1
|
Allen JC, Natta SS, Nasrin S, Toapanta FR, Tennant SM. Deletion of an immune evasion gene, steD, from a live Salmonella enterica serovar Typhimurium vaccine improves vaccine responses in aged mice. Front Immunol 2024; 15:1376734. [PMID: 38911854 PMCID: PMC11190192 DOI: 10.3389/fimmu.2024.1376734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 05/07/2024] [Indexed: 06/25/2024] Open
Abstract
Introduction Non-typhoidal Salmonella (NTS) generally causes self-limiting gastroenteritis. However, older adults (≥65 years) can experience more severe outcomes from NTS infection. We have previously shown that a live attenuated S. Typhimurium vaccine, CVD 1926 (I77 ΔguaBA ΔclpP ΔpipA ΔhtrA), was immunogenic in adult but not aged mice. Here we describe modification of CVD 1926 through deletion of steD, a Salmonella effector responsible for host immune escape, which we hypothesized would increase immunogenicity in aged mice. Methods Mel Juso and/or mutuDC cells were infected with S. Typhimurium I77, CVD 1926, and their respective steD mutants, and the MHC-II levels were evaluated. Aged (18-month-old) C57BL/6 mice received two doses of PBS, CVD 1926, or CVD 1926 ΔsteD perorally (109 CFU) and the number of FliC-specific CD4+ T cells were determined. Lastly, aged C57BL/6 mice received three doses of PBS, CVD 1926, or CVD 1926 ΔsteD perorally (109 CFU) and then were challenged perorally with wild-type S. Typhimurium SL1344 (108 CFU). These animals were also evaluated for antibody responses. Results MHC-II induction was higher in cells treated with steD mutants, compared to their respective parental strains. Compared to PBS-vaccinated mice, CVD 1926 ΔsteD elicited significantly more FliC-specific CD4+ T cells in the Peyer's Patches. There were no significant differences in FliC-specific CD4+ T cells in the Peyer's patches or spleen of CVD 1926- versus PBS-immunized mice. CVD 1926 and CVD 1926 ΔsteD induced similar serum and fecal anti-core and O polysaccharide antibody titers after three doses. After two immunizations, the proportion of seroconverters for CVD 1926 ΔsteD was 83% (10/12) compared to 42% (5/12) for CVD 1926. Compared to PBS-immunized mice, mice immunized with CVD 1926 ΔsteD had significantly lower S. Typhimurium counts in the spleen, cecum, and small intestine upon challenge. In contrast, there were no differences in bacterial loads in the tissues of PBS-vaccinated and CVD 1926-immunized animals. Conclusion These data suggest that the steD deletion enhanced the immunogenicity of our live attenuated S. Typhimurium vaccine. Deletion of immune evasion genes could be a potential strategy to improve the immunogenicity of live attenuated vaccines in older adults.
Collapse
Affiliation(s)
- Jessica C. Allen
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, United States
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Shanaliz S. Natta
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, United States
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Shamima Nasrin
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, United States
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Franklin R. Toapanta
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, United States
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Sharon M. Tennant
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, United States
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
| |
Collapse
|
2
|
Wang N, Scott TA, Kupz A, Shreenivas MM, Peres NG, Hocking DM, Yang C, Jebeli L, Beattie L, Groom JR, Pierce TP, Wakim LM, Bedoui S, Strugnell RA. Vaccine-induced inflammation and inflammatory monocytes promote CD4+ T cell-dependent immunity against murine salmonellosis. PLoS Pathog 2023; 19:e1011666. [PMID: 37733817 PMCID: PMC10547166 DOI: 10.1371/journal.ppat.1011666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 10/03/2023] [Accepted: 09/07/2023] [Indexed: 09/23/2023] Open
Abstract
Prior infection can generate protective immunity against subsequent infection, although the efficacy of such immunity can vary considerably. Live-attenuated vaccines (LAVs) are one of the most effective methods for mimicking this natural process, and analysis of their efficacy has proven instrumental in the identification of protective immune mechanisms. Here, we address the question of what makes a LAV efficacious by characterising immune responses to a LAV, termed TAS2010, which is highly protective (80-90%) against lethal murine salmonellosis, in comparison with a moderately protective (40-50%) LAV, BRD509. Mice vaccinated with TAS2010 developed immunity systemically and were protected against gut-associated virulent infection in a CD4+ T cell-dependent manner. TAS2010-vaccinated mice showed increased activation of Th1 responses compared with their BRD509-vaccinated counterparts, leading to increased Th1 memory populations in both lymphoid and non-lymphoid organs. The optimal development of Th1-driven immunity was closely correlated with the activation of CD11b+Ly6GnegLy6Chi inflammatory monocytes (IMs), the activation of which can be modulated proportionally by bacterial load in vivo. Upon vaccination with the LAV, IMs expressed T cell chemoattractant CXCL9 that attracted CD4+ T cells to the foci of infection, where IMs also served as a potent source of antigen presentation and Th1-promoting cytokine IL-12. The expression of MHC-II in IMs was rapidly upregulated following vaccination and then maintained at an elevated level in immune mice, suggesting IMs may have a role in sustained antigen stimulation. Our findings present a longitudinal analysis of CD4+ T cell development post-vaccination with an intracellular bacterial LAV, and highlight the benefit of inflammation in the development of Th1 immunity. Future studies focusing on the induction of IMs may reveal key strategies for improving vaccine-induced T cell immunity.
Collapse
Affiliation(s)
- Nancy Wang
- Department of Microbiology and Immunology, The University of Melbourne, at Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Timothy A. Scott
- Department of Microbiology and Immunology, The University of Melbourne, at Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Andreas Kupz
- Department of Microbiology and Immunology, The University of Melbourne, at Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Meghanashree M. Shreenivas
- Department of Microbiology and Immunology, The University of Melbourne, at Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Newton G. Peres
- Department of Microbiology and Immunology, The University of Melbourne, at Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Dianna M. Hocking
- Department of Microbiology and Immunology, The University of Melbourne, at Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Chenying Yang
- Department of Microbiology and Immunology, The University of Melbourne, at Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Leila Jebeli
- Department of Microbiology and Immunology, The University of Melbourne, at Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Lynette Beattie
- Department of Microbiology and Immunology, The University of Melbourne, at Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Joanna R. Groom
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Thomas P. Pierce
- Ludwig Institute for Cancer Research, Melbourne-Parkville Branch, Parkville, Victoria, Australia
| | - Linda M. Wakim
- Department of Microbiology and Immunology, The University of Melbourne, at Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Sammy Bedoui
- Department of Microbiology and Immunology, The University of Melbourne, at Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Richard A. Strugnell
- Department of Microbiology and Immunology, The University of Melbourne, at Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| |
Collapse
|
3
|
Marcial-Juárez E, Pérez-Toledo M, Nayar S, Pipi E, Alshayea A, Persaud R, Jossi SE, Lamerton R, Barone F, Henderson IR, Cunningham AF. Salmonella infection induces the reorganization of follicular dendritic cell networks concomitant with the failure to generate germinal centers. iScience 2023; 26:106310. [PMID: 36950118 PMCID: PMC10025972 DOI: 10.1016/j.isci.2023.106310] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 02/07/2023] [Accepted: 02/26/2023] [Indexed: 03/06/2023] Open
Abstract
Germinal centers (GCs) are sites where plasma and memory B cells form to generate high-affinity, Ig class-switched antibodies. Specialized stromal cells called follicular dendritic cells (FDCs) are essential for GC formation. During systemic Salmonella Typhimurium (STm) infection GCs are absent, whereas extensive extrafollicular and switched antibody responses are maintained. The mechanisms that underpin the absence of GC formation are incompletely understood. Here, we demonstrate that STm induces a reversible disruption of niches within the splenic microenvironment, including the T and B cell compartments and the marginal zone. Alongside these effects after infection, mature FDC networks are strikingly absent, whereas immature FDC precursors, including marginal sinus pre-FDCs (MadCAM-1+) and perivascular pre-FDCs (PDGFRβ+) are enriched. As normal FDC networks re-establish, extensive GCs become detectable throughout the spleen. Therefore, the reorganization of FDC networks and the loss of GC responses are key, parallel features of systemic STm infections.
Collapse
Affiliation(s)
- Edith Marcial-Juárez
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, West Midlands, B15 2TT, United Kingdom
| | - Marisol Pérez-Toledo
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, West Midlands, B15 2TT, United Kingdom
| | - Saba Nayar
- Rheumatology Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, West Midlands, B15 2TT, United Kingdom
| | - Elena Pipi
- Rheumatology Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, West Midlands, B15 2TT, United Kingdom
| | - Areej Alshayea
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, West Midlands, B15 2TT, United Kingdom
| | - Ruby Persaud
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, West Midlands, B15 2TT, United Kingdom
| | - Sian E. Jossi
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, West Midlands, B15 2TT, United Kingdom
| | - Rachel Lamerton
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, West Midlands, B15 2TT, United Kingdom
| | - Francesca Barone
- Rheumatology Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, West Midlands, B15 2TT, United Kingdom
- National Institute for Health Research (NIHR) Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust, UK and Sandwell and West Birmingham Trust, Birmingham, West Midlands, B15 2TH, United Kingdom
| | - Ian R. Henderson
- Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD4072, Australia
| | - Adam F. Cunningham
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, West Midlands, B15 2TT, United Kingdom
| |
Collapse
|
4
|
CD4+ T cell immunity to Salmonella is transient in the circulation. PLoS Pathog 2021; 17:e1010004. [PMID: 34695149 PMCID: PMC8568161 DOI: 10.1371/journal.ppat.1010004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 11/04/2021] [Accepted: 10/04/2021] [Indexed: 12/12/2022] Open
Abstract
While Salmonella enterica is seen as an archetypal facultative intracellular bacterial pathogen where protection is mediated by CD4+ T cells, identifying circulating protective cells has proved very difficult, inhibiting steps to identify key antigen specificities. Exploiting a mouse model of vaccination, we show that the spleens of C57BL/6 mice vaccinated with live-attenuated Salmonella serovar Typhimurium (S. Typhimurium) strains carried a pool of IFN-γ+ CD4+ T cells that could adoptively transfer protection, but only transiently. Circulating Salmonella-reactive CD4+ T cells expressed the liver-homing chemokine receptor CXCR6, accumulated over time in the liver and assumed phenotypic characteristics associated with tissue-associated T cells. Liver memory CD4+ T cells showed TCR selection bias and their accumulation in the liver could be inhibited by blocking CXCL16. These data showed that the circulation of CD4+ T cells mediating immunity to Salmonella is limited to a brief window after which Salmonella-specific CD4+ T cells migrate to peripheral tissues. Our observations highlight the importance of triggering tissue-specific immunity against systemic infections. Helper T cells are essential for controlling infections by bacterial pathogens, such as Salmonella enterica var Typhimurium (S. Typhimurium). While it is well-established that this role is related to their provision of IFN-γ, when and where helper T cells elicit their protective function in vivo remains unresolved. We identified a protective helper T cell population in the circulation of mice early after inoculation with growth-attenuated S. Typhimurium strains; this population waned overtime. We observed that circulating helper T cell immunity can adoptively protect naïve recipient mice against lethal S. Typhimurium infection when harvested from a short time-window. In comparing helper T cell responses between spleen and liver in Salmonella-infected mice, we have observed a previously uncharacterized trafficking of helper T cells to the liver followed by the residence of S. Typhimurium-specific T cell memory in the organ. Taken together these findings identify that protective immunity to Salmonella infections is transient in the circulation and the liver as a preferential site of helper T memory cells.
Collapse
|
5
|
Comparison of Lethal and Nonlethal Mouse Models of Orientia tsutsugamushi Infection Reveals T-Cell Population-Associated Cytokine Signatures Correlated with Lethality and Protection. Trop Med Infect Dis 2021; 6:tropicalmed6030121. [PMID: 34287349 PMCID: PMC8293330 DOI: 10.3390/tropicalmed6030121] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 06/25/2021] [Accepted: 06/30/2021] [Indexed: 11/19/2022] Open
Abstract
The antigenic diversity of Orientia tsutsugamushi as well as the interstrain difference(s) associated with virulence in mice impose the necessity to dissect the host immune response. In this study we compared the host response in lethal and non-lethal murine models of O. tsutsugamushi infection using the two strains, Karp (New Guinea) and Woods (Australia). The models included the lethal model: Karp intraperitoneal (IP) challenge; and the nonlethal models: Karp intradermal (ID), Woods IP, and Woods ID challenges. We monitored bacterial trafficking to the liver, lung, spleen, kidney, heart, and blood, and seroconversion during the 21-day challenge. Bacterial trafficking to all organs was observed in both the lethal and nonlethal models of infection, with significant increases in average bacterial loads observed in the livers and hearts of the lethal model. Multicolor flow cytometry was utilized to analyze the CD4+ and CD8+ T cell populations and their intracellular production of the cytokines IFNγ, TNF, and IL2 (single, double, and triple combinations) associated with both the lethal and nonlethal murine models of infection. The lethal model was defined by a cytokine signature of double- (IFNγ-IL2) and triple-producing (IL2-TNF-IFNγ) CD4+ T-cell populations; no multifunctional signature was identified in the CD8+ T-cell populations associated with the lethal model. In the nonlethal model, the cytokine signature was predominated by CD4+ and CD8+ T-cell populations associated with single (IL2) and/or double (IL2-TNF) populations of producers. The cytokine signatures associated with our lethal model will become depletion targets in future experiments; those signatures associated with our nonlethal model are hypothesized to be related to the protective nature of the nonlethal challenges.
Collapse
|
6
|
Fetal Macrophages Exposed to Salmonella Antigens Elicit Protective Immunity Against Overwhelming Salmonella Challenge in A Murine Model. Biomedicines 2021; 9:biomedicines9030245. [PMID: 33804435 PMCID: PMC8001423 DOI: 10.3390/biomedicines9030245] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 02/23/2021] [Accepted: 02/24/2021] [Indexed: 11/16/2022] Open
Abstract
Despite the evidence for fetal immunization following maternal infection, it remained a mystery how the fetal immune system was primed by vertically-transmitted pathogens or microbial antigens, especially before its full maturation. We previously demonstrated the capacity of fetal macrophages for endocytosing oncoprotein and allergens to bridge towards adaptive immunity in postnatal life. To investigate the immunological consequences of fetal contact with microbial antigens and the role of fetal macrophages in the defense against infection before T-cell development, we exposed gestational day 14 murine fetuses and their macrophages to flagellin and heat-killed Salmonella Typhimurium. Recipients with in utero exposure to Salmonella antigens or adoptive transfer of microbial antigen-loaded fetal macrophages were examined for immune responses to Salmonella antigens and resistance to virulent Salmonella challenge. Fetal exposure to microbial antigens or adoptive transfer of microbial antigen-loaded fetal macrophages could confer antigen-specific adaptive immunity. However, protective immunity against lethal Salmonella challenge was only granted to those receiving heat-killed Salmonella antigens, presenting as heightened recall responses of serum anti-lipopolysaccharide immunoglobulins and interferon-gamma. In immunized recipients surviving Salmonella challenge, their serum transfer to succeeding recipients provided immediate protection from lethal Salmonella challenge in preference to lymphocyte transfer, indicating a more active role of humoral immunity in the prevention of Salmonella invasiveness. Our study sheds insight on the role of fetal macrophages in immunogenicity to transplacental pathogens regardless of fetal lymphocyte maturity, paving the way for fetal macrophage therapies to enhance vaccine responsiveness or increase resistance to pathogenic microorganisms in perinatal life.
Collapse
|
7
|
Modifying bacterial flagellin to evade Nod-like Receptor CARD 4 recognition enhances protective immunity against Salmonella. Nat Microbiol 2020; 5:1588-1597. [PMID: 33106673 DOI: 10.1038/s41564-020-00801-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Accepted: 09/17/2020] [Indexed: 02/06/2023]
Abstract
Pattern recognition receptors (PRRs) expressed in antigen-presenting cells are thought to shape pathogen-specific immunity by inducing secretion of costimulatory cytokines during T-cell activation, yet data to support this notion in vivo are scarce. Here, we show that the cytosolic PRR Nod-like Receptor CARD 4 (NLRC4) suppresses, rather than facilitates, effector and memory CD4+ T-cell responses against Salmonella in mice. NLRC4 negatively regulates immunological memory by preventing delayed activation of the cytosolic PRR NLR pyrin domain 3 (NLRP3) that would otherwise amplify the production of cytokines important for the generation of Th1 immunity such as intereukin-18. Consistent with a role for NLRC4 in memory immunity, primary challenge with Salmonella expressing flagellin modified to largely evade NLRC4 recognition notably increases protection against lethal rechallenge. This finding suggests flagellin modification to reduce NLRC4 activation enhances protective immunity, which could have important implications for vaccine development against flagellated microbial pathogens.
Collapse
|
8
|
Rapaka RR, Wahid R, Fresnay S, Booth JS, Darton TC, Jones C, Waddington CS, Levine MM, Pollard AJ, Sztein MB. Human Salmonella Typhi exposure generates differential multifunctional cross-reactive T-cell memory responses against Salmonella Paratyphi and invasive nontyphoidal Salmonella. Clin Transl Immunology 2020; 9:e1178. [PMID: 33005416 PMCID: PMC7512505 DOI: 10.1002/cti2.1178] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 08/16/2020] [Accepted: 08/17/2020] [Indexed: 12/19/2022] Open
Abstract
Objective There are no vaccines for most of the major invasive Salmonella strains causing severe infection in humans. We evaluated the specificity of adaptive T memory cell responses generated after Salmonella Typhi exposure in humans against other major invasive Salmonella strains sharing capacity for dissemination. Methods T memory cells from eleven volunteers who underwent controlled oral challenge with wtS. Typhi were characterised by flow cytometry for cross‐reactive cellular cytokine/chemokine effector responses or evidence of degranulation upon stimulation with autologous B‐lymphoblastoid cells infected with either S. Typhi, Salmonella Paratyphi A (PA), S. Paratyphi B (PB) or an invasive nontyphoidal Salmonella strain of the S. Typhimurium serovar (iNTSTy). Results Blood T‐cell effector memory (TEM) responses after exposure to S. Typhi in humans evolve late, peaking weeks after infection in most volunteers. Induced multifunctional CD4+ Th1 and CD8+ TEM cells elicited after S. Typhi challenge were cross‐reactive with PA, PB and iNTSTy. The magnitude of multifunctional CD4+ TEM cell responses to S. Typhi correlated with induction of cross‐reactive multifunctional CD8+ TEM cells against PA, PB and iNTSTy. Highly multifunctional subsets and T central memory and T effector memory cells that re‐express CD45 (TEMRA) demonstrated less heterologous T‐cell cross‐reactivity, and multifunctional Th17 elicited after S. Typhi challenge was not cross‐reactive against other invasive Salmonella. Conclusion Gaps in cross‐reactive immune effector functions in human T‐cell memory compartments were highly dependent on invasive Salmonella strain, underscoring the importance of strain‐dependent vaccination in the design of T‐cell‐based vaccines for invasive Salmonella.
Collapse
Affiliation(s)
- Rekha R Rapaka
- Center for Vaccine Development and Global Health University of Maryland School of Medicine Baltimore MD USA.,Department of Medicine University of Maryland School of Medicine Baltimore MD USA
| | - Rezwanul Wahid
- Center for Vaccine Development and Global Health University of Maryland School of Medicine Baltimore MD USA.,Department of Pediatrics University of Maryland School of Medicine Baltimore MD USA
| | - Stephanie Fresnay
- Center for Vaccine Development and Global Health University of Maryland School of Medicine Baltimore MD USA.,Department of Pediatrics University of Maryland School of Medicine Baltimore MD USA.,Present address: Stephanie Fresnay GlaxoSmithKline Rockville MD USA
| | - Jayaum S Booth
- Center for Vaccine Development and Global Health University of Maryland School of Medicine Baltimore MD USA.,Department of Pediatrics University of Maryland School of Medicine Baltimore MD USA
| | - Thomas C Darton
- Oxford Vaccine Group Department of Paediatrics University of Oxford and the NIHR Oxford Biomedical Research Centre Oxford UK.,Present address: Thomas C Darton University of Sheffield Medical School Sheffield UK
| | - Claire Jones
- Oxford Vaccine Group Department of Paediatrics University of Oxford and the NIHR Oxford Biomedical Research Centre Oxford UK
| | - Claire S Waddington
- Oxford Vaccine Group Department of Paediatrics University of Oxford and the NIHR Oxford Biomedical Research Centre Oxford UK.,Present address: University of Cambridge Cambridge UK
| | - Myron M Levine
- Center for Vaccine Development and Global Health University of Maryland School of Medicine Baltimore MD USA.,Department of Medicine University of Maryland School of Medicine Baltimore MD USA.,Department of Pediatrics University of Maryland School of Medicine Baltimore MD USA
| | - Andrew J Pollard
- Oxford Vaccine Group Department of Paediatrics University of Oxford and the NIHR Oxford Biomedical Research Centre Oxford UK
| | - Marcelo B Sztein
- Center for Vaccine Development and Global Health University of Maryland School of Medicine Baltimore MD USA.,Department of Medicine University of Maryland School of Medicine Baltimore MD USA.,Department of Pediatrics University of Maryland School of Medicine Baltimore MD USA
| |
Collapse
|
9
|
Antibodies and Protection in Systemic Salmonella Infections: Do We Still Have More Questions than Answers? Infect Immun 2020; 88:IAI.00219-20. [PMID: 32601109 DOI: 10.1128/iai.00219-20] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Salmonella causes grave systemic infections in humans and other animals and provides a paradigm for other diseases in which the bacteria have both intracellular and extracellular lifestyles. New generations of vaccines rely on the essential contribution of the antibody responses for their protection. The quality, antigen specificity, and functions associated with antibody responses to this pathogen have been elusive for a long time. Recent approaches that combine studies in humans and genetically manipulated experimental models and that exploit awareness of the location and within-host life cycle of the pathogen are shedding light on how humoral immunity to Salmonella operates. However, this area of research remains full of controversy and discrepancies. The overall scenario indicates that antibodies are essential for resistance against systemic Salmonella infections and can express the highest protective function when operating in conjunction with cell-mediated immunity. Antigen specificity, isotype profile, Fc-gamma receptor usage, and complement activation are all intertwined factors that still arcanely influence antibody-mediated protection to Salmonella.
Collapse
|
10
|
Rossi O, Vlazaki M, Kanvatirth P, Restif O, Mastroeni P. Within-host spatiotemporal dynamic of systemic salmonellosis: Ways to track infection, reaction to vaccination and antimicrobial treatment. J Microbiol Methods 2020; 176:106008. [PMID: 32707153 DOI: 10.1016/j.mimet.2020.106008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Revised: 07/13/2020] [Accepted: 07/17/2020] [Indexed: 12/16/2022]
Abstract
During the last two decades our understanding of the complex in vivo host-pathogen interactions has increased due to technical improvements and new research tools. The rapid advancement of molecular biology, flow cytometry and microscopy techniques, combined with mathematical modelling, have empowered in-depth studies of systemic bacterial infections across scales from single molecules, to cells, to organs and systems to reach the whole organism level. By tracking subpopulations of bacteria in vivo using molecular or fluorescent tags, it has been possible to reconstruct the spread of infection within and between organs, allowing unprecedented quantification of the effects of antimicrobial treatment and vaccination. This review illustrates recent advances in the study of heterogeneous traits of the infection process and illustrate approaches to investigate the reciprocal interactions between antimicrobial treatments, bacterial growth/death as well as inter- and intra-organ spread. We also discuss how vaccines impact the in vivo behaviour of bacteria and how these findings can guide vaccine design and rational antimicrobial treatment.
Collapse
Affiliation(s)
- Omar Rossi
- University of Cambridge, Department of Veterinary Medicine, Madingley Road, CB3 0ES Cambridge, UK.
| | - Myrto Vlazaki
- University of Cambridge, Department of Veterinary Medicine, Madingley Road, CB3 0ES Cambridge, UK
| | - Panchali Kanvatirth
- University of Cambridge, Department of Veterinary Medicine, Madingley Road, CB3 0ES Cambridge, UK
| | - Olivier Restif
- University of Cambridge, Department of Veterinary Medicine, Madingley Road, CB3 0ES Cambridge, UK
| | - Pietro Mastroeni
- University of Cambridge, Department of Veterinary Medicine, Madingley Road, CB3 0ES Cambridge, UK.
| |
Collapse
|
11
|
Wang M, Qazi IH, Wang L, Zhou G, Han H. Salmonella Virulence and Immune Escape. Microorganisms 2020; 8:microorganisms8030407. [PMID: 32183199 PMCID: PMC7143636 DOI: 10.3390/microorganisms8030407] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 03/02/2020] [Accepted: 03/10/2020] [Indexed: 02/07/2023] Open
Abstract
Salmonella genus represents the most common foodborne pathogens causing morbidity, mortality, and burden of disease in all regions of the world. The introduction of antimicrobial agents and Salmonella-specific phages has been considered as an effective intervention strategy to reduce Salmonella contamination. However, data from the United States, European countries, and low- and middle-income countries indicate that Salmonella cases are still a commonly encountered cause of bacterial foodborne diseases globally. The control programs have not been successful and even led to the emergence of some multidrug-resistant Salmonella strains. It is known that the host immune system is able to effectively prevent microbial invasion and eliminate microorganisms. However, Salmonella has evolved mechanisms of resisting host physical barriers and inhibiting subsequent activation of immune response through their virulence factors. There has been a high interest in understanding how Salmonella interacts with the host. Therefore, in the present review, we characterize the functions of Salmonella virulence genes and particularly focus on the mechanisms of immune escape in light of evidence from the emerging mainstream literature.
Collapse
Affiliation(s)
- Mengyao Wang
- Beijing Key Laboratory for Animal Genetic Improvement, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (M.W.); (L.W.)
- Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Izhar Hyder Qazi
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China;
- Department of Veterinary Anatomy and Histology, Shaheed Benazir Bhutto University of Veterinary and Animal Sciences, Sakrand 67210, Pakistan
| | - Linli Wang
- Beijing Key Laboratory for Animal Genetic Improvement, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (M.W.); (L.W.)
- Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Guangbin Zhou
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China;
- Correspondence: (H.H.); (G.Z.)
| | - Hongbing Han
- Beijing Key Laboratory for Animal Genetic Improvement, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (M.W.); (L.W.)
- Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
- Correspondence: (H.H.); (G.Z.)
| |
Collapse
|
12
|
Salerno-Gonçalves R, Tettelin H, Luo D, Guo Q, Ardito MT, Martin WD, De Groot AS, Sztein MB. Differential functional patterns of memory CD4 + and CD8 + T-cells from volunteers immunized with Ty21a typhoid vaccine observed using a recombinant Escherichia coli system expressing S. Typhi proteins. Vaccine 2019; 38:258-270. [PMID: 31629569 DOI: 10.1016/j.vaccine.2019.10.020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 10/03/2019] [Accepted: 10/07/2019] [Indexed: 02/01/2023]
Abstract
It is widely accepted that CD4+ and CD8+ T-cells play a significant role in protection against Salmonella enterica serovar Typhi (S. Typhi), the causative agent of the typhoid fever. However, the antigen specificity of these T-cells remains largely unknown. Previously, we demonstrated the feasibility of using a recombinant Escherichia coli (E. coli) expression system to uncover the antigen specificity of CD4+ and CD8+ T cells. Here, we expanded these studies to include the evaluation of 12 additional S. Typhi proteins: 4 outer membrane proteins (OmpH, OmpL, OmpR, OmpX), 3 Vi-polysaccharide biosynthesis proteins (TviA, TviB, TviE), 3 cold shock proteins (CspA, CspB, CspC), and 2 conserved hypothetical proteins (Chp 1 and Chp2), all selected based on the bioinformatic analyses of the content of putative T-cell epitopes. CD4+ and CD8+ T cells from 15 adult volunteers, obtained before and 42 days after immunization with oral live attenuated Ty21a vaccine, were assessed for their functionality (i.e., production of cytokines and cytotoxic expression markers in response to stimulation with selected antigens) as measured by flow cytometry. Although volunteers differed on their T-cell antigen specificity, we observed T-cell immune responses against all S. Typhi proteins evaluated. These responses included 9 proteins, OmpH, OmpR, TviA, TviE, CspA, CspB, CspC, Chp 1 and Chp 2, which have not been previously reported to elicit T-cell responses. Interestingly, we also observed that, regardless of the protein, the functional patterns of the memory T-cells were different between CD4+ and CD8+ T cells. In sum, these studies demonstrated the feasibility of using bioinformatic analysis and the E. coli expressing system described here to uncover novel immunogenic T-cell proteins that could serve as potential targets for the production of protein-based vaccines.
Collapse
Affiliation(s)
- Rosângela Salerno-Gonçalves
- Center for Vaccine Development and Global Health (CVD), Department of Pediatrics, University of Maryland School of Medicine, 685 West Baltimore Street, HSF1, Baltimore, MD 21201, USA.
| | - Hervé Tettelin
- Department of Microbiology and Immunology and Institute for Genome Sciences (IGS), University of Maryland School of Medicine, 670 West Baltimore Street, HSF3, Baltimore, MD 21201, USA
| | - David Luo
- Center for Vaccine Development and Global Health (CVD), Department of Pediatrics, University of Maryland School of Medicine, 685 West Baltimore Street, HSF1, Baltimore, MD 21201, USA
| | - Qin Guo
- Department of Microbiology and Immunology and Institute for Genome Sciences (IGS), University of Maryland School of Medicine, 670 West Baltimore Street, HSF3, Baltimore, MD 21201, USA
| | - Matthew T Ardito
- Institute for Immunology and Informatics (iCubed), Department of Cell and Molecular Biology, University of Rhode Island, 80 Washington Street, Providence, RI, USA; EpiVax, Inc., 188 Valley Street Suite 424, Providence, RI, USA
| | - William D Martin
- Institute for Immunology and Informatics (iCubed), Department of Cell and Molecular Biology, University of Rhode Island, 80 Washington Street, Providence, RI, USA; EpiVax, Inc., 188 Valley Street Suite 424, Providence, RI, USA
| | - Anne S De Groot
- Institute for Immunology and Informatics (iCubed), Department of Cell and Molecular Biology, University of Rhode Island, 80 Washington Street, Providence, RI, USA; EpiVax, Inc., 188 Valley Street Suite 424, Providence, RI, USA
| | - Marcelo B Sztein
- Center for Vaccine Development and Global Health (CVD), Department of Pediatrics, University of Maryland School of Medicine, 685 West Baltimore Street, HSF1, Baltimore, MD 21201, USA
| |
Collapse
|
13
|
O’Brien VP, Dorsey DA, Hannan TJ, Hultgren SJ. Host restriction of Escherichia coli recurrent urinary tract infection occurs in a bacterial strain-specific manner. PLoS Pathog 2018; 14:e1007457. [PMID: 30543708 PMCID: PMC6292575 DOI: 10.1371/journal.ppat.1007457] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 11/05/2018] [Indexed: 11/29/2022] Open
Abstract
Urinary tract infections (UTI) are extremely common and can be highly recurrent, with 1-2% of women suffering from six or more recurrent episodes per year. The high incidence of recurrent UTI, including recurrent infections caused by the same bacterial strain that caused the first infection, suggests that at least some women do not mount a protective adaptive immune response to UTI. Here we observed in a mouse model of cystitis (bladder infection) that infection with two different clinical uropathogenic Escherichia coli (UPEC) isolates, UTI89 or CFT073, resulted in different kinetics of bacterial clearance and different susceptibility to same-strain recurrent infection. UTI89 and CFT073 both caused infections that persisted for at least two weeks in similar proportions of mice, but whereas UTI89 infections could persist indefinitely, CFT073 infections began to clear two weeks after inoculation and were uniformly cleared within eight weeks. Mice with a history of CFT073 cystitis lasting four weeks were protected against recurrent CFT073 infection after antibiotic therapy, but were not protected against challenge with UTI89. In contrast, mice with a history of UTI89 cystitis lasting four weeks were highly susceptible to challenge infection with either strain after antibiotic treatment. We found that depletion of CD4+ and CD8+ T cell subsets impaired the ability of the host to clear CFT073 infections and rendered mice with a history of CFT073 cystitis lasting four weeks susceptible to recurrent CFT073 cystitis upon challenge. Our findings demonstrate the complex interplay between the broad genetic diversity of UPEC and the host innate and adaptive immune responses during UTI. A better understanding of these host-pathogen interactions is urgently needed for effective drug and vaccine development in the era of increasing antibiotic resistance.
Collapse
Affiliation(s)
- Valerie P. O’Brien
- Department of Molecular Microbiology and Center for Women’s Infectious Disease Research, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Denise A. Dorsey
- Department of Molecular Microbiology and Center for Women’s Infectious Disease Research, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Thomas J. Hannan
- Department of Molecular Microbiology and Center for Women’s Infectious Disease Research, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Scott J. Hultgren
- Department of Molecular Microbiology and Center for Women’s Infectious Disease Research, Washington University School of Medicine, St. Louis, Missouri, United States of America
| |
Collapse
|
14
|
Overexpressing ovotransferrin and avian β-defensin-3 improves antimicrobial capacity of chickens and poultry products. Transgenic Res 2018; 28:51-76. [PMID: 30374651 DOI: 10.1007/s11248-018-0101-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Accepted: 10/22/2018] [Indexed: 02/08/2023]
Abstract
Zoonotic and foodborne diseases pose a significant burden, decreasing both human and animal health. Modifying chickens to overexpress antimicrobials has the potential to decrease bacterial growth on poultry products and boost chicken innate immunity. Chickens overexpressing either ovotransferrin or avian β-defensin-3 (AvβD3) were generated using Tol-2 transposons. Transgene expression at the RNA and protein level was seen in egg white, breast muscle, and serum. There were significant differences in the immune cell populations in the blood, bursa, and spleen associated with transgene expression including an increased proportion of CD8+ cells in the blood of ovotransferrin and AvβD3 transgenic birds. Expression of the antimicrobials inhibited the in vitro growth of human and chicken bacterial pathogens and spoilage bacteria. For example, transgene expression significantly reduced growth of aerobic and coliform bacteria in breast muscle and decreased the growth of Salmonella enterica in egg white. Overall these results indicate that overexpression of antimicrobials in the chicken can impact the immune system and increase the antimicrobial capacity of poultry products.
Collapse
|
15
|
Xu W, Zhou T, Zhou J, Qiang Z, Zhang J, Hua Z. Attenuated Salmonella VNP20009 mutant (ΔhtrA) is a promising candidate for bacteria-mediated tumour therapy in hosts with TNFR1 deficiency. Lett Appl Microbiol 2018; 67:97-103. [PMID: 29698572 DOI: 10.1111/lam.12999] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 04/13/2018] [Accepted: 04/14/2018] [Indexed: 12/11/2022]
Abstract
VNP20009 is a genetically modified strain of Salmonella typhimurium and has a good anticancer effect wildly used in tumour therapy on animal models. For its clinical application, an accurate bio-safety assessment on sensitive models is necessary. In this study, we use TNFR1 KO mice as a susceptive model to assess the virulence of bacterial VNP20009 and its derivative ΔhtrA. By intraperitoneal administration of Salmonella, the increased lethality was observed in TNFR1 KO mice infected with VNP20009, but not with ΔhtrA. We performed a systemically comparative analysis of their toxicity, and ΔhtrA shows a better bio-safety for TNFR1 KO mice. Since the macrophages with TNFR1 deficiency exhibit a reduced ability of bacteria clearance, ΔhtrA with lower survival ability in normal macrophages restores its viability in TNFR1 KO macrophages. Thus, ΔhtrA was further tested for its antitumour effect in TNFR1 KO mice bearing a B16F10 melanoma model. It displays a moderate antitumour effect, suggesting ΔhtrA instead of VNP20009 might be a promising candidate for bacteria-mediated tumour therapy specific to those with low immunity. SIGNIFICANT AND IMPACT OF THE STUDY VNP20009 is attenuated Salmonella with a good safety widely used for tumour-targeting bacterial therapies. Little is known about its toxicity in hosts with low immunity. This study is the first systemically comparative analysis of their toxicity of VNP20009 and its mutant ΔhtrA in TNFR1-KO mice. Research on toxicity of tumour-targeting Salmonella in mice with immunodeficiency can facilitate the optimization of bacterial therapies with reduced adverse effects in future clinical trials.
Collapse
Affiliation(s)
- W Xu
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - T Zhou
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - J Zhou
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Z Qiang
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - J Zhang
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China.,Changzhou High-Tech Research Institute of Nanjing University and Jiangsu TargetPharma Laboratories Inc., Changzhou, China
| | - Z Hua
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China.,Changzhou High-Tech Research Institute of Nanjing University and Jiangsu TargetPharma Laboratories Inc., Changzhou, China.,Shenzhen Research Institute of Nanjing University, Shenzhen, China
| |
Collapse
|
16
|
Kang E, Crouse A, Chevallier L, Pontier SM, Alzahrani A, Silué N, Campbell-Valois FX, Montagutelli X, Gruenheid S, Malo D. Enterobacteria and host resistance to infection. Mamm Genome 2018; 29:558-576. [PMID: 29785663 DOI: 10.1007/s00335-018-9749-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Accepted: 05/14/2018] [Indexed: 02/06/2023]
Abstract
Enterobacteriaceae are a large family of Gram-negative, non-spore-forming bacteria. Although many species exist as part of the natural flora of animals including humans, some members are associated with both intestinal and extraintestinal diseases. In this review, we focus on members of this family that have important roles in human disease: Salmonella, Escherichia, Shigella, and Yersinia, providing a brief overview of the disease caused by these bacteria, highlighting the contribution of animal models to our understanding of their pathogenesis and of host genetic determinants involved in susceptibility or resistance to infection.
Collapse
Affiliation(s)
- Eugene Kang
- Department of Microbiology and Immunology, McGill University, Montreal, QC, Canada
- McGill Research Center on Complex Traits, McGill University, Montreal, QC, Canada
| | - Alanna Crouse
- McGill Research Center on Complex Traits, McGill University, Montreal, QC, Canada
- Department of Human Genetics, McGill University, Montreal, QC, Canada
| | - Lucie Chevallier
- U955 - IMRB, Team 10 - Biology of the neuromuscular system, Inserm, École Nationale Vétérinaire d'Alfort, UPEC, Maisons-Alfort, France
- Mouse Genetics Laboratory, Department of Genomes and Genetics, Institut Pasteur, Paris, France
| | - Stéphanie M Pontier
- Department of Chemistry and Biomolecular Sciences, Centre for Chemical and Synthetic Biology, University of Ottawa, Ottawa, ON, Canada
| | - Ashwag Alzahrani
- Department of Chemistry and Biomolecular Sciences, Centre for Chemical and Synthetic Biology, University of Ottawa, Ottawa, ON, Canada
| | - Navoun Silué
- Department of Chemistry and Biomolecular Sciences, Centre for Chemical and Synthetic Biology, University of Ottawa, Ottawa, ON, Canada
| | - François-Xavier Campbell-Valois
- Department of Chemistry and Biomolecular Sciences, Centre for Chemical and Synthetic Biology, University of Ottawa, Ottawa, ON, Canada
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada
| | - Xavier Montagutelli
- U955 - IMRB, Team 10 - Biology of the neuromuscular system, Inserm, École Nationale Vétérinaire d'Alfort, UPEC, Maisons-Alfort, France
| | - Samantha Gruenheid
- Department of Microbiology and Immunology, McGill University, Montreal, QC, Canada
- McGill Research Center on Complex Traits, McGill University, Montreal, QC, Canada
| | - Danielle Malo
- McGill Research Center on Complex Traits, McGill University, Montreal, QC, Canada.
- Department of Human Genetics, McGill University, Montreal, QC, Canada.
- Department of Medicine, McGill University, Montreal, QC, Canada.
| |
Collapse
|
17
|
Use of a novel antigen expressing system to study the Salmonella enterica serovar Typhi protein recognition by T cells. PLoS Negl Trop Dis 2017; 11:e0005912. [PMID: 28873442 PMCID: PMC5600385 DOI: 10.1371/journal.pntd.0005912] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 09/15/2017] [Accepted: 08/28/2017] [Indexed: 11/19/2022] Open
Abstract
Salmonella enterica serovar Typhi (S. Typhi), the causative agent of the typhoid fever, is a pathogen of great public health importance. Typhoid vaccines have the potential to be cost-effective measures towards combating this disease, yet the antigens triggering host protective immune responses are largely unknown. Given the key role of cellular-mediated immunity in S. Typhi protection, it is crucial to identify S. Typhi proteins involved in T-cell responses. Here, cells from individuals immunized with Ty21a typhoid vaccine were collected before and after immunization and used as effectors. We also used an innovative antigen expressing system based on the infection of B-cells with recombinant Escherichia coli (E. coli) expressing one of four S. Typhi gene products (i.e., SifA, OmpC, FliC, GroEL) as targets. Using flow cytometry, we found that the pattern of response to specific S. Typhi proteins was variable. Some individuals responded to all four proteins while others responded to only one or two proteins. We next evaluated whether T-cells responding to recombinant E. coli also possess the ability to respond to purified proteins. We observed that CD4+ cell responses, but not CD8+ cell responses, to recombinant E. coli were significantly associated with the responses to purified proteins. Thus, our results demonstrate the feasibility of using an E. coli expressing system to uncover the antigen specificity of T-cells and highlight its applicability to vaccine studies. These results also emphasize the importance of selecting the stimuli appropriately when evaluating CD4+ and CD8+ cell responses. Salmonella enterica serovar Typhi (S. Typhi) is the causative agent of the life-threatening typhoid fever that affects 11.9–20.6 million individuals annually in low-income and middle-income countries. The T-cells, CD4+ and CD8+ T cells, play a significant role in protection against S. Typhi infection. Yet, the antigens triggering host protective immune responses recognized by these cells are largely unknown. To address this shortcoming, in this study we used an E. coli expression system methodology for identifying immunogenic proteins of S. Typhi. We found that although the pattern of response to individual S. Typhi proteins was variable among the typhoid vaccinees, the E. coli expressing system uncovered the antigen specificity of T-cells, and highlight its applicability to vaccine studies.
Collapse
|
18
|
Mass spectrometry imaging identifies palmitoylcarnitine as an immunological mediator during Salmonella Typhimurium infection. Sci Rep 2017; 7:2786. [PMID: 28584281 PMCID: PMC5459799 DOI: 10.1038/s41598-017-03100-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Accepted: 04/25/2017] [Indexed: 12/19/2022] Open
Abstract
Salmonella Typhimurium causes a self-limiting gastroenteritis that may lead to systemic disease. Bacteria invade the small intestine, crossing the intestinal epithelium from where they are transported to the mesenteric lymph nodes (MLNs) within migrating immune cells. MLNs are an important site at which the innate and adaptive immune responses converge but their architecture and function is severely disrupted during S. Typhimurium infection. To further understand host-pathogen interactions at this site, we used mass spectrometry imaging (MSI) to analyse MLN tissue from a murine model of S. Typhimurium infection. A molecule, identified as palmitoylcarnitine (PalC), was of particular interest due to its high abundance at loci of S. Typhimurium infection and MLN disruption. High levels of PalC localised to sites within the MLNs where B and T cells were absent and where the perimeter of CD169+ sub capsular sinus macrophages was disrupted. MLN cells cultured ex vivo and treated with PalC had reduced CD4+CD25+ T cells and an increased number of B220+CD19+ B cells. The reduction in CD4+CD25+ T cells was likely due to apoptosis driven by increased caspase-3/7 activity. These data indicate that PalC significantly alters the host response in the MLNs, acting as a decisive factor in infection outcome.
Collapse
|
19
|
Rossi O, Grant AJ, Mastroeni P. Effect of in vivo neutralization of tumor necrosis alpha on the efficacy of antibiotic treatment in systemic Salmonella enterica infections. Pathog Dis 2017; 75:ftx002. [PMID: 28087648 PMCID: PMC5353993 DOI: 10.1093/femspd/ftx002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Revised: 10/13/2016] [Accepted: 01/12/2017] [Indexed: 02/06/2023] Open
Abstract
Immunity can co-operate with antibiotics, but can also antagonize drug efficacy by segregating the bacteria to areas of the body that are less accessible to antimicrobials, and by selecting for subpopulations with low division rates that are often difficult to eradicate. We studied the effect of an anti-inflammatory/immunosuppressive anti-TNFα treatment, which accelerates bacterial growth in the tissues and inhibits or reverses the formation of granulomas, on the efficacy of ampicillin and ciprofloxacin during a systemic Salmonella enterica infection of the mouse. The anti-TNFα treatment neither precluded nor enhanced the efficacy of antibiotic treatment. However, the anti-TNFα treatment rendered the animals susceptible to the rapid relapse of the infection seen after cessation of the antibiotic treatment. Reactivation of an established infection, due to late administration of anti-TNFα antibodies, could be successfully controlled by antibiotics, but full clearance of the bacterial load from the tissues was not achieved. We conclude that the lack of TNFα does not preclude the efficacy of antibiotic treatment and must be monitored with care due to post-treatment relapses. Combinations of anti-cytokine compounds and antibiotic molecules may not be the best way to treat persistent infections with intracellular bacteria like Salmonella.
Collapse
Affiliation(s)
- Omar Rossi
- Department of Veterinary Medicine, University of Cambridge, Cambridge CB3 0ES, UK
| | | | | |
Collapse
|
20
|
Karunakaran KP, Yu H, Jiang X, Chan Q, Goldberg MF, Jenkins MK, Foster LJ, Brunham RC. Identification of MHC-Bound Peptides from Dendritic Cells Infected with Salmonella enterica Strain SL1344: Implications for a Nontyphoidal Salmonella Vaccine. J Proteome Res 2016; 16:298-306. [PMID: 27802388 DOI: 10.1021/acs.jproteome.6b00926] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Worldwide Salmonella enterica infections result in substantial morbidity and mortality and are the major cause of infant bacteremia in Sub-Saharan Africa. Diseases caused by Salmonella are treatable with antibiotics, but successful antibiotic treatment has become difficult due to antimicrobial resistance and collateral effects on the microbiome. An effective vaccine together with public health efforts may be a better strategy to control these infections. Protective immunity against Salmonella depends primarily on CD4 T-cell-mediated immune responses; therefore, identifying relevant T-cell antigens is necessary for Salmonella vaccine development. We previously used a dendritic-cell-based immunoproteomics approach in our laboratory to identify T-cell antigens. The testing of these antigens as vaccine candidates against Chlamydia infection in mice yielded positive results. We applied this technology in the present study by infecting murine bone-marrow-derived dendritic cells from C57BL/6 mice with Salmonella enterica strain SL1344, followed by immunoaffinity isolation of MHC class I and II molecules and elution of bound peptides. The sequences of the peptides were identified using tandem mass spectrometry. We identified 87 MHC class-II- and 23 MHC class-I-binding Salmonella-derived peptides. Four of the 12 highest scoring class-II-binding Salmonella peptides stimulated IFN-γ production by CD4+ T cells from the spleens of mice with persistent Salmonella infection. We conclude that antigens identified by MHC immunoproteomics will be useful for Salmonella immunobiology studies and are potential Salmonella vaccine candidates. Data have been deposited to the ProteomeXchange Consortium via the PRIDE partner repository with the data set identifier PXD004451.
Collapse
Affiliation(s)
- Karuna P Karunakaran
- Vaccine Research Laboratory, University of British Columbia Centre for Disease Control , Vancouver, British Columbia V5Z 4R4, Canada
| | - Hong Yu
- Vaccine Research Laboratory, University of British Columbia Centre for Disease Control , Vancouver, British Columbia V5Z 4R4, Canada
| | - Xiaozhou Jiang
- Vaccine Research Laboratory, University of British Columbia Centre for Disease Control , Vancouver, British Columbia V5Z 4R4, Canada
| | - Queenie Chan
- Department of Biochemistry and Molecular Biology, Centre for High-Throughput Biology, University of British Columbia , Vancouver, British Columbia V6T 1Z3, Canada
| | - Michael F Goldberg
- Center for Immunology, Department of Microbiology and Immunology, University of Minnesota Medical School , Minneapolis, Minnesota 55455, United States
| | - Marc K Jenkins
- Center for Immunology, Department of Microbiology and Immunology, University of Minnesota Medical School , Minneapolis, Minnesota 55455, United States
| | - Leonard J Foster
- Department of Biochemistry and Molecular Biology, Centre for High-Throughput Biology, University of British Columbia , Vancouver, British Columbia V6T 1Z3, Canada
| | - Robert C Brunham
- Vaccine Research Laboratory, University of British Columbia Centre for Disease Control , Vancouver, British Columbia V5Z 4R4, Canada
| |
Collapse
|
21
|
Cross-reactive multifunctional CD4+ T cell responses against Salmonella enterica serovars Typhi, Paratyphi A and Paratyphi B in humans following immunization with live oral typhoid vaccine Ty21a. Clin Immunol 2016; 173:87-95. [PMID: 27634430 DOI: 10.1016/j.clim.2016.09.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Revised: 08/25/2016] [Accepted: 09/09/2016] [Indexed: 12/11/2022]
Abstract
The live oral typhoid vaccine Ty21a elicits predominantly CD8+, as well as CD4+ T cells mediated immune responses. Clinical field studies showed that Ty21a is moderately effective against S. Typhi and S. Paratyphi B, but not S. Paratyphi A infections. In this study we describe the in depth characterization of S. Typhi, S. Paratyphi A and S. Paratyphi B cross-reactive CD4+ T cell responses elicited following immunization with Ty21a. PBMC samples were collected from 16 healthy volunteers before and 42/84days after Ty21a immunization and stimulated ex-vivo with Salmonella-infected targets. Multiparametric flow cytometry was used to detect the vaccine elicited Salmonella-specific responses in T effector/memory (TEM) and CD45RA+ T effector/memory (TEMRA) CD4+ cell subsets, by measuring CD4+ multifunctional (MF) cells that concomitantly produced IFN-γ, TNF-α, IL-2, MIP-1β, IL-17A and/or expressed CD107a. Post-vaccination increases in S. Typhi-specific MF cells were observed in CD4+ TEM and TEMRA subsets which predominantly produced IFN-γ and/or TNF-α, while IL-2 was produced by a smaller cell subset. A small proportion of those MF cells also produced MIP-1β, IL-17A and expressed CD107a (a marker associated with cytotoxicity). Approximately one third of these specific MF cells have the potential to migrate to the gut mucosa, as evidenced by co-expression of the gut-homing molecule integrin α4β7. In contrast to our previous observations with CD8+ T cells, MF CD4+ T cell responses to the different Salmonella serovars evaluated were similar in magnitude and characteristics. We conclude that although induction of cross-reactive CD4+ MF effector T cells suggest a possible role in Salmonella-immunity, these responses are unlikely to provide an immunological basis for the observed efficacy of Ty21a against S. Typhi and S. Paratyphi B, but not to S. Paratyphi A.
Collapse
|
22
|
Cinobufagin enhances the protective efficacy of formalin-inactivated Salmonella typhimurium vaccine through Th1 immune response. Microb Pathog 2016; 99:264-270. [PMID: 27574776 DOI: 10.1016/j.micpath.2016.08.036] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Revised: 08/24/2016] [Accepted: 08/25/2016] [Indexed: 11/23/2022]
Abstract
Cinobufagin (CBG), one active ingredient isolated from Venenum Bufonis, has been demonstrated to have immunoregulatory effect. The aim of this study was to investigate whether CBG can enhance the protective efficacy of formalin-inactivated Salmonella typhimurium (FIST) in mice. ICR mice were immunized with FIST (106 CFU/mouse) alone or mixed with CBG (10, 20, and 40 μg) or alum (200 μg) on day 1 and day 15. Two weeks after the second immunization, serum and spleen were sampled for measuring FIST-specific antibody levels, cytokine levels, and splenocyte proliferation. The results showed that CBG enhanced FIST-specific IgG and IgG2a, the levels of interferon-gamma (IFNγ) and nitric oxide (NO), and the splenocyte proliferation response induced by concanavalin A, lipopolysaccharide, and FIST. In vivo protection studies showed that CBG significantly decreased the bacterial burdens in the spleen and prolonged the survival time of FIST-immunized mice challenged with live Salmonella typhimurium. In vivo IFNγ neutralization led to a significant reduction in FIST-specific IgG2a and IFNγ levels, and in the protective efficacy in CBG/FIST-immunized mice. In conclusion, CBG enhances the protective efficacy of formalin-inactivated Salmonella typhimurium vaccine by promoting the Th1 immune response.
Collapse
|
23
|
Thonhauser KE, Raveh S, Thoß M, Penn DJ. Does multiple paternity influence offspring disease resistance? J Evol Biol 2016; 29:1142-50. [PMID: 26949230 PMCID: PMC4949575 DOI: 10.1111/jeb.12854] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Accepted: 03/04/2016] [Indexed: 11/29/2022]
Abstract
It has been suggested that polyandry allows females to increase offspring genetic diversity and reduce the prevalence and susceptibility of their offspring to infectious diseases. We tested this hypothesis in wild‐derived house mice (Mus musculus) by experimentally infecting the offspring from 15 single‐ and 15 multiple‐sired litters with two different strains of a mouse pathogen (Salmonella Typhimurium) and compared their ability to control infection. We found a high variation in individual infection resistance (measured with pathogen loads) and significant differences among families, suggesting genetic effects on Salmonella resistance, but we found no difference in prevalence or infection resistance between single‐ vs. multiple‐sired litters. We found a significant sex difference in infection resistance, but surprisingly, males were more resistant to infection than females. Also, infection resistance was correlated with weight loss during infection, although only for females, indicating that susceptibility to infection had more harmful health consequences for females than for males. To our knowledge, our findings provide the first evidence for sex‐dependent resistance to Salmonella infection in house mice. Our results do not support the hypothesis that multiple‐sired litters are more likely to survive infection than single‐sired litters; however, as we explain, additional studies are required before ruling out this hypothesis.
Collapse
Affiliation(s)
- K E Thonhauser
- Konrad Lorenz Institute of Ethology, Department of Integrative Biology and Evolution, University of Veterinary Medicine, Vienna, Austria
| | - S Raveh
- Konrad Lorenz Institute of Ethology, Department of Integrative Biology and Evolution, University of Veterinary Medicine, Vienna, Austria.,Department of Environmental Sciences, Zoology and Evolution, University of Basel, Basel, Switzerland
| | - M Thoß
- Konrad Lorenz Institute of Ethology, Department of Integrative Biology and Evolution, University of Veterinary Medicine, Vienna, Austria
| | - D J Penn
- Konrad Lorenz Institute of Ethology, Department of Integrative Biology and Evolution, University of Veterinary Medicine, Vienna, Austria
| |
Collapse
|
24
|
Wahid R, Fresnay S, Levine MM, Sztein MB. Immunization with Ty21a live oral typhoid vaccine elicits crossreactive multifunctional CD8+ T-cell responses against Salmonella enterica serovar Typhi, S. Paratyphi A, and S. Paratyphi B in humans. Mucosal Immunol 2015; 8:1349-59. [PMID: 25872480 PMCID: PMC4607552 DOI: 10.1038/mi.2015.24] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Accepted: 02/06/2015] [Indexed: 02/04/2023]
Abstract
Previously we have extensively characterized Salmonella enterica serovar Typhi (S. Typhi)-specific cell-mediated immune (CMI) responses in volunteers orally immunized with the licensed Ty21a typhoid vaccine. In this study we measured Salmonella-specific multifunctional (MF) CD8+ T-cell responses to further investigate whether Ty21a elicits crossreactive CMI against S. Paratyphi A and S. Paratyphi B that also cause enteric fever. Ty21a-elicited crossreactive CMI responses against all three Salmonella serotypes were predominantly observed in CD8+ T effector/memory (T(EM)) and, to a lesser extent, in CD8+CD45RA+ T(EM) (T(EMRA)) subsets. These CD8+ T-cell responses were largely mediated by MF cells coproducing interferon-γ and macrophage inflammatory protein-1β and expressing CD107a with or without tumor necrosis factor-α. Significant proportions of Salmonella-specific MF cells expressed the gut-homing molecule integrin α4β7. In most subjects, similar MF responses were observed to S. Typhi and S. Paratyphi B, but not to S. Paratyphi A. These results suggest that Ty21a elicits MF CMI responses against Salmonella that could be critical in clearing the infection. Moreover, because S. Paratyphi A is a major public concern and Ty21a was shown in field studies not to afford cross-protection to S. Paratyphi A, these results will be important in developing a S. Typhi/S. Paratyphi A bivalent vaccine against enteric fevers.
Collapse
Affiliation(s)
- Rezwanul Wahid
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Stephanie Fresnay
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Myron M. Levine
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Department of Medicine, Center for Vaccine Development, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Marcelo B. Sztein
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Department of Medicine, Center for Vaccine Development, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
25
|
Kinnear CL, Strugnell RA. Vaccination Method Affects Immune Response and Bacterial Growth but Not Protection in the Salmonella Typhimurium Animal Model of Typhoid. PLoS One 2015; 10:e0141356. [PMID: 26509599 PMCID: PMC4625024 DOI: 10.1371/journal.pone.0141356] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2015] [Accepted: 10/07/2015] [Indexed: 01/22/2023] Open
Abstract
Understanding immune responses elicited by vaccines, together with immune responses required for protection, is fundamental to designing effective vaccines and immunisation programs. This study examines the effects of the route of administration of a live attenuated vaccine on its interactions with, and stimulation of, the murine immune system as well as its ability to increase survival and provide protection from colonisation by a virulent challenge strain. We assess the effect of administration method using the murine model for typhoid, where animals are infected with S. Typhimurium. Mice were vaccinated either intravenously or orally with the same live attenuated S. Typhimurium strain and data were collected on vaccine strain growth, shedding and stimulation of antibodies and cytokines. Following vaccination, mice were challenged with a virulent strain of S. Typhimurium and the protection conferred by the different vaccination routes was measured in terms of challenge suppression and animal survival. The main difference in immune stimulation found in this study was the development of a secretory IgA response in orally-vaccinated mice, which was absent in IV vaccinated mice. While both strains showed similar protection in terms of challenge suppression in systemic organs (spleen and liver) as well as survival, they differed in terms of challenge suppression of virulent pathogens in gut-associated organs. This difference in gut colonisation presents important questions around the ability of vaccines to prevent shedding and transmission. These findings demonstrate that while protection conferred by two vaccines can appear to be the same, the mechanisms controlling the protection can differ and have important implications for infection dynamics within a population.
Collapse
Affiliation(s)
- Clare L. Kinnear
- Department of Microbiology and Immunology, The University of Melbourne, Melbourne, Victoria, Australia
- Department of Biosciences, The University of Melbourne, Melbourne, Victoria, Australia
- * E-mail:
| | - Richard A. Strugnell
- Department of Microbiology and Immunology, The University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
26
|
Abstract
The interaction betweenSalmonella and its host is complex and dynamic: the host mounts an immune defense against the pathogen, which in turn acts to reduce, evade, or exploit these responses to successfully colonize the host. Although the exact mechanisms mediating protective immunity are poorly understood, it is known that T cells are a critical component of immunity to Salmonella infection, and a robust T-cell response is required for both clearance of primary infection and resistance to subsequent challenge. B-cell functions, including but not limited to antibody production, are also required for generation of protective immunity. Additionally, interactions among host cells are essential. For example, antigen-presenting cells (including B cells) express cytokines that participate in CD4+ T cell activation and differentiation. Differentiated CD4+ T cells secrete cytokines that have both autocrine and paracrine functions, including recruitment and activation of phagocytes, and stimulation of B cell isotype class switching and affinity maturation. Multiple bacterium-directed mechanisms, including altered antigen expression and bioavailability and interference with antigen-presenting cell activation and function, combine to modify Salmonella's "pathogenic signature" in order to minimize its susceptibility to host immune surveillance. Therefore, a more complete understanding of adaptive immune responses may provide insights into pathogenic bacterial functions. Continued identification of adaptive immune targets will guide rational vaccine development, provide insights into host functions required to resist Salmonella infection, and correspondingly provide valuable reagents for defining the critical pathogenic capabilities of Salmonella that contribute to their success in causing acute and chronic infections.
Collapse
|
27
|
Mooney JP, Lee SJ, Lokken KL, Nanton MR, Nuccio SP, McSorley SJ, Tsolis RM. Transient Loss of Protection Afforded by a Live Attenuated Non-typhoidal Salmonella Vaccine in Mice Co-infected with Malaria. PLoS Negl Trop Dis 2015; 9:e0004027. [PMID: 26366739 PMCID: PMC4569369 DOI: 10.1371/journal.pntd.0004027] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Accepted: 08/03/2015] [Indexed: 11/19/2022] Open
Abstract
In immunocompetent individuals, non-typhoidal Salmonella serovars (NTS) are associated with gastroenteritis, however, there is currently an epidemic of NTS bloodstream infections in sub-Saharan Africa. Plasmodium falciparum malaria is an important risk factor for invasive NTS bloodstream in African children. Here we investigated whether a live, attenuated Salmonella vaccine could be protective in mice, in the setting of concurrent malaria. Surprisingly, mice acutely infected with the nonlethal malaria parasite Plasmodium yoelii 17XNL exhibited a profound loss of protective immunity to NTS, but vaccine-mediated protection was restored after resolution of malaria. Absence of protective immunity during acute malaria correlated with maintenance of antibodies to NTS, but a marked reduction in effector capability of Salmonella-specific CD4 and CD8 T cells. Further, increased expression of the inhibitory molecule PD1 was identified on memory CD4 T cells induced by vaccination. Blockade of IL-10 restored protection against S. Typhimurium, without restoring CD4 T cell effector function. Simultaneous blockade of CTLA-4, LAG3, and PDL1 restored IFN-γ production by vaccine-induced memory CD4 T cells but was not sufficient to restore protection. Together, these data demonstrate that malaria parasite infection induces a temporary loss of an established adaptive immune response via multiple mechanisms, and suggest that in the setting of acute malaria, protection against NTS mediated by live vaccines may be interrupted. In children, malaria is a predisposing factor for invasive bacterial infections with non-typhoidal Salmonella (NTS) serovars, a frequent cause of morbidity and mortality in sub-Saharan Africa. Since development of vaccines against NTS has been proposed as a strategy to protect African children against disseminated NTS infection, we interrogated the effect of malaria on vaccine-induced memory responses to NTS. Our results from a mouse infection model show that infection with malaria parasites temporarily suspends protective immunity conferred by a live, attenuated vaccine and suppresses adaptive immune responses to NTS that are mediated by T cells. These results suggest that in the setting of acute malaria, live attenuated NTS vaccines may lose their effectiveness.
Collapse
Affiliation(s)
- Jason P. Mooney
- Department of Microbiology & Immunology, School of Medicine, University of California Davis, Davis, California, United States of America
| | - Seung-Joo Lee
- Center for Comparative Medicine, Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California Davis, Davis, California, United States of America
| | - Kristen L. Lokken
- Department of Microbiology & Immunology, School of Medicine, University of California Davis, Davis, California, United States of America
| | - Minelva R. Nanton
- Center for Comparative Medicine, Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California Davis, Davis, California, United States of America
| | - Sean-Paul Nuccio
- Department of Microbiology & Immunology, School of Medicine, University of California Davis, Davis, California, United States of America
| | - Stephen J. McSorley
- Center for Comparative Medicine, Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California Davis, Davis, California, United States of America
| | - Renée M. Tsolis
- Department of Microbiology & Immunology, School of Medicine, University of California Davis, Davis, California, United States of America
- * E-mail:
| |
Collapse
|
28
|
Gilchrist JJ, MacLennan CA, Hill AVS. Genetic susceptibility to invasive Salmonella disease. Nat Rev Immunol 2015; 15:452-63. [PMID: 26109132 DOI: 10.1038/nri3858] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Invasive Salmonella disease, in the form of enteric fever and invasive non-typhoidal Salmonella (iNTS) disease, causes substantial morbidity and mortality in children and adults in the developing world. The study of genetic variations in humans and mice that influence susceptibility of the host to Salmonella infection provides important insights into immunity to Salmonella. In this Review, we discuss data that have helped to elucidate the host genetic determinants of human enteric fever and iNTS disease, alongside data from the mouse model of Salmonella infection. Considered together, these studies provide a detailed picture of the immunobiology of human invasive Salmonella disease.
Collapse
Affiliation(s)
- James J Gilchrist
- Wellcome Trust Centre for Human Genetics, Roosevelt Drive, University of Oxford, Oxford OX3 7BN, UK
| | - Calman A MacLennan
- 1] Jenner Institute, Nuffield Department of Medicine, Old Road Campus Research Building, Roosevelt Drive, University of Oxford, Oxford, OX3 7DQ, UK. [2] Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SA, UK
| | - Adrian V S Hill
- 1] Wellcome Trust Centre for Human Genetics, Roosevelt Drive, University of Oxford, Oxford OX3 7BN, UK. [2] Jenner Institute, Nuffield Department of Medicine, Old Road Campus Research Building, Roosevelt Drive, University of Oxford, Oxford, OX3 7DQ, UK
| |
Collapse
|
29
|
Abstract
Salmonella are a common source of food- or water-borne infection and cause a wide range of clinical disease in human and animal hosts. Salmonella are relatively easy to culture and manipulate in a laboratory setting, and the infection of laboratory animals induces robust innate and adaptive immune responses. Thus, immunologists have frequently turned to Salmonella infection models to expand understanding of host immunity to intestinal pathogens. In this review, I summarize current knowledge of innate and adaptive immunity to Salmonella and highlight features of this response that have emerged from recent studies. These include the heterogeneity of the antigen-specific T-cell response to intestinal infection, the prominence of microbial mechanisms to impede T- and B-cell responses, and the contribution of non-cognate pathways for elicitation of T-cell effector functions. Together, these different issues challenge an overly simplistic view of host-pathogen interaction during mucosal infection, but also allow deeper insight into the real-world dynamic of protective immunity to intestinal pathogens.
Collapse
Affiliation(s)
- Stephen J McSorley
- Center for Comparative Medicine, Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California Davis, Davis, CA, USA
| |
Collapse
|
30
|
Guadarrama C, Villaseñor T, Calva E. The Subtleties and Contrasts of the LeuO Regulator in Salmonella Typhi: Implications in the Immune Response. Front Immunol 2014; 5:581. [PMID: 25566242 PMCID: PMC4264507 DOI: 10.3389/fimmu.2014.00581] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Accepted: 10/30/2014] [Indexed: 01/15/2023] Open
Abstract
Salmonella are facultative intracellular pathogens. Salmonella infection occurs mainly by expression of two Salmonella pathogenicity Islands (SPI-1 and SPI-2). SPI-1 encodes transcriptional factors that participate in the expression of virulence factors encoded in the island. However, there are transcriptional factors encoded outside the island that also participate in the expression of SPI-1-encoded genes. Upon infection, bacteria are capable of avoiding the host immune response with several strategies that involve several virulence factors under the control of transcriptional regulators. Interestingly, LeuO a transcriptional global regulator which is encoded outside of any SPI, is proposed to be part of a complex regulatory network that involves expression of several genes that help bacteria to survive stress conditions and, also, induces the expression of porins that have been shown to be immunogens and can thus be considered as antigenic candidates for acellular vaccines. Hence, the understanding of the LeuO regulon implies a role of bacterial genetic regulation in determining the host immune response.
Collapse
Affiliation(s)
- Carmen Guadarrama
- Departamento de Microbiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México , Cuernavaca , Mexico
| | - Tomás Villaseñor
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México , Cuernavaca , Mexico
| | - Edmundo Calva
- Departamento de Microbiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México , Cuernavaca , Mexico
| |
Collapse
|
31
|
Reynolds CJ, Jones C, Blohmke CJ, Darton TC, Goudet A, Sergeant R, Maillere B, Pollard AJ, Altmann DM, Boyton RJ. The serodominant secreted effector protein of Salmonella, SseB, is a strong CD4 antigen containing an immunodominant epitope presented by diverse HLA class II alleles. Immunology 2014; 143:438-46. [PMID: 24891088 PMCID: PMC4212957 DOI: 10.1111/imm.12327] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Revised: 05/03/2014] [Accepted: 05/19/2014] [Indexed: 12/25/2022] Open
Abstract
Detailed characterization of the protective T-cell response in salmonellosis is a pressing unmet need in light of the global burden of human Salmonella infections and the likely contribution of CD4 T cells to immunity against this intracellular infection. In previous studies screening patient sera against antigen arrays, SseB was noteworthy as a serodominant target of adaptive immunity, inducing significantly raised antibody responses in HIV-seronegative compared with seropositive patients. SseB is a secreted protein, part of the Espa superfamily, localized to the bacterial surface and forming part of the translocon of the type III secretion system (T3SS) encoded by Salmonella pathogenicity island 2. We demonstrate here that SseB is also a target of CD4 T-cell immunity, generating a substantial response after experimental infection in human volunteers, with around 0·1% of the peripheral repertoire responding to it. HLA-DR/peptide binding studies indicate that this protein encompasses a number of peptides with ability to bind to several different HLA-DR alleles. Of these, peptide 11 (p11) was shown in priming of both HLA-DR1 and HLA-DR4 transgenic mice to contain an immunodominant CD4 epitope. Analysis of responses in human donors showed immunity focused on p11 and another epitope in peptide 2. The high frequency of SseB-reactive CD4 T cells and the broad applicability to diverse HLA genotypes coupled with previous observations of serodominance and protective vaccination in mouse challenge experiments, make SseB a plausible candidate for next-generation Salmonella vaccines.
Collapse
Affiliation(s)
- Catherine J Reynolds
- Section of Infectious Diseases and Immunity, Department of Medicine, Imperial College, Hammersmith Hospital, London, UK
| | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Natural and vaccine-mediated immunity to Salmonella Typhimurium is impaired by the helminth Nippostrongylus brasiliensis. PLoS Negl Trop Dis 2014; 8:e3341. [PMID: 25474738 PMCID: PMC4256288 DOI: 10.1371/journal.pntd.0003341] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2014] [Accepted: 10/14/2014] [Indexed: 02/06/2023] Open
Abstract
Background The impact of exposure to multiple pathogens concurrently or consecutively on immune function is unclear. Here, immune responses induced by combinations of the bacterium Salmonella Typhimurium (STm) and the helminth Nippostrongylus brasiliensis (Nb), which causes a murine hookworm infection and an experimental porin protein vaccine against STm, were examined. Methodology/Principal Findings Mice infected with both STm and Nb induced similar numbers of Th1 and Th2 lymphocytes compared with singly infected mice, as determined by flow cytometry, although lower levels of secreted Th2, but not Th1 cytokines were detected by ELISA after re-stimulation of splenocytes. Furthermore, the density of FoxP3+ T cells in the T zone of co-infected mice was lower compared to mice that only received Nb, but was greater than those that received STm. This reflected the intermediate levels of IL-10 detected from splenocytes. Co-infection compromised clearance of both pathogens, with worms still detectable in mice weeks after they were cleared in the control group. Despite altered control of bacterial and helminth colonization in co-infected mice, robust extrafollicular Th1 and Th2-reflecting immunoglobulin-switching profiles were detected, with IgG2a, IgG1 and IgE plasma cells all detected in parallel. Whilst extrafollicular antibody responses were maintained in the first weeks after co-infection, the GC response was less than that in mice infected with Nb only. Nb infection resulted in some abrogation of the longer-term development of anti-STm IgG responses. This suggested that prior Nb infection may modulate the induction of protective antibody responses to vaccination. To assess this we immunized mice with porins, which confer protection in an antibody-dependent manner, before challenging with STm. Mice that had resolved a Nb infection prior to immunization induced less anti-porin IgG and had compromised protection against infection. Conclusion These findings demonstrate that co-infection can radically alter the development of protective immunity during natural infection and in response to immunization. Vaccination studies in animal models have focused on understanding responses in young, previously naïve mice. In reality, humans are vaccinated or respond to infection in the context of a life-time of accumulated exposure to multiple, systemic infections and other vaccines, some of which are themselves attenuated live organisms. This is even more pronounced in areas that are endemic for infectious diseases. We wished to examine the impact infectious history can have on the immune response against infection and the efficacy of vaccination. To do this, we used two classes of pathogens that model clinically important invasive infections. One pathogen is the bacterium, Salmonella Typhimurium against which we have also developed an experimental porin vaccine, and the second is an invasive helminth, Nippostrongylus brasiliensis, that models aspects of hookworm infections. Our studies indicate that exposure to a second, unrelated pathogen can reduce the efficiency of immunity generated during natural infection and immunity generated after vaccination. These results are important as they help to identify potential strategies for improving immune-mediated control of infection and the success of vaccination in infection-endemic regions.
Collapse
|
33
|
Nanton MR, Lee SJ, Atif SM, Nuccio SP, Taylor JJ, Bäumler AJ, Way SS, McSorley SJ. Direct visualization of endogenous Salmonella-specific B cells reveals a marked delay in clonal expansion and germinal center development. Eur J Immunol 2014; 45:428-41. [PMID: 25346524 DOI: 10.1002/eji.201444540] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2014] [Revised: 09/30/2014] [Accepted: 10/21/2014] [Indexed: 11/11/2022]
Abstract
CD4(+) T cells and B cells are both essential for acquired immunity to Salmonella infection. It is well established that Salmonella inhibit host CD4(+) T-cell responses, but a corresponding inhibitory effect on B cells is less well defined. Here, we utilize an Ag tetramer and pull-down enrichment strategy to directly visualize OVA-specific B cells in mice, as they respond to infection with Salmonella-OVA. Surprisingly, OVA-specific B-cell expansion and germinal center formation was not detected until bacteria were cleared from the host. Furthermore, Salmonella infection also actively inhibited both B- and T-cell responses to the same coinjected Ag but this did not require the presence of iNOS. The Salmonella Pathogenicity Island 2 (SPI2) locus has been shown to be responsible for inhibition of Salmonella-specific CD4(+) T-cell responses, and an examination of SPI2-deficient bacteria demonstrated a recovery in B-cell expansion in infected mice. Together, these data suggest that Salmonella can simultaneously inhibit host B- and T-cell responses using SPI2-dependent mechanisms.
Collapse
Affiliation(s)
- Minelva R Nanton
- Center for Comparative Medicine, Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California Davis, Davis, CA, USA; Microbiology, Immunology, and Cancer Biology Graduate Program, University of Minnesota Medical School-Twin Cities, Minneapolis, MN, USA
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Bobat S, Cunningham AF. Bacterial infections and vaccines. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 828:75-98. [PMID: 25253028 DOI: 10.1007/978-1-4939-1489-0_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2023]
Affiliation(s)
- Saeeda Bobat
- The Institute of Microbiology and Infection, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham, West Midlands, UK,
| | | |
Collapse
|
35
|
Coward C, Restif O, Dybowski R, Grant AJ, Maskell DJ, Mastroeni P. The effects of vaccination and immunity on bacterial infection dynamics in vivo. PLoS Pathog 2014; 10:e1004359. [PMID: 25233077 PMCID: PMC4169467 DOI: 10.1371/journal.ppat.1004359] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Accepted: 07/25/2014] [Indexed: 01/31/2023] Open
Abstract
Salmonella enterica infections are a significant global health issue, and development of vaccines against these bacteria requires an improved understanding of how vaccination affects the growth and spread of the bacteria within the host. We have combined in vivo tracking of molecularly tagged bacterial subpopulations with mathematical modelling to gain a novel insight into how different classes of vaccines and branches of the immune response protect against secondary Salmonella enterica infections of the mouse. We have found that a live Salmonella vaccine significantly reduced bacteraemia during a secondary challenge and restrained inter-organ spread of the bacteria in the systemic organs. Further, fitting mechanistic models to the data indicated that live vaccine immunisation enhanced both the bacterial killing in the very early stages of the infection and bacteriostatic control over the first day post-challenge. T-cell immunity induced by this vaccine is not necessary for the enhanced bacteriostasis but is required for subsequent bactericidal clearance of Salmonella in the blood and tissues. Conversely, a non-living vaccine while able to enhance initial blood clearance and killing of virulent secondary challenge bacteria, was unable to alter the subsequent bacterial growth rate in the systemic organs, did not prevent the resurgence of extensive bacteraemia and failed to control the spread of the bacteria in the body. The bacterium Salmonella enterica causes gastroenteritis and the severe systemic diseases typhoid, paratyphoid fever and non-typhoidal septicaemia (NTS). Treatment of systemic disease with antibiotics is becoming increasingly difficult due to the acquisition of resistance. Licensed vaccines are available for the prevention of typhoid, but not paratyphoid fever or NTS. Vaccines can be either living (attenuated strains) or non-living (e.g. inactivated whole cells or surface polysaccharides) and these different classes potentially activate different components of the host immune system. Improvements in vaccine design require a better understanding of how different vaccine types differ in their ability to control a subsequent infection. We have improved a previously developed experimental system and mathematical model to investigate how these different vaccine types act. We show that the inactivated vaccine can only control bacterial numbers by a transient increase in bactericidal activity whereas the living vaccine is superior as it can induce an immune response that rapidly kills, then restrains the growth and spread of infecting bacteria.
Collapse
Affiliation(s)
- Chris Coward
- University of Cambridge, Department of Veterinary Medicine, Cambridge, United Kingdom
| | - Olivier Restif
- University of Cambridge, Department of Veterinary Medicine, Cambridge, United Kingdom
| | - Richard Dybowski
- University of Cambridge, Department of Veterinary Medicine, Cambridge, United Kingdom
| | - Andrew J Grant
- University of Cambridge, Department of Veterinary Medicine, Cambridge, United Kingdom
| | - Duncan J Maskell
- University of Cambridge, Department of Veterinary Medicine, Cambridge, United Kingdom
| | - Pietro Mastroeni
- University of Cambridge, Department of Veterinary Medicine, Cambridge, United Kingdom
| |
Collapse
|
36
|
Cellular requirements for systemic control of Salmonella enterica serovar Typhimurium infections in mice. Infect Immun 2014; 82:4997-5004. [PMID: 25225248 DOI: 10.1128/iai.02192-14] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The rational design of vaccines requires an understanding of the contributions of individual immune cell subsets to immunity. With this understanding, targeted vaccine delivery approaches and adjuvants can be developed to maximize vaccine efficiency and to minimize side effects (S. H. E. Kaufmann et al., Immunity 33:555-577, 2010; T. Ben-Yedidia and R. Arnon, Hum. Vaccines 1:95-101, 2005). We have addressed the contributions of different immune cell subsets and their ability to contribute to the control and clearance of the facultative intracellular pathogen Salmonella enterica serovar Typhimurium (S. Typhimurium) in a murine model. Using a systematic and reproducible model of experimental attenuated S. Typhimurium infection, we show that distinct lymphocyte deficiencies lead to one of four different infection outcomes: clearance, chronic infection, early death, or late death. Our study demonstrates a high level of functional redundancy in the ability of different lymphocyte subsets to provide interferon gamma (IFN-γ), a critical cytokine in Salmonella immunity. Whereas early control of the infection was entirely dependent on IFN-γ but not on any particular lymphocyte subset, clearance of the infection critically required CD4(+) T cells but appeared to be independent of IFN-γ. These data reinforce the idea of a bimodal immune response against Salmonella: an early T cell-independent but IFN-γ-dependent phase and a late T cell-dependent phase that may be IFN-γ independent.
Collapse
|
37
|
Haque SS. Immunological Evaluation of Formulated Drugs against Typhoid. Open Access Maced J Med Sci 2014. [DOI: 10.3889/oamjms.2014.069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
OBJECTIVES: Typhoid fever an important causes of illness and death, particularly among children and adolescents in south-central and Southeast Asia, where enteric fever is associated with poor sanitation and unsafe food and water. Cell-mediated immunity (CMI) plays an important role for the survival of the host in experimental salmonellosis. Nitric oxide (NO) is the one of the product of macrophages activated by cytokines, microbial compounds or both, is derived from the amino acid L-arginine by the enzymatic activity of inducible nitric oxide synthase (iNOS or NOS2) which acts as antimicrobial molecule.AIM: The aim was to examine the induction of DTH reaction in the animals treated with L-Arginine, ciprofloxacin and their combination followed by immunization with S. typhimurium cell lysate  as assessed by the footpad swelling test.RESULTS: The results of the present study showed that the induction of DTH reaction in the animals treated with L-Arginine, ciprofloxacin and their combination followed by immunization with S. typhimurium cell lysate using an antigen revealed that the treatment with combination increased foot pad swelling significantly as compared to saline treated control animals at 48 hour which was followed by a decrease of the swelling at 72 hour.CONCLUSIONS: Animal treated with L-arginine, ciprofloxacin and their combination showed increased cell mediated immune responses as evident by DTH response whereas groups (B+S) shows decreases CMI responses.
Collapse
|
38
|
Ross EA, Flores-Langarica A, Bobat S, Coughlan RE, Marshall JL, Hitchcock JR, Cook CN, Carvalho-Gaspar MM, Mitchell AM, Clarke M, Garcia P, Cobbold M, Mitchell TJ, Henderson IR, Jones ND, Anderson G, Buckley CD, Cunningham AF. Resolving Salmonella infection reveals dynamic and persisting changes in murine bone marrow progenitor cell phenotype and function. Eur J Immunol 2014; 44:2318-30. [PMID: 24825601 PMCID: PMC4209805 DOI: 10.1002/eji.201344350] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2013] [Revised: 04/07/2014] [Accepted: 05/08/2014] [Indexed: 11/15/2022]
Abstract
The generation of immune cells from BM precursors is a carefully regulated process. This is essential to limit the potential for oncogenesis and autoimmunity yet protect against infection. How infection modulates this is unclear. Salmonella can colonize systemic sites including the BM and spleen. This resolving infection has multiple IFN-γ-mediated acute and chronic effects on BM progenitors, and during the first week of infection IFN-γ is produced by myeloid, NK, NKT, CD4(+) T cells, and some lineage-negative cells. After infection, the phenotype of BM progenitors rapidly but reversibly alters, with a peak ∼ 30-fold increase in Sca-1(hi) progenitors and a corresponding loss of Sca-1(lo/int) subsets. Most strikingly, the capacity of donor Sca-1(hi) cells to reconstitute an irradiated host is reduced; the longer donor mice are exposed to infection, and Sca-1(hi) c-kit(int) cells have an increased potential to generate B1a-like cells. Thus, Salmonella can have a prolonged influence on BM progenitor functionality not directly related to bacterial persistence. These results reflect changes observed in leucopoiesis during aging and suggest that BM functionality can be modulated by life-long, periodic exposure to infection. Better understanding of this process could offer novel therapeutic opportunities to modulate BM functionality and promote healthy aging.
Collapse
Affiliation(s)
- Ewan A Ross
- MRC Centre for Immune Regulation, Institute for Microbiology and Infection, School of Immunity and Infection, Institute for Biomedical Research, Medical School, University of BirminghamEdgbaston, Birmingham, UK
| | - Adriana Flores-Langarica
- MRC Centre for Immune Regulation, Institute for Microbiology and Infection, School of Immunity and Infection, Institute for Biomedical Research, Medical School, University of BirminghamEdgbaston, Birmingham, UK
| | - Saeeda Bobat
- MRC Centre for Immune Regulation, Institute for Microbiology and Infection, School of Immunity and Infection, Institute for Biomedical Research, Medical School, University of BirminghamEdgbaston, Birmingham, UK
| | - Ruth E Coughlan
- MRC Centre for Immune Regulation, Institute for Microbiology and Infection, School of Immunity and Infection, Institute for Biomedical Research, Medical School, University of BirminghamEdgbaston, Birmingham, UK
| | - Jennifer L Marshall
- MRC Centre for Immune Regulation, Institute for Microbiology and Infection, School of Immunity and Infection, Institute for Biomedical Research, Medical School, University of BirminghamEdgbaston, Birmingham, UK
| | - Jessica R Hitchcock
- MRC Centre for Immune Regulation, Institute for Microbiology and Infection, School of Immunity and Infection, Institute for Biomedical Research, Medical School, University of BirminghamEdgbaston, Birmingham, UK
| | - Charlotte N Cook
- MRC Centre for Immune Regulation, Institute for Microbiology and Infection, School of Immunity and Infection, Institute for Biomedical Research, Medical School, University of BirminghamEdgbaston, Birmingham, UK
| | - Manuela M Carvalho-Gaspar
- MRC Centre for Immune Regulation, Institute for Microbiology and Infection, School of Immunity and Infection, Institute for Biomedical Research, Medical School, University of BirminghamEdgbaston, Birmingham, UK
| | - Andrea M Mitchell
- MRC Centre for Immune Regulation, Institute for Microbiology and Infection, School of Immunity and Infection, Institute for Biomedical Research, Medical School, University of BirminghamEdgbaston, Birmingham, UK
| | - Mary Clarke
- MRC Centre for Immune Regulation, Institute for Microbiology and Infection, School of Immunity and Infection, Institute for Biomedical Research, Medical School, University of BirminghamEdgbaston, Birmingham, UK
| | - Paloma Garcia
- MRC Centre for Immune Regulation, Institute for Microbiology and Infection, School of Immunity and Infection, Institute for Biomedical Research, Medical School, University of BirminghamEdgbaston, Birmingham, UK
| | - Mark Cobbold
- MRC Centre for Immune Regulation, Institute for Microbiology and Infection, School of Immunity and Infection, Institute for Biomedical Research, Medical School, University of BirminghamEdgbaston, Birmingham, UK
| | - Tim J Mitchell
- MRC Centre for Immune Regulation, Institute for Microbiology and Infection, School of Immunity and Infection, Institute for Biomedical Research, Medical School, University of BirminghamEdgbaston, Birmingham, UK
| | - Ian R Henderson
- MRC Centre for Immune Regulation, Institute for Microbiology and Infection, School of Immunity and Infection, Institute for Biomedical Research, Medical School, University of BirminghamEdgbaston, Birmingham, UK
| | - Nick D Jones
- MRC Centre for Immune Regulation, Institute for Microbiology and Infection, School of Immunity and Infection, Institute for Biomedical Research, Medical School, University of BirminghamEdgbaston, Birmingham, UK
| | - Graham Anderson
- MRC Centre for Immune Regulation, Institute for Microbiology and Infection, School of Immunity and Infection, Institute for Biomedical Research, Medical School, University of BirminghamEdgbaston, Birmingham, UK
| | - Christopher D Buckley
- MRC Centre for Immune Regulation, Institute for Microbiology and Infection, School of Immunity and Infection, Institute for Biomedical Research, Medical School, University of BirminghamEdgbaston, Birmingham, UK
| | - Adam F Cunningham
- MRC Centre for Immune Regulation, Institute for Microbiology and Infection, School of Immunity and Infection, Institute for Biomedical Research, Medical School, University of BirminghamEdgbaston, Birmingham, UK
| |
Collapse
|
39
|
Patel S, McCormick BA. Mucosal Inflammatory Response to Salmonella typhimurium Infection. Front Immunol 2014; 5:311. [PMID: 25071772 PMCID: PMC4082011 DOI: 10.3389/fimmu.2014.00311] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2014] [Accepted: 06/20/2014] [Indexed: 12/24/2022] Open
Abstract
The human intestinal epithelium consists of a single layer of epithelial cells that forms a barrier against food antigens and the resident microbiota within the lumen. This delicately balanced organ functions in a highly sophisticated manner to uphold the fidelity of the intestinal epithelium and to eliminate pathogenic microorganisms. On the luminal side, this barrier is fortified by a thick mucus layer, and on the serosal side exists the lamina propria containing a resident population of immune cells. Pathogens that are able to breach this barrier disrupt the healthy epithelial lining by interfering with the regulatory mechanisms that govern the normal balance of intestinal architecture and function. This disruption results in a coordinated innate immune response deployed to eliminate the intruder that includes the release of antimicrobial peptides, activation of pattern-recognition receptors, and recruitment of a variety of immune cells. In the case of Salmonella enterica serovar typhimurium (S. typhimurium) infection, induction of an inflammatory response has been linked to its virulence mechanism, the type III secretion system (T3SS). The T3SS secretes protein effectors that exploit the host’s cell biology to facilitate bacterial entry and intracellular survival, and to modulate the host immune response. As the role of the intestinal epithelium in initiating an immune response has been increasingly realized, this review will highlight recent research that details progress made in understanding mechanisms underlying the mucosal inflammatory response to Salmonella infection, and how such inflammatory responses impact pathogenic fitness of this organism.
Collapse
Affiliation(s)
- Samir Patel
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School , Worcester, MA , USA
| | - Beth A McCormick
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School , Worcester, MA , USA
| |
Collapse
|
40
|
Bai SP, Huang Y, Luo YH, Wang LL, Ding XM, Wang JP, Zeng QF, Zhang KY. Alteration in lymphocytes responses, cytokine and chemokine profiles in laying hens infected with Salmonella Typhimurium. Vet Immunol Immunopathol 2014; 160:235-43. [PMID: 24986046 DOI: 10.1016/j.vetimm.2014.05.015] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2014] [Revised: 05/06/2014] [Accepted: 05/15/2014] [Indexed: 11/24/2022]
Abstract
Salmonella Typhimurium has been reported to contaminate egg production across the world, but the exact nature of the immune mechanisms protective against Salmonella infection in laying hens has not been characterized at the molecular level. The experiment was conducted to determine Salmonella colonization and lymphocytes subpopulation in the ileum and spleen, and the mRNA expression of pro-inflammatory cytokines [interleukin (IL)-1β and IL-6], chemokine IL-8, and T helper (Th)1/Th2 cytokines [Interferon (IFN)-γ, IL-12 and IL-18; IL-4 and IL-10 respectively] in the cecal tonsil and spleen of Salmonella challenged hens. Forty Salmonella-free laying hens were challenged orally with Salmonella Typhimurium or phosphate-buffered saline (PBS; control). The Salmonella challenged or non-challenged hens (n=10) were sacrificed at 2 and 7 days post-infection (DPI). The lymphocyte subpopulation was determined via flow cytometric analysis in the ileum and spleen. The cecal tonsil and spleen samples were collected for mRNA expression through quantitative-RT-PCR. The Salmonella counts were higher (P<0.05) in the ileum than that in the spleen at 2 and 7DPI, and were higher (P<0.05) at 7DPI than that at 2DPI in the spleen. Salmonella challenge increased (P<0.05) ileal CD4+ and CD8α+ cells ratios at 2 and 7DPI, whereas it increased (P<0.05) splenic CD8α+ cells ratio only at 7DPI. The magnitude of increase in ileal CD8α+ cells ratio was higher (P<0.05) than that in CD4+ cells ratio. The mRNA expression of IL-1β, IL-6, IL-8, IFN-γ, IL-12 and IL-18 were significantly up-regulated in the cecal tonsil of Salmonella challenged hens, and the magnitude of increases in IL-6, IL-8 and IL-12 were significantly higher at 7DPI than that at 2DPI. However, Salmonella challenge increased (P<0.05) the mRNA expression of IL-1β, IL-10 and IL-18 at 2 and 7DPI, and IL-8 and IFN-γ mRNA only at 7DPI in the spleen. These findings demonstrated that there appeared the induction of cellular immune responses, and a Th1-cytokines reaction in the intestine and spleen of laying hens infected with Salmonella Typhimurium.
Collapse
Affiliation(s)
- Shi P Bai
- Institute of Animal Nutrition, Feed Engineering Research Centre of Sichuan Province, Sichuan Agricultural University, Ya'an 625014, PR China
| | - Yong Huang
- College of Veterinary Medicine, Sichuan Agricultural University, Ya'an, 625014, PR China
| | - Yu H Luo
- Institute of Animal Nutrition, Feed Engineering Research Centre of Sichuan Province, Sichuan Agricultural University, Ya'an 625014, PR China
| | - Lei L Wang
- Institute of Animal Nutrition, Feed Engineering Research Centre of Sichuan Province, Sichuan Agricultural University, Ya'an 625014, PR China
| | - Xue M Ding
- Institute of Animal Nutrition, Feed Engineering Research Centre of Sichuan Province, Sichuan Agricultural University, Ya'an 625014, PR China
| | - Jian P Wang
- Institute of Animal Nutrition, Feed Engineering Research Centre of Sichuan Province, Sichuan Agricultural University, Ya'an 625014, PR China
| | - Qiu F Zeng
- Institute of Animal Nutrition, Feed Engineering Research Centre of Sichuan Province, Sichuan Agricultural University, Ya'an 625014, PR China
| | - Ke Y Zhang
- Institute of Animal Nutrition, Feed Engineering Research Centre of Sichuan Province, Sichuan Agricultural University, Ya'an 625014, PR China.
| |
Collapse
|
41
|
Vaccination with a single CD4 T cell peptide epitope from a Salmonella type III-secreted effector protein provides protection against lethal infection. Infect Immun 2014; 82:2424-33. [PMID: 24686055 DOI: 10.1128/iai.00052-14] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Salmonella infections affect millions worldwide and remain a significant cause of morbidity and mortality. It is known from mouse studies that CD4 T cells are essential mediators of immunity against Salmonella infection, yet it is not clear whether targeting CD4 T cell responses directly with peptide vaccines against Salmonella can be effective in combating infection. Additionally, it is not known whether T cell responses elicited against Salmonella secreted effector proteins can provide protective immunity against infection. In this study, we investigated both of these possibilities using prime-boost immunization of susceptible mice with a single CD4 T cell peptide epitope from Salmonella secreted effector protein I (SseI), a component of the Salmonella type III secretion system. This immunization conferred significant protection against lethal oral infection, equivalent to that conferred by whole heat-killed Salmonella bacteria. Surprisingly, a well-characterized T cell epitope from the flagellar protein FliC afforded no protection compared to immunization with an irrelevant control peptide. The protective response appeared to be most associated with polyfunctional CD4 T cells raised against the SseI peptide, since no antibodies were produced against any of the peptides and very little CD8 T cell response was observed. Overall, this study demonstrates that eliciting CD4 T cell responses against components of the Salmonella type III secretion system can contribute to protection against infection and should be considered in the design of future Salmonella subunit vaccines.
Collapse
|
42
|
Regulatory T-cell vaccination independent of auto-antigen. Exp Mol Med 2014; 46:e82. [PMID: 24626168 PMCID: PMC3972794 DOI: 10.1038/emm.2014.4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2013] [Accepted: 12/06/2013] [Indexed: 12/21/2022] Open
Abstract
To date, efforts to treat autoimmune diseases have primarily focused on the disease symptoms rather than on the cause of the disease. In large part, this is attributed to not knowing the responsible auto-antigens (auto-Ags) for driving the self-reactivity coupled with the poor success of treating autoimmune diseases using oral tolerance methods. Nonetheless, if tolerogenic approaches or methods that stimulate regulatory T (Treg) cells can be devised, these could subdue autoimmune diseases. To forward such efforts, our approach with colonization factor antigen I (CFA/I) fimbriae is to establish bystander immunity to ultimately drive the development of auto-Ag-specific Treg cells. Using an attenuated Salmonella vaccine expressing CFA/I fimbriae, fimbriae-specific Treg cells were induced without compromising the vaccine's capacity to protect against travelers' diarrhea or salmonellosis. By adapting the vaccine's anti-inflammatory properties, it was found that it could also dampen experimental inflammatory diseases resembling multiple sclerosis (MS) and rheumatoid arthritis. Because of this bystander effect, disease-specific Treg cells are eventually induced to resolve disease. Interestingly, this same vaccine could elicit the required Treg cell subset for each disease. For MS-like disease, conventional CD25+ Treg cells are stimulated, but for arthritis CD39+ Treg cells are induced instead. This review article will examine the potential of treating autoimmune diseases without having previous knowledge of the auto-Ag using an innocuous antigen to stimulate Treg cells via the production of transforming growth factor-β and interleukin-10.
Collapse
|
43
|
Siggins MK, O'Shaughnessy CM, Pravin J, Cunningham AF, Henderson IR, Drayson MT, MacLennan CA. Differential timing of antibody-mediated phagocytosis and cell-free killing of invasive African Salmonella allows immune evasion. Eur J Immunol 2014; 44:1093-8. [PMID: 24375424 DOI: 10.1002/eji.201343529] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2013] [Revised: 10/17/2013] [Accepted: 12/19/2013] [Indexed: 11/07/2022]
Abstract
Nontyphoidal Salmonellae commonly cause fatal bacteraemia in African children lacking anti-Salmonella antibodies. These are facultative intracellular bacteria capable of cell-free and intracellular survival within macrophages. To better understand the relationship between extracellular and intracellular infection in blood and general mechanisms of Ab-related protection against Salmonella, we used human blood and sera to measure kinetics of Ab and complement deposition, serum-mediated bactericidal killing and phagocytosis of invasive African Salmonella enterica serovar Typhimurium D23580. Binding of antibodies peaked by 30 s, but C3 deposition lagged behind, peaking after 2-4 min. C5b-9 deposition was undetectable until between 2 and 6 min and peaked after 10 min, after which time an increase in serum-mediated killing occurred. In contrast, intracellular, opsonized Salmonellae were readily detectable within 5 min. By 10 min, around half of monocytes and most neutrophils contained bacteria. The same kinetics of serum-mediated killing and phagocytosis were observed with S. enterica Typhimurium laboratory strain SL1344, and the S. enterica Enteritidis African invasive isolate D24954 and laboratory strain PT4. The differential kinetics between cell-free killing and phagocytosis of invasive nontyphoidal Salmonella allows these bacteria to escape the blood and establish intracellular infection before they are killed by the membrane attack complex.
Collapse
Affiliation(s)
- Matthew K Siggins
- Medical Research Council Centre for Immune Regulation and Clinical Immunology Service, Institute of Biomedical Research, School of Immunity and Infection, College of Medicine and Dental Sciences, University of Birmingham, Birmingham, UK; Novartis Vaccines Institute for Global Health, Siena, Italy
| | | | | | | | | | | | | |
Collapse
|
44
|
Mastroeni P, Grant A. Dynamics of spread of Salmonella enterica in the systemic compartment. Microbes Infect 2013; 15:849-57. [PMID: 24183878 DOI: 10.1016/j.micinf.2013.10.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2013] [Accepted: 10/09/2013] [Indexed: 12/21/2022]
Abstract
Traditional microbiological and immunological tools, combined with modern imaging, and molecular and mathematical approaches, have revealed the dispersive nature of Salmonella infections. Bacterial escape from infected cells, spread in the tissues and attempts to restrain this process by the host give rise to fascinating scenarios that underpin the pathogenesis of salmonelloses.
Collapse
Affiliation(s)
- Pietro Mastroeni
- Department of Veterinary Medicine, University of Cambridge, Madingley Road, Cambridge CB3 0ES, United Kingdom.
| | | |
Collapse
|
45
|
Vigorito E, Kohlhaas S, Lu D, Leyland R. miR-155: an ancient regulator of the immune system. Immunol Rev 2013; 253:146-57. [PMID: 23550644 DOI: 10.1111/imr.12057] [Citation(s) in RCA: 235] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
MicroRNAs (miRNAs) are a newly recognized class of regulatory genes which repress the expression of protein-coding genes. Numerous studies have uncovered a complex role for miRNAs regulating many aspects of a variety of cellular processes including cell growth, differentiation, and lineage commitment. In the immune system, miR-155 is unique in its ability to shape the transcriptome of activated myeloid and lymphoid cells controlling diverse biological functions ranging from inflammation to immunological memory. Not surprisingly, a tight control of miR-155 expression is required to avoid malignant transformation, as evidenced by miR-155 overexpression in many cancers of B-cell origin. In this review, we discuss the potential of miR-155 as a molecular target for therapeutic intervention and discuss the function of miR-155 in the context of protective immunity. We first look back into the emergence of miR-155 in evolution, which is coincidental with the emergence of the ancestors of the antigen receptors. We then summarize what we have learned about the role of miR-155 in the regulation of lymphoid subsets at the cellular and molecular level in the context of recent progress in this field.
Collapse
Affiliation(s)
- Elena Vigorito
- Laboratory of Lymphocyte Signalling and Development, The Babraham Institute, Babraham Research Campus, Cambridge, UK.
| | | | | | | |
Collapse
|
46
|
Deobagkar-Lele M, Chacko SK, Victor ES, Kadthur JC, Nandi D. Interferon-γ- and glucocorticoid-mediated pathways synergize to enhance death of CD4(+) CD8(+) thymocytes during Salmonella enterica serovar Typhimurium infection. Immunology 2013. [PMID: 23186527 DOI: 10.1111/imm.12047] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Thymic atrophy is known to occur during infections; however, there is limited understanding of its causes and of the cross-talk between different pathways. This study investigates mechanisms involved in thymic atrophy during a model of oral infection by Salmonella enterica serovar Typhimurium (S. typhimurium). Significant death of CD4(+) CD8(+) thymocytes, but not of single-positive thymocytes or peripheral lymphocytes, is observed at later stages during infection with live, but not heat-killed, bacteria. The death of CD4(+) CD8(+) thymocytes is Fas-independent as shown by infection studies with lpr mice. However, apoptosis occurs with lowering of mitochondrial potential and higher caspase-3 activity. The amounts of cortisol, a glucocorticoid, and interferon-γ (IFN-γ), an inflammatory cytokine, increase upon infection. To investigate the functional roles of these molecules, studies were performed using Ifnγ(-/-) mice together with RU486, a glucocorticoid receptor antagonist. Treatment of C57BL/6 mice with RU486 does not affect colony-forming units (CFU), amounts of IFN-γ and mouse survival; however, there is partial rescue in thymocyte death. Upon infection, Ifnγ(-/-) mice display higher CFU and lower survival but more surviving thymocytes are recovered. However, there is no difference in cortisol amounts in C57BL/6 and Ifnγ(-/-) mice. Importantly, the number of CD4(+) CD8(+) thymocytes is significantly higher in Ifnγ(-/-) mice treated with RU486 along with lower caspase-3 activity and mitochondrial damage. Hence, endogenous glucocorticoid and IFN-γ-mediated pathways are parallel but synergize in an additive manner to induce death of CD4(+) CD8(+) thymocytes during S. typhimurium infection. The implications of this study for host responses during infection are discussed.
Collapse
|
47
|
A mathematical model representing cellular immune development and response to Salmonella of chicken intestinal tissue. J Theor Biol 2013; 330:75-87. [PMID: 23603730 DOI: 10.1016/j.jtbi.2013.04.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2012] [Revised: 04/05/2013] [Accepted: 04/09/2013] [Indexed: 12/28/2022]
Abstract
The aim of this study was to create a dynamic mathematical model of the development of the cellular branch of the intestinal immune system of poultry during the first 42 days of life and of its response towards an oral infection with Salmonella enterica serovar Enteritidis. The system elements were grouped in five important classes consisting of intra- and extracellular S. Enteritidis bacteria, macrophages, CD4+, and CD8+ cells. Twelve model variables were described by ordinary differential equations, including 50 parameters. Parameter values were estimated from literature or from own immunohistochemistry data. The model described the immune development in non-infected birds with an average R² of 0.87. The model showed less accuracy in reproducing the immune response to S. Enteritidis infection, with an average R² of 0.51, although model response did follow observed trends in time. Evaluation of the model against independent data derived from several infection trials showed strong/significant deviations from observed values. Nevertheless, it was shown that the model could be used to simulate the effect of varying input parameters on system elements response, such as the number of immune cells at hatch. Model simulations allowed one to study the sensitivity of the model outcome for varying model inputs. The initial number of immune cells at hatch was shown to have a profound impact on the predicted development in the number of systemic S. Enteritidis bacteria after infection. The theoretical contribution of this work is the identification of responses in system elements of the developing intestinal immune system of poultry obtaining a mathematical representation which allows one to explore the relationships between these elements under contrasting environmental conditions during different stages of intestinal development.
Collapse
|
48
|
Contribution of Thy1+ NK cells to protective IFN-γ production during Salmonella typhimurium infections. Proc Natl Acad Sci U S A 2013; 110:2252-7. [PMID: 23345426 DOI: 10.1073/pnas.1222047110] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
IFN-γ is critical for immunity against infections with intracellular pathogens, such as Salmonella enterica. However, which of the many cell types capable of producing IFN-γ controls Salmonella infections remains unclear. Using a mouse model of systemic Salmonella infection, we observed that only a lack of all lymphocytes or CD90 (Thy1)(+) cells, but not the absence of T cells, Retinoic acid-related orphan receptor (ROR)-γt-dependent lymphocytes, (NK)1.1(+) cells, natural killer T (NKT), and/or B cells alone, replicated the highly susceptible phenotype of IFN-γ-deficient mice to Salmonella infection. A combination of antibody depletions and adoptive transfer experiments revealed that early protective IFN-γ was provided by Thy1-expressing natural killer (NK) cells and that these cells improved antibacterial immunity through the provision of IFN-γ. Further analysis of NK cells producing IFN-γ in response to Salmonella indicated that less mature NK cells were more efficient at mediating antibacterial effector function than terminally differentiated NK cells. Inspired by recent reports of Thy1(+) NK cells contributing to immune memory, we analyzed their role in secondary protection against otherwise lethal WT Salmonella infections. Notably, we observed that a newly generated Salmonella vaccine strain not only conferred superior protection compared with conventional regimens but that this enhanced efficiency of recall immunity was afforded by incorporating CD4(-)CD8(-)Thy1(+) cells into the secondary response. Taken together, these findings demonstrate that Thy1-expressing NK cells play an important role in antibacterial immunity.
Collapse
|
49
|
Shippy DC, Fadl AA. Immunological characterization of a gidA mutant strain of Salmonella for potential use in a live-attenuated vaccine. BMC Microbiol 2012. [PMID: 23194372 PMCID: PMC3520829 DOI: 10.1186/1471-2180-12-286] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Background Salmonella is often associated with gastrointestinal disease outbreaks in humans throughout the world due to the consumption of contaminated food. Our previous studies have shown that deletion of glucose-inhibited division gene (gidA) significantly attenuated Salmonella enterica serovar Typhimurium (STM) virulence in both in vitro and in vivo models of infection. Most importantly, immunization with the gidA mutant protected mice from a lethal dose challenge of wild-type STM. In this study, we further characterize the gidA mutant STM strain for potential use in a live-attenuated vaccine. Results The protective efficacy of immunization with the gidA mutant was evaluated by challenging immunized mice with a lethal dose of wild-type STM. Sera levels of IgG2a and IgG1, passive transfer of sera and cells, and cytokine profiling were performed to study the induction of humoral and cellular immune responses induced by immunization with the gidA mutant strain. Additionally, a lymphocyte proliferation assay was performed to gauge the splenocyte survival in response to treatment with STM cell lysate. Mice immunized with the gidA mutant strain were fully protected from a lethal dose challenge of wild-type STM. Naïve mice receiving either cells or sera from immunized mice were partially protected from a lethal dose challenge of wild-type STM. The lymphocyte proliferation assay displayed a significant response of splenocytes from immunized mice when compared to splenocytes from non-immunized control mice. Furthermore, the immunized mice displayed significantly higher levels of IgG1 and IgG2a with a marked increase in IgG1. Additionally, immunization with the gidA mutant strain evoked higher levels of IL-2, IFN-γ, and IL-10 cytokines in splenocytes induced with STM cell lysate. Conclusions Together, the results demonstrate that immunization with the gidA mutant strain protects mice by inducing humoral and cellular immune responses with the humoral immune response potentially being the main mechanism of protection.
Collapse
Affiliation(s)
- Daniel C Shippy
- Department of Animal Sciences, University of Wisconsin-Madison, 1675 Observatory Dr, Madison, WI 53706, USA
| | | |
Collapse
|
50
|
Nanton MR, Way SS, Shlomchik MJ, McSorley SJ. Cutting edge: B cells are essential for protective immunity against Salmonella independent of antibody secretion. THE JOURNAL OF IMMUNOLOGY 2012; 189:5503-7. [PMID: 23150714 DOI: 10.4049/jimmunol.1201413] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Typhoid fever and nontyphoidal bacteremia caused by Salmonella remain critical human health problems. B cells are required for protective immunity to Salmonella, but the mechanism of protection remains unclear. In this study, we immunized wild-type, B cell-deficient, Ab-deficient, and class-switched Ab-deficient mice with attenuated Salmonella and examined protection against secondary infection. As expected, wild-type mice were protected and B cell-deficient mice succumbed to secondary infection. Interestingly, mice with B cells but lacking secreted Ab or class-switched Ab had little deficiency in resistance to Salmonella infection. The susceptibility of B cell-deficient mice correlated with marked reductions in CD4 T cell IFN-γ production after secondary infection. Taken together, these data suggest that the primary role of B cells in acquired immunity to Salmonella is via the development of protective T cell immunity.
Collapse
Affiliation(s)
- Minelva R Nanton
- Department of Pediatric Infectious Disease, Center for Infectious Diseases and Microbiology Translational Research, University of Minnesota Medical School-Twin Cities, Minneapolis, MN 55455, USA.
| | | | | | | |
Collapse
|