hyper-replication within the cytosol of epithelial cells. A recently published RNA-seq study provides an overview of cytosolic and vacuolar upregulated genes and highlights pagN vacuolar upregulation. Here, using transcription kinetics, protein production profile, and immunofluorescence microscopy, we showed that PagN is exclusively produced by Salmonella in SCV. Gentamicin protection and chloroquine resistance assays were performed to demonstrate that deletion of pagN affects Salmonella replication by affecting the cytosolic bacterial population. This study presents the first example of a Salmonella virulence factor expressed within the endocytic compartment, which has a significant impact on the dynamics of Salmonella cytosolic hyper-replication.
Collapse
Affiliation(s)
| | | | - Jin Yan
- IRSD - Institut de Recherche en Santé Digestive, ENVT, INRAE, INSERM, Université́ de Toulouse, UPS, Toulouse, France
- Department of Gastroenterology, The Second Xiangya Hospital of Central South University, China
- Research Center of Digestive Disease, Central South University, China
| | | | | | - Mégane Charton
- INRAE, Université de Tours, ISP, Nouzilly, France
- Service biologie vétérinaire et santé animale, Inovalys, Angers, France
| | - Yves Le Vern
- INRAE, Université de Tours, ISP, Nouzilly, France
| | | | - Guntram A. Grassl
- Institute of Medical Microbiology and Hospital Epidemiology, Hannover Medical School and German Center for Infection Research (DZIF), Hannover, Germany
| | | | - Agnès Wiedemann
- INRAE, Université de Tours, ISP, Nouzilly, France
- IRSD - Institut de Recherche en Santé Digestive, ENVT, INRAE, INSERM, Université́ de Toulouse, UPS, Toulouse, France
| |
Collapse
|
3
|
Julianingsih D, Tung CW, Thapa K, Biswas D. Unveiling the Potential Ways to Apply Citrus Oil to Control Causative Agents of Pullorum Disease and Fowl Typhoid in Floor Materials. Animals (Basel) 2023; 14:23. [PMID: 38200754 PMCID: PMC10778308 DOI: 10.3390/ani14010023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 12/15/2023] [Accepted: 12/18/2023] [Indexed: 01/12/2024] Open
Abstract
This study investigates the potential role of Cold-pressed Valencia Terpeneless citrus oil (CO), as a natural antimicrobial, in controlling causative agents of pullorum disease and fowl typhoid in floor materials for poultry farming, specifically wooden chips. The study addresses the issues that have arisen as a result of the reduction in antibiotic use in poultry farming, which has resulted in the re-emergence of bacterial diseases including salmonellosis. CO efficiently inhibits the growth of pathogens including various serovars of Salmonella enterica (SE), including SE serovar Gallinarum (S. Gallinarum) and SE serovar Pullorum (S. Pullorum), in a dose-dependent manner. Minimum Inhibitory Concentration (MIC) and Minimum Bactericidal Concentration (MBC) of CO showed potential for controlling diverse S. Gallinarum and S. Pullorum isolates. Growth inhibition assays demonstrated that 0.4% (v/w) CO eliminated S. Pullorum and S. Gallinarum from 24 h onwards, also impacting poultry gut microbiota and probiotic strains. Floor material simulation, specifically wooden chips treated with 0.4% CO, confirmed CO's effectiveness in preventing S. Gallinarum and S. Pullorum growth on poultry house floors. This study also investigated the effect of CO on the expression of virulence genes in S. Gallinarum and S. Pullorum. Specifically, the study revealed that the application of CO resulted in a downregulation trend in virulence genes, including spiA, invA, spaN, sitC, and sifA, in both S. Pullorum and S. Gallinarum, implying that CO may alter the pathogenicity of these bacterial pathogens. Overall, this study reveals that CO has the potential to be used as a natural antimicrobial in the prevention and management of Salmonella-related infections in chicken production, offering a viable alternative to control these re-emerging diseases.
Collapse
Affiliation(s)
- Dita Julianingsih
- Department of Animal and Avian Sciences, University of Maryland, College Park, MD 20742, USA; (D.J.); (C.-W.T.); (K.T.)
| | - Chuan-Wei Tung
- Department of Animal and Avian Sciences, University of Maryland, College Park, MD 20742, USA; (D.J.); (C.-W.T.); (K.T.)
| | - Kanchan Thapa
- Department of Animal and Avian Sciences, University of Maryland, College Park, MD 20742, USA; (D.J.); (C.-W.T.); (K.T.)
| | - Debabrata Biswas
- Department of Animal and Avian Sciences, University of Maryland, College Park, MD 20742, USA; (D.J.); (C.-W.T.); (K.T.)
- Biological Sciences Program, University of Maryland, College Park, MD 20742, USA
| |
Collapse
|
4
|
Chatterjee R, Chowdhury AR, Mukherjee D, Chakravortty D. From Eberthella typhi to Salmonella Typhi: The Fascinating Journey of the Virulence and Pathogenicity of Salmonella Typhi. ACS OMEGA 2023; 8:25674-25697. [PMID: 37521659 PMCID: PMC10373206 DOI: 10.1021/acsomega.3c02386] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Accepted: 06/30/2023] [Indexed: 08/01/2023]
Abstract
Salmonella Typhi (S. Typhi), the invasive typhoidal serovar of Salmonella enterica that causes typhoid fever in humans, is a severe threat to global health. It is one of the major causes of high morbidity and mortality in developing countries. According to recent WHO estimates, approximately 11-21 million typhoid fever illnesses occur annually worldwide, accounting for 0.12-0.16 million deaths. Salmonella infection can spread to healthy individuals by the consumption of contaminated food and water. Typhoid fever in humans sometimes is accompanied by several other critical extraintestinal complications related to the central nervous system, cardiovascular system, pulmonary system, and hepatobiliary system. Salmonella Pathogenicity Island-1 and Salmonella Pathogenicity Island-2 are the two genomic segments containing genes encoding virulent factors that regulate its invasion and systemic pathogenesis. This Review aims to shed light on a comparative analysis of the virulence and pathogenesis of the typhoidal and nontyphoidal serovars of S. enterica.
Collapse
Affiliation(s)
- Ritika Chatterjee
- Department
of Microbiology and Cell Biology, Division of Biological Sciences, Indian Institute of Science, Bangalore, Karnataka 560012, India
| | - Atish Roy Chowdhury
- Department
of Microbiology and Cell Biology, Division of Biological Sciences, Indian Institute of Science, Bangalore, Karnataka 560012, India
| | - Debapriya Mukherjee
- Department
of Microbiology and Cell Biology, Division of Biological Sciences, Indian Institute of Science, Bangalore, Karnataka 560012, India
| | - Dipshikha Chakravortty
- Department
of Microbiology and Cell Biology, Division of Biological Sciences, Indian Institute of Science, Bangalore, Karnataka 560012, India
- Centre
for Biosystems Science and Engineering, Indian Institute of Science, Bangalore, Karnataka 560012, India
| |
Collapse
|
5
|
Pillay TD, Hettiarachchi SU, Gan J, Diaz-Del-Olmo I, Yu XJ, Muench JH, Thurston TL, Pearson JS. Speaking the host language: how Salmonella effector proteins manipulate the host. MICROBIOLOGY (READING, ENGLAND) 2023; 169:001342. [PMID: 37279149 PMCID: PMC10333799 DOI: 10.1099/mic.0.001342] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Accepted: 05/10/2023] [Indexed: 06/08/2023]
Abstract
Salmonella injects over 40 virulence factors, termed effectors, into host cells to subvert diverse host cellular processes. Of these 40 Salmonella effectors, at least 25 have been described as mediating eukaryotic-like, biochemical post-translational modifications (PTMs) of host proteins, altering the outcome of infection. The downstream changes mediated by an effector's enzymatic activity range from highly specific to multifunctional, and altogether their combined action impacts the function of an impressive array of host cellular processes, including signal transduction, membrane trafficking, and both innate and adaptive immune responses. Salmonella and related Gram-negative pathogens have been a rich resource for the discovery of unique enzymatic activities, expanding our understanding of host signalling networks, bacterial pathogenesis as well as basic biochemistry. In this review, we provide an up-to-date assessment of host manipulation mediated by the Salmonella type III secretion system injectosome, exploring the cellular effects of diverse effector activities with a particular focus on PTMs and the implications for infection outcomes. We also highlight activities and functions of numerous effectors that remain poorly characterized.
Collapse
Affiliation(s)
- Timesh D. Pillay
- Centre for Bacterial Resistance Biology, Department of Infectious Disease, Imperial College, London SW7 2AZ, UK
- The Francis Crick Institute, London NW1 1AT, UK
| | - Sahampath U. Hettiarachchi
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Victoria, Australia
- Department of Microbiology, Monash University, Clayton, Victoria, Australia
| | - Jiyao Gan
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Victoria, Australia
| | - Ines Diaz-Del-Olmo
- Centre for Bacterial Resistance Biology, Department of Infectious Disease, Imperial College, London SW7 2AZ, UK
| | - Xiu-Jun Yu
- Centre for Bacterial Resistance Biology, Department of Infectious Disease, Imperial College, London SW7 2AZ, UK
| | - Janina H. Muench
- Centre for Bacterial Resistance Biology, Department of Infectious Disease, Imperial College, London SW7 2AZ, UK
- The Francis Crick Institute, London NW1 1AT, UK
| | - Teresa L.M. Thurston
- Centre for Bacterial Resistance Biology, Department of Infectious Disease, Imperial College, London SW7 2AZ, UK
- The Francis Crick Institute, London NW1 1AT, UK
| | - Jaclyn S. Pearson
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Victoria, Australia
- Department of Microbiology, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
6
|
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
|
7
|
Development and evaluation of an indirect enzyme-linked immunosorbent assay based on a recombinant SifA protein to detect Salmonella infection in poultry. Poult Sci 2023; 102:102513. [PMID: 36805395 PMCID: PMC9972565 DOI: 10.1016/j.psj.2023.102513] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 01/12/2023] [Accepted: 01/13/2023] [Indexed: 01/22/2023] Open
Abstract
Salmonella is an important zoonotic pathogen that not only endangers food safety and human health, but also causes considerable economic losses to the poultry industry. Therefore, it is essential to establish a rapid, sensitive, and specific diagnostic method for the early detection of Salmonella infection in poultry. In this study, we developed a novel enzyme-linked immunosorbent assay (ELISA) for the detection of anti-Salmonella antibodies using a recombinant SifA protein. Amino acid sequence comparison revealed that SifA is a relatively conserved secretory protein across Salmonella serotypes. Therefore, we hypothesized that SifA can serve as a detection antigen for diagnostic testing. The SifA protein was expressed in Escherichia coli and used as a coating antigen to establish an SifA-ELISA. Control sera from specific-pathogen-free (SPF) chickens infected with Salmonella or several other non-Salmonella pathogens were then tested using the SifA-ELISA. Specificity testing demonstrated that the SifA-ELISA could detect antibodies against 3 different serotypes of Salmonella, whereas antibodies against other non-Salmonella pathogens could not be detected. Compared to the SifA-ELISA, the Salmonella plate agglutination test (PAT) failed to detect antibodies in serum samples from chickens infected with Salmonella Typhimurium. This result suggests that our SifA-ELISA may be better than PAT at detecting Salmonella infection. Comparing clinical sera, we observed a similar rate of Salmonella positivity between SifA-ELISA and PAT (92.6%). In addition, anti-SifA antibodies were continuously detected during Salmonella infection of SPF chickens, demonstrating that SifA-ELISA could consistently detect high levels of antibodies for at least 8 wk. Furthermore, the intra-assay and interassay coefficients of variation (CV) of the SifA-ELISA were below 10%, which is considered acceptable. In summary, the SifA-ELISA established here is a promising and reliable method for detection of anti-Salmonella antibodies in poultry and may contribute to the early diagnosis of Salmonella infection.
Collapse
|
8
|
Fang Z, Méresse S. Endomembrane remodeling and dynamics in Salmonella infection. MICROBIAL CELL (GRAZ, AUSTRIA) 2022; 9:24-41. [PMID: 35127930 PMCID: PMC8796136 DOI: 10.15698/mic2022.02.769] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 12/08/2021] [Accepted: 12/10/2021] [Indexed: 11/13/2022]
Abstract
Salmonellae are bacteria that cause moderate to severe infections in humans, depending on the strain and the immune status of the infected host. These pathogens have the particularity of residing in the cells of the infected host. They are usually found in a vacuolar compartment that the bacteria shape with the help of effector proteins. Following invasion of a eukaryotic cell, the bacterial vacuole undergoes maturation characterized by changes in localization, composition and morphology. In particular, membrane tubules stretching over the microtubule cytoskeleton are formed from the bacterial vacuole. Although these tubules do not occur in all infected cells, they are functionally important and promote intracellular replication. This review focuses on the role and significance of membrane compartment remodeling observed in infected cells and the bacterial and host cell pathways involved.
Collapse
Affiliation(s)
- Ziyan Fang
- Aix Marseille University, CNRS, INSERM, CIML, Marseille, France
| | | |
Collapse
|
9
|
Fang Z, Fallet M, Moest T, Gorvel JP, Méresse S. The Salmonella effector SifA initiates a kinesin-1 and kinesin-3 recruitment process mirroring that mediated by Arl8a/b. J Cell Sci 2021; 135:273658. [PMID: 34878110 DOI: 10.1242/jcs.259183] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 11/29/2021] [Indexed: 11/20/2022] Open
Abstract
When intracellular, pathogenic Salmonella reside in a membrane compartment composed of interconnected vacuoles and tubules, the formation of which depends on the translocation of bacterial effectors into the host cell. Cytoskeletons and their molecular motors are prime targets for these effectors. In this study, we show that the microtubule molecular motor KIF1Bß, a member of the kinesin-3 family, is a key element for the establishment of the Salmonella replication niche as its absence is detrimental to the stability of bacterial vacuoles and the formation of associated tubules. Kinesin-3 interacts with the Salmonella effector SifA but also with SKIP, a host protein complexed to SifA. The interaction with SifA is essential for the recruitment of kinesin-3 on Salmonella vacuoles while that with SKIP is incidental. In the non-infectious context, however, the interaction with SKIP is essential for the recruitment and activity of kinesin-3 on a part of lysosomes. Finally, our results show that in infected cells, the presence of SifA establishes a kinesin-1 and kinesin-3 recruitment pathway that is analogous to and functions independently of that mediated by the Arl8a/b GTPases.
Collapse
Affiliation(s)
- Ziyan Fang
- Aix Marseille Univ, CNRS, INSERM, CIML, Marseille, France
| | - Mathieu Fallet
- Aix Marseille Univ, CNRS, INSERM, CIML, Marseille, France
| | - Tomas Moest
- Aix Marseille Univ, CNRS, INSERM, CIML, Marseille, France
| | | | | |
Collapse
|
10
|
Walch P, Selkrig J, Knodler LA, Rettel M, Stein F, Fernandez K, Viéitez C, Potel CM, Scholzen K, Geyer M, Rottner K, Steele-Mortimer O, Savitski MM, Holden DW, Typas A. Global mapping of Salmonella enterica-host protein-protein interactions during infection. Cell Host Microbe 2021; 29:1316-1332.e12. [PMID: 34237247 PMCID: PMC8561747 DOI: 10.1016/j.chom.2021.06.004] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 02/24/2021] [Accepted: 05/21/2021] [Indexed: 11/16/2022]
Abstract
Intracellular bacterial pathogens inject effector proteins to hijack host cellular processes and promote their survival and proliferation. To systematically map effector-host protein-protein interactions (PPIs) during infection, we generated a library of 32 Salmonella enterica serovar Typhimurium (STm) strains expressing chromosomally encoded affinity-tagged effectors and quantified PPIs in macrophages and epithelial cells. We identified 446 effector-host PPIs, 25 of which were previously described, and validated 13 by reciprocal co-immunoprecipitation. While effectors converged on the same host cellular processes, most had multiple targets, which often differed between cell types. We demonstrate that SseJ, SseL, and SifA modulate cholesterol accumulation at the Salmonella-containing vacuole (SCV) partially via the cholesterol transporter Niemann-Pick C1 protein. PipB recruits the organelle contact site protein PDZD8 to the SCV, and SteC promotes actin bundling by phosphorylating formin-like proteins. This study provides a method for probing host-pathogen PPIs during infection and a resource for interrogating STm effector mechanisms.
Collapse
Affiliation(s)
- Philipp Walch
- European Molecular Biology Laboratory (EMBL), Genome Biology Unit, Heidelberg, Germany; Heidelberg University, Faculty of Biosciences, Heidelberg, Germany
| | - Joel Selkrig
- European Molecular Biology Laboratory (EMBL), Genome Biology Unit, Heidelberg, Germany
| | - Leigh A Knodler
- Paul G. Allen School for Global Health, College of Veterinary Medicine, Washington State University, Pullman, USA; Laboratory of Intracellular Parasites, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Mandy Rettel
- EMBL, Proteomics Core Facility, Heidelberg, Germany
| | - Frank Stein
- EMBL, Proteomics Core Facility, Heidelberg, Germany
| | - Keith Fernandez
- European Molecular Biology Laboratory (EMBL), Genome Biology Unit, Heidelberg, Germany
| | - Cristina Viéitez
- European Molecular Biology Laboratory (EMBL), Genome Biology Unit, Heidelberg, Germany; EMBL European Bioinformatics Institute, (EMBL-EBI), Hinxton, UK
| | - Clément M Potel
- European Molecular Biology Laboratory (EMBL), Genome Biology Unit, Heidelberg, Germany
| | - Karoline Scholzen
- European Molecular Biology Laboratory (EMBL), Genome Biology Unit, Heidelberg, Germany
| | - Matthias Geyer
- Institute of Structural Biology, University of Bonn, Bonn, Germany
| | - Klemens Rottner
- Division of Molecular Cell Biology, Zoological Institute, TU Braunschweig, Braunschweig, Germany; Molecular Cell Biology Group, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Olivia Steele-Mortimer
- Laboratory of Intracellular Parasites, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Mikhail M Savitski
- European Molecular Biology Laboratory (EMBL), Genome Biology Unit, Heidelberg, Germany; EMBL, Proteomics Core Facility, Heidelberg, Germany
| | - David W Holden
- MRC Centre for Molecular Bacteriology and Infection, Imperial College, London, UK
| | - Athanasios Typas
- European Molecular Biology Laboratory (EMBL), Genome Biology Unit, Heidelberg, Germany.
| |
Collapse
|
11
|
Fang Z, Lagier M, Méresse S. Production of Murine Macrophages from Hoxb8-Immortalized Myeloblasts: Utility and Use in the Context of Salmonella Infection. Methods Mol Biol 2021; 2182:117-126. [PMID: 32894491 DOI: 10.1007/978-1-0716-0791-6_11] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
Salmonella enterica is a Gram-negative intracellular pathogen that causes a range of life-threatening diseases in humans and animals worldwide. In a systemic infection, the ability of Salmonella to survive/replicate in macrophages, particularly in the liver and spleen, is crucial for virulence. Transformed macrophage cell lines and primary macrophages prepared from mouse bone marrow are commonly used models for the study of Salmonella infection. However, these models raise technical or ethical issues that highlight the need for alternative methods. This chapter describes a technique for immortalizing early hematopoietic progenitor cells derived from wild-type or transgenic mice and using them to produce macrophages. It validates, through a specific example, the interest of this cellular approach for the study of Salmonella infection.
Collapse
Affiliation(s)
- Ziyan Fang
- Aix Marseille Univ, CNRS, INSERM, CIML, Marseille, France
| | - Margaux Lagier
- Aix Marseille Univ, CNRS, INSERM, CIML, Marseille, France
| | | |
Collapse
|
12
|
Self-Labeling Enzyme Tags for Translocation Analyses of Salmonella Effector Proteins. Methods Mol Biol 2020. [PMID: 32894488 DOI: 10.1007/978-1-0716-0791-6_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
Salmonella enterica is an invasive, facultative intracellular pathogen with a highly sophisticated intracellular lifestyle. Invasion and intracellular proliferation are dependent on the translocation of effector proteins by two distinct type III secretion systems (T3SS) into the host cell. To unravel host-pathogen interactions, dedicated imaging techniques visualizing Salmonella effector proteins during the infection are essential. Here we describe a new approach utilizing self-labeling enzyme (SLE) tags as a universal labeling tool for tracing effector proteins. This method is able to resolve the temporal and spatial dynamics of effector proteins in living cells. The method is applicable to conventional confocal fluorescence microscopy, but also to tracking and localization microscopy (TALM), and super-resolution microscopy (SRM) of single molecules, allowing the visualization of effector proteins beyond the optical diffraction limit.
Collapse
|
13
|
Shutinoski B, Hakimi M, Harmsen IE, Lunn M, Rocha J, Lengacher N, Zhou YY, Khan J, Nguyen A, Hake-Volling Q, El-Kodsi D, Li J, Alikashani A, Beauchamp C, Majithia J, Coombs K, Shimshek D, Marcogliese PC, Park DS, Rioux JD, Philpott DJ, Woulfe JM, Hayley S, Sad S, Tomlinson JJ, Brown EG, Schlossmacher MG. Lrrk2 alleles modulate inflammation during microbial infection of mice in a sex-dependent manner. Sci Transl Med 2020; 11:11/511/eaas9292. [PMID: 31554740 DOI: 10.1126/scitranslmed.aas9292] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Revised: 12/27/2018] [Accepted: 05/11/2019] [Indexed: 12/20/2022]
Abstract
Variants in the leucine-rich repeat kinase-2 (LRRK2) gene are associated with Parkinson's disease, leprosy, and Crohn's disease, three disorders with inflammation as an important component. Because of its high expression in granulocytes and CD68-positive cells, LRRK2 may have a function in innate immunity. We tested this hypothesis in two ways. First, adult mice were intravenously inoculated with Salmonella typhimurium, resulting in sepsis. Second, newborn mouse pups were intranasally infected with reovirus (serotype 3 Dearing), which induced encephalitis. In both mouse models, wild-type Lrrk2 expression was protective and showed a sex effect, with female Lrrk2-deficient animals not controlling infection as well as males. Mice expressing Lrrk2 carrying the Parkinson's disease-linked p.G2019S mutation controlled infection better, with reduced bacterial growth and longer animal survival during sepsis. This gain-of-function effect conferred by the p.G2019S mutation was mediated by myeloid cells and was abolished in animals expressing a kinase-dead Lrrk2 variant, p.D1994S. Mouse pups with reovirus-induced encephalitis that expressed the p.G2019S Lrrk2 mutation showed increased mortality despite lower viral titers. The p.G2019S mutant Lrrk2 augmented immune cell chemotaxis and generated more reactive oxygen species during virulent infection. Reovirus-infected brains from mice expressing the p.G2019S mutant Lrrk2 contained higher concentrations of α-synuclein. Animals expressing one or two p.D1994S Lrrk2 alleles showed lower mortality from reovirus-induced encephalitis. Thus, Lrrk2 alleles may alter the course of microbial infections by modulating inflammation, and this may be dependent on the sex and genotype of the host as well as the type of pathogen.
Collapse
Affiliation(s)
- Bojan Shutinoski
- Program in Neuroscience, Ottawa Hospital Research Institute, University of Ottawa, Ottawa, ON, Canada.,Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada
| | - Mansoureh Hakimi
- Program in Neuroscience, Ottawa Hospital Research Institute, University of Ottawa, Ottawa, ON, Canada.,Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Irene E Harmsen
- Program in Neuroscience, Ottawa Hospital Research Institute, University of Ottawa, Ottawa, ON, Canada
| | - Michaela Lunn
- Program in Neuroscience, Ottawa Hospital Research Institute, University of Ottawa, Ottawa, ON, Canada.,Department of Neuroscience, Carleton University, Ottawa, ON, Canada
| | - Juliana Rocha
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Nathalie Lengacher
- Program in Neuroscience, Ottawa Hospital Research Institute, University of Ottawa, Ottawa, ON, Canada
| | - Yi Yuan Zhou
- Program in Neuroscience, Ottawa Hospital Research Institute, University of Ottawa, Ottawa, ON, Canada
| | - Jasmine Khan
- Program in Neuroscience, Ottawa Hospital Research Institute, University of Ottawa, Ottawa, ON, Canada
| | - Angela Nguyen
- Program in Neuroscience, Ottawa Hospital Research Institute, University of Ottawa, Ottawa, ON, Canada
| | - Quinton Hake-Volling
- Program in Neuroscience, Ottawa Hospital Research Institute, University of Ottawa, Ottawa, ON, Canada
| | - Daniel El-Kodsi
- Program in Neuroscience, Ottawa Hospital Research Institute, University of Ottawa, Ottawa, ON, Canada
| | - Juan Li
- Program in Neuroscience, Ottawa Hospital Research Institute, University of Ottawa, Ottawa, ON, Canada
| | - Azadeh Alikashani
- Research Centre, Montreal Heart Institute, Montréal, QC, Canada.,Faculty of Medicine, Université de Montréal, Montréal, QC, Canada
| | - Claudine Beauchamp
- Research Centre, Montreal Heart Institute, Montréal, QC, Canada.,Faculty of Medicine, Université de Montréal, Montréal, QC, Canada
| | - Jay Majithia
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada
| | - Kevin Coombs
- Department of Medical Microbiology, University of Manitoba, Winnipeg, MB, Canada
| | - Derya Shimshek
- Novartis Institutes of BioMedical Research, Novartis Campus, CH-4056 Basel, Switzerland
| | - Paul C Marcogliese
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada.,University of Ottawa Brain and Mind Research Institute, University of Ottawa, Ottawa, ON, Canada
| | - David S Park
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada.,University of Ottawa Brain and Mind Research Institute, University of Ottawa, Ottawa, ON, Canada
| | - John D Rioux
- Research Centre, Montreal Heart Institute, Montréal, QC, Canada.,Faculty of Medicine, Université de Montréal, Montréal, QC, Canada
| | - Dana J Philpott
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - John M Woulfe
- Program in Neuroscience, Ottawa Hospital Research Institute, University of Ottawa, Ottawa, ON, Canada.,Department of Pathology and Laboratory Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Shawn Hayley
- Department of Neuroscience, Carleton University, Ottawa, ON, Canada
| | - Subash Sad
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada
| | - Julianna J Tomlinson
- Program in Neuroscience, Ottawa Hospital Research Institute, University of Ottawa, Ottawa, ON, Canada.,University of Ottawa Brain and Mind Research Institute, University of Ottawa, Ottawa, ON, Canada
| | - Earl G Brown
- Program in Neuroscience, Ottawa Hospital Research Institute, University of Ottawa, Ottawa, ON, Canada.,Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada
| | - Michael G Schlossmacher
- Program in Neuroscience, Ottawa Hospital Research Institute, University of Ottawa, Ottawa, ON, Canada. .,Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada.,University of Ottawa Brain and Mind Research Institute, University of Ottawa, Ottawa, ON, Canada.,Division of Neurology, Department of Medicine, Ottawa Hospital, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
14
|
Kehl A, Göser V, Reuter T, Liss V, Franke M, John C, Richter CP, Deiwick J, Hensel M. A trafficome-wide RNAi screen reveals deployment of early and late secretory host proteins and the entire late endo-/lysosomal vesicle fusion machinery by intracellular Salmonella. PLoS Pathog 2020; 16:e1008220. [PMID: 32658937 PMCID: PMC7377517 DOI: 10.1371/journal.ppat.1008220] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2019] [Revised: 07/23/2020] [Accepted: 05/19/2020] [Indexed: 12/17/2022] Open
Abstract
The intracellular lifestyle of Salmonella enterica is characterized by the formation of a replication-permissive membrane-bound niche, the Salmonella-containing vacuole (SCV). As a further consequence of the massive remodeling of the host cell endosomal system, intracellular Salmonella establish a unique network of various Salmonella-induced tubules (SIT). The bacterial repertoire of effector proteins required for the establishment for one type of these SIT, the Salmonella-induced filaments (SIF), is rather well-defined. However, the corresponding host cell proteins are still poorly understood. To identify host factors required for the formation of SIF, we performed a sub-genomic RNAi screen. The analyses comprised high-resolution live cell imaging to score effects on SIF induction, dynamics and morphology. The hits of our functional RNAi screen comprise: i) The late endo-/lysosomal SNARE (soluble N-ethylmaleimide-sensitive factor attachment protein receptor) complex, consisting of STX7, STX8, VTI1B, and VAMP7 or VAMP8, which is, in conjunction with RAB7 and the homotypic fusion and protein sorting (HOPS) tethering complex, a complete vesicle fusion machinery. ii) Novel interactions with the early secretory GTPases RAB1A and RAB1B, providing a potential link to coat protein complex I (COPI) vesicles and reinforcing recently identified ties to the endoplasmic reticulum. iii) New connections to the late secretory pathway and/or the recycling endosome via the GTPases RAB3A, RAB8A, and RAB8B and the SNAREs VAMP2, VAMP3, and VAMP4. iv) An unprecedented involvement of clathrin-coated structures. The resulting set of hits allowed us to characterize completely new host factor interactions, and to strengthen observations from several previous studies. The facultative intracellular pathogen Salmonella enterica serovar Typhimurium induces the reorganization of the endosomal system of mammalian host cells. This activity is dependent on translocated effector proteins of the pathogen. The host cell factors required for endosomal remodeling are only partially known. To identify such factors for the formation and dynamics of endosomal compartments in Salmonella-infected cells, we performed a live cell imaging-based RNAi screen to investigate the role of 496 mammalian proteins involved in cellular logistics. We identified that endosomal remodeling by intracellular Salmonella is dependent on host factors in the following functional classes: i) the late endo-/lysosomal SNARE (soluble N-ethylmaleimide-sensitive factor attachment protein receptor) complex, ii) the early secretory pathway, represented by regulator GTPases RAB1A and RAB1B, iii) the late secretory pathway and/or recycling endosomes represented by GTPases RAB3A, RAB8A, RAB8B, and the SNAREs VAMP2, VAMP3, and VAMP4, and iv) clathrin-coated structures. The identification of these new host factors provides further evidence for the complex manipulation of host cell transport functions by intracellular Salmonella and should enable detailed follow-up studies on the mechanisms involved.
Collapse
Affiliation(s)
- Alexander Kehl
- Division of Microbiology, University of Osnabrück, Osnabrück, Germany
- Division of Biophysics, University of Osnabrück, Osnabrück, Germany
- * E-mail: (AK); (MH)
| | - Vera Göser
- Division of Microbiology, University of Osnabrück, Osnabrück, Germany
| | - Tatjana Reuter
- Division of Microbiology, University of Osnabrück, Osnabrück, Germany
| | - Viktoria Liss
- Division of Microbiology, University of Osnabrück, Osnabrück, Germany
| | - Maximilian Franke
- Division of Microbiology, University of Osnabrück, Osnabrück, Germany
| | - Christopher John
- Division of Microbiology, University of Osnabrück, Osnabrück, Germany
| | | | - Jörg Deiwick
- Division of Microbiology, University of Osnabrück, Osnabrück, Germany
| | - Michael Hensel
- Division of Microbiology, University of Osnabrück, Osnabrück, Germany
- CellNanOs–Center for Cellular Nanoanalytics, Fachbereich Biologie/Chemie, Universität Osnabrück, Osnabrück, Germany
- * E-mail: (AK); (MH)
| |
Collapse
|
15
|
Knuff-Janzen K, Tupin A, Yurist-Doutsch S, Rowland JL, Finlay BB. Multiple Salmonella-pathogenicity island 2 effectors are required to facilitate bacterial establishment of its intracellular niche and virulence. PLoS One 2020; 15:e0235020. [PMID: 32584855 PMCID: PMC7316343 DOI: 10.1371/journal.pone.0235020] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 06/05/2020] [Indexed: 12/31/2022] Open
Abstract
The pathogenesis of Salmonella Typhimurium depends on the
bacterium’s ability to survive and replicate within host cells. The formation
and maintenance of a unique membrane-bound compartment, termed the
Salmonella-containing vacuole (SCV), is essential for
S. Typhimurium pathogenesis. SCV-bound S.
Typhimurium induces formation of filamentous tubules that radiate outwards from
the SCV, termed Salmonella-induced filaments (SIFs). SIF
formation is concomitant with the onset of replication within host epithelial
cells. SIF biogenesis, formation and maintenance of the SCV, and the
intracellular positioning of the SCV within the host cell requires translocation
of bacterial proteins (effectors) into the host cell. Effectors secreted by the
type III secretion system encoded on Salmonella pathogenicity
island 2 (T3SS2) function to interfere with host cellular processes and promote
both intracellular survival and replication of S. Typhimurium.
Seven T3SS2-secreted effectors, SifA, SopD2, PipB2, SteA, SseJ, SseF, and SseG
have previously been implicated to play complementary, redundant, and/or
antagonistic roles with respect to SIF biogenesis, intracellular positioning of
the SCV, and SCV membrane dynamics modulation during infection. We undertook a
systematic study to delineate the contribution of each effector to these
processes by (i) deleting all seven of these effectors in a single
S. Typhimurium strain; and (ii) deleting combinations of
multiple effectors based on putative effector function. Using this deletion
mutant library, we show that each of SIF biogenesis, intracellular SCV
localization, intramacrophage replication, colonization, and virulence depends
on the activities of multiple effectors. Together, our data demonstrates the
complex interplay between these seven effectors and highlights the necessity to
study T3SS2-secreted effectors as groups, rather than studies of individual
effectors.
Collapse
Affiliation(s)
- Katelyn Knuff-Janzen
- Michael Smith Laboratories, University of British Columbia, Vancouver,
British Columbia, Canada
- Department of Microbiology & Immunology, University of British
Columbia, Vancouver, British Columbia, Canada
| | - Audrey Tupin
- Michael Smith Laboratories, University of British Columbia, Vancouver,
British Columbia, Canada
- Department of Microbiology & Immunology, University of British
Columbia, Vancouver, British Columbia, Canada
| | - Sophie Yurist-Doutsch
- Michael Smith Laboratories, University of British Columbia, Vancouver,
British Columbia, Canada
- Department of Microbiology & Immunology, University of British
Columbia, Vancouver, British Columbia, Canada
| | - Jennifer L. Rowland
- Michael Smith Laboratories, University of British Columbia, Vancouver,
British Columbia, Canada
- Department of Microbiology & Immunology, University of British
Columbia, Vancouver, British Columbia, Canada
| | - B. Brett Finlay
- Michael Smith Laboratories, University of British Columbia, Vancouver,
British Columbia, Canada
- Department of Microbiology & Immunology, University of British
Columbia, Vancouver, British Columbia, Canada
- Department of Biochemistry and Molecular Biology, University of British
Columbia, Vancouver, British Columbia, Canada
- * E-mail:
| |
Collapse
|
16
|
Alberdi L, Vergnes A, Manneville JB, Tembo DL, Fang Z, Zhao Y, Schroeder N, Dumont A, Lagier M, Bassereau P, Redondo-Morata L, Gorvel JP, Méresse S. Regulation of kinesin-1 activity by the Salmonella enterica effectors PipB2 and SifA. J Cell Sci 2020; 133:133/9/jcs239863. [PMID: 32409568 DOI: 10.1242/jcs.239863] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 03/13/2020] [Indexed: 11/20/2022] Open
Abstract
Salmonella enterica is an intracellular bacterial pathogen. The formation of its replication niche, which is composed of a vacuole associated with a network of membrane tubules, depends on the secretion of a set of bacterial effector proteins whose activities deeply modify the functions of the eukaryotic host cell. By recruiting and regulating the activity of the kinesin-1 molecular motor, Salmonella effectors PipB2 and SifA play an essential role in the formation of the bacterial compartments. In particular, they allow the formation of tubules from the vacuole and their extension along the microtubule cytoskeleton, and thus promote membrane exchanges and nutrient supply. We have developed in vitro and in cellulo assays to better understand the specific role played by these two effectors in the recruitment and regulation of kinesin-1. Our results reveal a specific interaction between the two effectors and indicate that, contrary to what studies on infected cells suggested, interaction with PipB2 is sufficient to relieve the autoinhibition of kinesin-1. Finally, they suggest the involvement of other Salmonella effectors in the control of the activity of this molecular motor.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
| | | | - Jean-Baptiste Manneville
- Institut Curie, PSL Research University, CNRS, UMR 144, 26 rue d'Ulm, F-75005, Paris, France.,Sorbonne Université, UPMC University Paris 06, CNRS, UMR 144, 26 rue d'Ulm, F-75005, Paris, France
| | | | - Ziyan Fang
- Aix-Marseille Université, CNRS, INSERM, CIML, Marseille, France
| | - Yaya Zhao
- Aix-Marseille Université, CNRS, INSERM, CIML, Marseille, France
| | - Nina Schroeder
- Aix-Marseille Université, CNRS, INSERM, CIML, Marseille, France
| | - Audrey Dumont
- Aix-Marseille Université, CNRS, INSERM, CIML, Marseille, France
| | - Margaux Lagier
- Aix-Marseille Université, CNRS, INSERM, CIML, Marseille, France
| | - Patricia Bassereau
- Laboratoire Physico Chimie Curie, Institut Curie, PSL Research University, CNRS UMR168, 75005 Paris, France.,Sorbonne Université, 1 Place Jussieu, 75005 Paris, France
| | | | | | | |
Collapse
|
17
|
Göser V, Kehl A, Röder J, Hensel M. Role of the ESCRT‐III complex in controlling integrity of the
Salmonella
‐containing vacuole. Cell Microbiol 2020; 22:e13176. [DOI: 10.1111/cmi.13176] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 12/27/2019] [Accepted: 01/20/2020] [Indexed: 12/21/2022]
Affiliation(s)
- Vera Göser
- Abt. MikrobiologieFachbereich Biologie/Chemie, Universität Osnabrück Osnabrück Germany
| | - Alexander Kehl
- Abt. MikrobiologieFachbereich Biologie/Chemie, Universität Osnabrück Osnabrück Germany
- Institut für HygieneUniversität Münster Münster Germany
- CellNanOs, Center for Cellular NanoanalyticsFachbereich Biologie/Chemie, Universität Osnabrück Osnabrück Germany
| | - Jennifer Röder
- Abt. MikrobiologieFachbereich Biologie/Chemie, Universität Osnabrück Osnabrück Germany
| | - Michael Hensel
- Abt. MikrobiologieFachbereich Biologie/Chemie, Universität Osnabrück Osnabrück Germany
- CellNanOs, Center for Cellular NanoanalyticsFachbereich Biologie/Chemie, Universität Osnabrück Osnabrück Germany
| |
Collapse
|
18
|
Sit WY, Chen YA, Chen YL, Lai CH, Wang WC. Cellular evasion strategies of Helicobacter pylori in regulating its intracellular fate. Semin Cell Dev Biol 2020; 101:59-67. [PMID: 32033828 PMCID: PMC7102552 DOI: 10.1016/j.semcdb.2020.01.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 01/27/2020] [Indexed: 12/19/2022]
Abstract
Helicobacter pylori colonizes human stomach mucosa and its infection causes gastrointestinal diseases with variable severity. Bacterial infection stimulates autophagy, which is a part of innate immunity used to eliminate intracellular pathogens. Several intracellular bacteria have evolved multipronged strategies to circumvent this conserved system and thereby enhance their chance of intracellular survival. Nonetheless, studies on H. pylori have produced inconsistent results, showing either elevated or reduced clearance efficiency of intracellular bacteria through autophagy. In this review, we summarize recent studies on the mechanisms involved in autophagy induced by H. pylori and the fate of intracellular bacteria.
Collapse
Affiliation(s)
- Wei Yang Sit
- Biomedical Science and Engineering Center, National Tsing Hua University, Hsinchu, Taiwan; Institute of Molecular and Cellular Biology & Department of Life Science, National Tsing Hua University, Hsinchu, Taiwan
| | - Yu-An Chen
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA; Department of Microbiology and Immunology, Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan, Taiwan
| | - Yu-Lun Chen
- Biomedical Science and Engineering Center, National Tsing Hua University, Hsinchu, Taiwan; Institute of Molecular and Cellular Biology & Department of Life Science, National Tsing Hua University, Hsinchu, Taiwan
| | - Chih-Ho Lai
- Department of Microbiology and Immunology, Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan, Taiwan; Department of Microbiology, School of Medicine, China Medical University, Taichung, Taiwan; Department of Nursing, Asia University, Taichung, Taiwan; Molecular Infectious Disease Research Center, Department of Pediatrics, Chang Gung Memorial Hospital, Linkuo, Taiwan.
| | - Wen-Ching Wang
- Biomedical Science and Engineering Center, National Tsing Hua University, Hsinchu, Taiwan; Institute of Molecular and Cellular Biology & Department of Life Science, National Tsing Hua University, Hsinchu, Taiwan.
| |
Collapse
|
19
|
Beshiru A, Igbinosa IH, Igbinosa EO. Prevalence of Antimicrobial Resistance and Virulence Gene Elements of Salmonella Serovars From Ready-to-Eat (RTE) Shrimps. Front Microbiol 2019; 10:1613. [PMID: 31354693 PMCID: PMC6637730 DOI: 10.3389/fmicb.2019.01613] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Accepted: 06/28/2019] [Indexed: 01/02/2023] Open
Abstract
Gastrointestinal illnesses continue to be a global public health risk. Exposure to foodborne Salmonella directly or indirectly through consumption of ready-to-eat seafood can be an important route of infection to humans. This study was designed to estimate the population cell density, prevalence, virulence gene signatures, and antibiotic resistance of Salmonella serovars from ready-to-eat shrimps. Ready-to-eat (RTE) shrimp samples were obtained from different open markets in Delta and Edo States, Nigeria from November 2016 to October 2017. We employed classical and polymerase chain reaction (PCR) approaches. The mean Salmonella species enumerated from the RTE shrimps ranged from -0.301 to 5.434 log10 cfu/g with 210/1440 (14.58%) of the RTE shrimp samples harbored Salmonella species. After biochemical and PCR approach, the identified isolates were Salmonella Enteritidis 11(24.4%), Salmonella Typhimurium 14 (31.1%) and other Salmonella spp. 20 (44.4%). All Salmonella species recovered were resistant to penicillin and erythromycin with 100% sensitivity to cefotaxime, cephalothin, colistin, and polymyxin B. Findings on the multidrug-resistant (MDR) profile showed that a total of 9/14 (64.3%) of Salmonella Enteritidis were resistant to 5 antibiotics which belongs to 3 different groups of antimicrobials with a multiple antibiotic-resistant (MAR) index of 0.21; while 3/11 (27.3%) of Salmonella Typhimurium were resistant to 11 antibiotics which belongs to 7 different groups of antimicrobials with a MAR index of 0.46. Virulence genes (spiA, sipB, invA, sif A, fljB, and sefA) and resistance genes (class 1 and II integrase, sul2, catB3, flor, tmp, bla TEM, strB, dfr1, and tetC) were also detected in some of the Salmonella species with variable percentage. This study indicates that ready-to-eat shrimps are probable reservoirs harboring Salmonella strains. The identified Salmonella isolates which exhibited virulence determinants and antibiotic-resistant coupled with high MAR index constitute a consumer health risk to the communities.
Collapse
Affiliation(s)
- Abeni Beshiru
- Applied Microbial Processes and Environmental Health Research Group, Department of Microbiology, Faculty of Life Sciences, University of Benin, Benin City, Nigeria
| | - Isoken H. Igbinosa
- Department of Environmental Management and Toxicology, Faculty of Life Sciences, University of Benin, Benin City, Nigeria
| | - Etinosa O. Igbinosa
- Applied Microbial Processes and Environmental Health Research Group, Department of Microbiology, Faculty of Life Sciences, University of Benin, Benin City, Nigeria
- Sustainable Development Office, University of Benin, Benin City, Nigeria
| |
Collapse
|
20
|
Stamm CE, Pasko BL, Chaisavaneeyakorn S, Franco LH, Nair VR, Weigele BA, Alto NM, Shiloh MU. Screening Mycobacterium tuberculosis Secreted Proteins Identifies Mpt64 as a Eukaryotic Membrane-Binding Bacterial Effector. mSphere 2019; 4:e00354-19. [PMID: 31167949 PMCID: PMC6553557 DOI: 10.1128/msphere.00354-19] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 05/19/2019] [Indexed: 02/07/2023] Open
Abstract
Mycobacterium tuberculosis (Mtb), the causative agent of tuberculosis, is one of the most successful human pathogens. One reason for its success is that Mtb can reside within host macrophages, a cell type that normally functions to phagocytose and destroy infectious bacteria. However, Mtb is able to evade macrophage defenses in order to survive for prolonged periods of time. Many intracellular pathogens secrete virulence factors targeting host membranes and organelles to remodel their intracellular environmental niche. We hypothesized that Mtb secreted proteins that target host membranes are vital for Mtb to adapt to and manipulate the host environment for survival. Thus, we characterized 200 secreted proteins from Mtb for their ability to associate with eukaryotic membranes using a unique temperature-sensitive yeast screen and to manipulate host trafficking pathways using a modified inducible secretion screen. We identified five Mtb secreted proteins that both associated with eukaryotic membranes and altered the host secretory pathway. One of these secreted proteins, Mpt64, localized to the endoplasmic reticulum during Mtb infection of murine and human macrophages and impaired the unfolded protein response in macrophages. These data highlight the importance of secreted proteins in Mtb pathogenesis and provide a basis for further investigation into their molecular mechanisms.IMPORTANCE Advances have been made to identify secreted proteins of Mycobacterium tuberculosis during animal infections. These data, combined with transposon screens identifying genes important for M. tuberculosis virulence, have generated a vast resource of potential M. tuberculosis virulence proteins. However, the function of many of these proteins in M. tuberculosis pathogenesis remains elusive. We have integrated three cell biological screens to characterize nearly 200 M. tuberculosis secreted proteins for eukaryotic membrane binding, host subcellular localization, and interactions with host vesicular trafficking. In addition, we observed the localization of one secreted protein, Mpt64, to the endoplasmic reticulum (ER) during M. tuberculosis infection of macrophages. Interestingly, although Mpt64 is exported by the Sec pathway, its delivery into host cells was dependent upon the action of the type VII secretion system. Finally, we observed that Mpt64 impairs the ER-mediated unfolded protein response in macrophages.
Collapse
Affiliation(s)
- Chelsea E Stamm
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Breanna L Pasko
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Sujittra Chaisavaneeyakorn
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Luis H Franco
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Center for Autophagy Research, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Vidhya R Nair
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Bethany A Weigele
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Neal M Alto
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Michael U Shiloh
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
21
|
Patel S, Wall DM, Castillo A, McCormick BA. Caspase-3 cleavage of Salmonella type III secreted effector protein SifA is required for localization of functional domains and bacterial dissemination. Gut Microbes 2019; 10:172-187. [PMID: 30727836 PMCID: PMC6546311 DOI: 10.1080/19490976.2018.1506668] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
SifA is a bi-functional Type III Secretion System (T3SS) effector protein that plays an important role in Salmonella virulence. The N-terminal domain of SifA binds SifA-Kinesin-Interacting-Protein (SKIP), and via an interaction with kinesin, forms tubular membrane extensions called Sif filaments (Sifs) that emanate from the Salmonella Containing Vacuole (SCV). The C-terminal domain of SifA harbors a WxxxE motif that functions to mimic active host cell GTPases. Taken together, SifA functions in inducing endosomal tubulation in order to maintain the integrity of the SCV and promote bacterial dissemination. Since SifA performs multiple, unrelated functions, the objective of this study was to determine how each functional domain of SifA becomes processed. Our work demonstrates that a linker region containing a caspase-3 cleavage motif separates the two functional domains of SifA. To test the hypothesis that processing of SifA by caspase-3 at this particular site is required for function and proper localization of the effector protein domains, we developed two tracking methods to analyze the intracellular localization of SifA. We first adapted a fluorescent tag called phiLOV that allowed for type-III secretion system (T3SS) mediated delivery of SifA and observation of its intracellular colocalization with caspase-3. Additionally, we created a dual-tagging strategy that permitted tracking of each of the SifA functional domains following caspase-3 cleavage to different subcellular locations. The results of this study reveal that caspase-3 cleavage of SifA is required for the proper localization of functional domains and bacterial dissemination. Considering the importance of these events in Salmonella pathogenesis, we conclude that caspase-3 cleavage of effector proteins is a more broadly applicable effector processing mechanism utilized by Salmonella to invade and persist during infection.
Collapse
Affiliation(s)
- Samir Patel
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA, USA,CONTACT Beth McCormick Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, 368 Plantation Street AS8-2011, Worcester, MA 01605, USA
| | - Daniel M. Wall
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Antonio Castillo
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA, USA
| | - Beth A. McCormick
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA, USA
| |
Collapse
|
22
|
Moest T, Zhao W, Zhao Y, Schüssler JM, Yan W, Gorvel JP, Méresse S. Contribution of bacterial effectors and host proteins to the composition and function of Salmonella-induced tubules. Cell Microbiol 2018; 20:e12951. [PMID: 30212607 DOI: 10.1111/cmi.12951] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 08/31/2018] [Accepted: 09/04/2018] [Indexed: 12/17/2023]
Abstract
Cells infected with Salmonella are characterised by the appearance of membrane tubular structures that stretch from the bacterial vacuole. The formation of these tubules requires the translocation of Salmonella effector proteins within the infected cell. Different types of Salmonella-induced tubules with varying host protein compositions have been identified. This variability probably reflects the ability of these tubules to interact with different host compartments. Membrane tubules decorated with effector proteins but essentially devoid of host proteins and named LAMP1-negative (LNT) were observed. LNTs wrap around LAMP1-positive vesicles and may promote recruitment of lysosomal glycoproteins to bacterial vacuole and the formation of a replication niche. We conducted a biochemical and functional characterisation of LNTs. We show that the effector proteins SseF and SseG are necessary for their formation. The absence of these tubules is associated with decreased recruitment of LAMP1 to SCVs, decreased intracellular replication of Salmonella, and decreased virulence in mice. We found that the process leading to the recruitment of lysosomal glycoproteins to tubules involves the C-terminal domain of the effector protein SifA and the GTPase Arl8b. Overall, these data suggest that Salmonella-induced tubules promote the establishment of the replication niche by promoting recruitment of host proteins to the bacterial vacuole.
Collapse
Affiliation(s)
- Thomas Moest
- Aix Marseille Univ, CNRS, INSERM, CIML, Marseille, France
| | - Weidong Zhao
- Aix Marseille Univ, CNRS, INSERM, CIML, Marseille, France
| | - Yaya Zhao
- Aix Marseille Univ, CNRS, INSERM, CIML, Marseille, France
| | | | - Wen Yan
- Aix Marseille Univ, CNRS, INSERM, CIML, Marseille, France
| | | | | |
Collapse
|
23
|
Wang L, Yan J, Niu H, Huang R, Wu S. Autophagy and Ubiquitination in Salmonella Infection and the Related Inflammatory Responses. Front Cell Infect Microbiol 2018; 8:78. [PMID: 29594070 PMCID: PMC5861197 DOI: 10.3389/fcimb.2018.00078] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Accepted: 02/27/2018] [Indexed: 12/12/2022] Open
Abstract
Salmonellae are facultative intracellular pathogens that cause globally distributed diseases with massive morbidity and mortality in humans and animals. In the past decades, numerous studies were focused on host defenses against Salmonella infection. Autophagy has been demonstrated to be an important defense mechanism to clear intracellular pathogenic organisms, as well as a regulator of immune responses. Ubiquitin modification also has multiple effects on the host immune system against bacterial infection. It has been indicated that ubiquitination plays critical roles in recognition and clearance of some invading bacteria by autophagy. Additionally, the ubiquitination of autophagy proteins in autophagy flux and inflammation-related substance determines the outcomes of infection. However, many intracellular pathogens manipulate the ubiquitination system to counteract the host immunity. Salmonellae interfere with host responses via the delivery of ~30 effector proteins into cytosol to promote their survival and proliferation. Among them, some could link the ubiquitin-proteasome system with autophagy during infection and affect the host inflammatory responses. In this review, novel findings on the issue of ubiquitination and autophagy connection as the mechanisms of host defenses against Salmonella infection and the subverted processes are introduced.
Collapse
Affiliation(s)
- Lidan Wang
- Department of Microbiology, Medical College of Soochow University, Suzhou, China
| | - Jing Yan
- Department of Microbiology, Medical College of Soochow University, Suzhou, China
| | - Hua Niu
- Department of Microbiology, Medical College of Soochow University, Suzhou, China
| | - Rui Huang
- Department of Microbiology, Medical College of Soochow University, Suzhou, China
| | - Shuyan Wu
- Department of Microbiology, Medical College of Soochow University, Suzhou, China
| |
Collapse
|
24
|
Salmonella exploits the host endolysosomal tethering factor HOPS complex to promote its intravacuolar replication. PLoS Pathog 2017; 13:e1006700. [PMID: 29084291 PMCID: PMC5679646 DOI: 10.1371/journal.ppat.1006700] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Revised: 11/09/2017] [Accepted: 10/18/2017] [Indexed: 12/22/2022] Open
Abstract
Salmonella enterica serovar typhimurium extensively remodels the host late endocytic compartments to establish its vacuolar niche within the host cells conducive for its replication, also known as the Salmonella-containing vacuole (SCV). By maintaining a prolonged interaction with late endosomes and lysosomes of the host cells in the form of interconnected network of tubules (Salmonella-induced filaments or SIFs), Salmonella gains access to both membrane and fluid-phase cargo from these compartments. This is essential for maintaining SCV membrane integrity and for bacterial intravacuolar nutrition. Here, we have identified the multisubunit lysosomal tethering factor—HOPS (HOmotypic fusion and Protein Sorting) complex as a crucial host factor facilitating delivery of late endosomal and lysosomal content to SCVs, providing membrane for SIF formation, and nutrients for intravacuolar bacterial replication. Accordingly, depletion of HOPS subunits significantly reduced the bacterial load in non-phagocytic and phagocytic cells as well as in a mouse model of Salmonella infection. We found that Salmonella effector SifA in complex with its binding partner; SKIP, interacts with HOPS subunit Vps39 and mediates recruitment of this tethering factor to SCV compartments. The lysosomal small GTPase Arl8b that binds to, and promotes membrane localization of Vps41 (and other HOPS subunits) was also required for HOPS recruitment to SCVs and SIFs. Our findings suggest that Salmonella recruits the host late endosomal and lysosomal membrane fusion machinery to its vacuolar niche for access to host membrane and nutrients, ensuring its intracellular survival and replication. Intracellular pathogens have devised various strategies to subvert the host membrane trafficking pathways for their growth and survival inside the host cells. Salmonella is one such successful intracellular pathogen that redirects membrane and nutrients from the host endocytic compartments to its replicative niche known as the Salmonella-containing vacuole (SCV) via establishing an interconnected network of tubules (Salmonella-induced filaments or SIFs) that form a continuum with the SCVs. How Salmonella ensures a constant supply of endocytic cargo required for its survival and growth remained unexplored. Our work uncovers a strategy evolved by Salmonella wherein it secretes a bacterial effector into the host cytosol that recruits component of host vesicle fusion machinery-HOPS complex to SCVs and SIFs. HOPS complex promotes docking of the late endocytic compartments at the SCV membrane, prior to their fusion. Thus, depletion of HOPS subunits both in cultured cell lines as well as a mouse model inhibits Salmonella replication, likely due to reduced access to host membranes and nutrients by the vacuolar bacteria. These findings provide mechanistic insights into how this pathogen reroutes the host’s endocytic transport towards its vacuole, ensuring its own intracellular survival and replication.
Collapse
|
25
|
Namakchian M, Kassler K, Sticht H, Hensel M, Deiwick J. Structure-based functional analysis of effector protein SifA in living cells reveals motifs important for Salmonella intracellular proliferation. Int J Med Microbiol 2017; 308:84-96. [PMID: 28939436 DOI: 10.1016/j.ijmm.2017.09.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2017] [Revised: 09/02/2017] [Accepted: 09/04/2017] [Indexed: 02/01/2023] Open
Abstract
The facultative intracellular pathogen Salmonella enterica survives and replicates inside the Salmonella-containing vacuole (SCV) of mammalian host cells. SifA is a key effector protein translocated by a type III secretion system and involved in formation of Salmonella-induced filaments (SIF), extensive tubular endosomal compartments. Recruitment of LAMP1 (lysosomal-associated membrane protein 1)-positive membranes to SIF ensures integrity and dynamics of the membrane network. The binding of SifA to the host protein SKIP (SifA and kinesin interacting protein) was proposed as crucial for this function. Due to structural mimicry SifA has further been proposed to interact with G-proteins. We conducted a mutational study of SifA to identify domains and amino acid residues specifically relevant for intracellular replication and SIF formation. Mutations were designed based on the available structural data of SifA and its interface with SKIP, or modeled for SifA as putative guanine nucleotide exchange factor. We developed a live cell imaging-based approach for volume quantification of the SIF network that allowed determination of subtle changes in SIF network and performed a comprehensive analysis of mutant forms of SifA by this approach. We found that the SifA catalytic loop of WxxxE effectors is as important for SIF formation and intracellular proliferation as the SKIP interaction motif, or the CAAX motif for membrane anchoring of SifA.
Collapse
Affiliation(s)
| | - Kristin Kassler
- Institut für Biochemie, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Heinrich Sticht
- Institut für Biochemie, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Michael Hensel
- Abt. Mikrobiologie, Universität Osnabrück, Osnabrück, Germany.
| | - Jörg Deiwick
- Abt. Mikrobiologie, Universität Osnabrück, Osnabrück, Germany.
| |
Collapse
|
26
|
Use of a novel antigen expressing system to study the Salmonella enterica serovar Typhi protein recognition by T cells. PLoS Negl Trop Dis 2017; 11:e0005912. [PMID: 28873442 PMCID: PMC5600385 DOI: 10.1371/journal.pntd.0005912] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 09/15/2017] [Accepted: 08/28/2017] [Indexed: 11/19/2022] Open
Abstract
Salmonella enterica serovar Typhi (S. Typhi), the causative agent of the typhoid fever, is a pathogen of great public health importance. Typhoid vaccines have the potential to be cost-effective measures towards combating this disease, yet the antigens triggering host protective immune responses are largely unknown. Given the key role of cellular-mediated immunity in S. Typhi protection, it is crucial to identify S. Typhi proteins involved in T-cell responses. Here, cells from individuals immunized with Ty21a typhoid vaccine were collected before and after immunization and used as effectors. We also used an innovative antigen expressing system based on the infection of B-cells with recombinant Escherichia coli (E. coli) expressing one of four S. Typhi gene products (i.e., SifA, OmpC, FliC, GroEL) as targets. Using flow cytometry, we found that the pattern of response to specific S. Typhi proteins was variable. Some individuals responded to all four proteins while others responded to only one or two proteins. We next evaluated whether T-cells responding to recombinant E. coli also possess the ability to respond to purified proteins. We observed that CD4+ cell responses, but not CD8+ cell responses, to recombinant E. coli were significantly associated with the responses to purified proteins. Thus, our results demonstrate the feasibility of using an E. coli expressing system to uncover the antigen specificity of T-cells and highlight its applicability to vaccine studies. These results also emphasize the importance of selecting the stimuli appropriately when evaluating CD4+ and CD8+ cell responses. Salmonella enterica serovar Typhi (S. Typhi) is the causative agent of the life-threatening typhoid fever that affects 11.9–20.6 million individuals annually in low-income and middle-income countries. The T-cells, CD4+ and CD8+ T cells, play a significant role in protection against S. Typhi infection. Yet, the antigens triggering host protective immune responses recognized by these cells are largely unknown. To address this shortcoming, in this study we used an E. coli expression system methodology for identifying immunogenic proteins of S. Typhi. We found that although the pattern of response to individual S. Typhi proteins was variable among the typhoid vaccinees, the E. coli expressing system uncovered the antigen specificity of T-cells, and highlight its applicability to vaccine studies.
Collapse
|
27
|
Knuff K, Finlay BB. What the SIF Is Happening-The Role of Intracellular Salmonella-Induced Filaments. Front Cell Infect Microbiol 2017; 7:335. [PMID: 28791257 PMCID: PMC5524675 DOI: 10.3389/fcimb.2017.00335] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 07/10/2017] [Indexed: 11/29/2022] Open
Abstract
A common strategy among intracellular bacterial pathogens is to enter into a vacuolar environment upon host cell invasion. One such pathogen, Salmonella enterica, resides within the Salmonella-containing vacuole (SCV) inside epithelial cells and macrophages. Salmonella hijacks the host endosomal system to establish this unique intracellular replicative niche, forming a highly complex and dynamic network of Salmonella-induced filaments (SIFs). SIFs radiate outwards from the SCV upon onset of bacterial replication. SIF biogenesis is dependent on the activity of bacterial effector proteins secreted by the Salmonella-pathogenicity island-2 (SPI-2) encoded type III secretion system. While the presence of SIFs has been known for almost 25 years, their precise role during infection remains elusive. This review summarizes our current knowledge of SCV maturation and SIF biogenesis, and recent advances in our understanding of the role of SIFs inside cells.
Collapse
Affiliation(s)
- Katelyn Knuff
- Michael Smith Laboratories, University of British ColumbiaVancouver, BC, Canada.,Department of Microbiology and Immunology, University of British ColumbiaVancouver, BC, Canada
| | - B Brett Finlay
- Michael Smith Laboratories, University of British ColumbiaVancouver, BC, Canada.,Department of Microbiology and Immunology, University of British ColumbiaVancouver, BC, Canada.,Department of Biochemistry and Molecular Biology, University of British ColumbiaVancouver, BC, Canada
| |
Collapse
|
28
|
Ratner D, Orning MPA, Lien E. Bacterial secretion systems and regulation of inflammasome activation. J Leukoc Biol 2016; 101:165-181. [PMID: 27810946 DOI: 10.1189/jlb.4mr0716-330r] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Revised: 09/19/2016] [Accepted: 09/20/2016] [Indexed: 01/03/2023] Open
Abstract
Innate immunity is critical for host defenses against pathogens, but many bacteria display complex ways of interacting with innate immune signaling, as they may both activate and evade certain pathways. Gram-negative bacteria can exhibit specialized nanomachine secretion systems for delivery of effector proteins into mammalian cells. Bacterial types III, IV, and VI secretion systems (T3SS, T4SS, and T6SS) are known for their impact on caspase-1-activating inflammasomes, necessary for producing bioactive inflammatory cytokines IL-1β and IL-18, key participants of anti-bacterial responses. Here, we discuss how these secretion systems can mediate triggering and inhibition of inflammasome signaling. We propose that a fine balance between secretion system-mediated activation and inhibition can determine net activation of inflammasome activity and control inflammation, clearance, or spread of the infection.
Collapse
Affiliation(s)
- Dmitry Ratner
- Program in Innate Immunity, Division of Infectious Diseases and Immunology, Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, USA; and
| | - M Pontus A Orning
- Program in Innate Immunity, Division of Infectious Diseases and Immunology, Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, USA; and.,Centre of Molecular Inflammation Research, Department of Cancer Research and Molecular Medicine, Norges Teknisk-Naturvitenskapelige Universitet, Trondheim, Norway
| | - Egil Lien
- Program in Innate Immunity, Division of Infectious Diseases and Immunology, Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, USA; and .,Centre of Molecular Inflammation Research, Department of Cancer Research and Molecular Medicine, Norges Teknisk-Naturvitenskapelige Universitet, Trondheim, Norway
| |
Collapse
|
29
|
Abstract
Small GTPases of the Rab protein family control intracellular vesicular trafficking to allow their communication and maintenance. It is a common strategy for intracellular bacteria to exploit these pathways to shape their respective niches for survival. The subversion of Rabs for the generation of an intracellular environment favoring the pathogen has been described almost exclusively for intracellular bacteria that reside within bacterial containing vacuoles (BCVs). However, less is known about Rab subversion for bacteria that rupture the BCV to reach the host cytoplasm. Here, we provide recent examples of Rab targeting by both groups of intracellular bacteria with a special focus on Shigella, the causative agent of bacillary dysentery. Shigella recruits Rab11, the hallmark of the perinuclear recycling compartment to in situ formed macropinosomes at the entry foci via the bacterial effector IpgD. This leads to efficient BCV rupture and cytosolic escape. We discuss the concept of diverted recycling through host Rab GTPases that emerges as a novel pathogen strategy.
Collapse
Affiliation(s)
- Noelia López-Montero
- a Institut Pasteur, Research unit "Dynamics of host-pathogen interactions," Paris , France
| | - Jost Enninga
- a Institut Pasteur, Research unit "Dynamics of host-pathogen interactions," Paris , France
| |
Collapse
|
30
|
Zhao Y, Gorvel JP, Méresse S. Effector proteins support the asymmetric apportioning of Salmonella during cytokinesis. Virulence 2016; 7:669-78. [PMID: 27046257 PMCID: PMC4991364 DOI: 10.1080/21505594.2016.1173298] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Salmonella-infected cells are characterized by the presence of intra-cellular membranous tubules that emerge from bacterial vacuoles and extend along microtubules. The formation of Salmonella-induced tubules depends on the Salmonella pathogenicity island 2-encoded type III secretion system (T3SS-2) that translocates bacterial effector proteins inside host cells. Effector proteins have enzymatic activities or allow for hijacking of cellular functions. The role of Salmonella-induced tubules in virulence remains unclear but their absence is correlated with virulence defects. This study describes the presence of inter-cellular tubules that arise between daughter cells during cytokinesis. Inter-cellular tubules connect bacterial vacuoles originally present in the parent cell and that have been apportioned between daughters. Their formation requires a functional T3SS-2 and effector proteins. Our data establish a correlation between the formation of inter-cellular tubules and the asymmetric distribution of bacterial vacuoles in daughters. Thus, by manipulating the distribution of bacteria in cytokinetic cells, Salmonella T3SS-2 effector proteins may increase bacterial spreading and the systemic character of the infection.
Collapse
Affiliation(s)
- Yaya Zhao
- a Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université UM2 , Inserm, U1104, CNRS UMR7280, Marseille , France
| | - Jean-Pierre Gorvel
- a Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université UM2 , Inserm, U1104, CNRS UMR7280, Marseille , France
| | - Stéphane Méresse
- a Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université UM2 , Inserm, U1104, CNRS UMR7280, Marseille , France
| |
Collapse
|