1
|
Nolan LM, Webber MA, Filloux A. Throwing a spotlight on genomic dark matter: the power and potential of transposon-insertion sequencing. J Biol Chem 2025:110231. [PMID: 40378959 DOI: 10.1016/j.jbc.2025.110231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2025] [Revised: 04/29/2025] [Accepted: 05/06/2025] [Indexed: 05/19/2025] Open
Abstract
Linking genotype to phenotype is a central goal in biology. In the microbiological field, transposon mutagenesis is a technique that has been widely used since the 1970's to facilitate this connection. The development of modern 'omics approaches and next-generation sequencing, have allowed high-throughput association between genes and their putative function. In 2009, four different variations of modern transposon-insertion sequencing (TIS) approaches were published, being referred to as transposon-directed insertion-site sequencing (TraDIS), transposon sequencing (Tn-seq), insertion sequencing (INSeq) and high-throughput insertion tracking by deep sequencing (HITS). These approaches exploit a similar concept to allow estimation of the essentiality or contribution to fitness of each gene in any bacterial genome. The main rationale is to perform a comparative analysis of the abundance of specific transposon mutants under one or more selective conditions. The approaches themselves only vary in the transposon used for mutagenesis, and in the methodology used for sequencing library preparation. In this review, we discuss how TIS approaches have been used to facilitate a major shift in our fundamental understanding of bacterial biology in a range of areas. We focus on several aspects including pathogenesis, biofilm development, polymicrobial interactions in various ecosystems, and antimicrobial resistance. These studies have provided new insight into bacterial physiology and revealed predicted functions for hundreds of genes previously representing genomic 'dark matter'. We also discuss how TIS approaches have been used to understand complex bacterial systems and interactions and how future developments of TIS could continue to accelerate and enrich our understanding of bacterial biology.
Collapse
Affiliation(s)
- Laura M Nolan
- Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University, Singapore; School of Biological Sciences, Nanyang Technological University, Singapore; Quadram Institute Bioscience, Norwich Research Park, Norwich, UK.
| | - Mark A Webber
- Quadram Institute Bioscience, Norwich Research Park, Norwich, UK; Norwich Medical School, Norwich Research Park, Norwich, UK; Center for Microbial Interactions, Norwich Research Park, Norwich, UK
| | - Alain Filloux
- Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University, Singapore; School of Biological Sciences, Nanyang Technological University, Singapore; Lee Kon Chian School of Medicine, Nanyang Technological University, Singapore; Imperial, Centre for Bacterial Resistance Biology, London, UK
| |
Collapse
|
2
|
Li Y, Xu Y, Jin C, Qiu J, Jiao X, Pan Z, Guo Y. Salmonella-NLRP3 Inflammasome Crosstalk: Host Defense Activation Versus Bacterial Immune Evasion Strategies. J Inflamm Res 2025; 18:5133-5148. [PMID: 40255664 PMCID: PMC12009050 DOI: 10.2147/jir.s519902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Accepted: 04/10/2025] [Indexed: 04/22/2025] Open
Abstract
The innate immune system plays a crucial role in defending against Salmonella infection. Inflammasomes are macromolecular complexes that assemble in response to the recognition of pathogen- or danger-associated molecular patterns. These complexes serve as signaling platforms for the activation of inflammatory Caspases, which subsequently triggers the maturation and secretion of the pro-inflammatory cytokines IL-1β and IL-18. This process also initiates pyroptosis, a highly inflammatory form of programmed cell death characterized by lytic cell lysis. Salmonella are intracellular pathogens that proliferate within epithelial cells and macrophages, posing a significant public health risk in both developed and developing countries. During Salmonella infection, the canonical NLRP3 and NLRC4 inflammasome, as well as non-canonical inflammasome, are activated. Unlike NLRC4 and non-canonical inflammasomes, which play crucial roles during intestinal infection phases, the role of NLRP3 inflammasome in resisting Salmonella infection demonstrates a higher degree of complexity and uncertainty. Nonetheless, the activation of NLRP3 inflammasome, along with the downstream innate and adaptive responses, form a robust host immune barrier against potential pathogens. Therefore, successful pathogens must evolve multiple mechanisms to circumvent or counteract these immune barriers. Here we review and discuss the mechanisms of NLRP3 inflammasome activation triggered by intracellular Salmonella, as well as the multiple strategies employed by Salmonella to avoid or delay NLRP3 inflammasome activation. A deeper understanding of how NLRP3 inflammasomes recognize Salmonella and how pathogens evade NLRP3 activation has the potential to facilitate the development of novel prevention and control measures for Salmonella infection.
Collapse
Affiliation(s)
- Yuxuan Li
- School of Nursing School of Public Health, Yangzhou University, Jiangsu, People’s Republic of China
| | - Ying Xu
- The Department of Economics and Management, Jiangsu College of Tourism, Jiangsu, People’s Republic of China
| | - Cheng Jin
- School of Nursing School of Public Health, Yangzhou University, Jiangsu, People’s Republic of China
| | - Jiayi Qiu
- School of Nursing School of Public Health, Yangzhou University, Jiangsu, People’s Republic of China
| | - Xinan Jiao
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, Jiangsu, 225009, People’s Republic of China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, Jiangsu, 225009, People’s Republic of China
- Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture of China, Yangzhou University, Yangzhou, Jiangsu, People’s Republic of China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education, Yangzhou University, Yangzhou, Jiangsu, People’s Republic of China
| | - Zhiming Pan
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, Jiangsu, 225009, People’s Republic of China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, Jiangsu, 225009, People’s Republic of China
- Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture of China, Yangzhou University, Yangzhou, Jiangsu, People’s Republic of China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education, Yangzhou University, Yangzhou, Jiangsu, People’s Republic of China
| | - Yaxin Guo
- School of Nursing School of Public Health, Yangzhou University, Jiangsu, People’s Republic of China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, Jiangsu, 225009, People’s Republic of China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, Jiangsu, 225009, People’s Republic of China
- Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture of China, Yangzhou University, Yangzhou, Jiangsu, People’s Republic of China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education, Yangzhou University, Yangzhou, Jiangsu, People’s Republic of China
| |
Collapse
|
3
|
Egan MS, O'Rourke EA, Mageswaran SK, Zuo B, Martynyuk I, Demissie T, Hunter EN, Bass AR, Chang YW, Brodsky IE, Shin S. Inflammasomes primarily restrict cytosolic Salmonella replication within human macrophages. eLife 2025; 12:RP90107. [PMID: 40162563 PMCID: PMC11957546 DOI: 10.7554/elife.90107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/02/2025] Open
Abstract
Salmonella enterica serovar Typhimurium is a facultative intracellular pathogen that utilizes its type III secretion systems (T3SSs) to inject virulence factors into host cells and colonize the host. In turn, a subset of cytosolic immune receptors respond to T3SS ligands by forming multimeric signaling complexes called inflammasomes, which activate caspases that induce interleukin-1 (IL-1) family cytokine release and an inflammatory form of cell death called pyroptosis. Human macrophages mount a multifaceted inflammasome response to Salmonella infection that ultimately restricts intracellular bacterial replication. However, how inflammasomes restrict Salmonella replication remains unknown. We find that caspase-1 is essential for mediating inflammasome responses to Salmonella and restricting bacterial replication within human macrophages, with caspase-4 contributing as well. We also demonstrate that the downstream pore-forming protein gasdermin D (GSDMD) and Ninjurin-1 (NINJ1), a mediator of terminal cell lysis, play a role in controlling Salmonella replication in human macrophages. Notably, in the absence of inflammasome responses, we observed hyperreplication of Salmonella within the cytosol of infected cells as well as increased bacterial replication within vacuoles, suggesting that inflammasomes control Salmonella replication primarily within the cytosol and also within vacuoles. These findings reveal that inflammatory caspases and pyroptotic factors mediate inflammasome responses that restrict the subcellular localization of intracellular Salmonella replication within human macrophages.
Collapse
Affiliation(s)
- Marisa S Egan
- Department of Microbiology, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Emily A O'Rourke
- Department of Microbiology, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Shrawan Kumar Mageswaran
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
- Institute of Structural Biology, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Biao Zuo
- Institute of Structural Biology, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
- Electron Microscopy Resource Laboratory, Department of Biochemistry & Biophysics, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Inna Martynyuk
- Institute of Structural Biology, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
- Electron Microscopy Resource Laboratory, Department of Biochemistry & Biophysics, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Tabitha Demissie
- Department of Microbiology, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Emma N Hunter
- Department of Microbiology, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Antonia R Bass
- Department of Microbiology, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Yi-Wei Chang
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
- Institute of Structural Biology, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Igor E Brodsky
- Department of Pathobiology, University of Pennsylvania School of Veterinary MedicinePhiladelphiaUnited States
| | - Sunny Shin
- Department of Microbiology, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| |
Collapse
|
4
|
Mathieu-Bégné E, Gattis S, Ebert D. Genetic basis of resistance in hosts facing alternative infection strategies by a virulent bacterial pathogen. G3 (BETHESDA, MD.) 2025; 15:jkae302. [PMID: 39707909 PMCID: PMC11917490 DOI: 10.1093/g3journal/jkae302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 12/12/2024] [Accepted: 12/13/2024] [Indexed: 12/23/2024]
Abstract
Having alternative infection routes is thought to help parasites circumvent host resistance, provided that these routes are associated with different host resistance loci. This study tests this postulate by examining whether alternate infection routes of the parasite Pasteuria ramosa are linked to distinct resistance loci in its crustacean host, Daphnia magna. We focus on the P. ramosa isolate P15, which can attach and penetrate the host through either the hindgut or the foregut. Using a global panel of 174 D. magna genotypes supplemented with breeding experiments, we analyzed resistance patterns for each of these infection routes. Our findings confirm our hypothesis in D. magna, hindgut attachment is determined by the D locus, while foregut attachment is controlled by a newly identified G locus. We established a gene model for the G locus that indicated Mendelian segregation and epistatic interaction with at least one other resistance locus for P. ramosa, the C locus. Using genomic Pool-sequencing data, we localized the G locus within a known Pasteuria Resistance Complex on chromosome 4 of D. magna, whereas the D locus is on chromosome 7. Two candidate genes for the G locus, belonging to the Glycosyltransferase gene family, were identified. Our study sheds new light on host-parasite coevolution and enhances our understanding of how parasites evolve infection strategies.
Collapse
Affiliation(s)
- Eglantine Mathieu-Bégné
- Department of Environmental Sciences, Zoology, University of Basel, CH-4051 Basel, Switzerland
| | - Sabrina Gattis
- Department of Environmental Sciences, Zoology, University of Basel, CH-4051 Basel, Switzerland
- School of Zoology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
| | - Dieter Ebert
- Department of Environmental Sciences, Zoology, University of Basel, CH-4051 Basel, Switzerland
| |
Collapse
|
5
|
Worley MJ. Salmonella Type III Secretion System Effectors. Int J Mol Sci 2025; 26:2611. [PMID: 40141253 PMCID: PMC11942329 DOI: 10.3390/ijms26062611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Revised: 03/05/2025] [Accepted: 03/12/2025] [Indexed: 03/28/2025] Open
Abstract
Salmonella is estimated to infect between 200 million and over 1 billion people per year. The exact number is not known, as many cases go unreported. Integral to the pathogenesis of Salmonella, as well as numerous other Gram-negative pathogens, is its type III effectors. Salmonella possesses two distinct type III secretion systems, encoded by Salmonella pathogenicity island-1 and Salmonella pathogenicity island-2. Together, they secrete at least 49 type III effectors into host cells that are collectively responsible for many of the virulence attributes of this pathogen. These virulence factors facilitate the invasion of host cells, induce and attenuate inflammation, and change the migratory properties of infected phagocytes, among other things. The effects of all type III effectors on Salmonella virulence are discussed.
Collapse
Affiliation(s)
- Micah J Worley
- Department of Biology, University of Louisville, Louisville, KY 40292, USA
| |
Collapse
|
6
|
Newson JPM, Gürtler F, Piffaretti P, Meyer A, Sintsova A, Barthel M, Steiger Y, McHugh SC, Enz U, Alto NM, Sunagawa S, Hardt WD. Salmonella multimutants enable efficient identification of SPI-2 effector protein function in gut inflammation and systemic colonization. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.14.628483. [PMID: 39713370 PMCID: PMC11661221 DOI: 10.1101/2024.12.14.628483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
Salmonella enterica spp. rely on translocation of effector proteins through the SPI-2 encoded type III secretion system (T3SS) to achieve pathogenesis. More than 30 effectors contribute to manipulation of host cells through diverse mechanisms, but interdependency or redundancy between effectors complicates the discovery of effector phenotypes using single mutant strains. Here, we engineer six mutant strains to be deficient in cohorts of SPI-2 effector proteins, as defined by their reported function. Using various animal models of infection, we show that three principle phenotypes define the functional contribution of the SPI-2 T3SS to infection. Multimutant strains deficient for intracellular replication, for manipulation of host cell defences, or for expression of virulence plasmid effectors all showed strong attenuation in vivo, while mutants representing approximately half of the known effector complement showed phenotypes similar to the wild-type parent strain. By additionally removing the SPI-1 T3SS, we find cohorts of effector proteins that contribute to SPI-2 T3SS-driven enhancement of gut inflammation. Further, we provide an example of how iterative mutation can be used to find a minimal number of effector deletions required for attenuation, and thus establish that the SPI-2 effectors SopD2 and GtgE are critical for the promotion of gut inflammation and mucosal pathology. This strategy provides a powerful toolset for simultaneous parallel screening of all known SPI-2 effectors in a single experimental context, and further facilitates the identification of the responsible effectors, and thereby provides an efficient approach to study how individual effectors contribute to disease.
Collapse
Affiliation(s)
| | - Flavia Gürtler
- Institute of Microbiology, ETH Zurich, Zurich, Switzerland
- Current address: Epidemiology, Biostatistics & Prevention Institute, University of Zurich, Zurich, Switzerland
| | | | - Annina Meyer
- Institute of Microbiology, ETH Zurich, Zurich, Switzerland
- Current address: Institute of Food, Nutrition and Health, ETH Zurich, Zurich, Switzerland
| | - Anna Sintsova
- Institute of Microbiology, ETH Zurich, Zurich, Switzerland
| | - Manja Barthel
- Institute of Microbiology, ETH Zurich, Zurich, Switzerland
| | - Yves Steiger
- Institute of Microbiology, ETH Zurich, Zurich, Switzerland
| | - Sarah C McHugh
- Institute of Microbiology, ETH Zurich, Zurich, Switzerland
- Current address: Department of Fundamental Microbiology, University of Lausanne, Lausanne, Switzerland
| | - Ursina Enz
- Institute of Microbiology, ETH Zurich, Zurich, Switzerland
| | - Neal M Alto
- Department of Microbiology, University of Texas (UT) Southwestern Medical Center, Dallas, TX, United States
| | | | | |
Collapse
|
7
|
Han J, Aljahdali N, Zhao S, Tang H, Harbottle H, Hoffmann M, Frye JG, Foley SL. Infection biology of Salmonella enterica. EcoSal Plus 2024; 12:eesp00012023. [PMID: 38415623 PMCID: PMC11636313 DOI: 10.1128/ecosalplus.esp-0001-2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 07/31/2023] [Indexed: 02/29/2024]
Abstract
Salmonella enterica is the leading cause of bacterial foodborne illness in the USA, with an estimated 95% of salmonellosis cases due to the consumption of contaminated food products. Salmonella can cause several different disease syndromes, with the most common being gastroenteritis, followed by bacteremia and typhoid fever. Among the over 2,600 currently identified serotypes/serovars, some are mostly host-restricted and host-adapted, while the majority of serotypes can infect a broader range of host species and are associated with causing both livestock and human disease. Salmonella serotypes and strains within serovars can vary considerably in the severity of disease that may result from infection, with some serovars that are more highly associated with invasive disease in humans, while others predominantly cause mild gastroenteritis. These observed clinical differences may be caused by the genetic make-up and diversity of the serovars. Salmonella virulence systems are very complex containing several virulence-associated genes with different functions that contribute to its pathogenicity. The different clinical syndromes are associated with unique groups of virulence genes, and strains often differ in the array of virulence traits they display. On the chromosome, virulence genes are often clustered in regions known as Salmonella pathogenicity islands (SPIs), which are scattered throughout different Salmonella genomes and encode factors essential for adhesion, invasion, survival, and replication within the host. Plasmids can also carry various genes that contribute to Salmonella pathogenicity. For example, strains from several serovars associated with significant human disease, including Choleraesuis, Dublin, Enteritidis, Newport, and Typhimurium, can carry virulence plasmids with genes contributing to attachment, immune system evasion, and other roles. The goal of this comprehensive review is to provide key information on the Salmonella virulence, including the contributions of genes encoded in SPIs and plasmids during Salmonella pathogenesis.
Collapse
Affiliation(s)
- Jing Han
- National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, Arkansas, USA
| | - Nesreen Aljahdali
- National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, Arkansas, USA
- Biological Science Department, College of Science, King Abdul-Aziz University, Jeddah, Saudi Arabia
| | - Shaohua Zhao
- Center for Veterinary Medicine, U.S. Food and Drug Administration, Rockville, Maryland, USA
| | - Hailin Tang
- National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, Arkansas, USA
| | - Heather Harbottle
- Center for Veterinary Medicine, U.S. Food and Drug Administration, Rockville, Maryland, USA
| | - Maria Hoffmann
- Center for Food Safety and Applied Nutrition, U.S. Food and Drug Administration, College Park, Maryland, USA
| | - Jonathan G. Frye
- Agricutlutral Research Service, U.S. Department of Agriculture, Athens, Georgia, USA
| | - Steven L. Foley
- National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, Arkansas, USA
| |
Collapse
|
8
|
Egan MS, O’Rourke EA, Mageswaran SK, Zuo B, Martynyuk I, Demissie T, Hunter EN, Bass AR, Chang YW, Brodsky IE, Shin S. Inflammasomes primarily restrict cytosolic Salmonella replication within human macrophages. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.07.17.549348. [PMID: 37503120 PMCID: PMC10370064 DOI: 10.1101/2023.07.17.549348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Salmonella enterica serovar Typhimurium is a facultative intracellular pathogen that utilizes its type III secretion systems (T3SSs) to inject virulence factors into host cells and colonize the host. In turn, a subset of cytosolic immune receptors respond to T3SS ligands by forming multimeric signaling complexes called inflammasomes, which activate caspases that induce interleukin-1 (IL-1) family cytokine release and an inflammatory form of cell death called pyroptosis. Human macrophages mount a multifaceted inflammasome response to Salmonella infection that ultimately restricts intracellular bacterial replication. However, how inflammasomes restrict Salmonella replication remains unknown. We find that caspase-1 is essential for mediating inflammasome responses to Salmonella and restricting bacterial replication within human macrophages, with caspase-4 contributing as well. We also demonstrate that the downstream pore-forming protein gasdermin D (GSDMD) and Ninjurin-1 (NINJ1), a mediator of terminal cell lysis, play a role in controlling Salmonella replication in human macrophages. Notably, in the absence of inflammasome responses, we observed hyperreplication of Salmonella within the cytosol of infected cells as well as increased bacterial replication within vacuoles, suggesting that inflammasomes control Salmonella replication primarily within the cytosol and also within vacuoles. These findings reveal that inflammatory caspases and pyroptotic factors mediate inflammasome responses that restrict the subcellular localization of intracellular Salmonella replication within human macrophages.
Collapse
Affiliation(s)
- Marisa S. Egan
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Emily A. O’Rourke
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Shrawan Kumar Mageswaran
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Biao Zuo
- Electron Microscopy Resource Laboratory, Department of Biochemistry & Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Inna Martynyuk
- Electron Microscopy Resource Laboratory, Department of Biochemistry & Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Tabitha Demissie
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Emma N. Hunter
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Antonia R. Bass
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Yi-Wei Chang
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Igor E. Brodsky
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA
| | - Sunny Shin
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
9
|
Cheng G, Jian S, Li W, Yan L, Chen T, Cheng T, Liu Z, Ye G, Tang H, Zhang L. Epigallocatechin gallate protects mice from Salmonella enterica ser. Typhimurium infection by modulating bacterial virulence through quorum sensing inhibition. Front Cell Infect Microbiol 2024; 14:1432111. [PMID: 39479281 PMCID: PMC11521958 DOI: 10.3389/fcimb.2024.1432111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 09/23/2024] [Indexed: 11/02/2024] Open
Abstract
Salmonella enterica ser. Typhimurium is a common pathogen that poses a considerable public health threat, contributing to severe gastrointestinal diseases and widespread foodborne illnesses. The virulence of S. Typhimurium is regulated by quorum sensing (QS) and the type III secretion system (T3SS). This study investigated the inhibitory effects and anti-QS activity of epigallocatechin gallate (EGCG), which is a bioactive ingredient found in green tea, on the virulence of S. Typhimurium. In vitro bacterial experiments demonstrated that EGCG inhibited the production of autoinducers, biofilm formation, and flagellar activity by downregulating the expression of AI-1, AI-2, Salmonella pathogenicity islands (SPI)-1, SPI-2, and genes related to flagella, fimbriae, and curli fibers. In a mouse model of S. Typhimurium-induced enteritis, EGCG considerably reduced intestinal colonization by S. Typhimurium and alleviated intestinal damage. In conclusion, EGCG protects the intestines of mice infected with S. Typhimurium by inhibiting QS-induced virulence gene expression, demonstrating its potential as a therapeutic agent for controlling S. Typhimurium infections.
Collapse
Affiliation(s)
- Guoqiang Cheng
- Animal Experiment Center, Sichuan Academy of Chinese Medicine Sciences, Chengdu, China
| | - Shanqiu Jian
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Wen Li
- Department of Science, Sichuan Academy of Agricultural Sciences, Chengdu, China
| | - Liangchun Yan
- Animal Experiment Center, Sichuan Academy of Chinese Medicine Sciences, Chengdu, China
| | - Tiezhu Chen
- Animal Experiment Center, Sichuan Academy of Chinese Medicine Sciences, Chengdu, China
| | - Tingting Cheng
- Animal Experiment Center, Sichuan Academy of Chinese Medicine Sciences, Chengdu, China
| | - Zongxiu Liu
- Department of Innovation, Chengdu Qiankun Animal Pharmaceutical Co., Ltd, Chengdu, China
| | - Gang Ye
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Huaqiao Tang
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Li Zhang
- Animal Experiment Center, Sichuan Academy of Chinese Medicine Sciences, Chengdu, China
| |
Collapse
|
10
|
Yu XJ, Xie H, Li Y, Liu M, Hou R, Predeus AV, Perez Sepulveda BM, Hinton JCD, Holden DW, Thurston TLM. Modulation of Salmonella virulence by a novel SPI-2 injectisome effector that interacts with the dystrophin-associated protein complex. mBio 2024; 15:e0112824. [PMID: 38904384 PMCID: PMC11253597 DOI: 10.1128/mbio.01128-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 05/16/2024] [Indexed: 06/22/2024] Open
Abstract
The injectisome encoded by Salmonella pathogenicity island 2 (SPI-2) had been thought to translocate 28 effectors. Here, we used a proteomic approach to characterize the secretome of a clinical strain of invasive non-typhoidal Salmonella enterica serovar Enteritidis that had been mutated to cause hyper-secretion of the SPI-2 injectisome effectors. Along with many known effectors, we discovered the novel SseM protein. sseM is widely distributed among the five subspecies of Salmonella enterica, is found in many clinically relevant serovars, and is co-transcribed with pipB2, a SPI-2 effector gene. The translocation of SseM required a functional SPI-2 injectisome. Following expression in human cells, SseM interacted with five components of the dystrophin-associated protein complex (DAPC), namely, β-2-syntrophin, utrophin/dystrophin, α-catulin, α-dystrobrevin, and β-dystrobrevin. The interaction between SseM and β-2-syntrophin and α-dystrobrevin was verified in Salmonella Typhimurium-infected cells and relied on the postsynaptic density-95/discs large/zonula occludens-1 (PDZ) domain of β-2-syntrophin and a sequence corresponding to a PDZ-binding motif (PBM) in SseM. A ΔsseM mutant strain had a small competitive advantage over the wild-type strain in the S. Typhimurium/mouse model of systemic disease. This phenotype was complemented by a plasmid expressing wild-type SseM from S. Typhimurium or S. Enteritidis and was dependent on the PBM of SseM. Therefore, a PBM within a Salmonella effector mediates interactions with the DAPC and modulates the systemic growth of bacteria in mice. Furthermore, the ΔsseM mutant strain displayed enhanced replication in bone marrow-derived macrophages, demonstrating that SseM restrains intracellular bacterial growth to modulate Salmonella virulence. IMPORTANCE In Salmonella enterica, the injectisome machinery encoded by Salmonella pathogenicity island 2 (SPI-2) is conserved among the five subspecies and delivers proteins (effectors) into host cells, which are required for Salmonella virulence. The identification and functional characterization of SPI-2 injectisome effectors advance our understanding of the interplay between Salmonella and its host(s). Using an optimized method for preparing secreted proteins and a clinical isolate of the invasive non-typhoidal Salmonella enterica serovar Enteritidis strain D24359, we identified 22 known SPI-2 injectisome effectors and one new effector-SseM. SseM modulates bacterial growth during murine infection and has a sequence corresponding to a postsynaptic density-95/discs large/zonula occludens-1 (PDZ)-binding motif that is essential for interaction with the PDZ-containing host protein β-2-syntrophin and other components of the dystrophin-associated protein complex (DAPC). To our knowledge, SseM is unique among Salmonella effectors in containing a functional PDZ-binding motif and is the first bacterial protein to target the DAPC.
Collapse
Affiliation(s)
- Xiu-Jun Yu
- Department of Infectious Disease, Centre for Bacterial Resistance Biology, Imperial College London, London, United Kingdom
| | - Haixia Xie
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, Hubei, China
| | - Yan Li
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Mei Liu
- Department of Infectious Disease, Centre for Bacterial Resistance Biology, Imperial College London, London, United Kingdom
| | - Ruhong Hou
- Department of Infectious Disease, Centre for Bacterial Resistance Biology, Imperial College London, London, United Kingdom
| | - Alexander V. Predeus
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Blanca M. Perez Sepulveda
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Jay C. D. Hinton
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
| | - David W. Holden
- Department of Infectious Disease, Centre for Bacterial Resistance Biology, Imperial College London, London, United Kingdom
| | - Teresa L. M. Thurston
- Department of Infectious Disease, Centre for Bacterial Resistance Biology, Imperial College London, London, United Kingdom
| |
Collapse
|
11
|
Oke MT, D’Costa VM. Functional Divergence of the Paralog Salmonella Effector Proteins SopD and SopD2 and Their Contributions to Infection. Int J Mol Sci 2024; 25:4191. [PMID: 38673776 PMCID: PMC11050076 DOI: 10.3390/ijms25084191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 04/05/2024] [Accepted: 04/05/2024] [Indexed: 04/28/2024] Open
Abstract
Salmonella enterica is a leading cause of bacterial food-borne illness in humans and is responsible for millions of cases annually. A critical strategy for the survival of this pathogen is the translocation of bacterial virulence factors termed effectors into host cells, which primarily function via protein-protein interactions with host proteins. The Salmonella genome encodes several paralogous effectors believed to have arisen from duplication events throughout the course of evolution. These paralogs can share structural similarities and enzymatic activities but have also demonstrated divergence in host cell targets or interaction partners and contributions to the intracellular lifecycle of Salmonella. The paralog effectors SopD and SopD2 share 63% amino acid sequence similarity and extensive structural homology yet have demonstrated divergence in secretion kinetics, intracellular localization, host targets, and roles in infection. SopD and SopD2 target host Rab GTPases, which represent critical regulators of intracellular trafficking that mediate diverse cellular functions. While SopD and SopD2 both manipulate Rab function, these paralogs display differences in Rab specificity, and the effectors have also evolved multiple mechanisms of action for GTPase manipulation. Here, we highlight this intriguing pair of paralog effectors in the context of host-pathogen interactions and discuss how this research has presented valuable insights into effector evolution.
Collapse
Affiliation(s)
- Mosopefoluwa T. Oke
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada
- Centre for Infection, Immunity and Inflammation, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Vanessa M. D’Costa
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada
- Centre for Infection, Immunity and Inflammation, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| |
Collapse
|
12
|
Grote A, Piscon B, Manson AL, Adani B, Cohen H, Livny J, Earl AM, Gal-Mor O. Persistent Salmonella infections in humans are associated with mutations in the BarA/SirA regulatory pathway. Cell Host Microbe 2024; 32:79-92.e7. [PMID: 38211565 PMCID: PMC11410052 DOI: 10.1016/j.chom.2023.12.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 10/04/2023] [Accepted: 12/06/2023] [Indexed: 01/13/2024]
Abstract
Several bacterial pathogens, including Salmonella enterica, can cause persistent infections in humans by mechanisms that are poorly understood. By comparing genomes of isolates longitudinally collected from 256 prolonged salmonellosis patients, we identified repeated mutations in global regulators, including the barA/sirA two-component regulatory system, across multiple patients and Salmonella serovars. Comparative RNA-seq analysis revealed that distinct mutations in barA/sirA led to diminished expression of Salmonella pathogenicity islands 1 and 4 genes, which are required for Salmonella invasion and enteritis. Moreover, barA/sirA mutants were attenuated in an acute salmonellosis mouse model and induced weaker transcription of host immune responses. In contrast, in a persistent infection mouse model, these mutants exhibited long-term colonization and prolonged shedding. Taken together, these findings suggest that selection of mutations in global virulence regulators facilitates persistent Salmonella infection in humans, by attenuating Salmonella virulence and inducing a weaker host inflammatory response.
Collapse
Affiliation(s)
- Alexandra Grote
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Bar Piscon
- Infectious Diseases Research Laboratory, Sheba Medical Center, Tel-Hashomer, Israel; Department of Clinical Microbiology and Immunology, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Abigail L Manson
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Boaz Adani
- Infectious Diseases Research Laboratory, Sheba Medical Center, Tel-Hashomer, Israel
| | - Helit Cohen
- Infectious Diseases Research Laboratory, Sheba Medical Center, Tel-Hashomer, Israel
| | - Jonathan Livny
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
| | - Ashlee M Earl
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
| | - Ohad Gal-Mor
- Infectious Diseases Research Laboratory, Sheba Medical Center, Tel-Hashomer, Israel; Department of Clinical Microbiology and Immunology, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
13
|
Feng Z, Wang L, Guan Q, Chu X, Luo ZQ. Acinetobacter baumannii coordinates central metabolism, plasmid dissemination, and virulence by sensing nutrient availability. mBio 2023; 14:e0227623. [PMID: 37855599 PMCID: PMC10746170 DOI: 10.1128/mbio.02276-23] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 09/01/2023] [Indexed: 10/20/2023] Open
Abstract
IMPORTANCE Plasmid conjugation is known to be an energy-expensive process, but our understanding of the molecular linkage between conjugation and metabolism is limited. Our finding reveals that Acinetobacter baumannii utilizes a two-component system to co-regulate metabolism, plasmid transfer, and virulence by sensing reaction intermediates of key metabolic pathways, which suggests that nutrient availability dictates not only bacterial proliferation but also horizontal gene transfer. The identification of Dot/Icm-like proteins as components of a conjugation system involved in the dissemination of antibiotic-resistance genes by A. baumannii has provided important targets for the development of agents capable of inhibiting virulence and the spread of anti-microbial-resistance genes in bacterial communities.
Collapse
Affiliation(s)
- Zhengshan Feng
- Department of Respiratory Medicine, Center of Infectious Diseases and Pathogen Biology, Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, The First Hospital of Jilin University, Changchun, China
| | - Lidong Wang
- Department of Respiratory Medicine, Center of Infectious Diseases and Pathogen Biology, Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, The First Hospital of Jilin University, Changchun, China
| | - Qingtian Guan
- Bioinformatics Laboratory, The First Hospital of Jilin University, Changchun, China
| | - Xiao Chu
- Department of Respiratory Medicine, Center of Infectious Diseases and Pathogen Biology, Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, The First Hospital of Jilin University, Changchun, China
| | - Zhao-Qing Luo
- Department of Biological Sciences, Purdue University, West Lafayette, Indiana, USA
| |
Collapse
|
14
|
Bryant OJ, Lastovka F, Powell J, Chung BYW. The distinct translational landscapes of gram-negative Salmonella and gram-positive Listeria. Nat Commun 2023; 14:8167. [PMID: 38071303 PMCID: PMC10710512 DOI: 10.1038/s41467-023-43759-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Accepted: 11/17/2023] [Indexed: 12/18/2023] Open
Abstract
Translational control in pathogenic bacteria is fundamental to gene expression and affects virulence and other infection phenotypes. We used an enhanced ribosome profiling protocol coupled with parallel transcriptomics to capture accurately the global translatome of two evolutionarily distant pathogenic bacteria-the Gram-negative bacterium Salmonella and the Gram-positive bacterium Listeria. We find that the two bacteria use different mechanisms to translationally regulate protein synthesis. In Salmonella, in addition to the expected correlation between translational efficiency and cis-regulatory features such as Shine-Dalgarno (SD) strength and RNA secondary structure around the initiation codon, our data reveal an effect of the 2nd and 3rd codons, where the presence of tandem lysine codons (AAA-AAA) enhances translation in both Salmonella and E. coli. Strikingly, none of these features are seen in efficiently translated Listeria transcripts. Instead, approximately 20% of efficiently translated Listeria genes exhibit 70 S footprints seven nt upstream of the authentic start codon, suggesting that these genes may be subject to a novel translational initiation mechanism. Our results show that SD strength is not a direct hallmark of translational efficiency in all bacteria. Instead, Listeria has evolved additional mechanisms to control gene expression level that are distinct from those utilised by Salmonella and E. coli.
Collapse
Affiliation(s)
- Owain J Bryant
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QP, UK
- Centre for Structural Biology, National Cancer Institute, 21702, Frederick, MD, USA
| | - Filip Lastovka
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QP, UK
| | - Jessica Powell
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QP, UK
| | - Betty Y-W Chung
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QP, UK.
| |
Collapse
|
15
|
Martins IM, Seribelli AA, Machado Ribeiro TR, da Silva P, Lustri BC, Hernandes RT, Falcão JP, Moreira CG. Invasive non-typhoidal Salmonella (iNTS) aminoglycoside-resistant ST313 isolates feature unique pathogenic mechanisms to reach the bloodstream. INFECTION, GENETICS AND EVOLUTION : JOURNAL OF MOLECULAR EPIDEMIOLOGY AND EVOLUTIONARY GENETICS IN INFECTIOUS DISEASES 2023; 116:105519. [PMID: 37890808 DOI: 10.1016/j.meegid.2023.105519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 10/19/2023] [Accepted: 10/24/2023] [Indexed: 10/29/2023]
Abstract
Invasive non-typhoidal Salmonella (iNTS) from the clonal type ST313 (S. Typhimurium ST313) is the primary cause of invasive salmonellosis in Africa. Recently, in Brazil, iNTS ST313 strains have been isolated from different sources, but there is a lack of understanding of the mechanisms behind how these gut bacteria can break the gut barrier and reach the patient's bloodstream. Here, we compare 13 strains of S. Typhimurium ST313, previously unreported isolates, from human blood cultures, investigating aspects of virulence and mechanisms of resistance. Initially, RNAseq analyses between ST13-blood isolate and SL1344 (ST19) prototype revealed 15 upregulated genes directly related to cellular invasion and replication, such as sopD2, sifB, and pipB. Limited information is available about S. Typhimurium ST313 pathogenesis and epidemiology, especially related to the global distribution of strains. Herein, the correlation of strains isolated from different sources in Brazil was employed to compare clinical and non-clinical isolates, a total of 22 genomes were studied by single nucleotide polymorphism (SNPs). The epidemiological analysis of 22 genomes of S. Typhimurium ST313 strains grouped them into three distinct clusters (A, B, and C) by SNP analysis, where cluster A comprised five, group B six, and group C 11. The 13 clinical blood isolates were all resistant to streptomycin, 92.3% of strains were resistant to ampicillin and 15.39% were resistant to kanamycin. The resistance genes acrA, acrB, mdtK, emrB, emrR, mdsA, and mdsB related to the production of efflux pumps were detected in all (100%) strains studied, similar to pathogenic traits investigated. In conclusion, we evidenced that S. Typhimurium ST313 strains isolated in Brazil have unique epidemiology. The elevated frequencies of virulence genes such as sseJ, sopD2, and pipB are a major concern in these Brazilian isolates, showing a higher pathogenic potential.
Collapse
Affiliation(s)
- Isabela Mancini Martins
- Faculdade de Ciências Farmacêuticas de Araraquara, Universidade Estadual Paulista- UNESP- Departamento de Ciências Biológicas, Araraquara, SP, Brazil
| | - Amanda Aparecida Seribelli
- Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo- USP, Ribeirão Preto, SP, Brazil
| | - Tamara R Machado Ribeiro
- Faculdade de Ciências Farmacêuticas de Araraquara, Universidade Estadual Paulista- UNESP- Departamento de Ciências Biológicas, Araraquara, SP, Brazil
| | - Patrick da Silva
- Faculdade de Ciências Farmacêuticas de Araraquara, Universidade Estadual Paulista- UNESP- Departamento de Ciências Biológicas, Araraquara, SP, Brazil
| | - Bruna Cardinali Lustri
- Faculdade de Ciências Farmacêuticas de Araraquara, Universidade Estadual Paulista- UNESP- Departamento de Ciências Biológicas, Araraquara, SP, Brazil
| | - Rodrigo T Hernandes
- Instituto de Biociências, Universidade Estadual Paulista- UNESP, Botucatu, SP, Brazil
| | - Juliana Pfrimer Falcão
- Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo- USP, Ribeirão Preto, SP, Brazil.
| | - Cristiano Gallina Moreira
- Faculdade de Ciências Farmacêuticas de Araraquara, Universidade Estadual Paulista- UNESP- Departamento de Ciências Biológicas, Araraquara, SP, Brazil; Department of Biological Sciences, Louisiana State University, Baton Rouge, LA, USA.
| |
Collapse
|
16
|
Worley MJ. Salmonella Bloodstream Infections. Trop Med Infect Dis 2023; 8:487. [PMID: 37999606 PMCID: PMC10675298 DOI: 10.3390/tropicalmed8110487] [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: 09/19/2023] [Revised: 10/23/2023] [Accepted: 10/24/2023] [Indexed: 11/25/2023] Open
Abstract
Salmonella is a major foodborne pathogen of both animals and humans. This bacterium is responsible for considerable morbidity and mortality world-wide. Different serovars of this genus cause diseases ranging from self-limiting gastroenteritis to a potentially fatal systemic disease known as enteric fever. Gastrointestinal infections with Salmonella are usually self-limiting and rarely require medical intervention. Bloodstream infections, on the other hand, are often fatal even with hospitalization. This review describes the routes and underlying mechanisms of the extraintestinal dissemination of Salmonella and the chronic infections that sometimes result. It includes information on the pathogenicity islands and individual virulence factors involved in systemic dissemination as well as a discussion of the host factors that mediate susceptibility. Also, the major outbreaks of invasive Salmonella disease in the tropics are described.
Collapse
Affiliation(s)
- Micah J Worley
- Department of Biology, University of Louisville, Louisville, KY 40292, USA
| |
Collapse
|
17
|
Pandeya A, Zhang Y, Cui J, Yang L, Li J, Zhang G, Wu C, Li Z, Wei Y. Inflammasome activation and pyroptosis mediate coagulopathy and inflammation in Salmonella systemic infection. Microbiol Res 2023; 275:127460. [PMID: 37467711 PMCID: PMC10693354 DOI: 10.1016/j.micres.2023.127460] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Revised: 07/09/2023] [Accepted: 07/12/2023] [Indexed: 07/21/2023]
Abstract
Inflammasome activation is a critical defense mechanism against bacterial infection. Previous studies suggest that inflammasome activation protects against Salmonella oral infection. Here we find inflammasome activation plays a critical role in the pathogenesis of Salmonella systemic infection. We show that in a systemic infection model by i.p. injection of Salmonella, deficiency of caspase-1 or gasdermin-D prolonged survival time, reduced plasma concentrations of the proinflammatory cytokines IL-1β, IL-6 and TNFα. These deficiencies also protected against coagulopathy during Salmonella infection as evidenced by diminished prolongation of prothrombin time and increase in plasma thrombin-antithrombin complex concentrations in the caspase-1 or gasdermin-D deficient mice. Activation of the NAIP/NLRC4 inflammasome by flagellin and/or the components of the SPI1 type 3 secretion system played a critical role in Salmonella-induced coagulopathy. In the absence of flagellin and SPI1, the Salmonella mutant strain still triggered coagulopathy through the caspase-11/NLRP3 pathway. Our results reveal a previously undisclosed role of the inflammasomes and pyroptosis in the pathogenesis of Salmonella systemic infection.
Collapse
Affiliation(s)
- Ankit Pandeya
- Department of Pharmaceutical Sciences, Irma Lerma Rangel School of Pharmacy, Texas A&M University, College Station, TX, USA; Department of Chemistry, University of Kentucky, Lexington, KY, USA
| | - Yan Zhang
- Department of Pharmaceutical Sciences, Irma Lerma Rangel School of Pharmacy, Texas A&M University, College Station, TX, USA
| | - Jian Cui
- Saha Cardiovascular Research Center, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Ling Yang
- Department of Pharmaceutical Sciences, Irma Lerma Rangel School of Pharmacy, Texas A&M University, College Station, TX, USA
| | - Jeffery Li
- Department of Pharmaceutical Sciences, Irma Lerma Rangel School of Pharmacy, Texas A&M University, College Station, TX, USA
| | - Guoying Zhang
- Department of Pharmaceutical Sciences, Irma Lerma Rangel School of Pharmacy, Texas A&M University, College Station, TX, USA
| | - Congqing Wu
- Saha Cardiovascular Research Center, College of Medicine, University of Kentucky, Lexington, KY, USA; Department of Surgery, College of Medicine, University of Kentucky, Lexington, KY, USA; Department of Microbiology, Immunology, and Molecular Genetics, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Zhenyu Li
- Department of Pharmaceutical Sciences, Irma Lerma Rangel School of Pharmacy, Texas A&M University, College Station, TX, USA.
| | - Yinan Wei
- Department of Pharmaceutical Sciences, Irma Lerma Rangel School of Pharmacy, Texas A&M University, College Station, TX, USA.
| |
Collapse
|
18
|
Abstract
Type III secretion systems (T3SSs) are utilized by Gram-negative pathogens to enhance their pathogenesis. This secretion system is associated with the delivery of effectors through a needle-like structure from the bacterial cytosol directly into a target eukaryotic cell. These effector proteins then manipulate specific eukaryotic cell functions to benefit pathogen survival within the host. The obligate intracellular pathogens of the family Chlamydiaceae have a highly evolutionarily conserved nonflagellar T3SS that is an absolute requirement for their survival and propagation within the host with about one-seventh of the genome dedicated to genes associated with the T3SS apparatus, chaperones, and effectors. Chlamydiae also have a unique biphasic developmental cycle where the organism alternates between an infectious elementary body (EB) and replicative reticulate body (RB). T3SS structures have been visualized on both EBs and RBs. And there are effector proteins that function at each stage of the chlamydial developmental cycle, including entry and egress. This review will discuss the history of the discovery of chlamydial T3SS and the biochemical characterization of components of the T3SS apparatus and associated chaperones in the absence of chlamydial genetic tools. These data will be contextualized into how the T3SS apparatus functions throughout the chlamydial developmental cycle and the utility of heterologous/surrogate models to study chlamydial T3SS. Finally, there will be a targeted discussion on the history of chlamydial effectors and recent advances in the field.
Collapse
Affiliation(s)
- Elizabeth A. Rucks
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Durham Research Center II, Omaha, Nebraska, USA
| |
Collapse
|
19
|
Shetty D, Kenney LJ. A pH-sensitive switch activates virulence in Salmonella. eLife 2023; 12:e85690. [PMID: 37706506 PMCID: PMC10519707 DOI: 10.7554/elife.85690] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 09/13/2023] [Indexed: 09/15/2023] Open
Abstract
The transcriptional regulator SsrB acts as a switch between virulent and biofilm lifestyles of non-typhoidal Salmonella enterica serovar Typhimurium. During infection, phosphorylated SsrB activates genes on Salmonella Pathogenicity Island-2 (SPI-2) essential for survival and replication within the macrophage. Low pH inside the vacuole is a key inducer of expression and SsrB activation. Previous studies demonstrated an increase in SsrB protein levels and DNA-binding affinity at low pH; the molecular basis was unknown (Liew et al., 2019). This study elucidates its underlying mechanism and in vivo significance. Employing single-molecule and transcriptional assays, we report that the SsrB DNA-binding domain alone (SsrBc) is insufficient to induce acid pH-sensitivity. Instead, His12, a conserved residue in the receiver domain confers pH sensitivity to SsrB allosterically. Acid-dependent DNA binding was highly cooperative, suggesting a new configuration of SsrB oligomers at SPI-2-dependent promoters. His12 also plays a role in SsrB phosphorylation; substituting His12 reduced phosphorylation at neutral pH and abolished pH-dependent differences. Failure to flip the switch in SsrB renders Salmonella avirulent and represents a potential means of controlling virulence.
Collapse
Affiliation(s)
- Dasvit Shetty
- Mechanobiology Institute, National University of SingaporeSingaporeSingapore
| | - Linda J Kenney
- Mechanobiology Institute, National University of SingaporeSingaporeSingapore
- Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch at GalvestonGalvestonUnited States
- Sealy Center for Structural Biology and Molecular Biophysics, The University of Texas Medical Branch at GalvestonGalvestonUnited States
| |
Collapse
|
20
|
McGuckin MA, Davies JM, Felgner P, Wong KY, Giri R, He Y, Moniruzzaman M, Kryza T, Sajiir H, Hooper JD, Florin TH, Begun J, Oussalah A, Hasnain SZ, Hensel M, Sheng YH. MUC13 Cell Surface Mucin Limits Salmonella Typhimurium Infection by Protecting the Mucosal Epithelial Barrier. Cell Mol Gastroenterol Hepatol 2023; 16:985-1009. [PMID: 37660948 PMCID: PMC10630632 DOI: 10.1016/j.jcmgh.2023.08.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 08/25/2023] [Accepted: 08/28/2023] [Indexed: 09/05/2023]
Abstract
BACKGROUND & AIMS MUC13 cell surface mucin is highly expressed on the mucosal surface throughout the intestine, yet its role against bacterial infection is unknown. We investigated how MUC13 impacts Salmonella typhimurium (S Tm) infection and elucidated its mechanisms of action. METHODS Muc13-/- and wild-type littermate mice were gavaged with 2 isogenic strains of S Tm after pre-conditioning with streptomycin. We assessed clinical parameters, cecal histology, local and systemic bacterial load, and proinflammatory cytokines after infection. Cecal enteroids and epithelial cell lines were used to evaluate the mechanism of MUC13 activity after infection. The interaction between bacterial SiiE and MUC13 was assessed by using siiE-deficient Salmonella. RESULTS S Tm-infected Muc13-/- mice had increased disease activity, histologic damage, and higher local and systemic bacterial loads. Mechanistically, we found that S Tm binds to MUC13 through its giant SiiE adhesin and that MUC13 acts as a pathogen-binding decoy shed from the epithelial cell surface after pathogen engagement, limiting bacterial invasion. In addition, MUC13 reduces epithelial cell death and intestinal barrier breakdown by enhancing nuclear factor kappa B signaling during infection, independent of its decoy function. CONCLUSIONS We show for the first time that MUC13 plays a critical role in antimicrobial defense against pathogenic S Tm at the intestinal mucosal surface by both acting as a releasable decoy limiting bacterial invasion and reducing pathogen-induced cell death. This further implicates the cell surface mucin family in mucosal defense from bacterial infection.
Collapse
Affiliation(s)
- Michael A McGuckin
- Inflammatory Disease Biology and Therapeutics Group, Mater Research Institute - The University of Queensland, Translational Research Institute, Woolloongabba, Queensland, Australia; Faculty of Medicine Dentistry and Health Sciences, University of Melbourne, Parkville, Victoria, Australia.
| | - Julie M Davies
- Inflammatory Bowel Diseases Group, Mater Research Institute - The University of Queensland, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Pascal Felgner
- CellNanOs, Center for Cellular Nanoanalytics, Osnabrueck, Germany; Division Microbiology, Universitaet Osnabrueck, Osnabrueck, Germany
| | - Kuan Yau Wong
- Inflammatory Disease Biology and Therapeutics Group, Mater Research Institute - The University of Queensland, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Rabina Giri
- Inflammatory Bowel Diseases Group, Mater Research Institute - The University of Queensland, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Yaowu He
- Cancer Biology Group, Mater Research Institute-University of Queensland, Woolloongabba, Queensland, Australia
| | - Md Moniruzzaman
- Inflammatory Bowel Diseases Group, Mater Research Institute - The University of Queensland, Translational Research Institute, Woolloongabba, Queensland, Australia; School of Pharmacy, The University of Queensland, Woolloongabba, Queensland, Australia
| | - Thomas Kryza
- Cancer Biology Group, Mater Research Institute-University of Queensland, Woolloongabba, Queensland, Australia
| | - Haressh Sajiir
- Inflammatory Disease Biology and Therapeutics Group, Mater Research Institute - The University of Queensland, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - John D Hooper
- Cancer Biology Group, Mater Research Institute-University of Queensland, Woolloongabba, Queensland, Australia
| | - Timothy H Florin
- Inflammatory Bowel Diseases Group, Mater Research Institute - The University of Queensland, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Jakob Begun
- Inflammatory Bowel Diseases Group, Mater Research Institute - The University of Queensland, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Abderrahim Oussalah
- Department of Molecular Medicine, Division of Biochemistry, Molecular Biology, Nutrition, and Metabolism, University Hospital of Nancy, Nancy, France; University of Lorraine, INSERM UMR_S 1256, Nutrition, Genetics, and Environmental Risk Exposure (NGERE), Faculty of Medicine of Nancy, Nancy, France; Reference Center for Inborn Errors of Metabolism (ORPHA67872), University Hospital of Nancy, Nancy, France
| | - Sumaira Z Hasnain
- Inflammatory Disease Biology and Therapeutics Group, Mater Research Institute - The University of Queensland, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Michael Hensel
- CellNanOs, Center for Cellular Nanoanalytics, Osnabrueck, Germany; Division Microbiology, Universitaet Osnabrueck, Osnabrueck, Germany
| | - Yong H Sheng
- Inflammatory Disease Biology and Therapeutics Group, Mater Research Institute - The University of Queensland, Translational Research Institute, Woolloongabba, Queensland, Australia; Laboratory of B-Lymphocytes in Autoimmunity and Malignancies, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia.
| |
Collapse
|
21
|
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
|
22
|
Egan MS, Zhang J, Shin S. Human and mouse NAIP/NLRC4 inflammasome responses to bacterial infection. Curr Opin Microbiol 2023; 73:102298. [PMID: 37058933 PMCID: PMC10225321 DOI: 10.1016/j.mib.2023.102298] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 02/15/2023] [Accepted: 02/22/2023] [Indexed: 04/16/2023]
Abstract
Intracellular immune complexes known as inflammasomes sense breaches of cytosolic sanctity. Inflammasomes promote downstream proinflammatory events, including interleukin-1 (IL-1) family cytokine release and pyroptotic cell death. The nucleotide-binding leucine-rich repeat family, apoptosis inhibitory protein/nucleotide-binding leucine-rich repeat family, caspase recruitment domain (CARD) domain-containing protein 4 (NAIP/NLRC4) inflammasome is involved in a range of pathogenic and protective inflammatory processes in mammalian hosts. In particular, the NAIP/NLRC4 inflammasome responds to flagellin and components of the virulence-associated type III secretion (T3SS) apparatus in the host cytosol, thereby allowing it to be a critical mediator of host defense during bacterial infection. Notable species- and cell type-specific differences exist in NAIP/NLRC4 inflammasome responses to bacterial pathogens. With a focus on Salmonella enterica serovar Typhimurium as a model pathogen, we review differences between murine and human NAIP/NLRC4 inflammasome responses. Differences in NAIP/NLRC4 inflammasome responses across species and cell types may have arisen in part due to evolutionary pressures.
Collapse
Affiliation(s)
- Marisa S Egan
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Jenna Zhang
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Sunny Shin
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA.
| |
Collapse
|
23
|
Gibbs KD, Wang L, Yang Z, Anderson CE, Bourgeois JS, Cao Y, Gaggioli MR, Biel M, Puertollano R, Chen CC, Ko DC. Human variation impacting MCOLN2 restricts Salmonella Typhi replication by magnesium deprivation. CELL GENOMICS 2023; 3:100290. [PMID: 37228749 PMCID: PMC10203047 DOI: 10.1016/j.xgen.2023.100290] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 01/24/2023] [Accepted: 02/27/2023] [Indexed: 05/27/2023]
Abstract
Human genetic diversity can reveal critical factors in host-pathogen interactions. This is especially useful for human-restricted pathogens like Salmonella enterica serovar Typhi (S. Typhi), the cause of typhoid fever. One key defense during bacterial infection is nutritional immunity: host cells attempt to restrict bacterial replication by denying bacteria access to key nutrients or supplying toxic metabolites. Here, a cellular genome-wide association study of intracellular replication by S. Typhi in nearly a thousand cell lines from around the world-and extensive follow-up using intracellular S. Typhi transcriptomics and manipulation of magnesium availability-demonstrates that the divalent cation channel mucolipin-2 (MCOLN2 or TRPML2) restricts S. Typhi intracellular replication through magnesium deprivation. Mg2+ currents, conducted through MCOLN2 and out of endolysosomes, were measured directly using patch-clamping of the endolysosomal membrane. Our results reveal Mg2+ limitation as a key component of nutritional immunity against S. Typhi and as a source of variable host resistance.
Collapse
Affiliation(s)
- Kyle D. Gibbs
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, Durham, NC 27710, USA
| | - Liuyang Wang
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, Durham, NC 27710, USA
| | - Zhuo Yang
- Department of Pharmacy, Center for Drug Research, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Caroline E. Anderson
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, Durham, NC 27710, USA
| | - Jeffrey S. Bourgeois
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, Durham, NC 27710, USA
- University Program in Genetics and Genomics, Duke University, Durham, NC 27710, USA
| | - Yanlu Cao
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, Durham, NC 27710, USA
| | - Margaret R. Gaggioli
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, Durham, NC 27710, USA
| | - Martin Biel
- Department of Pharmacy, Center for Drug Research, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Rosa Puertollano
- Cell and Developmental Biology Center, National Heart, Lung, & Blood Institute, NIH, Bethesda, MD 20892, USA
| | - Cheng-Chang Chen
- Department of Clinical Laboratory Sciences and Medical Biotechnology, College of Medicine, National Taiwan University, Taipei 100, Taiwan
- Department of Laboratory Medicine, National Taiwan University Hospital, Taipei 100, Taiwan
| | - Dennis C. Ko
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, Durham, NC 27710, USA
- University Program in Genetics and Genomics, Duke University, Durham, NC 27710, USA
- Division of Infectious Diseases, Department of Medicine, School of Medicine, Duke University, Durham, NC 27710, USA
| |
Collapse
|
24
|
Salmonella effector SopB reorganizes cytoskeletal vimentin to maintain replication vacuoles for efficient infection. Nat Commun 2023; 14:478. [PMID: 36717589 PMCID: PMC9885066 DOI: 10.1038/s41467-023-36123-w] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 01/16/2023] [Indexed: 01/31/2023] Open
Abstract
A variety of intracellular bacteria modulate the host cytoskeleton to establish subcellular niches for replication. However, the role of intermediate filaments, which are crucial for mechanical strength and resilience of the cell, and in bacterial vacuole preservation remains unclear. Here, we show that Salmonella effector SopB reorganizes the vimentin network to form cage-like structures that surround Salmonella-containing vacuoles (SCVs). Genetic removal of vimentin markedly disrupts SCV organization, significantly reduces bacterial replication and cell death. Mechanistically, SopB uses its N-terminal Cdc42-binding domain to interact with and activate Cdc42 GTPase, which in turn recruits vimentin around SCVs. A high-content imaging-based screening identified that MEK1/2 inhibition led to vimentin dispersion. Our work therefore elucidates the signaling axis SopB-Cdc42-MEK1/2 as mobilizing host vimentin to maintain concrete SCVs and identifies a mechanism contributing to Salmonella replication. Importantly, Trametinib, a clinically-approved MEK1/2 inhibitor identified in the screen, displayed significant anti-infection efficacy against Salmonella both in vitro and in vivo, and may provide a therapeutic option for treating drug-tolerant salmonellosis.
Collapse
|
25
|
Klein JA, Powers TR, Knodler LA. Measurement of Salmonella enterica Internalization and Vacuole Lysis in Epithelial Cells. Methods Mol Biol 2023; 2692:209-220. [PMID: 37365470 DOI: 10.1007/978-1-0716-3338-0_14] [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] [Indexed: 06/28/2023]
Abstract
Establishment of an intracellular niche within mammalian cells is key to the pathogenesis of the gastrointestinal bacterium, Salmonella enterica serovar Typhimurium (S. Typhimurium). Here we will describe how to study the internalization of S. Typhimurium into human epithelial cells using the gentamicin protection assay. The assay takes advantage of the relatively poor penetration of gentamicin into mammalian cells; internalized bacteria are effectively protected from its antibacterial actions. A second assay, the chloroquine (CHQ) resistance assay, can be used to determine the proportion of internalized bacteria that have lysed or damaged their Salmonella-containing vacuole and are therefore residing within the cytosol. Its application to the quantification of cytosolic S. Typhimurium in epithelial cells will also be presented. Together, these protocols provide an inexpensive, rapid, and sensitive quantitative measure of bacterial internalization and vacuole lysis by S. Typhimurium.
Collapse
Affiliation(s)
- Jessica A Klein
- Paul G. Allen School for Global Health, College of Veterinary Medicine, Washington State University, Pullman, WA, USA
| | - TuShun R Powers
- Paul G. Allen School for Global Health, College of Veterinary Medicine, Washington State University, Pullman, WA, USA
| | - Leigh A Knodler
- Paul G. Allen School for Global Health, College of Veterinary Medicine, Washington State University, Pullman, WA, USA.
- Department of Microbiology and Molecular Genetics, University of Vermont, Burlington, USA.
| |
Collapse
|
26
|
Newson JP, Gaissmaier MS, McHugh SC, Hardt WD. Studying antibiotic persistence in vivo using the model organism Salmonella Typhimurium. Curr Opin Microbiol 2022; 70:102224. [PMID: 36335713 DOI: 10.1016/j.mib.2022.102224] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 09/22/2022] [Accepted: 10/02/2022] [Indexed: 11/06/2022]
Abstract
Antibiotic persistence permits a subpopulation of susceptible bacteria to survive lethal concentrations of bactericidal antibiotics. This prolongs antibiotic therapy, promotes the evolution of antibiotic-resistant pathogen strains and can select for pathogen virulence within infected hosts. Here, we review the literature exploring antibiotic persistence in vivo, and describe the consequences of recalcitrant subpopulations, with a focus on studies using the model pathogen Salmonella Typhimurium. In vitro studies have established a concise set of features distinguishing true persisters from other forms of bacterial recalcitrance to bactericidal antibiotics. We discuss how animal infection models are useful for exploring these features in vivo, and describe how technical challenges can sometimes prevent the conclusive identification of true antibiotic persistence within infected hosts. We propose using two complementary working definitions for studying antibiotic persistence in vivo: the strict definition for studying the mechanisms of persister formation, and an operative definition for functional studies assessing the links between invasive virulence and persistence as well as the consequences for horizontal gene transfer, or the emergence of antibiotic-resistant mutants. This operative definition will enable further study of how antibiotic persisters arise in vivo, and of how surviving populations contribute to diverse downstream effects such as pathogen transmission, horizontal gene transfer and the evolution of virulence and antibiotic resistance. Ultimately, such studies will help to improve therapeutic control of antibiotic- recalcitrant populations.
Collapse
Affiliation(s)
- Joshua Pm Newson
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Marla S Gaissmaier
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Sarah C McHugh
- Department of Fundamental Microbiology, University of Lausanne, Lausanne, Switzerland
| | - Wolf-Dietrich Hardt
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland.
| |
Collapse
|
27
|
Naseer N, Zhang J, Bauer R, Constant DA, Nice TJ, Brodsky IE, Rauch I, Shin S. Salmonella enterica Serovar Typhimurium Induces NAIP/NLRC4- and NLRP3/ASC-Independent, Caspase-4-Dependent Inflammasome Activation in Human Intestinal Epithelial Cells. Infect Immun 2022; 90:e0066321. [PMID: 35678562 PMCID: PMC9302179 DOI: 10.1128/iai.00663-21] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 05/23/2022] [Indexed: 01/09/2023] Open
Abstract
Salmonella enterica serovar Typhimurium is a Gram-negative pathogen that causes diseases ranging from gastroenteritis to systemic infection and sepsis. Salmonella uses type III secretion systems (T3SS) to inject effectors into host cells. While these effectors are necessary for bacterial invasion and intracellular survival, intracellular delivery of T3SS products also enables detection of translocated Salmonella ligands by cytosolic immune sensors. Some of these sensors form multimeric complexes called inflammasomes, which activate caspases that lead to interleukin-1 (IL-1) family cytokine release and pyroptosis. In particular, the Salmonella T3SS needle, inner rod, and flagellin proteins activate the NAIP/NLRC4 inflammasome in murine intestinal epithelial cells (IECs), which leads to restriction of bacterial replication and extrusion of infected IECs into the intestinal lumen, thereby preventing systemic dissemination of Salmonella. While these processes are quite well studied in mice, the role of the NAIP/NLRC4 inflammasome in human IECs remains unknown. Unexpectedly, we found the NAIP/NLRC4 inflammasome is dispensable for early inflammasome responses to Salmonella in both human IEC lines and enteroids. Additionally, NLRP3 and the adaptor protein ASC are not required for inflammasome activation in Caco-2 cells. Instead, we observed a necessity for caspase-4 and gasdermin D pore-forming activity in mediating inflammasome responses to Salmonella in Caco-2 cells. These findings suggest that unlike murine IECs, human IECs do not rely on NAIP/NLRC4 or NLRP3/ASC inflammasomes and instead primarily use caspase-4 to mediate inflammasome responses to Salmonella pathogenicity island 1 (SPI-1)-expressing Salmonella.
Collapse
Affiliation(s)
- Nawar Naseer
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Jenna Zhang
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Renate Bauer
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, Oregon, USA
- Department of Biosciences, Paris Lodron University of Salzburg, Salzburg, Austria
| | - David A. Constant
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, Oregon, USA
| | - Timothy J. Nice
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, Oregon, USA
| | - Igor E. Brodsky
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, Pennsylvania, USA
| | - Isabella Rauch
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, Oregon, USA
| | - Sunny Shin
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| |
Collapse
|
28
|
Integration of the Salmonella Typhimurium Methylome and Transcriptome Reveals That DNA Methylation and Transcriptional Regulation Are Largely Decoupled under Virulence-Related Conditions. mBio 2022; 13:e0346421. [PMID: 35658533 PMCID: PMC9239280 DOI: 10.1128/mbio.03464-21] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Despite being in a golden age of bacterial epigenomics, little work has systematically examined the plasticity and functional impacts of the bacterial DNA methylome. Here, we leveraged single-molecule, real-time sequencing (SMRT-seq) to examine the m6A DNA methylome of two Salmonella enterica serovar Typhimurium strains: 14028s and a ΔmetJ mutant with derepressed methionine metabolism, grown in Luria broth or medium that simulates the intracellular environment. We found that the methylome is remarkably static: >95% of adenosine bases retain their methylation status across conditions. Integration of methylation with transcriptomic data revealed limited correlation between changes in methylation and gene expression. Further, examination of the transcriptome in ΔyhdJ bacteria lacking the m6A methylase with the most dynamic methylation pattern in our data set revealed little evidence of YhdJ-mediated gene regulation. Curiously, despite G(m6A)TC motifs being particularly resistant to change across conditions, incorporating dam mutants into our analyses revealed two examples where changes in methylation and transcription may be linked across conditions. This includes the novel finding that the ΔmetJ motility defect may be partially driven by hypermethylation of the chemotaxis gene tsr. Together, these data redefine the S. Typhimurium epigenome as a highly stable system that has rare but important roles in transcriptional regulation. Incorporating these lessons into future studies will be critical as we progress through the epigenomic era.
Collapse
|
29
|
El Qaidi S, Scott NE, Hays MP, Hardwidge PR. Arginine glycosylation regulates UDP-GlcNAc biosynthesis in Salmonella enterica. Sci Rep 2022; 12:5293. [PMID: 35351940 PMCID: PMC8964723 DOI: 10.1038/s41598-022-09276-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 03/21/2022] [Indexed: 12/27/2022] Open
Abstract
The Salmonella enterica SseK1 protein is a type three secretion system effector that glycosylates host proteins during infection on specific arginine residues with N-acetyl glucosamine (GlcNAc). SseK1 also Arg-glycosylates endogenous bacterial proteins and we thus hypothesized that SseK1 activities might be integrated with regulating the intrabacterial abundance of UPD-GlcNAc, the sugar-nucleotide donor used by this effector. After searching for new SseK1 substrates, we found that SseK1 glycosylates arginine residues in the dual repressor-activator protein NagC, leading to increased DNA-binding affinity and enhanced expression of the NagC-regulated genes glmU and glmS. SseK1 also glycosylates arginine residues in GlmR, a protein that enhances GlmS activity. This Arg-glycosylation improves the ability of GlmR to enhance GlmS activity. We also discovered that NagC is a direct activator of glmR expression. Salmonella lacking SseK1 produce significantly reduced amounts of UDP-GlcNAc as compared with Salmonella expressing SseK1. Overall, we conclude that SseK1 up-regulates UDP-GlcNAc synthesis both by enhancing the DNA-binding activity of NagC and by increasing GlmS activity through GlmR glycosylation. Such regulatory activities may have evolved to maintain sufficient levels of UDP-GlcNAc for both bacterial cell wall precursors and for SseK1 to modify other bacterial and host targets in response to environmental changes and during infection.
Collapse
Affiliation(s)
- Samir El Qaidi
- College of Veterinary Medicine, Kansas State University, Manhattan, KS, 66506, USA
| | - Nichollas E Scott
- Department of Microbiology and Immunology, University of Melbourne Within the Peter Doherty Institute for Infection and Immunity, Melbourne, 3000, Australia
| | - Michael P Hays
- College of Veterinary Medicine, Kansas State University, Manhattan, KS, 66506, USA
| | - Philip R Hardwidge
- College of Veterinary Medicine, Kansas State University, Manhattan, KS, 66506, USA.
| |
Collapse
|
30
|
Cohen H, Adani B, Cohen E, Piscon B, Azriel S, Desai P, Bähre H, McClelland M, Rahav G, Gal-Mor O. The ancestral stringent response potentiator, DksA has been adapted throughout Salmonella evolution to orchestrate the expression of metabolic, motility, and virulence pathways. Gut Microbes 2022; 14:1997294. [PMID: 34923900 PMCID: PMC8726615 DOI: 10.1080/19490976.2021.1997294] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
DksA is a conserved RNA polymerase-binding protein known to play a key role in the stringent response of proteobacteria species, including many gastrointestinal pathogens. Here, we used RNA-sequencing of Escherichia coli, Salmonella bongori and Salmonella enterica serovar Typhimurium, together with phenotypic comparison to study changes in the DksA regulon, during Salmonella evolution. Comparative RNA-sequencing showed that under non-starved conditions, DksA controls the expression of 25%, 15%, and 20% of the E. coli, S. bongori, and S. enterica genes, respectively, indicating that DksA is a pleiotropic regulator, expanding its role beyond the canonical stringent response. We demonstrate that DksA is required for the growth of these three enteric bacteria species in minimal medium and controls the expression of the TCA cycle, glycolysis, pyrimidine biosynthesis, and quorum sensing. Interestingly, at multiple steps during Salmonella evolution, the type I fimbriae and various virulence genes encoded within SPIs 1, 2, 4, 5, and 11 have been transcriptionally integrated under the ancestral DksA regulon. Consequently, we show that DksA is necessary for host cells invasion by S. Typhimurium and S. bongori and for intracellular survival of S. Typhimurium in bone marrow-derived macrophages (BMDM). Moreover, we demonstrate regulatory inversion of the conserved motility-chemotaxis regulon by DksA, which acts as a negative regulator in E. coli, but activates this pathway in S. bongori and S. enterica. Overall, this study demonstrates the regulatory assimilation of multiple horizontally acquired virulence genes under the DksA regulon and provides new insights into the evolution of virulence genes regulation in Salmonella spp.
Collapse
Affiliation(s)
- Helit Cohen
- Sheba Medical Center, The Infectious Diseases Research Laboratory, Tel-Hashomer, Israel
| | - Boaz Adani
- Sheba Medical Center, The Infectious Diseases Research Laboratory, Tel-Hashomer, Israel
| | - Emiliano Cohen
- Sheba Medical Center, The Infectious Diseases Research Laboratory, Tel-Hashomer, Israel
| | - Bar Piscon
- Sheba Medical Center, The Infectious Diseases Research Laboratory, Tel-Hashomer, Israel,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel,Department of Clinical Microbiology and Immunology, Tel Aviv University, Tel Aviv, Israel
| | - Shalhevet Azriel
- Sheba Medical Center, The Infectious Diseases Research Laboratory, Tel-Hashomer, Israel
| | - Prerak Desai
- Janssen Research & Development, LLC, Raritan, New Jersey, USA,Department of Microbiology and Molecular Genetics, University of California, Irvine, California, USA
| | - Heike Bähre
- Hannover Medical School, Research Core Unit Metabolomics, Hannover, Germany
| | - Michael McClelland
- Department of Microbiology and Molecular Genetics, University of California, Irvine, California, USA
| | - Galia Rahav
- Sheba Medical Center, The Infectious Diseases Research Laboratory, Tel-Hashomer, Israel,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ohad Gal-Mor
- Sheba Medical Center, The Infectious Diseases Research Laboratory, Tel-Hashomer, Israel,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel,Department of Clinical Microbiology and Immunology, Tel Aviv University, Tel Aviv, Israel,Contact Ohad Gal-Mor The Infectious Diseases Research Laboratory Sheba Medical Cente, Tel-Hashomer, Israel
| |
Collapse
|
31
|
Pozdeev G, Beckett MC, Mogre A, Thomson NR, Dorman CJ. Reciprocally rewiring and repositioning the Integration Host Factor (IHF) subunit genes in Salmonella enterica serovar Typhimurium: impacts on physiology and virulence. Microb Genom 2022; 8. [PMID: 35166652 PMCID: PMC8942017 DOI: 10.1099/mgen.0.000768] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
The Integration Host Factor (IHF) is a heterodimeric nucleoid-associated protein that plays roles in bacterial nucleoid architecture and genome-wide gene regulation. The ihfA and ihfB genes encode the subunits and are located 350 kbp apart, in the Right replichore of the Salmonella chromosome. IHF is composed of one IhfA and one IhfB subunit. Despite this 1 : 1 stoichiometry, MS revealed that IhfB is produced in 2-fold excess over IhfA. We re-engineered Salmonella to exchange reciprocally the protein-coding regions of ihfA and ihfB, such that each relocated protein-encoding region was driven by the expression signals of the other's gene. MS showed that in this 'rewired' strain, IhfA is produced in excess over IhfB, correlating with enhanced stability of the hybrid ihfB-ihfA mRNA that was expressed from the ihfB promoter. Nevertheless, the rewired strain grew at a similar rate to the wild-type and was similar in competitive fitness. However, compared to the wild-type, it was less motile, had growth-phase-specific reductions in SPI-1 and SPI-2 gene expression, and was engulfed at a higher rate by RAW macrophage. Our data show that while exchanging the physical locations of its ihf genes and the rewiring of their regulatory circuitry are well tolerated in Salmonella, genes involved in the production of type 3 secretion systems exhibit dysregulation accompanied by altered phenotypes.
Collapse
Affiliation(s)
- German Pozdeev
- Department of Microbiology, Moyne Institute of Preventive Medicine, Trinity College Dublin, Dublin 2, Ireland
| | - Michael C Beckett
- Department of Microbiology, Moyne Institute of Preventive Medicine, Trinity College Dublin, Dublin 2, Ireland
| | - Aalap Mogre
- Department of Microbiology, Moyne Institute of Preventive Medicine, Trinity College Dublin, Dublin 2, Ireland
| | | | - Charles J Dorman
- Department of Microbiology, Moyne Institute of Preventive Medicine, Trinity College Dublin, Dublin 2, Ireland
| |
Collapse
|
32
|
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
|
33
|
Rana S, Maurya S, Mohapatra G, Singh S, Babar R, Chandrasekhar H, Chamoli G, Rathore D, Kshetrapal P, Srikanth CV. Activation of epigenetic regulator KDM6B by Salmonella Typhimurium enables chronic infections. Gut Microbes 2022; 13:1986665. [PMID: 34696686 PMCID: PMC8555538 DOI: 10.1080/19490976.2021.1986665] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Non-typhoidal Salmonella (NTS) infections result in self limiting gastroenteritis except in rare cases wherein manifestations of chronic infections can occur. Strategies employed by Salmonella to thrive in hostile environments of host during chronic infections are complex and multifaceted. In chronic state, a coordinated action of bacterial effectors allows reprogramming of macrophages to M2 subtype and thereby creating a permissible replicative niche. The mechanistic details of these processes are not fully known. In the current study we identified, histone H3-lysine 27 trimethylation (H3K27me3)-specific demethylase, KDM6B to be upregulated in both cell culture and in murine model of Salmonella infection. KDM6B recruitment upon infection exhibited an associated loss of overall H3K27me3 in host cells and was Salmonella SPI1 effectors coordinated. ChIP-qRT-PCR array analysis revealed several new gene promoter targets of KDM6B demethylase activity including PPARδ, a crucial regulator of fatty acid oxidation pathway and Salmonella-persistent infections. Furthermore, pharmacological inhibition of KDM6B demethylase activity with GSKJ4 in chronic Salmonella infection mice model led to a significant reduction in pathogen load and M2 macrophage polarization in peripheral lymphoid organs. The following work thus reveals Salmonella effector-mediated epigenetic reprogramming of macrophages responsible for its long-term survival and chronic carriage.
Collapse
Affiliation(s)
- Sarika Rana
- Laboratory of Gut Infection and Inflammation Biology, Regional Centre for Biotechnology, Faridabad, India,Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Sonalika Maurya
- Laboratory of Gut Infection and Inflammation Biology, Regional Centre for Biotechnology, Faridabad, India
| | - Gayatree Mohapatra
- Laboratory of Gut Infection and Inflammation Biology, Regional Centre for Biotechnology, Faridabad, India
| | - Savita Singh
- Maternal and Child Health, Translational Health Science and Technology Institute, Faridabad, India
| | - Rohan Babar
- Laboratory of Gut Infection and Inflammation Biology, Regional Centre for Biotechnology, Faridabad, India
| | - Hridya Chandrasekhar
- Laboratory of Gut Infection and Inflammation Biology, Regional Centre for Biotechnology, Faridabad, India
| | - Garima Chamoli
- Laboratory of Gut Infection and Inflammation Biology, Regional Centre for Biotechnology, Faridabad, India
| | - Deepak Rathore
- Maternal and Child Health, Translational Health Science and Technology Institute, Faridabad, India
| | - Pallavi Kshetrapal
- Maternal and Child Health, Translational Health Science and Technology Institute, Faridabad, India
| | - C. V. Srikanth
- Laboratory of Gut Infection and Inflammation Biology, Regional Centre for Biotechnology, Faridabad, India,CONTACT C. V. Srikanth Regional Centre for Biotechnology, 3rd Milestone Gurgaon Faridabad Expressway, Faridabad, India
| |
Collapse
|
34
|
Naseer N, Egan MS, Reyes Ruiz VM, Scott WP, Hunter EN, Demissie T, Rauch I, Brodsky IE, Shin S. Human NAIP/NLRC4 and NLRP3 inflammasomes detect Salmonella type III secretion system activities to restrict intracellular bacterial replication. PLoS Pathog 2022; 18:e1009718. [PMID: 35073381 PMCID: PMC8812861 DOI: 10.1371/journal.ppat.1009718] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 02/03/2022] [Accepted: 12/27/2021] [Indexed: 01/16/2023] Open
Abstract
Salmonella enterica serovar Typhimurium is a Gram-negative pathogen that uses two distinct type III secretion systems (T3SSs), termed Salmonella pathogenicity island (SPI)-1 and SPI-2, to deliver virulence factors into the host cell. The SPI-1 T3SS enables Salmonella to invade host cells, while the SPI-2 T3SS facilitates Salmonella's intracellular survival. In mice, a family of cytosolic immune sensors, including NAIP1, NAIP2, and NAIP5/6, recognizes the SPI-1 T3SS needle, inner rod, and flagellin proteins, respectively. Ligand recognition triggers assembly of the NAIP/NLRC4 inflammasome, which mediates caspase-1 activation, IL-1 family cytokine secretion, and pyroptosis of infected cells. In contrast to mice, humans encode a single NAIP that broadly recognizes all three ligands. The role of NAIP/NLRC4 or other inflammasomes during Salmonella infection of human macrophages is unclear. We find that although the NAIP/NLRC4 inflammasome is essential for detecting T3SS ligands in human macrophages, it is partially required for responses to infection, as Salmonella also activated the NLRP3 and CASP4/5 inflammasomes. Importantly, we demonstrate that combinatorial NAIP/NLRC4 and NLRP3 inflammasome activation restricts Salmonella replication in human macrophages. In contrast to SPI-1, the SPI-2 T3SS inner rod is not sensed by human or murine NAIPs, which is thought to allow Salmonella to evade host recognition and replicate intracellularly. Intriguingly, we find that human NAIP detects the SPI-2 T3SS needle protein. Critically, in the absence of both flagellin and the SPI-1 T3SS, the NAIP/NLRC4 inflammasome still controlled intracellular Salmonella burden. These findings reveal that recognition of Salmonella SPI-1 and SPI-2 T3SSs and engagement of both the NAIP/NLRC4 and NLRP3 inflammasomes control Salmonella infection in human macrophages.
Collapse
Affiliation(s)
- Nawar Naseer
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Marisa S. Egan
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Valeria M. Reyes Ruiz
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - William P. Scott
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, Oregon
| | - Emma N. Hunter
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Tabitha Demissie
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Isabella Rauch
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, Oregon
| | - Igor E. Brodsky
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, Pennsylvania
| | - Sunny Shin
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
- * E-mail:
| |
Collapse
|
35
|
Zakaria Z, Hassan L, Sharif Z, Ahmad N, Mohd Ali R, Amir Husin S, Mohamed Sohaimi N, Abu Bakar S, Garba B. Virulence Gene Profile, Antimicrobial Resistance and Multilocus Sequence Typing of Salmonella enterica Subsp. enterica Serovar Enteritidis from Chickens and Chicken Products. Animals (Basel) 2022; 12:ani12010097. [PMID: 35011203 PMCID: PMC8749576 DOI: 10.3390/ani12010097] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Revised: 11/01/2021] [Accepted: 11/04/2021] [Indexed: 01/19/2023] Open
Abstract
This study was undertaken to determine the virulence, antimicrobial resistance and molecular subtypes of Salmonella in the Central Region of Peninsular Malaysia. A total of 45 Salmonella Enteritidis were detected from live chicken (cloacal swab), and chicken products (fresh and ready-to-eat meat) samples upon cultural isolation and serotyping. Similarly, an antimicrobial susceptibility test based on the Kirby Bauer disk diffusion method as well as antimicrobial resistance AMR genes, virulence determinants and multilocus sequence typing (MLST) typing were conducted after the Whole Genome Sequencing and analysis of the isolates. The results indicate that sequence types ST1925 (63.7%), and ST11 (26.5%) were the predominant out of the seven sequence types identified (ST292, ST329, ST365, ST423 and ST2132). The phenotypic antimicrobial profile corresponds to the genotypic characterization in that the majority of the isolates that exhibited tetracycline, gentamycin and aminoglycoside resistance; they also possessed the tetC and blaTEM β-Lactam resistance genes. However, isolates from cloacal swabs showed the highest number of resistance genes compared to the chicken products (fresh and ready-to-eat meat) samples. Furthermore, most of the virulence genes were found to cluster in the Salmonella pathogenicity island (SPI). In this study, all the isolates were found to possess SPI-1, which codes for the type III secretion system, which functions as actin-binding proteins (SptP and SopE). The virulence plasmid (VP) genes (spvB, spvC) were present in all genotypes except ST365. The findings of this study, particularly with regard to the molecular subtypes and AMR profiles of the Salmonella Enteritidis serotype shows multidrug-resistance features as well as genetic characteristics indicative of high pathogenicity.
Collapse
Affiliation(s)
- Zunita Zakaria
- Institute of Bioscience, Universiti Putra Malaysia, Serdang 43400, Malaysia
- Department of Veterinary Pathology and Microbiology, Faculty of Veterinary Medicine, Universiti Putra Malaysia, Serdang 43400, Malaysia;
- Correspondence:
| | - Latiffah Hassan
- Department of Veterinary Laboratory Diagnostics, Faculty of Veterinary Medicine, Universiti Putra Malaysia, Serdang 43400, Malaysia; (L.H.); (N.M.S.)
| | - Zawiyah Sharif
- Food Safety and Quality Division, Ministry of Health, Putrajaya 62675, Malaysia; (Z.S.); (S.A.B.)
| | - Norazah Ahmad
- Veterinary Public Health Division, Department of Veterinary Services Malaysia, Putrajaya 62630, Malaysia; (N.A.); (R.M.A.)
| | - Rohaya Mohd Ali
- Veterinary Public Health Division, Department of Veterinary Services Malaysia, Putrajaya 62630, Malaysia; (N.A.); (R.M.A.)
| | - Suraya Amir Husin
- Medical Development Division, Ministry of Health, Putrajaya 62590, Malaysia;
| | - Norfitriah Mohamed Sohaimi
- Department of Veterinary Laboratory Diagnostics, Faculty of Veterinary Medicine, Universiti Putra Malaysia, Serdang 43400, Malaysia; (L.H.); (N.M.S.)
| | - Shafini Abu Bakar
- Food Safety and Quality Division, Ministry of Health, Putrajaya 62675, Malaysia; (Z.S.); (S.A.B.)
| | - Bashiru Garba
- Department of Veterinary Pathology and Microbiology, Faculty of Veterinary Medicine, Universiti Putra Malaysia, Serdang 43400, Malaysia;
- Faculty of Veterinary Medicine, Usmanu Danfodiyo University, Sultan Abubakar Road, City Campus Complex, Sokoto 840212, Nigeria
| |
Collapse
|
36
|
Rahmatelahi H, El-Matbouli M, Menanteau-Ledouble S. Delivering the pain: an overview of the type III secretion system with special consideration for aquatic pathogens. Vet Res 2021; 52:146. [PMID: 34924019 PMCID: PMC8684695 DOI: 10.1186/s13567-021-01015-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 11/08/2021] [Indexed: 11/10/2022] Open
Abstract
Gram-negative bacteria are known to subvert eukaryotic cell physiological mechanisms using a wide array of virulence factors, among which the type three-secretion system (T3SS) is often one of the most important. The T3SS constitutes a needle-like apparatus that the bacterium uses to inject a diverse set of effector proteins directly into the cytoplasm of the host cells where they can hamper the host cellular machinery for a variety of purposes. While the structure of the T3SS is somewhat conserved and well described, effector proteins are much more diverse and specific for each pathogen. The T3SS can remodel the cytoskeleton integrity to promote intracellular invasion, as well as silence specific eukaryotic cell signals, notably to hinder or elude the immune response and cause apoptosis. This is also the case in aquatic bacterial pathogens where the T3SS can often play a central role in the establishment of disease, although it remains understudied in several species of important fish pathogens, notably in Yersinia ruckeri. In the present review, we summarise what is known of the T3SS, with a special focus on aquatic pathogens and suggest some possible avenues for research including the potential to target the T3SS for the development of new anti-virulence drugs.
Collapse
Affiliation(s)
- Hadis Rahmatelahi
- Clinical Division of Fish Medicine, University of Veterinary Medicine, 1210, Vienna, Austria
| | - Mansour El-Matbouli
- Clinical Division of Fish Medicine, University of Veterinary Medicine, 1210, Vienna, Austria
| | - Simon Menanteau-Ledouble
- Clinical Division of Fish Medicine, University of Veterinary Medicine, 1210, Vienna, Austria.
- Department of Chemistry and Bioscience, Aalborg University, Fredrik Bajers Vej 7H, 9220, Aalborg Ø, Denmark.
| |
Collapse
|
37
|
Quantitative proteomic screen identifies annexin A2 as a host target for Salmonella pathogenicity island-2 effectors SopD2 and PipB2. Sci Rep 2021; 11:23630. [PMID: 34880286 PMCID: PMC8655068 DOI: 10.1038/s41598-021-02795-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 11/17/2021] [Indexed: 11/18/2022] Open
Abstract
Intracellular pathogens need to establish an intracellular replicative niche to promote survival and replication within the hostile environment inside the host cell. Salmonella enterica serovar Typhimurium (S. Typhimurium) initiates formation of the unique Salmonella-containing vacuole and an extensive network of Salmonella-induced tubules in order to survive and thrive within host cells. At least six effectors secreted by the type III secretion system encoded within Salmonella pathogenicity island-2 (SPI-2), namely SifA, SopD2, PipB2, SteA, SseJ, and SseF, purportedly manipulate host cell intracellular trafficking and establish the intracellular replicative niche for S. Typhimurium. The phenotypes of these effectors are both subtle and complex, complicating elucidation of the mechanism underpinning host cell manipulation by S. Typhimurium. In this work we used stable isotope labeling of amino acids in cell culture (SILAC) and a S. Typhimurium mutant that secretes increased amounts of effectors to identify cognate effector binding partners during infection. Using this method, we identified the host protein annexin A2 (AnxA2) as a binding partner for both SopD2 and PipB2 and were able to confirm its binding to SopD2 and PipB2 by reciprocal pull down, although there was a low level of non-specific binding of SopD2-2HA and PipB2-2HA to the Ni-Sepharose beads present. We further showed that knockdown of AnxA2 altered the intracellular positioning of the Salmonella containing vacuole (SCV). This suggests that AnxA2 plays a role in the subcellular positioning of the SCV which could potentially be mediated through protein–protein interactions with either SopD2 or PipB2. This demonstrates the value of studying effector interactions using proteomic techniques and natural effector delivery during infection rather than transfection.
Collapse
|
38
|
Uchiya KI, Isono S, Yoshimura M, Wajima T, Nikai T. Salmonella fimbrial protein StcD induces cyclooxygenase-2 expression via Toll-like receptor 4. JOURNAL OF MICROBIOLOGY, IMMUNOLOGY, AND INFECTION = WEI MIAN YU GAN RAN ZA ZHI 2021; 55:581-589. [PMID: 34801423 DOI: 10.1016/j.jmii.2021.11.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 11/02/2021] [Indexed: 12/23/2022]
Abstract
INTRODUCTION The genome of Salmonella enterica serovar Typhimurium contains 13 operons with homology to fimbrial genes. METHODS To investigate the involvement of these fimbrial gene clusters in the expression of cyclooxygenase-2 (COX-2), which is an inducible enzyme involved in the synthesis of prostanoids, in J774 macrophages infected with S. enterica serovar Typhimurium, we constructed strains carrying a mutation in genes encoding the putative subunit proteins in 12 fimbrial operons. RESULTS The level of COX-2 expression was lower in macrophages infected with fimA or stcA mutant Salmonella than in those infected with wild-type Salmonella. Therefore, we focused on putative subunit protein StcA and adhesive like protein StcD encoded in the stc operon. Treatment of macrophages with purified recombinant StcD protein, but not StcA, resulted in the activation of the mitogen-activated protein kinase and nuclear factor kappa B signaling pathways, leading to the expression of not only COX-2 but also of pro-inflammatory cytokines such as interleukin (IL)-1β, IL-6, and tumor necrosis factor alpha. The expression of StcD-induced COX-2 was inhibited by treatment with the Toll-like receptor 4 (TLR4) inhibitor TAK-242, but not by treatment with the lipopolysaccharide (LPS) antagonist polymyxin B. Furthermore, StcD treatment stimulated HEK293 cells expressing TLR4 in the presence of CD14 and MD-2. CONCLUSION StcD is a pathogen-associated molecular pattern of S. enterica serovar Typhimurium that is recognized by TLR4 and plays a significant role in the induction of COX-2 expression in macrophages.
Collapse
Affiliation(s)
- Kei-Ichi Uchiya
- Department of Microbiology, Faculty of Pharmacy, Meijo University, 150 Yagotoyama, Tempaku-ku, Nagoya 468-8503, Japan.
| | - Saki Isono
- Department of Microbiology, Faculty of Pharmacy, Meijo University, 150 Yagotoyama, Tempaku-ku, Nagoya 468-8503, Japan
| | - Misa Yoshimura
- Department of Microbiology, Faculty of Pharmacy, Meijo University, 150 Yagotoyama, Tempaku-ku, Nagoya 468-8503, Japan
| | - Takeaki Wajima
- Department of Microbiology, Faculty of Pharmacy, Meijo University, 150 Yagotoyama, Tempaku-ku, Nagoya 468-8503, Japan
| | - Toshiaki Nikai
- Department of Microbiology, Faculty of Pharmacy, Meijo University, 150 Yagotoyama, Tempaku-ku, Nagoya 468-8503, Japan
| |
Collapse
|
39
|
Chen D, Burford WB, Pham G, Zhang L, Alto LT, Ertelt JM, Winter MG, Winter SE, Way SS, Alto NM. Systematic reconstruction of an effector-gene network reveals determinants of Salmonella cellular and tissue tropism. Cell Host Microbe 2021; 29:1531-1544.e9. [PMID: 34536347 DOI: 10.1016/j.chom.2021.08.012] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 06/24/2021] [Accepted: 08/20/2021] [Indexed: 11/17/2022]
Abstract
The minimal genetic requirements for microbes to survive within multiorganism communities, including host-pathogen interactions, remain poorly understood. Here, we combined targeted gene mutagenesis with phenotype-guided genetic reassembly to identify a cooperative network of SPI-2 T3SS effector genes that are sufficient for Salmonella Typhimurium (STm) to cause disease in a natural host organism. Five SPI-2 effector genes support pathogen survival within the host cell cytoplasm by coordinating bacterial replication with Salmonella-containing vacuole (SCV) division. Unexpectedly, this minimal genetic repertoire does not support STm systemic infection of mice. In vivo screening revealed a second effector-gene network, encoded by the spv operon, that expands the life cycle of STm from growth in cells to deep-tissue colonization in a murine model of typhoid fever. Comparison between Salmonella infection models suggests how cooperation between effector genes drives tissue tropism in a pathogen group.
Collapse
Affiliation(s)
- Didi Chen
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Wesley B Burford
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Giang Pham
- Division of Infectious Diseases, Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Lishu Zhang
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Laura T Alto
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - James M Ertelt
- Division of Infectious Diseases, Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Maria G Winter
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Sebastian E Winter
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Sing Sing Way
- Division of Infectious Diseases, Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Neal M Alto
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
40
|
Groisman EA, Duprey A, Choi J. How the PhoP/PhoQ System Controls Virulence and Mg 2+ Homeostasis: Lessons in Signal Transduction, Pathogenesis, Physiology, and Evolution. Microbiol Mol Biol Rev 2021; 85:e0017620. [PMID: 34191587 PMCID: PMC8483708 DOI: 10.1128/mmbr.00176-20] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
The PhoP/PhoQ two-component system governs virulence, Mg2+ homeostasis, and resistance to a variety of antimicrobial agents, including acidic pH and cationic antimicrobial peptides, in several Gram-negative bacterial species. Best understood in Salmonella enterica serovar Typhimurium, the PhoP/PhoQ system consists o-regulated gene products alter PhoP-P amounts, even under constant inducing conditions. PhoP-P controls the abundance of hundreds of proteins both directly, by having transcriptional effects on the corresponding genes, and indirectly, by modifying the abundance, activity, or stability of other transcription factors, regulatory RNAs, protease regulators, and metabolites. The investigation of PhoP/PhoQ has uncovered novel forms of signal transduction and the physiological consequences of regulon evolution.
Collapse
Affiliation(s)
- Eduardo A. Groisman
- Department of Microbial Pathogenesis, Yale School of Medicine, New Haven, Connecticut, USA
- Yale Microbial Sciences Institute, West Haven, Connecticut, USA
| | - Alexandre Duprey
- Department of Microbial Pathogenesis, Yale School of Medicine, New Haven, Connecticut, USA
| | - Jeongjoon Choi
- Department of Microbial Pathogenesis, Yale School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
41
|
Reuter T, Scharte F, Franzkoch R, Liss V, Hensel M. Single cell analyses reveal distinct adaptation of typhoidal and non-typhoidal Salmonella enterica serovars to intracellular lifestyle. PLoS Pathog 2021; 17:e1009319. [PMID: 34143852 PMCID: PMC8244875 DOI: 10.1371/journal.ppat.1009319] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 06/30/2021] [Accepted: 05/14/2021] [Indexed: 01/06/2023] Open
Abstract
Salmonella enterica is a common foodborne, facultative intracellular enteropathogen. Human-restricted typhoidal S. enterica serovars Typhi (STY) or Paratyphi A (SPA) cause severe typhoid or paratyphoid fever, while many S. enterica serovar Typhimurium (STM) strains have a broad host range and in human hosts usually lead to a self-limiting gastroenteritis. Due to restriction of STY and SPA to primate hosts, experimental systems for studying the pathogenesis of typhoid and paratyphoid fever are limited. Therefore, STM infection of susceptible mice is commonly considered as model system for studying these diseases. The type III secretion system encoded by Salmonella pathogenicity island 2 (SPI2-T3SS) is a key factor for intracellular survival of Salmonella. Inside host cells, the pathogen resides within the Salmonella-containing vacuole (SCV) and induces tubular structures extending from the SCV, termed Salmonella-induced filaments (SIF). This study applies single cell analyses approaches, which are flow cytometry of Salmonella harboring dual fluorescent protein reporters, effector translocation, and correlative light and electron microscopy to investigate the fate and activities of intracellular STY and SPA. The SPI2-T3SS of STY and SPA is functional in translocation of effector proteins, SCV and SIF formation. However, only a low proportion of intracellular STY and SPA are actively deploying SPI2-T3SS and STY and SPA exhibited a rapid decline of protein biosynthesis upon experimental induction. A role of SPI2-T3SS for proliferation of STY and SPA in epithelial cells was observed, but not for survival or proliferation in phagocytic host cells. Our results indicate that reduced intracellular activities are factors of the stealth strategy of STY and SPA and facilitate systemic spread and persistence of the typhoidal Salmonella. Typhoidal Salmonella enterica serovars Typhi (STY) and Paratyphi A (SPA) cause a major disease burden to the human population. The restriction of these pathogens to human hosts limits experimental analyses of molecular mechanisms of diseases. S. enterica serovar Typhimurium is commonly used as surrogate model for typhoidal Salmonella (TS), and allowed the identification of virulence factors for intracellular lifestyle of S. enterica in mammalian host cells. If virulence factors, such as the Salmonella Pathogenicity Island 2-encoded type III secretion system (SPI2-T3SS) have similar roles for intracellular lifestyle of TS is largely unknown. We analyzed, on single cell level, the intracellular activities of STY and SPA in comparison to STM. STY and SPA deploy SPI2-T3SS to actively manipulate their host cells, but with far lower frequency than STM. Our work supports a model of TS as stealth pathogens that persist in host cells.
Collapse
Affiliation(s)
- Tatjana Reuter
- Abt. Mikrobiologie, Universität Osnabrück, Osnabrück, Germany
| | - Felix Scharte
- Abt. Mikrobiologie, Universität Osnabrück, Osnabrück, Germany
| | - Rico Franzkoch
- Abt. Mikrobiologie, Universität Osnabrück, Osnabrück, Germany
- iBiOs–integrated Bioimaging Facility Osnabrück, Universität Osnabrück, Osnabrück, Germany
| | - Viktoria Liss
- Abt. Mikrobiologie, Universität Osnabrück, Osnabrück, Germany
- iBiOs–integrated Bioimaging Facility Osnabrück, Universität Osnabrück, Osnabrück, Germany
| | - Michael Hensel
- Abt. Mikrobiologie, Universität Osnabrück, Osnabrück, Germany
- CellNanOs–Center of Cellular Nanoanalytics Osnabrück, Universität Osnabrück, Osnabrück, Germany
- * E-mail:
| |
Collapse
|
42
|
Ko D, Choi SH. Comparative genomics reveals an SNP potentially leading to phenotypic diversity of Salmonella enterica serovar Enteritidis. Microb Genom 2021; 7:000572. [PMID: 33952386 PMCID: PMC8209725 DOI: 10.1099/mgen.0.000572] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 03/29/2021] [Indexed: 12/25/2022] Open
Abstract
An SNP is a spontaneous genetic change having a potential to modify the functions of the original genes and to lead to phenotypic diversity of bacteria in nature. In this study, a phylogenetic analysis of Salmonella enterica serovar Enteritidis, a major food-borne pathogen, showed that eight strains of S. Enteritidis isolated in South Korea, including FORC_075 and FORC_078, have almost identical genome sequences. Interestingly, however, the abilities of FORC_075 to form biofilms and red, dry and rough (RDAR) colonies were significantly impaired, resulting in phenotypic differences among the eight strains. Comparative genomic analyses revealed that one of the non-synonymous SNPs unique to FORC_075 has occurred in envZ, which encodes a sensor kinase of the EnvZ/OmpR two-component system. The SNP in envZ leads to an amino acid change from Pro248 (CCG) in other strains including FORC_078 to Leu248 (CTG) in FORC_075. Allelic exchange of envZ between FORC_075 and FORC_078 identified that the SNP in envZ is responsible for the impaired biofilm- and RDAR colony-forming abilities of S. Enteritidis. Biochemical analyses demonstrated that the SNP in envZ significantly increases the phosphorylated status of OmpR in S. Enteritidis and alters the expression of the OmpR regulon. Phenotypic analyses further identified that the SNP in envZ decreases motility of S. Enteritidis but increases its adhesion and invasion to both human epithelial cells and murine macrophage cells. In addition to an enhancement of infectivity to the host cells, survival under acid stress was also elevated by the SNP in envZ. Together, these results suggest that the natural occurrence of the SNP in envZ could contribute to phenotypic diversity of S. Enteritidis, possibly improving its fitness and pathogenesis.
Collapse
Affiliation(s)
- Duhyun Ko
- National Research Laboratory of Molecular Microbiology and Toxicology, Department of Agricultural Biotechnology and Center for Food and Bioconvergence, Seoul National University, Seoul, Republic of Korea
| | - Sang Ho Choi
- National Research Laboratory of Molecular Microbiology and Toxicology, Department of Agricultural Biotechnology and Center for Food and Bioconvergence, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
43
|
Luk CH, Valenzuela C, Gil M, Swistak L, Bomme P, Chang YY, Mallet A, Enninga J. Salmonella enters a dormant state within human epithelial cells for persistent infection. PLoS Pathog 2021; 17:e1009550. [PMID: 33930101 PMCID: PMC8115778 DOI: 10.1371/journal.ppat.1009550] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 05/12/2021] [Accepted: 04/08/2021] [Indexed: 02/06/2023] Open
Abstract
Salmonella Typhimurium (S. Typhimurium) is an enteric bacterium capable of invading a wide range of hosts, including rodents and humans. It targets different host cell types showing different intracellular lifestyles. S. Typhimurium colonizes different intracellular niches and is able to either actively divide at various rates or remain dormant to persist. A comprehensive tool to determine these distinct S. Typhimurium lifestyles remains lacking. Here we developed a novel fluorescent reporter, Salmonella INtracellular Analyzer (SINA), compatible for fluorescence microscopy and flow cytometry in single-bacterium level quantification. This identified a S. Typhimurium subpopulation in infected epithelial cells that exhibits a unique phenotype in comparison to the previously documented vacuolar or cytosolic S. Typhimurium. This subpopulation entered a dormant state in a vesicular compartment distinct from the conventional Salmonella-containing vacuoles (SCV) as well as the previously reported niche of dormant S. Typhimurium in macrophages. The dormant S. Typhimurium inside enterocytes were viable and expressed Salmonella Pathogenicity Island 2 (SPI-2) virulence factors at later time points. We found that the formation of these dormant S. Typhimurium is not triggered by the loss of SPI-2 effector secretion but it is regulated by (p)ppGpp-mediated stringent response through RelA and SpoT. We predict that intraepithelial dormant S. Typhimurium represents an important pathogen niche and provides an alternative strategy for S. Typhimurium pathogenicity and its persistence. Salmonella Typhimurium is a clinically relevant bacterial pathogen that causes Salmonellosis. It can actively or passively invade various host cell types and reside in a Salmonella-containing vacuole (SCV) within host cells. The SCV can be remodeled into a replicative niche with the aid of Salmonella Type III Secretion System 2 (T3SS2) effectors or else, the SCV is ruptured for the access of the nutrient-rich host cytosol. Depending on the infected host cell type, S. Typhimurium undertake different lifestyles that are distinct by their subcellular localization, replication rate and metabolic rate. We present here a novel fluorescent reporter system that rapidly detects S. Typhimurium lifestyles using fluorescence microscopy and flow cytometry. We identified a dormant S. Typhimurium population within enterocyte that displays capacities in host cell persistence, dormancy exit and antibiotic tolerance. We deciphered the (p)ppGpp stringent response pathway that suppresses S. Typhimurium dormancy in enterocytes while promoting dormancy in macrophages, pinpointing a divergent physiological consequence regulated by the same set of S. Typhimurium molecular mediators. Altogether, our work demonstrated the potential of fluorescent reporters in facile bacterial characterization, and revealed a dormant S. Typhimurium population in human enterocytes that are phenotypically distinct from that observed in macrophages and fibroblasts.
Collapse
Affiliation(s)
- Chak Hon Luk
- Dynamics of Host-Pathogen Interactions Unit and UMR3691 CNRS, Institut Pasteur, Paris, France
- Université de Paris, Sorbonne Paris Cité, Paris, France
| | - Camila Valenzuela
- Dynamics of Host-Pathogen Interactions Unit and UMR3691 CNRS, Institut Pasteur, Paris, France
| | - Magdalena Gil
- Dynamics of Host-Pathogen Interactions Unit and UMR3691 CNRS, Institut Pasteur, Paris, France
| | - Léa Swistak
- Dynamics of Host-Pathogen Interactions Unit and UMR3691 CNRS, Institut Pasteur, Paris, France
- Université de Paris, Sorbonne Paris Cité, Paris, France
| | - Perrine Bomme
- Ultrastructural Bioimaging UTechS, C2RT, Institut Pasteur, Paris, France
| | - Yuen-Yan Chang
- Dynamics of Host-Pathogen Interactions Unit and UMR3691 CNRS, Institut Pasteur, Paris, France
| | - Adeline Mallet
- Ultrastructural Bioimaging UTechS, C2RT, Institut Pasteur, Paris, France
| | - Jost Enninga
- Dynamics of Host-Pathogen Interactions Unit and UMR3691 CNRS, Institut Pasteur, Paris, France
- Université de Paris, Sorbonne Paris Cité, Paris, France
- * E-mail:
| |
Collapse
|
44
|
Mónaco A, Canales-Huerta N, Jara-Wilde J, Härtel S, Chabalgoity JA, Moreno M, Scavone P. Salmonella Typhimurium Triggers Extracellular Traps Release in Murine Macrophages. Front Cell Infect Microbiol 2021; 11:639768. [PMID: 33981627 PMCID: PMC8107695 DOI: 10.3389/fcimb.2021.639768] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 04/06/2021] [Indexed: 12/23/2022] Open
Abstract
Salmonella comprises two species and more than 2500 serovars with marked differences in host specificity, and is responsible for a wide spectrum of diseases, ranging from localized gastroenteritis to severe life-threatening invasive disease. The initiation of the host inflammatory response, triggered by many Pathogen-Associated Molecular Patterns (PAMPs) that Salmonella possesses, recruits innate immune cells in order to restrain the infection at the local site. Neutrophils are known for killing bacteria through oxidative burst, amid other mechanisms. Amongst those mechanisms for controlling bacteria, the release of Extracellular Traps (ETs) represents a newly described pathway of programmed cell death known as ETosis. Particularly, Neutrophil Extracellular Traps (NETs) were first described in 2004 and since then, a number of reports have demonstrated their role as a novel defense mechanism against different pathogens. This released net-like material is composed of cellular DNA decorated with histones and cellular proteins. These structures have shown ability to trap, neutralize and kill different kinds of microorganisms, ranging from viruses and bacteria to fungi and parasites. Salmonella was one of the first microorganisms that were reported to be killed by NETs and several studies have confirmed the observation and deepened into its variants. Nevertheless, much less is known about their counterparts in other immune cells, e.g. Macrophage Extracellular Traps (METs) and Salmonella-induced MET release has never been reported so far. In this work, we observed the production of METs induced by Salmonella enterica serovar Typhimurium and recorded their effect on bacteria, showing for the first time that macrophages can also release extracellular DNA traps upon encounter with Salmonella Typhimurium. Additionally we show that METs effectively immobilize and reduce Salmonella survival in a few minutes, suggesting METs as a novel immune-mediated defense mechanism against Salmonella infection. Of note, this phenomenon was confirmed in primary macrophages, since MET release was also observed in bone marrow-derived macrophages infected with Salmonella. The evidence of this peculiar mechanism provides new incipient insights into macrophages´ role against Salmonella infection and can help to design new strategies for the clinical control of this transcendental pathogen.
Collapse
Affiliation(s)
- Amy Mónaco
- Departamento de Desarrollo Biotecnológico, Instituto de Higiene, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Nicole Canales-Huerta
- Laboratorio de Análisis de Imágenes Científicas SCIAN-Lab, Integrative Biology Program, Institute of Biomedical Sciences ICBM, Faculty of Medicine, University of Chile, Santiago de Chile, Chile.,Biomedical Neuroscience Institute BNI, Faculty of Medicine, University of Chile, Santiago de Chile, Chile
| | - Jorge Jara-Wilde
- Laboratorio de Análisis de Imágenes Científicas SCIAN-Lab, Integrative Biology Program, Institute of Biomedical Sciences ICBM, Faculty of Medicine, University of Chile, Santiago de Chile, Chile.,Biomedical Neuroscience Institute BNI, Faculty of Medicine, University of Chile, Santiago de Chile, Chile
| | - Steffen Härtel
- Laboratorio de Análisis de Imágenes Científicas SCIAN-Lab, Integrative Biology Program, Institute of Biomedical Sciences ICBM, Faculty of Medicine, University of Chile, Santiago de Chile, Chile.,Biomedical Neuroscience Institute BNI, Faculty of Medicine, University of Chile, Santiago de Chile, Chile.,Centro de Informática Médica y Telemedicina CIMT, Faculty of Medicine, University of Chile, Santiago de Chile, Chile.,National Center for Health Information Systems CENS, Santiago de Chile, Chile
| | - Jose Alejandro Chabalgoity
- Departamento de Desarrollo Biotecnológico, Instituto de Higiene, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - María Moreno
- Departamento de Desarrollo Biotecnológico, Instituto de Higiene, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Paola Scavone
- Departamento de Microbiología, Instituto de Investigaciones Biológicas Clemente Estable, Montevideo, Uruguay
| |
Collapse
|
45
|
Choi J, Groisman EA. Horizontally acquired regulatory gene activates ancestral regulatory system to promote Salmonella virulence. Nucleic Acids Res 2020; 48:10832-10847. [PMID: 33045730 PMCID: PMC7641745 DOI: 10.1093/nar/gkaa813] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 09/09/2020] [Accepted: 09/17/2020] [Indexed: 01/21/2023] Open
Abstract
Horizontally acquired genes are typically regulated by ancestral regulators. This regulation enables expression of horizontally acquired genes to be coordinated with that of preexisting genes. Here, we report a singular example of the opposite regulation: a horizontally acquired gene that controls an ancestral regulator, thereby promoting bacterial virulence. We establish that the horizontally acquired regulatory gene ssrB is necessary to activate the ancestral regulatory system PhoP/PhoQ of Salmonella enterica serovar Typhimurium (S. Typhimurium) in mildly acidic pH, which S. Typhimurium experiences inside macrophages. SsrB promotes phoP transcription by binding upstream of the phoP promoter. SsrB also increases ugtL transcription by binding to the ugtL promoter region, where it overcomes gene silencing by the heat-stable nucleoid structuring protein H-NS, enhancing virulence. The largely non-pathogenic species S. bongori failed to activate PhoP/PhoQ in mildly acidic pH because it lacks both the ssrB gene and the SsrB binding site in the target promoter. Low Mg2+ activated PhoP/PhoQ in both S. bongori and ssrB-lacking S. Typhimurium, indicating that the SsrB requirement for PhoP/PhoQ activation is signal-dependent. By controlling the ancestral genome, horizontally acquired genes are responsible for more crucial abilities, including virulence, than currently thought.
Collapse
Affiliation(s)
- Jeongjoon Choi
- Department of Microbial Pathogenesis, Yale School of Medicine, 295 Congress Avenue, New Haven, CT 06536, USA
| | - Eduardo A Groisman
- Department of Microbial Pathogenesis, Yale School of Medicine, 295 Congress Avenue, New Haven, CT 06536, USA.,Yale Microbial Sciences Institute, P.O. Box 27389, West Haven, CT 06516, USA
| |
Collapse
|
46
|
Antibodies and Protection in Systemic Salmonella Infections: Do We Still Have More Questions than Answers? Infect Immun 2020; 88:IAI.00219-20. [PMID: 32601109 DOI: 10.1128/iai.00219-20] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Salmonella causes grave systemic infections in humans and other animals and provides a paradigm for other diseases in which the bacteria have both intracellular and extracellular lifestyles. New generations of vaccines rely on the essential contribution of the antibody responses for their protection. The quality, antigen specificity, and functions associated with antibody responses to this pathogen have been elusive for a long time. Recent approaches that combine studies in humans and genetically manipulated experimental models and that exploit awareness of the location and within-host life cycle of the pathogen are shedding light on how humoral immunity to Salmonella operates. However, this area of research remains full of controversy and discrepancies. The overall scenario indicates that antibodies are essential for resistance against systemic Salmonella infections and can express the highest protective function when operating in conjunction with cell-mediated immunity. Antigen specificity, isotype profile, Fc-gamma receptor usage, and complement activation are all intertwined factors that still arcanely influence antibody-mediated protection to Salmonella.
Collapse
|
47
|
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
|
48
|
Gao R, Wang L, Ogunremi D. Virulence Determinants of Non-typhoidal Salmonellae. Microorganisms 2020. [DOI: 10.5772/intechopen.88904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
|
49
|
For the Greater (Bacterial) Good: Heterogeneous Expression of Energetically Costly Virulence Factors. Infect Immun 2020; 88:IAI.00911-19. [PMID: 32041785 DOI: 10.1128/iai.00911-19] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Bacterial populations are phenotypically heterogeneous, which allows subsets of cells to survive and thrive following changes in environmental conditions. For bacterial pathogens, changes within the host environment occur over the course of the immune response to infection and can result in exposure to host-derived, secreted antimicrobials or force direct interactions with immune cells. Many recent studies have shown host cell interactions promote virulence factor expression, forcing subsets of bacterial cells to battle the host response, while other bacteria reap the benefits of this pacification. It still remains unclear whether virulence factor expression is truly energetically costly within host tissues and whether expression is sufficient to impact the growth kinetics of virulence factor-expressing cells. However, it is clear that slow-growing subsets of bacteria emerge during infection and that these subsets are particularly difficult to eliminate with antibiotics. This minireview will focus on our current understanding of heterogenous virulence factor expression and discuss the evidence that supports or refutes the hypothesis that virulence factor expression is linked to slowed growth and antibiotic tolerance.
Collapse
|
50
|
Tsai CN, MacNair CR, Cao MPT, Perry JN, Magolan J, Brown ED, Coombes BK. Targeting Two-Component Systems Uncovers a Small-Molecule Inhibitor of Salmonella Virulence. Cell Chem Biol 2020; 27:793-805.e7. [PMID: 32413287 DOI: 10.1016/j.chembiol.2020.04.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 03/27/2020] [Accepted: 04/06/2020] [Indexed: 12/27/2022]
Abstract
Salmonella serovars are leading causes of gastrointestinal disease and have become increasingly resistant to fluoroquinolone and cephalosporin antibiotics. Overcoming this healthcare crisis requires new approaches in antibiotic discovery and the identification of unique bacterial targets. In this work, we describe a chemical genomics approach to identify inhibitors of Salmonella virulence. From a cell-based, promoter reporter screen of ∼50,000 small molecules, we identified dephostatin as a non-antibiotic compound that inhibits intracellular virulence factors and polymyxin resistance genes. Dephostatin disrupts signaling through both the SsrA-SsrB and PmrB-PmrA two-component regulatory systems and restores sensitivity to the last-resort antibiotic, colistin. Cell-based experiments and mouse models of infection demonstrate that dephostatin attenuates Salmonella virulence in vitro and in vivo, suggesting that perturbing regulatory networks is a promising strategy for the development of anti-infectives.
Collapse
Affiliation(s)
- Caressa N Tsai
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8S 4L8, Canada; Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Craig R MacNair
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8S 4L8, Canada; Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - My P T Cao
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, ON L8S 4L8, Canada; Department of Chemistry and Chemical Biology, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Jordyn N Perry
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8S 4L8, Canada; Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Jakob Magolan
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8S 4L8, Canada; Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, ON L8S 4L8, Canada; Department of Chemistry and Chemical Biology, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Eric D Brown
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8S 4L8, Canada; Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Brian K Coombes
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8S 4L8, Canada; Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, ON L8S 4L8, Canada.
| |
Collapse
|