1
|
Fernandez M, Yamanaka Y, Zangoui P, White MA, Kenney LJ. The sulfur assimilation pathway mitigates redox stress from acidic pH in Salmonella Typhi H58. mBio 2025:e0046725. [PMID: 40422406 DOI: 10.1128/mbio.00467-25] [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: 02/07/2025] [Accepted: 04/10/2025] [Indexed: 05/28/2025] Open
Abstract
Salmonella enterica serovar Typhi is the causative agent of typhoid fever, a human-restricted systemic infection. The rapidly disseminating multi-drug resistant H58 subclade is endemic in Africa, Asia, and Oceania, yet little is known regarding its intracellular behavior and virulence properties. It was of interest to understand the H58 response to host cell infection in terms of its response to acid stress and subsequent changes in gene regulation. We compared the H58 response in vitro and during infection of THP-1 human macrophages with the well-characterized response of Salmonella Typhimurium, which causes gastroenteritis. In S. Typhimurium infections, bacteria reside in an acidic intracellular vacuole and become acidified, driving the expression of pathogenicity island 2 genes (SPI-2). SPI-2 virulence factors modify the vacuole and enable bacterial replication. In response to acid stress, the sulfur assimilation pathway is highly upregulated and unique to H58. Replacing the Typhi cysK promoter with the Typhimurium promoter resulted in a cysK gene that was upregulated by acid stress in the H58 background, highlighting the differential regulation between the two serovars. In acidic conditions, H58 experienced much greater redox stress compared with S. Typhimurium, and the sulfur assimilation pathway was required to mitigate the redox stress. Higher redox stress modified the transcriptional regulator SsrB, resulting in diminished secretion of the SPI-2 virulence factor SifA. Our results highlight significant differences between S. Typhi H58 and S. Typhimurium and emphasize the importance of studying S. Typhi strains directly to understand their unique behavior during pathogenesis. IMPORTANCE In this study, we examined the clinically relevant, multi-drug resistant Salmonella Typhi strain H58, which is rapidly disseminating across Southeast Asia, Africa, and Oceania. It has heretofore been uncharacterized in terms of its gene regulation. Using human THP-1 macrophages, we discovered that S. Typhi strongly activates the sulfur utilization pathway in response to acid stress encountered in the vacuole once Typhi is inside host cells. Our novel findings were that S. Typhi experiences substantially higher redox stress compared with Typhimurium, and it requires the sulfur utilization pathway to mitigate this stress. This pathway is not upregulated in Typhimurium and represents a divergence in the response of these two serovars. We emphasize that S. Typhimurium is not a reasonable model for understanding H58, a serovar that is seriously impacting human health.
Collapse
Affiliation(s)
- Marion Fernandez
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Yuki Yamanaka
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
| | - Parisa Zangoui
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Mark Andrew White
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, Texas, USA
- Sealy Center for Structural Biology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Linda J Kenney
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, Texas, USA
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
- Sealy Center for Structural Biology, University of Texas Medical Branch, Galveston, Texas, USA
| |
Collapse
|
2
|
Guo J, He X, Bai Y, Sun H, Yang J. Virulence factors of Salmonella Typhi: interplay between the bacteria and host macrophages. Arch Microbiol 2025; 207:89. [PMID: 40095029 DOI: 10.1007/s00203-025-04297-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 02/20/2025] [Accepted: 03/05/2025] [Indexed: 03/19/2025]
Abstract
Salmonella Typhi (S. Typhi) is a Gram-negative bacterium that exclusively infects humans and causes typhoid fever- a major global public health concern responsible for approximately 9 million infections and 110,000 deaths annually. Macrophages, a key component of the innate immune system, play essential roles in pathogen clearance, antigen presentation, immune regulation, and tissue repair. As one of the primary targets of S. Typhi infection, macrophages significantly influence disease onset and progression. S. Typhi expresses a range of virulence factors, including the virulence-associated (Vi) capsule, outer membrane proteins (OMPs), flagella, fimbriae, type III secretion systems (T3SSs) and other genes encoded on Salmonella pathogenicity islands (SPIs), as well as toxins, regulatory factors, and virulence plasmids. These virulence factors facilitate S. Typhi's intracellular survival within macrophages by mediating processes such as adhesion, invasion, nutrient acquisition and immune evasion, ultimately enabling systemic infection. This review explores the role and molecular mechanisms of S. Typhi virulence factors in counteracting macrophage antimicrobial functions, providing insights for future research on typhoid pathogenesis and the development of potential therapeutic interventions.
Collapse
Affiliation(s)
- Jiayin Guo
- Cuiying Biomedical Research Center, The Second Hospital & Clinical Medical School, Lanzhou University, Cuiying Gate 82, Lanzhou, Gansu, 730030, China
| | - Xiaoe He
- Cuiying Biomedical Research Center, The Second Hospital & Clinical Medical School, Lanzhou University, Cuiying Gate 82, Lanzhou, Gansu, 730030, China
| | - Yanrui Bai
- Cuiying Biomedical Research Center, The Second Hospital & Clinical Medical School, Lanzhou University, Cuiying Gate 82, Lanzhou, Gansu, 730030, China
| | - Hui Sun
- Cuiying Biomedical Research Center, The Second Hospital & Clinical Medical School, Lanzhou University, Cuiying Gate 82, Lanzhou, Gansu, 730030, China
| | - Jing Yang
- Cuiying Biomedical Research Center, The Second Hospital & Clinical Medical School, Lanzhou University, Cuiying Gate 82, Lanzhou, Gansu, 730030, China.
| |
Collapse
|
3
|
Zhang Y, Xie Z, Yang F, Zhan L, Zhang Z, Sheng X, Xia L. Vi capsular polysaccharide of Salmonella enterica serovar Typhi disturbs autophagy to increase intracellular survival in macrophages. Microb Pathog 2025; 199:107265. [PMID: 39732413 DOI: 10.1016/j.micpath.2024.107265] [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: 11/03/2024] [Revised: 12/17/2024] [Accepted: 12/24/2024] [Indexed: 12/30/2024]
Abstract
The autophagy pathway plays a crucial role in resistance to bacterial infection in the host. Salmonella enterica serovar Typhi (S. Typhi), a human restricted pathogen, causes a systemic infection known as typhoid fever. Vi capsular polysaccharide not only forms a physical barrier on the surface of S. Typhi but also serves as an important virulence factor. Here, the effects of Vi expression on autophagy of host cells were investigated. Vi was highly expressed in super optimal broth (SOB) medium for 8 h and in the early stage of macrophage infection. Strains of S. Typhi with a mutation in Vi capsule, ΔtviA and ΔvexE, were constructed and showed lower intracellular survival in macrophage THP-1 cells compared with wild type strain. Western blot, immunofluorescence and flow cytometry were used to detected the autophagy level of macrophages infected by Vi mutant and wild type strains, respectively. Autophagy receptor p62 protein level significantly decreased and LC3-II protein level significantly increased in Vi mutant strains compared with wild type strain, which indicated increased autophagy in macrophages infected by Vi mutant strains. The qRT-PCR and Western blot results showed that not Nod1, but Nod2 and Galectin-8 were up-regulated in Vi mutant strains. In summary, we propose that Vi capsule of S. Typhi decreased autophagy of macrophages to increase its survival in host cells by decreasing the expression of Nod2 and Galectin-8.
Collapse
Affiliation(s)
- Ying Zhang
- Department of Biochemistry and Molecular Biology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212013, China
| | - Zhongyi Xie
- Department of Environmental Health, Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, Zhejiang, 310000, China
| | - Fanfan Yang
- Department of Microorganism Examination, Ningbo Center for Disease Control and Prevention, Ningbo, Zhejiang, 315000, China
| | - Lina Zhan
- Department of Biochemistry and Molecular Biology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212013, China
| | - Zuowei Zhang
- Department of Biochemistry and Molecular Biology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212013, China
| | - Xiumei Sheng
- Department of Biochemistry and Molecular Biology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212013, China
| | - Lin Xia
- Department of Laboratory Medicine, Affiliated Hospital, Jiangsu University, Zhenjiang, Jiangsu, 212001, China.
| |
Collapse
|
4
|
Krone L, Mahankali S, Geiger T. Cytolysin A is an intracellularly induced and secreted cytotoxin of typhoidal Salmonella. Nat Commun 2024; 15:8414. [PMID: 39341826 PMCID: PMC11438861 DOI: 10.1038/s41467-024-52745-0] [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: 01/16/2024] [Accepted: 09/20/2024] [Indexed: 10/01/2024] Open
Abstract
Typhoidal Salmonella enterica serovars, such as Typhi and Paratyphi A, cause severe systemic infections, thereby posing a significant threat as human-adapted pathogens. This study focuses on cytolysin A (ClyA), a virulence factor essential for bacterial dissemination within the human body. We show that ClyA is exclusively expressed by intracellular S. Paratyphi A within the Salmonella-containing vacuole (SCV), regulated by the PhoP/Q system and SlyA. ClyA localizes in the bacterial periplasm, suggesting potential secretion. Deletion of TtsA, an essential Type 10 Secretion System component, completely abolishes intracellular ClyA detection and its presence in host cell supernatants. Host cells infected with wild-type S. Paratyphi A contain substantial ClyA, with supernatants capable of lysing neighboring cells. Notably, ClyA selectively lyses macrophages and erythrocytes while sparing epithelial cells. These findings identify ClyA as an intracellularly induced cytolysin, dependent on the SCV environment and secreted via a Type 10 Secretion System, with specific cytolytic activity.
Collapse
Affiliation(s)
- Lena Krone
- Max von Pettenkofer-Institute, Chair for Medical Microbiology and Hygiene, Ludwig-Maximilians-Universität München (LMU Munich), Munich, Germany
| | - Srujita Mahankali
- Max von Pettenkofer-Institute, Chair for Medical Microbiology and Hygiene, Ludwig-Maximilians-Universität München (LMU Munich), Munich, Germany
| | - Tobias Geiger
- Max von Pettenkofer-Institute, Chair for Medical Microbiology and Hygiene, Ludwig-Maximilians-Universität München (LMU Munich), Munich, Germany.
| |
Collapse
|
5
|
Ackroyd BK, Dodson EJ, Mehboob J, Dowle AA, Thomas GH, Wilkinson AJ. Structure and ligand binding in the putative anti-microbial peptide transporter protein, YejA. MICROBIOLOGY (READING, ENGLAND) 2024; 170:001430. [PMID: 38334478 PMCID: PMC10924461 DOI: 10.1099/mic.0.001430] [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: 11/20/2023] [Accepted: 01/19/2024] [Indexed: 02/10/2024]
Abstract
YejABEF is an ATP-binding cassette transporter that is implicated in the sensitivity of Escherichia coli to anti-microbial peptides, the best-characterized example being microcin C, a peptide-nucleotide antibiotic that targets aspartyl-tRNA synthetase. Here the structure of the extracellular solute binding protein, YejA, has been determined, revealing an oligopeptide-binding protein fold enclosing a ligand-binding pocket larger than those of other peptide-binding proteins of known structure. Prominent electron density in this cavity defines an undecapeptide sequence LGEPRYAFNFN, an observation that is confirmed by mass spectrometry. In the structure, the peptide interactions with the protein are mediated by main chain hydrogen bonds with the exception of Arg5 whose guanidinium side chain makes a set of defining polar interactions with four YejA residues. More detailed characterization of purified recombinant YejA, by a combination of ESI and MALDI-mass spectrometry as well as thermal shift assays, reveals a set of YejA complexes containing overlapping peptides 10-19 residues in length. All contain the sequence LGEPRYAFN. Curiously, these peptides correspond to residues 8-26 of the mature YejA protein, which belong to a unique N-terminal extension that distinguishes YejA from other cluster C oligopeptide binding proteins of known structure. This 35-residue extension is well-ordered and packs across the surface of the protein. The undecapeptide ligand occupies only a fraction of the enclosed pocket volume suggesting the possibility that much larger peptides or peptide conjugates could be accommodated, though thermal shift assays of YejA binding to antimicrobial peptides and peptides unrelated to LGEPRYAFNFN have not provided evidence of binding. While the physiological significance of this 'auto-binding' is not clear, the experimental data suggest that it is not an artefact of the crystallization process and that it may have a function in the sensing of periplasmic or membrane stress.
Collapse
Affiliation(s)
- Bryony K. Ackroyd
- York Structural Biology Laboratory and York Biomedical Research Institute, Department of Chemistry, University of York, York YO10 5DD, UK
- Department of Biology and York Biomedical Research Institute, University of York, York YO10 5DD, UK
| | - Eleanor J. Dodson
- York Structural Biology Laboratory and York Biomedical Research Institute, Department of Chemistry, University of York, York YO10 5DD, UK
| | - Javeria Mehboob
- Department of Biology and York Biomedical Research Institute, University of York, York YO10 5DD, UK
| | - Adam A. Dowle
- Bioscience Technology Facility, Department of Biology, University of York, York YO10 5DD, UK
| | - Gavin H. Thomas
- Department of Biology and York Biomedical Research Institute, University of York, York YO10 5DD, UK
| | - Anthony J. Wilkinson
- York Structural Biology Laboratory and York Biomedical Research Institute, Department of Chemistry, University of York, York YO10 5DD, UK
| |
Collapse
|
6
|
Carey ME, Dyson ZA, Ingle DJ, Amir A, Aworh MK, Chattaway MA, Chew KL, Crump JA, Feasey NA, Howden BP, Keddy KH, Maes M, Parry CM, Van Puyvelde S, Webb HE, Afolayan AO, Alexander AP, Anandan S, Andrews JR, Ashton PM, Basnyat B, Bavdekar A, Bogoch II, Clemens JD, da Silva KE, De A, de Ligt J, Diaz Guevara PL, Dolecek C, Dutta S, Ehlers MM, Francois Watkins L, Garrett DO, Godbole G, Gordon MA, Greenhill AR, Griffin C, Gupta M, Hendriksen RS, Heyderman RS, Hooda Y, Hormazabal JC, Ikhimiukor OO, Iqbal J, Jacob JJ, Jenkins C, Jinka DR, John J, Kang G, Kanteh A, Kapil A, Karkey A, Kariuki S, Kingsley RA, Koshy RM, Lauer AC, Levine MM, Lingegowda RK, Luby SP, Mackenzie GA, Mashe T, Msefula C, Mutreja A, Nagaraj G, Nagaraj S, Nair S, Naseri TK, Nimarota-Brown S, Njamkepo E, Okeke IN, Perumal SPB, Pollard AJ, Pragasam AK, Qadri F, Qamar FN, Rahman SIA, Rambocus SD, Rasko DA, Ray P, Robins-Browne R, Rongsen-Chandola T, Rutanga JP, Saha SK, Saha S, Saigal K, Sajib MSI, Seidman JC, Shakya J, Shamanna V, Shastri J, Shrestha R, Sia S, Sikorski MJ, Singh A, Smith AM, Tagg KA, Tamrakar D, Tanmoy AM, Thomas M, Thomas MS, et alCarey ME, Dyson ZA, Ingle DJ, Amir A, Aworh MK, Chattaway MA, Chew KL, Crump JA, Feasey NA, Howden BP, Keddy KH, Maes M, Parry CM, Van Puyvelde S, Webb HE, Afolayan AO, Alexander AP, Anandan S, Andrews JR, Ashton PM, Basnyat B, Bavdekar A, Bogoch II, Clemens JD, da Silva KE, De A, de Ligt J, Diaz Guevara PL, Dolecek C, Dutta S, Ehlers MM, Francois Watkins L, Garrett DO, Godbole G, Gordon MA, Greenhill AR, Griffin C, Gupta M, Hendriksen RS, Heyderman RS, Hooda Y, Hormazabal JC, Ikhimiukor OO, Iqbal J, Jacob JJ, Jenkins C, Jinka DR, John J, Kang G, Kanteh A, Kapil A, Karkey A, Kariuki S, Kingsley RA, Koshy RM, Lauer AC, Levine MM, Lingegowda RK, Luby SP, Mackenzie GA, Mashe T, Msefula C, Mutreja A, Nagaraj G, Nagaraj S, Nair S, Naseri TK, Nimarota-Brown S, Njamkepo E, Okeke IN, Perumal SPB, Pollard AJ, Pragasam AK, Qadri F, Qamar FN, Rahman SIA, Rambocus SD, Rasko DA, Ray P, Robins-Browne R, Rongsen-Chandola T, Rutanga JP, Saha SK, Saha S, Saigal K, Sajib MSI, Seidman JC, Shakya J, Shamanna V, Shastri J, Shrestha R, Sia S, Sikorski MJ, Singh A, Smith AM, Tagg KA, Tamrakar D, Tanmoy AM, Thomas M, Thomas MS, Thomsen R, Thomson NR, Tupua S, Vaidya K, Valcanis M, Veeraraghavan B, Weill FX, Wright J, Dougan G, Argimón S, Keane JA, Aanensen DM, Baker S, Holt KE. Global diversity and antimicrobial resistance of typhoid fever pathogens: Insights from a meta-analysis of 13,000 Salmonella Typhi genomes. eLife 2023; 12:e85867. [PMID: 37697804 PMCID: PMC10506625 DOI: 10.7554/elife.85867] [Show More Authors] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 08/02/2023] [Indexed: 09/13/2023] Open
Abstract
BACKGROUND The Global Typhoid Genomics Consortium was established to bring together the typhoid research community to aggregate and analyse Salmonella enterica serovar Typhi (Typhi) genomic data to inform public health action. This analysis, which marks 22 years since the publication of the first Typhi genome, represents the largest Typhi genome sequence collection to date (n=13,000). METHODS This is a meta-analysis of global genotype and antimicrobial resistance (AMR) determinants extracted from previously sequenced genome data and analysed using consistent methods implemented in open analysis platforms GenoTyphi and Pathogenwatch. RESULTS Compared with previous global snapshots, the data highlight that genotype 4.3.1 (H58) has not spread beyond Asia and Eastern/Southern Africa; in other regions, distinct genotypes dominate and have independently evolved AMR. Data gaps remain in many parts of the world, and we show the potential of travel-associated sequences to provide informal 'sentinel' surveillance for such locations. The data indicate that ciprofloxacin non-susceptibility (>1 resistance determinant) is widespread across geographies and genotypes, with high-level ciprofloxacin resistance (≥3 determinants) reaching 20% prevalence in South Asia. Extensively drug-resistant (XDR) typhoid has become dominant in Pakistan (70% in 2020) but has not yet become established elsewhere. Ceftriaxone resistance has emerged in eight non-XDR genotypes, including a ciprofloxacin-resistant lineage (4.3.1.2.1) in India. Azithromycin resistance mutations were detected at low prevalence in South Asia, including in two common ciprofloxacin-resistant genotypes. CONCLUSIONS The consortium's aim is to encourage continued data sharing and collaboration to monitor the emergence and global spread of AMR Typhi, and to inform decision-making around the introduction of typhoid conjugate vaccines (TCVs) and other prevention and control strategies. FUNDING No specific funding was awarded for this meta-analysis. Coordinators were supported by fellowships from the European Union (ZAD received funding from the European Union's Horizon 2020 research and innovation programme under the Marie Sklodowska-Curie grant agreement No 845681), the Wellcome Trust (SB, Wellcome Trust Senior Fellowship), and the National Health and Medical Research Council (DJI is supported by an NHMRC Investigator Grant [GNT1195210]).
Collapse
Affiliation(s)
- Megan E Carey
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), University of Cambridge School of Clinical Medicine, Cambridge Biomedical CampusCambridgeUnited Kingdom
- Department of Infection Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical MedicineLondonUnited Kingdom
- IAVI, Chelsea & Westminster HospitalLondonUnited Kingdom
| | - Zoe A Dyson
- Department of Infection Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical MedicineLondonUnited Kingdom
- Department of Infectious Diseases, Central Clinical School, Monash UniversityMelbourneAustralia
- Wellcome Sanger Institute, Wellcome Genome CampusHinxtonUnited Kingdom
| | - Danielle J Ingle
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, The University of MelbourneMelbourneAustralia
| | | | - Mabel K Aworh
- Nigeria Field Epidemiology and Laboratory Training ProgrammeAbujaNigeria
- College of Veterinary Medicine, North Carolina State UniversityRaleighUnited States
| | | | - Ka Lip Chew
- National University HospitalSingaporeSingapore
| | - John A Crump
- Centre for International Health, University of OtagoDunedinNew Zealand
| | - Nicholas A Feasey
- Department of Clinical Sciences, Liverpool School of Tropical MedicineLiverpoolUnited Kingdom
- Malawi-Liverpool Wellcome Programme, Kamuzu University of Health SciencesBlantyreMalawi
| | - Benjamin P Howden
- Centre for Pathogen Genomics, Department of Microbiology and Immunology, University of Melbourne at Doherty Institute for Infection and ImmunityMelbourneAustralia
- Microbiological Diagnostic Unit Public Health Laboratory, The University of Melbourne at the Peter Doherty Institute for Infection and ImmunityMelbourneAustralia
| | | | - Mailis Maes
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), University of Cambridge School of Clinical Medicine, Cambridge Biomedical CampusCambridgeUnited Kingdom
| | - Christopher M Parry
- Department of Clinical Sciences, Liverpool School of Tropical MedicineLiverpoolUnited Kingdom
| | - Sandra Van Puyvelde
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), University of Cambridge School of Clinical Medicine, Cambridge Biomedical CampusCambridgeUnited Kingdom
- University of AntwerpAntwerpBelgium
| | - Hattie E Webb
- Centers for Disease Control and PreventionAtlantaUnited States
| | - Ayorinde Oluwatobiloba Afolayan
- Global Health Research Unit (GHRU) for the Genomic Surveillance of Antimicrobial Resistance, Faculty of Pharmacy, University of IbadanIbadanNigeria
| | | | - Shalini Anandan
- Department of Clinical Microbiology, Christian Medical CollegeVelloreIndia
| | - Jason R Andrews
- Division of Infectious Diseases and Geographic Medicine, Stanford UniversityStanfordUnited States
| | - Philip M Ashton
- Malawi-Liverpool Wellcome ProgrammeBlantyreMalawi
- Institute of Infection, Veterinary and Ecological Sciences, University of LiverpoolLiverpoolUnited Kingdom
| | - Buddha Basnyat
- Oxford University Clinical Research Unit NepalKathmanduNepal
| | | | - Isaac I Bogoch
- Department of Medicine, Division of Infectious Diseases, University of TorontoTorontoCanada
| | - John D Clemens
- International Vaccine InstituteSeoulRepublic of Korea
- International Centre for Diarrhoeal Disease ResearchDhakaBangladesh
- UCLA Fielding School of Public HealthLos AngelesUnited States
- Korea UniversitySeoulRepublic of Korea
| | - Kesia Esther da Silva
- Division of Infectious Diseases and Geographic Medicine, Stanford UniversityStanfordUnited States
| | - Anuradha De
- Topiwala National Medical CollegeMumbaiIndia
| | - Joep de Ligt
- ESR, Institute of Environmental Science and Research Ltd., PoriruaWellingtonNew Zealand
| | | | - Christiane Dolecek
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of OxfordOxfordUnited Kingdom
- Mahidol Oxford Tropical Medicine Research Unit, Mahidol UniversityBangkokThailand
| | - Shanta Dutta
- ICMR - National Institute of Cholera & Enteric DiseasesKolkataIndia
| | - Marthie M Ehlers
- Department of Medical Microbiology, Faculty of Health Sciences, University of PretoriaPretoriaSouth Africa
- Department of Medical Microbiology, Tshwane Academic Division, National Health Laboratory ServicePretoriaSouth Africa
| | | | | | - Gauri Godbole
- United Kingdom Health Security AgencyLondonUnited Kingdom
| | - Melita A Gordon
- Institute of Infection, Veterinary and Ecological Sciences, University of LiverpoolLiverpoolUnited Kingdom
| | - Andrew R Greenhill
- Federation University AustraliaChurchillAustralia
- Papua New Guinea Institute of Medical ResearchGorokaPapua New Guinea
| | - Chelsey Griffin
- Centers for Disease Control and PreventionAtlantaUnited States
| | - Madhu Gupta
- Post Graduate Institute of Medical Education and ResearchChandigarhIndia
| | | | - Robert S Heyderman
- Research Department of Infection, Division of Infection and Immunity, University College LondonLondonUnited Kingdom
| | | | - Juan Carlos Hormazabal
- Bacteriologia, Subdepartamento de Enfermedades Infecciosas, Departamento de Laboratorio Biomedico, Instituto de Salud Publica de Chile (ISP)SantiagoChile
| | - Odion O Ikhimiukor
- Global Health Research Unit (GHRU) for the Genomic Surveillance of Antimicrobial Resistance, Faculty of Pharmacy, University of IbadanIbadanNigeria
| | - Junaid Iqbal
- Department of Pediatrics and Child Health, Aga Khan UniversityKarachiPakistan
| | - Jobin John Jacob
- Department of Clinical Microbiology, Christian Medical CollegeVelloreIndia
| | - Claire Jenkins
- United Kingdom Health Security AgencyLondonUnited Kingdom
| | | | - Jacob John
- Department of Community Health, Christian Medical CollegeVelloreIndia
| | - Gagandeep Kang
- Department of Community Health, Christian Medical CollegeVelloreIndia
| | - Abdoulie Kanteh
- Medical Research Council Unit The Gambia at London School Hygiene & Tropical MedicineFajaraGambia
| | - Arti Kapil
- All India Institute of Medical SciencesDelhiIndia
| | | | - Samuel Kariuki
- Centre for Microbiology Research, Kenya Medical Research InstituteNairobiKenya
| | | | | | - AC Lauer
- Centers for Disease Control and PreventionAtlantaUnited States
| | - Myron M Levine
- Center for Vaccine Development and Global Health (CVD), University of Maryland School of Medicine, Baltimore, Maryland, USABaltimoreUnited States
| | | | - Stephen P Luby
- Division of Infectious Diseases and Geographic Medicine, Stanford UniversityStanfordUnited States
| | - Grant Austin Mackenzie
- Medical Research Council Unit The Gambia at London School Hygiene & Tropical MedicineFajaraGambia
| | - Tapfumanei Mashe
- National Microbiology Reference LaboratoryHarareZimbabwe
- World Health OrganizationHarareZimbabwe
| | | | - Ankur Mutreja
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), University of Cambridge School of Clinical Medicine, Cambridge Biomedical CampusCambridgeUnited Kingdom
| | - Geetha Nagaraj
- Central Research Laboratory, Kempegowda Institute of Medical SciencesBengaluruIndia
| | | | - Satheesh Nair
- United Kingdom Health Security AgencyLondonUnited Kingdom
| | | | | | | | - Iruka N Okeke
- Global Health Research Unit (GHRU) for the Genomic Surveillance of Antimicrobial Resistance, Faculty of Pharmacy, University of IbadanIbadanNigeria
| | | | - Andrew J Pollard
- Oxford Vaccine Group, Department of Paediatrics, University of OxfordOxfordUnited Kingdom
- The NIHR Oxford Biomedical Research CentreOxfordUnited Kingdom
| | | | - Firdausi Qadri
- International Centre for Diarrhoeal Disease ResearchDhakaBangladesh
| | - Farah N Qamar
- Department of Pediatrics and Child Health, Aga Khan UniversityKarachiPakistan
| | | | - Savitra Devi Rambocus
- Microbiological Diagnostic Unit Public Health Laboratory, The University of Melbourne at the Peter Doherty Institute for Infection and ImmunityMelbourneAustralia
| | - David A Rasko
- Department of Microbiology and Immunology, University of Maryland School of MedicineBaltimoreUnited States
- Institute for Genome Sciences, University of Maryland School of MedicineBaltimoreUnited States
| | - Pallab Ray
- Post Graduate Institute of Medical Education and ResearchChandigarhIndia
| | - Roy Robins-Browne
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, The University of MelbourneMelbourneAustralia
- Murdoch Children’s Research Institute, Royal Children’s HospitalParkvilleAustralia
| | | | | | | | | | | | - Mohammad Saiful Islam Sajib
- Child Health Research FoundationDhakaBangladesh
- Institute of Biodiversity, Animal Health and Comparative Medicine, University of GlasgowGlasgowUnited Kingdom
| | | | - Jivan Shakya
- Dhulikhel HospitalDhulikhelNepal
- Institute for Research in Science and TechnologyKathmanduNepal
| | - Varun Shamanna
- Central Research Laboratory, Kempegowda Institute of Medical SciencesBengaluruIndia
| | - Jayanthi Shastri
- Topiwala National Medical CollegeMumbaiIndia
- Kasturba Hospital for Infectious DiseasesMumbaiIndia
| | - Rajeev Shrestha
- Center for Infectious Disease Research & Surveillance, Dhulikhel Hospital, Kathmandu University HospitalDhulikhelNepal
| | - Sonia Sia
- Research Institute for Tropical Medicine, Department of HealthMuntinlupa CityPhilippines
| | - Michael J Sikorski
- Center for Vaccine Development and Global Health (CVD), University of Maryland School of Medicine, Baltimore, Maryland, USABaltimoreUnited States
- Department of Microbiology and Immunology, University of Maryland School of MedicineBaltimoreUnited States
- Institute for Genome Sciences, University of Maryland School of MedicineBaltimoreUnited States
| | | | - Anthony M Smith
- Centre for Enteric Diseases, National Institute for Communicable DiseasesJohannesburgSouth Africa
| | - Kaitlin A Tagg
- Centers for Disease Control and PreventionAtlantaUnited States
| | - Dipesh Tamrakar
- Center for Infectious Disease Research & Surveillance, Dhulikhel Hospital, Kathmandu University HospitalDhulikhelNepal
| | | | - Maria Thomas
- Christian Medical College, LudhianaLudhianaIndia
| | | | | | | | - Siaosi Tupua
- Ministry of Health, Government of SamoaApiaSamoa
| | | | - Mary Valcanis
- Microbiological Diagnostic Unit Public Health Laboratory, The University of Melbourne at the Peter Doherty Institute for Infection and ImmunityMelbourneAustralia
| | | | | | - Jackie Wright
- ESR, Institute of Environmental Science and Research Ltd., PoriruaWellingtonNew Zealand
| | - Gordon Dougan
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), University of Cambridge School of Clinical Medicine, Cambridge Biomedical CampusCambridgeUnited Kingdom
| | - Silvia Argimón
- Centre for Genomic Pathogen Surveillance, Big Data Institute, University of OxfordOxfordUnited Kingdom
| | - Jacqueline A Keane
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), University of Cambridge School of Clinical Medicine, Cambridge Biomedical CampusCambridgeUnited Kingdom
| | - David M Aanensen
- Centre for Genomic Pathogen Surveillance, Big Data Institute, University of OxfordOxfordUnited Kingdom
| | - Stephen Baker
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), University of Cambridge School of Clinical Medicine, Cambridge Biomedical CampusCambridgeUnited Kingdom
- IAVI, Chelsea & Westminster HospitalLondonUnited Kingdom
| | - Kathryn E Holt
- Department of Infection Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical MedicineLondonUnited Kingdom
- Department of Infectious Diseases, Central Clinical School, Monash UniversityMelbourneAustralia
| |
Collapse
|
7
|
Hamblin M, Schade R, Narasimhan R, Monack DM. Salmonella enterica serovar Typhi uses two type 3 secretion systems to replicate in human macrophages and colonize humanized mice. mBio 2023; 14:e0113723. [PMID: 37341487 PMCID: PMC10470537 DOI: 10.1128/mbio.01137-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 05/04/2023] [Indexed: 06/22/2023] Open
Abstract
Salmonella enterica serovar Typhi (S. Typhi) is a human-restricted pathogen that replicates in macrophages. In this study, we investigated the roles of the S. Typhi type 3 secretion systems (T3SSs) encoded on Salmonella pathogenicity islands (SPI)-1 (T3SS-1) and SPI-2 (T3SS-2) during human macrophage infection. We found that mutants of S. Typhi deficient for both T3SSs were defective for intramacrophage replication as measured by flow cytometry, viable bacterial counts, and live time-lapse microscopy. T3SS-secreted proteins PipB2 and SifA contributed to S. Typhi replication and were translocated into the cytosol of human macrophages through both T3SS-1 and T3SS-2, demonstrating functional redundancy for these secretion systems. Importantly, an S. Typhi mutant strain that is deficient for both T3SS-1 and T3SS-2 was severely attenuated in the ability to colonize systemic tissues in a humanized mouse model of typhoid fever. Overall, this study establishes a critical role for S. Typhi T3SSs during its replication within human macrophages and during systemic infection of humanized mice. IMPORTANCE Salmonella enterica serovar Typhi is a human-restricted pathogen that causes typhoid fever. Understanding the key virulence mechanisms that facilitate S. Typhi replication in human phagocytes will enable rational vaccine and antibiotic development to limit the spread of this pathogen. While S. Typhimurium replication in murine models has been studied extensively, there is limited information available about S. Typhi replication in human macrophages, some of which directly conflict with findings from S. Typhimurium murine models. This study establishes that both of S. Typhi's two type 3 secretion systems (T3SS-1 and T3SS-2) contribute to intramacrophage replication and virulence.
Collapse
Affiliation(s)
- Meagan Hamblin
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California, USA
| | - Ruth Schade
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California, USA
| | - Ramya Narasimhan
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California, USA
| | - Denise M. Monack
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
8
|
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: 10] [Impact Index Per Article: 5.0] [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
|
9
|
Hamblin M, Schade R, Narasimhan R, Monack DM. Salmonella enterica serovar Typhi uses two type 3 secretion systems to replicate in human macrophages and to colonize humanized mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.06.543980. [PMID: 37333307 PMCID: PMC10274799 DOI: 10.1101/2023.06.06.543980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Salmonella enterica serovar Typhi ( S. Typhi) is a human-restricted pathogen that replicates in macrophages. In this study, we investigated the roles of the S. Typhi Type 3 secretion systems (T3SSs) encoded on Salmonella Pathogenicity Islands (SPI) -1 (T3SS-1) and -2 (T3SS-2) during human macrophage infection. We found that mutants of S . Typhi deficient for both T3SSs were defective for intramacrophage replication as measured by flow cytometry, viable bacterial counts, and live time-lapse microscopy. T3SS-secreted proteins PipB2 and SifA contributed to S. Typhi replication and were translocated into the cytosol of human macrophages through both T3SS-1 and -2, demonstrating functional redundancy for these secretion systems. Importantly, an S . Typhi mutant strain that is deficient for both T3SS-1 and -2 was severely attenuated in the ability to colonize systemic tissues in a humanized mouse model of typhoid fever. Overall, this study establishes a critical role for S. Typhi T3SSs during its replication within human macrophages and during systemic infection of humanized mice. Importance Salmonella enterica serovar Typhi is a human-restricted pathogen that causes typhoid fever. Understanding the key virulence mechanisms that facilitate S. Typhi replication in human phagocytes will enable rational vaccine and antibiotic development to limit spread of this pathogen. While S. Typhimurium replication in murine models has been studied extensively, there is limited information available about S. Typhi replication in human macrophages, some of which directly conflicts with findings from S. Typhimurium murine models. This study establishes that both of S. Typhi's two Type 3 Secretion Systems (T3SS-1 and -2) contribute to intramacrophage replication and virulence.
Collapse
|
10
|
Xu Y, Abdelhamid AG, Sabag-Daigle A, Sovic MG, Ahmer BM, Yousef AE. The Role of Egg Yolk in Modulating the Virulence of Salmonella Enterica Serovar Enteritidis. Front Cell Infect Microbiol 2022; 12:903979. [PMID: 35774398 PMCID: PMC9237210 DOI: 10.3389/fcimb.2022.903979] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 04/15/2022] [Indexed: 11/13/2022] Open
Abstract
Contribution of food vehicles to pathogenicity of disease-causing microorganisms is an important but overlooked research field. The current study was initiated to reveal the relationship between virulence of Salmonella enterica serovar Enteritidis and egg yolk as a hosting medium. Mice were orally challenged with Salmonella Enteritidis cultured in egg yolk or tryptic soy broth (TSB). Additionally, mice were challenged with Salmonella Enteritidis cultured in TSB, followed by administration of sterile egg yolk, to discern the difference between pre-growth of the pathogen and its mere presence in egg yolk during infection. The pathogen's Lethal dose 50 (LD50) was the lowest when grown in yolk (2.8×102 CFU), compared to 1.1×103 CFU in TSB, and 4.6×103 CFU in TSB followed by administration of sterile yolk. Additionally, mice that orally received Salmonella Enteritidis grown in egg yolk expressed a high death rate. These findings were supported by transcriptional analysis results. Expression of promoters of virulence-related genes (sopB and sseA) in genetically modified Salmonella Enteritidis reporter strains was significantly higher (p < 0.05) when the bacterium was grown in the yolk, compared to that grown in TSB. Sequencing of RNA (RNA-seq) revealed 204 differentially transcribed genes in Salmonella Enteritidis grown in yolk vs. TSB. Yolk-grown Salmonella Enteritidis exhibited upregulated virulence pathways, including type III secretion systems, epithelial cell invasion, and infection processes; these observations were confirmed by RT-qPCR results. The transcriptomic analysis suggested that upregulation of virulence machinery of Salmonella Enteritidis grown in egg yolk was related to increased iron uptake, biotin utilization, flagellar biosynthesis, and export of virulence proteins encoded on Salmonella pathogenicity island 1, 2, 4, and 5. These biological responses may have acted in concert to increase the virulence of Salmonella infection in mice. In conclusion, growth in egg yolk enhanced Salmonella Enteritidis virulence, indicating the significance of this food vehicle to the risk assessment of salmonellosis.
Collapse
Affiliation(s)
- Yumin Xu
- Department of Food Science and Technology, The Ohio State University, Columbus, OH, United States
| | - Ahmed G. Abdelhamid
- Department of Food Science and Technology, The Ohio State University, Columbus, OH, United States
- Botany and Microbiology Department, Faculty of Science, Benha University, Benha, Egypt
| | - Anice Sabag-Daigle
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, United States
| | - Michael G. Sovic
- Center for Applied Plant Sciences, The Ohio State University, Columbus, OH, United States
| | - Brian M.M. Ahmer
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, United States
| | - Ahmed E. Yousef
- Department of Food Science and Technology, The Ohio State University, Columbus, OH, United States
- Department of Microbiology, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
11
|
Wang H, Xie Z, Yang F, Wang Y, Jiang H, Huang X, Zhang Y. Salmonella enterica serovar Typhi influences inflammation and autophagy in macrophages. Braz J Microbiol 2022; 53:525-534. [PMID: 35274232 PMCID: PMC9151981 DOI: 10.1007/s42770-022-00719-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 02/01/2022] [Indexed: 02/01/2023] Open
Abstract
Salmonella enterica serovar Typhi (S. Typhi) is a human enteropathogen that can survive in macrophages and cause systemic infection. Autophagy and inflammation are two important immune responses of macrophages that contribute to the elimination of pathogens. However, Salmonella has derived many strategies to evade inflammation and autophagy. This study investigated inflammation-related NF-κB signaling pathways and autophagy in S. Typhi-infected macrophages. RNA-seq and quantitative real-time PCR indicated that mRNA levels of NF-κB signaling pathway and autophagy-related genes were dynamically influenced in S. Typhi-infected macrophages. Western blots revealed that S. Typhi activated the NF-κB signaling pathway and induced the expression of inhibitor protein IκBζ. In addition, S. Typhi enhanced autophagy during early stages of infection and may inhibit autophagy during late stages of infection. Thus, we propose that S. Typhi can influence the NF-κB signaling pathway and autophagy in macrophages.
Collapse
Affiliation(s)
- Huiyun Wang
- JiangYin Hospital Affiliated to Nanjing University of Chinese Medicine, Jiangyin, 214400, Jiangsu, China
| | - Zhongyi Xie
- Department of Biochemistry and Molecular Biology, Jiangsu University School of Medicine, 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, China
| | - Fanfan Yang
- Department of Biochemistry and Molecular Biology, Jiangsu University School of Medicine, 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, China
| | - Yurou Wang
- Department of Biochemistry and Molecular Biology, Jiangsu University School of Medicine, 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, China
| | - Haiqiang Jiang
- JiangYin Hospital Affiliated to Nanjing University of Chinese Medicine, Jiangyin, 214400, Jiangsu, China
| | - Xinxiang Huang
- Department of Biochemistry and Molecular Biology, Jiangsu University School of Medicine, 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, China
| | - Ying Zhang
- Department of Biochemistry and Molecular Biology, Jiangsu University School of Medicine, 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, China.
| |
Collapse
|
12
|
Kirthika P, Jawalagatti V, Senevirathne A, Lee JH. Coordinated interaction between Lon protease and catalase-peroxidase regulates virulence and oxidative stress management during Salmonellosis. Gut Microbes 2022; 14:2064705. [PMID: 35438052 PMCID: PMC9037549 DOI: 10.1080/19490976.2022.2064705] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
This study investigates the interplay between Lon protease and catalase-peroxidase (KatG) in relation to virulence modulation and the response to oxidative stress in Salmonella Typhimurium (ST). Proteomic comparison of ST wild-type and lon deletion mutant led to the recognition of a highly expressed KatG protein product among five other protein candidates that were significantly affected by lon deletion. By employing a bacterium two-hybrid assay (B2H), we demonstrated that the catalytic domain of Lon protease potentially interacts with the KatG protein that leads to proteolytic cleavage. Assessment of virulence gene expression in single and double lon and katG mutants revealed katG to be a potential positive modulator of both Salmonella pathogenicity Island-1 (SPI-1) and -2, while lon significantly affected SPI-1 genes. ST double deletion mutant, ∆lon∆katG was more susceptible to survival defects within macrophage-like cells and exhibited meager colonization of the mouse spleen compared to the single deletion mutants. The findings reveal a previously unknown function of Lon and KatG interaction in Salmonella virulence. Taken together, our experiments demonstrate the importance of Lon and KatG to cope with oxidative stress, for intracellular survival and in vivo virulence of Salmonella.
Collapse
Affiliation(s)
- Perumalraja Kirthika
- Department of Public Health, College of Veterinary Medicine, Jeonbuk National University, Republic of Korea,Biochemistry & Molecular Biology Department, Mayo Clinic, Rochester, Minnesota, USA 55905
| | - Vijayakumar Jawalagatti
- Department of Public Health, College of Veterinary Medicine, Jeonbuk National University, Republic of Korea,Urology Department, Mayo Clinic, Rochester, Minnesota, USA 55905
| | - Amal Senevirathne
- Department of Public Health, College of Veterinary Medicine, Jeonbuk National University, Republic of Korea
| | - John Hwa Lee
- Department of Public Health, College of Veterinary Medicine, Jeonbuk National University, Republic of Korea,CONTACT John Hwa Lee College of Veterinary Medicine, Jeonbuk National University,Iksan campus, 54595, South Korea
| |
Collapse
|
13
|
Jiang L, Yang W, Jiang X, Yao T, Wang L, Yang B. Virulence-related O islands in enterohemorrhagic Escherichia coli O157:H7. Gut Microbes 2022; 13:1992237. [PMID: 34711138 PMCID: PMC8565820 DOI: 10.1080/19490976.2021.1992237] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Enterohemorrhagic Escherichia coli (EHEC) O157:H7 is a principally foodborne pathogen linked to serious diseases, including bloody diarrhea, hemorrhagic colitis, and hemolytic uremic syndrome. Comparative genomics analysis revealed that EHEC O157 contains 177 unique genomic islands, termed O islands, compared with the nonpathogenic E. coli K-12 laboratory strain. These O islands contribute largely to the pathogenicity of EHEC O157:H7 by providing numerous virulence factors, effectors, virulence regulatory proteins, and virulence regulatory sRNAs. The present review aimed to provide a comprehensive understanding of the research progress on the function of O islands, especially focusing on virulence-related O islands.
Collapse
Affiliation(s)
- Lingyan Jiang
- TEDA Institute of Biological Sciences and Biotechnology, Nankai University, TEDA, Tianjin, P. R. China
| | - Wen Yang
- TEDA Institute of Biological Sciences and Biotechnology, Nankai University, TEDA, Tianjin, P. R. China
| | - Xinlei Jiang
- School of Environmental and Municipal Engineering, Tianjin Chengjian University, Tianjin, P. R. China
| | - Ting Yao
- TEDA Institute of Biological Sciences and Biotechnology, Nankai University, TEDA, Tianjin, P. R. China
| | - Lu Wang
- TEDA Institute of Biological Sciences and Biotechnology, Nankai University, TEDA, Tianjin, P. R. China
| | - Bin Yang
- TEDA Institute of Biological Sciences and Biotechnology, Nankai University, TEDA, Tianjin, P. R. China,CONTACT Bin Yang TEDA Institute of Biological Sciences and Biotechnology, Nankai University, TEDA, Tianjin300457, P. R. China
| |
Collapse
|
14
|
Cohn AR, Cheng RA, Orsi RH, Wiedmann M. Moving Past Species Classifications for Risk-Based Approaches to Food Safety: Salmonella as a Case Study. FRONTIERS IN SUSTAINABLE FOOD SYSTEMS 2021. [DOI: 10.3389/fsufs.2021.652132] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
15
|
Mendoza-Mejía BD, Medina-Aparicio L, Serrano-Fujarte I, Vázquez A, Calva E, Hernández-Lucas I. Salmonella enterica serovar Typhi genomic regions involved in low pH resistance and in invasion and replication in human macrophages. ANN MICROBIOL 2021. [DOI: 10.1186/s13213-021-01629-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Purpose
Salmonella enterica serovar Typhi, the etiological agent of typhoid fever, causes a systemic life-threatening disease. To carry out a successful infection process, this bacterium needs to survive alkaline and acid pH conditions presented in the mouth, stomach, small intestine, and gallbladder. Therefore, in this work, a genetic screening to identify S. Typhi genes involved in acid and circumneutral pH resistance was performed.
Methods
A collection of S. Typhi mutants deleted of fragments ranging from 6 to 80 kb were obtained by the Datsenko and Wanner method. Bacterial growth rate assays of each mutant were performed to identify S. Typhi genes involved in circumneutral and acid pH resistance. S. Typhi mutants deficient to growth at specific pH were evaluated in their capacity to invade and replicate in phagocytic cells.
Results
In this work, it is reported that S. Typhi ∆F4 (pH 4.5), S. Typhi ∆F44 (pH 4.5, 5.5, and 6.5), and S. Typhi ∆F73 (pH 4.5, 5.5, 6.5, and 7.5) were deficient to grow in the pH indicated. These three mutant strains were also affected in their ability to invade and replicate in human macrophages.
Conclusions
S. Typhi contains defined genomic regions that influence the survival at specific pH values, as well as the invasion and replication inside human cells. Thus, this genetic information probably allows the bacteria to survive in different human compartments for an efficient infection cycle.
Collapse
|
16
|
Piccini G, Montomoli E. Pathogenic signature of invasive non-typhoidal Salmonella in Africa: implications for vaccine development. Hum Vaccin Immunother 2020; 16:2056-2071. [PMID: 32692622 PMCID: PMC7553687 DOI: 10.1080/21645515.2020.1785791] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
Invasive non-typhoidal Salmonella (iNTS) infections are a leading cause of bacteremia in Sub-Saharan Africa (sSA), thereby representing a major public health threat. Salmonella Typhimurium clade ST313 and Salmonella Enteriditis lineages associated with Western and Central/Eastern Africa are among the iNTS serovars which are of the greatest concern due to their case-fatality rate, especially in children and in the immunocompromised population. Identification of pathogen-associated features and host susceptibility factors that increase the risk for invasive non-typhoidal salmonellosis would be instrumental for the design of targeted prevention strategies, which are urgently needed given the increasing spread of multidrug-resistant iNTS in Africa. This review summarizes current knowledge of bacterial traits and host immune responses associated with iNTS infections in sSA, then discusses how this knowledge can guide vaccine development while providing a summary of vaccine candidates in preclinical and early clinical development.
Collapse
Affiliation(s)
| | - Emanuele Montomoli
- VisMederi srl , Siena, Italy.,Department of Molecular and Developmental Medicine, University of Siena , Siena, Italy
| |
Collapse
|
17
|
Abstract
Reverse-transcription quantitative real-time polymerase chain reaction (RT-qPCR) using fluorescent DNA-binding dyes is now a gold-standard methodology to study bacterial gene expression through relative quantitation of target mRNAs under specific experimental conditions, and recent developments in the technology allow for gene expression analysis in single cells. Nevertheless, several critical steps of the RT-qPCR protocol need to be carefully addressed in order to obtain reliable results, particularly regarding RNA sample quality and appropriate choice of reference genes. Besides, accurate reporting of study conditions is essential, as recommended by the MIQE guidelines. Herein, we provide a practical approach to quantitation of the transcript levels of bacterial genes using RT-qPCR, including a general protocol for obtaining good-quality bacterial RNA and a discussion on the selection and validation of candidate bacterial reference genes for data normalization.
Collapse
|
18
|
Cao L, Wang J, Sun L, Kong Z, Wu Q, Wang Z. Transcriptional analysis reveals the relativity of acid tolerance and antimicrobial peptide resistance of Salmonella. Microb Pathog 2019; 136:103701. [PMID: 31472260 DOI: 10.1016/j.micpath.2019.103701] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 08/27/2019] [Accepted: 08/28/2019] [Indexed: 01/20/2023]
Abstract
The objective of this study was to comprehensively identify the target genes induced by acid stimulation in Salmonella, and to clarify the relativity of acid tolerance and antimicrobial peptide resistance. A clinical S. Typhimurium strain, S6, was selected and performed a transcriptome analysis under the acid tolerance response. In total, we found 1461 genes to be differentially expressed, including 721 up-regulated and 740 down-regulated genes. Functional annotation revealed differentially expressed genes to be associated with regulation, metabolism, transport, virulence, and motility. Interestingly, KEGG pathway analysis demonstrated that the induced genes by acid were enriched in cationic antimicrobial peptide resistance, sulfur relay system, ABC transporters, and two-component system pathway. Therein, PhoQ belonging to the two-component system PhoP-PhoQ that promotes virulence by detecting the macrophage phagosome and controls the transcript levels of many genes associated with the resistance to AMPs; MarA, a multiple antibiotic resistance factor; SapA, one of the encoding gene of sapABCDF operon that confers resistance to small cationic peptides of Salmonella; YejB, one of the encoding gene of yejABEF operon that confers resistance to antimicrobial peptides and contributes to the virulence of Salmonella, were all induced by acid stimulation, and could potentially explain that there is a correlation between acid tolerance and AMPs resistance, and finally affects the virulence of intracellular pathogenic bacteria.
Collapse
Affiliation(s)
- Li Cao
- Beijing Key Laboratory of Traditional Chinese Veterinary Medicine, Animal Science and Technology College, Beijing University of Agriculture, Beijing, 102206, China
| | - Jiawei Wang
- Beijing Key Laboratory of Traditional Chinese Veterinary Medicine, Animal Science and Technology College, Beijing University of Agriculture, Beijing, 102206, China
| | - Lu Sun
- Beijing Key Laboratory of Traditional Chinese Veterinary Medicine, Animal Science and Technology College, Beijing University of Agriculture, Beijing, 102206, China
| | - Zian Kong
- Beijing Key Laboratory of Traditional Chinese Veterinary Medicine, Animal Science and Technology College, Beijing University of Agriculture, Beijing, 102206, China
| | - Qingmin Wu
- Key Laboratory of Animal Epidemiology and Zoonosis of the Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Zhen Wang
- Beijing Key Laboratory of Traditional Chinese Veterinary Medicine, Animal Science and Technology College, Beijing University of Agriculture, Beijing, 102206, China.
| |
Collapse
|
19
|
Karlinsey JE, Stepien TA, Mayho M, Singletary LA, Bingham-Ramos LK, Brehm MA, Greiner DL, Shultz LD, Gallagher LA, Bawn M, Kingsley RA, Libby SJ, Fang FC. Genome-wide Analysis of Salmonella enterica serovar Typhi in Humanized Mice Reveals Key Virulence Features. Cell Host Microbe 2019; 26:426-434.e6. [PMID: 31447308 DOI: 10.1016/j.chom.2019.08.001] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 07/03/2019] [Accepted: 07/31/2019] [Indexed: 11/29/2022]
Abstract
Salmonella enterica serovar Typhi causes typhoid fever only in humans. Murine infection with S. Typhimurium is used as a typhoid model, but its relevance to human typhoid is limited. Non-obese diabetic-scid IL2rγnull mice engrafted with human hematopoietic stem cells (hu-SRC-SCID) are susceptible to lethal S. Typhi infection. In this study, we use a high-density S. Typhi transposon library in hu-SRC-SCID mice to identify virulence loci using transposon-directed insertion site sequencing (TraDIS). Vi capsule, lipopolysaccharide (LPS), and aromatic amino acid biosynthesis were essential for virulence, along with the siderophore salmochelin. However, in contrast to the murine S. Typhimurium model, neither the PhoPQ two-component system nor the SPI-2 pathogenicity island was required for lethal S. Typhi infection, nor was the CdtB typhoid toxin. These observations highlight major differences in the pathogenesis of typhoid and non-typhoidal Salmonella infections and demonstrate the utility of humanized mice for understanding the pathogenesis of a human-specific pathogen.
Collapse
Affiliation(s)
- Joyce E Karlinsey
- Department of Microbiology, University of Washington, Seattle, WA 98195, USA
| | - Taylor A Stepien
- Department of Global Health, University of Washington, Seattle, WA 98195, USA
| | | | | | | | - Michael A Brehm
- Program in Molecular Medicine and the Diabetes Center of Excellence, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Dale L Greiner
- Program in Molecular Medicine and the Diabetes Center of Excellence, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | | | - Larry A Gallagher
- Department of Genome Sciences, School of Medicine, University of Washington, Seattle, WA 98195, USA
| | - Matt Bawn
- Quadram Institute Bioscience, Norwich, UK; Earlham Institute, Norwich, UK
| | - Robert A Kingsley
- Quadram Institute Bioscience, Norwich, UK; School of Biological Science, University of East Anglia, Norwich, UK
| | - Stephen J Libby
- Department of Laboratory Medicine, University of Washington, Seattle, WA 98195, USA
| | - Ferric C Fang
- Department of Microbiology, University of Washington, Seattle, WA 98195, USA; Department of Global Health, University of Washington, Seattle, WA 98195, USA; Department of Laboratory Medicine, University of Washington, Seattle, WA 98195, USA.
| |
Collapse
|
20
|
Xu J, Preciado-Llanes L, Aulicino A, Decker CM, Depke M, Gesell Salazar M, Schmidt F, Simmons A, Huang WE. Single-Cell and Time-Resolved Profiling of Intracellular Salmonella Metabolism in Primary Human Cells. Anal Chem 2019; 91:7729-7737. [PMID: 31117406 PMCID: PMC7006958 DOI: 10.1021/acs.analchem.9b01010] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
![]()
The
intracellular pathogen Salmonella enterica has evolved
an array of traits for propagation and invasion of the
intestinal layers. It remains largely elusive how Salmonella adjusts its metabolic states to survive inside immune host cells.
In this study, single-cell Raman biotechnology combined with deuterium
isotope probing (Raman-DIP) have been applied to reveal metabolic
changes of the typhoidal Salmonella Typhi Ty2, the
nontyphoidal Salmonella Typhimurium LT2, and a clinical
isolate Typhimurium D23580. By initially labeling the Salmonella strains with deuterium, we employed reverse labeling to track their
metabolic changes in the time-course infection of THP-1 cell line,
human monocyte-derived dendritic cells (MoDCs) and macrophages (Mf).
We found that, in comparison with a noninvasive serovar, the invasive Salmonella strains Ty2 and D23580 have downregulated metabolic
activity inside human macrophages and dendritic cells and used lipids
as alternative carbon source, perhaps a strategy to escape from the
host immune response. Proteomic analysis using high sensitivity mass
spectrometry validated the findings of Raman-DIP analysis.
Collapse
Affiliation(s)
- Jiabao Xu
- Department of Engineering Science , University of Oxford , Parks Road , Oxford OX1 3PJ , United Kingdom
| | - Lorena Preciado-Llanes
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine , University of Oxford , Oxford OX3 9DS , United Kingdom.,Translational Gastroenterology Unit, John Radcliffe Hospital , Headington, Oxford OX3 9DU , United Kingdom
| | - Anna Aulicino
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine , University of Oxford , Oxford OX3 9DS , United Kingdom.,Translational Gastroenterology Unit, John Radcliffe Hospital , Headington, Oxford OX3 9DU , United Kingdom
| | - Christoph Martin Decker
- Interfaculty Institute for Genetics and Functional Genomics , University Medicine Greifswald , Felix-Hausdorff-Str. 8 , 17475 Greifswald , Germany
| | - Maren Depke
- Interfaculty Institute for Genetics and Functional Genomics , University Medicine Greifswald , Felix-Hausdorff-Str. 8 , 17475 Greifswald , Germany
| | - Manuela Gesell Salazar
- Interfaculty Institute for Genetics and Functional Genomics , University Medicine Greifswald , Felix-Hausdorff-Str. 8 , 17475 Greifswald , Germany
| | - Frank Schmidt
- Interfaculty Institute for Genetics and Functional Genomics , University Medicine Greifswald , Felix-Hausdorff-Str. 8 , 17475 Greifswald , Germany.,Proteomics Core, Weill Cornel Medicine-Qatar , Education City , PO 24144 Doha , Qatar
| | - Alison Simmons
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine , University of Oxford , Oxford OX3 9DS , United Kingdom.,Translational Gastroenterology Unit, John Radcliffe Hospital , Headington, Oxford OX3 9DU , United Kingdom
| | - Wei E Huang
- Department of Engineering Science , University of Oxford , Parks Road , Oxford OX1 3PJ , United Kingdom
| |
Collapse
|
21
|
Salmonella Proteomic Profiling during Infection Distinguishes the Intracellular Environment of Host Cells. mSystems 2019; 4:mSystems00314-18. [PMID: 30984873 PMCID: PMC6456673 DOI: 10.1128/msystems.00314-18] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 03/14/2019] [Indexed: 02/06/2023] Open
Abstract
Salmonella Typhimurium is one of the leading causes of foodborne bacterial infection. Nevertheless, how Salmonella adapts to distinct types of host cells during infection remains poorly understood. By contrasting intracellular Salmonella proteomes from both infected macrophages and epithelial cells, we found striking proteomic signatures specific to particular types of host cells. Notably, Salmonella proteomic remodeling exhibited quicker kinetics in macrophages than in epithelial cells with respect to bacterial virulence and flagellar and chemotaxis systems. Furthermore, we unveiled high levels of induction of bacterial histidine biosynthesis in macrophages but not in epithelial cells, which is attributable to differing intracellular levels of this amino acid. Intriguingly, we found that a defective hisG gene renders a Salmonella strain hypersensitive to histidine shortage in macrophages. Overall, our work reveals specific Salmonella adaptation mechanisms in distinct host cells, which should aid in the development of novel anti-infection strategies. Essential to bacterial pathogenesis, Salmonella enterica serovar Typhimurium (S. Typhimurium) has evolved the capacity to quickly sense and adapt to specific intracellular environment within distinct host cells. Here we examined S. Typhimurium proteomic remodeling within macrophages, allowing direct comparison with our previous studies in epithelial cells. In addition to many shared features, our data revealed proteomic signatures highly specific to one type of host cells. Notably, intracellular S. Typhimurium differentially regulates the two type III secretion systems (T3SSs) far more quickly in macrophages than in epithelial cells; bacterial flagellar and chemotaxis systems degenerate more quickly in macrophages than in HeLa cells as well. Importantly, our comparative analysis uncovered high levels of induction of bacterial histidine biosynthesis in macrophages but not in epithelial cells. Targeted metabolomic measurements revealed markedly lower histidine levels within macrophages. Intriguingly, further functional studies established that histidine biosynthesis that is defective (due to a hisG mutation) renders the bacterium (strain SL1344) hypersensitive to intracellular shortage of this amino acid. Indeed, another S. Typhimurium strain, namely, strain 14028s, with a fully functional biosynthetic pathway exhibited only minor induction of the his operon within infected macrophages. Our work thus provided novel insights into S. Typhimurium adaptation mechanisms within distinct host cells and also provided an elegant paradigm where proteomic profiling of intracellular pathogens is utilized to discriminate specific host environments (e.g., on the basis of nutrient availability). IMPORTANCESalmonella Typhimurium is one of the leading causes of foodborne bacterial infection. Nevertheless, how Salmonella adapts to distinct types of host cells during infection remains poorly understood. By contrasting intracellular Salmonella proteomes from both infected macrophages and epithelial cells, we found striking proteomic signatures specific to particular types of host cells. Notably, Salmonella proteomic remodeling exhibited quicker kinetics in macrophages than in epithelial cells with respect to bacterial virulence and flagellar and chemotaxis systems. Furthermore, we unveiled high levels of induction of bacterial histidine biosynthesis in macrophages but not in epithelial cells, which is attributable to differing intracellular levels of this amino acid. Intriguingly, we found that a defective hisG gene renders a Salmonella strain hypersensitive to histidine shortage in macrophages. Overall, our work reveals specific Salmonella adaptation mechanisms in distinct host cells, which should aid in the development of novel anti-infection strategies.
Collapse
|
22
|
Liaquat S, Sarwar Y, Ali A, Haque A, Farooq M, Martinez-Ballesteros I, Laorden L, Garaizar J, Bikandi J. Virulotyping of Salmonella enterica serovar Typhi isolates from Pakistan: Absence of complete SPI-10 in Vi negative isolates. PLoS Negl Trop Dis 2018; 12:e0006839. [PMID: 30500817 PMCID: PMC6267989 DOI: 10.1371/journal.pntd.0006839] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Accepted: 09/13/2018] [Indexed: 12/30/2022] Open
Abstract
The pathogenesis of Salmonella enterica serovar Typhi (S. Typhi), the cause of typhoid fever in humans, is mainly attributed to the acquisition of horizontally acquired DNA elements. Salmonella pathogenicity islands (SPIs) are indubitably the most important form of horizontally acquired DNA with respect to pathogenesis of this bacterium. The insertion or deletion of any of these transferrable SPIs may have impact on the virulence potential of S. Typhi. In this study, the virulence potential and genetic relatedness of 35 S. Typhi isolates, collected from 2004 to 2013 was determined by identification of SPI and non-SPI virulence factors through a combination of techniques including virulotyping, Whole Genome Sequencing (WGS), and Variable Number of Tandem Repeats (VNTR) profiling. In order to determine the virulence potential of local S. Typhi isolates, 56 virulence related genes were studied by PCR. These genes are located in the core as well as accessory genome (SPIs and plasmid). Major variations among studied virulence determinants were found in case of SPI-7 and SPI-10 associated genes. On the basis of presence of virulence related genes, the studied S. Typhi isolates from Pakistan were clustered into two virulotypes Vi-positive and Vi-negative. Interestingly, SPI-7 and SPI-10 were collectively absent or present in Vi-negative and Vi-positive strains, respectively. Two Vi-negative and 11 Vi-positive S. Typhi strains were also analyzed by whole genome sequencing (WGS) and their results supported the PCR results. Genetic diversity was tested by VNTR-based molecular typing. All 35 isolates were clustered into five groups. Overall, all Vi-negative isolates were placed in a single group (T5) whereas Vi-positive isolates were grouped into four types. Vi-negative and Vi-positive isolates were mutually exclusive. This is the first report on the comparative distribution of SPI and non-SPI related virulence genes in Vi-negative and Vi-positive S. Typhi isolates with an important finding that SPI-10 is absent in all Vi-negative isolates. The distribution of virulence factors in S. Typhi can vary in isolates from different geographical regions and can have significant effect on the disease control. In this study, we have checked the distribution of 56 reported virulence associated factors in 35 local isolates of S. Typhi to identify any variations that can help in designing effective control strategies for typhoid. We have identified four naturally occurring variants which are simultaneously lacking SPI-7 and SPI-10, two adjacently located pathogenicity islands on S. Typhi chromosome. These isolates are not producing Vi capsular antigen hence the Vi based vaccines will not be effective against them. These findings highlight the need to develop typhoid vaccines specifically effective in Pakistan.
Collapse
Affiliation(s)
- Sadia Liaquat
- Enteric Pathogen Laboratory, Health Biotechnology Division, National Institute for Biotechnology and Genetic Engineering (NIBGE), Faisalabad, Pakistan affiliated with Pakistan Institute of Engineering and Applied Sciences (PIEAS), Islamabad, Pakistan
- Departments of Bioinformatics and Biotechnology, Government College University Faisalabad, Faisalabad, Pakistan
| | - Yasra Sarwar
- Enteric Pathogen Laboratory, Health Biotechnology Division, National Institute for Biotechnology and Genetic Engineering (NIBGE), Faisalabad, Pakistan affiliated with Pakistan Institute of Engineering and Applied Sciences (PIEAS), Islamabad, Pakistan
- * E-mail:
| | - Aamir Ali
- Enteric Pathogen Laboratory, Health Biotechnology Division, National Institute for Biotechnology and Genetic Engineering (NIBGE), Faisalabad, Pakistan affiliated with Pakistan Institute of Engineering and Applied Sciences (PIEAS), Islamabad, Pakistan
| | - Abdul Haque
- Akhuwat Faisalabad Institute For Research In Science And Technology, Faisalabad, Pakistan
| | - Muhammad Farooq
- Enteric Pathogen Laboratory, Health Biotechnology Division, National Institute for Biotechnology and Genetic Engineering (NIBGE), Faisalabad, Pakistan affiliated with Pakistan Institute of Engineering and Applied Sciences (PIEAS), Islamabad, Pakistan
| | - Ilargi Martinez-Ballesteros
- Department of Immunology, Microbiology and Parasitology, University of the Basque Country, (UPV/EHU), Vitoria-Gasteiz, Spain
| | - Lorena Laorden
- Department of Immunology, Microbiology and Parasitology, University of the Basque Country, (UPV/EHU), Vitoria-Gasteiz, Spain
| | - Javier Garaizar
- Department of Immunology, Microbiology and Parasitology, University of the Basque Country, (UPV/EHU), Vitoria-Gasteiz, Spain
| | - Joseba Bikandi
- Department of Immunology, Microbiology and Parasitology, University of the Basque Country, (UPV/EHU), Vitoria-Gasteiz, Spain
| |
Collapse
|
23
|
Matilla MA, Krell T. The effect of bacterial chemotaxis on host infection and pathogenicity. FEMS Microbiol Rev 2018; 42:4563582. [PMID: 29069367 DOI: 10.1093/femsre/fux052] [Citation(s) in RCA: 177] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Accepted: 10/19/2017] [Indexed: 12/26/2022] Open
Abstract
Chemotaxis enables microorganisms to move according to chemical gradients. Although this process requires substantial cellular energy, it also affords key physiological benefits, including enhanced access to growth substrates. Another important implication of chemotaxis is that it also plays an important role in infection and disease, as chemotaxis signalling pathways are broadly distributed across a variety of pathogenic bacteria. Furthermore, current research indicates that chemotaxis is essential for the initial stages of infection in different human, animal and plant pathogens. This review focuses on recent findings that have identified specific bacterial chemoreceptors and corresponding chemoeffectors associated with pathogenicity. Pathogenicity-related chemoeffectors are either host and niche-specific signals or intermediates of the host general metabolism. Plant pathogens were found to contain an elevated number of chemotaxis signalling genes and functional studies demonstrate that these genes are critical for their ability to enter the host. The expanding body of knowledge of the mechanisms underlying chemotaxis in pathogens provides a foundation for the development of new therapeutic strategies capable of blocking infection and preventing disease by interfering with chemotactic signalling pathways.
Collapse
Affiliation(s)
- Miguel A Matilla
- Department of Environmental Protection, Estación Experimental del Zaidín, Consejo Superior de Investigaciones Científicas, 18008 Granada, Spain
| | - Tino Krell
- Department of Environmental Protection, Estación Experimental del Zaidín, Consejo Superior de Investigaciones Científicas, 18008 Granada, Spain
| |
Collapse
|
24
|
Dufresne K, Saulnier-Bellemare J, Daigle F. Functional Analysis of the Chaperone-Usher Fimbrial Gene Clusters of Salmonella enterica serovar Typhi. Front Cell Infect Microbiol 2018; 8:26. [PMID: 29473020 PMCID: PMC5809473 DOI: 10.3389/fcimb.2018.00026] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Accepted: 01/19/2018] [Indexed: 01/11/2023] Open
Abstract
The human-specific pathogen Salmonella enterica serovar Typhi causes typhoid, a major public health issue in developing countries. Several aspects of its pathogenesis are still poorly understood. S. Typhi possesses 14 fimbrial gene clusters including 12 chaperone-usher fimbriae (stg, sth, bcf, fim, saf, sef, sta, stb, stc, std, ste, and tcf). These fimbriae are weakly expressed in laboratory conditions and only a few are actually characterized. In this study, expression of all S. Typhi chaperone-usher fimbriae and their potential roles in pathogenesis such as interaction with host cells, motility, or biofilm formation were assessed. All S. Typhi fimbriae were better expressed in minimal broth. Each system was overexpressed and only the fimbrial gene clusters without pseudogenes demonstrated a putative major subunits of about 17 kDa on SDS-PAGE. Six of these (Fim, Saf, Sta, Stb, Std, and Tcf) also show extracellular structure by electron microscopy. The impact of fimbrial deletion in a wild-type strain or addition of each individual fimbrial system to an S. Typhi afimbrial strain were tested for interactions with host cells, biofilm formation and motility. Several fimbriae modified bacterial interactions with human cells (THP-1 and INT-407) and biofilm formation. However, only Fim fimbriae had a deleterious effect on motility when overexpressed. Overall, chaperone-usher fimbriae seem to be an important part of the balance between the different steps (motility, adhesion, host invasion and persistence) of S. Typhi pathogenesis.
Collapse
Affiliation(s)
- Karine Dufresne
- Department of Microbiology, Infectiology and Immunology, Université de Montréal, Montreal, QC, Canada
| | - Julie Saulnier-Bellemare
- Department of Microbiology, Infectiology and Immunology, Université de Montréal, Montreal, QC, Canada
| | - France Daigle
- Department of Microbiology, Infectiology and Immunology, Université de Montréal, Montreal, QC, Canada
| |
Collapse
|
25
|
Bi-valent polysaccharides of Vi capsular and O9 O-antigen in attenuated Salmonella Typhimurium induce strong immune responses against these two antigens. NPJ Vaccines 2018; 3:1. [PMID: 29354293 PMCID: PMC5760606 DOI: 10.1038/s41541-017-0041-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Revised: 12/01/2017] [Accepted: 12/08/2017] [Indexed: 11/17/2022] Open
Abstract
Salmonella Typhi is the causative agent of typhoid fever in humans, responsible for approximately 21 million infections and 222,000 deaths globally each year. The current licensed vaccines provide moderate protection to recipients aged >2 years. Prior work on typhoid vaccines has focused on injectable Vi capsular polysaccharide or Vi–protein conjugates and live, oral attenuated S. Typhi vaccines to induce humoral anti-Vi antibodies, while the value and importance of anti-O9 antibodies is less well established. In this study, we constructed a S. Typhimurium strain that synthesizes Vi capsular antigen in vivo and produces the immunodominant O9-antigen polysaccharide instead of its native O4-antigen. The live recombinant attenuated S. Typhimurium mutants were effective in stimulating anti-Vi and anti-O9 antibodies in a mouse model, and the surface Vi capsular expression did not affect the immune responses against the O9 O-antigen polysaccharide. Moreover, the resulting anti-Vi and anti-O9 antibodies were effective at killing S. Typhi and other Salmonella spp. expressing Vi or O9 antigen polysaccharides and provided efficient protection against lethal challenge by S. Typhimurium and S. Enteritidis. Our work highlights the strategy of developing live attenuated S. Typhimurium vaccines to prevent typhoid fever by targeting the both Vi capsular and O9 O-polysaccharide antigens simultaneously. An attenuated strain of modified Salmonella Typhimurium bacteria could answer calls for a more effective typhoid fever vaccine. Current vaccines against typhoid-causing Salmonella Typhi are only moderately effective and potentially ineffective in children under 5 years. Qingke Kong and Roy Curtiss, leading a team of US and Chinese researchers, developed an attenuated version of the less-pathogenic S. Typhimurium that, when orally dosed in mice, expresses bacterial sugar-chain molecules known to elicit a strong immune response. In an in vitro assay, the antibodies produced by the mice in response to these molecules killed S. Typhi and related Salmonella bacteria with similar surface molecules, indicating a potential cross-protective ability. Further research would reveal whether this two-pronged live vaccine has the potential to protect in vivo, in live animals and in humans.
Collapse
|
26
|
Bent ZW, Poorey K, LaBauve AE, Hamblin R, Williams KP, Meagher RJ. A Rapid Spin Column-Based Method to Enrich Pathogen Transcripts from Eukaryotic Host Cells Prior to Sequencing. PLoS One 2016; 11:e0168788. [PMID: 28002481 PMCID: PMC5176299 DOI: 10.1371/journal.pone.0168788] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Accepted: 12/06/2016] [Indexed: 02/04/2023] Open
Abstract
When analyzing pathogen transcriptomes during the infection of host cells, the signal-to-background (pathogen-to-host) ratio of nucleic acids (NA) in infected samples is very small. Despite the advancements in next-generation sequencing, the minute amount of pathogen NA makes standard RNA-seq library preps inadequate for effective gene-level analysis of the pathogen in cases with low bacterial loads. In order to provide a more complete picture of the pathogen transcriptome during an infection, we developed a novel pathogen enrichment technique, which can enrich for transcripts from any cultivable bacteria or virus, using common, readily available laboratory equipment and reagents. To evenly enrich for pathogen transcripts, we generate biotinylated pathogen-targeted capture probes in an enzymatic process using the entire genome of the pathogen as a template. The capture probes are hybridized to a strand-specific cDNA library generated from an RNA sample. The biotinylated probes are captured on a monomeric avidin resin in a miniature spin column, and enriched pathogen-specific cDNA is eluted following a series of washes. To test this method, we performed an in vitro time-course infection using Klebsiella pneumoniae to infect murine macrophage cells. K. pneumoniae transcript enrichment efficiency was evaluated using RNA-seq. Bacterial transcripts were enriched up to ~400-fold, and allowed the recovery of transcripts from ~2000–3600 genes not observed in untreated control samples. These additional transcripts revealed interesting aspects of K. pneumoniae biology including the expression of putative virulence factors and the expression of several genes responsible for antibiotic resistance even in the absence of drugs.
Collapse
Affiliation(s)
- Zachary W. Bent
- Systems Biology Department, Sandia National Laboratories, Livermore, California, United States of America
- * E-mail: (ZWB); (RJM)
| | - Kunal Poorey
- Systems Biology Department, Sandia National Laboratories, Livermore, California, United States of America
| | - Annette E. LaBauve
- Biotechnology and Bioengineering Department, Sandia National Laboratories, Livermore, California, United States of America
| | - Rachelle Hamblin
- Systems Biology Department, Sandia National Laboratories, Livermore, California, United States of America
| | - Kelly P. Williams
- Systems Biology Department, Sandia National Laboratories, Livermore, California, United States of America
| | - Robert J. Meagher
- Biotechnology and Bioengineering Department, Sandia National Laboratories, Livermore, California, United States of America
- * E-mail: (ZWB); (RJM)
| |
Collapse
|
27
|
Wang Z, Bie P, Cheng J, Lu L, Cui B, Wu Q. The ABC transporter YejABEF is required for resistance to antimicrobial peptides and the virulence of Brucella melitensis. Sci Rep 2016; 6:31876. [PMID: 27550726 PMCID: PMC4994006 DOI: 10.1038/srep31876] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Accepted: 07/28/2016] [Indexed: 02/06/2023] Open
Abstract
The ability to resist the killing effects of host antimicrobial peptides (AMPs) plays a vital role in the virulence of pathogens. The Brucella melitensis NI genome has a gene cluster that encodes ABC transport. In this study, we constructed yejA1, yejA2, yejB, yejE, yejF, and whole yej operon deletion mutants, none of which exhibited discernible growth defect in TSB or minimal medium. Unlike their parental strain, the mutants showed a significantly increased sensitivity to acidic stress. The NIΔyejE and NIΔyejABEF mutants were also more sensitive than B. melitensis NI to polymyxin B, and the expression of yej operon genes was induced by polymyxin B. Moreover, cell and mouse infection assays indicated that NIΔyejE and NIΔyejABEF have restricted invasion and replication abilities inside macrophages and are rapidly cleared from the spleens of infected mice. These findings indicate that the ABC transporter YejABEF is required for the virulence of Brucella, suggesting that resistance to host antimicrobials is a key mechanism for Brucella to persistently survive in vivo. This study provided insights that led us to further investigate the potential correlation of AMP resistance with the mechanisms of immune escape and persistent infection by pathogens.
Collapse
Affiliation(s)
- Zhen Wang
- Beijing Key Laboratory of Traditional Chinese Veterinary Medicine, Animal Science and Technology College, Beijing University of Agriculture, Beijing 102206, China
| | - Pengfei Bie
- Key Laboratory of Animal Epidemiology and Zoonosis of the Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Jie Cheng
- Beijing Key Laboratory of Traditional Chinese Veterinary Medicine, Animal Science and Technology College, Beijing University of Agriculture, Beijing 102206, China
| | - Lin Lu
- Beijing Key Laboratory of Traditional Chinese Veterinary Medicine, Animal Science and Technology College, Beijing University of Agriculture, Beijing 102206, China
| | - Buyun Cui
- State Key Laboratory for Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Qingmin Wu
- Key Laboratory of Animal Epidemiology and Zoonosis of the Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| |
Collapse
|
28
|
Velásquez JC, Hidalgo AA, Villagra N, Santiviago CA, Mora GC, Fuentes JA. SPI-9 of Salmonella enterica serovar Typhi is constituted by an operon positively regulated by RpoS and contributes to adherence to epithelial cells in culture. Microbiology (Reading) 2016; 162:1367-1378. [DOI: 10.1099/mic.0.000319] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Affiliation(s)
- Juan C. Velásquez
- Laboratorio de Genética y Patogénesis Bacteriana, Departamento de Ciencias Biológicas, Facultad de Ciencias Biológicas, Universidad Andres Bello, República 217, Santiago, Chile
| | - Alejandro A. Hidalgo
- Laboratorio de Patogénesis Molecular y Antimicrobianos, Facultad de Medicina, Universidad Andres Bello, Echaurren 183, Santiago, Chile
| | - Nicolás Villagra
- Laboratorio de Patogénesis Molecular y Antimicrobianos, Facultad de Medicina, Universidad Andres Bello, Echaurren 183, Santiago, Chile
| | - Carlos A. Santiviago
- Laboratorio de Genética y Patogénesis Bacteriana, Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santos Dumont 964, Independencia, Santiago, Chile
| | - Guido C. Mora
- Laboratorio de Patogénesis Molecular y Antimicrobianos, Facultad de Medicina, Universidad Andres Bello, Echaurren 183, Santiago, Chile
| | - Juan A. Fuentes
- Laboratorio de Genética y Patogénesis Bacteriana, Departamento de Ciencias Biológicas, Facultad de Ciencias Biológicas, Universidad Andres Bello, República 217, Santiago, Chile
| |
Collapse
|
29
|
Lamberti Y, Cafiero JH, Surmann K, Valdez H, Holubova J, Večerek B, Sebo P, Schmidt F, Völker U, Rodriguez ME. Proteome analysis of Bordetella pertussis isolated from human macrophages. J Proteomics 2016; 136:55-67. [DOI: 10.1016/j.jprot.2016.02.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Revised: 02/05/2016] [Accepted: 02/07/2016] [Indexed: 12/13/2022]
|
30
|
An R, Grewal PS. Comparative Analysis of Xenorhabdus koppenhoeferi Gene Expression during Symbiotic Persistence in the Host Nematode. PLoS One 2016; 11:e0145739. [PMID: 26745883 PMCID: PMC4706420 DOI: 10.1371/journal.pone.0145739] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Accepted: 12/08/2015] [Indexed: 01/24/2023] Open
Abstract
Species of Xenorhabdus and Photorhabdus bacteria form mutualistic associations with Steinernema and Heterorhabditis nematodes, respectively and serve as model systems for studying microbe-animal symbioses. Here, we profiled gene expression of Xenorhabdus koppenhoeferi during their symbiotic persistence in the newly formed infective juveniles of the host nematode Steinernema scarabaei through the selective capture of transcribed sequences (SCOTS). The obtained gene expression profile was then compared with other nematode-bacteria partnerships represented by Steinernema carpocapsae-Xenorhabdus nematophila and Heterorhabditis bacteriophora-Photorhabdus temperata. A total of 29 distinct genes were identified to be up-regulated and 53 were down-regulated in X. koppenhoeferi while in S. scarabaei infective juveniles. Of the identified genes, 8 of the up-regulated and 14 of the down-regulated genes were similarly expressed in X. nematophila during persistence in its host nematode S. carpocapsae. However, only one from each of these up- and down-regulated genes was common to the mutualistic partnership between the bacterium P. temperata and the nematode H. bacteriophora. Interactive network analysis of the shared genes between X. koppenhoeferi and X. nematophila demonstrated that the up-regulated genes were mainly involved in bacterial survival and the down-regulated genes were more related to bacterial virulence and active growth. Disruption of two selected genes pta (coding phosphotransacetylase) and acnB (coding aconitate hydratase) in X. nematophila with shared expression signature with X. koppenhoeferi confirmed that these genes are important for bacterial persistence in the nematode host. The results of our comparative analyses show that the two Xenorhabdus species share a little more than a quarter of the transcriptional mechanisms during persistence in their nematode hosts but these features are quite different from those used by P. temperata bacteria in their nematode host H. bacteriophora.
Collapse
Affiliation(s)
- Ruisheng An
- Department of Entomology and Plant Pathology, University of Tennessee, 2505 E. J. Chapman Drive, Knoxville, TN, 37996, United States of America
| | - Parwinder S. Grewal
- Department of Entomology and Plant Pathology, University of Tennessee, 2505 E. J. Chapman Drive, Knoxville, TN, 37996, United States of America
| |
Collapse
|
31
|
Ryan D, Pati NB, Ojha UK, Padhi C, Ray S, Jaiswal S, Singh GP, Mannala GK, Schultze T, Chakraborty T, Suar M. Global transcriptome and mutagenic analyses of the acid tolerance response of Salmonella enterica serovar Typhimurium. Appl Environ Microbiol 2015; 81:8054-65. [PMID: 26386064 PMCID: PMC4651094 DOI: 10.1128/aem.02172-15] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Accepted: 09/01/2015] [Indexed: 01/18/2023] Open
Abstract
Salmonella enterica serovar Typhimurium (S. Typhimurium) is one of the leading causative agents of food-borne bacterial gastroenteritis. Swift invasion through the intestinal tract and successful establishment in systemic organs are associated with the adaptability of S. Typhimurium to different stress environments. Low-pH stress serves as one of the first lines of defense in mammalian hosts, which S. Typhimurium must efficiently overcome to establish an infection. Therefore, a better understanding of the molecular mechanisms underlying the adaptability of S. Typhimurium to acid stress is highly relevant. In this study, we have performed a transcriptome analysis of S. Typhimurium under the acid tolerance response (ATR) and found a large number of genes (∼47%) to be differentially expressed (more than 1.5-fold or less than -1.5-fold; P < 0.01). Functional annotation revealed differentially expressed genes to be associated with regulation, metabolism, transport and binding, pathogenesis, and motility. Additionally, our knockout analysis of a subset of differentially regulated genes facilitated the identification of proteins that contribute to S. Typhimurium ATR and virulence. Mutants lacking genes encoding the K(+) binding and transport protein KdpA, hypothetical protein YciG, the flagellar hook cap protein FlgD, and the nitrate reductase subunit NarZ were significantly deficient in their ATRs and displayed varied in vitro virulence characteristics. This study offers greater insight into the transcriptome changes of S. Typhimurium under the ATR and provides a framework for further research on the subject.
Collapse
Affiliation(s)
- Daniel Ryan
- School of Biotechnology, KIIT University, Bhubaneswar, Odisha, India
| | - Niladri Bhusan Pati
- Institute of Medical Microbiology, German Centre of Infection Research, Site Giessen-Marburg-Langen, Justus-Liebig-University Giessen, Giessen, Germany
| | - Urmesh K Ojha
- School of Biotechnology, KIIT University, Bhubaneswar, Odisha, India
| | | | - Shilpa Ray
- School of Biotechnology, KIIT University, Bhubaneswar, Odisha, India
| | - Sangeeta Jaiswal
- School of Biotechnology, KIIT University, Bhubaneswar, Odisha, India
| | - Gajinder P Singh
- School of Biotechnology, KIIT University, Bhubaneswar, Odisha, India
| | - Gopala K Mannala
- Institute of Medical Microbiology, German Centre of Infection Research, Site Giessen-Marburg-Langen, Justus-Liebig-University Giessen, Giessen, Germany
| | - Tilman Schultze
- Institute of Medical Microbiology, German Centre of Infection Research, Site Giessen-Marburg-Langen, Justus-Liebig-University Giessen, Giessen, Germany
| | - Trinad Chakraborty
- Institute of Medical Microbiology, German Centre of Infection Research, Site Giessen-Marburg-Langen, Justus-Liebig-University Giessen, Giessen, Germany
| | - Mrutyunjay Suar
- School of Biotechnology, KIIT University, Bhubaneswar, Odisha, India
| |
Collapse
|
32
|
Srikumar S, Kröger C, Hébrard M, Colgan A, Owen SV, Sivasankaran SK, Cameron ADS, Hokamp K, Hinton JCD. RNA-seq Brings New Insights to the Intra-Macrophage Transcriptome of Salmonella Typhimurium. PLoS Pathog 2015; 11:e1005262. [PMID: 26561851 PMCID: PMC4643027 DOI: 10.1371/journal.ppat.1005262] [Citation(s) in RCA: 184] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2015] [Accepted: 10/17/2015] [Indexed: 11/18/2022] Open
Abstract
Salmonella enterica serovar Typhimurium is arguably the world’s best-understood bacterial pathogen. However, crucial details about the genetic programs used by the bacterium to survive and replicate in macrophages have remained obscure because of the challenge of studying gene expression of intracellular pathogens during infection. Here, we report the use of deep sequencing (RNA-seq) to reveal the transcriptional architecture and gene activity of Salmonella during infection of murine macrophages, providing new insights into the strategies used by the pathogen to survive in a bactericidal immune cell. We characterized 3583 transcriptional start sites that are active within macrophages, and highlight 11 of these as candidates for the delivery of heterologous antigens from Salmonella vaccine strains. A majority (88%) of the 280 S. Typhimurium sRNAs were expressed inside macrophages, and SPI13 and SPI2 were the most highly expressed pathogenicity islands. We identified 31 S. Typhimurium genes that were strongly up-regulated inside macrophages but expressed at very low levels during in vitro growth. The SalComMac online resource allows the visualisation of every transcript expressed during bacterial replication within mammalian cells. This primary transcriptome of intra-macrophage S.-Typhimurium describes the transcriptional start sites and the transcripts responsible for virulence traits, and catalogues the sRNAs that may play a role in the regulation of gene expression during infection. The burden of Salmonellosis remains unacceptably high throughout the world and control measures have had limited success. Because Salmonella bacteria can be transmitted from the wider environment to animals and humans, the bacteria encounter diverse environments that include food, water, plant surfaces and the extracellular and intracellular phases of infection of eukaryotic hosts. An intricate transcriptional network has evolved to respond to a variety of environmental signals and control the “right time/ right place” expression of virulence genes. To understand how transcription is rewired during intracellular infection, we determined the primary transcriptome of Salmonella enterica serovar Typhimurium within murine macrophages. We report the coding genes, sRNAs and transcriptional start sites that are expressed within macrophages at 8 hours after infection, and use these to infer gene function. We identified gene promoters that are specifically expressed within macrophages and could drive the intracellular delivery of antigens by S. Typhimurium vaccine strains. These data contribute to our understanding of the mechanisms used by Salmonella to regulate virulence gene expression whilst replicating inside mammalian cells.
Collapse
Affiliation(s)
- Shabarinath Srikumar
- Department of Microbiology, School of Genetics and Microbiology, Moyne Institute of Preventive Medicine, Trinity College, Dublin, Ireland
- Institute of Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| | - Carsten Kröger
- Department of Microbiology, School of Genetics and Microbiology, Moyne Institute of Preventive Medicine, Trinity College, Dublin, Ireland
- Institute of Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| | - Magali Hébrard
- Department of Microbiology, School of Genetics and Microbiology, Moyne Institute of Preventive Medicine, Trinity College, Dublin, Ireland
| | - Aoife Colgan
- Department of Microbiology, School of Genetics and Microbiology, Moyne Institute of Preventive Medicine, Trinity College, Dublin, Ireland
| | - Siân V. Owen
- Institute of Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| | - Sathesh K. Sivasankaran
- Department of Microbiology, School of Genetics and Microbiology, Moyne Institute of Preventive Medicine, Trinity College, Dublin, Ireland
| | | | - Karsten Hokamp
- Department of Genetics, School of Genetics and Microbiology, Smurfit Institute of Genetics, Trinity College, Dublin, Ireland
| | - Jay C. D. Hinton
- Department of Microbiology, School of Genetics and Microbiology, Moyne Institute of Preventive Medicine, Trinity College, Dublin, Ireland
- Institute of Integrative Biology, University of Liverpool, Liverpool, United Kingdom
- * E-mail:
| |
Collapse
|
33
|
Abstract
Human salmonellosis is generally associated with Salmonella enterica from subspecies enterica (subspecies I). Acute infections can present in one of four ways: enteric fever, gastroenteritis, bacteremia, or extraintestinal focal infection. As with other infectious diseases, the course and outcome of the infection depend on a variety of factors, including the infecting organism, the inoculating dose, and the immune status and genetic background of the host. For serovarsTyphi and Paratyphi A there is a clear association between the genetic background of the serovar and systemic infection in humans. For serovars Paratyphi B and Paratyphi C, a good clinical description of the host and detailed population genetics of the pathogen are necessary before more detailed genetic studies of novel virulence factors,or host factors,can be initiated. For the nontyphoidalserovars (NTS) the situation is less clear. Serovars Typhimurium and Enteritidis are the most common within the food chain, and so the large number of invasive infections associated with these serovars is most likely due to exposure rather than to increased virulence of the pathogen. In Africa, however, a closely related group of strains of serovar Typhimurium, associated with HIV infection, may have become host adapted tohumans, suggesting that not all isolates called "Typhimurium" should be considered as a single group. Here we review current knowledge of the salmonellae for which invasive disease in humans is an important aspect of their population biology.
Collapse
|
34
|
Ballesté-Delpierre C, Fàbrega A, Ferrer-Navarro M, Mathur R, Ghosh S, Vila J. Attenuation of in vitro host-pathogen interactions in quinolone-resistant Salmonella Typhi mutants. J Antimicrob Chemother 2015; 71:111-22. [PMID: 26446080 DOI: 10.1093/jac/dkv299] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Accepted: 08/22/2015] [Indexed: 12/25/2022] Open
Abstract
OBJECTIVES The relationship between quinolone resistance acquisition and invasion impairment has been studied in some Salmonella enterica serovars. However, little information has been reported regarding the invasive human-restricted pathogen Salmonella Typhi. The aim of this study was to investigate the molecular mechanisms of quinolone resistance acquisition and its impact on virulence in this serovar. METHODS Two antibiotic-resistant mutants (Ty_c1 and Ty_c2) were generated from a Salmonella Typhi clinical isolate (Ty_wt). The three strains were compared in terms of antimicrobial susceptibility, molecular mechanisms of resistance, gene expression of virulence-related factors, ability to invade eukaryotic cells (human epithelial cells and macrophages) and cytokine production. RESULTS Multidrug resistance in Ty_c2 was attributed to AcrAB/TolC overproduction, decreased OmpF (both mediated by the mar regulon) and decreased OmpC. The two mutants showed a gradually reduced expression of virulence-related genes (invA, hilA, hilD, fliC and fimA), correlating with decreased motility, reduced infection of HeLa cells and impaired uptake by and intracellular survival in human macrophages. Moreover, Ty_c2 also showed reduced tviA expression. Additionally, we revealed a significant reduction in TNF-α and IL-1β production and decreased NF-κB activation. CONCLUSIONS In this study, we provide an in-depth characterization of the molecular mechanisms of antibiotic resistance in the Salmonella Typhi serovar and evidence that acquisition of antimicrobial resistance is concomitantly detected with a loss of virulence (epithelial cell invasion, macrophage phagocytosis and cytokine production). We suggest that the low prevalence of clinical isolates of Salmonella Typhi highly resistant to ciprofloxacin is due to poor immunogenicity and impaired dissemination ability of these isolates.
Collapse
Affiliation(s)
- Clara Ballesté-Delpierre
- ISGlobal, Barcelona Ctr. Int. Health Res. (CRESIB), Hospital Clínic - Universitat de Barcelona, Rosselló 149-153 Barcelona, 08036, Spain
| | - Anna Fàbrega
- ISGlobal, Barcelona Ctr. Int. Health Res. (CRESIB), Hospital Clínic - Universitat de Barcelona, Rosselló 149-153 Barcelona, 08036, Spain
| | - Mario Ferrer-Navarro
- ISGlobal, Barcelona Ctr. Int. Health Res. (CRESIB), Hospital Clínic - Universitat de Barcelona, Rosselló 149-153 Barcelona, 08036, Spain
| | - Ramkumar Mathur
- Department of Microbiology and Immunology, College of Physicians and Surgeons, Columbia University, 701 West 168th Street, New York City, NY 10032, USA
| | - Sankar Ghosh
- Department of Microbiology and Immunology, College of Physicians and Surgeons, Columbia University, 701 West 168th Street, New York City, NY 10032, USA
| | - Jordi Vila
- ISGlobal, Barcelona Ctr. Int. Health Res. (CRESIB), Hospital Clínic - Universitat de Barcelona, Rosselló 149-153 Barcelona, 08036, Spain
| |
Collapse
|
35
|
Berrocal L, Fuentes JA, Trombert AN, Jofré MR, Villagra NA, Valenzuela LM, Mora GC. stg fimbrial operon from S. Typhi STH2370 contributes to association and cell disruption of epithelial and macrophage-like cells. Biol Res 2015; 48:34. [PMID: 26149381 PMCID: PMC4494162 DOI: 10.1186/s40659-015-0024-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Accepted: 06/23/2015] [Indexed: 11/10/2022] Open
Abstract
Background Salmonella enterica serovar Typhi (S. Typhi) stg operon, encoding a chaperone/usher fimbria (CU), contributes to an increased adherence to human epithelial cells. However, one report suggests that the presence of the Stg fimbria impairs the monocyte—bacteria association, as deduced by the lower level of invasion to macrophage-like cells observed when the stg fimbrial cluster was overexpressed. Nevertheless, since other CU fimbrial structures increase the entry of S. Typhi into macrophages, and considering that transcriptomic analyses revealed that stg operon is indeed expressed in macrophages, we reassessed the role of the stg operon in the interaction between S. Typhi strain STH2370 and human cells, including macrophage-like cells and mononuclear cells directly taken from human peripheral blood. Results We compared S. Typhi STH2370 WT, a Chilean clinical strain, and the S. Typhi STH2370 Δstg mutant with respect to association and invasion using epithelial and macrophage-like cells. We observed that deletion of stg operon reduced the association and invasion of S. Typhi, in both cellular types. The presence of the cloned stg operon restored the WT phenotype in all the cases. Moreover, we compared Salmonella enterica sv. Typhimurium 14028s (S. Typhimurium, a serovar lacking stg operon) and S. Typhimurium heterologously expressing S. Typhi stg. We found that the latter presents an increased cell disruption of polarized epithelial cells and an increased association in both epithelial and macrophage-like cells. Conclusions S. Typhi stg operon encodes a functional adhesin that participates in the interaction bacteria—eukaryotic cells, including epithelial cells and macrophages-like cells. The phenotypes associated to stg operon include increased association and consequent invasion in bacteria—eukaryotic cells, and cell disruption. Electronic supplementary material The online version of this article (doi:10.1186/s40659-015-0024-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Liliana Berrocal
- Laboratorio de Microbiología, Facultad de Ciencias Biológicas, Universidad Andres Bello, República 217, Santiago, Chile. .,Escuela de Medicina, Facultad de Medicina, Universidad Finis Terrae, Santiago, Av. Pedro de Valdivia 1509, Providencia, Santiago, Chile.
| | - Juan A Fuentes
- Laboratorio de Microbiología, Facultad de Ciencias Biológicas, Universidad Andres Bello, República 217, Santiago, Chile.
| | - A Nicole Trombert
- Laboratorio de Microbiología, Facultad de Ciencias Biológicas, Universidad Andres Bello, República 217, Santiago, Chile. .,Centro de Genómica y Bioinformática, Facultad de Ciencias, Universidad Mayor, Santiago, Camino La Pirámide 5750, Huechuraba, Santiago, Chile.
| | - Matías R Jofré
- Laboratorio de Microbiología, Facultad de Ciencias Biológicas, Universidad Andres Bello, República 217, Santiago, Chile. .,Escuela de Medicina, Facultad de Medicina, Universidad Finis Terrae, Santiago, Av. Pedro de Valdivia 1509, Providencia, Santiago, Chile.
| | - Nicolás A Villagra
- Laboratorio de Microbiología, Facultad de Medicina, Universidad Andres Bello, República 313, Santiago, Chile.
| | - Luis M Valenzuela
- Laboratorio de Microbiología, Facultad de Ciencias Biológicas, Universidad Andres Bello, República 217, Santiago, Chile.
| | - Guido C Mora
- Laboratorio de Microbiología, Facultad de Medicina, Universidad Andres Bello, República 313, Santiago, Chile.
| |
Collapse
|
36
|
Schunder E, Gillmaier N, Kutzner E, Eisenreich W, Herrmann V, Lautner M, Heuner K. Amino Acid Uptake and Metabolism of Legionella pneumophila Hosted by Acanthamoeba castellanii. J Biol Chem 2015; 289:21040-54. [PMID: 24904060 DOI: 10.1074/jbc.m114.570085] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Legionella pneumophila survives and replicates within a Legionella-containing vacuole (LCV) of amoebae and macrophages. Less is known about the carbon metabolism of the bacteria within the LCV. We have now analyzed the transfer and usage of amino acids from the natural host organism Acanthamoeba castellanii to Legionella pneumophila under in vivo (LCV) conditions. For this purpose, A. castellanii was 13C-labeled by incubation in buffer containing [U-(13)C(6)]glucose. Subsequently, these 13C-prelabeled amoebae were infected with L. pneumophila wild type or some mutants defective in putative key enzymes or regulators of carbon metabolism. 13C-Isotopologue compositions of amino acids from bacterial and amoebal proteins were then determined by mass spectrometry. In a comparative approach, the profiles documented the efficient uptake of Acanthamoeba amino acids into the LCV and further into L. pneumophila where they served as precursors for bacterial protein biosynthesis. More specifically, A. castellanii synthesized from exogenous [U-13C6]glucose unique isotopologue mixtures of several amino acids including Phe and Tyr, which were also observed in the same amino acids from LCV-grown L. pneumophila. Minor but significant differences were only detected in the isotopologue profiles of Ala, Asp, and Glu from the amoebal or bacterial protein fractions, respectively, indicating partial de novo synthesis of these amino acids by L. pneumophila. The similar isotopologue patterns in amino acids from L. pneumophila wild type and the mutants under study reflected the robustness of amino acid usage in the LCV of A. castellannii.
Collapse
|
37
|
de Paiva JB, Leite JL, da Silva LPM, Rojas TCG, de Pace F, Conceição RA, Sperandio V, da Silveira WD. Influence of the major nitrite transporter NirC on the virulence of a Swollen Head Syndrome avian pathogenic E. coli (APEC) strain. Vet Microbiol 2014; 175:123-31. [PMID: 25487442 DOI: 10.1016/j.vetmic.2014.11.015] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Revised: 11/13/2014] [Accepted: 11/18/2014] [Indexed: 12/21/2022]
Abstract
Avian Pathogenic Escherichia coli (APEC) strains are extra-intestinal E. coli that infect poultry and cause diseases. Nitrite is a central branch-point in bacterial nitrogen metabolism and is used as a cytotoxin by macrophages. Unlike nitric oxide (NO), nitrite cannot diffuse across bacterial membrane cells. The NirC protein acts as a specific channel to facilitate the transport of nitrite into Salmonella and E. coli cells for nitrogen metabolism and cytoplasmic detoxification. NirC is also required for the pathogenicity of Salmonella by downregulating the production of NO by the host macrophages. Based on an in vitro microarray that revealed the overexpression of the nirC gene in APEC strain SCI-07, we constructed a nirC-deficient SCI-07 strain (ΔnirC) and evaluated its virulence potential using in vivo and in vitro assays. The final cumulative mortalities caused by mutant and wild-type (WT) were similar; while the ΔnirC caused a gradual increase in the mortality rate during the seven days recorded, the WT caused mortality up to 24h post-infection (hpi). Counts of the ΔnirC cells in the spleen, lung and liver were higher than those of the WT after 48 hpi but similar at 24 hpi. Although similar number of ΔnirC and WT cells was observed in macrophages at 3 hpi, there was higher number of ΔnirC cells at 16 hpi. The cell adhesion ability of the ΔnirC strain was about half the WT level in the presence and absence of alpha-D-mannopyranoside. These results indicate that the nirC gene influences the pathogenicity of SCI-07 strain.
Collapse
Affiliation(s)
- Jacqueline Boldrin de Paiva
- Department of Genetics, Evolution and Bioagents, Institute of Biology, State University of Campinas-UNICAMP, PO Box 6109, Campinas, SP, Brazil.
| | - Janaína Luisa Leite
- Department of Genetics, Evolution and Bioagents, Institute of Biology, State University of Campinas-UNICAMP, PO Box 6109, Campinas, SP, Brazil
| | - Livia Pilatti Mendes da Silva
- Department of Genetics, Evolution and Bioagents, Institute of Biology, State University of Campinas-UNICAMP, PO Box 6109, Campinas, SP, Brazil
| | - Thais Cabrera Galvão Rojas
- Department of Genetics, Evolution and Bioagents, Institute of Biology, State University of Campinas-UNICAMP, PO Box 6109, Campinas, SP, Brazil
| | - Fernanda de Pace
- Department of Clinical Medicine, Faculty of Medical Sciences, State University of Campinas-UNICAMP, Campinas, SP, Brazil
| | - Rogério Arcuri Conceição
- Department of Genetics, Evolution and Bioagents, Institute of Biology, State University of Campinas-UNICAMP, PO Box 6109, Campinas, SP, Brazil
| | - Vanessa Sperandio
- University of Texas Southwestern Medical Center, Dallas, TX 75390-9048, USA
| | - Wanderley Dias da Silveira
- Department of Genetics, Evolution and Bioagents, Institute of Biology, State University of Campinas-UNICAMP, PO Box 6109, Campinas, SP, Brazil.
| |
Collapse
|
38
|
Ortega AD, Quereda JJ, Pucciarelli MG, García-del Portillo F. Non-coding RNA regulation in pathogenic bacteria located inside eukaryotic cells. Front Cell Infect Microbiol 2014; 4:162. [PMID: 25429360 PMCID: PMC4228915 DOI: 10.3389/fcimb.2014.00162] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Accepted: 10/20/2014] [Indexed: 01/06/2023] Open
Abstract
Intracellular bacterial pathogens have evolved distinct lifestyles inside eukaryotic cells. Some pathogens coexist with the infected cell in an obligate intracellular state, whereas others transit between the extracellular and intracellular environment. Adaptation to these intracellular lifestyles is regulated in both space and time. Non-coding small RNAs (sRNAs) are post-transcriptional regulatory molecules that fine-tune important processes in bacterial physiology including cell envelope architecture, intermediate metabolism, bacterial communication, biofilm formation, and virulence. Recent studies have shown production of defined sRNA species by intracellular bacteria located inside eukaryotic cells. The molecules targeted by these sRNAs and their expression dynamics along the intracellular infection cycle remain, however, poorly characterized. Technical difficulties linked to the isolation of “intact” intracellular bacteria from infected host cells might explain why sRNA regulation in these specialized pathogens is still a largely unexplored field. Transition from the extracellular to the intracellular lifestyle provides an ideal scenario in which regulatory sRNAs are intended to participate; so much work must be done in this direction. This review focuses on sRNAs expressed by intracellular bacterial pathogens during the infection of eukaryotic cells, strategies used with these pathogens to identify sRNAs required for virulence, and the experimental technical challenges associated to this type of studies. We also discuss varied techniques for their potential application to study RNA regulation in intracellular bacterial infections.
Collapse
Affiliation(s)
- Alvaro D Ortega
- Centro Nacional de Biotecnología - Consejo Superior de Investigaciones Científicas (CNB-CSIC) Madrid, Spain
| | - Juan J Quereda
- Centro Nacional de Biotecnología - Consejo Superior de Investigaciones Científicas (CNB-CSIC) Madrid, Spain
| | - M Graciela Pucciarelli
- Centro Nacional de Biotecnología - Consejo Superior de Investigaciones Científicas (CNB-CSIC) Madrid, Spain ; Departamento de Biología Molecular, Universidad Autónoma de Madrid, Centro de Biología Molecular 'Severo Ochoa' (CBMSO-CSIC) Madrid, Spain
| | | |
Collapse
|
39
|
Wang Y, Yi L, Wang S, Lu C, Ding C. Selective capture of transcribed sequences in the functional gene analysis of microbial pathogens. Appl Microbiol Biotechnol 2014; 98:9983-92. [PMID: 25381492 DOI: 10.1007/s00253-014-6190-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Revised: 10/23/2014] [Accepted: 10/25/2014] [Indexed: 01/26/2023]
Abstract
Selective capture of transcribed sequences (SCOTS) is an effective method to identify bacterial genes differentially expressed during different biological processes, including pathogenic interactions with a host species. The method can be used to elucidate molecular mechanisms driving and maintaining such interactions. The method is a powerful genetic tool that overcomes limitations found in other methods, by working with small amounts of mRNA and allowing for the separation of bacterial cDNA from host cDNA. It has been increasingly used in the discovery of genes involved in the bacterium-host interaction. In this review, we briefly introduce the SCOTS method, outline the technical advances offered in the method, and focus on the method's applications in several microbial pathogens.
Collapse
Affiliation(s)
- Yang Wang
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China,
| | | | | | | | | |
Collapse
|
40
|
Wilharm G, Heider C. Interrelationship between type three secretion system and metabolism in pathogenic bacteria. Front Cell Infect Microbiol 2014; 4:150. [PMID: 25386411 PMCID: PMC4209828 DOI: 10.3389/fcimb.2014.00150] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Accepted: 10/07/2014] [Indexed: 11/13/2022] Open
Abstract
Before the advent of molecular biology methods, studies of pathogens were dominated by analyses of their metabolism. Development of molecular biology techniques then enabled the identification and functional characterisation of the fascinating toolbox of virulence factors. Increasing, genomic and proteomic approaches form the basis for a more systemic view on pathogens' functions in the context of infection. Re-emerging interest in the metabolism of pathogens and hosts further expands our view of infections. There is increasing evidence that virulence functions and metabolism of pathogens are extremely intertwined. Type three secretion systems (T3SSs) are major virulence determinants of many Gram-negative pathogens and it is the objective of this review to illustrate the intertwined relationship between T3SSs and the metabolism of the pathogens deploying them.
Collapse
|
41
|
Dougan G, Baker S. Salmonella entericaSerovar Typhi and the Pathogenesis of Typhoid Fever. Annu Rev Microbiol 2014; 68:317-36. [DOI: 10.1146/annurev-micro-091313-103739] [Citation(s) in RCA: 191] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Gordon Dougan
- The Wellcome Trust Sanger Institute, The Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SA, United Kingdom;
| | - Stephen Baker
- The Hospital for Tropical Diseases, Wellcome Trust Major Overseas Program, Oxford University, Clinical Research Unit, Ho Chi Minh City, Vietnam;
- Centre for Tropical Medicine, Oxford University, Oxford OX3 7FZ, United Kingdom
- The London School of Hygiene and Tropical Medicine, London WC1E 7HT, United Kingdom
| |
Collapse
|
42
|
Dhal PK, Barman RK, Saha S, Das S. Dynamic modularity of host protein interaction networks in Salmonella Typhi infection. PLoS One 2014; 9:e104911. [PMID: 25144185 PMCID: PMC4140748 DOI: 10.1371/journal.pone.0104911] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2014] [Accepted: 07/17/2014] [Indexed: 01/08/2023] Open
Abstract
Background Salmonella Typhi is a human-restricted pathogen, which causes typhoid fever and remains a global health problem in the developing countries. Although previously reported host expression datasets had identified putative biomarkers and therapeutic targets of typhoid fever, the underlying molecular mechanism of pathogenesis remains incompletely understood. Methods We used five gene expression datasets of human peripheral blood from patients suffering from S. Typhi or other bacteremic infections or non-infectious disease like leukemia. The expression datasets were merged into human protein interaction network (PIN) and the expression correlation between the hubs and their interacting proteins was measured by calculating Pearson Correlation Coefficient (PCC) values. The differences in the average PCC for each hub between the disease states and their respective controls were calculated for studied datasets. The individual hubs and their interactors with expression, PCC and average PCC values were treated as dynamic subnetworks. The hubs that showed unique trends of alterations specific to S. Typhi infection were identified. Results We identified S. Typhi infection-specific dynamic subnetworks of the host, which involve 81 hubs and 1343 interactions. The major enriched GO biological process terms in the identified subnetworks were regulation of apoptosis and biological adhesions, while the enriched pathways include cytokine signalling in the immune system and downstream TCR signalling. The dynamic nature of the hubs CCR1, IRS2 and PRKCA with their interactors was studied in detail. The difference in the dynamics of the subnetworks specific to S. Typhi infection suggests a potential molecular model of typhoid fever. Conclusions Hubs and their interactors of the S. Typhi infection-specific dynamic subnetworks carrying distinct PCC values compared with the non-typhoid and other disease conditions reveal new insight into the pathogenesis of S. Typhi.
Collapse
Affiliation(s)
- Paltu Kumar Dhal
- Biomedical Informatics Centre, National Institute of Cholera and Enteric Diseases, Kolkata, West Bengal, India
| | - Ranjan Kumar Barman
- Biomedical Informatics Centre, National Institute of Cholera and Enteric Diseases, Kolkata, West Bengal, India
| | - Sudipto Saha
- Bioinformatics Centre, Bose Institute, Kolkata, West Bengal, India
| | - Santasabuj Das
- Biomedical Informatics Centre, National Institute of Cholera and Enteric Diseases, Kolkata, West Bengal, India; Division of Clinical Medicine, National Institute of Cholera and Enteric Diseases, Kolkata, West Bengal, India
| |
Collapse
|
43
|
Jofré MR, Rodríguez LM, Villagra NA, Hidalgo AA, Mora GC, Fuentes JA. RpoS integrates CRP, Fis, and PhoP signaling pathways to control Salmonella Typhi hlyE expression. BMC Microbiol 2014; 14:139. [PMID: 24885225 PMCID: PMC4105832 DOI: 10.1186/1471-2180-14-139] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Accepted: 05/21/2014] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND SPI-18 is a pathogenicity island found in some Salmonella enterica serovars, including S. Typhi. SPI-18 harbors two ORFs organized into an operon, hlyE and taiA genes, both implicated in virulence. Regarding the hlyE regulation in S. Typhi, it has been reported that RpoS participates as transcriptional up-regulator under low pH and high osmolarity. In addition, CRP down-regulates hlyE expression during exponential growth. Previously, it has been suggested that there is another factor related to catabolite repression, different from CRP, involved in the down-regulation of hlyE. Moreover, PhoP-dependent hlyE up-regulation has been reported in bacteria cultured simultaneously under low pH and low concentration of Mg2+. Nevertheless, the relative contribution of each environmental signal is not completely clear. In this work we aimed to better understand the regulation of hlyE in S. Typhi and the integration of different environmental signals through global regulators. RESULTS We found that Fis participates as a CRP-independent glucose-dependent down-regulator of hlyE. Also, Fis and CRP seem to exert the repression over hlyE through down-regulating rpoS. Moreover, PhoP up-regulates hlyE expression via rpoS under low pH and low Mg2+ conditions. CONCLUSIONS All these results together show that, at least under the tested conditions, RpoS is the central regulator in the hlyE regulatory network, integrating multiple environmental signals and global regulators.
Collapse
Affiliation(s)
| | | | | | | | | | - Juan A Fuentes
- Facultad de Ciencias Biológicas, Universidad Andres Bello, Santiago, Chile.
| |
Collapse
|
44
|
Cameron ADS, Kröger C, Quinn HJ, Scally IK, Daly AJ, Kary SC, Dorman CJ. Transmission of an oxygen availability signal at the Salmonella enterica serovar Typhimurium fis promoter. PLoS One 2013; 8:e84382. [PMID: 24358360 PMCID: PMC3865300 DOI: 10.1371/journal.pone.0084382] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2013] [Accepted: 11/14/2013] [Indexed: 11/19/2022] Open
Abstract
The nucleoid-associated protein FIS is a global regulator of gene expression and chromosome structure in Escherichia coli and Salmonella enterica. Despite the importance of FIS for infection and intracellular invasion, very little is known about the regulation of S. enterica fis expression. Under standard laboratory growth conditions, fis is highly expressed during rapid growth but is then silenced as growth slows. However, if cells are cultured in non-aerated conditions, fis expression is sustained during stationary phase. This led us to test whether the redox-sensing transcription factors ArcA and FNR regulate S. enterica fis. Deletion of FNR had no detectable effect, whereas deletion of ArcA had the unexpected effect of further elevating fis expression in stationary phase. ArcA required RpoS for induction of fis expression, suggesting that ArcA indirectly affects fis expression. Other putative regulators were found to play diverse roles: FIS acted directly as an auto-repressor (as expected), whereas CRP had little direct effect on fis expression. Deleting regions of the fis promoter led to the discovery of a novel anaerobically-induced transcription start site (Pfis-2) upstream of the primary transcription start site (Pfis-1). Promoter truncation also revealed that the shortest functional fis promoter was incapable of sustained expression. Moreover, fis expression was observed to correlate directly with DNA supercoiling in non-aerated conditions. Thus, the full-length S. enterica fis promoter region may act as a topological switch that is sensitive to stress-induced duplex destabilisation and up-regulates expression in non-aerated conditions.
Collapse
Affiliation(s)
- Andrew D. S. Cameron
- Department of Microbiology, Moyne Institute of Preventive Medicine, School of Genetics and Microbiology, Trinity College Dublin, Dublin, Ireland
- Department of Biology, University of Regina, Regina, SK, Canada
| | - Carsten Kröger
- Department of Microbiology, Moyne Institute of Preventive Medicine, School of Genetics and Microbiology, Trinity College Dublin, Dublin, Ireland
| | - Heather J. Quinn
- Department of Microbiology, Moyne Institute of Preventive Medicine, School of Genetics and Microbiology, Trinity College Dublin, Dublin, Ireland
| | - Isobel K. Scally
- Department of Microbiology, Moyne Institute of Preventive Medicine, School of Genetics and Microbiology, Trinity College Dublin, Dublin, Ireland
| | - Anne J. Daly
- Department of Microbiology, Moyne Institute of Preventive Medicine, School of Genetics and Microbiology, Trinity College Dublin, Dublin, Ireland
| | - Stefani C. Kary
- Department of Biology, University of Regina, Regina, SK, Canada
| | - Charles J. Dorman
- Department of Microbiology, Moyne Institute of Preventive Medicine, School of Genetics and Microbiology, Trinity College Dublin, Dublin, Ireland
- * E-mail:
| |
Collapse
|
45
|
Humphrys MS, Creasy T, Sun Y, Shetty AC, Chibucos MC, Drabek EF, Fraser CM, Farooq U, Sengamalay N, Ott S, Shou H, Bavoil PM, Mahurkar A, Myers GSA. Simultaneous transcriptional profiling of bacteria and their host cells. PLoS One 2013; 8:e80597. [PMID: 24324615 PMCID: PMC3851178 DOI: 10.1371/journal.pone.0080597] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2013] [Accepted: 10/14/2013] [Indexed: 02/06/2023] Open
Abstract
We developed an RNA-Seq-based method to simultaneously capture prokaryotic and eukaryotic expression profiles of cells infected with intracellular bacteria. As proof of principle, this method was applied to Chlamydia trachomatis-infected epithelial cell monolayers in vitro, successfully obtaining transcriptomes of both C. trachomatis and the host cells at 1 and 24 hours post-infection. Chlamydiae are obligate intracellular bacterial pathogens that cause a range of mammalian diseases. In humans chlamydiae are responsible for the most common sexually transmitted bacterial infections and trachoma (infectious blindness). Disease arises by adverse host inflammatory reactions that induce tissue damage & scarring. However, little is known about the mechanisms underlying these outcomes. Chlamydia are genetically intractable as replication outside of the host cell is not yet possible and there are no practical tools for routine genetic manipulation, making genome-scale approaches critical. The early timeframe of infection is poorly understood and the host transcriptional response to chlamydial infection is not well defined. Our simultaneous RNA-Seq method was applied to a simplified in vitro model of chlamydial infection. We discovered a possible chlamydial strategy for early iron acquisition, putative immune dampening effects of chlamydial infection on the host cell, and present a hypothesis for Chlamydia-induced fibrotic scarring through runaway positive feedback loops. In general, simultaneous RNA-Seq helps to reveal the complex interplay between invading bacterial pathogens and their host mammalian cells and is immediately applicable to any bacteria/host cell interaction.
Collapse
Affiliation(s)
- Michael S. Humphrys
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Todd Creasy
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Yezhou Sun
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Amol C. Shetty
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Marcus C. Chibucos
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
- Department of Microbiology & Immunology, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Elliott F. Drabek
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Claire M. Fraser
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
- Department of Microbiology & Immunology, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Umar Farooq
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Naomi Sengamalay
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Sandy Ott
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Huizhong Shou
- Department of Microbiology & Immunology, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Patrik M. Bavoil
- Department of Microbiology & Immunology, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
- Department of Microbial Pathogenesis, University of Maryland Dental School, Baltimore, Maryland, United States of America
| | - Anup Mahurkar
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Garry S. A. Myers
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
- Department of Microbiology & Immunology, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
- Department of Microbial Pathogenesis, University of Maryland Dental School, Baltimore, Maryland, United States of America
- * E-mail:
| |
Collapse
|
46
|
Sheng X, Zhang H, Xia Q, Xu S, Xu H, Huang X. Mig-14 plays an important role in influencing gene expression of Salmonella enterica serovar Typhi, which contributes to cell invasion under hyperosmotic conditions. Res Microbiol 2013; 164:903-12. [DOI: 10.1016/j.resmic.2013.07.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2013] [Accepted: 07/22/2013] [Indexed: 10/26/2022]
|
47
|
Mastroeni P, Grant A. Dynamics of spread of Salmonella enterica in the systemic compartment. Microbes Infect 2013; 15:849-57. [PMID: 24183878 DOI: 10.1016/j.micinf.2013.10.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2013] [Accepted: 10/09/2013] [Indexed: 12/21/2022]
Abstract
Traditional microbiological and immunological tools, combined with modern imaging, and molecular and mathematical approaches, have revealed the dispersive nature of Salmonella infections. Bacterial escape from infected cells, spread in the tissues and attempts to restrain this process by the host give rise to fascinating scenarios that underpin the pathogenesis of salmonelloses.
Collapse
Affiliation(s)
- Pietro Mastroeni
- Department of Veterinary Medicine, University of Cambridge, Madingley Road, Cambridge CB3 0ES, United Kingdom.
| | | |
Collapse
|
48
|
Hammarlöf DL, Canals R, Hinton JCD. The FUN of identifying gene function in bacterial pathogens; insights from Salmonella functional genomics. Curr Opin Microbiol 2013; 16:643-51. [PMID: 24021902 DOI: 10.1016/j.mib.2013.07.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2013] [Accepted: 07/12/2013] [Indexed: 02/01/2023]
Abstract
The availability of thousands of genome sequences of bacterial pathogens poses a particular challenge because each genome contains hundreds of genes of unknown function (FUN). How can we easily discover which FUN genes encode important virulence factors? One solution is to combine two different functional genomic approaches. First, transcriptomics identifies bacterial FUN genes that show differential expression during the process of mammalian infection. Second, global mutagenesis identifies individual FUN genes that the pathogen requires to cause disease. The intersection of these datasets can reveal a small set of candidate genes most likely to encode novel virulence attributes. We demonstrate this approach with the Salmonella infection model, and propose that a similar strategy could be used for other bacterial pathogens.
Collapse
Affiliation(s)
- Disa L Hammarlöf
- Institute of Integrative Biology, University of Liverpool, Crown Street, Liverpool L69 7ZB, United Kingdom
| | | | | |
Collapse
|
49
|
Charles RC, Sultana T, Alam MM, Yu Y, Wu-Freeman Y, Bufano MK, Rollins SM, Tsai L, Harris JB, LaRocque RC, Leung DT, Brooks WA, Nga TVT, Dongol S, Basnyat B, Calderwood SB, Farrar J, Khanam F, Gunn JS, Qadri F, Baker S, Ryan ET. Identification of immunogenic Salmonella enterica serotype Typhi antigens expressed in chronic biliary carriers of S. Typhi in Kathmandu, Nepal. PLoS Negl Trop Dis 2013; 7:e2335. [PMID: 23936575 PMCID: PMC3731212 DOI: 10.1371/journal.pntd.0002335] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2013] [Accepted: 06/12/2013] [Indexed: 11/21/2022] Open
Abstract
Background Salmonella enterica serotype Typhi can colonize and persist in the biliary tract of infected individuals, resulting in a state of asymptomatic chronic carriage. Chronic carriers may act as persistent reservoirs of infection within a community and may introduce infection to susceptible individuals and new communities. Little is known about the interaction between the host and pathogen in the biliary tract of chronic carriers, and there is currently no reliable diagnostic assay to identify asymptomatic S. Typhi carriage. Methodology/Principal Findings To study host-pathogen interactions in the biliary tract during S. Typhi carriage, we applied an immunoscreening technique called in vivo-induced antigen technology (IVIAT), to identify potential biomarkers unique to carriers. IVIAT identifies humorally immunogenic bacterial antigens expressed uniquely in the in vivo environment, and we hypothesized that S. Typhi surviving in the biliary tract of humans may express a distinct antigenic profile. Thirteen S. Typhi antigens that were immunoreactive in carriers, but not in healthy individuals from a typhoid endemic area, were identified. The identified antigens included a number of putative membrane proteins, lipoproteins, and hemolysin-related proteins. YncE (STY1479), an uncharacterized protein with an ATP-binding motif, gave prominent responses in our screen. The response to YncE in patients whose biliary tract contained S. Typhi was compared to responses in patients whose biliary tract did not contain S. Typhi, patients with acute typhoid fever, and healthy controls residing in a typhoid endemic area. Seven of 10 (70%) chronic carriers, 0 of 8 bile culture-negative controls (0%), 0 of 8 healthy Bangladeshis (0%), and 1 of 8 (12.5%) Bangladeshis with acute typhoid fever had detectable anti-YncE IgG in blood. IgA responses were also present. Conclusions/Significance Further evaluation of YncE and other antigens identified by IVIAT could lead to the development of improved diagnostic assays to identify asymptomatic S. Typhi carriers. Salmonella enterica serotype Typhi is the cause of typhoid fever and infects over 21 million individuals and causes 200,000 deaths each year. With adequate treatment, most patients recover from their acute stage of illness and clear infection. However, a small percentage of S. Typhi infected individuals develop a chronic but asymptomatic infection in the biliary tract that can persist for decades. Since S. Typhi is a human-restricted pathogen, chronic carriers may act as reservoirs of infection. Correctly identifying and treating asymptomatic chronic carriers could be critical for ultimate control of typhoid fever. Using an immunoscreening technique called in vivo-induced antigen technology (IVIAT), we have identified potential biomarkers unique to S. Typhi chronic carriers. Further evaluation of these antigens could lead to the development of improved diagnostic assays to detect asymptomatic S. Typhi carriers in typhoid endemic zones, and to an improved understanding of the pathogenesis of S. Typhi in the chronic carrier state.
Collapse
Affiliation(s)
- Richelle C Charles
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, Massachusetts, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Identification of genes transcribed by Streptococcus equi ssp. zooepidemicus in infected porcine lung. Microb Pathog 2013; 59-60:7-12. [DOI: 10.1016/j.micpath.2013.02.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2012] [Revised: 01/31/2013] [Accepted: 02/04/2013] [Indexed: 01/03/2023]
|