1
|
Buzilă ER, Dorneanu OS, Trofin F, Sima CM, Iancu LS. Assessing Salmonella Typhi Pathogenicity and Prevention: The Crucial Role of Vaccination in Combating Typhoid Fever. Int J Mol Sci 2025; 26:3981. [PMID: 40362220 PMCID: PMC12071698 DOI: 10.3390/ijms26093981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2025] [Revised: 04/17/2025] [Accepted: 04/21/2025] [Indexed: 05/15/2025] Open
Abstract
Enteric fever is caused by Salmonella enterica serovar Typhi (S. Typhi) and Salmonella enterica serovar Paratyphi (S. Paratyphi) A, B, and C. Globally, an estimated 11 to 21 million cases of typhoid and paratyphoid fever occur annually, with approximately 130,000-160,000 deaths, most of which are reported in South/Southeast Asia and sub-Saharan Africa. The antibiotic susceptibility of S. Typhi strains varies between countries within broad limits, from 3% to 97% for ampicillin, 9% to 95% for ciprofloxacin, 4% to 94% for chloramphenicol (India vs. Pakistan), and 0% to 99% for ceftriaxone (India vs. Iraq). With S. Typhi increasingly exhibiting resistance to antibiotics, vaccination becomes an essential preventive measure. Currently, three vaccines are licensed for typhoid fever: the typhoid conjugate vaccine (TCV), live-attenuated oral vaccine Ty21a (Ty21a), and Vi capsular polysaccharide vaccine (Vi-CPS). While no specific vaccine exists for paratyphoid fever, the genetic and antigenic similarities between S. Paratyphi and S. Typhi offer potential for the development of such a vaccine. Early studies show promising results, demonstrating both safety and immunogenicity in preclinical trials. Whole genome sequencing (WGS) provides a powerful tool for assigning genotypes, identifying plasmids, comparing genetic elements, and investigating molecular factors that contribute to antibiotic resistance and virulence.
Collapse
Affiliation(s)
- Elena Roxana Buzilă
- Microbiology Discipline, Preventive Medicine and Interdisciplinarity Department, University of Medicine and Pharmacy “Grigore T. Popa”, 700115 Iasi, Romania; (E.R.B.); (F.T.); (C.M.S.); (L.S.I.)
- Iasi Regional Center for Public Health, National Institute of Public Health, 700465 Iasi, Romania
| | - Olivia Simona Dorneanu
- Microbiology Discipline, Preventive Medicine and Interdisciplinarity Department, University of Medicine and Pharmacy “Grigore T. Popa”, 700115 Iasi, Romania; (E.R.B.); (F.T.); (C.M.S.); (L.S.I.)
- Clinical Hospital of Infectious Diseases “Sf. Parascheva”, 700116 Iasi, Romania
| | - Felicia Trofin
- Microbiology Discipline, Preventive Medicine and Interdisciplinarity Department, University of Medicine and Pharmacy “Grigore T. Popa”, 700115 Iasi, Romania; (E.R.B.); (F.T.); (C.M.S.); (L.S.I.)
- “Sf. Spiridon” Clinical Emergency Hospital, 700111 Iasi, Romania
| | - Cristina Mihaela Sima
- Microbiology Discipline, Preventive Medicine and Interdisciplinarity Department, University of Medicine and Pharmacy “Grigore T. Popa”, 700115 Iasi, Romania; (E.R.B.); (F.T.); (C.M.S.); (L.S.I.)
- Clinical Hospital of Infectious Diseases “Sf. Parascheva”, 700116 Iasi, Romania
| | - Luminița Smaranda Iancu
- Microbiology Discipline, Preventive Medicine and Interdisciplinarity Department, University of Medicine and Pharmacy “Grigore T. Popa”, 700115 Iasi, Romania; (E.R.B.); (F.T.); (C.M.S.); (L.S.I.)
- Iasi Regional Center for Public Health, National Institute of Public Health, 700465 Iasi, Romania
| |
Collapse
|
2
|
Raman V, Hall CL, Wetherby VE, Whitney SA, Van Dessel N, Forbes NS. Controlling intracellular protein delivery, tumor colonization and tissue distribution using flhDC in clinically relevant ΔsseJ Salmonella. Mol Ther 2025; 33:649-669. [PMID: 39741404 PMCID: PMC11852948 DOI: 10.1016/j.ymthe.2024.12.038] [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: 04/30/2024] [Revised: 09/17/2024] [Accepted: 12/27/2024] [Indexed: 01/03/2025] Open
Abstract
Effectively targeting intracellular pathways in cancers requires a system that specifically delivers to tumors and internalizes into cancer cells. To achieve this goal, we developed intracellular-delivering (ID) Salmonella with controllable expression of flhDC to regulate flagella production and cell invasion. We hypothesized that controlling flhDC would overcome the poor colonization seen in prior clinical trials. To test this hypothesis, we incorporated the aspirin-responsive Psal promoter and tuned flhDC expression with ssra degradation tags. In tumor-bearing mice, controlling flhDC increased protein release, tissue dispersion, and tumor colonization more than 10 million times. We discovered that inducing flhDC increases escape from intracellular vacuoles; however, deleting sseJ prevented escape and further increased protein delivery. Delivering constitutively active caspase-3 with ID-f-s Salmonella (ΔsseJ and induced Psal-flhDC) induced cell death in pancreatic, breast, and liver cancer cells and reduced the growth of breast tumors. This clinically ready strain preferentially colonized metastatic breast tissue 280 and 800 times more than surrounding healthy tissue in the lung and liver, respectively. By precisely controlling tumor colonization and cell invasion, this strain overcomes critical limitations of bacterial therapy and will enable treatment of many hard-to-treat cancers.
Collapse
Affiliation(s)
- Vishnu Raman
- Department of Chemical Engineering, University of Massachusetts, Amherst, Amherst, MA 01003, USA; Ernest Pharmaceuticals, Inc., Hadley, MA 01035, USA
| | - Christopher L Hall
- Department of Chemical Engineering, University of Massachusetts, Amherst, Amherst, MA 01003, USA; Ernest Pharmaceuticals, Inc., Hadley, MA 01035, USA
| | | | - Samantha A Whitney
- Department of Chemical Engineering, University of Massachusetts, Amherst, Amherst, MA 01003, USA
| | | | - Neil S Forbes
- Department of Chemical Engineering, University of Massachusetts, Amherst, Amherst, MA 01003, USA; Molecular and Cell Biology Program, University of Massachusetts, Amherst, Amherst, MA 01003, USA; Institute for Applied Life Science, University of Massachusetts, Amherst, Amherst, MA 01003, USA; Department of Microbiology, University of Massachusetts, Amherst, Amherst, MA 01003, USA.
| |
Collapse
|
3
|
Han J, Aljahdali N, Zhao S, Tang H, Harbottle H, Hoffmann M, Frye JG, Foley SL. Infection biology of Salmonella enterica. EcoSal Plus 2024; 12:eesp00012023. [PMID: 38415623 PMCID: PMC11636313 DOI: 10.1128/ecosalplus.esp-0001-2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 07/31/2023] [Indexed: 02/29/2024]
Abstract
Salmonella enterica is the leading cause of bacterial foodborne illness in the USA, with an estimated 95% of salmonellosis cases due to the consumption of contaminated food products. Salmonella can cause several different disease syndromes, with the most common being gastroenteritis, followed by bacteremia and typhoid fever. Among the over 2,600 currently identified serotypes/serovars, some are mostly host-restricted and host-adapted, while the majority of serotypes can infect a broader range of host species and are associated with causing both livestock and human disease. Salmonella serotypes and strains within serovars can vary considerably in the severity of disease that may result from infection, with some serovars that are more highly associated with invasive disease in humans, while others predominantly cause mild gastroenteritis. These observed clinical differences may be caused by the genetic make-up and diversity of the serovars. Salmonella virulence systems are very complex containing several virulence-associated genes with different functions that contribute to its pathogenicity. The different clinical syndromes are associated with unique groups of virulence genes, and strains often differ in the array of virulence traits they display. On the chromosome, virulence genes are often clustered in regions known as Salmonella pathogenicity islands (SPIs), which are scattered throughout different Salmonella genomes and encode factors essential for adhesion, invasion, survival, and replication within the host. Plasmids can also carry various genes that contribute to Salmonella pathogenicity. For example, strains from several serovars associated with significant human disease, including Choleraesuis, Dublin, Enteritidis, Newport, and Typhimurium, can carry virulence plasmids with genes contributing to attachment, immune system evasion, and other roles. The goal of this comprehensive review is to provide key information on the Salmonella virulence, including the contributions of genes encoded in SPIs and plasmids during Salmonella pathogenesis.
Collapse
Affiliation(s)
- Jing Han
- National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, Arkansas, USA
| | - Nesreen Aljahdali
- National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, Arkansas, USA
- Biological Science Department, College of Science, King Abdul-Aziz University, Jeddah, Saudi Arabia
| | - Shaohua Zhao
- Center for Veterinary Medicine, U.S. Food and Drug Administration, Rockville, Maryland, USA
| | - Hailin Tang
- National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, Arkansas, USA
| | - Heather Harbottle
- Center for Veterinary Medicine, U.S. Food and Drug Administration, Rockville, Maryland, USA
| | - Maria Hoffmann
- Center for Food Safety and Applied Nutrition, U.S. Food and Drug Administration, College Park, Maryland, USA
| | - Jonathan G. Frye
- Agricutlutral Research Service, U.S. Department of Agriculture, Athens, Georgia, USA
| | - Steven L. Foley
- National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, Arkansas, USA
| |
Collapse
|
4
|
Abstract
The pathogen Salmonella enterica encompasses a range of bacterial serovars that cause intestinal inflammation and systemic infections in humans. Mice are a widely used infection model due to their relative simplicity and versatility. Here, we provide standardized protocols for culturing the prolific zoonotic pathogen S. enterica serovar Typhimurium for intragastric inoculation of mice to model colitis or systemic dissemination, along with techniques for direct extraintestinal infection. Furthermore, we present procedures for quantifying pathogen burden and for characterizing the immune response by analyzing tissue pathology, inflammatory markers, and immune cells from intestinal tissues. © 2023 The Authors. Current Protocols published by Wiley Periodicals LLC. Basic Protocol 1: Murine colitis model utilizing oral streptomycin pretreatment and oral S. Typhimurium administration Basic Protocol 2: Intraperitoneal injection of S. Typhimurium for modeling extraintestinal infection Support Protocol 1: Preparation of S. Typhimurium inoculum Support Protocol 2: Preparation of mixed S. Typhimurium inoculum for competitive infection Basic Protocol 3: Assessment of S. Typhimurium burden Support Protocol 3: Preservation and pathological assessment of S. Typhimurium-infected tissues Support Protocol 4: Measurement of inflammatory marker expression in intestinal tissues by qPCR Support Protocol 5: Preparation of intestinal content for inflammatory marker quantification by ELISA Support Protocol 6: Immune cell isolation from Salmonella-infected intestinal tissues.
Collapse
Affiliation(s)
- Gregory T. Walker
- Division of Host-Microbe Systems & Therapeutics, Department of Pediatrics, University of California San Diego, La Jolla, CA 92093, USA
| | - Romana R. Gerner
- Division of Host-Microbe Systems & Therapeutics, Department of Pediatrics, University of California San Diego, La Jolla, CA 92093, USA
- Technical University Munich, TUM School of Life Sciences Weihenstephan, ZIEL – Institute for Food & Health, Freising, Germany
- Department of Internal Medicine III, School of Medicine, University Hospital rechts der Isar, Technical University of Munich, Munich, Germany
| | - Sean-Paul Nuccio
- Division of Host-Microbe Systems & Therapeutics, Department of Pediatrics, University of California San Diego, La Jolla, CA 92093, USA
| | - Manuela Raffatellu
- Division of Host-Microbe Systems & Therapeutics, Department of Pediatrics, University of California San Diego, La Jolla, CA 92093, USA
- Center for Microbiome Innovation, University of California San Diego, La Jolla, CA 92093, USA
- Chiba University-UC San Diego Center for Mucosal Immunology, Allergy, and Vaccines (CU-UCSD cMAV), La Jolla, CA 92093, USA
| |
Collapse
|
5
|
The Vi Capsular Polysaccharide of Salmonella Typhi Promotes Macrophage Phagocytosis by Binding the Human C-Type Lectin DC-SIGN. mBio 2022; 13:e0273322. [PMID: 36286551 PMCID: PMC9765441 DOI: 10.1128/mbio.02733-22] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Capsular polysaccharides are common virulence factors of extracellular, but not intracellular bacterial pathogens, due to the antiphagocytic properties of these surface structures. It is therefore paradoxical that Salmonella enterica subspecies enterica serovar Typhi, an intracellular pathogen, synthesizes a virulence-associated (Vi) capsule, which exhibits antiphagocytic properties. Here, we show that the Vi capsular polysaccharide has different functions when S. Typhi interacts with distinct subsets of host phagocytes. The Vi capsular polysaccharide allowed S. Typhi to selectively evade phagocytosis by human neutrophils while promoting human macrophage phagocytosis. A screen of C-type lectin receptors identified human DC-SIGN as the receptor involved in macrophage binding and phagocytosis of capsulated S. Typhi. Consistent with the anti-inflammatory activity of DC-SIGN, purified Vi capsular polysaccharide reduced inflammatory responses in macrophages. These data suggest that binding of the human C-type lectin receptor DC-SIGN by the Vi capsular polysaccharide contributes to the pathogenesis of typhoid fever. IMPORTANCE Salmonella enterica subspecies enterica serovar Typhi is the causative agent of typhoid fever. The recent emergence of S. Typhi strains which are resistant to antibiotic therapy highlights the importance of vaccination in managing typhoid fever. The virulence-associated (Vi) capsular polysaccharide is an effective vaccine against typhoid fever, but the role the capsule plays during pathogenesis remains incompletely understood. Here, we identify the human C-type lectin receptor DC-SIGN as the receptor for the Vi capsular polysaccharide. Binding of capsulated S. Typhi to DC-SIGN resulted in phagocytosis of the pathogen by macrophages and induction of an anti-inflammatory cytokine response. Thus, the interaction of the Vi capsular polysaccharide with human DC-SIGN contributes to the pathogenesis of typhoid fever and should be further investigated in the context of vaccine development.
Collapse
|
6
|
Cohen H, Hoede C, Scharte F, Coluzzi C, Cohen E, Shomer I, Mallet L, Holbert S, Serre RF, Schiex T, Virlogeux-Payant I, Grassl GA, Hensel M, Chiapello H, Gal-Mor O. Intracellular Salmonella Paratyphi A is motile and differs in the expression of flagella-chemotaxis, SPI-1 and carbon utilization pathways in comparison to intracellular S. Typhimurium. PLoS Pathog 2022; 18:e1010425. [PMID: 35381053 PMCID: PMC9012535 DOI: 10.1371/journal.ppat.1010425] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 04/15/2022] [Accepted: 03/09/2022] [Indexed: 12/21/2022] Open
Abstract
Although Salmonella Typhimurium (STM) and Salmonella Paratyphi A (SPA) belong to the same phylogenetic species, share large portions of their genome and express many common virulence factors, they differ vastly in their host specificity, the immune response they elicit, and the clinical manifestations they cause. In this work, we compared their intracellular transcriptomic architecture and cellular phenotypes during human epithelial cell infection. While transcription induction of many metal transport systems, purines, biotin, PhoPQ and SPI-2 regulons was similar in both intracellular SPA and STM, we identified 234 differentially expressed genes that showed distinct expression patterns in intracellular SPA vs. STM. Surprisingly, clear expression differences were found in SPI-1, motility and chemotaxis, and carbon (mainly citrate, galactonate and ethanolamine) utilization pathways, indicating that these pathways are regulated differently during their intracellular phase. Concurring, on the cellular level, we show that while the majority of STM are non-motile and reside within Salmonella-Containing Vacuoles (SCV), a significant proportion of intracellular SPA cells are motile and compartmentalized in the cytosol. Moreover, we found that the elevated expression of SPI-1 and motility genes by intracellular SPA results in increased invasiveness of SPA, following exit from host cells. These findings demonstrate unexpected flagellum-dependent intracellular motility of a typhoidal Salmonella serovar and intriguing differences in intracellular localization between typhoidal and non-typhoidal salmonellae. We propose that these differences facilitate new cycles of host cell infection by SPA and may contribute to the ability of SPA to disseminate beyond the intestinal lamina propria of the human host during enteric fever. Salmonella enterica is a ubiquitous, facultative intracellular animal and human pathogen. Although non-typhoidal Salmonella (NTS) and typhoidal Salmonella serovars belong to the same phylogenetic species and share many virulence factors, the disease they cause in humans is very different. While the underlying mechanisms for these differences are not fully understood, one possible reason expected to contribute to their different pathogenicity is a distinct expression pattern of genes involved in host-pathogen interactions. Here, we compared the global gene expression and intracellular phenotypes, during human epithelial cell infection of S. Paratyphi A (SPA) and S. Typhimurium (STM), as prototypical serovars of typhoidal and NTS, respectively. Interestingly, we identified different expression patterns in key virulence and metabolic pathways, cytosolic motility and increased reinvasion of SPA, following exit from infected cells. We hypothesize that these differences contribute to the invasive and systemic disease developed following SPA infection in humans.
Collapse
Affiliation(s)
- Helit Cohen
- The Infectious Diseases Research Laboratory, Sheba Medical Center, Tel-Hashomer, Israel
| | - Claire Hoede
- Université Fédérale de Toulouse, INRAE, BioinfOmics, UR MIAT, GenoToul Bioinformatics facility, 31326, Castanet-Tolosan, France
| | - Felix Scharte
- Abt. Mikrobiologie, Universität Osnabrück, Osnabrück, Germany
| | - Charles Coluzzi
- INRAE, Université Paris-Saclay, MaIAGE, Jouy-en-Josas, France
| | - Emiliano Cohen
- The Infectious Diseases Research Laboratory, Sheba Medical Center, Tel-Hashomer, Israel
| | - Inna Shomer
- The Infectious Diseases Research Laboratory, Sheba Medical Center, Tel-Hashomer, Israel
| | - Ludovic Mallet
- Université Fédérale de Toulouse, INRAE, BioinfOmics, UR MIAT, GenoToul Bioinformatics facility, 31326, Castanet-Tolosan, France
| | | | | | - Thomas Schiex
- Université Fédérale de Toulouse, ANITI, INRAE, Toulouse, France
| | | | - Guntram A. Grassl
- Institute of Medical Microbiology and Hospital Epidemiology, Hannover Medical School and German Center for Infection Research (DZIF), Hanover, Germany
| | - Michael Hensel
- Abt. Mikrobiologie, Universität Osnabrück, Osnabrück, Germany
- CellNanOs–Center of Cellular Nanoanalytics Osnabrück, Universität Osnabrück, Osnabrück, Germany
- * E-mail: (MH); (HC); (OG-M)
| | - Hélène Chiapello
- Université Fédérale de Toulouse, INRAE, BioinfOmics, UR MIAT, GenoToul Bioinformatics facility, 31326, Castanet-Tolosan, France
- INRAE, Université Paris-Saclay, MaIAGE, Jouy-en-Josas, France
- * E-mail: (MH); (HC); (OG-M)
| | - Ohad Gal-Mor
- The Infectious Diseases Research Laboratory, Sheba Medical Center, Tel-Hashomer, Israel
- Department of Clinical Microbiology and Immunology, Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
- * E-mail: (MH); (HC); (OG-M)
| |
Collapse
|
7
|
Chen D, Burford WB, Pham G, Zhang L, Alto LT, Ertelt JM, Winter MG, Winter SE, Way SS, Alto NM. Systematic reconstruction of an effector-gene network reveals determinants of Salmonella cellular and tissue tropism. Cell Host Microbe 2021; 29:1531-1544.e9. [PMID: 34536347 DOI: 10.1016/j.chom.2021.08.012] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 06/24/2021] [Accepted: 08/20/2021] [Indexed: 11/17/2022]
Abstract
The minimal genetic requirements for microbes to survive within multiorganism communities, including host-pathogen interactions, remain poorly understood. Here, we combined targeted gene mutagenesis with phenotype-guided genetic reassembly to identify a cooperative network of SPI-2 T3SS effector genes that are sufficient for Salmonella Typhimurium (STm) to cause disease in a natural host organism. Five SPI-2 effector genes support pathogen survival within the host cell cytoplasm by coordinating bacterial replication with Salmonella-containing vacuole (SCV) division. Unexpectedly, this minimal genetic repertoire does not support STm systemic infection of mice. In vivo screening revealed a second effector-gene network, encoded by the spv operon, that expands the life cycle of STm from growth in cells to deep-tissue colonization in a murine model of typhoid fever. Comparison between Salmonella infection models suggests how cooperation between effector genes drives tissue tropism in a pathogen group.
Collapse
Affiliation(s)
- Didi Chen
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Wesley B Burford
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Giang Pham
- Division of Infectious Diseases, Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Lishu Zhang
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Laura T Alto
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - James M Ertelt
- Division of Infectious Diseases, Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Maria G Winter
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Sebastian E Winter
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Sing Sing Way
- Division of Infectious Diseases, Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Neal M Alto
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
8
|
Developing Cyclic Peptomers as Broad-Spectrum Type III Secretion System Inhibitors in Gram-Negative Bacteria. Antimicrob Agents Chemother 2021; 65:e0169020. [PMID: 33875435 PMCID: PMC8373237 DOI: 10.1128/aac.01690-20] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Antibiotic-resistant bacteria are an emerging global health threat. New antimicrobials are urgently needed. The injectisome type III secretion system (T3SS), required by dozens of Gram-negative bacteria for virulence but largely absent from nonpathogenic bacteria, is an attractive antimicrobial target. We previously identified synthetic cyclic peptomers, inspired by the natural product phepropeptin D, that inhibit protein secretion through the Yersinia Ysc and Pseudomonas aeruginosa Psc T3SSs but do not inhibit bacterial growth. Here, we describe the identification of an isomer, 4EpDN, that is 2-fold more potent (50% inhibitory concentration [IC50] of 4 μM) than its parental compound. Furthermore, 4EpDN inhibited the Yersinia Ysa and the Salmonella SPI-1 T3SSs, suggesting that this cyclic peptomer has broad efficacy against evolutionarily distant injectisome T3SSs. Indeed, 4EpDN strongly inhibited intracellular growth of Chlamydia trachomatis in HeLa cells, which requires the T3SS. 4EpDN did not inhibit the unrelated twin arginine translocation (Tat) system, nor did it impact T3SS gene transcription. Moreover, although the injectisome and flagellar T3SSs are evolutionarily and structurally related, the 4EpDN cyclic peptomer did not inhibit secretion of substrates through the Salmonella flagellar T3SS, indicating that cyclic peptomers broadly but specifically target the injectisome T3SS. 4EpDN reduced the number of T3SS needles detected on the surface of Yersinia pseudotuberculosis as detected by microscopy. Collectively, these data suggest that cyclic peptomers specifically inhibit the injectisome T3SS from a variety of Gram-negative bacteria, possibly by preventing complete T3SS assembly.
Collapse
|
9
|
Interactions between Salmonella and host macrophages - Dissecting NF-κB signaling pathway responses. Microb Pathog 2021; 154:104846. [PMID: 33711426 DOI: 10.1016/j.micpath.2021.104846] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 12/18/2020] [Accepted: 02/22/2021] [Indexed: 01/07/2023]
Abstract
Salmonella not only invades host cells, but also replicates intracellularly to cause a range of diseases, including gastroenteritis and systemic infections such as typhoid fever. The body's first line of defense against pathogens is the innate immune response system that can protect against Salmonella invasion and replication. Nuclear factor κB (NF-κB) is an important transcriptional regulator that plays an important role in host inflammatory responses to pathogens. Both the canonical and non-canonical NF-κB signaling pathways are activated by Salmonella in many different ways through its virulence factors, leading to the release of inflammatory factors and the activation of inflammatory responses in mammalian hosts. Equally, Salmonella, as an enteropathogen, has accordingly evolved strategies to disturb NF-κB activation, such as secreting some effector proteins by type III secretion systems as well as inducing host cells to express NF-κB pathway inhibitors, allowing it to colonize and persistently infect the hosts. This review focuses on how Salmonella activates NF-κB signaling pathway and the strategies used by Salmonella to interfere with the NF-κB pathway activation.
Collapse
|
10
|
J Barton A, Hill J, J Blohmke C, J Pollard A. Host restriction, pathogenesis and chronic carriage of typhoidal Salmonella. FEMS Microbiol Rev 2021; 45:6159486. [PMID: 33733659 PMCID: PMC8498562 DOI: 10.1093/femsre/fuab014] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 03/03/2021] [Indexed: 12/16/2022] Open
Abstract
While conjugate vaccines against typhoid fever have recently been recommended by the World Health Organization for deployment, the lack of a vaccine against paratyphoid, multidrug resistance and chronic carriage all present challenges for the elimination of enteric fever. In the past decade, the development of in vitro and human challenge models has resulted in major advances in our understanding of enteric fever pathogenesis. In this review, we summarise these advances, outlining mechanisms of host restriction, intestinal invasion, interactions with innate immunity and chronic carriage, and discuss how this knowledge may progress future vaccines and antimicrobials.
Collapse
Affiliation(s)
- Amber J Barton
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford OX3 7LE, UK.,National Institute for Health Research (NIHR) Oxford Biomedical Research Centre, Oxford OX4 2PG, UK.,Department of Clinical Research, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London WC1E 7HT, UK
| | - Jennifer Hill
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford OX3 7LE, UK.,National Institute for Health Research (NIHR) Oxford Biomedical Research Centre, Oxford OX4 2PG, UK
| | - Christoph J Blohmke
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford OX3 7LE, UK.,National Institute for Health Research (NIHR) Oxford Biomedical Research Centre, Oxford OX4 2PG, UK
| | - Andrew J Pollard
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford OX3 7LE, UK.,National Institute for Health Research (NIHR) Oxford Biomedical Research Centre, Oxford OX4 2PG, UK
| |
Collapse
|
11
|
Mylona E, Sanchez-Garrido J, Hoang Thu TN, Dongol S, Karkey A, Baker S, Shenoy AR, Frankel G. Very long O-antigen chains of Salmonella Paratyphi A inhibit inflammasome activation and pyroptotic cell death. Cell Microbiol 2021; 23:e13306. [PMID: 33355403 PMCID: PMC8609438 DOI: 10.1111/cmi.13306] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 12/02/2020] [Accepted: 12/18/2020] [Indexed: 11/28/2022]
Abstract
Salmonella Paratyphi A (SPtA) remains one of the leading causes of enteric (typhoid) fever. Yet, despite the recent increased rate of isolation from patients in Asia, our understanding of its pathogenesis is incomplete. Here we investigated inflammasome activation in human macrophages infected with SPtA. We found that SPtA induces GSDMD‐mediated pyroptosis via activation of caspase‐1, caspase‐4 and caspase‐8. Although we observed no cell death in the absence of a functional Salmonella pathogenicity island‐1 (SPI‐1) injectisome, HilA‐mediated overexpression of the SPI‐1 regulon enhances pyroptosis. SPtA expresses FepE, an LPS O‐antigen length regulator, which induces the production of very long O‐antigen chains. Using a ΔfepE mutant we established that the very long O‐antigen chains interfere with bacterial interactions with epithelial cells and impair inflammasome‐mediated macrophage cell death. Salmonella Typhimurium (STm) serovar has a lower FepE expression than SPtA, and triggers higher pyroptosis, conversely, increasing FepE expression in STm reduced pyroptosis. These results suggest that differential expression of FepE results in serovar‐specific inflammasome modulation, which mirrors the pro‐ and anti‐inflammatory strategies employed by STm and SPtA, respectively. Our studies point towards distinct mechanisms of virulence of SPtA, whereby it attenuates inflammasome‐mediated detection through the elaboration of very long LPS O‐polysaccharides.
Collapse
Affiliation(s)
- Elli Mylona
- Department of Life Sciences, MRC Centre for Molecular Bacteriology and Infection, Imperial College London, London, UK.,Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Department of Medicine, University of Cambridge, Cambridge, UK
| | - Julia Sanchez-Garrido
- Department of Life Sciences, MRC Centre for Molecular Bacteriology and Infection, Imperial College London, London, UK
| | - Trang Nguyen Hoang Thu
- Oxford University Clinical Research Unit, The Hospital for Tropical Diseases, Ho Chi Minh City, Vietnam
| | - Sabina Dongol
- Oxford University Clinical Research Unit, Patan Academy of Health Sciences, Kathmandu, Nepal
| | - Abhilasha Karkey
- Oxford University Clinical Research Unit, Patan Academy of Health Sciences, Kathmandu, Nepal
| | - Stephen Baker
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Department of Medicine, University of Cambridge, Cambridge, UK
| | - Avinash R Shenoy
- Department of Infectious Disease, MRC Centre for Molecular Bacteriology and Infection, Imperial College London, London, UK
| | - Gad Frankel
- Department of Life Sciences, MRC Centre for Molecular Bacteriology and Infection, Imperial College London, London, UK
| |
Collapse
|
12
|
Pan P, Zou F, He C, He Q, Yin J. Characterization and protective efficacy of a sptP mutant of Salmonella Paratyphi A. IMMUNITY INFLAMMATION AND DISEASE 2020; 8:774-781. [PMID: 33135379 PMCID: PMC7654428 DOI: 10.1002/iid3.369] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Revised: 10/15/2020] [Accepted: 10/21/2020] [Indexed: 01/24/2023]
Abstract
Background Salmonella Paratyphi A causes paratyphoid A, a severe systemic disease of people and remains a major public health problem in many parts of the world. In the interest of researching the roles of sptP on Salmonella Paratyphi A and developing a live‐attenuated vaccine candidate, an sptP mutant of Salmonella Paratyphi A SPA017 (SPA017ΔsptP) was constructed, and then its characterization, immunogenicity, and protective ability were evaluated. Results The deletion of sptP had no effect on growth and biochemical properties. Adhesion and invasion assays showed that the lack of sptP did not affect the adhesion of Salmonella Paratyphi A, but the invasive ability of the mutant strain was significantly decreased, the half‐lethal dose (LD50) of the mutant strain was 1.43 × 104 times of the parent strain in intraperitoneally injected mice. Single intraperitoneal vaccination with SPA017ΔsptP (1 × 105 CFU) in mice did not affect the body weight or elicit clinical symptoms relative to the control group, SPA017ΔsptP bacteria were isolated from livers and spleens of vaccinated mice at 14 days postvaccination. Notably, specific humoral and cellular immune responses were significantly induced. The protective assessment showed that the mutant strain could provide high‐level protection against subsequent challenge with the wild‐type SPA017 strain. Conclusions These results demonstrated that SptP plays an essential role in the pathogenicity of Salmonella Paratyphi A, and Salmonella Paratyphi A lacking sptP is immunogenic and protective in mice.
Collapse
Affiliation(s)
- Pengtao Pan
- Medical College, Xinxiang University, Xinxiang, China
| | - Fanyu Zou
- Medical College, Xinxiang University, Xinxiang, China
| | - Chuanshan He
- Second Hospital, Shanxi Medical University, Taiyuan, China
| | - Qunli He
- Medical College, Zhengzhou University of Industrial Technology, Zhengzhou, China
| | - Junlei Yin
- Medical College, Xinxiang University, Xinxiang, China
| |
Collapse
|
13
|
Protection conferred by typhoid fever against recurrent typhoid fever in urban Kolkata. PLoS Negl Trop Dis 2020; 14:e0008530. [PMID: 32804950 PMCID: PMC7430703 DOI: 10.1371/journal.pntd.0008530] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 06/27/2020] [Indexed: 11/19/2022] Open
Abstract
We evaluated the protection conferred by a first documented visit for clinical care of typhoid fever against recurrent typhoid fever prompting a visit. This study takes advantage of multi-year follow-up of a population with endemic typhoid participating in a cluster-randomized control trial of Vi capsular polysaccharide typhoid vaccine in Kolkata, India. A population of 70,566 individuals, of whom 37,673 were vaccinated with one dose of either Vi vaccine or a control (Hepatitis A) vaccine, were observed for four years. Surveillance detected 315 first typhoid visits, among whom 4 developed subsequent typhoid, 3 due to reinfection, defined using genomic criteria and corresponding to -124% (95% CI: -599, 28) protection by the initial illness. Point estimates of protection conferred by an initial illness were negative or negligible in both vaccinated and non-vaccinated subjects, though confidence intervals around the point estimates were wide. These data provide little support for a protective immunizing effect of clinically treated typhoid illness, though modest levels of protection cannot be excluded.
Collapse
|
14
|
Aviv G, Cornelius A, Davidovich M, Cohen H, Suwandi A, Galeev A, Steck N, Azriel S, Rokney A, Valinsky L, Rahav G, Grassl GA, Gal-Mor O. Differences in the expression of SPI-1 genes pathogenicity and epidemiology between the emerging Salmonella enterica serovar Infantis and the model Salmonella enterica serovar Typhimurium. J Infect Dis 2020; 220:1071-1081. [PMID: 31062854 DOI: 10.1093/infdis/jiz235] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2018] [Accepted: 05/06/2019] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND Salmonella enterica serovar Infantis (S. Infantis) is one of the ubiquitous serovars of the bacterial pathogen S. enterica and recently has been emerging in many countries worldwide. Nonetheless, not much is known about its epidemiology, host adaptation, and virulence. METHODS Epidemiological and molecular approaches were used together with tissue-culture and mouse models to conduct phenotypic comparison with the model S. enterica serovar Typhimurium. RESULTS We show that S. Infantis is more frequently associated with infections in infants <2 years old and prone to cause significantly less invasive infections than serovar Typhimurium. Moreover, although S. Infantis adheres better to host cells and highly colonizes mouse intestines soon after infection, it is significantly less invasive and induces much lower inflammation and disease in vivo than S. Typhimurium. These differences were associated with lower expression of Salmonella pathogenicity island (SPI) 1 genes in S. Infantis than in S. Typhimurium. CONCLUSIONS Our results demonstrate previously unknown differences in the epidemiology, virulence pathway expression, and pathogenicity between two highly abundant Salmonella serovars and suggest that native variation in the expression of the SPI-1 regulon is likely to contribute to epidemiological and virulence variation between genetically similar nontyphoidal Salmonella serovars.
Collapse
Affiliation(s)
- Gili Aviv
- The Infectious Diseases Research Laboratory, Sheba Medical Center, Tel-Hashomer.,Department of Clinical Microbiology and Immunology, Jerusalem, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Jerusalem, Israel
| | | | | | - Helit Cohen
- The Infectious Diseases Research Laboratory, Sheba Medical Center, Tel-Hashomer
| | - Abdulhadi Suwandi
- Institute for Medical Microbiology and Hospital Epidemiology, Hannover Medical School and German Center for Infection Research, Partner Site Hannover-Braunschweig
| | - Alibek Galeev
- Institute for Medical Microbiology and Hospital Epidemiology, Hannover Medical School and German Center for Infection Research, Partner Site Hannover-Braunschweig
| | | | - Shalhevet Azriel
- The Infectious Diseases Research Laboratory, Sheba Medical Center, Tel-Hashomer
| | - Assaf Rokney
- Central Laboratories, Ministry of Health, Jerusalem, Israel
| | - Lea Valinsky
- Central Laboratories, Ministry of Health, Jerusalem, Israel
| | - Galia Rahav
- The Infectious Diseases Research Laboratory, Sheba Medical Center, Tel-Hashomer.,Sackler Faculty of Medicine, Tel Aviv University, Jerusalem, Israel
| | - Guntram A Grassl
- Research Center Borstel, Germany.,Institute for Medical Microbiology and Hospital Epidemiology, Hannover Medical School and German Center for Infection Research, Partner Site Hannover-Braunschweig
| | - Ohad Gal-Mor
- The Infectious Diseases Research Laboratory, Sheba Medical Center, Tel-Hashomer.,Department of Clinical Microbiology and Immunology, Jerusalem, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Jerusalem, Israel
| |
Collapse
|
15
|
Biswas K, Ilyas H, Datta A, Bhunia A. NMR Assisted Antimicrobial Peptide Designing: Structure Based Modifications and Functional Correlation of a Designed Peptide VG16KRKP. Curr Med Chem 2020; 27:1387-1404. [PMID: 31232231 DOI: 10.2174/0929867326666190624090817] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 05/16/2019] [Accepted: 05/29/2019] [Indexed: 01/08/2023]
Abstract
Antimicrobial Peptides (AMPs), within their realm incorporate a diverse group of structurally and functionally varied peptides, playing crucial roles in innate immunity. Over the last few decades, the field of AMP has seen a huge upsurge, mainly owing to the generation of the so-called drug resistant 'superbugs' as well as limitations associated with the existing antimicrobial agents. Due to their resilient biological properties, AMPs can very well form the sustainable alternative for nextgeneration therapeutic agents. Certain drawbacks associated with existing AMPs are, however, issues of major concern, circumventing which are imperative. These limitations mainly include proteolytic cleavage and hence poor stability inside the biological systems, reduced activity due to inadequate interaction with the microbial membrane, and ineffectiveness because of inappropriate delivery among others. In this context, the application of naturally occurring AMPs as an efficient prototype for generating various synthetic and designed counterparts has evolved as a new avenue in peptide-based therapy. Such designing approaches help to overcome the drawbacks of the parent AMPs while retaining the inherent activity. In this review, we summarize some of the basic NMR structure based approaches and techniques which aid in improving the activity of AMPs, using the example of a 16-residue dengue virus fusion protein derived peptide, VG16KRKP. Using first principle based designing technique and high resolution NMR-based structure characterization we validate different types of modifications of VG16KRKP, highlighting key motifs, which optimize its activity. The approaches and designing techniques presented can support our peers in their drug development work.
Collapse
Affiliation(s)
- Karishma Biswas
- Department of Biophysics, Bose Institute, P-1/12 CIT Scheme VII(M), Kolkata 700054, India
| | - Humaira Ilyas
- Department of Biophysics, Bose Institute, P-1/12 CIT Scheme VII(M), Kolkata 700054, India
| | - Aritreyee Datta
- Department of Biophysics, Bose Institute, P-1/12 CIT Scheme VII(M), Kolkata 700054, India
| | - Anirban Bhunia
- Department of Biophysics, Bose Institute, P-1/12 CIT Scheme VII(M), Kolkata 700054, India
| |
Collapse
|
16
|
Verma S, Senger S, Cherayil BJ, Faherty CS. Spheres of Influence: Insights into Salmonella Pathogenesis from Intestinal Organoids. Microorganisms 2020; 8:microorganisms8040504. [PMID: 32244707 PMCID: PMC7232497 DOI: 10.3390/microorganisms8040504] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 03/27/2020] [Accepted: 03/28/2020] [Indexed: 12/19/2022] Open
Abstract
The molecular complexity of host-pathogen interactions remains poorly understood in many infectious diseases, particularly in humans due to the limited availability of reliable and specific experimental models. To bridge the gap between classical two-dimensional culture systems, which often involve transformed cell lines that may not have all the physiologic properties of primary cells, and in vivo animal studies, researchers have developed the organoid model system. Organoids are complex three-dimensional structures that are generated in vitro from primary cells and can recapitulate key in vivo properties of an organ such as structural organization, multicellularity, and function. In this review, we discuss how organoids have been deployed in exploring Salmonella infection in mice and humans. In addition, we summarize the recent advancements that hold promise to elevate our understanding of the interactions and crosstalk between multiple cell types and the microbiota with Salmonella. These models have the potential for improving clinical outcomes and future prophylactic and therapeutic intervention strategies.
Collapse
Affiliation(s)
- Smriti Verma
- Mucosal Immunology and Biology Research Center, Division of Pediatric Gastroenterology and Nutrition, Massachusetts General Hospital, Charlestown Navy Yard, Boston, 02129 MA, USA; (S.S.); (B.J.C.); (C.S.F.)
- Harvard Medical School, Boston, 02115 MA, USA
- Correspondence: ; Tel.: +1-617-726-7991
| | - Stefania Senger
- Mucosal Immunology and Biology Research Center, Division of Pediatric Gastroenterology and Nutrition, Massachusetts General Hospital, Charlestown Navy Yard, Boston, 02129 MA, USA; (S.S.); (B.J.C.); (C.S.F.)
- Harvard Medical School, Boston, 02115 MA, USA
| | - Bobby J. Cherayil
- Mucosal Immunology and Biology Research Center, Division of Pediatric Gastroenterology and Nutrition, Massachusetts General Hospital, Charlestown Navy Yard, Boston, 02129 MA, USA; (S.S.); (B.J.C.); (C.S.F.)
- Harvard Medical School, Boston, 02115 MA, USA
| | - Christina S. Faherty
- Mucosal Immunology and Biology Research Center, Division of Pediatric Gastroenterology and Nutrition, Massachusetts General Hospital, Charlestown Navy Yard, Boston, 02129 MA, USA; (S.S.); (B.J.C.); (C.S.F.)
- Harvard Medical School, Boston, 02115 MA, USA
| |
Collapse
|
17
|
Lv Q, Li S, Wei H, Wen Z, Wang Y, Tang T, Wang J, Xia L, Deng X. Identification of the natural product paeonol derived from peony bark as an inhibitor of the Salmonella enterica serovar Typhimurium type III secretion system. Appl Microbiol Biotechnol 2020; 104:1673-1682. [PMID: 31897522 DOI: 10.1007/s00253-019-10290-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 11/20/2019] [Accepted: 11/28/2019] [Indexed: 12/18/2022]
Abstract
Salmonella enterica serovar Typhimurium (S. Typhimurium) is an important zoonotic pathogen in public health and food safety. The type III secretion system (T3SS) encoded by Salmonella pathogenicity island (SPI) is a sophisticated molecular machine that facilitates active invasion, intracellular replication, and host inflammation. Due to increasing antibiotic resistance, new therapeutic strategies that target the Salmonella T3SS have received considerable attention. In this study, paeonol was identified as an inhibitor of the S. Typhimurium T3SS. Paeonol significantly blocked the translocation of SipA into host cells and suppressed the expression of effector proteins without affecting bacterial growth in the effective concentration range. Additionally, S. Typhimurium-mediated cell injury and invasion levels were significantly reduced after treatment with paeonol, without cytotoxicity. Most importantly, the comprehensive protective effect of paeonol was confirmed in an S. Typhimurium mouse infection model. Preliminary mechanistic studies suggest that paeonol inhibits the expression of effector proteins by reducing the transcription level of the SPI-1 regulatory pathway gene hilA. This work provides proof that paeonol could be used as a potential drug to treat infections caused by Salmonella.
Collapse
Affiliation(s)
- Qianghua Lv
- Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, 130062, China
| | - Shufang Li
- Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, 130062, China
| | - Hanlu Wei
- Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, 130062, China
| | - Zhongmei Wen
- Department of Respiratory Medicine, The First Hospital of Jilin University, Jilin University, Changchun, 130021, Jilin, China
| | - Yanling Wang
- Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, 130062, China.,Qingdao Vland Biological Limited Co., Ltd., Qingdao, 266001, Shandong, China
| | - Tianzhong Tang
- Hubei Wudang Animal Pharmaceutical Co., Ltd., Shiyan, 442100, Hubei, China
| | - Jianfeng Wang
- Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, 130062, China
| | - Lining Xia
- College of Veterinary Medicine, Xinjiang Agricultural University, Urumqi, 830052, China.
| | - Xuming Deng
- Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun, 130062, China.
| |
Collapse
|
18
|
Berri M, Hogan D, Saade G, Roche S, Velge P, Virlogeux-Payant I, Meurens F. IPEC-1 variable immune response to different serovars of Salmonella enterica subsp. enterica. Vet Immunol Immunopathol 2019; 220:109989. [PMID: 31841890 DOI: 10.1016/j.vetimm.2019.109989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 11/14/2019] [Accepted: 12/01/2019] [Indexed: 11/16/2022]
Abstract
Salmonella is a genus of Gram-negative bacteria in the Enterobacteriaceae family causing various illnesses. The ability of the different serovars of Salmonella enterica subsp. enterica to infect a host and to induce pathology relies in part on their cellular and molecular interactions with the intestinal epithelium. In the current study, an in vitro approach using non-polarized or polarized IPEC-1 porcine intestinal epithelial cells were used in order to assess the relation between adhesion, invasion, and induction of the immune response as a function of the serotype of Salmonella. Five serovars, Choleraesuis (host-adapted), Typhimurium (ubiquitous), Typhisuis (host-restricted), which are relevant for pig infection, and Dublin and Gallinarum, which are host-restricted or host-adapted, were studied. A strong variation was observed in the percentages of adhesion and invasion amongst the S. enterica serovars used to interact with the non-polarized and polarized cells. Subsequently, differences were identified between serovars in terms of immune response induced. Serovars Typhimurium and Typhisuis induced a strong innate immune response four and half hours after the beginning of cell stimulation while Choleraesuis, Gallinarum, and Dublin did not. A strong inflammatory response could limit the spread of the porcine serovars to the gut while, with a weak response, bacteria may not be constrained by the immune response enabling severe systemic diseases. Different repertoires of adhesion factors and of secreted protein effectors between Salmonella serovars interacting with IPEC-1 cells probably explains the differences in their early pathogenic behaviours.
Collapse
Affiliation(s)
- Mustapha Berri
- ISP, INRAE, Université François Rabelais de Tours, UMR 1282, 37380, Nouzilly, France
| | | | | | - Sylvie Roche
- ISP, INRAE, Université François Rabelais de Tours, UMR 1282, 37380, Nouzilly, France
| | - Philippe Velge
- ISP, INRAE, Université François Rabelais de Tours, UMR 1282, 37380, Nouzilly, France
| | | | | |
Collapse
|
19
|
Hotinger JA, May AE. Animal Models of Type III Secretion System-Mediated Pathogenesis. Pathogens 2019; 8:pathogens8040257. [PMID: 31766664 PMCID: PMC6963218 DOI: 10.3390/pathogens8040257] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 11/18/2019] [Accepted: 11/20/2019] [Indexed: 01/22/2023] Open
Abstract
The type III secretion system (T3SS) is a conserved virulence factor used by many Gram-negative pathogenic bacteria and has become an important target for anti-virulence drugs. Most T3SS inhibitors to date have been discovered using in vitro screening assays. Pharmacokinetics and other important characteristics of pharmaceuticals cannot be determined with in vitro assays alone. In vivo assays are required to study pathogens in their natural environment and are an important step in the development of new drugs and vaccines. Animal models are also required to understand whether T3SS inhibition will enable the host to clear the infection. This review covers selected animal models (mouse, rat, guinea pig, rabbit, cat, dog, pig, cattle, primates, chicken, zebrafish, nematode, wax moth, flea, fly, and amoeba), where T3SS activity and infectivity have been studied in relation to specific pathogens (Escherichia coli, Salmonella spp., Pseudomonas spp., Shigella spp., Bordetella spp., Vibrio spp., Chlamydia spp., and Yersinia spp.). These assays may be appropriate for those researching T3SS inhibition.
Collapse
|
20
|
Hiyoshi H, Wangdi T, Lock G, Saechao C, Raffatellu M, Cobb BA, Bäumler AJ. Mechanisms to Evade the Phagocyte Respiratory Burst Arose by Convergent Evolution in Typhoidal Salmonella Serovars. Cell Rep 2019; 22:1787-1797. [PMID: 29444431 PMCID: PMC5826628 DOI: 10.1016/j.celrep.2018.01.016] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2017] [Revised: 12/20/2017] [Accepted: 01/05/2018] [Indexed: 12/22/2022] Open
Abstract
Typhoid fever caused by Salmonella enterica serovar (S.) Typhi differs in its clinical presentation from gastroenteritis caused by S. Typhimurium and other non-typhoidal Salmonella serovars. The different clinical presentations are attributed in part to the virulence-associated capsular polysaccharide (Vi antigen) of S. Typhi, which prevents phagocytes from triggering a respiratory burst by preventing antibody-mediated complement activation. Paradoxically, the Vi antigen is absent from S. Paratyphi A, which causes a disease that is indistinguishable from typhoid fever. Here, we show that evasion of the phagocyte respiratory burst by S. Paratyphi A required very long O antigen chains containing the O2 antigen to inhibit antibody binding. We conclude that the ability to avoid the phagocyte respiratory burst is a property distinguishing typhoidal from non-typhoidal Salmonella serovars that was acquired by S. Typhi and S. Paratyphi A independently through convergent evolution.
Collapse
Affiliation(s)
- Hirotaka Hiyoshi
- Department of Medical Microbiology and Immunology, School of Medicine, University of California, Davis, Davis, CA 95616, USA
| | - Tamding Wangdi
- Department of Medical Microbiology and Immunology, School of Medicine, University of California, Davis, Davis, CA 95616, USA.
| | - Gabriel Lock
- Department of Medical Microbiology and Immunology, School of Medicine, University of California, Davis, Davis, CA 95616, USA
| | - Cheng Saechao
- Department of Medical Microbiology and Immunology, School of Medicine, University of California, Davis, Davis, CA 95616, USA
| | - Manuela Raffatellu
- Department of Pediatrics, School of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Brian A Cobb
- Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Andreas J Bäumler
- Department of Medical Microbiology and Immunology, School of Medicine, University of California, Davis, Davis, CA 95616, USA.
| |
Collapse
|
21
|
Urdaneta V, Casadesús J. Host-pathogen interactions in typhoid fever: the model is the message. ANNALS OF TRANSLATIONAL MEDICINE 2018; 6:S38. [PMID: 30613613 PMCID: PMC6291556 DOI: 10.21037/atm.2018.09.52] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 09/20/2018] [Indexed: 01/22/2023]
Affiliation(s)
- Verónica Urdaneta
- Departamento de Genética, Facultad de Biología, Universidad de Sevilla, Sevilla, Spain
| | - Josep Casadesús
- Departamento de Genética, Facultad de Biología, Universidad de Sevilla, Sevilla, Spain
| |
Collapse
|
22
|
Tanner JR, Kingsley RA. Evolution of Salmonella within Hosts. Trends Microbiol 2018; 26:986-998. [PMID: 29954653 PMCID: PMC6249985 DOI: 10.1016/j.tim.2018.06.001] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 05/22/2018] [Accepted: 06/01/2018] [Indexed: 11/18/2022]
Abstract
Within-host evolution has resulted in thousands of variants of Salmonella that exhibit remarkable diversity in host range and disease outcome, from broad host range to exquisite host restriction, causing gastroenteritis to disseminated disease such as typhoid fever. Within-host evolution is a continuing process driven by genomic variation that occurs during each infection, potentiating adaptation to a new niche resulting from changes in animal husbandry, the use of antimicrobials, and emergence of immune compromised populations. We discuss key advances in our understanding of the evolution of Salmonella within the host, inferred from (i) the process of host adaptation of Salmonella pathovars in the past, and (ii) direct observation of the generation of variation and selection of beneficial traits during single infections. Salmonella is a bacterial pathogen with remarkable diversity in its host range and pathogenicity due to past within-host evolution in vertebrate species that modified ancestral mechanisms of pathogenesis. Variation arising during infection includes point mutations, new genes acquired through horizontal gene transfer (HGT), deletions, and genomic rearrangements. Beneficial mutations increase in frequency within the host and, if they retain the ability to be transmitted to subsequent hosts, may become fixed in the population. Whole-genome sequencing of sequential isolates from clinical infections reveals within-host HGT and point mutations that impact therapy and clinical management. HGT is the primary mechanism for evolution in prokaryotes and is synergised by complex networks of transfer involving the microbiome. Within-host evolution of Salmonella, resulting in new pathovars, can proceed in the absence of HGT.
Collapse
Affiliation(s)
- Jennifer R Tanner
- Quadram Institute Bioscience, Norwich Research Park, Colney, Norwich, UK
| | - Robert A Kingsley
- Quadram Institute Bioscience, Norwich Research Park, Colney, Norwich, UK.
| |
Collapse
|
23
|
Hiyoshi H, Tiffany CR, Bronner DN, Bäumler AJ. Typhoidal Salmonella serovars: ecological opportunity and the evolution of a new pathovar. FEMS Microbiol Rev 2018; 42:527-541. [DOI: 10.1093/femsre/fuy024] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Accepted: 05/19/2018] [Indexed: 12/20/2022] Open
Affiliation(s)
- Hirotaka Hiyoshi
- Department of Medial Microbiology and Immunology, School of Medicine, University of California Davis, Davis, CA 95616, USA
| | - Connor R Tiffany
- Department of Medial Microbiology and Immunology, School of Medicine, University of California Davis, Davis, CA 95616, USA
| | - Denise N Bronner
- Department of Medial Microbiology and Immunology, School of Medicine, University of California Davis, Davis, CA 95616, USA
| | - Andreas J Bäumler
- Department of Medial Microbiology and Immunology, School of Medicine, University of California Davis, Davis, CA 95616, USA
| |
Collapse
|
24
|
Nickerson KP, Senger S, Zhang Y, Lima R, Patel S, Ingano L, Flavahan WA, Kumar DKV, Fraser CM, Faherty CS, Sztein MB, Fiorentino M, Fasano A. Salmonella Typhi Colonization Provokes Extensive Transcriptional Changes Aimed at Evading Host Mucosal Immune Defense During Early Infection of Human Intestinal Tissue. EBioMedicine 2018; 31:92-109. [PMID: 29735417 PMCID: PMC6013756 DOI: 10.1016/j.ebiom.2018.04.005] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 04/02/2018] [Accepted: 04/05/2018] [Indexed: 12/29/2022] Open
Abstract
Commensal microorganisms influence a variety of host functions in the gut, including immune response, glucose homeostasis, metabolic pathways and oxidative stress, among others. This study describes how Salmonella Typhi, the pathogen responsible for typhoid fever, uses similar strategies to escape immune defense responses and survive within its human host. To elucidate the early mechanisms of typhoid fever, we performed studies using healthy human intestinal tissue samples and "mini-guts," organoids grown from intestinal tissue taken from biopsy specimens. We analyzed gene expression changes in human intestinal specimens and bacterial cells both separately and after colonization. Our results showed mechanistic strategies that S. Typhi uses to rearrange the cellular machinery of the host cytoskeleton to successfully invade the intestinal epithelium, promote polarized cytokine release and evade immune system activation by downregulating genes involved in antigen sampling and presentation during infection. This work adds novel information regarding S. Typhi infection pathogenesis in humans, by replicating work shown in traditional cell models, and providing new data that can be applied to future vaccine development strategies.
Collapse
Affiliation(s)
- K P Nickerson
- Department of Pediatric Gastroenterology, Mucosal Immunology and Biology Research Center, Massachusetts General Hospital, Boston, MA, United States; Department of Pediatrics, Harvard Medical School, Harvard University, Boston, MA, United States.
| | - S Senger
- Department of Pediatric Gastroenterology, Mucosal Immunology and Biology Research Center, Massachusetts General Hospital, Boston, MA, United States; Department of Pediatrics, Harvard Medical School, Harvard University, Boston, MA, United States
| | - Y Zhang
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, United States
| | - R Lima
- Department of Pediatric Gastroenterology, Mucosal Immunology and Biology Research Center, Massachusetts General Hospital, Boston, MA, United States
| | - S Patel
- Department of Pediatric Gastroenterology, Mucosal Immunology and Biology Research Center, Massachusetts General Hospital, Boston, MA, United States
| | - L Ingano
- Department of Pediatric Gastroenterology, Mucosal Immunology and Biology Research Center, Massachusetts General Hospital, Boston, MA, United States
| | - W A Flavahan
- Department of Pathology, Massachusetts General Hospital, Boston, MA, United States
| | - D K V Kumar
- Department for the Neuroscience of Genetics and Aging, Massachusetts General Hospital, Boston, MA, United States
| | - C M Fraser
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, United States
| | - C S Faherty
- Department of Pediatric Gastroenterology, Mucosal Immunology and Biology Research Center, Massachusetts General Hospital, Boston, MA, United States; Department of Pediatrics, Harvard Medical School, Harvard University, Boston, MA, United States
| | - M B Sztein
- Center for Vaccine Development, Department of Pediatrics, University of Maryland, Baltimore, MD, United States
| | - M Fiorentino
- Department of Pediatric Gastroenterology, Mucosal Immunology and Biology Research Center, Massachusetts General Hospital, Boston, MA, United States; Department of Pediatrics, Harvard Medical School, Harvard University, Boston, MA, United States
| | - A Fasano
- Department of Pediatric Gastroenterology, Mucosal Immunology and Biology Research Center, Massachusetts General Hospital, Boston, MA, United States; Department of Pediatrics, Harvard Medical School, Harvard University, Boston, MA, United States.
| |
Collapse
|
25
|
The QseG Lipoprotein Impacts the Virulence of Enterohemorrhagic Escherichia coli and Citrobacter rodentium and Regulates Flagellar Phase Variation in Salmonella enterica Serovar Typhimurium. Infect Immun 2018; 86:IAI.00936-17. [PMID: 29358334 DOI: 10.1128/iai.00936-17] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 01/12/2018] [Indexed: 11/20/2022] Open
Abstract
The QseEF histidine kinase/response regulator system modulates expression of enterohemorrhagic Escherichia coli (EHEC) and Salmonella enterica serovar Typhimurium virulence genes in response to the host neurotransmitters epinephrine and norepinephrine. qseG, which encodes an outer membrane lipoprotein, is cotranscribed with qseEF in these enteric pathogens, but there is little knowledge of its role in virulence. Here, we found that in EHEC QseG interacts with the type III secretion system (T3SS) gate protein SepL and modulates the kinetics of attaching and effacing (AE) lesion formation on tissue-cultured cells. Moreover, an EHEC ΔqseG mutant had reduced intestinal colonization in an infant rabbit model. Additionally, in Citrobacter rodentium, an AE lesion-forming pathogen like EHEC, QseG is required for full virulence in a mouse model. In S Typhimurium, we found that QseG regulates the phase switch between the two flagellin types, FliC and FljB. In an S Typhimurium ΔqseG mutant, the phase-variable promoter for fljB is preferentially switched into the "on" position, leading to overproduction of this phase two flagellin. In infection of tissue-cultured cells, the S Typhimurium ΔqseG mutant provokes increased inflammatory cytokine production versus the wild type; in vivo, in a murine infection model, the ΔqseG strain caused a more severe inflammatory response and was attenuated versus the wild-type strain. Collectively, our findings demonstrate that QseG is important for full virulence in several enteric pathogens and controls flagellar phase variation in S Typhimurium, and they highlight both the complexity and conservation of the regulatory networks that control the virulence of enteric pathogens.
Collapse
|
26
|
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: 21] [Impact Index Per Article: 3.0] [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
|
27
|
Chowdhury R, Ilyas H, Ghosh A, Ali H, Ghorai A, Midya A, Jana NR, Das S, Bhunia A. Multivalent gold nanoparticle-peptide conjugates for targeting intracellular bacterial infections. NANOSCALE 2017; 9:14074-14093. [PMID: 28901372 DOI: 10.1039/c7nr04062h] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Although nanoparticle-tagged antimicrobal peptides have gained considerable importance in recent years, their structure-function correlation has not yet been explored. Here, we have studied the mechanism of action of a designed antimicrobial peptide, VG16KRKP (VARGWKRKCPLFGKGG), delivered via gold nanoparticle tagging against Salmonella infection by combining biological experiments with high- and low-resolution spectroscopic techniques. In comparison with the free VG16KRKP peptide or gold nanoparticle alone, the conjugated variant, Au-VG16KRKP, is non-cytotoxic to eukaryotic cells, but exhibits strong bacteriolytic activity in culture. Au-VG16KRKP can penetrate host epithelial and macrophage cells as well as interact with intracellular S. Typhi LPS under both in vitro and in vivo conditions. Treatment of mice with Au-VG16KRKP post-infection with S. Typhi resulted in reduced intracellular bacterial recovery and highly enhanced protection against S. Typhi challenge. The three-dimensional high resolution structure of nanoparticle conjugated VG16KRKP depicted the generation of a well-separated amphipathic structure with slight aggregation, responsible for the increase of the local concentration of the peptide, thus leading to potent activity. This is the first report on the structural and functional characterization of a nanoparticle conjugated synthetic antimicrobial peptide that can kill intracellular pathogens and eventually protect against S. Typhi challenge in vivo.
Collapse
Affiliation(s)
- Rimi Chowdhury
- Division of Clinical Medicine, National Institute of Cholera and Enteric Diseases, P-33 CIT Road Scheme XM, Beliaghata, Kolkata-700010, India.
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Spiga L, Winter MG, Furtado de Carvalho T, Zhu W, Hughes ER, Gillis CC, Behrendt CL, Kim J, Chessa D, Andrews-Polymenis HL, Beiting DP, Santos RL, Hooper LV, Winter SE. An Oxidative Central Metabolism Enables Salmonella to Utilize Microbiota-Derived Succinate. Cell Host Microbe 2017; 22:291-301.e6. [PMID: 28844888 DOI: 10.1016/j.chom.2017.07.018] [Citation(s) in RCA: 132] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Revised: 06/23/2017] [Accepted: 07/28/2017] [Indexed: 12/31/2022]
Abstract
The mucosal inflammatory response induced by Salmonella serovar Typhimurium creates a favorable niche for this gut pathogen. Conventional wisdom holds that S. Typhimurium undergoes an incomplete tricarboxylic acid (TCA) cycle in the anaerobic mammalian gut. One change during S. Typhimurium-induced inflammation is the production of oxidized compounds by infiltrating neutrophils. We show that inflammation-derived electron acceptors induce a complete, oxidative TCA cycle in S. Typhimurium, allowing the bacteria to compete with the microbiota for colonization. A complete TCA cycle facilitates utilization of the microbiota-derived fermentation product succinate as a carbon source. S. Typhimurium succinate utilization genes contribute to efficient colonization in conventionally raised mice, but provide no growth advantage in germ-free mice. Mono-association of gnotobiotic mice with Bacteroides, a major succinate producer, restores succinate utilization in S. Typhimurium. Thus, oxidative central metabolism enables S. Typhimurium to utilize a variety of carbon sources, including microbiota-derived succinate.
Collapse
Affiliation(s)
- Luisella Spiga
- Department of Microbiology, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Maria G Winter
- Department of Microbiology, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Tatiane Furtado de Carvalho
- Departamento de Clínica e Cirurgia Veterinárias, Escola de Veterinária, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Wenhan Zhu
- Department of Microbiology, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Elizabeth R Hughes
- Department of Microbiology, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Caroline C Gillis
- Department of Microbiology, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Cassie L Behrendt
- Department of Immunology, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jiwoong Kim
- Department of Clinical Science, Quantitative Biomedical Research Center, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Daniela Chessa
- Department of Biomedical Science, School of Medicine, University of Sassari, Sassari, Italy
| | - Helene L Andrews-Polymenis
- Department of Microbial Pathogenesis and Immunology, College of Medicine, Texas A&M University System Health Science Center, Bryan, TX 77807, USA
| | - Daniel P Beiting
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Renato L Santos
- Departamento de Clínica e Cirurgia Veterinárias, Escola de Veterinária, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Lora V Hooper
- Department of Immunology, UT Southwestern Medical Center, Dallas, TX 75390, USA; Howard Hughes Medical Institute, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Sebastian E Winter
- Department of Microbiology, UT Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
29
|
New Insights into the Roles of Long Polar Fimbriae and Stg Fimbriae in Salmonella Interactions with Enterocytes and M Cells. Infect Immun 2017. [PMID: 28630073 DOI: 10.1128/iai.00172-17] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Salmonella enterica serovar Typhi causes the systemic disease typhoid fever. After ingestion, it adheres to and invades the host epithelium while evading the host innate immune response, causing little if any inflammation. Conversely, Salmonella enterica serovar Typhimurium causes gastroenteritis in humans and thrives in the inflamed gut. Upon entering the host, S Typhimurium preferentially colonizes Peyer's patches, a lymphoid organ in which microfold cells (M cells) overlay an arrangement of B cells, T cells, and antigen-presenting cells. Both serovars can adhere to and invade M cells and enterocytes, and it has been assumed that S Typhi also preferentially targets M cells. In this study, we present data supporting the alternative hypothesis that S Typhi preferentially targets enterocytes. Using a tissue culture M cell model, we examined S Typhi strains with a deletion in the stg fimbriae. The stg deletion resulted in increased adherence to M cells and, as expected, decreased adherence to Caco-2 cells. Adherence to M cells could be further enhanced by introduction of the long polar fimbriae (Lpf), which facilitate adherence of S Typhimurium to M cells. Deletion of stg and/or introduction of lpf enhanced M cell invasion as well, leading to significant increases in secretion of interleukin 8. These results suggest that S Typhi may preferentially target enterocytes in vivo.
Collapse
|
30
|
Pérez-Morales D, Banda MM, Chau NYE, Salgado H, Martínez-Flores I, Ibarra JA, Ilyas B, Coombes BK, Bustamante VH. The transcriptional regulator SsrB is involved in a molecular switch controlling virulence lifestyles of Salmonella. PLoS Pathog 2017; 13:e1006497. [PMID: 28704543 PMCID: PMC5562331 DOI: 10.1371/journal.ppat.1006497] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Revised: 08/18/2017] [Accepted: 06/28/2017] [Indexed: 11/22/2022] Open
Abstract
The evolution of bacterial pathogenicity, heavily influenced by horizontal gene transfer, provides new virulence factors and regulatory connections that alter bacterial phenotypes. Salmonella pathogenicity islands 1 and 2 (SPI-1 and SPI-2) are chromosomal regions that were acquired at different evolutionary times and are essential for Salmonella virulence. In the intestine of mammalian hosts, Salmonella expresses the SPI-1 genes that mediate its invasion to the gut epithelium. Once inside the cells, Salmonella down-regulates the SPI-1 genes and induces the expression of the SPI-2 genes, which favor its intracellular replication. The mechanism by which the invasion machinery is deactivated following successful invasion of host cells is not known. Here, we show that the SPI-2 encoded transcriptional regulator SsrB, which positively controls SPI-2, acts as a dual regulator that represses expression of SPI-1 during intracellular stages of infection. The mechanism of this SPI-1 repression by SsrB was direct and acts upon the hilD and hilA regulatory genes. The phenotypic effect of this molecular switch activity was a significant reduction in invasion ability of S. enterica serovar Typhimurium while promoting the expression of genes required for intracellular survival. During mouse infections, Salmonella mutants lacking SsrB had high levels of hilA (SPI-1) transcriptional activity whereas introducing a constitutively active SsrB led to significant hilA repression. Thus, our results reveal a novel SsrB-mediated mechanism of transcriptional crosstalk between SPI-1 and SPI-2 that helps Salmonella transition to the intracellular lifestyle. Salmonella infect humans and a wide range of mammalian hosts. Successful infection requires the bacteria to sense their surroundings and regulate gene expression in a way that maximizes fitness in that particular environment. The two major lifestyles of Salmonella include extracellular stages and intracellular stages of host cell infection; however, the molecular mechanisms of how Salmonella transitions between these two lifestyles are not completely understood. Here we show that the transcriptional regulator SsrB functions in a dual capacity, activating genes required for intracellular survival while simultaneously repressing genes needed for extracellular stages of infection. Our data highlight how regulatory crosstalk is selective during infection, presumably because it helps facilitate rapid transitions in bacterial lifestyles that ultimately promote bacterial survival and replication.
Collapse
Affiliation(s)
- Deyanira Pérez-Morales
- Departamento de Microbiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, México
| | - María M. Banda
- Departamento de Microbiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, México
| | - N. Y Elizabeth Chau
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| | - Heladia Salgado
- Programa de Genómica Computacional, Centro de Ciencias Genómicas, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, México
| | - Irma Martínez-Flores
- Programa de Genómica Computacional, Centro de Ciencias Genómicas, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, México
| | - J. Antonio Ibarra
- Departamento de Microbiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México, México
| | - Bushra Ilyas
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| | - Brian K. Coombes
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| | - Víctor H. Bustamante
- Departamento de Microbiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, México
- * E-mail:
| |
Collapse
|
31
|
Eisenreich W, Rudel T, Heesemann J, Goebel W. To Eat and to Be Eaten: Mutual Metabolic Adaptations of Immune Cells and Intracellular Bacterial Pathogens upon Infection. Front Cell Infect Microbiol 2017; 7:316. [PMID: 28752080 PMCID: PMC5508010 DOI: 10.3389/fcimb.2017.00316] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 06/26/2017] [Indexed: 12/11/2022] Open
Abstract
Intracellular bacterial pathogens (IBPs) invade and replicate in different cell types including immune cells, in particular of the innate immune system (IIS) during infection in the acute phase. However, immune cells primarily function as essential players in the highly effective and integrated host defense systems comprising the IIS and the adaptive immune system (AIS), which cooperatively protect the host against invading microbes including IBPs. As countermeasures, the bacterial pathogens (and in particular the IBPs) have developed strategies to evade or reprogram the IIS at various steps. The intracellular replication capacity and the anti-immune defense responses of the IBP's as well as the specific antimicrobial responses of the immune cells of the innate and the AIS depend on specific metabolic programs of the IBPs and their host cells. The metabolic programs of the immune cells supporting or counteracting replication of the IBPs appear to be mutually exclusive. Indeed, recent studies show that upon interaction of naïve, metabolically quiescent immune cells with IBPs, different metabolic activation processes occur which may result in the provision of a survival and replication niche for the pathogen or its eradication. It is therefore likely that within a possible host cell population subsets exist that are metabolically programmed for pro- or anti-microbial conditions. These metabolic programs may be triggered by the interactions between different bacterial agonistic components and host cell receptors. In this review, we summarize the current status in the field and discuss metabolic adaptation processes within immune cells of the IIS and the IBPs that support or restrict the intracellular replication of the pathogens.
Collapse
Affiliation(s)
- Wolfgang Eisenreich
- Department of Chemistry, Chair of Biochemistry, Technische Universität MünchenGarching, Germany
| | - Thomas Rudel
- Department of Microbiology, Biocenter, University of WürzburgWürzburg, Germany
| | - Jürgen Heesemann
- Max von Pettenkofer-Institute, Chair of Medical Microbiology and Hospital Epidemiology, Ludwig Maximilian University of MunichMünchen, Germany
| | - Werner Goebel
- Max von Pettenkofer-Institute, Chair of Medical Microbiology and Hospital Epidemiology, Ludwig Maximilian University of MunichMünchen, Germany
| |
Collapse
|
32
|
Fresnay S, McArthur MA, Magder LS, Darton TC, Jones C, Waddington CS, Blohmke CJ, Angus B, Levine MM, Pollard AJ, Sztein MB. Importance of Salmonella Typhi-Responsive CD8+ T Cell Immunity in a Human Typhoid Fever Challenge Model. Front Immunol 2017; 8:208. [PMID: 28303138 PMCID: PMC5332428 DOI: 10.3389/fimmu.2017.00208] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Accepted: 02/15/2017] [Indexed: 01/25/2023] Open
Abstract
Typhoid fever, caused by the human-restricted organism Salmonella enterica serovar Typhi (S. Typhi), constitutes a major global health problem. The development of improved attenuated vaccines is pressing, but delayed by the lack of appropriate preclinical models. Herein, we report that high levels of S. Typhi-responsive CD8+ T cells at baseline significantly correlate with an increased risk of disease in humans challenged with a high dose (~104 CFU) wild-type S. Typhi. Typhoid fever development was associated with higher multifunctional S. Typhi-responsive CD8+ T effector memory cells at baseline. Early decreases of these cells in circulation following challenge were observed in both S. Typhi-responsive integrin α4β7− and integrin α4β7+ CD8+ T effector memory (TEM) cells, suggesting their potential to home to both mucosal and extra-intestinal sites. Participants with higher baseline levels of S. Typhi-responsive CD8+ T memory cells had a higher risk of acquiring disease, but among those who acquired disease, those with a higher baseline responses took longer to develop disease. In contrast, protection against disease was associated with low or absent S. Typhi-responsive T cells at baseline and no changes in circulation following challenge. These data highlight the importance of pre-existing S. Typhi-responsive immunity in predicting clinical outcome following infection with wild-type S. Typhi and provide novel insights into the complex mechanisms involved in protective immunity to natural infection in a stringent human model with a high challenge dose. They also contribute important information on the immunological responses to be assessed in the appraisal and selection of new generation typhoid vaccines.
Collapse
Affiliation(s)
- Stephanie Fresnay
- Center for Vaccine Development, University of Maryland School of Medicine , Baltimore, MD , USA
| | - Monica A McArthur
- Center for Vaccine Development, University of Maryland School of Medicine , Baltimore, MD , USA
| | - Laurence S Magder
- Department of Epidemiology and Public Health, University of Maryland School of Medicine , Baltimore, MD , USA
| | - Thomas C Darton
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, NIHR Oxford Biomedical Research Centre , Oxford , UK
| | - Claire Jones
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, NIHR Oxford Biomedical Research Centre , Oxford , UK
| | - Claire S Waddington
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, NIHR Oxford Biomedical Research Centre , Oxford , UK
| | - Christoph J Blohmke
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, NIHR Oxford Biomedical Research Centre , Oxford , UK
| | - Brian Angus
- Nuffield Department of Medicine, University of Oxford , Oxford , UK
| | - Myron M Levine
- Center for Vaccine Development, University of Maryland School of Medicine , Baltimore, MD , USA
| | - Andrew J Pollard
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, NIHR Oxford Biomedical Research Centre , Oxford , UK
| | - Marcelo B Sztein
- Center for Vaccine Development, University of Maryland School of Medicine , Baltimore, MD , USA
| |
Collapse
|
33
|
Costa LF, Mol JPS, Silva APC, Macêdo AA, Silva TMA, Alves GES, Winter S, Winter MG, Velazquez EM, Byndloss MX, Bäumler AJ, Tsolis RM, Paixão TA, Santos RL. Iron acquisition pathways and colonization of the inflamed intestine by Salmonella enterica serovar Typhimurium. Int J Med Microbiol 2016; 306:604-610. [PMID: 27760693 PMCID: PMC5140723 DOI: 10.1016/j.ijmm.2016.10.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Revised: 10/11/2016] [Accepted: 10/12/2016] [Indexed: 12/11/2022] Open
Abstract
Salmonella enterica serotype Typhimurium is able to expand in the lumen of the inflamed intestine through mechanisms that have not been fully resolved. Here we utilized streptomycin-pretreated mice and dextran sodium sulfate (DSS)-treated mice to investigate how pathways for S. Typhimurium iron acquisition contribute to pathogen expansion in the inflamed intestine. Competitive infection with an iron uptake-proficient S. Typhimurium strain and mutant strains lacking tonB feoB, feoB, tonB or iroN in streptomycin pretreated mice demonstrated that ferric iron uptake requiring IroN and TonB conferred a fitness advantage during growth in the inflamed intestine. However, the fitness advantage conferred by ferrous iron uptake mechanisms was independent of inflammation and was only apparent in models where the normal microbiota composition had been disrupted by antibiotic treatment.
Collapse
Affiliation(s)
- Luciana F Costa
- Departamento de Patologia, Instituto de Ciências Biológicas da Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Juliana P S Mol
- Departamento de Clínica e Cirurgia Veterinária, Escola de Veterinária da Universidade Federal Minas Gerais, Belo Horizonte, MG, Brazil
| | - Ana Patricia C Silva
- Departamento de Clínica e Cirurgia Veterinária, Escola de Veterinária da Universidade Federal Minas Gerais, Belo Horizonte, MG, Brazil
| | - Auricélio A Macêdo
- Departamento de Clínica e Cirurgia Veterinária, Escola de Veterinária da Universidade Federal Minas Gerais, Belo Horizonte, MG, Brazil
| | - Teane M A Silva
- Departamento de Patologia, Instituto de Ciências Biológicas da Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Geraldo E S Alves
- Departamento de Clínica e Cirurgia Veterinária, Escola de Veterinária da Universidade Federal Minas Gerais, Belo Horizonte, MG, Brazil
| | - Sebastian Winter
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Maria G Winter
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Eric M Velazquez
- Department of Medical Microbiology and Immunology, University of California at Davis, Davis, CA, USA
| | - Mariana X Byndloss
- Department of Medical Microbiology and Immunology, University of California at Davis, Davis, CA, USA
| | - Andreas J Bäumler
- Department of Medical Microbiology and Immunology, University of California at Davis, Davis, CA, USA
| | - Renée M Tsolis
- Department of Medical Microbiology and Immunology, University of California at Davis, Davis, CA, USA.
| | - Tatiane A Paixão
- Departamento de Patologia, Instituto de Ciências Biológicas da Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Renato L Santos
- Departamento de Clínica e Cirurgia Veterinária, Escola de Veterinária da Universidade Federal Minas Gerais, Belo Horizonte, MG, Brazil.
| |
Collapse
|
34
|
Zhang H, Zhu Y, Xie X, Wang M, Du H, Xu S, Zhang Y, Gong M, Ni B, Xu H, Huang X. Identification and Characterization of a Gene stp17 Located on the Linear Plasmid pBSSB1 as an Enhanced Gene of Growth and Motility in Salmonella enterica Serovar Typhi. Front Cell Infect Microbiol 2016; 6:110. [PMID: 27761429 PMCID: PMC5050219 DOI: 10.3389/fcimb.2016.00110] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2016] [Accepted: 09/08/2016] [Indexed: 11/13/2022] Open
Abstract
The linear plasmid pBSSB1 mediates the flagellar phase variation in H:z66 positive Salmonella enterica serovar Typhi (S. Typhi). The gene named stp17 (S. Typhi plasmid number 17 gene) is located on pBSSB1 and encodes the protein STP17. The expression pattern at the protein-level and function of STP17 remains unknown. In this study, the recombinant protein STP17His6 was expressed, purified and used to prepare the polyclonal anti-STP17 antibody. We detected protein-level expression of stp17 in S. Typhi and further investigated the protein expression characteristics of stp17 in different growth phases by western blot analysis. The effects of STP17 on bacterial growth and motility were analyzed. In addition, the structure of STP17 was predicted and the active site of STP17 was identified by site-directed mutagenesis. The results showed that STP17 was expressed stably in the wild type strain of S. Typhi. STP17 expression at the protein level peaks when cultures reach an OD600 value of 1.2. The growth rate and motility of the Δstp17 strain were significantly decreased compared with the wild type strain (P < 0.05) and this phenotype was restored in the stp17 complementary strain. Moreover, the growth rate and motility of the stp17 over-expression strain was greater than the wild type strain. STP17 contains nine Helix segments, six Stand segments and some Coil segments in the secondary structural level. The top-ranked 3-D structure of STP17 predicted by I-TASSER contains a putative ATPase domain and the amino acid residues of GLY16, GLY19, LYS20, ASN133, LYS157, and LYS158 may be the active site residues of STP17. Finally, STP17 was able to catalyze the ATP to ADP reaction, suggesting that STP17 may be an ATPase. To our knowledge, this is the first report describing the protein expression characteristics of STP17 in S. Typhi, showing that STP17 promotes bacterial growth and motility, which may be associated with its potential ATPase activity.
Collapse
Affiliation(s)
- Haifang Zhang
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow UniversitySuzhou, China
- School of Medicine, Jiangsu UniversityZhenjiang, China
| | - Yunxia Zhu
- School of Medicine, Jiangsu UniversityZhenjiang, China
- Department of Clinical Laboratory, Shanghai East Hospital, School of Medicine, Tongji UniversityShanghai, China
| | - Xiaofang Xie
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow UniversitySuzhou, China
| | - Min Wang
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow UniversitySuzhou, China
| | - Hong Du
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow UniversitySuzhou, China
| | - Shungao Xu
- School of Medicine, Jiangsu UniversityZhenjiang, China
| | - Ying Zhang
- School of Medicine, Jiangsu UniversityZhenjiang, China
| | - Mingyu Gong
- School of Medicine, Jiangsu UniversityZhenjiang, China
| | - Bin Ni
- School of Medicine, Jiangsu UniversityZhenjiang, China
| | - Huaxi Xu
- School of Medicine, Jiangsu UniversityZhenjiang, China
| | | |
Collapse
|
35
|
Prado Acosta M, Ruzal SM, Cordo SM. S-layer proteins from Lactobacillus sp. inhibit bacterial infection by blockage of DC-SIGN cell receptor. Int J Biol Macromol 2016; 92:998-1005. [PMID: 27498415 DOI: 10.1016/j.ijbiomac.2016.07.096] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Revised: 07/27/2016] [Accepted: 07/28/2016] [Indexed: 12/18/2022]
Abstract
Many species of Lactobacillus sp. possess Surface(s) layer proteins in their envelope. Among other important characteristics S-layer from Lactobacillus acidophilus binds to the cellular receptor DC-SIGN (Dendritic Cell-Specific Intercellular adhesion molecule-3-Grabbing Non-integrin; CD209), which is involved in adhesion and infection of several families of bacteria. In this report we investigate the activity of new S-layer proteins from the Lactobacillus family (Lactobacillus acidophilus, Lactobacillus brevis, Lactobacillus helveticus and Lactobacillus kefiri) over the infection of representative microorganisms important to human health. After the treatment of DC-SIGN expressing cells with these proteins, we were able to diminish bacterial infection by up to 79% in both gram negative and mycobacterial models. We discovered that pre-treatment of the bacteria with S-layers from Lactobacillus acidophilus and Lactobacillus brevis reduced bacteria viability but also prevent infection by the pathogenic bacteria. We also proved the importance of the glycosylation of the S-layer from Lactobacillus kefiri in the binding to the receptor and thus inhibition of infection. This novel characteristic of the S-layers proteins may contribute to the already reported pathogen exclusion activity for these Lactobacillus probiotic strains; and might be also considered as a novel enzymatic antimicrobial agents to inhibit bacterial infection and entry to host cells.
Collapse
Affiliation(s)
- Mariano Prado Acosta
- Laboratorio de Bacterias Gram Positivas, Departamento de Química Biológica-IQUIBICEN, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires-CONICET, Argentina.
| | - Sandra M Ruzal
- Laboratorio de Bacterias Gram Positivas, Departamento de Química Biológica-IQUIBICEN, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires-CONICET, Argentina
| | - Sandra M Cordo
- Laboratorio de Virología, Departamento de Química Biológica-IQUIBICEN, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires-CONICET, Argentina.
| |
Collapse
|
36
|
Del Bel Belluz L, Guidi R, Pateras IS, Levi L, Mihaljevic B, Rouf SF, Wrande M, Candela M, Turroni S, Nastasi C, Consolandi C, Peano C, Tebaldi T, Viero G, Gorgoulis VG, Krejsgaard T, Rhen M, Frisan T. The Typhoid Toxin Promotes Host Survival and the Establishment of a Persistent Asymptomatic Infection. PLoS Pathog 2016; 12:e1005528. [PMID: 27055274 PMCID: PMC4824513 DOI: 10.1371/journal.ppat.1005528] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2015] [Accepted: 03/04/2016] [Indexed: 02/06/2023] Open
Abstract
Bacterial genotoxins, produced by several Gram-negative bacteria, induce DNA damage in the target cells. While the responses induced in the host cells have been extensively studied in vitro, the role of these effectors during the course of infection remains poorly characterized. To address this issue, we assessed the effects of the Salmonella enterica genotoxin, known as typhoid toxin, in in vivo models of murine infection. Immunocompetent mice were infected with isogenic S. enterica, serovar Typhimurium (S. Typhimurium) strains, encoding either a functional or an inactive typhoid toxin. The presence of the genotoxic subunit was detected 10 days post-infection in the liver of infected mice. Unexpectedly, its expression promoted the survival of the host, and was associated with a significant reduction of severe enteritis in the early phases of infection. Immunohistochemical and transcriptomic analysis confirmed the toxin-mediated suppression of the intestinal inflammatory response. The presence of a functional typhoid toxin further induced an increased frequency of asymptomatic carriers. Our data indicate that the typhoid toxin DNA damaging activity increases host survival and favours long-term colonization, highlighting a complex cross-talk between infection, DNA damage response and host immune response. These findings may contribute to understand why such effectors have been evolutionary conserved and horizontally transferred among Gram-negative bacteria.
Collapse
Affiliation(s)
- Lisa Del Bel Belluz
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Riccardo Guidi
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Ioannis S. Pateras
- Department of Histology and Embryology, School of Medicine, University of Athens, Athens, Greece
| | - Laura Levi
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Boris Mihaljevic
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Syed Fazle Rouf
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Marie Wrande
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Marco Candela
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Silvia Turroni
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Claudia Nastasi
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Clarissa Consolandi
- Institute of Biomedical Technologies, Italian National Research Council, Segrate, Milan, Italy
| | - Clelia Peano
- Institute of Biomedical Technologies, Italian National Research Council, Segrate, Milan, Italy
| | - Toma Tebaldi
- Centre for Integrative Biology University of Trento, Trento, Italy
| | | | - Vassilis G. Gorgoulis
- Department of Histology and Embryology, School of Medicine, University of Athens, Athens, Greece
- Biomedical Research Foundation, Academy of Athens, Athens, Greece
- Institute for Cancer Sciences, University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom
- Manchester Centre for Cellular Metabolism, University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Thorbjørn Krejsgaard
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Mikael Rhen
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Teresa Frisan
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
- * E-mail:
| |
Collapse
|
37
|
Differences in Host Cell Invasion and Salmonella Pathogenicity Island 1 Expression between Salmonella enterica Serovar Paratyphi A and Nontyphoidal S. Typhimurium. Infect Immun 2016; 84:1150-1165. [PMID: 26857569 DOI: 10.1128/iai.01461-15] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Accepted: 02/01/2016] [Indexed: 11/20/2022] Open
Abstract
Active invasion into nonphagocytic host cells is central to Salmonella enterica pathogenicity and dependent on multiple genes within Salmonella pathogenicity island 1 (SPI-1). Here, we explored the invasion phenotype and the expression of SPI-1 in the typhoidal serovarS Paratyphi A compared to that of the nontyphoidal serovarS Typhimurium. We demonstrate that while S. Typhimurium is equally invasive under both aerobic and microaerobic conditions, S. Paratyphi A invades only following growth under microaerobic conditions. Transcriptome sequencing (RNA-Seq), reverse transcription-PCR (RT-PCR), Western blot, and secretome analyses established that S. Paratyphi A expresses much lower levels of SPI-1 genes and secretes lesser amounts of SPI-1 effector proteins than S. Typhimurium, especially under aerobic growth. Bypassing the native SPI-1 regulation by inducible expression of the SPI-1 activator, HilA, considerably elevated SPI-1 gene expression, host cell invasion, disruption of epithelial integrity, and induction of proinflammatory cytokine secretion by S. Paratyphi A but not by S. Typhimurium, suggesting that SPI-1 expression is naturally downregulated inS Paratyphi A. Using streptomycin-treated mice, we were able to establish substantial intestinal colonization byS Paratyphi A and showed moderately higher pathology and intestinal inflammation in mice infected with S. Paratyphi A overexpressing hilA Collectively, our results reveal unexpected differences in SPI-1 expression between S. Paratyphi A andS Typhimurium, indicate that S. Paratyphi A host cell invasion is suppressed under aerobic conditions, and suggest that lower invasion in aerobic sites and suppressed expression of immunogenic SPI-1 components contributes to the restrained inflammatory infection elicited by S. Paratyphi A.
Collapse
|
38
|
Poultry body temperature contributes to invasion control through reduced expression of Salmonella pathogenicity island 1 genes in Salmonella enterica serovars Typhimurium and Enteritidis. Appl Environ Microbiol 2015; 81:8192-201. [PMID: 26386070 DOI: 10.1128/aem.02622-15] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Accepted: 09/15/2015] [Indexed: 01/17/2023] Open
Abstract
Salmonella enterica serovars Typhimurium (S. Typhimurium) and Enteritidis (S. Enteritidis) are foodborne pathogens, and outbreaks are often associated with poultry products. Chickens are typically asymptomatic when colonized by these serovars; however, the factors contributing to this observation are uncharacterized. Whereas symptomatic mammals have a body temperature between 37°C and 39°C, chickens have a body temperature of 41°C to 42°C. Here, in vivo experiments using chicks demonstrated that numbers of viable S. Typhimurium or S. Enteritidis bacteria within the liver and spleen organ sites were ≥4 orders of magnitude lower than those within the ceca. When similar doses of S. Typhimurium or S. Enteritidis were given to C3H/HeN mice, the ratio of the intestinal concentration to the liver/spleen concentration was 1:1. In the avian host, this suggested poor survival within these tissues or a reduced capacity to traverse the host epithelial layer and reach liver/spleen sites or both. Salmonella pathogenicity island 1 (SPI-1) promotes localization to liver/spleen tissues through invasion of the epithelial cell layer. Following in vitro growth at 42°C, SPI-1 genes sipC, invF, and hilA and the SPI-1 rtsA activator were downregulated compared to expression at 37°C. Overexpression of the hilA activators fur, fliZ, and hilD was capable of inducing hilA-lacZ at 37°C but not at 42°C despite the presence of similar levels of protein at the two temperatures. In contrast, overexpression of either hilC or rtsA was capable of inducing hilA and sipC at 42°C. These data indicate that physiological parameters of the poultry host, such as body temperature, have a role in modulating expression of virulence.
Collapse
|
39
|
Rivera-Chávez F, Bäumler AJ. The Pyromaniac Inside You: Salmonella Metabolism in the Host Gut. Annu Rev Microbiol 2015; 69:31-48. [PMID: 26002180 DOI: 10.1146/annurev-micro-091014-104108] [Citation(s) in RCA: 108] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
A metabolically diverse microbial community occupies all available nutrient-niches in the lumen of the mammalian intestine, making it difficult for pathogens to establish themselves in this highly competitive environment. Salmonella serovars sidestep the competition by using their virulence factors to coerce the host into creating a novel nutrient-niche. Inflammation-derived nutrients available in this new niche support a bloom of Salmonella serovars, thereby ensuring transmission of the pathogen to the next susceptible host by the fecal-oral route. Here we review the anaerobic food chain that characterizes resident gut-associated microbial communities along with the winning metabolic strategy Salmonella serovars use to edge out competing microbes in the inflamed intestine.
Collapse
Affiliation(s)
- Fabian Rivera-Chávez
- Department of Medical Microbiology and Immunology, School of Medicine, University of California, Davis, California 95616;
| | - Andreas J Bäumler
- Department of Medical Microbiology and Immunology, School of Medicine, University of California, Davis, California 95616;
| |
Collapse
|
40
|
Now you see me, now you don't: the interaction of Salmonella with innate immune receptors. Nat Rev Microbiol 2015; 13:206-16. [PMID: 25749454 DOI: 10.1038/nrmicro3428] [Citation(s) in RCA: 112] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Salmonella enterica serovars are associated with an estimated 1 million deaths annually and are also useful model organisms for investigating the mechanisms of host-bacterium interactions. The insights gained from studies on non-typhoidal Salmonella (NTS) serovars have provided a fascinating overview of the mechanisms by which the innate immune system detects and responds to bacterial pathogens. However, specific virulence factors and changes in virulence gene regulation in S. enterica subsp. enterica serovar Typhi alter the innate immune responses to this pathogen. In this Review, we compare and contrast the interactions of S. Typhi and NTS serovars with host innate immune receptors and discuss why the disease manifestations associated with S. Typhi infection differ considerably from those associated with the closely related NTS serovars.
Collapse
|
41
|
The flagellar regulator TviA reduces pyroptosis by Salmonella enterica serovar Typhi. Infect Immun 2015; 83:1546-55. [PMID: 25644011 DOI: 10.1128/iai.02803-14] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
To discern virulent from innocuous microbes, the innate immune system senses events associated with bacterial access to immunoprivileged sites such as the host cell cytosol. One such pathway is triggered by the cytosolic delivery of flagellin, the major subunit of the flagellum, by bacterial secretion systems. This leads to inflammasome activation and subsequent proinflammatory cell death (pyroptosis) of the infected phagocyte. In this study, we demonstrate that the causative agent of typhoid fever, Salmonella enterica serovar Typhi, can partially subvert this critical innate immune recognition event. The transcriptional regulator TviA, which is absent from Salmonella serovars associated with human gastroenteritis, repressed the expression of flagellin during infection of human macrophage-like (THP-1) cells. This mechanism allowed S. Typhi to dampen inflammasome activation, leading to reduced interleukin-1β (IL-1β) secretion and diminished cell death. Likewise, the introduction of the tviA gene in nontyphoidal Salmonella enterica serovar Typhimurium reduced flagellin-induced pyroptosis. These data suggest that gene regulation of virulence factors enables S. Typhi to evade innate immune recognition by concealing a pathogen-induced process from being sensed by the inflammasome.
Collapse
|
42
|
Wiedemann A, Virlogeux-Payant I, Chaussé AM, Schikora A, Velge P. Interactions of Salmonella with animals and plants. Front Microbiol 2015; 5:791. [PMID: 25653644 PMCID: PMC4301013 DOI: 10.3389/fmicb.2014.00791] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Accepted: 12/22/2014] [Indexed: 12/16/2022] Open
Abstract
Salmonella enterica species are Gram-negative bacteria, which are responsible for a wide range of food- and water-borne diseases in both humans and animals, thereby posing a major threat to public health. Recently, there has been an increasing number of reports, linking Salmonella contaminated raw vegetables and fruits with food poisoning. Many studies have shown that an essential feature of the pathogenicity of Salmonella is its capacity to cross a number of barriers requiring invasion of a large variety of cells and that the extent of internalization may be influenced by numerous factors. However, it is poorly understood how Salmonella successfully infects hosts as diversified as animals or plants. The aim of this review is to describe the different stages required for Salmonella interaction with its hosts: (i) attachment to host surfaces; (ii) entry processes; (iii) multiplication; (iv) suppression of host defense mechanisms; and to point out similarities and differences between animal and plant infections.
Collapse
Affiliation(s)
- Agnès Wiedemann
- Institut National de la Recherche Agronomique, UMR1282 Infectiologie et Santé Publique Nouzilly, France ; UMR1282 Infectiologie et Santé Publique, Université François Rabelais Tours, France
| | - Isabelle Virlogeux-Payant
- Institut National de la Recherche Agronomique, UMR1282 Infectiologie et Santé Publique Nouzilly, France ; UMR1282 Infectiologie et Santé Publique, Université François Rabelais Tours, France
| | - Anne-Marie Chaussé
- Institut National de la Recherche Agronomique, UMR1282 Infectiologie et Santé Publique Nouzilly, France ; UMR1282 Infectiologie et Santé Publique, Université François Rabelais Tours, France
| | - Adam Schikora
- Institute for Phytopathology, Research Center for BioSystems, Land Use and Nutrition (IFZ), Justus Liebig University Giessen Giessen, Germany
| | - Philippe Velge
- Institut National de la Recherche Agronomique, UMR1282 Infectiologie et Santé Publique Nouzilly, France ; UMR1282 Infectiologie et Santé Publique, Université François Rabelais Tours, France
| |
Collapse
|
43
|
|
44
|
Carden S, Okoro C, Dougan G, Monack D. Non-typhoidal Salmonella Typhimurium ST313 isolates that cause bacteremia in humans stimulate less inflammasome activation than ST19 isolates associated with gastroenteritis. Pathog Dis 2014; 73:ftu023. [PMID: 25808600 PMCID: PMC4399442 DOI: 10.1093/femspd/ftu023] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/19/2014] [Indexed: 02/07/2023] Open
Abstract
Salmonella is an enteric pathogen that causes a range of diseases in humans. Non-typhoidal Salmonella (NTS) serovars such as Salmonella enterica serovar Typhimurium generally cause a self-limiting gastroenteritis whereas typhoidal serovars cause a systemic disease, typhoid fever. However, S. Typhimurium isolates within the multi-locus sequence type ST313 have emerged in sub-Saharan Africa as a major cause of bacteremia in humans. The S. Typhimurium ST313 lineage is phylogenetically distinct from classical S. Typhimurium lineages, such as ST19, that cause zoonotic gastroenteritis worldwide. Previous studies have shown that the ST313 lineage has undergone genome degradation when compared to the ST19 lineage, similar to that observed for typhoidal serovars. Currently, little is known about phenotypic differences between ST313 isolates and other NTS isolates. We find that representative ST313 isolates invade non-phagocytic cells less efficiently than the classical ST19 isolates that are more commonly associated with gastroenteritis. In addition, ST313 isolates induce less Caspase-1-dependent macrophage death and IL-1β release than ST19 isolates. ST313 isolates also express relatively lower levels of mRNA of the genes encoding the SPI-1 effector sopE2 and the flagellin, fliC, providing possible explanations for the decrease in invasion and inflammasome activation. The ST313 isolates have invasion and inflammatory phenotypes that are intermediate; more invasive and inflammatory than Salmonella enterica serovar Typhi and less than ST19 isolates associated with gastroenteritis. This suggests that both phenotypically and at the genomic level ST313 isolates are evolving signatures that facilitate a systemic lifestyle in humans.
Collapse
Affiliation(s)
- Sarah Carden
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Chinyere Okoro
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SA, UK
| | - Gordon Dougan
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SA, UK
| | - Denise Monack
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| |
Collapse
|
45
|
Chowdhury R, Mandal RS, Ta A, Das S. An AIL family protein promotes type three secretion system-1-independent invasion and pathogenesis of Salmonella enterica serovar Typhi. Cell Microbiol 2014; 17:486-503. [PMID: 25308535 DOI: 10.1111/cmi.12379] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Revised: 09/25/2014] [Accepted: 10/04/2014] [Indexed: 02/05/2023]
Abstract
Adhesion and invasion of Intestinal Epithelial Cells (IECs) are critical for the pathogenesis of Salmonella Typhi, the aetiological agent of human typhoid fever. While type three secretion system-1 (T3SS-1) is a major invasion apparatus of Salmonella, independent invasion mechanisms were described for non-typhoidal Salmonellae. Here, we show that T2942, an AIL-like protein of S. Typhi Ty2 strain, is required for adhesion and invasion of cultured IECs. That invasion was T3SS-1 independent was proved by ectopic expression of T2942 in the non-invasive E. coli BL21 and double-mutant Ty2 (Ty2Δt2942ΔinvG) strains. Laminin and fibronectin were identified as the host-binding partners of T2942 with higher affinity for laminin. Standalone function of T2942 was confirmed by cell adhesion of the recombinant protein, while the protein or anti-T2942 antiserum blocked adhesion/invasion of S. Typhi, indicating specificity. A 20-amino acid extracellular loop was required for invasion, while several loop regions of T2942 contributed to adhesion. Further, T2942 cooperates with laminin-binding T2544 for adhesion and T3SS-1 for invasion. Finally, T2942 was required and synergistically worked with T3SS-1 for pathogenesis of S. Typhi in mice. Considering wide distribution of T2942 among clinical strains, the protein or the 20-mer peptide may be suitable for vaccine development.
Collapse
Affiliation(s)
- Rimi Chowdhury
- Division of Clinical Medicine, National Institute of Cholera and Enteric Diseases, P-33 Scheme XM C.I.T. Road, Beliaghata Kolkata, 700010, India
| | | | | | | |
Collapse
|