1
|
You Y, Zhao X, Jie J, Xie Y, Hao Z, He Q, Zhou Y. Construction and evaluation of a Salmonella Paratyphi A vaccine candidate based on a poxA gene mutation. Gene 2025; 933:148952. [PMID: 39299530 DOI: 10.1016/j.gene.2024.148952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 09/13/2024] [Accepted: 09/16/2024] [Indexed: 09/22/2024]
Abstract
Salmonella Paratyphi A, the pathogen of paratyphoid A accounts for an obviously growing proportion of cases in many areas. Therefore, development of specific paratyphoid A vaccines is needed. In the present study, the poxA gene of Salmonella Paratyphi A, encoding the aminoacyl-tRNA synthetase, was deleted successfully by the method of lambda Red recombination system, the resulting strain, ΔpoxA was characterized in respect of growth, adhesion and invasion, virulence, immunogenicity and protective efficacy. It was found that the growth of the ΔpoxA strain was significantly delayed compared with the wild type strain, the mutant ΔpoxA was less invasive to Caco-2 BBE epithelioid cells and THP-1 macrophages than the wild type strain, strain ΔpoxA was attenuated at least 1000-fold in mice, significant immune response and efficient protection were provided by the mutant ΔpoxA after oral immunization. It is concluded that the Salmonella Paratyphi A poxA deletion mutant ΔpoxA can be used as a live oral vaccine candidate against paratyphoid A.
Collapse
Affiliation(s)
- Yonghe You
- School of Basic Medical Sciences, Sanquan College of Xinxiang Medical University, Xinxiang, China
| | - Xiaohui Zhao
- School of Basic Medical Sciences, Sanquan College of Xinxiang Medical University, Xinxiang, China
| | - Jiayue Jie
- Department of Basic Medical Sciences, Zhengzhou Medical and Health Vocational College, Zhengzhou, China
| | - Yongsheng Xie
- School of Basic Medical Sciences, Sanquan College of Xinxiang Medical University, Xinxiang, China
| | - Zhenhua Hao
- School of Basic Medical Sciences, Sanquan College of Xinxiang Medical University, Xinxiang, China
| | - Qunli He
- Department of Basic Medical Sciences, Zhengzhou Medical and Health Vocational College, Zhengzhou, China; School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China.
| | - Yanlin Zhou
- School of Basic Medical Sciences, Sanquan College of Xinxiang Medical University, Xinxiang, China.
| |
Collapse
|
2
|
Muturi P, Wachira P, Wagacha M, Mbae C, Kavai SM, Mugo MM, Mohamed M, González JF, Kariuki S, Gunn JS. Salmonella Typhi Haplotype 58 biofilm formation and genetic variation in isolates from typhoid fever patients with gallstones in an endemic setting in Kenya. Front Cell Infect Microbiol 2024; 14:1468866. [PMID: 39606745 PMCID: PMC11599249 DOI: 10.3389/fcimb.2024.1468866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 10/09/2024] [Indexed: 11/29/2024] Open
Abstract
Although typhoid fever has largely been eliminated in high-income countries, it remains a major global public health concern especially among low- and middle-income countries. The causative agent, Salmonella enterica serovar Typhi (S. Typhi), is a human restricted pathogen with a limited capacity to replicate outside the human host. Human carriers, 90% of whom have gallstones in their gallbladder, continue to shed the pathogen for an ill-defined period of time after treatment. The genetic mechanisms involved in establishing the carrier state are poorly understood, but S. Typhi is thought to undergo specific genetic changes within the gallbladder as an adaptive mechanism. In the current study, we aimed to identify the genetic differences in longitudinal clinical S. Typhi isolates from asymptomatic carriers with gallstones in a typhoid endemic setting in Nairobi, Kenya. Whole-genome sequences were analyzed from 22 S. Typhi isolates, 20 from stool samples, and 2 from blood samples, all genotype 4.3.1 (H58). Out of this, 19 strains were from four patients also diagnosed with gallstones, of whom three had typhoid symptoms and continued to shed S. Typhi after treatment. All isolates had point mutations in the quinolone resistance-determining region (QRDR), and only sub-lineage 4.3.1.2.EA3 encoded multidrug resistance genes. There was no variation in antimicrobial resistance patterns among strains from the same patient/household. Non-multidrug resistant (MDR) isolates formed significantly stronger biofilms in vitro than the MDR isolates, p<0.001. A point mutation within the treB gene (treB A383T) was observed in strains isolated after clinical resolution from patients living in 75% of the households. For missense mutations in Vi capsular polysaccharide genes, tviE P263S was also observed in 18% of the isolates. This study provides insights into the role of typhoid carriage, biofilm formation, AMR genes, and genetic variations in S. Typhi during asymptomatic carriage.
Collapse
Affiliation(s)
- Peter Muturi
- Centre for Microbiology Research, Kenya Medical Research Institute, Nairobi, Kenya
- Department of Biology, University of Nairobi, Nairobi, Kenya
| | - Peter Wachira
- Department of Biology, University of Nairobi, Nairobi, Kenya
| | - Maina Wagacha
- Department of Biology, University of Nairobi, Nairobi, Kenya
| | - Cecilia Mbae
- Centre for Microbiology Research, Kenya Medical Research Institute, Nairobi, Kenya
| | - Susan M. Kavai
- Centre for Microbiology Research, Kenya Medical Research Institute, Nairobi, Kenya
| | - Michael M. Mugo
- Centre for Microbiology Research, Kenya Medical Research Institute, Nairobi, Kenya
| | - Musa Mohamed
- Department of Medical Services, Ministry of Health, Nairobi, Kenya
| | - Juan F. González
- Center for Microbial Pathogenesis, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH, United States
| | - Samuel Kariuki
- Centre for Microbiology Research, Kenya Medical Research Institute, Nairobi, Kenya
- Eastern Africa Office, Drugs for Neglected Diseases initiative, Nairobi, Kenya
| | - John S. Gunn
- Center for Microbial Pathogenesis, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH, United States
- Infectious Diseases Institute, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
3
|
Muturi P, Wachira P, Wagacha M, Mbae C, Kavai S, Mugo M, Muhammed M, González JF, Kariuki S, Gunn JS. Salmonella Typhi Haplotype 58 (H58) Biofilm Formation and Genetic Variation in Typhoid Fever Patients with Gallstones in an Endemic Setting in Kenya. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.06.03.24308409. [PMID: 38883710 PMCID: PMC11177912 DOI: 10.1101/2024.06.03.24308409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2024]
Abstract
The causative agent of typhoid fever, Salmonella enterica serovar Typhi, is a human restricted pathogen. Human carriers, 90% of whom have gallstones in their gallbladder, continue to shed the pathogen after treatment. The genetic mechanisms involved in establishing the carrier state are poorly understood, but S. Typhi is thought to undergo specific genetic changes within the gallbladder as an adaptive mechanism. In the current study, we aimed to identify biofilm forming ability and the genetic differences in longitudinal clinical S. Typhi isolates from asymptomatic carriers with gallstones in Nairobi, Kenya. Whole genome sequences were analyzed from 22 S. Typhi isolates, 20 from stool and 2 from blood samples, all genotype 4.3.1 (H58). Nineteen strains were from four patients also diagnosed with gallstones, of whom, three had typhoid symptoms and continued to shed S. Typhi after treatment. All isolates had point mutations in the quinolone resistance determining region (QRDR) and only sub-lineage 4.3.1.2EA3 encoded multidrug resistance genes. There was no variation in antimicrobial resistance patterns among strains from the same patient/household. Non-multidrug resistant (MDR), isolates formed significantly stronger biofilms in vitro than the MDR isolates, p<0.001. A point mutation within the treB gene (treB A383T) was observed in strains isolated after clinical resolution from patients living in 75% of the households. Missense mutations in Vi capsular polysaccharide genes, tviE P263S was also observed in 18% of the isolates. This study provides insights into the role of typhoid carriage, biofilm formation, AMR genes and genetic variations in S. Typhi from asymptomatic carriers.
Collapse
Affiliation(s)
- Peter Muturi
- Centre for Microbiology Research, Kenya Medical Research Institute
- Department of Biology, University of Nairobi, Kenya
| | | | | | - Cecilia Mbae
- Centre for Microbiology Research, Kenya Medical Research Institute
| | - Susan Kavai
- Centre for Microbiology Research, Kenya Medical Research Institute
| | - Michael Mugo
- Centre for Microbiology Research, Kenya Medical Research Institute
| | | | - Juan F. González
- Center for Microbial Pathogenesis, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH, USA
| | - Samuel Kariuki
- Centre for Microbiology Research, Kenya Medical Research Institute
- Wellcome Sanger Institute, Cambridge, United Kingdom
- Drugs for Neglected Diseases initiative Eastern Africa, Nairobi, Kenya
| | - John S. Gunn
- Center for Microbial Pathogenesis, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH, USA
- Infectious Diseases Institute, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
4
|
Stepien TA, Singletary LA, Guerra FE, Karlinsey JE, Libby SJ, Jaslow SL, Gaggioli MR, Gibbs KD, Ko DC, Brehm MA, Greiner DL, Shultz LD, Fang FC. Nuclear factor kappa B-dependent persistence of Salmonella Typhi and Paratyphi in human macrophages. mBio 2024; 15:e0045424. [PMID: 38497655 PMCID: PMC11005419 DOI: 10.1128/mbio.00454-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 02/21/2024] [Indexed: 03/19/2024] Open
Abstract
Salmonella serovars Typhi and Paratyphi cause a prolonged illness known as enteric fever, whereas other serovars cause acute gastroenteritis. Mechanisms responsible for the divergent clinical manifestations of nontyphoidal and enteric fever Salmonella infections have remained elusive. Here, we show that S. Typhi and S. Paratyphi A can persist within human macrophages, whereas S. Typhimurium rapidly induces apoptotic macrophage cell death that is dependent on Salmonella pathogenicity island 2 (SPI2). S. Typhi and S. Paratyphi A lack 12 specific SPI2 effectors with pro-apoptotic functions, including nine that target nuclear factor κB (NF-κB). Pharmacologic inhibition of NF-κB or heterologous expression of the SPI2 effectors GogA or GtgA restores apoptosis of S. Typhi-infected macrophages. In addition, the absence of the SPI2 effector SarA results in deficient signal transducer and activator of transcription 1 (STAT1) activation and interleukin 12 production, leading to impaired TH1 responses in macrophages and humanized mice. The absence of specific nontyphoidal SPI2 effectors may allow S. Typhi and S. Paratyphi A to cause chronic infections. IMPORTANCE Salmonella enterica is a common cause of gastrointestinal infections worldwide. The serovars Salmonella Typhi and Salmonella Paratyphi A cause a distinctive systemic illness called enteric fever, whose pathogenesis is incompletely understood. Here, we show that enteric fever Salmonella serovars lack 12 specific virulence factors possessed by nontyphoidal Salmonella serovars, which allow the enteric fever serovars to persist within human macrophages. We propose that this fundamental difference in the interaction of Salmonella with human macrophages is responsible for the chronicity of typhoid and paratyphoid fever, suggesting that targeting the nuclear factor κB (NF-κB) complex responsible for macrophage survival could facilitate the clearance of persistent bacterial infections.
Collapse
Affiliation(s)
- Taylor A. Stepien
- Department of Global Health, University of Washington, Seattle, Washington, USA
| | | | - Fermin E. Guerra
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
| | - Joyce E. Karlinsey
- Department of Microbiology, University of Washington, Seattle, Washington, USA
| | - Stephen J. Libby
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
| | - Sarah L. Jaslow
- Department of Molecular Genetics and Microbiology, Duke University, Durham, North Carolina, USA
| | - Margaret R. Gaggioli
- Department of Molecular Genetics and Microbiology, Duke University, Durham, North Carolina, USA
| | - Kyle D. Gibbs
- Department of Molecular Genetics and Microbiology, Duke University, Durham, North Carolina, USA
| | - Dennis C. Ko
- Department of Molecular Genetics and Microbiology, Duke University, Durham, North Carolina, USA
| | - Michael A. Brehm
- Department of Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Dale L. Greiner
- Department of Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | | | - Ferric C. Fang
- Department of Global Health, University of Washington, Seattle, Washington, USA
- Department of Microbiology, University of Washington, Seattle, Washington, USA
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
| |
Collapse
|
5
|
Liu RH, Sun AQ, Liao Y, Tang ZX, Zhang SH, Shan X, Hu JT. Lactiplantibacillus plantarum Regulated Intestinal Microbial Community and Cytokines to Inhibit Salmonella typhimurium Infection. Probiotics Antimicrob Proteins 2023; 15:1355-1370. [PMID: 36074298 DOI: 10.1007/s12602-022-09987-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/24/2022] [Indexed: 11/25/2022]
Abstract
Lactic acid bacteria (LAB) are recognized as food-grade safe microorganisms and have many beneficial effects. LAB could maintain the host intestinal homeostasis and regulate intestinal microbial community to exert antibacterial effects. In this study, Lactiplantibacillus plantarum (L. plantarum, Lp01) strain isolated from pig intestine was orally administered to C57BL/6 mice, and mice were then infected with Salmonella typhimurium (ATCC14028). The protective effects of L. plantarum were evaluated by monitoring body weight loss, survival rates, bacterial loads in tissue, colon histopathology analysis, and cytokine secretion. 16S rRNA gene sequencing was also utilized to detect the dynamics of the blind gut microbial community in mice. We found that L. plantarum could significantly reduce the body weight loss and improve the survival rates. The survival rate in the L. P-Sty group was up to 67.5%, which was much higher than that in the STY group (25%). Counting of bacterial loads displayed that the colony-forming unit (CFU) of S. typhimurium in the spleen (p < 0.05) and the liver (p < 0.05) from L. P-Sty group both decreased, compared with STY group. Intestinal histopathology showed that it alleviated the intestinal injury caused by Salmonella, inhibited the secretion of pro-inflammatory cytokines, and promoted anti-inflammatory cytokines (p < 0. 01). In addition, L. plantarum also significantly ameliorated the intestinal gut microbiome disturbance caused by Salmonella. It displayed an obvious increase of beneficial bacteria including Lactobacillus and Bacteroidetes and reduction of pathogenic bacteria like Proteobacteria. In conclusion, L. plantarum could regulate microbial community to inhibit Salmonella typhimurium infection.
Collapse
Affiliation(s)
- Rui-Han Liu
- College of Animal Science and Technology, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - An-Qi Sun
- College of Animal Science and Technology, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Ye Liao
- College of Animal Science and Technology, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Zheng-Xu Tang
- College of Animal Science and Technology, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Shi-Han Zhang
- College of Animal Science and Technology, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Xin Shan
- College of Animal Science and Technology, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Jing-Tao Hu
- College of Animal Science and Technology, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Jilin Agricultural University, Changchun, China.
| |
Collapse
|
6
|
Thurston TLM, Holden DW. The Salmonella Typhi SPI-2 injectisome enigma. MICROBIOLOGY (READING, ENGLAND) 2023; 169:001405. [PMID: 37862087 PMCID: PMC10634361 DOI: 10.1099/mic.0.001405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 10/11/2023] [Indexed: 10/21/2023]
Abstract
The Salmonella pathogenicity island 2 (SPI-2)-encoded type III secretion system (injectisome) is assembled following uptake of bacteria into vacuoles in mammalian cells. The injectisome translocates virulence proteins (effectors) into infected cells. Numerous studies have established the requirement for a functional SPI-2 injectisome for growth of Salmonella Typhimurium in mouse macrophages, but the results of similar studies involving Salmonella Typhi and human-derived macrophages are not consistent. It is important to clarify the functions of the S. Typhi SPI-2 injectisome, not least because an inactivated SPI-2 injectisome forms the basis for live attenuated S. Typhi vaccines that have undergone extensive trials in humans. Intracellular expression of injectisome genes and effector delivery take longer in the S. Typhi/human macrophage model than for S. Typhimurium and we propose that this could explain the conflicting results. Furthermore, strains of both S. Typhimurium and S. Typhi contain intact genes for several 'core' effectors. In S. Typhimurium these cooperate to regulate the vacuole membrane and contribute to intracellular bacterial replication; similar functions are therefore likely in S. Typhi.
Collapse
Affiliation(s)
- Teresa L. M. Thurston
- Department of Infectious Disease, Centre for Bacterial Resistance Biology, Imperial College London, London, SW7 2AZ, UK
| | - David W. Holden
- Department of Infectious Disease, Centre for Bacterial Resistance Biology, Imperial College London, London, SW7 2AZ, UK
| |
Collapse
|
7
|
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
|
8
|
Wang D, Zheng Y, Fan Y, He Y, Liu K, Deng S, Liu Y. Sodium Humate-Derived Gut Microbiota Ameliorates Intestinal Dysfunction Induced by Salmonella Typhimurium in Mice. Microbiol Spectr 2023; 11:e0534822. [PMID: 37067423 PMCID: PMC10269575 DOI: 10.1128/spectrum.05348-22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Accepted: 03/21/2023] [Indexed: 04/18/2023] Open
Abstract
Salmonella is a foodborne pathogen that is one of the main causes of gastroenteric disease in humans and animals. As a natural organic substance, sodium humate (HNa) possesses antibacterial, antidiarrheal, and anti-inflammatory properties. However, it is unclear whether the HNa and HNa-derived microbiota exert alleviative effects on Salmonella enterica serovar Typhimurium-induced enteritis. We found that treatment with HNa disrupted the cell wall of S. Typhimurium and decreased the virulence gene expression. Next, we explored the effect of HNa presupplementation on S. Typhimurium-induced murine enteritis. The results revealed that HNa ameliorated intestinal pathological damage. In addition, we observed that presupplementation with HNa enhanced intestinal barrier function via modulating gut microbiota, downregulating toll-like receptor 4 (TLR4)/nuclear factor kappa-B (NF-κB) and NOD-like receptor protein 3 (NLRP3) signaling pathways, regulating intestinal mucosal immunity, and enhancing tight junction protein expression. To further validate the effect of HNa-derived microbiota on S. Typhimurium-induced enteritis, we performed fecal microbiota transplantation and found that HNa-derived microbiota also alleviated S. Typhimurium-induced intestinal damage. It is noteworthy that both HNa and HNa-derived microbiota improved the liver injury caused by S. Typhimurium infection. Collectively, this is the first study to confirm that HNa could alleviate S. Typhimurium-induced enteritis in a gut microbiota-dependent manner. This study provides a new perspective on HNa as a potential drug to prevent and treat salmonellosis. IMPORTANCE Salmonella Typhimurium is an important zoonotic pathogen, widely distributed in nature. S. Typhimurium is one of the leading causes of foodborne illnesses worldwide, and more than 350,000 people died from Salmonella infection each year, which poses a substantial risk to public health and causes a considerable economic loss. Here, we found that the S. Typhimurium infection caused severe intestinal and liver damage. In addition, we first found that sodium humate (HNa) and HNa-derived gut microbiota can alleviate S. Typhimurium infection-induced intestinal damage. These findings extend the knowledge about the public health risk and pathogenic mechanisms of S. Typhimurium.
Collapse
Affiliation(s)
- Dong Wang
- Heilongjiang Key Laboratory of Experimental Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
- College of Veterinary Medicine, Shandong Agricultural University, Tai’an, China
| | - Yingce Zheng
- Heilongjiang Key Laboratory of Experimental Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Yuying Fan
- Heilongjiang Key Laboratory of Experimental Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Yanjun He
- Heilongjiang Key Laboratory of Experimental Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Kexin Liu
- Heilongjiang Key Laboratory of Experimental Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Shouxiang Deng
- Heilongjiang Key Laboratory of Experimental Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Yun Liu
- Heilongjiang Key Laboratory of Experimental Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| |
Collapse
|
9
|
Zala SM, Church B, Potts WK, Knauer F, Penn DJ. Female scent accelerates growth of juvenile male mice. Sci Rep 2023; 13:7371. [PMID: 37147391 PMCID: PMC10163255 DOI: 10.1038/s41598-023-34548-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 05/03/2023] [Indexed: 05/07/2023] Open
Abstract
Exposing female house mice (Mus musculus) to male urinary scent accelerates their sexual development (Vandenbergh effect). Here, we tested whether exposing juvenile male mice to females' urine similarly influences male growth and size of their sexual organs. We exposed three-week old male house mice to female urine or water (control) for ca. three months. We found that female-exposed males grew significantly faster and gained more body mass than controls, despite all males being reared on a controlled diet, but we detected no differences in males' muscle mass or sexual organs. In contrast, exposing juvenile males to male urine had no effect their growth. We tested whether the males' accelerated growth imposed functional trade-offs on males' immune resistance to an experimental infection. We challenged the same male subjects with an avirulent bacterial pathogen (Salmonella enterica), but found no evidence that faster growth impacted their bacterial clearance, body mass or survival during infection compared to controls. Our results provide the first evidence to our knowledge that juvenile male mice accelerate their growth when exposed to the urine of adult females, though we found no evidence that increased growth had negative trade-offs on immune resistance to infectious disease.
Collapse
Affiliation(s)
- Sarah M Zala
- Konrad Lorenz Institute of Ethology, University of Veterinary Medicine, Vienna, Savoyenstrasse 1a, 1160, Vienna, Austria.
- Department of Biology, University of Utah, 257 S. 1400 E., Salt Lake City, UT, 84112, USA.
| | - Brian Church
- Department of Biology, University of Utah, 257 S. 1400 E., Salt Lake City, UT, 84112, USA
| | - Wayne K Potts
- Department of Biology, University of Utah, 257 S. 1400 E., Salt Lake City, UT, 84112, USA
| | - Felix Knauer
- Research Institute of Wildlife Ecology, University of Veterinary Medicine, Vienna, 1160, Vienna, Austria
| | - Dustin J Penn
- Konrad Lorenz Institute of Ethology, University of Veterinary Medicine, Vienna, Savoyenstrasse 1a, 1160, Vienna, Austria
- Department of Biology, University of Utah, 257 S. 1400 E., Salt Lake City, UT, 84112, USA
| |
Collapse
|
10
|
Sedivy-Haley K, Blimkie T, Falsafi R, Lee AHY, Hancock REW. A transcriptomic analysis of the effects of macrophage polarization and endotoxin tolerance on the response to Salmonella. PLoS One 2022; 17:e0276010. [PMID: 36240188 PMCID: PMC9565388 DOI: 10.1371/journal.pone.0276010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Accepted: 09/27/2022] [Indexed: 11/19/2022] Open
Abstract
Salmonella is an intracellular pathogen causing significant morbidity and mortality. Its ability to grow inside macrophages is important to virulence, and is dependent on the activation state of the macrophages. Classically activated M1 macrophages are non-permissive for Salmonella growth, while alternatively activated M2 macrophages are permissive for Salmonella growth. Here we showed that endotoxin-primed macrophages (MEP), such as those associated with sepsis, showed similar levels of Salmonella resistance to M1 macrophages after 2 hr of intracellular infection, but at the 4 hr and 24 hr time points were susceptible like M2 macrophages. To understand this mechanistically, transcriptomic sequencing, RNA-Seq, was performed. This showed that M1 and MEP macrophages that had not been exposed to Salmonella, demonstrated a process termed here as primed activation, in expressing relatively higher levels of particular anti-infective genes and pathways, including the JAK-STAT (Janus kinase-signal transducer and activator of transcription) pathway. In contrast, in M2 macrophages these genes and pathways were largely expressed only in response to infection. Conversely, in response to infection, M1 macrophages, but not MEP macrophages, modulated additional genes known to be associated with susceptibility to Salmonella infection, possibly contributing to the differences in resistance at later time points. Application of the JAK inhibitor Ruxolitinib before infection reduced resistance in M1 macrophages, supporting the importance of early JAK-STAT signalling in M1 resistance to Salmonella.
Collapse
Affiliation(s)
- Katharine Sedivy-Haley
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Travis Blimkie
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Reza Falsafi
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Amy Huei-Yi Lee
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Robert E W Hancock
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
11
|
Becerra-Báez EI, Meza-Toledo SE, Muñoz-López P, Flores-Martínez LF, Fraga-Pérez K, Magaño-Bocanegra KJ, Juárez-Hernández U, Mateos-Chávez AA, Luria-Pérez R. Recombinant Attenuated Salmonella enterica as a Delivery System of Heterologous Molecules in Cancer Therapy. Cancers (Basel) 2022; 14:cancers14174224. [PMID: 36077761 PMCID: PMC9454573 DOI: 10.3390/cancers14174224] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 08/09/2022] [Accepted: 08/28/2022] [Indexed: 11/17/2022] Open
Abstract
Simple Summary Cancer is among the main causes of death of millions of individuals worldwide. Although survival has improved with conventional treatments, the appearance of resistant cancer cells leads to patient relapses. It is, therefore, necessary to find new antitumor therapies that can completely eradicate transformed cells. Bacteria-based tumor therapy represents a promising alternative treatment, particularly the use of live-attenuated Salmonella enterica, with its potential use as a delivery system of antitumor heterologous molecules such as tumor-associated antigens, cytotoxic molecules, immunomodulatory molecules, pro-apoptotic proteins, nucleic acids, and nanoparticles. In this review, we present the state of the art of current preclinical and clinical research on the use of Salmonella enterica as a potential therapeutic ally in the war against cancer. Abstract Over a century ago, bacterial extracts were found to be useful in cancer therapy, but this treatment modality was obviated for decades. Currently, in spite of the development and advances in chemotherapies and radiotherapy, failure of these conventional treatments still represents a major issue in the complete eradication of tumor cells and has led to renewed approaches with bacteria-based tumor therapy as an alternative treatment. In this context, live-attenuated bacteria, particularly Salmonella enterica, have demonstrated tumor selectivity, intrinsic oncolytic activity, and the ability to induce innate or specific antitumor immune responses. Moreover, Salmonella enterica also has strong potential as a delivery system of tumor-associated antigens, cytotoxic molecules, immunomodulatory molecules, pro-apoptotic proteins, and nucleic acids into eukaryotic cells, in a process known as bactofection and antitumor nanoparticles. In this review, we present the state of the art of current preclinical and clinical research on the use of Salmonella enterica as a potential therapeutic ally in the war against cancer.
Collapse
Affiliation(s)
- Elayne Irene Becerra-Báez
- Unit of Investigative Research on Hemato-Oncological Diseases, Children’s Hospital of Mexico Federico Gomez, Mexico City 06720, Mexico
- Departamento de Bioquímica, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 11340, Mexico
| | - Sergio Enrique Meza-Toledo
- Departamento de Bioquímica, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 11340, Mexico
| | - Paola Muñoz-López
- Unit of Investigative Research on Hemato-Oncological Diseases, Children’s Hospital of Mexico Federico Gomez, Mexico City 06720, Mexico
- Departamento de Microbiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 11340, Mexico
| | - Luis Fernando Flores-Martínez
- Unit of Investigative Research on Hemato-Oncological Diseases, Children’s Hospital of Mexico Federico Gomez, Mexico City 06720, Mexico
- Departamento de Microbiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 11340, Mexico
| | - Karla Fraga-Pérez
- Unit of Investigative Research on Hemato-Oncological Diseases, Children’s Hospital of Mexico Federico Gomez, Mexico City 06720, Mexico
| | - Kevin Jorge Magaño-Bocanegra
- Unit of Investigative Research on Hemato-Oncological Diseases, Children’s Hospital of Mexico Federico Gomez, Mexico City 06720, Mexico
- Department of Molecular Biomedicine, Center for Research and Advanced Studies of the National Polytechnic Institute, Mexico City 07360, Mexico
| | - Uriel Juárez-Hernández
- Unit of Investigative Research on Hemato-Oncological Diseases, Children’s Hospital of Mexico Federico Gomez, Mexico City 06720, Mexico
- Department of Molecular Biomedicine, Center for Research and Advanced Studies of the National Polytechnic Institute, Mexico City 07360, Mexico
| | - Armando Alfredo Mateos-Chávez
- Unit of Investigative Research on Hemato-Oncological Diseases, Children’s Hospital of Mexico Federico Gomez, Mexico City 06720, Mexico
| | - Rosendo Luria-Pérez
- Unit of Investigative Research on Hemato-Oncological Diseases, Children’s Hospital of Mexico Federico Gomez, Mexico City 06720, Mexico
- Correspondence: ; Tel.: +52-55-52289917 (ext. 4401)
| |
Collapse
|
12
|
Abstract
Typhoid fever is caused primarily by the enteric microbe Salmonella enterica serovar Typhi and remains a major global health problem with approximately 14 million new infections and 136,000 fatalities annually. While there are antibiotic options available to treat the disease, the global increase in multidrug-resistant strains necessitates alternative therapeutic options. Host-targeted therapeutics present a promising anti-infective strategy against intracellular bacterial pathogens. A cell-based assay identified a compound that inhibits Salmonella proliferation in infected cells, 2-(3-hydroxypropyl)-1-(3-phenoxyphenyl)-1,2-dihydrochromeno[2,3-c]pyrrole-3,9-dione (KH-1), which is devoid of direct activity against Salmonella. The compound inhibits the growth of both antibiotic-sensitive and -resistant Salmonella strains inside macrophages and reduces lactate dehydrogenase (LDH) release from Salmonella-infected cells. Subsequent screening of KH-1 commercial analogs identified 2-(4-fluorobenzyl)-1-(3-phenoxyphenyl)-1,2-dihydrochromeno[2,3-c] pyrrole-3,9-dione (KH-1-2), which is more effective in controlling Salmonella growth inside macrophages. In vitro KH-1-2 treatment of Salmonella infection resulted in an 8- to 10-fold reduction in bacterial load in infected macrophages. In combination with suboptimal ciprofloxacin treatment, KH-1-2 further reduces Salmonella growth inside macrophages. The toxicity and efficacy of KH-1-2 in controlling Salmonella infection were examined in vivo using a mouse model of typhoid fever. No significant compound-related clinical signs and histological findings of the liver, spleen, or kidney were observed from uninfected mice that were intraperitoneally treated with KH-1-2. KH-1-2 significantly protected mice from a lethal dose of infection by an antibiotic-resistant Salmonella strain. Thus, our study provides support that this is a promising lead compound for the development of a novel host-targeted therapeutic agent to control typhoid fever. IMPORTANCESalmonella spp. cause significant morbidity and mortality worldwide. Typhoidal spp. (e.g., S. Typhi) cause a systemic disease typically treated with antibiotics. However, growing antibiotic resistance is resulting in increased treatment failures. We screened a compound library for those that would reduce Salmonella-induced macrophage toxicity, identifying compound KH-1. KH-1 has no direct effects on the bacteria but limits Salmonella survival in macrophages and protects against lethal infection in a mouse model of typhoid fever. A suboptimal concentration of ciprofloxacin worked in conjunction with the compound to further decrease Salmonella survival in macrophages. An analog (KH-1-2) was identified that possessed increased activity in vitro in macrophages and in vivo against both antibiotic-sensitive and -resistant strains. Thus, we report the identification of a lead compound that may be a useful scaffold as a host-directed antimicrobial against typhoid fever.
Collapse
|
13
|
Zhao C, Chen H, Liang H, Zhao X, Tang W, Wei M, Li Y, Zhang J, Yu X, Chen G, Zhu H, Jiang L, Zhang X. Lactobacillus plantarum RS-09 Induces M1-Type Macrophage Immunity Against Salmonella Typhimurium Challenge via the TLR2/NF-κB Signalling Pathway. Front Pharmacol 2022; 13:832245. [PMID: 35355723 PMCID: PMC8959098 DOI: 10.3389/fphar.2022.832245] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 01/26/2022] [Indexed: 12/30/2022] Open
Abstract
Lactobacillus plantarum can interact with macrophages against bacterial enteropathy due to its potential ability to modulate macrophage polarization. However, this mechanism is not completely understood. TLR2 can recognize microbial components and trigger macrophage cytokine responses to different gram-positive strains. The aim of this study was to investigate whether probiotic Lactobacillus plantarum RS-09 can induce macrophage polarization against Salmonella Typhimurium infection via TLR2 signalling. BALB/c mice were preadministered RS-09 continuously for 7 days and then infected with Salmonella Typhimurium ATCC14028. Mouse RAW264.7 mononuclear macrophages were stimulated with RS-09 and coincubated with ATCC14028 or PBS controls. The results of the in vivo study indicated that RS-09 could relieve S. Typhimurium-induced splenomegaly, body weight loss and death rate. RS-09 also limited the colonization and translocation of S. Typhimurium in the gastrointestinal tract and thereby protected against infection. We also observed that RS-09 upregulated the production of M1 macrophage characteristics (e.g., CD11c and IL-6) against S. Typhimurium. Furthermore, RS-09 induced the expression of TLR2 in macrophages. In an in vitro study, treatment of RAW264.7 cells with RS-09 either concurrently with or before S. Typhimurium challenge enhanced the secretion of Reactive oxygen species and Nitric oxide. This effect was related to TLR2 and NF-κB activation. Based on these findings, Lactobacillus plantarum RS-09 was shown to modulate M1 macrophage polarization and induce TLR2-linked NF-κB signalling activity in the innate immune response to S. Typhimurium infection.
Collapse
Affiliation(s)
- Chenpei Zhao
- School of Life Sciences, Ludong University, Yantai, China
| | - Huan Chen
- School of Life Sciences, Ludong University, Yantai, China
| | - Hao Liang
- Department of Microbiology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xiaoyu Zhao
- School of Life Sciences, Ludong University, Yantai, China
- Shandong Provincial Key Laboratory of Quality Safty Monitoring and Risk Assessment for Animal Products, Jinan, China
| | - Wenli Tang
- Shandong Provincial Key Laboratory of Quality Safty Monitoring and Risk Assessment for Animal Products, Jinan, China
| | - Maolian Wei
- Shandong Provincial Key Laboratory of Quality Safty Monitoring and Risk Assessment for Animal Products, Jinan, China
| | - Youzhi Li
- Shandong Provincial Key Laboratory of Quality Safty Monitoring and Risk Assessment for Animal Products, Jinan, China
| | - Jianlong Zhang
- School of Life Sciences, Ludong University, Yantai, China
- Shandong Provincial Key Laboratory of Quality Safty Monitoring and Risk Assessment for Animal Products, Jinan, China
| | - Xin Yu
- School of Life Sciences, Ludong University, Yantai, China
- Shandong Provincial Key Laboratory of Quality Safty Monitoring and Risk Assessment for Animal Products, Jinan, China
- Yantai Key Laboratory of Animal Pathogenetic Microbiology and Immunology, Yantai, China
| | - Guozhong Chen
- School of Life Sciences, Ludong University, Yantai, China
- Yantai Key Laboratory of Animal Pathogenetic Microbiology and Immunology, Yantai, China
- Shandong Aquaculture Environmental Control Engineering Laboratory, Yantai, China
| | - Hongwei Zhu
- School of Life Sciences, Ludong University, Yantai, China
- Shandong Provincial Key Laboratory of Quality Safty Monitoring and Risk Assessment for Animal Products, Jinan, China
- Shandong Aquaculture Environmental Control Engineering Laboratory, Yantai, China
| | - Linlin Jiang
- School of Life Sciences, Ludong University, Yantai, China
- Shandong Provincial Key Laboratory of Quality Safty Monitoring and Risk Assessment for Animal Products, Jinan, China
- Shandong Aquaculture Environmental Control Engineering Laboratory, Yantai, China
- *Correspondence: Linlin Jiang, ; Xingxiao Zhang,
| | - Xingxiao Zhang
- School of Life Sciences, Ludong University, Yantai, China
- Shandong Aquaculture Environmental Control Engineering Laboratory, Yantai, China
- *Correspondence: Linlin Jiang, ; Xingxiao Zhang,
| |
Collapse
|
14
|
Aryl Hydrocarbon Receptor Activation by Benzo[ a]pyrene Prevents Development of Septic Shock and Fatal Outcome in a Mouse Model of Systemic Salmonella enterica Infection. Cells 2022; 11:cells11040737. [PMID: 35203386 PMCID: PMC8870598 DOI: 10.3390/cells11040737] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 02/11/2022] [Accepted: 02/13/2022] [Indexed: 02/04/2023] Open
Abstract
This study focused on immunomodulatory effects of aryl hydrocarbon receptor (AhR) activation through benzo[a]pyrene (BaP) during systemic bacterial infection. Using a well-established mouse model of systemic Salmonella enterica (S.E.) infection, we studied the influence of BaP on the cellular and humoral immune response and the outcome of disease. BaP exposure significantly reduced mortality, which is mainly caused by septic shock. Surprisingly, the bacterial burden in BaP-exposed surviving mice was significantly higher compared to non-exposed mice. During the early phase of infection (days 1-3 post-infection (p.i.)), the transcription of proinflammatory factors (i.e., IL-12, IFN-γ, TNF-α, IL-1β, IL-6, IL-18) was induced faster under BaP exposure. Moreover, BaP supported the activity of antigen-presenting cells (i.e., CD64 (FcγRI), MHC II, NO radicals, phagocytosis) at the site of infection. However, early in infection, the anti-inflammatory cytokines IL-10 and IL-22 were also locally and systemically upregulated in BaP-exposed S.E.-infected mice. BaP-exposure resulted in long-term persistence of salmonellae up to day 90 p.i., which was accompanied by significantly elevated S.E.-specific antibody responses (i.e., IgG1, IgG2c). In summary, these data suggest that BaP-induced AhR activation is capable of preventing a fatal outcome of systemic S.E. infection, but may result in long-term bacterial persistence, which, in turn, may support the development of chronic inflammation.
Collapse
|
15
|
N-acyl-homoserine lactone produced by Rahnella inusitata isolated from the gut of Galleria mellonella influences Salmonella phenotypes. Braz J Microbiol 2022; 53:819-829. [PMID: 35048318 PMCID: PMC9151966 DOI: 10.1007/s42770-022-00681-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 01/10/2022] [Indexed: 02/01/2023] Open
Abstract
The most studied mechanism of quorum sensing in Gram-negative bacteria is mediated by autoinducer 1 (AI-1), namely, acyl-homoserine lactone (AHL). This system allows communication among different bacterial species and regulates the expression of virulence genes in many pathogens. Although AHL-producing bacteria have been detected in the intestines of humans and other animals, no report was found about AHL-producing bacteria in the insect gut and the possible effects of these autoinducers on enteropathogenic bacteria. Therefore, this study aimed to identify AHL-producing bacteria in the gut of larvae of Galleria mellonella and to evaluate the influence of this quorum sensing signal on the regulation of adhesion and motility phenotypes in the intestinal pathogen Salmonella. Sequencing of the 16S rRNA gene, 16S rRNA gene-based phylogenetic analyses, and phenotypic characterization of gut isolates was performed. The profile of AHLs produced by the isolates was determined using thin-layer chromatography (TLC) and revealed with the biosensor strain Chromobacterium violaceum CV026. Sequencing, phylogenetic analyses and phenotypic characterization of gut isolates showed that the three AHL-producing strains belong to the species Rahnella inusitata, named GM34, GM56, and GM60. The TLC showed that R. inusitata produces a six-carbon AHL. In the presence of cell-free extract of R. inusitata containing AHL and under anaerobic conditions, Salmonella enterica increased the adhesion to stainless steel coupons and presented swarming motility. Extracts from the culture medium of R. inusitata isolates containing AHL increased the adhesion on stainless steel coupons and swarming motility of Salmonella enterica serovar Enteritidis PT4 under anaerobic conditions. The results suggest the possibility of communication between members of the G. mellonella intestinal microbiota with pathogens such as Salmonella.
Collapse
|
16
|
Li J, Claudi B, Fanous J, Chicherova N, Cianfanelli FR, Campbell RAA, Bumann D. Tissue compartmentalization enables Salmonella persistence during chemotherapy. Proc Natl Acad Sci U S A 2021; 118:e2113951118. [PMID: 34911764 PMCID: PMC8713819 DOI: 10.1073/pnas.2113951118] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/11/2021] [Indexed: 12/14/2022] Open
Abstract
Antimicrobial chemotherapy can fail to eradicate the pathogen, even in the absence of antimicrobial resistance. Persisting pathogens can subsequently cause relapsing diseases. In vitro studies suggest various mechanisms of antibiotic persistence, but their in vivo relevance remains unclear because of the difficulty of studying scarce pathogen survivors in complex host tissues. Here, we localized and characterized rare surviving Salmonella in mouse spleen using high-resolution whole-organ tomography. Chemotherapy cleared >99.5% of the Salmonella but was inefficient against a small Salmonella subset in the white pulp. Previous models could not explain these findings: drug exposure was adequate, Salmonella continued to replicate, and host stresses induced only limited Salmonella drug tolerance. Instead, antimicrobial clearance required support of Salmonella-killing neutrophils and monocytes, and the density of such cells was lower in the white pulp than in other spleen compartments containing higher Salmonella loads. Neutrophil densities declined further during treatment in response to receding Salmonella loads, resulting in insufficient support for Salmonella clearance from the white pulp and eradication failure. However, adjunctive therapies sustaining inflammatory support enabled effective clearance. These results identify uneven Salmonella tissue colonization and spatiotemporal inflammation dynamics as main causes of Salmonella persistence and establish a powerful approach to investigate scarce but impactful pathogen subsets in complex host environments.
Collapse
Affiliation(s)
- Jiagui Li
- Biozentrum, University of Basel, CH-4056 Basel, Switzerland
| | | | - Joseph Fanous
- Biozentrum, University of Basel, CH-4056 Basel, Switzerland
| | | | | | | | - Dirk Bumann
- Biozentrum, University of Basel, CH-4056 Basel, Switzerland
| |
Collapse
|
17
|
Dorvigny BM, Tavares LS, de Almeida IA, Santana LN, de Souza Silva E, de Souza JKU, Soares AF, da Silva Júnior VA, Lima-Filho JV. Antiinflammatory and antiinfective effect of caffeine in a mouse model of disseminated salmonellosis. Phytother Res 2021; 36:1652-1663. [PMID: 34910341 DOI: 10.1002/ptr.7349] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 11/25/2021] [Accepted: 11/27/2021] [Indexed: 11/09/2022]
Abstract
Caffeine has been reported for its antiinflammatory properties by stimulating phagocytosis. In this study, we investigated the antiinflammatory and antiinfective potential of caffeine in murine macrophage cell cultures and Swiss mice infected with virulent Salmonella enterica serotype typhimurium. Peritoneal macrophages (pMØ) were treated with caffeine on 96-well plates for 24 hr and then infected with Salmonella for 4 hr. In another experiment, the pMØ were first infected with the bacterium for 4 hr and then treated with caffeine for 24 hr. In addition, Swiss mice were inoculated, intraperitoneally, with S. typhimurium and then received caffeine intravenously. Control groups received phosphate-buffered saline (PBS) or dexamethasone. We found that treatments with caffeine increased the macrophage cell viability and reduced the intracellular bacterial load. The administration of caffeine to Swiss mice reduced the infiltration of leukocytes into the peritoneal cavity after the bacterial challenge. Furthermore, the bacterial burdens in the peritoneal fluid, bloodstream, spleen, and liver were decreased by caffeine treatment. The expression levels of tumor necrosis factor-alpha (TNF-α), interleukin-1β (IL-1β), IL-6, and inducible nitric oxide synthase (iNOs) were down-regulated after infection in caffeine-treated mice. We can conclude that caffeine has both antiinflammatory and antiinfective properties that can be useful for management of bacterial infections along with antibiotics.
Collapse
Affiliation(s)
| | | | | | - Lucas Nunes Santana
- Department of Biology, Federal Rural University of Pernambuco, Recife, Brazil
| | | | | | - Anísio Francisco Soares
- Department of Animal Morphology and Physiology, Federal Rural University of Pernambuco, Recife, Brazil
| | | | | |
Collapse
|
18
|
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
|
19
|
Foster N, Tang Y, Berchieri A, Geng S, Jiao X, Barrow P. Revisiting Persistent Salmonella Infection and the Carrier State: What Do We Know? Pathogens 2021; 10:1299. [PMID: 34684248 PMCID: PMC8537056 DOI: 10.3390/pathogens10101299] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 09/28/2021] [Accepted: 09/29/2021] [Indexed: 11/17/2022] Open
Abstract
One characteristic of the few Salmonella enterica serovars that produce typhoid-like infections is that disease-free persistent infection can occur for months or years in a small number of individuals post-convalescence. The bacteria continue to be shed intermittently which is a key component of the epidemiology of these infections. Persistent chronic infection occurs despite high levels of circulating specific IgG. We have reviewed the information on the basis for persistence in S. Typhi, S. Dublin, S. Gallinarum, S. Pullorum, S. Abortusovis and also S. Typhimurium in mice as a model of persistence. Persistence appears to occur in macrophages in the spleen and liver with shedding either from the gall bladder and gut or the reproductive tract. The involvement of host genetic background in defining persistence is clear from studies with the mouse but less so with human and poultry infections. There is increasing evidence that the organisms (i) modulate the host response away from the typical Th1-type response normally associated with immune clearance of an acute infection to Th2-type or an anti-inflammatory response, and that (ii) the bacteria modulate transformation of macrophage from M1 to M2 type. The bacterial factors involved in this are not yet fully understood. There are early indications that it might be possible to remodulate the response back towards a Th1 response by using cytokine therapy.
Collapse
Affiliation(s)
- Neil Foster
- SRUC Aberdeen Campus, Craibstone Estate, Ferguson Building, Aberdeen AB21 9YA, UK
| | - Ying Tang
- Institute of Molecular Physiology, Shenzhen Bay Laboratory, Shenzhen 518055, China;
| | - Angelo Berchieri
- Departamento de Patologia Veterinária, Faculdade de Ciências Agrárias e Veterinárias, Univ Estadual Paulista, Via de Acesso Paulo Donato Castellane, s/n, 14884-900 Jaboticabal, SP, Brazil;
| | - Shizhong Geng
- Jiangsu Key Laboratory of Zoonosis, Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China; (S.G.); (X.J.)
| | - Xinan Jiao
- Jiangsu Key Laboratory of Zoonosis, Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China; (S.G.); (X.J.)
| | - Paul Barrow
- School of Veterinary Medicine, University of Surrey, Daphne Jackson Road, Guildford GU2 7AL, UK;
| |
Collapse
|
20
|
Prevalence and risk factors for Salmonella spp. contamination of slaughtered chickens in Taiwan. Prev Vet Med 2021; 196:105476. [PMID: 34482151 DOI: 10.1016/j.prevetmed.2021.105476] [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: 03/24/2021] [Revised: 07/07/2021] [Accepted: 08/30/2021] [Indexed: 01/03/2023]
Abstract
The present study was designed to estimate the prevalence of Salmonella contamination in Taiwanese broilers at slaughter and to identify risk factors associated with the presence of Salmonella in processed batches of broilers. Carcass rinse samples from 362 batches of broilers were collected from 45 chicken abattoirs in Taiwan between February 2013 and November 2014. Univariate analyses and multivariable logistic regression analyses were conducted to identify putative risk factors for contamination. Salmonella was detected in 32.6 % (95 % CI: 30.4-34.8) of individual broilers and 56.4 % (95 % CI: 51.1-61.5) of the sampled batches. The multivariable logistic regression model identified season (July to November) (OR = 1.95; 95 % CI: 1.2-3.2) as increasing the risk of infection. Abattoirs in the southern region (Taichung and Kaohsiung) (OR = 0.45; 95 % CI: 0.3-0.8); batches scalded for > 90 s (OR = 0.2; 95 % CI: 0.1-0.3) and batches of commercial white broilers (BR) (OR = 0.21; 95 % CI: 0.1-0.4) all had a decreased risk of contamination compared to abattoirs from the northern region, scalding < 90 s and Taiwan native chickens (TNC), respectively. This study highlights the influence of environmental conditions and poultry breed on the risk of Salmonella contamination of chickens during slaughter.
Collapse
|
21
|
Zhou Y, Ye X, Wang B, Ying J, Zeng Z, Tang L, Wang Q, Zou P, Zhan X, Fu L. Protective Effects of Pidotimod Against Salmonella Infections. Int J Pept Res Ther 2021. [DOI: 10.1007/s10989-021-10186-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
22
|
Post A, Kaboré B, Berendsen M, Diallo S, Traore O, Arts RJW, Netea MG, Joosten LAB, Tinto H, Jacobs J, de Mast Q, van der Ven A. Altered Ex-Vivo Cytokine Responses in Children With Asymptomatic Plasmodium falciparum Infection in Burkina Faso: An Additional Argument to Treat Asymptomatic Malaria? Front Immunol 2021; 12:614817. [PMID: 34177883 PMCID: PMC8220162 DOI: 10.3389/fimmu.2021.614817] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 05/17/2021] [Indexed: 11/30/2022] Open
Abstract
Introduction Patients with clinical malaria have an increased risk for bacterial bloodstream infections. We hypothesized that asymptomatic malaria parasitemia increases susceptibility for bacterial infections through an effect on the innate immune system. We measured circulating cytokine levels and ex-vivo cytokine production capacity in asymptomatic malaria and compared with controls. Methods Data were collected from asymptomatic participants <5 years old with and without positive malaria microscopy, as well as from hospitalized patients <5 years old with clinical malaria, bacteremia, or malaria/bacteremia co-infections in a malaria endemic region of Burkina Faso. Circulating cytokines (TNF-α, IFN-γ, IL-6, IL-10) were measured using multiplex assays. Whole blood from asymptomatic participants with and without positive malaria microscopy were ex-vivo stimulated with S. aureus, E. coli LPS and Salmonella Typhimurium; cytokine concentrations (TNF-α, IFN-γ, IL-1β, IL-6, IL-10) were measured on supernatants using ELISA. Results Included were children with clinical malaria (n=118), bacteremia (n=22), malaria and bacteremia co-infection (n=9), asymptomatic malaria (n=125), and asymptomatic controls (n=237). Children with either clinical or asymptomatic malaria had higher plasma cytokine concentrations than controls. Cytokine concentrations correlated positively with malaria parasite density with the strongest correlation for IL-10 in both asymptomatic (r=0.63) and clinical malaria (r=0.53). Patients with bacteremia had lower circulating IL-10, TNF-α and IFN-γ and higher IL-6 concentrations, compared to clinical malaria. Ex-vivo whole blood cytokine production to LPS and S. aureus was significantly lower in asymptomatic malaria compared to controls. Whole blood IFN-γ and IL-10 production in response to Salmonella was also lower in asymptomatic malaria. Interpretation In children with asymptomatic malaria, cytokine responses upon ex-vivo bacterial stimulation are downregulated. Further studies are needed to explore if the suggested impaired innate immune response to bacterial pathogens also translates into impaired control of pathogens such as Salmonella spp.
Collapse
Affiliation(s)
- Annelies Post
- Department of Internal Medicine, Radboud Centre for Infectious Diseases, Radboud University Medical Centre, Nijmegen, Netherlands
| | - Berenger Kaboré
- Department of Internal Medicine, Radboud Centre for Infectious Diseases, Radboud University Medical Centre, Nijmegen, Netherlands.,IRSS/Clinical Research Unit of Nanoro (CRUN), Nanoro, Burkina Faso
| | - Mike Berendsen
- Department of Internal Medicine, Radboud Centre for Infectious Diseases, Radboud University Medical Centre, Nijmegen, Netherlands.,Bandim Health Project, Institute of Clinical Research, University of Southern Denmark and Odense University Hospital, Odense, Denmark
| | - Salou Diallo
- IRSS/Clinical Research Unit of Nanoro (CRUN), Nanoro, Burkina Faso
| | - Ousmane Traore
- IRSS/Clinical Research Unit of Nanoro (CRUN), Nanoro, Burkina Faso
| | - Rob J W Arts
- Department of Internal Medicine, Radboud Centre for Infectious Diseases, Radboud University Medical Centre, Nijmegen, Netherlands
| | - Mihai G Netea
- Department of Internal Medicine, Radboud Centre for Infectious Diseases, Radboud University Medical Centre, Nijmegen, Netherlands.,Department for Genomics & Immunoregulation, Life and Medical Sciences Institute (LIMES), University of Bonn, Bonn, Germany
| | - Leo A B Joosten
- Department of Internal Medicine, Radboud Centre for Infectious Diseases, Radboud University Medical Centre, Nijmegen, Netherlands
| | - Halidou Tinto
- IRSS/Clinical Research Unit of Nanoro (CRUN), Nanoro, Burkina Faso.,Institut Supérieur des Sciences de la Santé, Université Nazi Boni de Bobo-Dioulasso, Bobo-Dioulasso, Burkina Faso
| | - Jan Jacobs
- Department of Clinical Sciences, Institute of Tropical Medicine, Antwerp, Belgium.,Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
| | - Quirijn de Mast
- Department of Internal Medicine, Radboud Centre for Infectious Diseases, Radboud University Medical Centre, Nijmegen, Netherlands
| | - André van der Ven
- Department of Internal Medicine, Radboud Centre for Infectious Diseases, Radboud University Medical Centre, Nijmegen, Netherlands
| |
Collapse
|
23
|
Duan B, Shao L, Liu R, Msuthwana P, Hu J, Wang C. Lactobacillus rhamnosus GG defense against Salmonella enterica serovar Typhimurium infection through modulation of M1 macrophage polarization. Microb Pathog 2021; 156:104939. [PMID: 33964416 DOI: 10.1016/j.micpath.2021.104939] [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] [Received: 12/26/2020] [Revised: 04/24/2021] [Accepted: 04/26/2021] [Indexed: 01/28/2023]
Abstract
Lactobacillus rhamnosus GG (LGG), a model probiotic strain, plays an important role in immune regulatory activity to prevent and treat intestinal inflammation or diarrhea. However, the effect of the immune modulation of LGG on macrophages to prevent Salmonella infection has not been thoroughly studied. In this study, C57BL/6 mice were pre-administered LGG for 7 days continuously, and then infected with Salmonella enterica subsp. enterica serovar Typhimurium (S. Typhimurium). The results of the in vivo study indicated that LGG could reduce body weight loss, death rate and intestinal inflammatory response caused by S. Typhimurium. LGG also limited S. Typhimurium dissemination to liver and spleen, and thereby protected against infection. In vitro study, we observed that LGG enhanced the phagocytic and bactericidal ability of macrophages and upregulated M1 macrophage characters (e.g. iNOS, NO and IL-12) against S. Typhimurium. In addition, LGG also elevated IL-10 secretion, which was helpful to ameliorate intestinal inflammatory injury caused by S. Typhimurium. In conclusion, LGG could modulate M1 macrophage polarization and offer protective effects against S. Typhimurium infection.
Collapse
Affiliation(s)
- Bingjie Duan
- College of Veterinary Medicine, Jilin Agricultural University, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Changchun, China
| | - Lina Shao
- College of Veterinary Medicine, Jilin Agricultural University, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Changchun, China
| | - Ruihan Liu
- College of Veterinary Medicine, Jilin Agricultural University, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Changchun, China
| | - Petunia Msuthwana
- College of Veterinary Medicine, Jilin Agricultural University, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Changchun, China
| | - Jingtao Hu
- College of Veterinary Medicine, Jilin Agricultural University, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Changchun, China.
| | - Chunfeng Wang
- College of Veterinary Medicine, Jilin Agricultural University, Jilin Provincial Engineering Research Center of Animal Probiotics, Key Laboratory of Animal Production and Product Quality Safety of Ministry of Education, Changchun, China.
| |
Collapse
|
24
|
Praveen C, Bhatia SS, Alaniz RC, Droleskey RE, Cohen ND, Jesudhasan PR, Pillai SD. Assessment of microbiological correlates and immunostimulatory potential of electron beam inactivated metabolically active yet non culturable (MAyNC) Salmonella Typhimurium. PLoS One 2021; 16:e0243417. [PMID: 33861743 PMCID: PMC8051754 DOI: 10.1371/journal.pone.0243417] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Accepted: 11/23/2020] [Indexed: 12/02/2022] Open
Abstract
This study investigates the microbiological and immunological basis underlying the efficacy of electron beam-inactivated immune modulators. The underlying hypothesis is that exposure to eBeam-based ionization reactions inactivate microorganisms without modifying their antigenic properties and thereby creating immune modulators. The immunological correlates of protection induced by such eBeam based Salmonella Typhimurium (EBST) immune modulators in dendritic cell (DC) (in vitro) and mice (in vivo) models were assessed. The EBST stimulated innate pro inflammatory response (TNFα) and maturation (MHC-II, CD40, CD80 and CD86) of DC. Immuno-stimulatory potential of EBST was on par with both a commercial Salmonella vaccine, and live Salmonella cells. The EBST cells did not multiply under permissive in vitro and in vivo conditions. However, EBST cells remained metabolically active. EBST immunized mice developed Salmonella-specific CD4+ T-cells that produced the Th1 cytokine IFNγ at a level similar to that induced by the live attenuated vaccine (AroA- ST) formulation. The EBST retained stable immunogenic properties for several months at room temperature, 4°C, and -20°C as well as after lyophilization. Therefore, such eBeam-based immune modulators have potential as vaccine candidates since they offer the safety of a “killed” vaccine, while retaining the immunogenicity of an “attenuated” vaccine. The ability to store eBeam based immune modulators at room temperature without loss of potency is also noteworthy.
Collapse
Affiliation(s)
- Chandni Praveen
- National Center for Electron Beam Research-an International Atomic Energy Agency (IAEA) Collaborating Centre for Electron Beam Technology, Texas A&M University, College Station, TX, United States of America
| | - Sohini S. Bhatia
- National Center for Electron Beam Research-an International Atomic Energy Agency (IAEA) Collaborating Centre for Electron Beam Technology, Texas A&M University, College Station, TX, United States of America
| | - Robert C. Alaniz
- Department of Microbial Pathogenesis and Immunology, Texas A&M University Health Science Center, College Station, TX, United States of America
- * E-mail: (SDP); (RCA)
| | - Robert E. Droleskey
- Food and Feed Safety Research Unit, Southern Plains Agricultural Research Center, USDA-ARS, College Station, TX, United States of America
| | - Noah D. Cohen
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, United States of America
| | - Palmy R. Jesudhasan
- Poultry Production and Product Safety, USDA-ARS, University of Arkansas, Fayetteville, AR, United States of America
| | - Suresh D. Pillai
- National Center for Electron Beam Research-an International Atomic Energy Agency (IAEA) Collaborating Centre for Electron Beam Technology, Texas A&M University, College Station, TX, United States of America
- * E-mail: (SDP); (RCA)
| |
Collapse
|
25
|
Harrell JE, Hahn MM, D'Souza SJ, Vasicek EM, Sandala JL, Gunn JS, McLachlan JB. Salmonella Biofilm Formation, Chronic Infection, and Immunity Within the Intestine and Hepatobiliary Tract. Front Cell Infect Microbiol 2021; 10:624622. [PMID: 33604308 PMCID: PMC7885405 DOI: 10.3389/fcimb.2020.624622] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Accepted: 12/17/2020] [Indexed: 12/12/2022] Open
Abstract
Within the species of Salmonella enterica, there is significant diversity represented among the numerous subspecies and serovars. Collectively, these account for microbes with variable host ranges, from common plant and animal colonizers to extremely pathogenic and human-specific serovars. Despite these differences, many Salmonella species find commonality in the ability to form biofilms and the ability to cause acute, latent, or chronic disease. The exact outcome of infection depends on many factors such as the growth state of Salmonella, the environmental conditions encountered at the time of infection, as well as the infected host and immune response elicited. Here, we review the numerous biofilm lifestyles of Salmonella (on biotic and abiotic surfaces) and how the production of extracellular polymeric substances not only enhances long-term persistence outside the host but also is an essential function in chronic human infections. Furthermore, careful consideration is made for the events during initial infection that allow for gut transcytosis which, in conjunction with host immune functions, often determine the progression of disease. Both typhoidal and non-typhoidal salmonellae can cause chronic and/or secondary infections, thus the adaptive immune responses to both types of bacteria are discussed with particular attention to the differences between Salmonella Typhi, Salmonella Typhimurium, and invasive non-typhoidal Salmonella that can result in differential immune responses. Finally, while strides have been made in our understanding of immunity to Salmonella in the lymphoid organs, fewer definitive studies exist for intestinal and hepatobiliary immunity. By examining our current knowledge and what remains to be determined, we provide insight into new directions in the field of Salmonella immunity, particularly as it relates to chronic infection.
Collapse
Affiliation(s)
- Jaikin E Harrell
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA, United States
| | - Mark M Hahn
- Center for Microbial Pathogenesis, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, United States.,Infectious Diseases Institute, The Ohio State University, Columbus, OH, United States
| | - Shaina J D'Souza
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA, United States
| | - Erin M Vasicek
- Center for Microbial Pathogenesis, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, United States.,Infectious Diseases Institute, The Ohio State University, Columbus, OH, United States
| | - Jenna L Sandala
- Center for Microbial Pathogenesis, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, United States.,Infectious Diseases Institute, The Ohio State University, Columbus, OH, United States
| | - John S Gunn
- Center for Microbial Pathogenesis, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, United States.,Infectious Diseases Institute, The Ohio State University, Columbus, OH, United States.,Department of Pediatrics, College of Medicine, The Ohio State University, Columbus, OH, United States
| | - James B McLachlan
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA, United States
| |
Collapse
|
26
|
Nikam PS, Kingston JJ, Belagal Motatis AK. Oral co-administration of bivalent protein r-BL with U-Omp19 elicits mucosal immune responses and reduces S. Typhimurium shedding in BALB/c mice. Immunol Lett 2021; 231:61-67. [PMID: 33460704 DOI: 10.1016/j.imlet.2021.01.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Revised: 12/15/2020] [Accepted: 01/11/2021] [Indexed: 01/18/2023]
Abstract
The increase in international food trade and travel has dramatically increased the global incidences of Salmonellosis. In the light of widespread resistance to frontline antibiotics, oral vaccines remain the most reliable alternative. In this study, the fusion protein, r-BL was rationally constructed by splicing the Salmonella Typhimurium sseB and ompL genes through G4S linker by over-lap extension PCR. The oral coadministration of r-BL with B. abortus U-Omp19 protein with known protease inhibitor activity resulted in significant increase of mucosal IgA titres to antilog 4.5051 (p < 0.0001) and 4.806 (p < 0.0001) in the fecal samples and intestinal washes respectively. Antibody isotyping of the intestinal washes demonstrated increase in mucosal IgM, IgG1 and IgG2a isotypes also and demonstrated a significant reduction in fecal shedding of S. Typhimurium in challenge study. The r-BL + U-Omp19 treated mice demonstrated a complete termination of Salmonella fecal shedding by the 12th day of challenge as compared to other study groups. In summary, the bivalent protein r-BL when administered with the mucosal adjuvant U-Omp19 was successful in triggering mucosal arm of the immune system which forms the first line of defence in combating the infections caused by the enteric pathogen like Salmonella.
Collapse
Affiliation(s)
- Pradnya Sukhadev Nikam
- Department of Microbiology, Defence Food Research Laboratory, Siddarthanagar, Mysuru, Karnataka, 570011, India.
| | - Joseph J Kingston
- Department of Microbiology, Defence Food Research Laboratory, Siddarthanagar, Mysuru, Karnataka, 570011, India.
| | - Anil Kumar Belagal Motatis
- Department of Microbiology, Defence Food Research Laboratory, Siddarthanagar, Mysuru, Karnataka, 570011, India.
| |
Collapse
|
27
|
Luiz de Freitas L, Pereira da Silva F, Fernandes KM, Carneiro DG, Licursi de Oliveira L, Martins GF, Dantas Vanetti MC. The virulence of Salmonella Enteritidis in Galleria mellonella is improved by N-dodecanoyl-homoserine lactone. Microb Pathog 2021; 152:104730. [PMID: 33444697 DOI: 10.1016/j.micpath.2021.104730] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 01/01/2021] [Accepted: 01/03/2021] [Indexed: 01/18/2023]
Abstract
Salmonella is a food and waterborne pathogen responsible for outbreaks worldwide, and it can survive during passage through the stomach and inside host phagocytic cells. Virulence genes are required for infection and survival in macrophages, and some are under the regulation of the quorum sensing (QS) system. This study investigated the influence of the autoinducer 1 (AI-1), N-dodecanoyl-homoserine lactone (C12-HSL), on the virulence of Salmonella PT4 using Galleria mellonella as an infection model. Salmonella PT4 was grown in the presence and absence of C12-HSL under anaerobic conditions for 7 h, and the expression of rpoS, arcA, arcB, and invA genes was evaluated. After the inoculation of G. mellonella with the median lethal dose (LD50) of Salmonella PT4, the survival of bacteria inside the larvae and their health status (health index scoring) were monitored, as well as the pigment, nitric oxide (NO), superoxide dismutase (SOD), and catalase (CAT) production. Also, the hemocyte viability, the induction of caspase-3, and microtubule-associated light chain 3 (LC3) protein in hemocytes were evaluated. Salmonella PT4 growing in the presence of C12-HSL showed increased rpoS, arcA, arcB, and invA expression and promoted higher larvae mortality and worse state of health after 24 h of infection. The C12-HSL also increased the persistence of Salmonella PT4 in the hemolymph and in the hemocytes. The highest pigmentation, NO production, and antioxidant enzymes were verified in the larva hemolymph infected with Salmonella PT4 grown with C12-HSL. Hemocytes from larvae infected with Salmonella PT4 grown with C12-HSL showed lower viability and higher production of caspase-3 and LC3. Taken together, these findings suggest that C12-HSL could be involved in the virulence of Salmonella PT4.
Collapse
Affiliation(s)
- Leonardo Luiz de Freitas
- Departmento de Microbiologia, Universidade Federal de Viçosa (UFV), Viçosa, Minas Gerais, Brazil
| | | | - Kenner Morais Fernandes
- Departamento de Biologia Geral, Universidade Federal de Viçosa (UFV), Viçosa, Minas Gerais, Brazil
| | - Deisy Guimarães Carneiro
- Departmento de Microbiologia, Universidade Federal de Viçosa (UFV), Viçosa, Minas Gerais, Brazil
| | | | - Gustavo Ferreira Martins
- Departamento de Biologia Geral, Universidade Federal de Viçosa (UFV), Viçosa, Minas Gerais, Brazil
| | | |
Collapse
|
28
|
Lee HJ, Hong WG, Woo Y, Ahn JH, Ko HJ, Kim H, Moon S, Hahn TW, Jung YM, Song DK, Jung YJ. Lysophosphatidylcholine Enhances Bactericidal Activity by Promoting Phagosome Maturation via the Activation of the NF-κB Pathway during Salmonella Infection in Mouse Macrophages. Mol Cells 2020; 43:989-1001. [PMID: 33250450 PMCID: PMC7772511 DOI: 10.14348/molcells.2020.0030] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 10/29/2020] [Accepted: 11/03/2020] [Indexed: 12/13/2022] Open
Abstract
Salmonella enterica serovar Typhimurium (S. Typhimurium) is a facultative intracellular pathogen that causes salmonellosis and mortality worldwide. S. Typhimurium infects macrophages and survives within phagosomes by avoiding the phagosome-lysosome fusion system. Phagosomes sequentially acquire different Rab GTPases during maturation and eventually fuse with acidic lysosomes. Lysophosphatidylcholine (LPC) is a bioactive lipid that is associated with the generation of chemoattractants and reactive oxygen species (ROS). In our previous study, LPC controlled the intracellular growth of Mycobacterium tuberculosis by promoting phagosome maturation. In this study, to verify whether LPC enhances phagosome maturation and regulates the intracellular growth of S. Typhimurium, macrophages were infected with S. Typhimurium. LPC decreased the intracellular bacterial burden, but it did not induce cytotoxicity in S. Typhimuriuminfected cells. In addition, combined administration of LPC and antibiotic significantly reduced the bacterial burden in the spleen and the liver. The ratios of the colocalization of intracellular S. Typhimurium with phagosome maturation markers, such as early endosome antigen 1 (EEA1) and lysosome-associated membrane protein 1 (LAMP-1), were significantly increased in LPC-treated cells. The expression level of cleaved cathepsin D was rapidly increased in LPCtreated cells during S. Typhimurium infection. Treatment with LPC enhanced ROS production, but it did not affect nitric oxide production in S. Typhimurium-infected cells. LPC also rapidly triggered the phosphorylation of IκBα during S. Typhimurium infection. These results suggest that LPC can improve phagosome maturation via ROS-induced activation of NF-κB pathway and thus may be developed as a therapeutic agent to control S. Typhimurium growth.
Collapse
Affiliation(s)
- Hyo-Ji Lee
- Department of Biological Sciences and Institute of Life Sciences, Kangwon National University, Chuncheon 2434, Korea
- Kangwon Radiation Convergence Research Support Center, Kangwon National University, Chuncheon 24341, Korea
| | - Wan-Gi Hong
- BIT Medical Convergence Graduate Program, Kangwon National University, Chuncheon 4341, Korea
| | - Yunseo Woo
- Department of Biological Sciences and Institute of Life Sciences, Kangwon National University, Chuncheon 2434, Korea
- Kangwon Radiation Convergence Research Support Center, Kangwon National University, Chuncheon 24341, Korea
| | - Jae-Hee Ahn
- Department of Pharmacy, Kangwon National University, Chuncheon 2441, Korea
| | - Hyun-Jeong Ko
- Department of Pharmacy, Kangwon National University, Chuncheon 2441, Korea
- Kangwon Radiation Convergence Research Support Center, Kangwon National University, Chuncheon 24341, Korea
| | - Hyeran Kim
- Department of Biological Sciences and Institute of Life Sciences, Kangwon National University, Chuncheon 2434, Korea
| | - Sungjin Moon
- Department of Biological Sciences and Institute of Life Sciences, Kangwon National University, Chuncheon 2434, Korea
- Kangwon Radiation Convergence Research Support Center, Kangwon National University, Chuncheon 24341, Korea
| | - Tae-Wook Hahn
- Department of Veterinary Medicine, Kangwon National University, Chuncheon 231, Korea
| | - Young Mee Jung
- Department of Chemistry, Kangwon National University, Chuncheon 24341, Korea
- Kangwon Radiation Convergence Research Support Center, Kangwon National University, Chuncheon 24341, Korea
| | - Dong-Keun Song
- Department of Pharmacology, College of Medicine, Hallym University, Chuncheon 24252, Korea
| | - Yu-Jin Jung
- Department of Biological Sciences and Institute of Life Sciences, Kangwon National University, Chuncheon 2434, Korea
- BIT Medical Convergence Graduate Program, Kangwon National University, Chuncheon 4341, Korea
- Kangwon Radiation Convergence Research Support Center, Kangwon National University, Chuncheon 24341, Korea
| |
Collapse
|
29
|
Elsner RA, Shlomchik MJ. IL-12 Blocks Tfh Cell Differentiation during Salmonella Infection, thereby Contributing to Germinal Center Suppression. Cell Rep 2020; 29:2796-2809.e5. [PMID: 31775046 DOI: 10.1016/j.celrep.2019.10.069] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 08/21/2019] [Accepted: 10/16/2019] [Indexed: 12/13/2022] Open
Abstract
Germinal centers (GC) are crucial for the formation of long-lived humoral immunity. Many pathogens suppress GC, including Salmonella enterica serovar Typhimurium (STm), but the mechanisms driving suppression remain unknown. We report that neither plasmablasts nor STm-specific B cells are required for GC suppression in mice. Rather, we identify that interleukin-12 (IL-12), but not interferon-γ (IFN-γ), directly suppresses T follicular helper (Tfh) cell differentiation of T cells intrinsically. Administering recombinant IL-12 during nitrophenyl-Chicken Gamma Globulin (NP-CGG) immunization also suppresses Tfh cell differentiation and GC B cells, indicating that IL-12 is sufficient to suppress Tfh cell differentiation independent of STm infection. Recombinant IL-12 induces high levels of T-bet, and T-bet is necessary for Tfh cell suppression. Therefore, IL-12 induced during STm infection in mice contributes to GC suppression via suppression of Tfh cell differentiation. More broadly, these data suggest that IL-12 can tailor the proportions of humoral (Tfh cell) and cellular (T helper type 1 [Th1] cell) immunity to the infection, with implications for IL-12 targeting therapies in autoimmunity and vaccination.
Collapse
Affiliation(s)
- Rebecca A Elsner
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15216, USA
| | - Mark J Shlomchik
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15216, USA.
| |
Collapse
|
30
|
Salmonella enterica serovar Typhi exposure elicits ex vivo cell-type-specific epigenetic changes in human gut cells. Sci Rep 2020; 10:13581. [PMID: 32788681 PMCID: PMC7423951 DOI: 10.1038/s41598-020-70492-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 06/22/2020] [Indexed: 01/25/2023] Open
Abstract
Salmonella enterica serovar Typhi (S. Typhi) causes substantial morbidity and mortality worldwide, particularly among young children. Humans develop an array of mucosal immune responses following S. Typhi infection. Whereas the cellular mechanisms involved in S. Typhi infection have been intensively studied, very little is known about the early chromatin modifications occurring in the human gut microenvironment that influence downstream immune responses. To address this gap in knowledge, cells isolated from human terminal ileum exposed ex vivo to the wild-type S. Typhi strain were stained with a 33-metal-labeled antibody panel for mass cytometry analyses of the early chromatin modifications modulated by S. Typhi. We measured the cellular levels of 6 classes of histone modifications, and 1 histone variant in 11 major cell subsets (i.e., B, CD3 + T, CD4 + T, CD8 + T, NK, TCR-γδ, Mucosal associated invariant (MAIT), and NKT cells as well as monocytes, macrophages, and epithelial cells). We found that arginine methylation might regulate the early-differentiation of effector-memory CD4+ T-cells following exposure to S. Typhi. We also found S. Typhi-induced post-translational modifications in histone methylation and acetylation associated with epithelial cells, NKT, MAIT, TCR-γδ, Monocytes, and CD8 + T-cells that are related to both gene activation and silencing.
Collapse
|
31
|
Piccini G, Montomoli E. Pathogenic signature of invasive non-typhoidal Salmonella in Africa: implications for vaccine development. Hum Vaccin Immunother 2020; 16:2056-2071. [PMID: 32692622 PMCID: PMC7553687 DOI: 10.1080/21645515.2020.1785791] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
Invasive non-typhoidal Salmonella (iNTS) infections are a leading cause of bacteremia in Sub-Saharan Africa (sSA), thereby representing a major public health threat. Salmonella Typhimurium clade ST313 and Salmonella Enteriditis lineages associated with Western and Central/Eastern Africa are among the iNTS serovars which are of the greatest concern due to their case-fatality rate, especially in children and in the immunocompromised population. Identification of pathogen-associated features and host susceptibility factors that increase the risk for invasive non-typhoidal salmonellosis would be instrumental for the design of targeted prevention strategies, which are urgently needed given the increasing spread of multidrug-resistant iNTS in Africa. This review summarizes current knowledge of bacterial traits and host immune responses associated with iNTS infections in sSA, then discusses how this knowledge can guide vaccine development while providing a summary of vaccine candidates in preclinical and early clinical development.
Collapse
Affiliation(s)
| | - Emanuele Montomoli
- VisMederi srl , Siena, Italy.,Department of Molecular and Developmental Medicine, University of Siena , Siena, Italy
| |
Collapse
|
32
|
Esmailnia E, Amani J, Gargari SLM. Identification of novel vaccine candidate against Salmonella enterica serovar Typhi by reverse vaccinology method and evaluation of its immunization. Genomics 2020; 112:3374-3381. [PMID: 32565239 DOI: 10.1016/j.ygeno.2020.06.022] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 05/14/2020] [Accepted: 06/10/2020] [Indexed: 11/17/2022]
Abstract
Salmonella enterica serovar Typhi (S. Typhi) is an essential enteric fever causing bacterium worldwide. Due to the emergence of multidrug-resistant strains, urgently attention is needed to prevent the global spread of them. Vaccination is an alternative approach to control these kinds of infections. Currently available commercial vaccines have significant limitations such as non-recommendation for children below six years of age and poor long-term efficacy. Thus, the development of a new vaccine overcoming these limitations is immediately required. Reverse Vaccinology (RV) is one of the most robust approaches for direct screening of genome sequence assemblies to identify new protein-based vaccines. The present study aimed to identify potential vaccine candidates against S. Typhi by using the RV approach. Immunogenicity of the best candidate against S. Typhi was further investigated. The proteome of S. Typhi strain Ty2 was analyzed to identify the most immunogenic, conserved, and protective surface proteins. Among the predicted vaccine candidates, steD (fimbrial subunit) was the best for qualifying all the applied criteria. The synthetic steD gene was expressed in E.coli, and the mice were immunized with purified recombinant steD protein and then challenged with a lethal dose of S. Typhi. Immunized animals generated high protein-specific antibody titers and demonstrated 70% survival following lethal dose challenge with S. Typhi. Pretreatment of the S. Typhi cells with immunized mice antisera significantly decreased their adhesion to Caco-2 cells. Altogether, steD as a protective antigen could induce a robust and long term and protective immunity in immunized mice against S. Typhi challenge.
Collapse
Affiliation(s)
- Ehsan Esmailnia
- Department of Biology, Faculty of Basic Sciences, Shahed University, Tehran, Iran
| | - Jafar Amani
- Applied Microbiology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | | |
Collapse
|
33
|
Jneid B, Rouaix A, Féraudet-Tarisse C, Simon S. SipD and IpaD induce a cross-protection against Shigella and Salmonella infections. PLoS Negl Trop Dis 2020; 14:e0008326. [PMID: 32463817 PMCID: PMC7282677 DOI: 10.1371/journal.pntd.0008326] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 06/09/2020] [Accepted: 04/26/2020] [Indexed: 01/05/2023] Open
Abstract
Salmonella and Shigella species are food- and water-borne pathogens that are responsible for enteric infections in both humans and animals and are still the major cause of morbidity and mortality in the emerging countries. The existence of multiple Salmonella and Shigella serotypes as well as the emergence of strains resistant to antibiotics require the development of broadly protective therapies. Those bacteria utilize a Type III Secretion System (T3SS), necessary for their pathogenicity. The structural proteins composing the T3SS are common to all virulent Salmonella and Shigella spp., particularly the needle-tip proteins SipD (Salmonella) and IpaD (Shigella). We investigated the immunogenicity and protective efficacy of SipD and IpaD administered by intranasal and intragastric routes, in a mouse model of Salmonella enterica serotype Typhimurium (S. Typhimurium) intestinal challenge. Robust IgG (in all immunization routes) and IgA (in intranasal and oral immunization routes) antibody responses were induced against both proteins. Mice immunized with SipD or IpaD were protected against lethal intestinal challenge with S. Typhimurium or Shigella flexneri (100 Lethal Dose 50%). We have shown that SipD and IpaD are able to induce a cross-protection in a murine model of infection by Salmonella and Shigella. We provide the first demonstration that Salmonella and Shigella T3SS SipD and IpaD are promising antigens for the development of a cross-protective Salmonella-Shigella vaccine. These results open the way to the development of cross-protective therapeutic molecules.
Collapse
Affiliation(s)
- Bakhos Jneid
- Université Paris Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), SPI, Gif-sur-Yvette, France
| | - Audrey Rouaix
- Université Paris Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), SPI, Gif-sur-Yvette, France
| | - Cécile Féraudet-Tarisse
- Université Paris Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), SPI, Gif-sur-Yvette, France
| | - Stéphanie Simon
- Université Paris Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS), SPI, Gif-sur-Yvette, France
- * E-mail:
| |
Collapse
|
34
|
A Loss-of-Function Mutation in the Integrin Alpha L ( Itgal) Gene Contributes to Susceptibility to Salmonella enterica Serovar Typhimurium Infection in Collaborative Cross Strain CC042. Infect Immun 2019; 88:IAI.00656-19. [PMID: 31636138 DOI: 10.1128/iai.00656-19] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 10/05/2019] [Indexed: 12/18/2022] Open
Abstract
Salmonella is an intracellular bacterium found in the gastrointestinal tract of mammalian, avian, and reptilian hosts. Mouse models have been extensively used to model in vivo distinct aspects of human Salmonella infections and have led to the identification of several host susceptibility genes. We have investigated the susceptibility of Collaborative Cross strains to intravenous infection with Salmonella enterica serovar Typhimurium as a model of human systemic invasive infection. In this model, strain CC042/GeniUnc (CC042) mice displayed extreme susceptibility with very high bacterial loads and mortality. CC042 mice showed lower spleen weights and decreased splenocyte numbers before and after infection, affecting mostly CD8+ T cells, B cells, and all myeloid cell populations, compared with control C57BL/6J mice. CC042 mice also had lower thymus weights with a reduced total number of thymocytes and double-negative and double-positive (CD4+, CD8+) thymocytes compared to C57BL/6J mice. Analysis of bone marrow-resident hematopoietic progenitors showed a strong bias against lymphoid-primed multipotent progenitors. An F2 cross between CC042 and C57BL/6N mice identified two loci on chromosome 7 (Stsl6 and Stsl7) associated with differences in bacterial loads. In the Stsl7 region, CC042 carried a loss-of-function variant, unique to this strain, in the integrin alpha L (Itgal) gene, the causative role of which was confirmed by a quantitative complementation test. Notably, Itgal loss of function increased the susceptibility to S. Typhimurium in a (C57BL/6J × CC042)F1 mouse background but not in a C57BL/6J mouse inbred background. These results further emphasize the utility of the Collaborative Cross to identify new host genetic variants controlling susceptibility to infections and improve our understanding of the function of the Itgal gene.
Collapse
|
35
|
Cerny O, Holden DW. Salmonella SPI-2 type III secretion system-dependent inhibition of antigen presentation and T cell function. Immunol Lett 2019; 215:35-39. [PMID: 30771380 DOI: 10.1016/j.imlet.2019.01.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 01/09/2019] [Accepted: 01/10/2019] [Indexed: 12/17/2022]
Abstract
Salmonella enterica serovars infect a broad range of mammalian hosts, including humans, causing both gastrointestinal and systemic diseases. Effective immune responses to Salmonella infections depend largely on CD4+ T cell activation by dendritic cells (DCs). Bacteria are internalised by intestinal DCs and respond by translocating effectors of the Salmonella pathogenicity island 2 (SPI-2) type III secretion system (T3SS) into host cells. In this review, we discuss processes that are hijacked by SPI-2 T3SS effectors and how this affects DC biology and the activation of T cell responses.
Collapse
Affiliation(s)
- Ondrej Cerny
- MRC Centre for Molecular Bacteriology and Infection, Imperial College London, SW7 2AZ, UK
| | - David W Holden
- MRC Centre for Molecular Bacteriology and Infection, Imperial College London, SW7 2AZ, UK.
| |
Collapse
|
36
|
Unexpected Role of CD8 T Cells in Accelerated Clearance of Salmonella enterica Serovar Typhimurium from H-2 Congenic mice. Infect Immun 2019; 87:IAI.00588-19. [PMID: 31427450 DOI: 10.1128/iai.00588-19] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 07/31/2019] [Indexed: 12/21/2022] Open
Abstract
Salmonella infection can cause gastroenteritis in healthy individuals or a serious, systemic infection in immunocompromised patients and has a global impact. CD4 Th1 cells represent the main lymphocyte population that participates in bacterial clearance during both primary and secondary infections in mice of the H-2b haplotype. Previous studies have used congenic mice to examine the function of major histocompatibility complex (MHC) molecules in elimination of this pathogen from the host. In this study, we further characterized the ability of H-2b, H-2k, and H-2u molecules to influence adaptive immunity to Salmonella in MHC congenic mice. By depleting different cell populations during infection, we unexpectedly found that CD8 T cells, in addition to CD4 T cells, play a major role in accelerated clearance of bacteria from H-2k congenic hosts. Our data suggest that CD8 T cells accelerate clearance in some MHC congenic mouse strains and could therefore represent an unexpected contributor to the protective efficacy of Salmonella vaccines outside the typical studies in C57BL/6 mice.
Collapse
|
37
|
Mu H, Bai H, Sun F, Liu Y, Lu C, Qiu Y, Chen P, Yang Y, Kong L, Duan J. Pathogen-targeting glycovesicles as a therapy for salmonellosis. Nat Commun 2019; 10:4039. [PMID: 31492864 PMCID: PMC6731243 DOI: 10.1038/s41467-019-12066-z] [Citation(s) in RCA: 20] [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: 12/15/2018] [Accepted: 08/16/2019] [Indexed: 12/16/2022] Open
Abstract
Antibiotic therapy is usually not recommended for salmonellosis, as it is associated with prolonged fecal carriage without reducing symptom duration or severity. Here we show that antibiotics encapsulated in hydrogen sulfide (H2S)-responsive glycovesicles may be potentially useful for the treatment of salmonellosis. The antibiotics are released in the presence of Salmonella, which is known to produce H2S. This approach prevents the quick absorption of antibiotics into the bloodstream, allows localized targeting of the pathogen in the gut, and alleviates disease symptoms in a mouse infection model. In addition, it reduces antibiotic-induced changes in the gut microbiota, and increases the abundance of potentially beneficial lactobacilli due to the release of prebiotic xylooligosaccharide analogs.
Collapse
Affiliation(s)
- Haibo Mu
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, College of Chemistry & Pharmacy, Northwest A&F University, 712100, Yangling, Shaanxi, China
| | - Hu Bai
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, College of Chemistry & Pharmacy, Northwest A&F University, 712100, Yangling, Shaanxi, China
| | - Feifei Sun
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, College of Chemistry & Pharmacy, Northwest A&F University, 712100, Yangling, Shaanxi, China
| | - Yinyin Liu
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, College of Chemistry & Pharmacy, Northwest A&F University, 712100, Yangling, Shaanxi, China
| | - Chunbo Lu
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, College of Chemistry & Pharmacy, Northwest A&F University, 712100, Yangling, Shaanxi, China
| | - Yuanhao Qiu
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, College of Chemistry & Pharmacy, Northwest A&F University, 712100, Yangling, Shaanxi, China
| | - Peng Chen
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, College of Chemistry & Pharmacy, Northwest A&F University, 712100, Yangling, Shaanxi, China
| | - Yu Yang
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, College of Chemistry & Pharmacy, Northwest A&F University, 712100, Yangling, Shaanxi, China
| | - Lili Kong
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, College of Chemistry & Pharmacy, Northwest A&F University, 712100, Yangling, Shaanxi, China
| | - Jinyou Duan
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, College of Chemistry & Pharmacy, Northwest A&F University, 712100, Yangling, Shaanxi, China.
| |
Collapse
|
38
|
Bailleul G, Guabiraba R, Virlogeux-Payant I, Lantier I, Trotereau J, Gilbert FB, Wiedemann A, Trotereau A, Velge P, Schouler C, Lalmanach AC. Systemic Administration of Avian Defensin 7: Distribution, Cellular Target, and Antibacterial Potential in Mice. Front Microbiol 2019; 10:541. [PMID: 30972041 PMCID: PMC6444188 DOI: 10.3389/fmicb.2019.00541] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Accepted: 03/01/2019] [Indexed: 12/20/2022] Open
Abstract
Defensins are natural antimicrobial peptides. The avian beta-defensin AvBD7 isolated from the chicken bone marrow possess broad antibacterial spectrum and strong resistance to proteolysis. However, its ability to fight systemic infections of major concern for public health, such as salmonellosis, is unknown. As a first approach, fluorescence labeling of AvBD7 allowed to track its systemic distribution after intraperitoneal injection in mice using whole body live imaging. It was associated to peritoneal cells and to deeper organs such as the liver. In the next step, the use of labeled AvBD7 allowed to observe its interaction with murine macrophages in culture. After incubation, it was able to penetrate inside the cells through an endocytosis-like mechanism. Furthermore, natural AvBD7 contributed to the control of intracellular multiplication of a multidrug resistant Salmonella strain, after incubation with infected macrophages. Finally, administration in a model of systemic lethal Salmonella infection in mice led to significant improvement of mouse survival, consistently with significant reduction of the liver bacterial load. In conclusion, the results reveal a hitherto unknown intracellular antibacterial effect of AvBD7 in Salmonella target cells and support AvBD7 as a candidate of interest for the treatment of infectious diseases caused by multidrug-resistant pathogenic Enterobacteriaceae.
Collapse
Affiliation(s)
- Geoffrey Bailleul
- ISP, INRA, Université de Tours, UMR 1282 Centre INRA Val de Loire, Nouzilly, France
| | - Rodrigo Guabiraba
- ISP, INRA, Université de Tours, UMR 1282 Centre INRA Val de Loire, Nouzilly, France
| | | | - Isabelle Lantier
- ISP, INRA, Université de Tours, UMR 1282 Centre INRA Val de Loire, Nouzilly, France
| | - Jérôme Trotereau
- ISP, INRA, Université de Tours, UMR 1282 Centre INRA Val de Loire, Nouzilly, France
| | - Florence B Gilbert
- ISP, INRA, Université de Tours, UMR 1282 Centre INRA Val de Loire, Nouzilly, France
| | - Agnès Wiedemann
- ISP, INRA, Université de Tours, UMR 1282 Centre INRA Val de Loire, Nouzilly, France
| | - Angélina Trotereau
- ISP, INRA, Université de Tours, UMR 1282 Centre INRA Val de Loire, Nouzilly, France
| | - Philippe Velge
- ISP, INRA, Université de Tours, UMR 1282 Centre INRA Val de Loire, Nouzilly, France
| | - Catherine Schouler
- ISP, INRA, Université de Tours, UMR 1282 Centre INRA Val de Loire, Nouzilly, France
| | | |
Collapse
|
39
|
FBXO7 sensitivity of phenotypic traits elucidated by a hypomorphic allele. PLoS One 2019; 14:e0212481. [PMID: 30840666 PMCID: PMC6402633 DOI: 10.1371/journal.pone.0212481] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 02/04/2019] [Indexed: 11/19/2022] Open
Abstract
FBXO7 encodes an F box containing protein that interacts with multiple partners to facilitate numerous cellular processes and has a canonical role as part of an SCF E3 ubiquitin ligase complex. Mutation of FBXO7 is responsible for an early onset Parkinsonian pyramidal syndrome and genome-wide association studies have linked variants in FBXO7 to erythroid traits. A putative orthologue in Drosophila, nutcracker, has been shown to regulate the proteasome, and deficiency of nutcracker results in male infertility. Therefore, we reasoned that modulating Fbxo7 levels in a murine model could provide insights into the role of this protein in mammals. We used a targeted gene trap model which retained 4-16% residual gene expression and assessed the sensitivity of phenotypic traits to gene dosage. Fbxo7 hypomorphs showed regenerative anaemia associated with a shorter erythrocyte half-life, and male mice were infertile. Alterations to T cell phenotypes were also observed, which intriguingly were both T cell intrinsic and extrinsic. Hypomorphic mice were also sensitive to infection with Salmonella, succumbing to a normally sublethal challenge. Despite these phenotypes, Fbxo7 hypomorphs were produced at a normal Mendelian ratio with a normal lifespan and no evidence of neurological symptoms. These data suggest that erythrocyte survival, T cell development and spermatogenesis are particularly sensitive to Fbxo7 gene dosage.
Collapse
|
40
|
Wahid R, Kotloff KL, Levine MM, Sztein MB. Cell mediated immune responses elicited in volunteers following immunization with candidate live oral Salmonella enterica serovar Paratyphi A attenuated vaccine strain CVD 1902. Clin Immunol 2019; 201:61-69. [PMID: 30849494 DOI: 10.1016/j.clim.2019.03.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 02/22/2019] [Accepted: 03/03/2019] [Indexed: 01/01/2023]
Abstract
The incidence of Salmonella enterica serovar Paratyphi A (PA) infection is on the rise and no licensed vaccines are available. We evaluated cell mediated immune (CMI) responses elicited in volunteers following immunization with a single dose (109 or 1010 cfu) of a novel attenuated live oral PA-vaccine strain (CVD 1902). Results showed increases in PA-lipopolysaccharide-specific IgG- and/or IgA B-memory cells and production of IFN-γ, TNF-α, IL-10, IL-23 and RANTES following stimulation with PA-antigens by peripheral blood mononuclear cells obtained 28 days post immunization. Flow cytometry assays revealed that vaccine elicited PA-specific CD8+ and/or CD4+ T effector/memory cells were predominantly multifunctional concomitantly expressing CD107a and/or producing IFN-γ, TNF-α and/or IL-2. Similar proportions of these MF cells expressed, or not, the gut homing marker integrin α4β7. The results suggest that immunization with CVD 1902 elicits CMI responses against PA supporting its further evaluation as a potential vaccine candidate against paratyphoid A fever.
Collapse
Affiliation(s)
- Rezwanul Wahid
- Department of Pediatrics, Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA.
| | - Karen L Kotloff
- Department of Pediatrics, Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA; Department of Medicine, Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Myron M Levine
- Department of Pediatrics, Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA; Department of Medicine, Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Marcelo B Sztein
- Department of Pediatrics, Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA; Department of Medicine, Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
41
|
Bumann D. Salmonella Single-Cell Metabolism and Stress Responses in Complex Host Tissues. Microbiol Spectr 2019; 7:10.1128/microbiolspec.bai-0009-2019. [PMID: 30953427 PMCID: PMC11588158 DOI: 10.1128/microbiolspec.bai-0009-2019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Indexed: 01/12/2023] Open
Abstract
Systemic Salmonella enterica infections are a major cause of mortality worldwide and are becoming increasingly untreatable. Recent single-cell data from a mouse model of typhoid fever show that the host immune system actually eradicates many Salmonella cells, while other Salmonella organisms thrive at the same time in the same tissue, causing lethal disease progression. The surviving Salmonella cells have highly heterogeneous metabolism, growth rates, and exposure to various stresses. Emerging evidence suggests that similarly heterogeneous host-pathogen encounters might be a key feature of many infectious diseases. This heterogeneity offers fascinating opportunities for research and application. If we understand the mechanisms that determine the disparate local outcomes, we might be able to develop entirely novel strategies for infection control by broadening successful host antimicrobial attacks and closing permissive niches in which pathogens can thrive. This review describes suitable technologies, a current working model of heterogeneous host-Salmonella interactions, the impact of diverse Salmonella subsets on antimicrobial chemotherapy, and major open questions and challenges.
Collapse
Affiliation(s)
- Dirk Bumann
- Focal Area Infection Biology, Biozentrum, University of Basel, Basel, Switzerland
| |
Collapse
|
42
|
Fu A, Mo Q, Wu Y, Wang B, Liu R, Tang L, Zeng Z, Zhang X, Li W. Protective effect of Bacillus amyloliquefaciens against Salmonella via polarizing macrophages to M1 phenotype directly and to M2 depended on microbiota. Food Funct 2019; 10:7653-7666. [DOI: 10.1039/c9fo01651a] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Bacillus amyloliquefaciens SC06 (BaSC06), a potential probiotic, plays a positive role in animal growth performance and immune function.
Collapse
Affiliation(s)
- Aikun Fu
- Key Laboratory of Animal Molecular Nutrition of Education of Ministry
- College of Animal Sciences
- Zhejiang University
- Hangzhou
- China
| | - Qiufen Mo
- Key Laboratory of Animal Molecular Nutrition of Education of Ministry
- College of Animal Sciences
- Zhejiang University
- Hangzhou
- China
| | - Yanping Wu
- Key Laboratory of Animal Molecular Nutrition of Education of Ministry
- College of Animal Sciences
- Zhejiang University
- Hangzhou
- China
| | - Baikui Wang
- Key Laboratory of Animal Molecular Nutrition of Education of Ministry
- College of Animal Sciences
- Zhejiang University
- Hangzhou
- China
| | - Rongrong Liu
- Key Laboratory of Animal Molecular Nutrition of Education of Ministry
- College of Animal Sciences
- Zhejiang University
- Hangzhou
- China
| | - Li Tang
- Key Laboratory of Animal Molecular Nutrition of Education of Ministry
- College of Animal Sciences
- Zhejiang University
- Hangzhou
- China
| | - Zhonghua Zeng
- Key Laboratory of Animal Molecular Nutrition of Education of Ministry
- College of Animal Sciences
- Zhejiang University
- Hangzhou
- China
| | - Xiaoping Zhang
- China National Bamboo Research Center
- Key Laboratory of Resources and Utilization of Bamboo of State Forestry Administration
- Hangzhou
- China
| | - Weifen Li
- Key Laboratory of Animal Molecular Nutrition of Education of Ministry
- College of Animal Sciences
- Zhejiang University
- Hangzhou
- China
| |
Collapse
|
43
|
Mooney JP, Galloway LJ, Riley EM. Malaria, anemia, and invasive bacterial disease: A neutrophil problem? J Leukoc Biol 2018; 105:645-655. [PMID: 30570786 PMCID: PMC6487965 DOI: 10.1002/jlb.3ri1018-400r] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 11/29/2018] [Accepted: 12/01/2018] [Indexed: 12/16/2022] Open
Abstract
Invasive bacterial disease is well described in immunocompromised hosts, including those with malaria infection. One bacterial infection frequently observed in children with Plasmodium falciparum infection is nontyphoidal salmonella (NTS) infection, in which a typically intestinal infection becomes systemic with serious, often fatal, consequences. In this review, we consider the role of malaria‐induced immunoregulatory responses in tipping the balance from tissue homeostasis during malaria infection to risk of invasive NTS. Also, neutrophils are crucial in the clearance of NTS but their ability to mount an oxidative burst and kill intracellular Salmonella is severely compromised during, and for some time after, an acute malaria infection. Here, we summarize the evidence linking malaria and invasive NTS infections; describe the role of neutrophils in clearing NTS infections; review evidence for neutrophil dysfunction in malaria infections; and explore roles of heme oxygenase‐1, IL‐10, and complement in mediating this dysfunction. Finally, given the epidemiological evidence that low density, subclinical malaria infections pose a risk for invasive NTS infections, we consider whether the high prevalence of such infections might underlie the very high incidence of invasive bacterial disease across much of sub‐Saharan Africa.
Collapse
Affiliation(s)
- Jason P Mooney
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian, United Kingdom
| | - Lauren J Galloway
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian, United Kingdom
| | - Eleanor M Riley
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian, United Kingdom
| |
Collapse
|
44
|
Schramm G, Suwandi A, Galeev A, Sharma S, Braun J, Claes AK, Braubach P, Grassl GA. Schistosome Eggs Impair Protective Th1/Th17 Immune Responses Against Salmonella Infection. Front Immunol 2018; 9:2614. [PMID: 30487793 PMCID: PMC6246638 DOI: 10.3389/fimmu.2018.02614] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 10/24/2018] [Indexed: 01/19/2023] Open
Abstract
Countries with a high incidence of helminth infections are characterized by high morbidity and mortality to infections with intracellular pathogens such as Salmonella. Some patients with Salmonella-Schistosoma co-infections develop a so-called “chronic septicemic salmonellosis,” with prolonged fever and enlargement of the liver and spleen. These effects are most likely due to the overall immunoregulatory activities of schistosomes such as induction of Tregs, Bregs, alternatively activated macrophages, and degradation of antibodies. However, detailed underlying mechanisms are not very well investigated. Here, we show that intraperitoneal application of live Schistosoma mansoni eggs prior to infection with Salmonella Typhimurium in mice leads to an impairment of IFN-γ and IL-17 responses together with a higher bacterial load compared to Salmonella infection alone. S. mansoni eggs were found in granulomas in the visceral peritoneum attached to the colon. Immunohistological staining revealed IPSE/alpha-1, a glycoprotein secreted from live schistosome eggs, and recruited basophils around the eggs. Noteworthy, IPSE/alpha-1 is known to trigger IL-4 and IL-13 release from basophils which in turn is known to suppress Th1/Th17 responses. Therefore, our data support a mechanism of how schistosomes impair a protective immune response against Salmonella infection and increase our understanding of helminth-bacterial co-infections.
Collapse
Affiliation(s)
- Gabriele Schramm
- Experimental Pneumology, Research Center Borstel, Airway Research Center North, Member of the German Center for Lung Research (DZL), Borstel, Germany
| | - Abdulhadi Suwandi
- Institute of Medical Microbiology and Hospital Epidemiology and German Center for Infection Research (DZIF), Partner Site Hannover, Hannover Medical School, Hannover, Germany
| | - Alibek Galeev
- Institute of Medical Microbiology and Hospital Epidemiology and German Center for Infection Research (DZIF), Partner Site Hannover, Hannover Medical School, Hannover, Germany
| | - Samriti Sharma
- Institute of Medical Microbiology and Hospital Epidemiology and German Center for Infection Research (DZIF), Partner Site Hannover, Hannover Medical School, Hannover, Germany
| | - Janin Braun
- Institute for Experimental Medicine, Christian-Albrechts-University of Kiel, Kiel, Germany.,Research Center Borstel, Borstel, Germany
| | - Anne-Kathrin Claes
- Institute for Experimental Medicine, Christian-Albrechts-University of Kiel, Kiel, Germany.,Research Center Borstel, Borstel, Germany
| | - Peter Braubach
- Institute for Pathology, Hannover Medical School, Hannover, Germany
| | - Guntram A Grassl
- Institute of Medical Microbiology and Hospital Epidemiology and German Center for Infection Research (DZIF), Partner Site Hannover, Hannover Medical School, Hannover, Germany.,Institute for Experimental Medicine, Christian-Albrechts-University of Kiel, Kiel, Germany.,Research Center Borstel, Borstel, Germany
| |
Collapse
|
45
|
Zhen W, Shao Y, Gong X, Wu Y, Geng Y, Wang Z, Guo Y. Effect of dietary Bacillus coagulans supplementation on growth performance and immune responses of broiler chickens challenged by Salmonella enteritidis. Poult Sci 2018; 97:2654-2666. [PMID: 29660095 DOI: 10.3382/ps/pey119] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Accepted: 03/12/2018] [Indexed: 12/27/2022] Open
Abstract
This study was conducted to evaluate the protective efficacy of dietary Bacillus coagulans (B. coagulans) supplementation in birds receiving Salmonella enteritidis (SE). Two hundred and forty 1-day-old Cobb broilers were randomly assigned to 2 × 2 factorial arrangements of treatments with 2 levels of dietary B. coagulans (0 or 400 mg/kg) and 2 levels of SE challenge (0 or 1 × 109 SE between d 9 to 11). Results showed that SE infection did not affect growth performance, but caused intestinal inflammation and barrier function impairment by reducing intestinal goblet cells and beneficial bacteria numbers, increasing cecal Salmonella colonization and liver Salmonella invasion, downregulating jejunal mucin-2 (at 7 and 17 d post-infection, DPI), TLR2 (at 7 and 17 DPI), TLR4 (at 17 DPI), TNFSF15 (at 7 and 17 DPI) gene mRNA levels, and upregulating jejunal IFN-γ mRNA levels (at 17 DPI) compared to uninfected birds. Moreover, SE infection also elevated the concentration of jejunal anti-Salmonella IgA and sera anti-Salmonella IgG compared to uninfected birds. However, chickens received B. coagulans diets showed significant increase in body weight gain and weight gain to feed intake ratio from d 15 to 21, alkaline phosphatase activity (at 7 DPI), cecal Lactobacilli and Bifidobacterium numbers (at 7 DPI; at 17 DPI), villous height: crypt ratio (at 17 DPI), and goblet cell numbers (at 7 and 17 DPI), whereas exhibiting reduced jejunal crypt depth (at 17 DPI), cecal Escherichia coli (at 7, 17, and 31 DPI), and Salmonella (at 7 and 17 DPI) levels compared with the non-supplemented birds, regardless of SE infection. In addition, B. coagulans supplement upregulated lysozyme mRNA levels (at 17 DPI), downregulated IFN-γ mRNA levels (at 7 and 17 DPI), showed an increased trend in Fowlicidin-2 mRNA levels (at 7 DPI) and a reduced trend in liver Salmonella load compared to the non-supplemented control. These data indicated that B. coagulans has a protective effect in SE infected broilers.
Collapse
Affiliation(s)
- Wenrui Zhen
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Yujing Shao
- College of Biology, China Agricultural University, Beijing, China
| | - Xiuyan Gong
- School of Agriculture, Ningxia University, Yinchuan, China
| | - Yuanyuan Wu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Yanqiang Geng
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Zhong Wang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Yuming Guo
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| |
Collapse
|
46
|
Optimal protection against Salmonella infection requires noncirculating memory. Proc Natl Acad Sci U S A 2018; 115:10416-10421. [PMID: 30254173 DOI: 10.1073/pnas.1808339115] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
While CD4 Th1 cells are required for resistance to intramacrophage infections, adoptive transfer of Th1 cells is insufficient to protect against Salmonella infection. Using an epitope-tagged vaccine strain of Salmonella, we found that effective protection correlated with expanded Salmonella-specific memory CD4 T cells in circulation and nonlymphoid tissues. However, naive mice that previously shared a blood supply with vaccinated partners lacked T cell memory with characteristics of tissue residence and did not acquire robust protective immunity. Using a YFP-IFN-γ reporter system, we identified Th1 cells in the liver of immunized mice that displayed markers of tissue residence, including P2X7, ARTC2, LFA-1, and CD101. Adoptive transfer of liver memory cells after ARTC2 blockade increased protection against highly virulent bacteria. Taken together, these data demonstrate that noncirculating memory Th1 cells are a vital component of immunity to Salmonella infection and should be the focus of vaccine strategies.
Collapse
|
47
|
Oliveira J, Costa K, Acurcio L, Sandes S, Cassali G, Uetanabaro A, Costa A, Nicoli J, Neumann E, Porto A. In vitro and in vivo evaluation of two potential probiotic lactobacilli isolated from cocoa fermentation (Theobroma cacao L.). J Funct Foods 2018. [DOI: 10.1016/j.jff.2018.05.055] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
48
|
Gupta V, Kaur M, Datta P, Chander J. Chronic urinary carrier state due to Salmonella Typhi causing urinary tract infection in an immunocompetent healthy woman. Trop Doct 2018; 48:236-238. [DOI: 10.1177/0049475517752205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Enteric fever caused by Salmonella Typhi is a global public health problem. With adequate treatment, most patients recover from the acute phase; however, 2–4% develop a chronic carrier state acting as reservoir of infection by continued shedding of bacteria in faeces and urine. Recovery of S. Typhi from urine is rare, even in endemic areas. The three main causes of bacteriuria arise following a recent episode of typhoid fever, in chronic carrier states involving the urinary system and occasionally following localised urinary tract infection (UTI) due to S. Typhi. Symptomatic Salmonella UTI is mostly encountered in an immunocompromised patient with some underlying structural abnormality involving the urinary tract. We report a case of symptomatic UTI caused by Salmonella Typhi in a 50-year-old immunocompetent woman in a chronic carrier state without any known urological abnormality.
Collapse
Affiliation(s)
- Varsha Gupta
- Professor, Department of Microbiology, Government Medical College Hospital, Chandigarh, India
| | - Mandeep Kaur
- Demonstrator, Department of Microbiology, Government Medical College Hospital, Chandigarh, India
| | - Priya Datta
- Assistant Professor, Department of Microbiology, Government Medical College Hospital, Chandigarh, India
| | - Jagdish Chander
- Professor and Head, Department of Microbiology, Government Medical College Hospital, Chandigarh, India
| |
Collapse
|
49
|
Alturki NA, McComb S, Ariana A, Rijal D, Korneluk RG, Sun SC, Alnemri E, Sad S. Triad3a induces the degradation of early necrosome to limit RipK1-dependent cytokine production and necroptosis. Cell Death Dis 2018; 9:592. [PMID: 29789521 PMCID: PMC5964080 DOI: 10.1038/s41419-018-0672-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Revised: 04/24/2018] [Accepted: 04/27/2018] [Indexed: 12/31/2022]
Abstract
Understanding the molecular signaling in programmed cell death is vital to a practical understanding of inflammation and immune cell function. Here we identify a previously unrecognized mechanism that functions to downregulate the necrosome, a central signaling complex involved in inflammation and necroptosis. We show that RipK1 associates with RipK3 in an early necrosome, independent of RipK3 phosphorylation and MLKL-induced necroptotic death. We find that formation of the early necrosome activates K48-ubiquitin-dependent proteasomal degradation of RipK1, Caspase-8, and other necrosomal proteins. Our results reveal that the E3-ubiquitin ligase Triad3a promotes this negative feedback loop independently of typical RipK1 ubiquitin editing enzymes, cIAPs, A20, or CYLD. Finally, we show that Triad3a-dependent necrosomal degradation limits necroptosis and production of inflammatory cytokines. These results reveal a new mechanism of shutting off necrosome signaling and may pave the way to new strategies for therapeutic manipulation of inflammatory responses.
Collapse
Affiliation(s)
- Norah A Alturki
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Canada.,Applied Medical science, King Saud University, Riyadh, Saudi Arabia
| | - Scott McComb
- Human Health and Therapeutics, National Research Council of Canada, Ottawa, Canada
| | - Ardeshir Ariana
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Canada
| | - Dikchha Rijal
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Canada
| | - Robert G Korneluk
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Canada.,Children's Hospital of Eastern Ontario Research Institute, Ontario, Canada
| | - Shao-Cong Sun
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Emad Alnemri
- Thomas Jefferson University, Philadelphia, PA, USA
| | - Subash Sad
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Canada. .,Centre for Infection, Immunity and Inflammation, University of Ottawa, Ontario, Canada.
| |
Collapse
|
50
|
Yang Y, Tellez G, Latorre JD, Ray PM, Hernandez X, Hargis BM, Ricke SC, Kwon YM. Salmonella Excludes Salmonella in Poultry: Confirming an Old Paradigm Using Conventional and Barcode-Tagging Approaches. Front Vet Sci 2018; 5:101. [PMID: 29868621 PMCID: PMC5964308 DOI: 10.3389/fvets.2018.00101] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 04/23/2018] [Indexed: 12/16/2022] Open
Abstract
Salmonella is one of the major foodborne bacterial pathogens, and the consumption of contaminated chicken meats isa primary route of Salmonella transmission into human food chains. However, the mechanism of Salmonella transmission within the chicken flock is not fully understood, including competition among Salmonella strains during chicken infection. The purpose of the present study was to evaluate the competitive exclusion (CE) between different or same Salmonella species consecutively challenged through the oral route. Two different approaches were used to evaluate the CE effect, including tracking Salmonella colonization by wild-type strains with difference in natural antibiotic resistance or DNA barcode-tagged isogenic strains. When day-of-hatch chicks were administered by wild-type S. Typhimurium (ST) on day 1, followed by infection on day 2 by S. Enteritidis (SE) or vice versa, most of the birds were colonized only by the first strains administered (82% by ST or 83% by SE). When similar experiments were performed using two different isogenic barcode-tagged SE strains, Illumina sequencing analysis of the barcode region showed that the first barcode-tagged strains administered were dominant strains, ranging from 92 to 99% of the Salmonella recovered from ceca. These results provide quantitative evidence supporting the CE theory that oral administration of Salmonella will produce predominant inhibition over the subsequent colonization of ceca by the following administration one day later by different or same Salmonella species. We also showed that the use of barcode-tagged isogenic strains in combination with deep profiling of barcodes by Illumina sequencing can serve as a quantitative method for studying complex dynamics of Salmonella infection, transmission and colonization in poultry.
Collapse
Affiliation(s)
- Yichao Yang
- Department of Poultry Science, University of Arkansas, Fayetteville, AR , United States
| | - Guillermo Tellez
- Department of Poultry Science, University of Arkansas, Fayetteville, AR , United States
| | - Juan D Latorre
- Department of Poultry Science, University of Arkansas, Fayetteville, AR , United States
| | - Pamela M Ray
- Department of Veterinary Pathobiology, Texas A&M University, College Station, TX, United States
| | - Xochitl Hernandez
- Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Billy M Hargis
- Department of Poultry Science, University of Arkansas, Fayetteville, AR , United States.,Cell and Molecular Biology Program, University of Arkansas, Fayetteville, AR, United States
| | - Steven C Ricke
- Cell and Molecular Biology Program, University of Arkansas, Fayetteville, AR, United States.,Center of Food Safety, Department of Food Science, University of Arkansas, Fayetteville, AR, United States, 72704, USA
| | - Young Min Kwon
- Department of Poultry Science, University of Arkansas, Fayetteville, AR , United States.,Cell and Molecular Biology Program, University of Arkansas, Fayetteville, AR, United States
| |
Collapse
|