1
|
Guerra FE, Karlinsey JE, Libby SJ, Fang FC. Evasion of serum antibodies and complement by Salmonella Typhi and Paratyphi A. PLoS Pathog 2025; 21:e1012917. [PMID: 40315236 DOI: 10.1371/journal.ppat.1012917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Accepted: 04/18/2025] [Indexed: 05/04/2025] Open
Abstract
Nontyphoidal and enteric fever serovars of Salmonella enterica display distinctive interactions with serum antibodies and the complement system, which initiate the host immune response to invading microbes. This study examines the contributions of lipopolysaccharide O-antigen (O-ag) and the S. Typhi Vi polysaccharide capsule to serum resistance, complement activation and deposition, and immunoglobulin (Ig) binding in nontyphoidal S. enterica serovar Typhimurium and the enteric fever serovars S. Typhi and S. Paratyphi A. Although all three serovars are resistant to serum killing, S. Typhi and S. Paratyphi A exhibit lower levels of Ig binding, complement binding and complement activation compared to S. Typhimurium. In S. Typhimurium, WzzB-dependent long O-antigen (L O-ag) production with 16-to-35 repeating O-ag units, and FepE-dependent very long O-antigen (VL O-ag) production with over 100 repeating O-ag units, are required for serum resistance but do not prevent IgM binding or complement deposition. S. Typhi lacks VL O-ag, but its production of Vi capsule inhibits IgM binding and complement deposition, while acting in concert with L O-ag to resist serum killing. In S. Paratyphi A, L O-ag production is deficient due to a hypofunctional WzzB protein, but this is compensated by greater quantities of VL O-ag, which are required for serum resistance. Restoration of WzzB function by exchange with the S. Typhimurium or S. Typhi wzzB alleles can restore L O-ag production in S. Paratyphi A but decreases VL O-ag production, resulting in increased IgM binding. Replacement of the S. Paratyphi A O2-type polysaccharide with the S. Typhi O9 polysaccharide further increases IgM binding of S. Paratyphi A, which enhances complement activation but not complement deposition. Lastly, a gene duplication of rfbV in S. Paratyphi A is necessary for higher levels of VL O-ag and resistance to complement deposition and antibody binding. Collectively, these observations demonstrate fundamental differences between nontyphoidal and enteric fever Salmonella serovars in their interactions with innate immune effectors. Whereas nontyphoidal S. Typhimurium elicits, exploits and withstands the host acute inflammatory response, the enteric fever serovars S. Typhi and S. Paratyphi A evade it by limiting antibody recognition and complement activation and deposition.
Collapse
Affiliation(s)
- Fermin E Guerra
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, United States of America
| | - Joyce E Karlinsey
- Department of Microbiology, University of Washington, Seattle, Washington, United States of America
| | - Stephen J Libby
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, United States of America
| | - Ferric C Fang
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, United States of America
- Department of Microbiology, University of Washington, Seattle, Washington, United States of America
| |
Collapse
|
2
|
Lee S, Leclercq LD, Guerardel Y, Szymanski CM, Hurtaux T, Doering TL, Katayama T, Fujita K, Aoki K, Aoki-Kinoshita KF. MicroGlycoDB: A database of microbial glycans using Semantic Web technologies. BBA ADVANCES 2024; 6:100126. [PMID: 39720162 PMCID: PMC11667048 DOI: 10.1016/j.bbadva.2024.100126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 11/25/2024] [Accepted: 11/26/2024] [Indexed: 12/26/2024] Open
Abstract
Glycoconjugates are present on microbial surfaces and play critical roles in modulating interactions with the environment and the host. Extensive research on microbial glycans, including elucidating the structural diversity of the glycan moieties of glycoconjugates and polysaccharides, has been carried out to investigate the function of glycans in modulating the interactions between the host and microbes, to explore their potential applications in the therapeutic targeting of pathogenic species, and in the use as probiotics in gut microbiomes. However, glycan-related information is dispersed across numerous databases and a vast amount of literature, which makes it laborious and time-consuming to identify and gather the relevant information about microbial glycosylation. This challenge can be addressed by a comprehensive database, which could offer insight into the fundamental processes underlying glycosylation. We have developed a MicroGlycoDB database to provide integrated glycan information on important model microorganisms. The data is described using Semantic Web Technologies, which allow microbial glycan data to be represented in a structured format accessible by machines, thus facilitating data sharing and integration with other resources that catalog features such as pathways, diseases, or interactions. This semantic data based on ontologies will contribute to the discovery of new knowledge in the field of microbiology, along with the expansion of information on the glycosylation of other microorganisms.
Collapse
Affiliation(s)
- Sunmyoung Lee
- Glycan and Life Systems Integration Center (GaLSIC), Soka University, Hachioji, Tokyo, Japan
| | - Louis-David Leclercq
- French National Center for Scientific Research (CNRS), University of Lille, Lille, France
| | - Yann Guerardel
- French National Center for Scientific Research (CNRS), University of Lille, Lille, France
| | | | - Thomas Hurtaux
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Tamara L. Doering
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Takane Katayama
- Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Kiyotaka Fujita
- The United Graduate School of Agricultural Sciences, Kagoshima University, Kagoshima, Japan
| | - Kazuhiro Aoki
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Kiyoko F. Aoki-Kinoshita
- Glycan and Life Systems Integration Center (GaLSIC), Soka University, Hachioji, Tokyo, Japan
- Graduate School of Science and Engineering, Soka University, Hachioji, Tokyo, Japan
| |
Collapse
|
3
|
Carreto-Binaghi LE, Sztein MB, Booth JS. Role of cellular effectors in the induction and maintenance of IgA responses leading to protective immunity against enteric bacterial pathogens. Front Immunol 2024; 15:1446072. [PMID: 39324143 PMCID: PMC11422102 DOI: 10.3389/fimmu.2024.1446072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Accepted: 08/26/2024] [Indexed: 09/27/2024] Open
Abstract
The mucosal immune system is a critical first line of defense to infectious diseases, as many pathogens enter the body through mucosal surfaces, disrupting the balanced interactions between mucosal cells, secretory molecules, and microbiota in this challenging microenvironment. The mucosal immune system comprises of a complex and integrated network that includes the gut-associated lymphoid tissues (GALT). One of its primary responses to microbes is the secretion of IgA, whose role in the mucosa is vital for preventing pathogen colonization, invasion and spread. The mechanisms involved in these key responses include neutralization of pathogens, immune exclusion, immune modulation, and cross-protection. The generation and maintenance of high affinity IgA responses require a delicate balance of multiple components, including B and T cell interactions, innate cells, the cytokine milieu (e.g., IL-21, IL-10, TGF-β), and other factors essential for intestinal homeostasis, including the gut microbiota. In this review, we will discuss the main cellular components (e.g., T cells, innate lymphoid cells, dendritic cells) in the gut microenvironment as mediators of important effector responses and as critical players in supporting B cells in eliciting and maintaining IgA production, particularly in the context of enteric infections and vaccination in humans. Understanding the mechanisms of humoral and cellular components in protection could guide and accelerate the development of more effective mucosal vaccines and therapeutic interventions to efficiently combat mucosal infections.
Collapse
Affiliation(s)
- Laura E. Carreto-Binaghi
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, United States
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, United States
- Laboratorio de Inmunobiologia de la Tuberculosis, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City, Mexico
| | - Marcelo B. Sztein
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, United States
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, United States
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
- Tumor Immunology and Immunotherapy Program, University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD, United States
| | - Jayaum S. Booth
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, United States
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, United States
| |
Collapse
|
4
|
Lee GY, Song J. Single missense mutations in Vi capsule synthesis genes confer hypervirulence to Salmonella Typhi. Nat Commun 2024; 15:5258. [PMID: 38898034 PMCID: PMC11187135 DOI: 10.1038/s41467-024-49590-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 06/07/2024] [Indexed: 06/21/2024] Open
Abstract
Many bacterial pathogens, including the human exclusive pathogen Salmonella Typhi, express capsular polysaccharides as a crucial virulence factor. Here, through S. Typhi whole genome sequence analyses and functional studies, we found a list of single point mutations that make S. Typhi hypervirulent. We discovered a single point mutation in the Vi biosynthesis enzymes that control Vi polymerization or acetylation is enough to result in different capsule variants of S. Typhi. All variant strains are pathogenic, but the hyper Vi capsule variants are particularly hypervirulent, as demonstrated by the high morbidity and mortality rates observed in infected mice. The hypo Vi capsule variants have primarily been identified in Africa, whereas the hyper Vi capsule variants are distributed worldwide. Collectively, these studies increase awareness about the existence of different capsule variants of S. Typhi, establish a solid foundation for numerous future studies on S. Typhi capsule variants, and offer valuable insights into strategies to combat capsulated bacteria.
Collapse
Affiliation(s)
- Gi Young Lee
- Department of Microbiology and Immunology, Cornell University College of Veterinary Medicine, Ithaca, NY, 14853, USA
| | - Jeongmin Song
- Department of Microbiology and Immunology, Cornell University College of Veterinary Medicine, Ithaca, NY, 14853, USA.
| |
Collapse
|
5
|
Lee GY, Song J. Single missense mutations in Vi capsule synthesis genes confer hypervirulence to Salmonella Typhi. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.12.28.573590. [PMID: 38260632 PMCID: PMC10802248 DOI: 10.1101/2023.12.28.573590] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Many bacterial pathogens, including the human exclusive pathogen Salmonella Typhi, express capsular polysaccharides as a crucial virulence factor. Here, through S. Typhi whole genome sequence analyses and functional studies, we found a list of single point mutations that make S . Typhi hypervirulent. We discovered a single point mutation in the Vi biosynthesis enzymes that control the length or acetylation of Vi is enough to create different capsule variants of S. Typhi. All variant strains are pathogenic, but the hyper-capsule variants are particularly hypervirulent, as demonstrated by the high morbidity and mortality rates observed in infected mice. The hypo-capsule variants have primarily been identified in Africa, whereas the hyper-capsule variants are distributed worldwide. Collectively, these studies increase awareness about the existence of different capsule variants of S. Typhi, establish a solid foundation for numerous future studies on S. Typhi capsule variants, and offer valuable insights into strategies to combat capsulated bacteria.
Collapse
|
6
|
Raman V, Deshpande CP, Khanduja S, Howell LM, Van Dessel N, Forbes NS. Build-a-bug workshop: Using microbial-host interactions and synthetic biology tools to create cancer therapies. Cell Host Microbe 2023; 31:1574-1592. [PMID: 37827116 DOI: 10.1016/j.chom.2023.09.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 08/16/2023] [Accepted: 09/12/2023] [Indexed: 10/14/2023]
Abstract
Many systemically administered cancer therapies exhibit dose-limiting toxicities that reduce their effectiveness. To increase efficacy, bacterial delivery platforms have been developed that improve safety and prolong treatment. Bacteria are a unique class of therapy that selectively colonizes most solid tumors. As delivery vehicles, bacteria have been genetically modified to express a range of therapies that match multiple cancer indications. In this review, we describe a modular "build-a-bug" method that focuses on five design characteristics: bacterial strain (chassis), therapeutic compound, delivery method, immune-modulating features, and genetic control circuits. We emphasize how fundamental research into gut microbe pathogenesis has created safe bacterial therapies, some of which have entered clinical trials. The genomes of gut microbes are fertile grounds for discovery of components to improve delivery and modulate host immune responses. Future work coupling these delivery vehicles with insights from gut microbes could lead to the next generation of microbial cancer therapy.
Collapse
Affiliation(s)
- Vishnu Raman
- Department of Chemical Engineering, University of Massachusetts, Amherst, Amherst, MA, USA; Ernest Pharmaceuticals, LLC, Hadley, MA, USA
| | - Chinmay P Deshpande
- Department of Chemical Engineering, University of Massachusetts, Amherst, Amherst, MA, USA
| | - Shradha Khanduja
- Department of Chemical Engineering, University of Massachusetts, Amherst, Amherst, MA, USA
| | - Lars M Howell
- Department of Chemical Engineering, University of Massachusetts, Amherst, Amherst, MA, USA
| | | | - Neil S Forbes
- Department of Chemical Engineering, University of Massachusetts, Amherst, Amherst, MA, USA; Molecular and Cell Biology Program, University of Massachusetts, Amherst, Amherst, MA, USA; Institute for Applied Life Science, University of Massachusetts, Amherst, Amherst, MA, USA.
| |
Collapse
|
7
|
Chatterjee R, Chowdhury AR, Mukherjee D, Chakravortty D. From Eberthella typhi to Salmonella Typhi: The Fascinating Journey of the Virulence and Pathogenicity of Salmonella Typhi. ACS OMEGA 2023; 8:25674-25697. [PMID: 37521659 PMCID: PMC10373206 DOI: 10.1021/acsomega.3c02386] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Accepted: 06/30/2023] [Indexed: 08/01/2023]
Abstract
Salmonella Typhi (S. Typhi), the invasive typhoidal serovar of Salmonella enterica that causes typhoid fever in humans, is a severe threat to global health. It is one of the major causes of high morbidity and mortality in developing countries. According to recent WHO estimates, approximately 11-21 million typhoid fever illnesses occur annually worldwide, accounting for 0.12-0.16 million deaths. Salmonella infection can spread to healthy individuals by the consumption of contaminated food and water. Typhoid fever in humans sometimes is accompanied by several other critical extraintestinal complications related to the central nervous system, cardiovascular system, pulmonary system, and hepatobiliary system. Salmonella Pathogenicity Island-1 and Salmonella Pathogenicity Island-2 are the two genomic segments containing genes encoding virulent factors that regulate its invasion and systemic pathogenesis. This Review aims to shed light on a comparative analysis of the virulence and pathogenesis of the typhoidal and nontyphoidal serovars of S. enterica.
Collapse
Affiliation(s)
- Ritika Chatterjee
- Department
of Microbiology and Cell Biology, Division of Biological Sciences, Indian Institute of Science, Bangalore, Karnataka 560012, India
| | - Atish Roy Chowdhury
- Department
of Microbiology and Cell Biology, Division of Biological Sciences, Indian Institute of Science, Bangalore, Karnataka 560012, India
| | - Debapriya Mukherjee
- Department
of Microbiology and Cell Biology, Division of Biological Sciences, Indian Institute of Science, Bangalore, Karnataka 560012, India
| | - Dipshikha Chakravortty
- Department
of Microbiology and Cell Biology, Division of Biological Sciences, Indian Institute of Science, Bangalore, Karnataka 560012, India
- Centre
for Biosystems Science and Engineering, Indian Institute of Science, Bangalore, Karnataka 560012, India
| |
Collapse
|
8
|
Walker GT, Gerner RR, Nuccio SP, Raffatellu M. Murine Models of Salmonella Infection. Curr Protoc 2023; 3:e824. [PMID: 37478288 PMCID: PMC10372748 DOI: 10.1002/cpz1.824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/23/2023]
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
|
9
|
Identification of collaborative cross mouse strains permissive to Salmonella enterica serovar Typhi infection. Sci Rep 2023; 13:393. [PMID: 36624251 PMCID: PMC9829673 DOI: 10.1038/s41598-023-27400-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Accepted: 01/02/2023] [Indexed: 01/11/2023] Open
Abstract
Salmonella enterica serovar Typhi is the causative agent of typhoid fever restricted to humans and does not replicate in commonly used inbred mice. Genetic variation in humans is far greater and more complex than that in a single inbred strain of mice. The Collaborative Cross (CC) is a large panel of recombinant inbred strains which has a wider range of genetic diversity than laboratory inbred mouse strains. We found that the CC003/Unc and CC053/Unc strains are permissive to intraperitoneal but not oral route of S. Typhi infection and show histopathological changes characteristic of human typhoid. These CC strains are immunocompetent, and immunization induces antigen-specific responses that can kill S. Typhi in vitro and control S. Typhi in vivo. Our results indicate that CC003/Unc and CC053/Unc strains can help identify the genetic basis for typhoid susceptibility, S. Typhi virulence mechanism(s) in vivo, and serve as a preclinical mammalian model system to identify effective vaccines and therapeutics strategies.
Collapse
|
10
|
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
|
11
|
Alugupalli AS, Cravens MP, Walker JA, Gulandijany D, Dickinson GS, Debes GF, Schifferli DM, Bäumler AJ, Alugupalli KR, Alugupalli KR. The Lack of Natural IgM Increases Susceptibility and Impairs Anti-Vi Polysaccharide IgG Responses in a Mouse Model of Typhoid. Immunohorizons 2022; 6:807-816. [PMID: 36480484 DOI: 10.4049/immunohorizons.2200088] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 11/02/2022] [Indexed: 12/13/2022] Open
Abstract
Circulating IgM present in the body prior to any apparent Ag exposure is referred to as natural IgM. Natural IgM provides protective immunity against a variety of pathogens. Salmonella enterica serovar Typhi (S. Typhi) is the causative agent of typhoid fever in humans. Because mice are not permissive to S. Typhi infection, we employed a murine model of typhoid using S. enterica serovar Typhimurium expressing the Vi polysaccharide (ViPS) of S. Typhi (S. Typhimurium strain RC60) to evaluate the role of natural IgM in pathogenesis. We found that natural mouse IgM binds to S. Typhi and S. Typhimurium. The severity of S. Typhimurium infection in mice is dependent on presence of the natural resistance-associated macrophage protein 1 (Nramp1) allele; therefore, we infected mice deficient in secreted form of IgM (sIgM) on either a Nramp1-resistant (129S) or -susceptible (C57BL/6J) background. We found that the lack of natural IgM results in a significantly increased susceptibility and an exaggerated liver pathology regardless of the route of infection or the Nramp1 allele. Reconstitution of sIgM-/- mice with normal mouse serum or purified polyclonal IgM restored the resistance to that of sIgM+/+ mice. Furthermore, immunization of sIgM-/- mice with heat-killed S. Typhi induced a significantly reduced anti-ViPS IgG and complement-dependent bactericidal activity against S. Typhi in vitro, compared with that of sIgM+/+ mice. These findings indicate that natural IgM is an important factor in reducing the typhoid severity and inducing an optimal anti-ViPS IgG response to vaccination.
Collapse
Affiliation(s)
- Akhil S Alugupalli
- Department of Microbiology and Immunology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA.,Department of Microbiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA
| | - Matthew P Cravens
- Department of Microbiology and Immunology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA
| | - Justin A Walker
- Department of Microbiology and Immunology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA
| | - Dania Gulandijany
- Department of Microbiology and Immunology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA
| | - Gregory S Dickinson
- Department of Microbiology and Immunology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA
| | - Gudrun F Debes
- Department of Microbiology and Immunology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA
| | - Dieter M Schifferli
- Department of Microbiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA
| | - Andreas J Bäumler
- Department of Medical Microbiology and Immunology, School of Medicine, University of California at Davis, Davis, CA; and
| | - Kishore R Alugupalli
- Department of Microbiology and Immunology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA.,Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA
| | - Kishore R Alugupalli
- Department of Microbiology and Immunology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA.,Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA
| |
Collapse
|
12
|
Sun L, Cheng L, Ma Y, Lei P, Wang R, Gu Y, Li S, Zhang F, Xu H. Exopolysaccharides from Pantoea alhagi NX-11 specifically improve its root colonization and rice salt resistance. Int J Biol Macromol 2022; 209:396-404. [PMID: 35413311 DOI: 10.1016/j.ijbiomac.2022.04.015] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 03/31/2022] [Accepted: 04/03/2022] [Indexed: 12/13/2022]
Abstract
Plant growth-promoting rhizobacteria (PGPR) and their extracellular polymers such as exopolysaccharides can enhance rice salt stress resistance, however, the relevant mechanism remains unclear. In this study, an exopolysaccharides-deficient strain, named ΔpspD, was obtained from Pantoea alhagi NX-11 by chromosomal pspD deletion. The yield and characteristics of ΔpspD exopolysaccharides was obviously different from P. alhagi NX-11 exopolysaccharides (PAPS). Subsequently, hydroponic experiments showed that NX-11 or PAPS could enhance rice salt tolerance, but ΔpspD could not. Furthermore, it was found that PAPS promoted P. alhagi rhizosphere colonization through a direct effect on biofilm formation, as well as through an indirect impact of enhancing the abilities of biofilm formation and chemotaxis by altering rice root exudates. Importantly, the effect of PAPS in promoting the root colonization of NX-11 was specific. Through transcriptome and RT-qPCR analysis, we revealed that this specificity correlated with PAPS-induced lectin overexpression. The specificity between exopolysaccharides and the host microorganism ensures the colonization of the latter, and prevents other microorganisms from hitchhiking to the rice roots.
Collapse
Affiliation(s)
- Liang Sun
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Food Science and Light Industry, Nanjing Tech University, Nanjing 211816, China
| | - Lifangyu Cheng
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Food Science and Light Industry, Nanjing Tech University, Nanjing 211816, China
| | - Yuhang Ma
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Food Science and Light Industry, Nanjing Tech University, Nanjing 211816, China
| | - Peng Lei
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Food Science and Light Industry, Nanjing Tech University, Nanjing 211816, China.
| | - Rui Wang
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Food Science and Light Industry, Nanjing Tech University, Nanjing 211816, China
| | - Yian Gu
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Food Science and Light Industry, Nanjing Tech University, Nanjing 211816, China.
| | - Sha Li
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Food Science and Light Industry, Nanjing Tech University, Nanjing 211816, China
| | - Fuhai Zhang
- Agricultural and Rural Bureau of Yantai, Yantai 264000, China
| | - Hong Xu
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Food Science and Light Industry, Nanjing Tech University, Nanjing 211816, China.
| |
Collapse
|
13
|
Glycan-mediated molecular interactions in bacterial pathogenesis. Trends Microbiol 2022; 30:254-267. [PMID: 34274195 PMCID: PMC8758796 DOI: 10.1016/j.tim.2021.06.011] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 06/10/2021] [Accepted: 06/22/2021] [Indexed: 02/06/2023]
Abstract
Glycans are expressed on the surface of nearly all host and bacterial cells. Not surprisingly, glycan-mediated molecular interactions play a vital role in bacterial pathogenesis and host responses against pathogens. Glycan-mediated host-pathogen interactions can benefit the pathogen, host, or both. Here, we discuss (i) bacterial glycans that play a critical role in bacterial colonization and/or immune evasion, (ii) host glycans that are utilized by bacteria for pathogenesis, and (iii) bacterial and host glycans involved in immune responses against pathogens. We further discuss (iv) opportunities and challenges for transforming these research findings into more effective antibacterial strategies, and (v) technological advances in glycoscience that have helped to accelerate progress in research. These studies collectively offer valuable insights into new perspectives on antibacterial strategies that may effectively tackle the drug-resistant pathogens that are rapidly spreading globally.
Collapse
|
14
|
Hiyoshi H, English BC, Diaz-Ochoa VE, Wangdi T, Zhang LF, Sakaguchi M, Haneda T, Tsolis RM, Bäumler AJ. Virulence factors perforate the pathogen-containing vacuole to signal efferocytosis. Cell Host Microbe 2022; 30:163-170.e6. [PMID: 34951948 PMCID: PMC8831471 DOI: 10.1016/j.chom.2021.12.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 10/20/2021] [Accepted: 11/30/2021] [Indexed: 12/11/2022]
Abstract
Intracellular pathogens commonly reside within macrophages to find shelter from humoral defenses, but host cell death can expose them to the extracellular milieu. We find intracellular pathogens solve this dilemma by using virulence factors to generate a complement-dependent find-me signal that initiates uptake by a new phagocyte through efferocytosis. During macrophage death, Salmonella uses a type III secretion system to perforate the membrane of the pathogen-containing vacuole (PCV), thereby triggering complement deposition on bacteria entrapped in pore-induced intracellular traps (PITs). In turn, complement activation signals neutrophil efferocytosis, a process that shelters intracellular bacteria from the respiratory burst. Similarly, Brucella employs its type IV secretion system to perforate the PCV membrane, which induces complement deposition on bacteria entrapped in PITs. Collectively, this work identifies virulence factor-induced perforation of the PCV as a strategy of intracellular pathogens to generate a find-me signal for efferocytosis.
Collapse
Affiliation(s)
- Hirotaka Hiyoshi
- Department of Bacteriology, Institute of Tropical Medicine, Nagasaki University, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan; Department of Medical Microbiology and Immunology, University of California at Davis, One Shields Ave, Davis, CA 95616, USA
| | - Bevin C English
- Department of Medical Microbiology and Immunology, University of California at Davis, One Shields Ave, Davis, CA 95616, USA
| | - Vladimir E Diaz-Ochoa
- Department of Medical Microbiology and Immunology, University of California at Davis, One Shields Ave, Davis, CA 95616, USA
| | - Tamding Wangdi
- Department of Medical Microbiology and Immunology, University of California at Davis, One Shields Ave, Davis, CA 95616, USA
| | - Lillian F Zhang
- Department of Medical Microbiology and Immunology, University of California at Davis, One Shields Ave, Davis, CA 95616, USA
| | - Miako Sakaguchi
- Central Laboratory, Institute of Tropical Medicine, Nagasaki University, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan
| | - Takeshi Haneda
- Laboratory of Microbiology, School of Pharmacy, Kitasato University, 5-9-1, Shirokane, Minato-ku, Tokyo 108-8641, Japan
| | - Renée M Tsolis
- Department of Medical Microbiology and Immunology, University of California at Davis, One Shields Ave, Davis, CA 95616, USA
| | - Andreas J Bäumler
- Department of Medical Microbiology and Immunology, University of California at Davis, One Shields Ave, Davis, CA 95616, USA.
| |
Collapse
|
15
|
Sharma A, Alajangi HK, Pisignano G, Sood V, Singh G, Barnwal RP. RNA thermometers and other regulatory elements: Diversity and importance in bacterial pathogenesis. WILEY INTERDISCIPLINARY REVIEWS. RNA 2022; 13:e1711. [PMID: 35037405 DOI: 10.1002/wrna.1711] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Revised: 11/09/2021] [Accepted: 12/16/2021] [Indexed: 01/11/2023]
Abstract
Survival of microorganisms depends to a large extent on environmental conditions and the occupied host. By adopting specific strategies, microorganisms can thrive in the surrounding environment and, at the same time, preserve their viability. Evading the host defenses requires several mechanisms compatible with the host survival which include the production of RNA thermometers to regulate the expression of genes responsible for heat or cold shock as well as of those involved in virulence. Microorganisms have developed a variety of molecules in response to the environmental changes in temperature and even more specifically to the host they invade. Among all, RNA-based regulatory mechanisms are the most common ones, highlighting the importance of such molecules in gene expression control and novel drug development by suitable structure-based alterations. This article is categorized under: RNA Structure and Dynamics > RNA Structure, Dynamics and Chemistry RNA in Disease and Development > RNA in Disease RNA Structure and Dynamics > Influence of RNA Structure in Biological Systems.
Collapse
Affiliation(s)
- Akanksha Sharma
- Department of Biophysics, Panjab University, Chandigarh, India.,University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India
| | - Hema Kumari Alajangi
- Department of Biophysics, Panjab University, Chandigarh, India.,University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India
| | | | - Vikas Sood
- Department of Biochemistry, Jamia Hamdard, New Delhi, India
| | - Gurpal Singh
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India
| | | |
Collapse
|
16
|
Dahora LC, Verheul MK, Williams KL, Jin C, Stockdale L, Cavet G, Giladi E, Hill J, Kim D, Leung Y, Bobay BG, Spicer LD, Sawant S, Rijpkema S, Dennison SM, Alam SM, Pollard AJ, Tomaras GD. Salmonella Typhi Vi capsule prime-boost vaccination induces convergent and functional antibody responses. Sci Immunol 2021; 6:eabj1181. [PMID: 34714686 PMCID: PMC9960181 DOI: 10.1126/sciimmunol.abj1181] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Vaccine development to prevent Salmonella Typhi infections has accelerated over the past decade, resulting in licensure of new vaccines, which use the Vi polysaccharide (Vi PS) of the bacterium conjugated to an unrelated carrier protein as the active component. Antibodies elicited by these vaccines are important for mediating protection against typhoid fever. However, the characteristics of protective and functional Vi antibodies are unknown. In this study, we investigated the human antibody repertoire, avidity maturation, epitope specificity, and function after immunization with a single dose of Vi-tetanus toxoid conjugate vaccine (Vi-TT) and after a booster with plain Vi PS (Vi-PS). The Vi-TT prime induced an IgG1-dominant response, whereas the Vi-TT prime followed by the Vi-PS boost induced IgG1 and IgG2 antibody production. B cells from recipients who received both prime and boost showed evidence of convergence, with shared V gene usage and CDR3 characteristics. The detected Vi antibodies showed heterogeneous avidity ranging from 10 μM to 500 pM, with no evidence of affinity maturation after the boost. Vi-specific antibodies mediated Fc effector functions, which correlated with antibody dissociation kinetics but not with association kinetics. We identified antibodies induced by prime and boost vaccines that recognized subdominant epitopes, indicated by binding to the de–O-acetylated Vi backbone. These antibodies also mediated Fc-dependent functions, such as complement deposition and monocyte phagocytosis. Defining strategies on how to broaden epitope targeting for S. Typhi Vi and enriching for antibody Fc functions that protect against typhoid fever will advance the design of high-efficacy Vi vaccines for protection across diverse populations.
Collapse
Affiliation(s)
- Lindsay C. Dahora
- Center for Human Systems Immunology, Duke University, Durham, NC, USA.,Department of Immunology, Duke University, Durham, NC, USA.,Corresponding author. (L.C.D.); (G.D.T.)
| | - Marije K. Verheul
- Oxford Vaccine Group, Department of Pediatrics, University of Oxford and NIHR Oxford Biomedical Research Center, Oxford, UK
| | | | - Celina Jin
- Oxford Vaccine Group, Department of Pediatrics, University of Oxford and NIHR Oxford Biomedical Research Center, Oxford, UK
| | - Lisa Stockdale
- Oxford Vaccine Group, Department of Pediatrics, University of Oxford and NIHR Oxford Biomedical Research Center, Oxford, UK
| | | | | | - Jennifer Hill
- Oxford Vaccine Group, Department of Pediatrics, University of Oxford and NIHR Oxford Biomedical Research Center, Oxford, UK
| | | | | | - Benjamin G. Bobay
- Department of Biochemistry, Duke University, Durham, NC, USA.,Department of Radiology, Duke University, Durham, NC, USA.,Duke University NMR Center, Duke University Medical Center, Durham, NC, USA
| | - Leonard D. Spicer
- Department of Biochemistry, Duke University, Durham, NC, USA.,Department of Radiology, Duke University, Durham, NC, USA.,Duke University NMR Center, Duke University Medical Center, Durham, NC, USA
| | - Sheetal Sawant
- Center for Human Systems Immunology, Duke University, Durham, NC, USA.,Department of Surgery, Duke University, Durham, NC, USA
| | - Sjoerd Rijpkema
- Division of Bacteriology, National Institute of Biological Standards and Control, Potters Bar, UK
| | - S. Moses Dennison
- Center for Human Systems Immunology, Duke University, Durham, NC, USA.,Department of Surgery, Duke University, Durham, NC, USA
| | - S. Munir Alam
- Department of Medicine, Duke University, Durham, NC, USA.,Department of Pathology, Duke University, Durham, NC, USA.,Duke Human Vaccine Institute, Duke University, Durham, NC, USA
| | - Andrew J. Pollard
- Oxford Vaccine Group, Department of Pediatrics, University of Oxford and NIHR Oxford Biomedical Research Center, Oxford, UK
| | - Georgia D. Tomaras
- Center for Human Systems Immunology, Duke University, Durham, NC, USA.,Department of Immunology, Duke University, Durham, NC, USA.,Department of Surgery, Duke University, Durham, NC, USA.,Department of Molecular Genetics and Microbiology, Duke University, Durham, NC, USA.,Corresponding author. (L.C.D.); (G.D.T.)
| |
Collapse
|
17
|
Johnson M, Stockdale L, de Haan N, Wuhrer M, Nouta J, Koeleman CAM, Clarke J, Marinou S, Shakya M, Colin-Jones R, Theiss-Nyland K, Voysey M, Jin C, Pant D, Jones E, Kelly S, Dongol S, Karkey A, Shrestha S, Basnyat B, Hill J, Pollard AJ. Association of Antibody-Dependent Neutrophil Phagocytosis With Distinct Antibody Glycosylation Profiles Following Typhoid Vaccination. FRONTIERS IN TROPICAL DISEASES 2021. [DOI: 10.3389/fitd.2021.742804] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Typhoid Vi-conjugate vaccines (Vi-TCV) have been developed to control typhoid fever in children in endemic regions. Previously, in a human challenge model of typhoid, Vi-TCV was administered prior to deliberate ingestion of Salmonella Typhi by healthy adult volunteers in the UK. Vi-specific antibody-dependent neutrophil phagocytosis (ADNP) was associated with protection against enteric fever in this model, but it is not known if ADNP is induced by vaccination of children. We measured ADNP in a cohort of Nepalese children receiving a Vi-TCV in a field study to investigate whether functional antibody responses were also present in children in an endemic setting. Furthermore, we investigated relationships between the functional antibody measures and other properties of the antibody response, including Vi-IgG and IgA titres, and Fc region glycosylation. Antibody-dependent neutrophil phagocytosis significantly increased in children aged 9 months to 15 years between the day of vaccination and 28 days following administration of Vi-TCV (D28). The magnitude of ADNP was also comparable with the levels of ADNP induced by plasma from vaccinated UK adults. Neither IgG nor IgA antibody titres significantly correlated with ADNP scores at D28; however, increased vaccine-induced ADNP was associated with decreased levels of IgG1 sialylation. These data suggest that vaccination with Vi-TCV produces functional antibody responses in children, which associate with specific glycosylation patterns of the Fc region.
Collapse
|
18
|
Abstract
Polysaccharides are often the most abundant antigens found on the extracellular surfaces of bacterial cells. These polysaccharides play key roles in interactions with the outside world, and for many bacterial pathogens, they represent what is presented to the human immune system. As a result, many vaccines have been or currently are being developed against carbohydrate antigens. In this review, we explore the diversity of capsular polysaccharides (CPS) in Salmonella and other selected bacterial species and explain the classification and function of CPS as vaccine antigens. Despite many vaccines being developed using carbohydrate antigens, the low immunogenicity and the diversity of infecting strains and serovars present an antigen formulation challenge to manufacturers. Vaccines tend to focus on common serovars or have changing formulations over time, reflecting the trends in human infection, which can be costly and time-consuming. We summarize the approaches to generate carbohydrate-based vaccines for Salmonella, describe vaccines that are in development and emphasize the need for an effective vaccine against non-typhoidal Salmonella strains.
Collapse
|
19
|
Ahn C, Yang YA, Neupane DP, Nguyen T, Richards AF, Sim JH, Mantis NJ, Song J. Mechanisms of typhoid toxin neutralization by antibodies targeting glycan receptor binding and nuclease subunits. iScience 2021; 24:102454. [PMID: 34113815 PMCID: PMC8169802 DOI: 10.1016/j.isci.2021.102454] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Revised: 03/09/2021] [Accepted: 04/19/2021] [Indexed: 12/18/2022] Open
Abstract
Nearly all clinical isolates of Salmonella Typhi, the cause of typhoid fever, are antibiotic resistant. All S. Typhi isolates secrete an A2B5 exotoxin called typhoid toxin to benefit the pathogen during infection. Here, we demonstrate that antibiotic-resistant S. Typhi secretes typhoid toxin continuously during infection regardless of antibiotic treatment. We characterize typhoid toxin antibodies targeting glycan-receptor-binding PltB or nuclease CdtB, which neutralize typhoid toxin in vitro and in vivo, as demonstrated by using typhoid toxin secreted by antibiotic-resistant S. Typhi during human cell infection and lethal dose typhoid toxin challenge to mice. TyTx11 generated in this study neutralizes typhoid toxin effectively, comparable to TyTx4 that binds to all PltB subunits available per holotoxin. Cryoelectron microscopy explains that the binding of TyTx11 to CdtB makes this subunit inactive through CdtB catalytic-site conformational change. The identified toxin-neutralizing epitopes are conserved across all S. Typhi clinical isolates, offering critical insights into typhoid toxin-neutralizing strategies.
Collapse
Affiliation(s)
- Changhwan Ahn
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| | - Yi-An Yang
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| | - Durga P. Neupane
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| | - Tri Nguyen
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| | | | - Ji Hyun Sim
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| | - Nicholas J. Mantis
- Department of Biomedical Sciences, University at Albany, Albany, NY 12222, USA
- Division of Infectious Diseases, Wadsworth Center, New York State Department of Health, Albany, NY 12208, USA
| | - Jeongmin Song
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| |
Collapse
|
20
|
Enteric Fever Diagnosis: Current Challenges and Future Directions. Pathogens 2021; 10:pathogens10040410. [PMID: 33915749 PMCID: PMC8065732 DOI: 10.3390/pathogens10040410] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 03/23/2021] [Accepted: 03/27/2021] [Indexed: 12/21/2022] Open
Abstract
Enteric fever is a life-threatening systemic febrile disease caused by Salmonella enterica serovars Typhi and Paratyphi (S. Typhi and S. Paratyphi). Unfortunately, the burden of the disease remains high primarily due to the global spread of various drug-resistant Salmonella strains despite continuous advancement in the field. An accurate diagnosis is critical for effective control of the disease. However, enteric fever diagnosis based on clinical presentations is challenging due to overlapping symptoms with other febrile illnesses that are also prevalent in endemic areas. Current laboratory tests display suboptimal sensitivity and specificity, and no diagnostic methods are available for identifying asymptomatic carriers. Several research programs have employed systemic approaches to identify more specific biomarkers for early detection and asymptomatic carrier detection. This review discusses the pros and cons of currently available diagnostic tests for enteric fever, the advancement of research toward improved diagnostic tests, and the challenges of discovering new ideal biomarkers and tests.
Collapse
|
21
|
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
|
22
|
Brewer SM, Twittenhoff C, Kortmann J, Brubaker SW, Honeycutt J, Massis LM, Pham THM, Narberhaus F, Monack DM. A Salmonella Typhi RNA thermosensor regulates virulence factors and innate immune evasion in response to host temperature. PLoS Pathog 2021; 17:e1009345. [PMID: 33651854 PMCID: PMC7954313 DOI: 10.1371/journal.ppat.1009345] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Revised: 03/12/2021] [Accepted: 01/28/2021] [Indexed: 12/20/2022] Open
Abstract
Sensing and responding to environmental signals is critical for bacterial pathogens to successfully infect and persist within hosts. Many bacterial pathogens sense temperature as an indication they have entered a new host and must alter their virulence factor expression to evade immune detection. Using secondary structure prediction, we identified an RNA thermosensor (RNAT) in the 5' untranslated region (UTR) of tviA encoded by the typhoid fever-causing bacterium Salmonella enterica serovar Typhi (S. Typhi). Importantly, tviA is a transcriptional regulator of the critical virulence factors Vi capsule, flagellin, and type III secretion system-1 expression. By introducing point mutations to alter the mRNA secondary structure, we demonstrate that the 5' UTR of tviA contains a functional RNAT using in vitro expression, structure probing, and ribosome binding methods. Mutational inhibition of the RNAT in S. Typhi causes aberrant virulence factor expression, leading to enhanced innate immune responses during infection. In conclusion, we show that S. Typhi regulates virulence factor expression through an RNAT in the 5' UTR of tviA. Our findings demonstrate that limiting inflammation through RNAT-dependent regulation in response to host body temperature is important for S. Typhi's "stealthy" pathogenesis.
Collapse
Affiliation(s)
- Susan M. Brewer
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California, United States of America
| | | | - Jens Kortmann
- Genentech, Inc., South San Francisco, California, United States of America
| | - Sky W. Brubaker
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California, United States of America
| | - Jared Honeycutt
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California, United States of America
| | - Liliana Moura Massis
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California, United States of America
| | - Trung H. M. Pham
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California, United States of America
| | | | - Denise M. Monack
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California, United States of America
| |
Collapse
|
23
|
Harrell JE, Hahn MM, D'Souza SJ, Vasicek EM, Sandala JL, Gunn JS, McLachlan JB. Salmonella Biofilm Formation, Chronic Infection, and Immunity Within the Intestine and Hepatobiliary Tract. Front Cell Infect Microbiol 2021; 10:624622. [PMID: 33604308 PMCID: PMC7885405 DOI: 10.3389/fcimb.2020.624622] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Accepted: 12/17/2020] [Indexed: 12/12/2022] Open
Abstract
Within the species of Salmonella enterica, there is significant diversity represented among the numerous subspecies and serovars. Collectively, these account for microbes with variable host ranges, from common plant and animal colonizers to extremely pathogenic and human-specific serovars. Despite these differences, many Salmonella species find commonality in the ability to form biofilms and the ability to cause acute, latent, or chronic disease. The exact outcome of infection depends on many factors such as the growth state of Salmonella, the environmental conditions encountered at the time of infection, as well as the infected host and immune response elicited. Here, we review the numerous biofilm lifestyles of Salmonella (on biotic and abiotic surfaces) and how the production of extracellular polymeric substances not only enhances long-term persistence outside the host but also is an essential function in chronic human infections. Furthermore, careful consideration is made for the events during initial infection that allow for gut transcytosis which, in conjunction with host immune functions, often determine the progression of disease. Both typhoidal and non-typhoidal salmonellae can cause chronic and/or secondary infections, thus the adaptive immune responses to both types of bacteria are discussed with particular attention to the differences between Salmonella Typhi, Salmonella Typhimurium, and invasive non-typhoidal Salmonella that can result in differential immune responses. Finally, while strides have been made in our understanding of immunity to Salmonella in the lymphoid organs, fewer definitive studies exist for intestinal and hepatobiliary immunity. By examining our current knowledge and what remains to be determined, we provide insight into new directions in the field of Salmonella immunity, particularly as it relates to chronic infection.
Collapse
Affiliation(s)
- Jaikin E Harrell
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA, United States
| | - Mark M Hahn
- Center for Microbial Pathogenesis, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, United States.,Infectious Diseases Institute, The Ohio State University, Columbus, OH, United States
| | - Shaina J D'Souza
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA, United States
| | - Erin M Vasicek
- Center for Microbial Pathogenesis, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, United States.,Infectious Diseases Institute, The Ohio State University, Columbus, OH, United States
| | - Jenna L Sandala
- Center for Microbial Pathogenesis, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, United States.,Infectious Diseases Institute, The Ohio State University, Columbus, OH, United States
| | - John S Gunn
- Center for Microbial Pathogenesis, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, United States.,Infectious Diseases Institute, The Ohio State University, Columbus, OH, United States.,Department of Pediatrics, College of Medicine, The Ohio State University, Columbus, OH, United States
| | - James B McLachlan
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA, United States
| |
Collapse
|
24
|
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
|
25
|
Salmonella Extracellular Polymeric Substances Modulate Innate Phagocyte Activity and Enhance Tolerance of Biofilm-Associated Bacteria to Oxidative Stress. Microorganisms 2020; 8:microorganisms8020253. [PMID: 32070067 DOI: 10.3390/microorganisms8020253] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 01/28/2020] [Accepted: 02/10/2020] [Indexed: 12/13/2022] Open
Abstract
Salmonella enterica serovar Typhi causes 14.3 million acute cases of typhoid fever that are responsible for 136,000 deaths each year. Chronic infections occur in 3%-5% of those infected and S. Typhi persists primarily in the gallbladder by forming biofilms on cholesterol gallstones, but how these bacterial communities evade host immunity is not known. Salmonella biofilms produce several extracellular polymeric substances (EPSs) during chronic infection, which are hypothesized to prevent pathogen clearance either by protecting biofilm-associated bacteria from direct humoral attack or by modulating innate phagocyte interaction with biofilms. Using wild-type and EPS-deficient planktonic and biofilm Salmonella, the direct attack hypothesis was tested by challenging biofilms with human serum and antimicrobial peptides. Biofilms were found to be tolerant to these molecules, but these phenotypes were independent of the tested EPSs. By examining macrophage and neutrophil responses, new roles for biofilm-associated capsular polysaccharides and slime polysaccharides were identified. The S. Typhi Vi antigen was found to modulate innate immunity by reducing macrophage nitric oxide production and neutrophil reactive oxygen species (ROS) production. The slime polysaccharides colanic acid and cellulose were found to be immune-stimulating and represent a key difference between non-typhoidal serovars and typhoidal serovars, which do not express colanic acid. Furthermore, biofilm tolerance to the exogenously-supplied ROS intermediates hydrogen peroxide (H2O2) and hypochlorite (ClO) indicated an additional role of the capsular polysaccharides for both serovars in recalcitrance to H2O2 but not ClO, providing new understanding of the stalemate that arises during chronic infections and offering new directions for mechanistic and clinical studies.
Collapse
|
26
|
Dahora LC, Jin C, Spreng RL, Feely F, Mathura R, Seaton KE, Zhang L, Hill J, Jones E, Alam SM, Dennison SM, Pollard AJ, Tomaras GD. IgA and IgG1 Specific to Vi Polysaccharide of Salmonella Typhi Correlate With Protection Status in a Typhoid Fever Controlled Human Infection Model. Front Immunol 2019; 10:2582. [PMID: 31781100 PMCID: PMC6852708 DOI: 10.3389/fimmu.2019.02582] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 10/17/2019] [Indexed: 01/09/2023] Open
Abstract
Vaccination against Salmonella Typhi using the Vi capsular polysaccharide, a T-cell independent antigen, can protect from the development of typhoid fever. This implies that antibodies to Vi alone can protect in the absence of a T cell-mediated immune response; however, protective Vi antibodies have not been well-characterized. We hypothesized that variability in the biophysical properties of vaccine-elicited antibodies, including subclass distribution and avidity, may impact protective outcomes. To interrogate the relationship between antibody properties and protection against typhoid fever, we analyzed humoral responses from participants in a vaccine efficacy (VE) trial using a controlled human infection model (CHIM) who received either a purified Vi polysaccharide (Vi-PS) or Vi tetanus toxoid conjugate (Vi-TT) vaccine followed by oral challenge with live S. Typhi. We determined the avidity, overall magnitude, and vaccine-induced fold-change in magnitude from before immunization to day of challenge of Vi IgA and IgG subclass antibodies. Amongst those who received the Vi-PS vaccine, Vi IgA magnitude (FDR p = 0.01) and fold-change (FDR p = 0.02) were significantly higher in protected individuals compared with those individuals who developed disease ("diagnosed"). In the Vi-TT vaccine group, the responses of protected individuals had higher fold-change in Vi IgA (FDR p = 0.06) and higher Vi IgG1 avidity (FDR p = 0.058) than the diagnosed Vi-TT vaccinees, though these findings were not significant at p < 0.05. Overall, protective antibody signatures differed between the Vi-PS and Vi-TT vaccines, thus, we conclude that although the Vi-PS and Vi-TT vaccines were observed to have similar efficacies, these vaccines may protect through different mechanisms. These data will inform studies on mechanisms of protection against typhoid fever, including identification of antibody effector functions, as well as informing future vaccination strategies.
Collapse
Affiliation(s)
- Lindsay C Dahora
- Duke Human Vaccine Institute, Duke University, Durham, NC, United States.,Department of Immunology, Duke University, Durham, NC, United States
| | - Celina Jin
- Oxford Vaccine Group, Department of Paediatrics, The NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
| | - Rachel L Spreng
- Duke Human Vaccine Institute, Duke University, Durham, NC, United States.,Department of Medicine, Duke University, Durham, NC, United States
| | - Frederick Feely
- Duke Human Vaccine Institute, Duke University, Durham, NC, United States.,Department of Surgery, Duke University, Durham, NC, United States
| | - Ryan Mathura
- Duke Human Vaccine Institute, Duke University, Durham, NC, United States.,Department of Surgery, Duke University, Durham, NC, United States
| | - Kelly E Seaton
- Duke Human Vaccine Institute, Duke University, Durham, NC, United States.,Department of Surgery, Duke University, Durham, NC, United States
| | - Lu Zhang
- Duke Human Vaccine Institute, Duke University, Durham, NC, United States.,Department of Surgery, Duke University, Durham, NC, United States
| | - Jennifer Hill
- Oxford Vaccine Group, Department of Paediatrics, The NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
| | - Elizabeth Jones
- Oxford Vaccine Group, Department of Paediatrics, The NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
| | - S Munir Alam
- Duke Human Vaccine Institute, Duke University, Durham, NC, United States.,Department of Medicine, Duke University, Durham, NC, United States.,Department of Pathology, Duke University, Durham, NC, United States
| | - S Moses Dennison
- Duke Human Vaccine Institute, Duke University, Durham, NC, United States.,Department of Surgery, Duke University, Durham, NC, United States
| | - Andrew J Pollard
- Oxford Vaccine Group, Department of Paediatrics, The NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
| | - Georgia D Tomaras
- Duke Human Vaccine Institute, Duke University, Durham, NC, United States.,Department of Immunology, Duke University, Durham, NC, United States.,Department of Surgery, Duke University, Durham, NC, United States.,Molecular Genetics and Microbiology, Duke University, Durham, NC, United States
| |
Collapse
|
27
|
Salerno-Goncalves R, Kayastha D, Fasano A, Levine MM, Sztein MB. Crosstalk between leukocytes triggers differential immune responses against Salmonella enterica serovars Typhi and Paratyphi. PLoS Negl Trop Dis 2019; 13:e0007650. [PMID: 31412039 PMCID: PMC6709971 DOI: 10.1371/journal.pntd.0007650] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 08/26/2019] [Accepted: 07/23/2019] [Indexed: 12/30/2022] Open
Abstract
Enteric fevers, caused by the Salmonella enterica serovars Typhi (ST), Paratyphi A (PA) and Paratyphi B (PB), are life-threatening illnesses exhibiting very similar clinical symptoms but with distinct epidemiologies, geographical distributions and susceptibilities to antimicrobial treatment. Nevertheless, the mechanisms by which the host recognizes pathogens with high levels of homology, such as these bacterial serovars, remain poorly understood. Using a three-dimensional organotypic model of the human intestinal mucosa and PA, PB, and ST, we observed significant differences in the secretion patterns of pro-inflammatory cytokines and chemokines elicited by these serovars. These cytokines/chemokines were likely to be co-regulated and influenced the function of epithelial cells, such as the production of IL-8. We also found differing levels of polymorphonuclear leukocyte (PMN) migration among various infection conditions that either included or excluded lymphocytes and macrophages (Mϕ), strongly suggesting feedback mechanisms among these cells. Blocking experiments showed that IL-1β, IL-6, IL-8, TNF-α and CCL3 cytokines were involved in the differential regulation of migration patterns. We conclude that the crosstalk among the lymphocytes, Mϕ, PMN and epithelial cells is cytokine/chemokine-dependent and bacterial-serotype specific, and plays a pivotal role in orchestrating the functional efficiency of the innate cells and migratory characteristics of the leukocytes.
Collapse
Affiliation(s)
- Rosangela Salerno-Goncalves
- Center for Vaccine Development and Global Health, Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, United States of America
| | - Darpan Kayastha
- Center for Vaccine Development and Global Health, Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, United States of America
| | - Alessio Fasano
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital for Children, Boston, MA, United States of America
| | - Myron M. Levine
- Center for Vaccine Development and Global Health, Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, United States of America
| | - Marcelo B. Sztein
- Center for Vaccine Development and Global Health, Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, United States of America
| |
Collapse
|
28
|
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
|
29
|
Hausmann A, Hardt WD. The Interplay between Salmonella enterica Serovar Typhimurium and the Intestinal Mucosa during Oral Infection. Microbiol Spectr 2019; 7:10.1128/microbiolspec.bai-0004-2019. [PMID: 30953432 PMCID: PMC11588296 DOI: 10.1128/microbiolspec.bai-0004-2019] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Indexed: 12/28/2022] Open
Abstract
Bacterial infection results in a dynamic interplay between the pathogen and its host. The underlying interactions are multilayered, and the cellular responses are modulated by the local environment. The intestine is a particularly interesting tissue regarding host-pathogen interaction. It is densely colonized by commensal microbes and a portal of entry for ingested pathogens. This necessitates constant monitoring of microbial stimuli in order to maintain homeostasis during encounters with benign microbiota and to trigger immune defenses in response to bacterial pathogens. Homeostasis is maintained by physical barriers (the mucus layer and epithelium), chemical defenses (antimicrobial peptides), and innate immune responses (NLRC4 inflammasome), which keep the bacteria from reaching the sterile lamina propria. Intestinal pathogens represent potent experimental tools to probe these barriers and decipher how pathogens can circumvent them. The streptomycin mouse model of oral Salmonella enterica serovar Typhimurium infection provides a well-characterized, robust experimental system for such studies. Strikingly, each stage of the gut tissue infection poses a different set of challenges to the pathogen and requires tight control of virulence factor expression, host response modulation, and cooperation between phenotypic subpopulations. Therefore, successful infection of the intestinal tissue relies on a delicate and dynamic balance between responses of the pathogen and its host. These mechanisms can be deciphered to their full extent only in realistic in vivo infection models.
Collapse
Affiliation(s)
- Annika Hausmann
- Institute of Microbiology, D-BIOL ETH Zurich, Zurich, Switzerland
| | | |
Collapse
|
30
|
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
|
31
|
Francis EA, Heinrich V. Mechanistic Understanding of Single-Cell Behavior is Essential for Transformative Advances in Biomedicine. THE YALE JOURNAL OF BIOLOGY AND MEDICINE 2018; 91:279-289. [PMID: 30258315 PMCID: PMC6153630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Most current efforts to advance medical technology proceed along one of two tracks. The first is dedicated to the improvement of clinical tasks through the incremental refinement of medical instruments. The second comprises engineering endeavors to support basic science studies that often only remotely relate to human medicine. Here we survey emerging research approaches that aim to populate the sprawling frontier between these tracks. We focus on interdisciplinary single-live-cell techniques that have overcome limitations of traditional biological methods to successfully address vital questions about medically relevant cellular behavior. Most of the presented case studies are based on the controlled manipulation of nonadherent human immune cells using one or more micropipettes. The included studies have (i) examined one-on-one encounters of immune cells with real or model pathogens, (ii) assessed the physiological role of the expandable surface area of immune cells, and (iii) started to dissect the spatiotemporal organization of signaling processes within these cells. The unique aptitude of such single-live-cell studies to fill conspicuous gaps in our quantitative understanding of medically relevant cause-effect relationships provides a sound basis for new insights that will inform and drive future biomedical innovation.
Collapse
Affiliation(s)
| | - Volkmar Heinrich
- To whom all correspondence should be addressed: Volkmar Heinrich, Department of Biomedical Engineering, University of California, Davis, 451 Health Sciences Drive, Davis, CA 95616; Tel: 530-754-6644,
| |
Collapse
|
32
|
Johnson R, Mylona E, Frankel G. TyphoidalSalmonella: Distinctive virulence factors and pathogenesis. Cell Microbiol 2018; 20:e12939. [DOI: 10.1111/cmi.12939] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 07/06/2018] [Accepted: 07/13/2018] [Indexed: 01/13/2023]
Affiliation(s)
- Rebecca Johnson
- MRC Centre for Molecular Bacteriology and Infection, Department of Life Sciences; Imperial College London; London UK
| | - Elli Mylona
- MRC Centre for Molecular Bacteriology and Infection, Department of Life Sciences; Imperial College London; London UK
| | - Gad Frankel
- MRC Centre for Molecular Bacteriology and Infection, Department of Life Sciences; Imperial College London; London UK
| |
Collapse
|
33
|
Why Is Eradicating Typhoid Fever So Challenging: Implications for Vaccine and Therapeutic Design. Vaccines (Basel) 2018; 6:vaccines6030045. [PMID: 30042307 PMCID: PMC6160957 DOI: 10.3390/vaccines6030045] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Revised: 07/19/2018] [Accepted: 07/23/2018] [Indexed: 01/22/2023] Open
Abstract
Salmonella enterica serovar Typhi (S. Typhi) and S. Paratyphi, namely typhoidal Salmonellae, are the cause of (para) typhoid fever, which is a devastating systemic infectious disease in humans. In addition, the spread of multidrug-resistant (MDR) and extensively drug-resistant (XDR) S. Typhi in many low and middle-income countries poses a significant risk to human health. While currently available typhoid vaccines and therapeutics are efficacious, they have some limitations. One important limitation is the lack of controlling individuals who chronically carry S. Typhi. However, due to the strict host specificity of S. Typhi to humans, S. Typhi research is hampered. As a result, our understanding of S. Typhi pathogenesis is incomplete, thereby delaying the development and improvement of prevention and treatment strategies. Nonetheless, to better combat and contain S. Typhi, it is vital to develop a vaccine and therapy for controlling both acutely and chronically infected individuals. This review discusses how scientists are trying to combat typhoid fever, why it is so challenging to do so, which approaches show promise, and what we know about the pathogenesis of S. Typhi chronic infection.
Collapse
|
34
|
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
|
35
|
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
|
36
|
Pandya KD, Palomo-Caturla I, Walker JA, K Sandilya V, Zhong Z, Alugupalli KR. An Unmutated IgM Response to the Vi Polysaccharide of Salmonella Typhi Contributes to Protective Immunity in a Murine Model of Typhoid. THE JOURNAL OF IMMUNOLOGY 2018; 200:4078-4084. [PMID: 29743315 DOI: 10.4049/jimmunol.1701348] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Accepted: 04/16/2018] [Indexed: 11/19/2022]
Abstract
T cell-dependent B cell responses typically develop in germinal centers. Abs generated during such responses are isotype switched and have a high affinity to the Ag because of somatic hypermutation of Ab genes. B cell responses to purified polysaccharides are T cell independent and do not result in the formation of bona fide germinal centers, and the dominant Ab isotype produced during such responses is IgM with very few or no somatic mutations. Activation-induced cytidine deaminase (AID) is required for both somatic hypermutation and Ig isotype switching in humans and mice. To test the extent to which unmutated polysaccharide-specific IgM confers protective immunity, we immunized wildtype and AID-/- mice with either heat-killed Salmonella enterica serovar Typhi (S. Typhi) or purified Vi polysaccharide (ViPS). We found that wildtype and AID-/- mice immunized with heat-killed S. Typhi generated similar anti-ViPS IgM responses. As expected, wildtype, but not AID-/- mice generated ViPS-specific IgG. However, the differences in the Ab-dependent killing of S. Typhi mediated by the classical pathway of complement activation were not statistically significant. In ViPS-immunized wildtype and AID-/- mice, the ViPS-specific IgM levels and S. Typhi bactericidal Ab titers at 7 but not at 28 d postimmunization were also comparable. To test the protective immunity conferred by these immunizations, mice were challenged with a chimeric S. Typhimurium strain expressing ViPS. Compared with their naive counterparts, immunized wildtype and AID-/- mice exhibited significantly reduced bacterial burden regardless of the route of infection. These data indicate that an unmutated IgM response to ViPS contributes to protective immunity to S. Typhi.
Collapse
Affiliation(s)
- Kalgi D Pandya
- Department of Microbiology and Immunology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107; and
| | - Isabel Palomo-Caturla
- Department of Microbiology and Immunology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107; and
| | - Justin A Walker
- Department of Microbiology and Immunology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107; and
| | - Vijay K Sandilya
- Department of Microbiology and Immunology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107; and
| | - Zhijiu Zhong
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107
| | - Kishore R Alugupalli
- Department of Microbiology and Immunology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107; and .,Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107
| |
Collapse
|
37
|
Francis EA, Heinrich V. Extension of chemotactic pseudopods by nonadherent human neutrophils does not require or cause calcium bursts. Sci Signal 2018. [PMID: 29535263 DOI: 10.1126/scisignal.aal4289] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Global bursts in free intracellular calcium (Ca2+) are among the most conspicuous signaling events in immune cells. To test the common view that Ca2+ bursts mediate rearrangement of the actin cytoskeleton in response to the activation of G protein-coupled receptors, we combined single-cell manipulation with fluorescence imaging and monitored the Ca2+ concentration in individual human neutrophils during complement-mediated chemotaxis. By decoupling purely chemotactic pseudopod formation from cell-substrate adhesion, we showed that physiological concentrations of anaphylatoxins, such as C5a, induced nonadherent human neutrophils to form chemotactic pseudopods but did not elicit Ca2+ bursts. By contrast, pathological or supraphysiological concentrations of C5a often triggered Ca2+ bursts, but pseudopod protrusion stalled or reversed in such cases, effectively halting chemotaxis, similar to sepsis-associated neutrophil paralysis. The maximum increase in cell surface area during pseudopod extension in pure chemotaxis was much smaller-by a factor of 8-than the known capacity of adherent human neutrophils to expand their surface. Because the measured rise in cortical tension was not sufficient to account for this difference, we attribute the limited deformability to a reduced ability of the cytoskeleton to generate protrusive force in the absence of cell adhesion. Thus, we hypothesize that Ca2+ bursts in neutrophils control a mechanistic switch between two distinct modes of cytoskeletal organization and dynamics. A key element of this switch appears to be the expedient coordination of adhesion-dependent lock or release events of cytoskeletal membrane anchors.
Collapse
Affiliation(s)
- Emmet A Francis
- Department of Biomedical Engineering, University of California Davis, Davis, CA 95616, USA
| | - Volkmar Heinrich
- Department of Biomedical Engineering, University of California Davis, Davis, CA 95616, USA.
| |
Collapse
|
38
|
Dufresne K, Saulnier-Bellemare J, Daigle F. Functional Analysis of the Chaperone-Usher Fimbrial Gene Clusters of Salmonella enterica serovar Typhi. Front Cell Infect Microbiol 2018; 8:26. [PMID: 29473020 PMCID: PMC5809473 DOI: 10.3389/fcimb.2018.00026] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Accepted: 01/19/2018] [Indexed: 01/11/2023] Open
Abstract
The human-specific pathogen Salmonella enterica serovar Typhi causes typhoid, a major public health issue in developing countries. Several aspects of its pathogenesis are still poorly understood. S. Typhi possesses 14 fimbrial gene clusters including 12 chaperone-usher fimbriae (stg, sth, bcf, fim, saf, sef, sta, stb, stc, std, ste, and tcf). These fimbriae are weakly expressed in laboratory conditions and only a few are actually characterized. In this study, expression of all S. Typhi chaperone-usher fimbriae and their potential roles in pathogenesis such as interaction with host cells, motility, or biofilm formation were assessed. All S. Typhi fimbriae were better expressed in minimal broth. Each system was overexpressed and only the fimbrial gene clusters without pseudogenes demonstrated a putative major subunits of about 17 kDa on SDS-PAGE. Six of these (Fim, Saf, Sta, Stb, Std, and Tcf) also show extracellular structure by electron microscopy. The impact of fimbrial deletion in a wild-type strain or addition of each individual fimbrial system to an S. Typhi afimbrial strain were tested for interactions with host cells, biofilm formation and motility. Several fimbriae modified bacterial interactions with human cells (THP-1 and INT-407) and biofilm formation. However, only Fim fimbriae had a deleterious effect on motility when overexpressed. Overall, chaperone-usher fimbriae seem to be an important part of the balance between the different steps (motility, adhesion, host invasion and persistence) of S. Typhi pathogenesis.
Collapse
Affiliation(s)
- Karine Dufresne
- Department of Microbiology, Infectiology and Immunology, Université de Montréal, Montreal, QC, Canada
| | - Julie Saulnier-Bellemare
- Department of Microbiology, Infectiology and Immunology, Université de Montréal, Montreal, QC, Canada
| | - France Daigle
- Department of Microbiology, Infectiology and Immunology, Université de Montréal, Montreal, QC, Canada
| |
Collapse
|
39
|
Reciprocal Regulation of OmpR and Hfq and Their Regulatory Actions on the Vi Polysaccharide Capsular Antigen in Salmonella enterica Serovar Typhi. Curr Microbiol 2018; 75:773-778. [DOI: 10.1007/s00284-018-1447-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2017] [Accepted: 01/31/2018] [Indexed: 11/30/2022]
|
40
|
MacKenzie KD, Palmer MB, Köster WL, White AP. Examining the Link between Biofilm Formation and the Ability of Pathogenic Salmonella Strains to Colonize Multiple Host Species. Front Vet Sci 2017; 4:138. [PMID: 29159172 PMCID: PMC5581909 DOI: 10.3389/fvets.2017.00138] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2017] [Accepted: 08/09/2017] [Indexed: 12/11/2022] Open
Abstract
Salmonella are important pathogens worldwide and a predominant number of human infections are zoonotic in nature. The ability of strains to form biofilms, which is a multicellular behavior characterized by the aggregation of cells, is predicted to be a conserved strategy for increased persistence and survival. It may also contribute to the increasing number of infections caused by ingestion of contaminated fruits and vegetables. There is a correlation between biofilm formation and the ability of strains to colonize and replicate within the intestines of multiple host species. These strains predominantly cause localized gastroenteritis infections in humans. In contrast, there are salmonellae that cause systemic, disseminated infections in a select few host species; these “invasive” strains have a narrowed host range, and most are unable to form biofilms. This includes host-restricted Salmonella serovar Typhi, which are only able to infect humans, and atypical gastroenteritis strains associated with the opportunistic infection of immunocompromised patients. From the perspective of transmission, biofilm formation is advantageous for ensuring pathogen survival in the environment. However, from an infection point of view, biofilm formation may be an anti-virulence trait. We do not know if the capacity to form biofilms prevents a strain from accessing the systemic compartments within the host or if loss of the biofilm phenotype reflects a change in a strain’s interaction with the host. In this review, we examine the connections between biofilm formation, Salmonella disease states, degrees of host adaptation, and how this might relate to different transmission patterns. A better understanding of the dynamic lifecycle of Salmonella will allow us to reduce the burden of livestock and human infections caused by these important pathogens.
Collapse
Affiliation(s)
- Keith D MacKenzie
- Vaccine and Infectious Disease Organization-International Vaccine Centre, Saskatoon, SK, Canada.,Department of Microbiology and Immunology, University of Saskatchewan, Saskatoon, SK, Canada
| | - Melissa B Palmer
- Vaccine and Infectious Disease Organization-International Vaccine Centre, Saskatoon, SK, Canada.,Department of Microbiology and Immunology, University of Saskatchewan, Saskatoon, SK, Canada
| | - Wolfgang L Köster
- Vaccine and Infectious Disease Organization-International Vaccine Centre, Saskatoon, SK, Canada.,Department of Veterinary Microbiology, University of Saskatchewan, Saskatoon, SK, Canada
| | - Aaron P White
- Vaccine and Infectious Disease Organization-International Vaccine Centre, Saskatoon, SK, Canada.,Department of Microbiology and Immunology, University of Saskatchewan, Saskatoon, SK, Canada
| |
Collapse
|
41
|
Abstract
Many bacterial pathogens can cause acute infections that are cleared with the onset of adaptive immunity, but a subset of these pathogens can establish persistent, and sometimes lifelong, infections. While bacteria that cause chronic infections are phylogenetically diverse, they share common features in their interactions with the host that enable a protracted period of colonization. This article will compare the persistence strategies of two chronic pathogens from the Proteobacteria, Brucella abortus and Salmonella enterica serovar Typhi, to consider how these two pathogens, which are very different at the genomic level, can utilize common strategies to evade immune clearance to cause chronic intracellular infections of the mononuclear phagocyte system.
Collapse
|
42
|
Abstract
Salmonella enterica subspecies enterica includes several serovars infecting both humans and other animals and leading to typhoid fever or gastroenteritis. The high prevalence of associated morbidity and mortality, together with an increased emergence of multidrug-resistant strains, is a current global health issue that has prompted the development of vaccination strategies that confer protection against most serovars. Currently available systemic vaccine approaches have major limitations, including a reduced effectiveness in young children and a lack of cross-protection among different strains. Having studied host-pathogen interactions, microbiologists and immunologists argue in favor of topical gastrointestinal administration for improvement in vaccine efficacy. Here, recent advances in this field are summarized, including mechanisms of bacterial uptake at the intestinal epithelium, the assessment of protective host immunity, and improved animal models that closely mimic infection in humans. The pros and cons of existing vaccines are presented, along with recent progress made with novel formulations. Finally, new candidate antigens and their relevance in the refined design of anti-Salmonella vaccines are discussed, along with antigen vectorization strategies such as nanoparticles or secretory immunoglobulins, with a focus on potentiating mucosal vaccine efficacy.
Collapse
|
43
|
Chong A, Lee S, Yang YA, Song J. The Role of Typhoid Toxin in Salmonella Typhi Virulence
. THE YALE JOURNAL OF BIOLOGY AND MEDICINE 2017; 90:283-290. [PMID: 28656014 PMCID: PMC5482304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/29/2022]
Abstract
Unlike many of the nontyphoidal Salmonella serovars such as S. Typhimurium that cause restricted gastroenteritis, Salmonella Typhi is unique in that it causes life-threatening typhoid fever in humans. Despite the vast difference in disease outcomes that S. Typhi and S. Typhimurium cause in humans, there are few genomic regions that are unique to S. Typhi. Of these regions, the most notable is the small locus encoding typhoid toxin, an AB toxin that has several distinct characteristics that contribute to S. Typhi's pathogenicity. As a result, typhoid toxin and its role in S. Typhi virulence have been studied in an effort to gain insight into potential treatment and prevention strategies. Given the rise of multidrug-resistant strains, research in this area has become increasingly important. This article discusses the current understanding of typhoid toxin and potential directions for future research endeavors in order to better understand the contribution of typhoid toxin to S. Typhi virulence.
Collapse
Affiliation(s)
- Alexander Chong
- Department of Microbiology and Immunology, Cornell University College of Veterinary Medicine, Ithaca, NY
| | - Sohyoung Lee
- Department of Microbiology and Immunology, Cornell University College of Veterinary Medicine, Ithaca, NY
| | - Yi-An Yang
- Department of Microbiology and Immunology, Cornell University College of Veterinary Medicine, Ithaca, NY
| | - Jeongmin Song
- Department of Microbiology and Immunology, Cornell University College of Veterinary Medicine, Ithaca, NY
| |
Collapse
|
44
|
Heinrich V, Simpson WD, Francis EA. Analytical Prediction of the Spatiotemporal Distribution of Chemoattractants around Their Source: Theory and Application to Complement-Mediated Chemotaxis. Front Immunol 2017; 8:578. [PMID: 28603522 PMCID: PMC5445147 DOI: 10.3389/fimmu.2017.00578] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 05/01/2017] [Indexed: 11/13/2022] Open
Abstract
The ability of motile immune cells to detect and follow gradients of chemoattractant is critical to numerous vital functions, including their recruitment to sites of infection and-in emerging immunotherapeutic applications-to malignant tumors. Facilitated by a multitude of chemotactic receptors, the cells navigate a maze of stimuli to home in on their target. Distinct chemotactic processes direct this navigation at particular times and cell-target distances. The expedient coordination of this spatiotemporal hierarchy of chemotactic stages is the central element of a key paradigm of immunotaxis. Understanding this hierarchy is an enormous interdisciplinary challenge that requires, among others, quantitative insight into the shape, range, and dynamics of the profiles of chemoattractants around their sources. We here present a closed-form solution to a diffusion-reaction problem that describes the evolution of the concentration gradient of chemoattractant under various conditions. Our ready-to-use mathematical prescription captures many biological situations reasonably well and can be explored with standard graphing software, making it a valuable resource for every researcher studying chemotaxis. We here apply this mathematical model to characterize the chemoattractant cloud of anaphylatoxins that forms around bacterial and fungal pathogens in the presence of host serum. We analyze the spatial reach, rate of formation, and rate of dispersal of this locator cloud under realistic physiological conditions. Our analysis predicts that simply being small is an effective protective strategy of pathogens against complement-mediated discovery by host immune cells over moderate-to-large distances. Leveraging our predictions against single-cell, pure-chemotaxis experiments that use human immune cells as biosensors, we are able to explain the limited distance over which the cells recognize microbes. We conclude that complement-mediated chemotaxis is a universal, but short-range, homing mechanism by which chemotaxing immune cells can implement a last-minute course correction toward pathogenic microbes. Thus, the integration of theory and experiments provides a sound mechanistic explanation of the primary role of complement-mediated chemotaxis within the hierarchy of immunotaxis, and why other chemotactic processes are required for the successful recruitment of immune cells over large distances.
Collapse
Affiliation(s)
- Volkmar Heinrich
- Department of Biomedical Engineering, University of California at Davis, Davis, CA, United States
| | - Wooten D Simpson
- Department of Biomedical Engineering, University of California at Davis, Davis, CA, United States
| | - Emmet A Francis
- Department of Biomedical Engineering, University of California at Davis, Davis, CA, United States
| |
Collapse
|
45
|
An insider's perspective: Bacteroides as a window into the microbiome. Nat Microbiol 2017; 2:17026. [PMID: 28440278 DOI: 10.1038/nmicrobiol.2017.26] [Citation(s) in RCA: 399] [Impact Index Per Article: 49.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 01/31/2017] [Indexed: 12/22/2022]
Abstract
Over the last decade, our appreciation for the contribution of resident gut microorganisms-the gut microbiota-to human health has surged. However, progress is limited by the sheer diversity and complexity of these microbial communities. Compounding the challenge, the majority of our commensal microorganisms are not close relatives of Escherichia coli or other model organisms and have eluded culturing and manipulation in the laboratory. In this Review, we discuss how over a century of study of the readily cultured, genetically tractable human gut Bacteroides has revealed important insights into the biochemistry, genomics and ecology that make a gut bacterium a gut bacterium. While genome and metagenome sequences are being produced at breakneck speed, the Bacteroides provide a significant 'jump-start' on uncovering the guiding principles that govern microbiota-host and inter-bacterial associations in the gut that will probably extend to many other members of this ecosystem.
Collapse
|
46
|
Rivera-Chávez F, Lopez CA, Bäumler AJ. Oxygen as a driver of gut dysbiosis. Free Radic Biol Med 2017; 105:93-101. [PMID: 27677568 DOI: 10.1016/j.freeradbiomed.2016.09.022] [Citation(s) in RCA: 198] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 09/09/2016] [Accepted: 09/23/2016] [Indexed: 12/13/2022]
Abstract
Changes in the composition of gut-associated microbial communities may underlie many inflammatory and allergic diseases. However, the processes that help maintain a stable community structure are poorly understood. Here we review topical work elucidating the nutrient-niche occupied by facultative anaerobic bacteria of the family Enterobacteriaceae, whose predominance within the gut-associated microbial community is a common marker of dysbiosis. A paucity of exogenous respiratory electron acceptors limits growth of Enterobacteriaceae within a balanced gut-associated microbial community. However, recent studies suggest that the availability of oxygen in the large bowel is markedly elevated by changes in host physiology that accompany antibiotic treatment or infection with enteric pathogens, such as Salmonella serovars or attaching and effacing (AE) pathogens. The resulting increase in oxygen availability, alone or in conjunction with other electron acceptors, drives an uncontrolled luminal expansion of Enterobacteriaceae. Insights into the underlying mechanisms provide important clues about factors that control the balance between the host and its resident microbial communities.
Collapse
Affiliation(s)
- Fabian Rivera-Chávez
- Department of Medical Microbiology and Immunology, School of Medicine, University of California Davis, One Shields Ave, Davis, CA 95616, USA
| | - Christopher A Lopez
- Department of Medical Microbiology and Immunology, School of Medicine, University of California Davis, One Shields Ave, Davis, CA 95616, USA
| | - Andreas J Bäumler
- Department of Medical Microbiology and Immunology, School of Medicine, University of California Davis, One Shields Ave, Davis, CA 95616, USA.
| |
Collapse
|
47
|
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
|
48
|
Quantifying the Sensitivity of Human Immune Cells to Chemoattractant. Biophys J 2017; 112:834-837. [PMID: 28185642 DOI: 10.1016/j.bpj.2017.01.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Revised: 01/07/2017] [Accepted: 01/13/2017] [Indexed: 11/21/2022] Open
Abstract
The efficient recruitment of immune cells is a vital cornerstone of our defense against infections and a key challenge of immunotherapeutic applications. It relies on the ability of chemotaxing cells to prioritize their responses to different stimuli. For example, immune cells are known to abandon gradients of host-cell-produced cytokines in favor of complement-derived anaphylatoxins, which then guide the cells toward nearby pathogen surfaces. The aptitude to triage stimuli depends on the cells' specific sensitivities to different chemoattractants. We here use human neutrophils as uniquely capable biodetectors to map out the anaphylatoxic cloud that surrounds microbes in the presence of host serum. We quantify the neutrophil sensitivity in terms of the ratio between the chemoattractant concentration c and the production rate j0 of the chemoattractant at the source surface. An integrative experimental/theoretical approach allows us to estimate the c/j0-threshold at which human neutrophils first detect nearby β-glucan surfaces as c/j0 ≈ 0.0044 s/μm.
Collapse
|
49
|
Hu X, Chen Z, Xiong K, Wang J, Rao X, Cong Y. Vi capsular polysaccharide: Synthesis, virulence, and application. Crit Rev Microbiol 2016; 43:440-452. [PMID: 27869515 DOI: 10.1080/1040841x.2016.1249335] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Vi capsular polysaccharide, a linear homopolymer of α-1,4-linked N-acetylgalactosaminuronate, is characteristically produced by Salmonella enterica serovar Typhi. The Vi capsule covers the surface of the producing bacteria and serves as an virulence factor via inhibition of complement-mediated killing and promoting resistance against phagocytosis. Furthermore, Vi also represents a predominant protective antigen and plays a key role in the development of vaccines against typhoid fever. Herein, we reviewed the latest advances associated with the Vi polysaccharide, from its synthesis and transport within bacterial cells, mechanisms involved in virulence, immunological characteristics, and applications in vaccine, as well as its purification and detection methods.
Collapse
Affiliation(s)
- Xiaomei Hu
- a Department of Microbiology , Third Military Medical University , Chongqing , China
| | - Zhijin Chen
- a Department of Microbiology , Third Military Medical University , Chongqing , China
| | - Kun Xiong
- a Department of Microbiology , Third Military Medical University , Chongqing , China
| | - Jing Wang
- a Department of Microbiology , Third Military Medical University , Chongqing , China
| | - Xiancai Rao
- a Department of Microbiology , Third Military Medical University , Chongqing , China
| | - Yanguang Cong
- a Department of Microbiology , Third Military Medical University , Chongqing , China
| |
Collapse
|
50
|
Galen JE, Buskirk AD, Tennant SM, Pasetti MF. Live Attenuated Human Salmonella Vaccine Candidates: Tracking the Pathogen in Natural Infection and Stimulation of Host Immunity. EcoSal Plus 2016; 7:10.1128/ecosalplus.ESP-0010-2016. [PMID: 27809955 PMCID: PMC5119766 DOI: 10.1128/ecosalplus.esp-0010-2016] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Indexed: 04/08/2023]
Abstract
Salmonellosis, caused by members of the genus Salmonella, is responsible for considerable global morbidity and mortality in both animals and humans. In this review, we will discuss the pathogenesis of Salmonella enterica serovar Typhi and Salmonella enterica serovar Typhimurium, focusing on human Salmonella infections. We will trace the path of Salmonella through the body, including host entry sites, tissues and organs affected, and mechanisms involved in both pathogenesis and stimulation of host immunity. Careful consideration of the natural progression of disease provides an important context in which attenuated live oral vaccines can be rationally designed and developed. With this in mind, we will describe a series of attenuated live oral vaccines that have been successfully tested in clinical trials and demonstrated to be both safe and highly immunogenic. The attenuation strategies summarized in this review offer important insights into further development of attenuated vaccines against other Salmonella for which live oral candidates are currently unavailable.
Collapse
Affiliation(s)
- James E Galen
- Center for Vaccine Development, Institute for Global Health, University of Maryland School of Medicine, Baltimore, MD 21201
| | - Amanda D Buskirk
- Center for Vaccine Development, Institute for Global Health, University of Maryland School of Medicine, Baltimore MD 21201
| | - Sharon M Tennant
- Center for Vaccine Development, Institute for Global Health, University of Maryland School of Medicine, Baltimore MD 21201
| | - Marcela F Pasetti
- Center for Vaccine Development, Institute for Global Health, University of Maryland School of Medicine, Baltimore MD 21201
| |
Collapse
|