1
|
Charity OJ, Thilliez G, Al-Khanaq H, Acton L, Kolenda R, Bawn M, Petrovska L, Kingsley RA. Reversible excision of the wzy locus in Salmonella Typhimurium may aid recovery following phage predation. PLoS Genet 2025; 21:e1011688. [PMID: 40315401 DOI: 10.1371/journal.pgen.1011688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 04/11/2025] [Indexed: 05/04/2025] Open
Abstract
Bacteriophage (phage) are promising novel antimicrobials but a key challenge to their effective implementation is the rapid emergence of phage resistance. An improved understanding of phage-host interactions is therefore needed. The Anderson phage typing scheme differentiates closely related strains of Salmonella enterica serovar Typhimurium (S. Typhimurium) based on sensitivity to a panel of phage preparations. Switches in phage type are indicative of changes in phage sensitivity and inform on the dynamics of phage interaction with their host bacteria. We investigated the molecular basis of switches between the relatively phage sensitive S. Typhimurium DT8 and phage resistant DT30 strains that are present in the same phylogenetic clade. DT30 strains emerged from DT8 strains predominantly by deletion of a genomic region affecting the wzy locus encoding an O-antigen polymerase. The deletion site was flanked by two perfect direct repeats designated attL and attR. During broth culture in the presence of a typing phage that used O-antigen as primary receptor the Dwzy genotype increased in frequency compared with culture in the absence of phage and removal of attL prevented deletion of the wzy locus. Co-culture of S. Typhimurium DT8 with a strain lacking wzy resulted in reversion of the latter to wild type. We propose a model in which reversible deletion of the wzy locus enables recovery of S. Typhimurium DT8 following predation by phage that use O-antigen as their primary receptor. This was consistent with ancestral state reconstruction of DT8 and DT30 phylogeny that supported a model of reversible transition from DT8 to DT30 in natural populations.
Collapse
Affiliation(s)
- Oliver Jd Charity
- Quadram Institute Bioscience, Norwich, United Kingdom
- University of East Anglia, Norwich Research Park, Norwich, United Kingdom
| | - Gaetan Thilliez
- Quadram Institute Bioscience, Norwich, United Kingdom
- School of Biotechnology, Dublin City University, Dublin, Ireland
| | | | - Luke Acton
- Quadram Institute Bioscience, Norwich, United Kingdom
- University of East Anglia, Norwich Research Park, Norwich, United Kingdom
| | - Rafał Kolenda
- Quadram Institute Bioscience, Norwich, United Kingdom
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Wrocław University of Environmental and Life Sciences, Wrocław, Poland
| | - Matt Bawn
- Quadram Institute Bioscience, Norwich, United Kingdom
- Earlham Institute, Norwich, United Kingdom
- School of Natural and Environmental Sciences, Newcastle University, Newcastle, United Kingdom
| | - Liljana Petrovska
- Animal & Plant Health Agency (APHA), Weybridge, London, United Kingdom
| | - Robert A Kingsley
- Quadram Institute Bioscience, Norwich, United Kingdom
- University of East Anglia, Norwich Research Park, Norwich, United Kingdom
| |
Collapse
|
2
|
Mcdonald JB, Gan E, Cain J, Thoduka SG, Lee J, Wade B, Mauri M, Cuccui J, Wren BW, Packer NH, Londrigan SL, Fritzlar S, Mohotti S, Underwood GJ, Andrews DM, Van TTH, Moore RJ. Immunological and pathobiological characteristics of a novel live Salmonella Typhimurium-vectored Campylobacter vaccine candidate for layer chickens. Front Vet Sci 2025; 12:1518231. [PMID: 40191081 PMCID: PMC11969459 DOI: 10.3389/fvets.2025.1518231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Accepted: 01/03/2025] [Indexed: 04/09/2025] Open
Abstract
Introduction Spotty liver disease (SLD) poses a significant economic and animal welfare threat to the global cage-free egg industry, primarily due to infection by the emerging pathogen Campylobacter hepaticus. SLD can lead to a significant decline in egg production and increased mortality rates. Antibiotics remain the most effective measure for controlling the disease. However, the rise of antibiotic resistance is a growing global concern for public health, promoting efforts to reduce antibiotic usage in animal production. Poultry vaccination offers an alternative approach to decreasing C. hepaticus levels. Although autogenous vaccines are in use in some countries with limited efficacy, no vaccine is currently licensed for widespread use. Methods This study developed and characterized a live Salmonella Typhimurium vector strain designed to deliver the conserved Campylobacter N-glycan heptasaccharide as a target antigen against C. hepaticus. Results The replacement of the S. Typhimurium aroA gene with the Campylobacter pgl locus attenuated the vaccine strain, allowing the conjugation of the heptasaccharide to S. Typhimurium endogenous lipopolysaccharide (LPS). Commercial layer hens vaccinated with the S. Typhimurium strain producing the Campylobacter heptasaccharide induced significantly higher IgY antibody titres specific to the Campylobacter heptasaccharide compared to the birds vaccinated with the vector strain not expressing the heptasaccharide. Modification of the S. Typhimurium endogenous LPS with the heptasaccharide had no significant impact on IgY antibody responses against S. Typhimurium. Discussion This study provides evidence that using S. Typhimurium to deliver Campylobacter heptasaccharide is a feasible approach to providing bi-valent immunogenicity against both S. Typhimurium and C. hepaticus.
Collapse
Affiliation(s)
| | - Emily Gan
- Bioproperties Pty Ltd, RMIT University, Bundoora, VIC, Australia
| | - Joel Cain
- School of Natural Sciences, Macquarie University, Sydney, NSW, Australia
- ARC Centre of Excellence in Synthetic Biology, Macquarie University, Sydney, NSW, Australia
| | - Sapna G. Thoduka
- Bioproperties Pty Ltd, RMIT University, Bundoora, VIC, Australia
| | - Joseph Lee
- School of Science, RMIT University, Bundoora, VIC, Australia
| | - Ben Wade
- School of Science, RMIT University, Bundoora, VIC, Australia
| | - Marta Mauri
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Jon Cuccui
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Brendan W. Wren
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Nicolle H. Packer
- School of Natural Sciences, Macquarie University, Sydney, NSW, Australia
- ARC Centre of Excellence in Synthetic Biology, Macquarie University, Sydney, NSW, Australia
| | - Sarah L. Londrigan
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Svenja Fritzlar
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Sameera Mohotti
- Bioproperties Pty Ltd, RMIT University, Bundoora, VIC, Australia
| | | | | | - Thi Thu Hao Van
- School of Science, RMIT University, Bundoora, VIC, Australia
| | - Robert J. Moore
- School of Science, RMIT University, Bundoora, VIC, Australia
| |
Collapse
|
3
|
Santamaria de Souza N, Cherrak Y, Andersen TB, Vetsch M, Barthel M, Kroon S, Bakkeren E, Schubert C, Christen P, Kiefer P, Vorholt JA, Nguyen BD, Hardt WD. Context-dependent change in the fitness effect of (in)organic phosphate antiporter glpT during Salmonella Typhimurium infection. Nat Commun 2025; 16:1912. [PMID: 39994176 PMCID: PMC11850910 DOI: 10.1038/s41467-025-56851-5] [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: 06/05/2024] [Accepted: 02/04/2025] [Indexed: 02/26/2025] Open
Abstract
Salmonella enterica is a frequent cause of foodborne diseases, which is attributed to its adaptability. Even within a single host, expressing a gene can be beneficial in certain infection stages but neutral or even detrimental in others as previously shown for flagellins. Mutants deficient for the conserved glycerol-3-phosphate and phosphate antiporter glpT have been shown to be positively selected in nature, clinical, and laboratory settings. This suggests that different selective pressures select for the presence or absence of GlpT in a context dependent fashion, a phenomenon known as antagonistic pleiotropy. Using mutant libraries and reporters, we investigated the fitness of glpT-deficient mutants during murine orogastric infection. While glpT-deficient mutants thrive during initial growth in the gut lumen, where GlpT's capacity to import phosphate is disadvantageous, they are counter-selected by macrophages. The dichotomy showcases the need to study the spatial and temporal heterogeneity of enteric pathogens' fitness across distinct lifestyles and niches. Insights into the differential adaptation during infection may reveal opportunities for therapeutic interventions.
Collapse
Affiliation(s)
| | - Yassine Cherrak
- Department of Biology, Institute of Microbiology, ETH Zürich, Zürich, Switzerland
| | - Thea Bill Andersen
- Department of Biology, Institute of Microbiology, ETH Zürich, Zürich, Switzerland
| | - Michel Vetsch
- Department of Biology, Institute of Microbiology, ETH Zürich, Zürich, Switzerland
| | - Manja Barthel
- Department of Biology, Institute of Microbiology, ETH Zürich, Zürich, Switzerland
| | - Sanne Kroon
- Department of Biology, Institute of Microbiology, ETH Zürich, Zürich, Switzerland
| | - Erik Bakkeren
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Christopher Schubert
- Department of Biology, Institute of Microbiology, ETH Zürich, Zürich, Switzerland
| | - Philipp Christen
- Department of Biology, Institute of Microbiology, ETH Zürich, Zürich, Switzerland
| | - Patrick Kiefer
- Department of Biology, Institute of Microbiology, ETH Zürich, Zürich, Switzerland
| | - Julia A Vorholt
- Department of Biology, Institute of Microbiology, ETH Zürich, Zürich, Switzerland
| | - Bidong D Nguyen
- Department of Biology, Institute of Microbiology, ETH Zürich, Zürich, Switzerland
| | - Wolf-Dietrich Hardt
- Department of Biology, Institute of Microbiology, ETH Zürich, Zürich, Switzerland.
| |
Collapse
|
4
|
Kitchens SR, Wang C, Price SB. Bridging Classical Methodologies in Salmonella Investigation with Modern Technologies: A Comprehensive Review. Microorganisms 2024; 12:2249. [PMID: 39597638 PMCID: PMC11596670 DOI: 10.3390/microorganisms12112249] [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: 09/30/2024] [Revised: 11/01/2024] [Accepted: 11/03/2024] [Indexed: 11/29/2024] Open
Abstract
Advancements in genomics and machine learning have significantly enhanced the study of Salmonella epidemiology. Whole-genome sequencing has revolutionized bacterial genomics, allowing for detailed analysis of genetic variation and aiding in outbreak investigations and source tracking. Short-read sequencing technologies, such as those provided by Illumina, have been instrumental in generating draft genomes that facilitate serotyping and the detection of antimicrobial resistance. Long-read sequencing technologies, including those from Pacific Biosciences and Oxford Nanopore Technologies, offer the potential for more complete genome assemblies and better insights into genetic diversity. In addition to these sequencing approaches, machine learning techniques like decision trees and random forests provide powerful tools for pattern recognition and predictive modeling. Importantly, the study of bacteriophages, which interact with Salmonella, offers additional layers of understanding. Phages can impact Salmonella population dynamics and evolution, and their integration into Salmonella genomics research holds promise for novel insights into pathogen control and epidemiology. This review revisits the history of Salmonella and its pathogenesis and highlights the integration of these modern methodologies in advancing our understanding of Salmonella.
Collapse
Affiliation(s)
| | | | - Stuart B. Price
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, 1130 Wire Road, Auburn, AL 36849-5519, USA; (S.R.K.); (C.W.)
| |
Collapse
|
5
|
Devi MB, Bhattacharya A, Kumar A, Singh CT, Das S, Sarma HK, Mukherjee AK, Khan MR. Potential probiotic Lactiplantibacillus plantarum strains alleviate TNF-α by regulating ADAM17 protein and ameliorate gut integrity through tight junction protein expression in in vitro model. Cell Commun Signal 2024; 22:520. [PMID: 39468700 PMCID: PMC11514838 DOI: 10.1186/s12964-024-01900-7] [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: 08/01/2024] [Accepted: 10/19/2024] [Indexed: 10/30/2024] Open
Abstract
BACKGROUND Lactiplantibacillus species are extensively studied for their ability to regulate host immune responses and functional therapeutic potentials. Nevertheless, there is a lack of understanding on the mechanisms of interactions with the hosts during immunoregulatory activities. METHODS Two Lactiplantibacillus plantarum strains MKMB01 and MKMB02 were tested for probiotic potential following Indian Council of Medical Research (ICMR) guidelines. Human colorectal adenocarcinoma cells such as HT-29, caco-2, and human monocytic cell THP-1 were also used to study the potential of MKMB01 and MKMB02 in regulating the host immune response when challenged with enteric pathogen Salmonella enterica typhimurium. Cells were pre-treated with MKMB01 and MKMB02 for 4 h and then stimulated with Salmonella. qRT-PCR and ELISA were used to analyze the genes and protein expression. Confocal microscopy and field emission scanning electron microscopy (FESEM) were used to visualize the effects. An Agilent Seahorse XF analyzer was used to determine real-time mitochondrial functioning. RESULTS Both probiotic strains could defend against Salmonella by maintaining gut integrity via expressing tight junction proteins (TJPs), MUC-2, and toll-like receptors (TLRs) negative regulators such as single Ig IL-1-related receptor (SIGIRR), toll-interacting protein (Tollip), interleukin-1 receptor-associated kinase (IRAK)-M, A20, and anti-inflammatory transforming growth factor-β and interleukin-10. Both strains also downregulated the expression of pro-inflammatory cytokines/chemokines interleukin-1β, monocyte chemoattractant protein (MCP)-1, tumor necrosis factor-alpha (TNF-α), interleukin 6, and nitric oxide (NO). Moreover, TNF-α sheddase protein, a disintegrin and metalloproteinase domain 17 (ADAM17), and its regulator iRhom2 were downregulated by both strains. Moreover, the bacteria also ameliorated Salmonella-induced mitochondrial dysfunction by restoring bioenergetic profiles, such as non-mitochondrial respiration, spare respiratory capacity (SRC), basal respiration, adenosine triphosphate (ATP) production, and maximal respiration. CONCLUSIONS MKMB01 and MKMB02 can reduce pathogen-induced gut-associated disorders and therefore should be further explored for their probiotic potential.
Collapse
Affiliation(s)
- M Bidyarani Devi
- Division of Life Sciences, Institute of Advanced Study in Science and Technology (IASST), Guwahati, 781035, Assam, India
- Department of Biotechnology, Gauhati University, Guwahati, 781014, Assam, India
| | - Anupam Bhattacharya
- Division of Life Sciences, Institute of Advanced Study in Science and Technology (IASST), Guwahati, 781035, Assam, India
| | - Arun Kumar
- Division of Life Sciences, Institute of Advanced Study in Science and Technology (IASST), Guwahati, 781035, Assam, India
| | - Chingtham Thanil Singh
- Division of Life Sciences, Institute of Advanced Study in Science and Technology (IASST), Guwahati, 781035, Assam, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Santanu Das
- Division of Life Sciences, Institute of Advanced Study in Science and Technology (IASST), Guwahati, 781035, Assam, India
| | - Hridip Kumar Sarma
- Department of Biotechnology, Gauhati University, Guwahati, 781014, Assam, India
| | - Ashis K Mukherjee
- Division of Life Sciences, Institute of Advanced Study in Science and Technology (IASST), Guwahati, 781035, Assam, India
- Microbial Biotechnology and Protein Research laboratory, Department of Molecular Biology and Biotechnology, Tezpur University, Tezpur, 784028, Assam, India
| | - Mojibur R Khan
- Division of Life Sciences, Institute of Advanced Study in Science and Technology (IASST), Guwahati, 781035, Assam, India.
| |
Collapse
|
6
|
Aganja RP, Sivasankar C, Lee JH. AI-2 quorum sensing controlled delivery of cytolysin-A by tryptophan auxotrophic low-endotoxic Salmonella and its anticancer effects in CT26 mice with colon cancer. J Adv Res 2024; 61:83-100. [PMID: 37689243 PMCID: PMC11258660 DOI: 10.1016/j.jare.2023.09.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 08/16/2023] [Accepted: 09/04/2023] [Indexed: 09/11/2023] Open
Abstract
INTRODUCTION The limitations of conventional cancer therapies necessitate target-oriented, highly invasive, and safe treatment approaches. Hence, the intrinsic anti-tumor activity of Salmonella can offer better options to combat cancers. OBJECTIVES This study aims to utilize attenuated Salmonella and deliver cytolytic protein cytolysin A (ClyA) under quorum sensing (QS) signaling for precise localized expression in tumors but not in healthy organs. METHODS The therapeutic delivery strain was imposed with tryptophan auxotroph for selective colonization in tumors by trpA and trpE deletion, and lipid-A and O-antigen were altered by pagL and rfaL deletions using lambda red recombination method. The strain was transformed with the designed QS-controlled ClyA expression vector which was validated by western blot. The in vivo passaged therapeutic strain was used for treatment four times at a weekly interval, with a dose of 5 × 106 CFU/mouse for cancer therapy. RESULTS The attenuated strain induced minimal endotoxicity-related cytokines TNF-α, IL-1β, and IFN-γ and exhibited adequate colonization despite earlier exposure in mice. The QS-controlled ClyA expression was confirmed by western blot from bacterial cultures grown at different cell densities. The results demonstrated that the in vivo passaged strain preferentially colonized the tumor after vacating the spleen, liver, and lung, leaving no outward histological scars. The anti-cancer effect of the designed construct was evaluated in the murine CT26 colon cancer model. The expression of ClyA increased tumoricidal activity by 67 % compared to vector control. CONCLUSION Hence, the anti-tumor effect of the engineered Salmonella strain was improved by ClyA expression via QS activation after achieving the threshold bacterial cell density. Further, immunohistochemical staining of the tumor and other organs corroborated the QS-controlled tumor-specific expression of ClyA. Overall, the results imply that the developed anti-cancer Salmonella has low endotoxicity and QS-controlled expression of ClyA as beneficial safety elements and supports recurrent Salmonella inoculation by O-antigen deficiency.
Collapse
Affiliation(s)
- Ram Prasad Aganja
- Department of Veterinary Public Health, College of Veterinary Medicine, Jeonbuk National University, Iksan 54596, South Korea
| | - Chandran Sivasankar
- Department of Veterinary Public Health, College of Veterinary Medicine, Jeonbuk National University, Iksan 54596, South Korea
| | - John Hwa Lee
- Department of Veterinary Public Health, College of Veterinary Medicine, Jeonbuk National University, Iksan 54596, South Korea.
| |
Collapse
|
7
|
Liu Q, Ding J, Zhang X, Bian X, Li M, Chen J, Liu C, Chen X, Liu X, Chen Y, Zhang W, Lei M, Yuan H, Wen Y, Kong Q. Construction and characterization of Aeromonas hydrophila crp and fur deletion mutants and evaluation of its potential as live-attenuated vaccines in crucian carp. FISH & SHELLFISH IMMUNOLOGY 2024; 146:109380. [PMID: 38244821 DOI: 10.1016/j.fsi.2024.109380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 01/06/2024] [Accepted: 01/14/2024] [Indexed: 01/22/2024]
Abstract
Aeromonas hydrophila (A. hydrophila) is a typical zoonotic pathogenic bacterium that infects humans, animals, and fish. It has been reported that the Fur, a Fe2+ regulatory protein, and the Crp, a cAMP receptor protein, play important roles in bacterial virulence in many bacteria, but no research has been investigated on A. hydrophila. In this study, the Δfur and Δcrp mutant strains were constructed by the suicide plasmid method. These two mutant strains exhibited a slightly diminished bacterial growth and also were observed some alterations in the number of outer membrane proteins, and the disappearance of hemolysis in the Δcrp strain. Animal experiments of crucian carp showed that the Δfur and Δcrp mutant strains significantly decreased virulence compared to the wild-type strain, and both mutant strains were able to induce good immune responses by two kinds of administration routes of intraperitoneal immunization (i.p) and immersion immunization, and the protection rates through intraperitoneal injection of Δfur and Δcrp to crucian carp were as high as 83.3 % and 73.3 %, respectively, and immersion immunization route of Δfur and Δcrp to crucian carp provided protection as high as 40 % and 20 %, respectively. These two mutant strains showed abilities to induce changes in enzymatic activities of the non-specific enzymes SOD, LZM, AKP, and ACP in crucian carp. Together, these results indicated the Δfur and Δcrp mutants were safe and effective candidate vaccine strains, showing good protection against the wild-type A. hydrophila challenge.
Collapse
Affiliation(s)
- Qing Liu
- College of Veterinary Medicine, Southwest University, Chongqing, China; Yibin Academy of Southwest University, Yibin, China.
| | - Jianjun Ding
- College of Veterinary Medicine, Southwest University, Chongqing, China; Yibin Academy of Southwest University, Yibin, China
| | - Xiaofen Zhang
- College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Xiaoping Bian
- College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Mengru Li
- College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Jin Chen
- College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Chengying Liu
- College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Xin Chen
- College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Xinyu Liu
- College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Yaolin Chen
- College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Wenjin Zhang
- College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Meihong Lei
- College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Haoxiang Yuan
- College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Yusong Wen
- College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Qingke Kong
- College of Veterinary Medicine, Southwest University, Chongqing, China.
| |
Collapse
|
8
|
Zhu S, Bradfield CJ, Maminska A, Park ES, Kim BH, Kumar P, Huang S, Kim M, Zhang Y, Bewersdorf J, MacMicking JD. Native architecture of a human GBP1 defense complex for cell-autonomous immunity to infection. Science 2024; 383:eabm9903. [PMID: 38422126 DOI: 10.1126/science.abm9903] [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: 10/27/2021] [Accepted: 01/17/2024] [Indexed: 03/02/2024]
Abstract
All living organisms deploy cell-autonomous defenses to combat infection. In plants and animals, large supramolecular complexes often activate immune proteins for protection. In this work, we resolved the native structure of a massive host-defense complex that polymerizes 30,000 guanylate-binding proteins (GBPs) over the surface of gram-negative bacteria inside human cells. Construction of this giant nanomachine took several minutes and remained stable for hours, required guanosine triphosphate hydrolysis, and recruited four GBPs plus caspase-4 and Gasdermin D as a cytokine and cell death immune signaling platform. Cryo-electron tomography suggests that GBP1 can adopt an extended conformation for bacterial membrane insertion to establish this platform, triggering lipopolysaccharide release that activated coassembled caspase-4. Our "open conformer" model provides a dynamic view into how the human GBP1 defense complex mobilizes innate immunity to infection.
Collapse
Affiliation(s)
- Shiwei Zhu
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT 06510, USA
- Yale Systems Biology Institute, West Haven, CT 06477, USA
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT 06510, USA
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Clinton J Bradfield
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT 06510, USA
- Yale Systems Biology Institute, West Haven, CT 06477, USA
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT 06510, USA
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Agnieszka Maminska
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT 06510, USA
- Yale Systems Biology Institute, West Haven, CT 06477, USA
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT 06510, USA
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Eui-Soon Park
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT 06510, USA
- Yale Systems Biology Institute, West Haven, CT 06477, USA
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT 06510, USA
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Bae-Hoon Kim
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT 06510, USA
- Yale Systems Biology Institute, West Haven, CT 06477, USA
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT 06510, USA
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Pradeep Kumar
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT 06510, USA
- Yale Systems Biology Institute, West Haven, CT 06477, USA
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT 06510, USA
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Shuai Huang
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT 06510, USA
- Yale Systems Biology Institute, West Haven, CT 06477, USA
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT 06510, USA
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Minjeong Kim
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT 06510, USA
- Yale Systems Biology Institute, West Haven, CT 06477, USA
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT 06510, USA
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Yongdeng Zhang
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Joerg Bewersdorf
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06510, USA
- Yale Nanobiology Institute, West Haven, CT 06477, USA
| | - John D MacMicking
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT 06510, USA
- Yale Systems Biology Institute, West Haven, CT 06477, USA
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT 06510, USA
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06510, USA
| |
Collapse
|
9
|
Senevirathne A, Hewawaduge C, Lee JH. Assessment of environmental safety and protective efficacy of O-antigen deficient DIVA capable Salmonella Enteritidis against chicken salmonellosis. Poult Sci 2024; 103:103354. [PMID: 38154449 PMCID: PMC10788308 DOI: 10.1016/j.psj.2023.103354] [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: 08/25/2023] [Revised: 10/12/2023] [Accepted: 12/01/2023] [Indexed: 12/30/2023] Open
Abstract
In this study, we incorporated deletion of the O-antigen ligase gene to an attenuated Salmonella Enteritidis (SE) strain, JOL919 (SE PS; Δlon ΔcpxR), using the Lambda-Red recombination method and evaluated the safety and immunological aspects of the novel genotype, JOL2381 (SE VS: Δlon, ΔcpxR, ΔrfaL). Assessment of fecal shedding and organ persistence following administration via oral and IM routes revealed that the SE VS was safer than its parent strain, SE PS. Immunological assays confirmed that immunization via the oral route with SE PS was superior to the SE VS. However, chickens immunized with SE PS and SE VS strains via the IM route showed higher humoral and cell-mediated immune responses. Compared to PBS control, the IM route of immunization with SE VS resulted in a higher IgY antibody titer and expansion of CD4+ and CD8+ T-cell populations, which resulted in the clearance of Salmonella from the liver and splenic tissues. Furthermore, deletion of the O-antigen ligase gene caused lower production of LPS-specific antibodies in the host, promoting DIVA functionality and making it a plausible candidate for field utilization. Due to significant protection, high attenuation, and environmental safety concerns, the present SE VS strain is an ideal choice to prevent chicken salmonellosis and ensure public health.
Collapse
Affiliation(s)
- Amal Senevirathne
- College of Veterinary Medicine, Jeonbuk National University, 54596 Iksan, Republic of Korea
| | - Chamith Hewawaduge
- College of Veterinary Medicine, Jeonbuk National University, 54596 Iksan, Republic of Korea
| | - John Hwa Lee
- College of Veterinary Medicine, Jeonbuk National University, 54596 Iksan, Republic of Korea.
| |
Collapse
|
10
|
Lamichhane B, Mawad AMM, Saleh M, Kelley WG, Harrington PJ, Lovestad CW, Amezcua J, Sarhan MM, El Zowalaty ME, Ramadan H, Morgan M, Helmy YA. Salmonellosis: An Overview of Epidemiology, Pathogenesis, and Innovative Approaches to Mitigate the Antimicrobial Resistant Infections. Antibiotics (Basel) 2024; 13:76. [PMID: 38247636 PMCID: PMC10812683 DOI: 10.3390/antibiotics13010076] [Citation(s) in RCA: 25] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 12/24/2023] [Accepted: 01/10/2024] [Indexed: 01/23/2024] Open
Abstract
Salmonella is a major foodborne pathogen and a leading cause of gastroenteritis in humans and animals. Salmonella is highly pathogenic and encompasses more than 2600 characterized serovars. The transmission of Salmonella to humans occurs through the farm-to-fork continuum and is commonly linked to the consumption of animal-derived food products. Among these sources, poultry and poultry products are primary contributors, followed by beef, pork, fish, and non-animal-derived food such as fruits and vegetables. While antibiotics constitute the primary treatment for salmonellosis, the emergence of antibiotic resistance and the rise of multidrug-resistant (MDR) Salmonella strains have highlighted the urgency of developing antibiotic alternatives. Effective infection management necessitates a comprehensive understanding of the pathogen's epidemiology and transmission dynamics. Therefore, this comprehensive review focuses on the epidemiology, sources of infection, risk factors, transmission dynamics, and the host range of Salmonella serotypes. This review also investigates the disease characteristics observed in both humans and animals, antibiotic resistance, pathogenesis, and potential strategies for treatment and control of salmonellosis, emphasizing the most recent antibiotic-alternative approaches for infection control.
Collapse
Affiliation(s)
- Bibek Lamichhane
- Department of Veterinary Science, Martin-Gatton College of Agriculture, Food and Environment, University of Kentucky, Lexington, KY 40546, USA
| | - Asmaa M. M. Mawad
- Botany and Microbiology Department, Faculty of Science, Assiut University, Assiut 71516, Egypt
| | - Mohamed Saleh
- Department of Veterinary Science, Martin-Gatton College of Agriculture, Food and Environment, University of Kentucky, Lexington, KY 40546, USA
| | - William G. Kelley
- Department of Veterinary Science, Martin-Gatton College of Agriculture, Food and Environment, University of Kentucky, Lexington, KY 40546, USA
| | - Patrick J. Harrington
- Department of Veterinary Science, Martin-Gatton College of Agriculture, Food and Environment, University of Kentucky, Lexington, KY 40546, USA
| | - Cayenne W. Lovestad
- Department of Veterinary Science, Martin-Gatton College of Agriculture, Food and Environment, University of Kentucky, Lexington, KY 40546, USA
| | - Jessica Amezcua
- Department of Veterinary Science, Martin-Gatton College of Agriculture, Food and Environment, University of Kentucky, Lexington, KY 40546, USA
| | - Mohamed M. Sarhan
- Faculty of Pharmacy, King Salman International University (KSIU), Ras Sudr 8744304, Egypt
| | - Mohamed E. El Zowalaty
- Veterinary Medicine and Food Security Research Group, Medical Laboratory Sciences Program, Faculty of Health Sciences, Abu Dhabi Women’s Campus, Higher Colleges of Technology, Abu Dhabi 41012, United Arab Emirates
| | - Hazem Ramadan
- Hygiene and Zoonoses Department, Faculty of Veterinary Medicine, Mansoura University, Mansoura 35516, Egypt
| | - Melissa Morgan
- Department of Animal and Food Sciences, Martin-Gatton College of Agriculture, Food and Environment, University of Kentucky, Lexington, KY 40546, USA
| | - Yosra A. Helmy
- Department of Veterinary Science, Martin-Gatton College of Agriculture, Food and Environment, University of Kentucky, Lexington, KY 40546, USA
| |
Collapse
|
11
|
Benefo EO, Karanth S, Pradhan AK. A machine learning approach to identifying Salmonella stress response genes in isolates from poultry processing. Food Res Int 2024; 175:113635. [PMID: 38128977 DOI: 10.1016/j.foodres.2023.113635] [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: 06/07/2023] [Revised: 10/21/2023] [Accepted: 10/24/2023] [Indexed: 12/23/2023]
Abstract
We explored the potential of machine learning to identify significant genes associated with Salmonella stress response during poultry processing using whole genome sequencing (WGS) data. The Salmonella isolates (n = 177) used in this study were obtained from various chicken sources (skin before chiller, chicken carcass before chiller, frozen chicken, and post-chill chicken carcass). Six machine learning algorithms (random forest, neural network, cost-sensitive learning, logit boost, and support vector machine linear and radial kernels) were trained on Salmonella WGS data, and model fit was assessed using standard evaluation metrics such as the area under the receiver operating characteristic (AUROC) curve and confusion matrix statistics. All models achieved high performances based on the AUROC metric, with logit boost showing the best performance with an AUROC score of 0.904, sensitivity of 0.889, and specificity of 0.920. The significant genes identified included ybtX, which encodes a Yersiniabactin-associated zinc transporter, and the transferase-encoding genes yccK and thiS. Additionally, genes coding for cold (cspA, cspD, and cspE) and heat shock (rpoH and rpoE) responses were identified. Other significant genes included those involved in lipopolysaccharide biosynthesis (irp1, waaD, rfc, and rfbX), DNA repair and replication (traI), biofilm formation (ccdA and fyuA), and cellular metabolism (irtA).
Collapse
Affiliation(s)
- Edmund O Benefo
- Department of Nutrition and Food Science, University of Maryland, College Park, MD 20742, USA
| | - Shraddha Karanth
- Department of Nutrition and Food Science, University of Maryland, College Park, MD 20742, USA
| | - Abani K Pradhan
- Department of Nutrition and Food Science, University of Maryland, College Park, MD 20742, USA; Center for Food Safety and Security Systems, University of Maryland, College Park, MD 20742, USA.
| |
Collapse
|
12
|
Bian X, Liu Q, Chen Y, Zhang W, Li M, Zhang X, Yang L, Liao Y, Kong Q. Immunogenicity and cross-protective efficacy induced by delayed attenuated Salmonella with regulated length of lipopolysaccharide in mice. Gut Microbes 2024; 16:2424983. [PMID: 39529227 PMCID: PMC11559367 DOI: 10.1080/19490976.2024.2424983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 10/24/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024] Open
Abstract
Non-typhoidal Salmonella enterica (NTS) is a major global foodborne pathogen that poses a major public health concern worldwide, and no vaccines are available for protecting against infection of multiple Salmonella serotypes, therefore, the development of Salmonella vaccines to provide broad protection is valuable. In this work, we aimed to regulate lipopolysaccharide (LPS) synthesis of live Salmonella in vivo for exposing conserved protein antigens on the outer membrane while maintaining smooth LPS patterns in vitro to keep their original ability to invade host cells for inducing cross-protection against infection of multiple Salmonella serotypes. We generated a series of mutants defective in genes to affect the length of LPS. These mutants exhibit in vivo regulated-delayed attenuation and altered length of LPS, and all these mutants were derived from SW067 (ΔpagL7 ΔpagP81::Plpp lpxE ΔlpxR9 Δfur9) containing ∆pagP81::Plpp lpxE mutation to reduce their endotoxic activity. Animal experiments demonstrated that all regulated delayed attenuated mutants exhibited reduced ability to colonize the organs of the mice, and SW114 (waaI), SW116 (waaJ), SW118 (waaL), and SW120 (wbaP) induced a significant production of IgG and IgA against OMPs isolated from S. Typhimurium, S. Enteritidis, and S. Choleraesuis. SW114 (waaI), SW116 (waaJ), and SW118 (waaL) were capable of conferring significant protection against infection of wild-type S. Enteritidis and S. Choleraesuis, with SW118 (waaL) triggering significant CD4+ T-cell responses as well as the B220low IgG+ BM cell. In conclusion, regulated delayed attenuated Salmonella vaccines with the whole core oligosaccharides of LPS showed a goo.d ability to expose conserved outer antigens and to trigger strong cross-immune responses against both homologous and heterologous Salmonella infections. These results give new insight into the development of the Salmonella vaccine against multiple serotypes of Salmonella.
Collapse
Affiliation(s)
- Xiaoping Bian
- College of Veterinary Medicine, Southwest University, Beibei, Chongqing, China
- Yibin Academy of Southwest University, Yibin, Sichuan, China
| | - Qing Liu
- College of Veterinary Medicine, Southwest University, Beibei, Chongqing, China
- Yibin Academy of Southwest University, Yibin, Sichuan, China
| | - Yaolin Chen
- College of Veterinary Medicine, Southwest University, Beibei, Chongqing, China
| | - Wenjin Zhang
- College of Veterinary Medicine, Southwest University, Beibei, Chongqing, China
| | - Mengru Li
- College of Veterinary Medicine, Southwest University, Beibei, Chongqing, China
| | - Xiaofen Zhang
- College of Veterinary Medicine, Southwest University, Beibei, Chongqing, China
| | - Liu Yang
- National Center of Technology Innovation for Pigs, Rongchang, Chongqing, China
| | - Yonghong Liao
- College of Veterinary Medicine, Southwest University, Beibei, Chongqing, China
| | - Qingke Kong
- College of Veterinary Medicine, Southwest University, Beibei, Chongqing, China
- Yibin Academy of Southwest University, Yibin, Sichuan, China
| |
Collapse
|
13
|
Abstract
This review is focused on describing and analyzing means by which Salmonella enterica serotype strains have been genetically modified with the purpose of developing safe, efficacious vaccines to present Salmonella-induced disease in poultry and to prevent Salmonella colonization of poultry to reduce transmission through the food chain in and on eggs and poultry meat. Emphasis is on use of recently developed means to generate defined deletion mutations to eliminate genetic sequences conferring antimicrobial resistance or residual elements that might lead to genetic instability. Problems associated with prior means to develop vaccines are discussed with presentation of various means by which these problems have been lessened, if not eliminated. Practical considerations are also discussed in hope of facilitating means to move lab-proven successful vaccination procedures and vaccine candidates to the marketplace to benefit the poultry industry.
Collapse
Affiliation(s)
- Roy Curtiss
- College of Veterinary Medicine, University of Florida, Gainesville, Florida,
| |
Collapse
|
14
|
Zhou G, Ma Q, Li Q, Wang S, Shi H. The disruption of LPS in Salmonella Typhimurium provides partial protection against Salmonella Choleraesuis as a live attenuated vaccine. Vaccine 2023; 41:5951-5956. [PMID: 37666697 DOI: 10.1016/j.vaccine.2023.08.078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 07/28/2023] [Accepted: 08/28/2023] [Indexed: 09/06/2023]
Abstract
Interference with the normal synthesis of LPS was shown to enhance immune responses to conserved outer membrane proteins. In the present study, we have constructed three vaccine candidates by deleting four genes (rfaL, rfbB, rffG and wzy) associated with LPS synthesis in the wild-type strain UK-1. Virulence assessment showed that after oral immunization of BALB/c mice, all mutant strains were attenuated and had significantly reduced ability to colonize host tissues compared to the wild-type strain. In addition, all three vaccine candidates induced elevated humoral, mucosal and cellular immune responses against S. Typhimurium and S. Choleraesuis OMPs compared to the PBS-treated group. Finally, immunization of mice with the rSC0136 vaccine candidate strain provided 100 % and 40 % protection against S. Typhimurium and S. Choleraesuis challenge, respectively. These results suggest that the deletion of LPS synthesis-related genes may be an effective strategy against homologous serotypes, but provides only partial protection against heterologous serotypes.
Collapse
Affiliation(s)
- Guodong Zhou
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, Jiangsu, China; Jiangsu Co-innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China.
| | - Qifeng Ma
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, Jiangsu, China; Jiangsu Co-innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China.
| | - Quan Li
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, Jiangsu, China; Jiangsu Co-innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China.
| | - Shifeng Wang
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL 32611-0880, USA.
| | - Huoying Shi
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, Jiangsu, China; Jiangsu Co-innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, Yangzhou University (JIRLAAPS), Yangzhou, China.
| |
Collapse
|
15
|
Chen GY, Thorup NR, Miller AJ, Li YC, Ayres JS. Cooperation between physiological defenses and immune resistance produces asymptomatic carriage of a lethal bacterial pathogen. SCIENCE ADVANCES 2023; 9:eadg8719. [PMID: 37352357 PMCID: PMC10289649 DOI: 10.1126/sciadv.adg8719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 05/18/2023] [Indexed: 06/25/2023]
Abstract
Animals evolved two defense strategies to survive infections. Antagonistic strategies include immune resistance mechanisms that operate to kill invading pathogens. Cooperative or physiological defenses mediate host adaptation to the infected state, limiting physiological damage and disease, without killing the pathogen, and have been shown to cause asymptomatic carriage and transmission of lethal pathogens. Here, we demonstrate that physiological defenses cooperate with the adaptive immune response to generate long-term asymptomatic carriage of the lethal enteric murine pathogen, Citrobacter rodentium. Asymptomatic carriage of genetically virulent C. rodentium provided immune resistance against subsequent infections. Immune protection was dependent on systemic antibody responses and pathogen virulence behavior rather than the recognition of specific virulent antigens. Last, we demonstrate that an avirulent strain of C. rodentium in the field has background mutations in genes that are important for LPS structure. Our work reveals insight into how asymptomatic infections can arise mechanistically with immune resistance, mediating exclusion of phenotypically virulent enteric pathogen to promote asymptomatic carriage.
Collapse
Affiliation(s)
- Grischa Y. Chen
- Molecular and Systems Physiology Lab, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
- Gene Expression Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
- NOMIS Center for Immunobiology and Microbial Pathogenesis, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Natalia R. Thorup
- Molecular and Systems Physiology Lab, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
- Gene Expression Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
- NOMIS Center for Immunobiology and Microbial Pathogenesis, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Abigail J. Miller
- Molecular and Systems Physiology Lab, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
- Gene Expression Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
- NOMIS Center for Immunobiology and Microbial Pathogenesis, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Yao-Cheng Li
- Gene Expression Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Janelle S. Ayres
- Molecular and Systems Physiology Lab, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
- Gene Expression Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
- NOMIS Center for Immunobiology and Microbial Pathogenesis, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| |
Collapse
|
16
|
Fan X, Zheng S, Chen C, Lin L, Wang H, Shen Y, Pan Y, Li C. Sialidase facilitates Porphyromonas gingivalis immune evasion by reducing M1 polarization, antigen presentation, and phagocytosis of infected macrophages. Front Cell Infect Microbiol 2023; 13:1173899. [PMID: 37325520 PMCID: PMC10266273 DOI: 10.3389/fcimb.2023.1173899] [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: 02/25/2023] [Accepted: 05/15/2023] [Indexed: 06/17/2023] Open
Abstract
Background Porphyromonas gingivalis (P. gingivalis), a major pathogen of periodontitis, can evade host immune defenses. Previously, we found that P. gingivalis W83 sialidase gene mutant strain (ΔPG0352) was more easily cleared by macrophages. The aims of this study were to investigate the effects of sialidase in P. gingivalis on the polarization, antigen presentation, and phagocytosis of infected macrophages and to clarify the mechanism of P. gingivalis immune evasion. Methods Human monocytes U937 were differentiated to macrophages and infected with P. gingivalis W83, ΔPG0352, comΔPG0352, and Escherichia coli (E. coli). The phagocytosis of macrophages was observed by transmission electron microscopy and flow cytometry. ELISA or Griess reaction were used to examine the levels of interleukin-12 (IL-12), inducible nitric oxide synthase (iNOS) and interleukin-10 (IL-10), and the expressions of CD68, CD80 and CD206 were determined by flow cytometry. The expression of major histocompatibility complex-II (MHC-II) was detected by immunofluorescence. A rat periodontitis model was established to determine the M1 and M2 polarization of macrophages. Results Compare with P. gingivalis W83, ΔPG0352 increased the levels of IL-12, iNOS, CD80, and MHC-II and inhibited the levels of IL-10 and CD206. Macrophages phagocytosed 75.4% of ΔPG0352 and 59.5% of P. gingivalis W83. In the rat periodontitis model, the levels of M1 and M2 macrophages in P. gingivalis W83 group were both higher than those in ΔPG0352 group, while the ratio of M1/M2 was higher in the ΔPG0352 group. Alveolar bone absorption was lower in ΔPG0352 group. Conclusion Sialidase facilitates P. gingivalis immune evasion by reducing M1 polarization, antigen presentation, and phagocytosis of infected macrophages.
Collapse
Affiliation(s)
- Xiaomiao Fan
- Department of Periodontology, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Disease, Shenyang, Liaoning, China
- Department of Periodontics, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, Guangdong, China
| | - Shaowen Zheng
- Department of Periodontology, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Disease, Shenyang, Liaoning, China
| | - Chen Chen
- Department of Periodontology, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Disease, Shenyang, Liaoning, China
| | - Li Lin
- Department of Periodontology, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Disease, Shenyang, Liaoning, China
| | - Hongyan Wang
- Department of Periodontology, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Disease, Shenyang, Liaoning, China
| | - Yuqin Shen
- Department of Periodontics, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, Guangdong, China
| | - Yaping Pan
- Department of Periodontology, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Disease, Shenyang, Liaoning, China
| | - Chen Li
- Department of Periodontology, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Disease, Shenyang, Liaoning, China
| |
Collapse
|
17
|
Dodge GJ, Bernstein HM, Imperiali B. A generalizable protocol for expression and purification of membrane-bound bacterial phosphoglycosyl transferases in liponanoparticles. Protein Expr Purif 2023; 207:106273. [PMID: 37068720 DOI: 10.1016/j.pep.2023.106273] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 04/12/2023] [Accepted: 04/14/2023] [Indexed: 04/19/2023]
Abstract
Phosphoglycosyl transferases (PGTs) are among the first membrane-bound enzymes involved in the biosynthesis of bacterial glycoconjugates. Robust expression and purification protocols for an abundant subfamily of PGTs remains lacking. Recent advancements in detergent-free methods for membrane protein solubilization open the door for purification of difficult membrane proteins directly from cell membranes into native-like liponanoparticles. By leveraging autoinduction, in vivo SUMO tag cleavage, styrene maleic acid co-polymer liponanoparticles (SMALPs), and Strep-Tag purification, we have established a robust workflow for expression and purification of previously unobtainable PGTs. The material generated from this workflow is extremely pure and can be directly visualized by Cryogenic Electron Microscopy (CryoEM). The methods presented here promise to be generalizable to additional membrane proteins recombinantly expressed in E. coli and should be of interest to the greater membrane proteomics community.
Collapse
Affiliation(s)
- Greg J Dodge
- Department of Biology and Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Hannah M Bernstein
- Department of Biology and Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Barbara Imperiali
- Department of Biology and Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA.
| |
Collapse
|
18
|
Clarke OE, Pelling H, Bennett V, Matsumoto T, Gregory GE, Nzakizwanayo J, Slate AJ, Preston A, Laabei M, Bock LJ, Wand ME, Ikebukuro K, Gebhard S, Sutton JM, Jones BV. Lipopolysaccharide structure modulates cationic biocide susceptibility and crystalline biofilm formation in Proteus mirabilis. Front Microbiol 2023; 14:1150625. [PMID: 37089543 PMCID: PMC10113676 DOI: 10.3389/fmicb.2023.1150625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 03/06/2023] [Indexed: 04/08/2023] Open
Abstract
Chlorhexidine (CHD) is a cationic biocide used ubiquitously in healthcare settings. Proteus mirabilis, an important pathogen of the catheterized urinary tract, and isolates of this species are often described as "resistant" to CHD-containing products used for catheter infection control. To identify the mechanisms underlying reduced CHD susceptibility in P. mirabilis, we subjected the CHD tolerant clinical isolate RS47 to random transposon mutagenesis and screened for mutants with reduced CHD minimum inhibitory concentrations (MICs). One mutant recovered from these screens (designated RS47-2) exhibited ~ 8-fold reduction in CHD MIC. Complete genome sequencing of RS47-2 showed a single mini-Tn5 insert in the waaC gene involved in lipopolysaccharide (LPS) inner core biosynthesis. Phenotypic screening of RS47-2 revealed a significant increase in cell surface hydrophobicity and serum susceptibility compared to the wildtype, and confirmed defects in LPS production congruent with waaC inactivation. Disruption of waaC was also associated with increased susceptibility to a range of other cationic biocides but did not affect susceptibility to antibiotics tested. Complementation studies showed that repression of smvA efflux activity in RS47-2 further increased susceptibility to CHD and other cationic biocides, reducing CHD MICs to values comparable with the most CHD susceptible isolates characterized. The formation of crystalline biofilms and blockage of urethral catheters was also significantly attenuated in RS47-2. Taken together, these data show that aspects of LPS structure and upregulation of the smvA efflux system function in synergy to modulate susceptibility to CHD and other cationic biocides, and that LPS structure is also an important factor in P. mirabilis crystalline biofilm formation.
Collapse
Affiliation(s)
- O. E. Clarke
- Department of Life Sciences, University of Bath, Bath, United Kingdom
| | - H. Pelling
- Department of Life Sciences, University of Bath, Bath, United Kingdom
| | - V. Bennett
- Department of Life Sciences, University of Bath, Bath, United Kingdom
| | - T. Matsumoto
- Department of Biotechnology and Life Sciences, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - G. E. Gregory
- Department of Life Sciences, University of Bath, Bath, United Kingdom
| | - J. Nzakizwanayo
- Department of Life Sciences, University of Bath, Bath, United Kingdom
| | - A. J. Slate
- Department of Life Sciences, University of Bath, Bath, United Kingdom
| | - A. Preston
- Department of Life Sciences, University of Bath, Bath, United Kingdom
| | - M. Laabei
- Department of Life Sciences, University of Bath, Bath, United Kingdom
| | - L. J. Bock
- United Kingdom Health Security Agency, Salisbury, United Kingdom
| | - M. E. Wand
- United Kingdom Health Security Agency, Salisbury, United Kingdom
| | - K. Ikebukuro
- Department of Biotechnology and Life Sciences, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - S. Gebhard
- Department of Life Sciences, University of Bath, Bath, United Kingdom
| | - J. M. Sutton
- United Kingdom Health Security Agency, Salisbury, United Kingdom
| | - B. V. Jones
- Department of Life Sciences, University of Bath, Bath, United Kingdom
| |
Collapse
|
19
|
Comparison of Phenotype and Genotype Virulence and Antimicrobial Factors of Salmonella Typhimurium Isolated from Human Milk. Int J Mol Sci 2023; 24:ijms24065135. [PMID: 36982209 PMCID: PMC10048834 DOI: 10.3390/ijms24065135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 03/03/2023] [Accepted: 03/04/2023] [Indexed: 03/11/2023] Open
Abstract
Salmonella is a common foodborne infection. Many serovars belonging to Salmonella enterica subsp. enterica are present in the gut of various animal species. They can cause infection in human infants via breast milk or cross-contamination with powdered milk. In the present study, Salmonella BO was isolated from human milk in accordance with ISO 6579-1:2017 standards and sequenced using whole-genome sequencing (WGS), followed by serosequencing and genotyping. The results also allowed its pathogenicity to be predicted. The WGS results were compared with the bacterial phenotype. The isolated strain was found to be Salmonella enterica subsp. enterica serovar Typhimurium 4:i:1,2_69M (S. Typhimurium 69M); it showed a very close similarity to S. enterica subsp. enterica serovar Typhimurium LT2. Bioinformatics sequence analysis detected eleven SPIs (SPI-1, SPI-2, SPI-3, SPI-4, SPI-5, SPI-9, SPI-12, SPI-13, SPI-14, C63PI, CS54_island). Significant changes in gene sequences were noted, causing frameshift mutations in yeiG, rfbP, fumA, yeaL, ybeU (insertion) and lpfD, avrA, ratB, yacH (deletion). The sequences of several proteins were significantly different from those coded in the reference genome; their three-dimensional structure was predicted and compared with reference proteins. Our findings indicate the presence of a number of antimicrobial resistance genes that do not directly imply an antibiotic resistance phenotype.
Collapse
|
20
|
Chen GY, Thorup NR, Miller AJ, Li YC, Ayres JS. Cooperation between physiological defenses and immune resistance produces asymptomatic carriage of a lethal bacterial pathogen. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.22.525099. [PMID: 36711884 PMCID: PMC9882269 DOI: 10.1101/2023.01.22.525099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Animals have evolved two defense strategies to survive infections. Antagonistic strategies include mechanisms of immune resistance that operate to sense and kill invading pathogens. Cooperative or physiological defenses mediate host adaptation to the infected state, limiting physiological damage and disease, without killing the pathogen, and have been shown to cause asymptomatic carriage and transmission of lethal pathogens. Here we demonstrate that physiological defenses cooperate with the adaptive immune response to generate long-term asymptomatic carriage of the lethal enteric murine pathogen, Citrobacter rodentium. Asymptomatic carriage of genetically virulent C. rodentium provided immune resistance against subsequent infections. Host immune protection was dependent on systemic antibody responses and pathogen virulence behavior, rather than the recognition of specific virulent factor antigens. Finally, we demonstrate that an avirulent strain of C. rodentium in the field has background mutations in two genes that are important for LPS structure. Our work reveals novel insight into how asymptomatic infections can arise mechanistically with immune resistance, mediating exclusion of phenotypically virulent enteric pathogen to promote asymptomatic carriage.
Collapse
Affiliation(s)
- Grischa Y Chen
- Molecular and Systems Physiology Lab, The Salk Institute for Biological Studies, La Jolla, CA 92037
- Gene Expression Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037
- NOMIS Center for Immunobiology and Microbial Pathogenesis, The Salk Institute for Biological Studies, La Jolla, CA 92037
| | - Natalia R Thorup
- Molecular and Systems Physiology Lab, The Salk Institute for Biological Studies, La Jolla, CA 92037
- Gene Expression Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037
- NOMIS Center for Immunobiology and Microbial Pathogenesis, The Salk Institute for Biological Studies, La Jolla, CA 92037
| | - Abigail J Miller
- Molecular and Systems Physiology Lab, The Salk Institute for Biological Studies, La Jolla, CA 92037
- Gene Expression Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037
- NOMIS Center for Immunobiology and Microbial Pathogenesis, The Salk Institute for Biological Studies, La Jolla, CA 92037
| | - Yao-Cheng Li
- Gene Expression Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037
| | - Janelle S Ayres
- Molecular and Systems Physiology Lab, The Salk Institute for Biological Studies, La Jolla, CA 92037
- Gene Expression Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037
- NOMIS Center for Immunobiology and Microbial Pathogenesis, The Salk Institute for Biological Studies, La Jolla, CA 92037
| |
Collapse
|
21
|
Han Y, Luo P, Zeng H, Wang P, Xu J, Chen P, Chen X, Chen Y, Cao Q, Zhai R, Xia J, Deng S, Cheng A, Cheng C, Song H. The effect of O-antigen length determinant wzz on the immunogenicity of Salmonella Typhimurium for Escherichia coli O2 O-polysaccharides delivery. Vet Res 2023; 54:15. [PMID: 36849993 PMCID: PMC9969949 DOI: 10.1186/s13567-023-01142-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Accepted: 12/15/2022] [Indexed: 03/01/2023] Open
Abstract
Attenuated Salmonella Typhimurium is a promising antigen delivery system for live vaccines such as polysaccharides. The length of polysaccharides is a well-known key factor in modulating the immune response induced by glycoconjugates. However, the relationship between the length of Lipopolysaccharide (LPS) O-antigen (OAg) and the immunogenicity of S. Typhimurium remains unclear. In this study, we assessed the effect of OAg length determined by wzzST on Salmonella colonization, cell membrane permeability, antimicrobial activity, and immunogenicity by comparing the S. Typhimurium wild-type ATCC14028 strain to those with various OAg lengths of the ΔwzzST mutant and ΔwzzST::wzzECO2. The analysis of the OAg length distribution revealed that, except for the very long OAg, the short OAg length of 2-7 repeat units (RUs) was obtained from the ΔwzzST mutant, the intermediate OAg length of 13-21 RUs was gained from ΔwzzST::wzzECO2, and the long OAg length of over 20 RUs was gained from the wild-type. In addition, we found that the OAg length affected Salmonella colonization, cell permeability, and antibiotic resistance. Immunization of mice revealed that shortening the OAg length by altering wzzST had an effect on serum bactericidal ability, complement deposition, and humoral immune response. S. Typhimurium mutant strain ΔwzzST::wzzECO2 possessed good immunogenicity and was the optimum option for delivering E. coli O2 O-polysaccharides. Furthermore, the attenuated strain ATCC14028 ΔasdΔcrpΔcyaΔrfbPΔwzzST::wzzECO2-delivered E. coli O2 OAg gene cluster outperforms the ATCC14028 ΔasdΔcrpΔcyaΔrfbP in terms of IgG eliciting, cytokine expression, and immune protection in chickens. This study sheds light on the role of OAg length in Salmonella characteristics, which may have a potential application in optimizing the efficacy of delivered polysaccharide vaccines.
Collapse
Affiliation(s)
- Yue Han
- grid.443483.c0000 0000 9152 7385Key Laboratory of Applied Technology On Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Research Center for Animal Health Diagnostics & Advanced Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China-Australia Joint Laboratory for Animal, Health Big Data Analytics, College of Animal Science and Technology & College of Veterinary Medicine, Zhejiang A & F University, 666 Wusu Street, Hangzhou, 311300 China ,grid.80510.3c0000 0001 0185 3134Institute of Preventive Veterinary Medicine, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130 China
| | - Ping Luo
- grid.443483.c0000 0000 9152 7385Key Laboratory of Applied Technology On Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Research Center for Animal Health Diagnostics & Advanced Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China-Australia Joint Laboratory for Animal, Health Big Data Analytics, College of Animal Science and Technology & College of Veterinary Medicine, Zhejiang A & F University, 666 Wusu Street, Hangzhou, 311300 China
| | - Huan Zeng
- grid.443483.c0000 0000 9152 7385Key Laboratory of Applied Technology On Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Research Center for Animal Health Diagnostics & Advanced Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China-Australia Joint Laboratory for Animal, Health Big Data Analytics, College of Animal Science and Technology & College of Veterinary Medicine, Zhejiang A & F University, 666 Wusu Street, Hangzhou, 311300 China
| | - Pu Wang
- grid.443483.c0000 0000 9152 7385Key Laboratory of Applied Technology On Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Research Center for Animal Health Diagnostics & Advanced Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China-Australia Joint Laboratory for Animal, Health Big Data Analytics, College of Animal Science and Technology & College of Veterinary Medicine, Zhejiang A & F University, 666 Wusu Street, Hangzhou, 311300 China
| | - Jiali Xu
- grid.443483.c0000 0000 9152 7385Key Laboratory of Applied Technology On Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Research Center for Animal Health Diagnostics & Advanced Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China-Australia Joint Laboratory for Animal, Health Big Data Analytics, College of Animal Science and Technology & College of Veterinary Medicine, Zhejiang A & F University, 666 Wusu Street, Hangzhou, 311300 China
| | - Pengju Chen
- Henan Institute of Morden Chinese Veterinary Medicine, Zhengzhou, 450002 China ,Shangdong Xindehui Biotechnology Co., Ltd, Yunchengxian, 274700 China
| | - Xindan Chen
- grid.443483.c0000 0000 9152 7385Key Laboratory of Applied Technology On Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Research Center for Animal Health Diagnostics & Advanced Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China-Australia Joint Laboratory for Animal, Health Big Data Analytics, College of Animal Science and Technology & College of Veterinary Medicine, Zhejiang A & F University, 666 Wusu Street, Hangzhou, 311300 China
| | - Yuji Chen
- grid.443483.c0000 0000 9152 7385Key Laboratory of Applied Technology On Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Research Center for Animal Health Diagnostics & Advanced Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China-Australia Joint Laboratory for Animal, Health Big Data Analytics, College of Animal Science and Technology & College of Veterinary Medicine, Zhejiang A & F University, 666 Wusu Street, Hangzhou, 311300 China
| | - Qiyu Cao
- grid.443483.c0000 0000 9152 7385Key Laboratory of Applied Technology On Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Research Center for Animal Health Diagnostics & Advanced Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China-Australia Joint Laboratory for Animal, Health Big Data Analytics, College of Animal Science and Technology & College of Veterinary Medicine, Zhejiang A & F University, 666 Wusu Street, Hangzhou, 311300 China
| | - Ruidong Zhai
- grid.443483.c0000 0000 9152 7385Key Laboratory of Applied Technology On Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Research Center for Animal Health Diagnostics & Advanced Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China-Australia Joint Laboratory for Animal, Health Big Data Analytics, College of Animal Science and Technology & College of Veterinary Medicine, Zhejiang A & F University, 666 Wusu Street, Hangzhou, 311300 China
| | - Jing Xia
- grid.443483.c0000 0000 9152 7385Key Laboratory of Applied Technology On Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Research Center for Animal Health Diagnostics & Advanced Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China-Australia Joint Laboratory for Animal, Health Big Data Analytics, College of Animal Science and Technology & College of Veterinary Medicine, Zhejiang A & F University, 666 Wusu Street, Hangzhou, 311300 China
| | - Simin Deng
- grid.443483.c0000 0000 9152 7385Key Laboratory of Applied Technology On Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Research Center for Animal Health Diagnostics & Advanced Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China-Australia Joint Laboratory for Animal, Health Big Data Analytics, College of Animal Science and Technology & College of Veterinary Medicine, Zhejiang A & F University, 666 Wusu Street, Hangzhou, 311300 China
| | - Anchun Cheng
- grid.80510.3c0000 0001 0185 3134Institute of Preventive Veterinary Medicine, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130 China
| | - Changyong Cheng
- Key Laboratory of Applied Technology On Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Research Center for Animal Health Diagnostics & Advanced Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China-Australia Joint Laboratory for Animal, Health Big Data Analytics, College of Animal Science and Technology & College of Veterinary Medicine, Zhejiang A & F University, 666 Wusu Street, Hangzhou, 311300, China.
| | - Houhui Song
- Key Laboratory of Applied Technology On Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Research Center for Animal Health Diagnostics & Advanced Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China-Australia Joint Laboratory for Animal, Health Big Data Analytics, College of Animal Science and Technology & College of Veterinary Medicine, Zhejiang A & F University, 666 Wusu Street, Hangzhou, 311300, China.
| |
Collapse
|
22
|
Won K, Kim D, Shin D, Hur J, Lee HK, Heo J, Oh JD. High-throughput sequencing-based metagenomic and transcriptomic analysis of intestine in piglets infected with salmonella. JOURNAL OF ANIMAL SCIENCE AND TECHNOLOGY 2022; 64:1144-1172. [PMID: 36812005 PMCID: PMC9890335 DOI: 10.5187/jast.2022.e73] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 08/17/2022] [Accepted: 09/05/2022] [Indexed: 11/05/2022]
Abstract
Salmonella enterica serovar Typhimurium isolate HJL777 is a virulent bacterial strain in pigs. The high rate of salmonella infection are at high risk of non-typhoidal salmonella gastroenteritis development. Salmonellosis is most common in young pigs. We investigated changes in gut microbiota and biological function in piglets infected with salmonella via analysis of rectal fecal metagenome and intestinal transcriptome using 16S rRNA and RNA sequencing. We identified a decrease in Bacteroides and increase in harmful bacteria such as Spirochaetes and Proteobacteria by microbial community analysis. We predicted that reduction of Bacteroides by salmonella infection causes proliferation of salmonella and harmful bacteria that can cause an intestinal inflammatory response. Functional profiling of microbial communities in piglets with salmonella infection showed increasing lipid metabolism associated with proliferation of harmful bacteria and inflammatory responses. Transcriptome analysis identified 31 differentially expressed genes. Using gene ontology and Innate Immune Database analysis, we identified that BGN, DCN, ZFPM2 and BPI genes were involved in extracellular and immune mechanisms, specifically salmonella adhesion to host cells and inflammatory responses during infection. We confirmed alterations in gut microbiota and biological function during salmonella infection in piglets. Our findings will help prevent disease and improve productivity in the swine industry.
Collapse
Affiliation(s)
- KyeongHye Won
- Department of Animal Biotechnology,
College of Agricultural and Life Sciences, Jeonbuk National
University, Jeonju 54896, Korea
| | - Dohyun Kim
- Department of Animal Biotechnology,
College of Agricultural and Life Sciences, Jeonbuk National
University, Jeonju 54896, Korea
| | - Donghyun Shin
- Department of Agricultural Convergence
Technology, Jeonbuk National University, Jeonju 54896,
Korea
| | - Jin Hur
- Department of Veterinary Public Health,
College of Veterinary Medicine, Jeonbuk National University,
Iksan 54596, Korea
| | - Hak-Kyo Lee
- Department of Animal Biotechnology,
College of Agricultural and Life Sciences, Jeonbuk National
University, Jeonju 54896, Korea,Department of Agricultural Convergence
Technology, Jeonbuk National University, Jeonju 54896,
Korea
| | - Jaeyoung Heo
- Department of Animal Biotechnology,
College of Agricultural and Life Sciences, Jeonbuk National
University, Jeonju 54896, Korea,Corresponding author: Jaeyoung Heo,
Department of Animal Biotechnology, College of Agricultural and Life Sciences,
Jeonbuk National University, Jeonju 54896, Korea. Tel: +82-63-270-2549, E-mail:
| | - Jae-Don Oh
- Department of Animal Biotechnology,
College of Agricultural and Life Sciences, Jeonbuk National
University, Jeonju 54896, Korea,Corresponding author: Jae-Don Oh,
Department of Animal Biotechnology, College of Agricultural and Life Sciences,
Jeonbuk National University, Jeonju 54896, Korea. Tel: +82-63-270-5931, E-mail:
| |
Collapse
|
23
|
Fitness Trade-Offs in Phage Cocktail-Resistant Salmonella enterica Serovar Enteritidis Results in Increased Antibiotic Susceptibility and Reduced Virulence. Microbiol Spectr 2022; 10:e0291422. [PMID: 36165776 PMCID: PMC9603643 DOI: 10.1128/spectrum.02914-22] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
The rapid emergence of phage-resistant bacterial mutants is a major challenge for phage therapy. Phage cocktails have been considered one approach to mitigate this issue. However, the synergistic effect of randomly selected phages in the cocktails is ambiguous. Here, we rationally designed a phage cocktail consisting of four phages that utilize the lipopolysaccharide (LPS) O antigen, the LPS outer core, the LPS inner core, and the outer membrane proteins BtuB and TolC on the Salmonella enterica serovar Enteritidis cell surface as receptors. We demonstrated that the four-phage cocktail could significantly delay the emergence of phage-resistant bacterial mutants compared to the single phage. To investigate the fitness costs associated with phage resistance, we characterized a total of 80 bacterial mutants resistant to a single phage or the four-phage cocktail. We observed that mutants resistant to the four-phage cocktail were more sensitive to several antibiotics than the single-phage-resistant mutants. In addition, all mutants resistant to the four-phage cocktail had significantly reduced virulence compared to wild-type strains. Our mouse model of Salmonella Enteritidis infection also indicated that the four-phage cocktail exhibited an enhanced therapeutic effect. Together, our work demonstrates an efficient strategy to design phage cocktails by combining phages with different bacterial receptors, which can steer the evolution of phage-resistant strains toward clinically exploitable phenotypes. IMPORTANCE The selection pressure of phage promotes bacterial mutation, which results in a fitness cost. Such fitness trade-offs are related to the host receptor of the phage; therefore, we can utilize knowledge of bacterial receptors used by phages as a criterion for designing phage cocktails. Here, we evaluated the efficacy of a phage cocktail made up of phages that target four different receptors on Salmonella Enteritidis through in vivo and in vitro experiments. Importantly, we found that pressure from phage cocktails with different receptors can drive phage-resistant bacterial mutants to evolve in a direction that entails more severe fitness costs, resulting in reduced virulence and increased susceptibility to antibiotics. These findings suggest that phage cocktail therapy using combinations of phages targeting different important receptors (e.g., LPS or the efflux pump AcrAB-TolC) on the host surface can steer the host bacteria toward more detrimental surface mutations than single-phage therapy, resulting in more favorable therapeutic outcomes.
Collapse
|
24
|
Recent Advances in Bacteria-Based Cancer Treatment. Cancers (Basel) 2022; 14:cancers14194945. [PMID: 36230868 PMCID: PMC9563255 DOI: 10.3390/cancers14194945] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 09/29/2022] [Accepted: 10/03/2022] [Indexed: 12/04/2022] Open
Abstract
Simple Summary Cancer refers to a disease involving abnormal cells that proliferate uncontrollably and can invade normal body tissue. It was estimated that at least 9 million patients are killed by cancer annually. Recent studies have demonstrated that bacteria play a significant role in cancer treatment and prevention. Owing to its unique mechanism of abundant pathogen-associated molecular patterns in antitumor immune responses and preferentially accumulating and proliferating within tumors, bacteria-based cancer immunotherapy has recently attracted wide attention. We aim to illustrate that naïve bacteria and their components can serve as robust theranostic agents for cancer eradication. In addition, we summarize the recent advances in efficient antitumor treatments by genetically engineering bacteria and bacteria-based nanoparticles. Further, possible future perspectives in bacteria-based cancer immunotherapy are also inspected. Abstract Owing to its unique mechanism of abundant pathogen-associated molecular patterns in antitumor immune responses, bacteria-based cancer immunotherapy has recently attracted wide attention. Compared to traditional cancer treatments such as surgery, chemotherapy, radiotherapy, and phototherapy, bacteria-based cancer immunotherapy exhibits the versatile capabilities for suppressing cancer thanks to its preferentially accumulating and proliferating within tumors. In particular, bacteria have demonstrated their anticancer effect through the toxins, and other active components from the cell membrane, cell wall, and dormant spores. More importantly, the design of engineering bacteria with detoxification and specificity is essential for the efficacy of bacteria-based cancer therapeutics. Meanwhile, bacteria can deliver the cytokines, antibody, and other anticancer theranostic nanoparticles to tumor microenvironments by regulating the expression of the bacterial genes or chemical and physical loading. In this review, we illustrate that naïve bacteria and their components can serve as robust theranostic agents for cancer eradication. In addition, we summarize the recent advances in efficient antitumor treatments by genetically engineering bacteria and bacteria-based nanoparticles. Further, possible future perspectives in bacteria-based cancer immunotherapy are also inspected.
Collapse
|
25
|
Aganja RP, Sivasankar C, Hewawaduge C, Lee JH. Safety assessment of compliant, highly invasive, lipid A-altered, O-antigen-defected Salmonella strains as prospective vaccine delivery systems. Vet Res 2022; 53:76. [PMID: 36183131 PMCID: PMC9526937 DOI: 10.1186/s13567-022-01096-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 08/22/2022] [Indexed: 11/23/2022] Open
Abstract
In the present study, two prospective Salmonella delivery strains, JOL2782 and JOL2837, were developed by gene deletions of lon and cpxR, which are related to cellular adhesion and intracellular survival. Additionally, sifA deletion was introduced for JOL2782, which confers immune susceptibility and improves antigen delivery. Similarly, the rfaL deletion and lpxE substitution for pagL were accomplished in JOL2837 to reduce virulence and endotoxicity. Thus, enhanced adhesion and invasion and reduced intracellular survival were attained. Furthermore, aspartic acid auxotrophic (asd) was deleted to impose Darwinian selection on retention of the foreign antigen-expressing plasmid. Both delivery strains induced sufficient cytokine expression, but the level was significantly lower than that of the wild-type strain; the lowest cytokine expression was induced by the JOL2837 strain, indicating reduced endotoxicity. In parallel, IgG production was significantly enhanced by both delivery strains. Thus, the innate and adaptive immunogenicity of the strains was ensured. The environmental safety of these strains was ascertained through faecal dissemination assays. The nonpathogenicity of these strains to the host was confirmed by body weight monitoring, survival assays, and morphological and histological assessments of the vital organs. The in vitro assay in murine and human cell lines and in vivo safety assessments in mice suggest that these novel strains possess safety, invasiveness, and immunogenicity, making them ideal delivery strains. Overall, the results clearly showed that strain JOL2782 with sifA deletion had higher invasiveness, demonstrating superior vaccine deliverability, while JOL2837 with lpxE substitution for pagL and rfaL deletion had outstanding safety potential with drastically abridged endotoxicity.
Collapse
Affiliation(s)
- Ram Prasad Aganja
- Department of Veterinary Public Health, College of Veterinary Medicine, Jeonbuk National University, Iksan, 54596, South Korea
| | - Chandran Sivasankar
- Department of Veterinary Public Health, College of Veterinary Medicine, Jeonbuk National University, Iksan, 54596, South Korea
| | - Chamith Hewawaduge
- Department of Veterinary Public Health, College of Veterinary Medicine, Jeonbuk National University, Iksan, 54596, South Korea
| | - John Hwa Lee
- Department of Veterinary Public Health, College of Veterinary Medicine, Jeonbuk National University, Iksan, 54596, South Korea.
| |
Collapse
|
26
|
Wang F, Wang L, Ge H, Wang X, Guo Y, Xu Z, Geng S, Jiao X, Chen X. Safety of the Salmonella enterica serotype Dublin strain Sdu189-derived live attenuated vaccine—A pilot study. Front Vet Sci 2022; 9:986332. [PMID: 36246339 PMCID: PMC9554587 DOI: 10.3389/fvets.2022.986332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 08/30/2022] [Indexed: 11/15/2022] Open
Abstract
Salmonella enterica serovar Dublin (S. Dublin) is an important zoonotic pathogen with high invasiveness. In the prevention and control of the Salmonella epidemic, the live attenuated vaccine plays a very important role. To prevent and control the epidemic of S. Dublin in cattle farms, the development of more effective vaccines is necessary. In this study, we constructed two gene deletion mutants, Sdu189ΔspiC and Sdu189ΔspiCΔaroA, with the parental strain S. Dublin Sdu189. The immunogenicity and protective efficacy were evaluated in the mice model. First, both mutant strains were much less virulent than the parental strain, as determined by the 50% lethal dose (LD50) for specific pathogen-free (SPF) 6-week-old female BALB/c mice. Second, the specific IgG antibody level and the expression level of cytokine TNF-α, IFN-γ, IL-4, and IL-18 were increased significantly in the vaccinated mice compared to the control group. In addition, the deletion strains were cleared rapidly from organs of immunized mice within 14 d after immunization, while the parental strain could still be detected in the spleen and liver after 21 d of infection. Compared with the parental strain infected group, no obvious lesions were detected in the liver, spleen, and cecum of the deletion strain vaccinated groups of mice. Immunization with Sdu189ΔspiC and Sdu189ΔspiCΔaroA both provided 100% protection against subsequent challenges with the wild-type Sdu189 strain. These results demonstrated that these two deletion strains showed the potential as live attenuated vaccines against S. Dublin infection. The present study established a foundation for screening a suitable live attenuated Salmonella vaccine.
Collapse
Affiliation(s)
- Fuzhong Wang
- Jiangsu Key Laboratory of Zoonosis/Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
- Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality of Ministry of Agriculture and Rural Affairs, Yangzhou University, Yangzhou, China
| | - Lei Wang
- Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality of Ministry of Agriculture and Rural Affairs, Yangzhou University, Yangzhou, China
| | - Haojie Ge
- Jiangsu Key Laboratory of Zoonosis/Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
| | - Xiaobo Wang
- Jiangsu Key Laboratory of Zoonosis/Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
| | - Yaxin Guo
- Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality of Ministry of Agriculture and Rural Affairs, Yangzhou University, Yangzhou, China
| | - Zhengzhong Xu
- Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality of Ministry of Agriculture and Rural Affairs, Yangzhou University, Yangzhou, China
| | - 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, China
| | - Xin'an Jiao
- Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality of Ministry of Agriculture and Rural Affairs, Yangzhou University, Yangzhou, China
- Xin'an Jiao
| | - Xiang Chen
- Jiangsu Key Laboratory of Zoonosis/Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
- *Correspondence: Xiang Chen
| |
Collapse
|
27
|
Attenuated Salmonella Typhimurium with truncated LPS and outer membrane-displayed RGD peptide for cancer therapy. Biomed Pharmacother 2022; 155:113682. [PMID: 36095964 DOI: 10.1016/j.biopha.2022.113682] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 09/01/2022] [Accepted: 09/07/2022] [Indexed: 11/21/2022] Open
Abstract
Gram-negative, facultatively anaerobic bacteria Salmonella Typhimurium is a candidate agent or delivery vector for cancer therapy. Effective targeted therapies in addition to radiotherapy, chemotherapy and surgery have been urgently needed as an alternative or supplement. This study expected to further improve the tumor-targeting ability of Salmonella bacteria through genetic modifications. Based on an auxotrophic Salmonella bacterial strain (D2), we constructed Salmonella mutants with altered LPS length to facilitate displaying the RGD4C targeting peptide on the outer membrane surface of Salmonella. The expression of RGD4C peptide in fusion with OmpA was identified by outer membrane protein extraction and WB detection in different mutant strains. However, flow cytometry analysis following immunofluorescence staining demonstrated that the extracellular length of Salmonella LPS did affect the surface display of RGD4C peptide. The strain D2-RGD4C that synthesized intact LPS including lipid A, core oligosaccharides and O antigen polysaccharides could hardly display RGD4C peptide, showing the same fluorescence signal intensity as the strains not expressing RGD4C peptide. Among different strains, D2 ∆rfaJ-RGD4C that synthesized truncated LPS including lipid A and partial core oligosaccharides was capable of displaying RGD4C peptide most efficiently and showed the highest ability to target HUVECs expressing αV integrin and tumor tissue with abundant neovascularization. Animal experiments also demonstrated that this tumor-targeting attenuated Salmonella strain to simultaneously deliver endostatin and TRAIL, two agents with different anti-tumor activities, could significantly inhibit tumor growth and prolong mouse survival. Thus, our studies revealed that Salmonella could be genetically engineered to improve its tumor targeting via the truncation of LPS and surface display of targeting peptides, thereby eliciting superior anti-tumor effects through targeted delivery of drug molecules.
Collapse
|
28
|
Senevirathne A, Hewawaduge C, Sivasankar C, Lee JH. Prospective lipid-A altered live attenuated Salmonella Gallinarum confers protectivity, DIVA capability, safety and low endotoxicity against fowl typhoid. Vet Microbiol 2022; 274:109572. [PMID: 36113357 DOI: 10.1016/j.vetmic.2022.109572] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Revised: 09/06/2022] [Accepted: 09/08/2022] [Indexed: 10/31/2022]
Abstract
The present study describes creating an attenuated Salmonella Gallinarum (SG) strain with reduced endotoxicity to prevent fowl typhoid. The strain was attenuated by deleting the lon, cpxR, and rfaL virulence-related genes. Endotoxicity was reduced by deleting the pagL open reading frame and replacing it with the lpxE gene derived from Francisella tularencis. Both events, (1) deletion of the pagL and (2) introduction of the lpxE genes, conferred reduced endotoxicity by detoxifying the lipid A structure. The detoxified SG strain (SGVSdt) was well tolerated in 7-day-old chicks when administered orally at 1 × 108 CFU/bird and in 14-day-old birds administered 1 × 107 CFU/bird subcutaneously. Parenteral immunization of detoxified vaccine strain was completely safe in birds and free of environmental contamination. Subcutaneous immunization conferred disease protection and induced humoral and cell-mediated immune responses marked by Th1-skewed patterns similar to those produced by the commercial SG9R vaccine strain. Compared with the SG9R-based vaccine, the SGVSdt construct generated significantly fewer inflammatory TNF-α responses while significantly inducing IFN-γ cytokine levels as an indication of an adaptive antibacterial response. The differentiating infected from vaccinated animals (DIVA) capability was on par with the predecessor SGVS. This study presents an appealing biological strategy to minimize lipid A-mediated endotoxicity without compromising protective efficacy against the SG challenge. Reduced endotoxicity permits the utilization of higher inoculation doses to maximize protection against fowl typhoid.
Collapse
Affiliation(s)
- Amal Senevirathne
- College of Veterinary Medicine, Jeonbuk National University, Iksan Campus, 54596, Republic of Korea
| | - Chamith Hewawaduge
- College of Veterinary Medicine, Jeonbuk National University, Iksan Campus, 54596, Republic of Korea
| | - Chandran Sivasankar
- College of Veterinary Medicine, Jeonbuk National University, Iksan Campus, 54596, Republic of Korea
| | - John Hwa Lee
- College of Veterinary Medicine, Jeonbuk National University, Iksan Campus, 54596, Republic of Korea.
| |
Collapse
|
29
|
Senevirathne A, Hewawaduge C, Lee JH. Assessing an O-antigen deficient, live attenuated Salmonella Gallinarium strain that is DIVA compatible, environmentally safe, and protects chickens against fowl typhoid. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2022; 133:104433. [PMID: 35568244 DOI: 10.1016/j.dci.2022.104433] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 04/25/2022] [Accepted: 05/06/2022] [Indexed: 06/15/2023]
Abstract
The objective of the present study was to create a highly attenuated, safe Salmonella Gallinarium (SG) vaccine strain for chicken vaccination against fowl typhoid (FT) diseases. The SG vaccine strain (SGVS) consists of three virulence-related gene deletions, namely, lon, cpxR, and rfaL. The parent strain (SGPS) with Δlon ΔcpxR genotype was utilized as the host strain for in-frame rfaL gene deletion by lambda red recombination. The SGVS was highly attenuated with improved environmental safety by zero fecal contamination beyond seven days for both oral and intramuscular immunization routes. Upon inoculation into 1-month-old young chicken, no vaccine-induced adverse behaviors were observed and did not cause a chronic state of infection as the SG wild-type strain did. Immunization of chicken elicited both humoral and cell-mediated immune responses demarcated by, IgY antibody assessment, T-cell responses in peripheral blood mononuclear cells, and the induction of immunomodulatory cytokines, IFN-γ, IL-2, IL-12, and IL-4 to resemble both Th1 and Th2 type of immune responses. The immunological assessment revealed a high level of efficacy of the SGVS when inoculated via the IM route than the oral route. The strain was less cytotoxic with reduced cytotoxicity on chicken macrophages and was DIVA capable with minimum reactivity of immunized serum with purified SG lipopolysaccharides. The challenge study could generate 70% protection in chicken for SGVS, whereas no birds were protected in the PBS challenged group. The protection levels were evident in histopathological assessment of spleen and liver specimens and also the external appearance of the spleen with reduced lesions on immunized groups. Further experiments may be warranted to dose and route optimization for further increase in the protection level derived by present SGVS.
Collapse
Affiliation(s)
- Amal Senevirathne
- College of Veterinary Medicine, Jeonbuk National University, Iksan Campus, 54596, South Korea
| | - Chamith Hewawaduge
- College of Veterinary Medicine, Jeonbuk National University, Iksan Campus, 54596, South Korea
| | - John Hwa Lee
- College of Veterinary Medicine, Jeonbuk National University, Iksan Campus, 54596, South Korea.
| |
Collapse
|
30
|
Liang K, Zhang R, Luo H, Zhang J, Tian Z, Zhang X, Zhang Y, Ali MK, Kong Q. Optimized Attenuated Salmonella Typhimurium Suppressed Tumor Growth and Improved Survival in Mice. Front Microbiol 2022; 12:774490. [PMID: 35003007 PMCID: PMC8733734 DOI: 10.3389/fmicb.2021.774490] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 12/02/2021] [Indexed: 01/03/2023] Open
Abstract
The gram-negative facultative anaerobic bacteria Salmonella enterica serovar Typhimurium (hereafter S. Typhimurium) has always been considered as one candidate of anti-tumor agents or vectors for delivering drug molecules. In this study, we compared several widely studied S. Typhimurium strains in their anti-tumor properties aiming to screen out the best one for further optimization and use in cancer therapy. In terms of the motility, virulence and anti-tumor efficacy, the three strains 14028, SL1344, and UK-1 were similar and obviously better than LT-2, and UK-1 showed the best phenotypes among them. Therefore, the strain UK-1 (D) was selected for the following studies. Its auxotrophic mutant strain (D1) harboring ∆aroA and ∆purM mutations was further optimized through the modification of lipid A structure, generating a new strain named D2 with stronger immunostimulatory activity. Finally, the ∆asd derivative of D2 was utilized as one live vector to deliver anti-tumor molecules including the angiogenesis inhibitor endostatin and apoptosis inducer TRAIL and the therapeutic and toxic-side effects were evaluated in mouse models of colon carcinoma and melanoma. After intraperitoneal infection, engineered Salmonella bacteria equipped with endostatin and/or TRAIL significantly suppressed the tumor growth and prolonged survival of tumor-bearing mice compared to PBS or bacteria carrying the empty plasmid. Consistently, immunohistochemical studies confirmed the colonization of Salmonella bacteria and the expression of anti-tumor molecules inside tumor tissue, which were accompanied by the increase of cell apoptosis and suppression of tumor angiogenesis. These results demonstrated that the beneficial anti-tumor efficacy of attenuated S. Typhimurium bacteria could be improved through delivery of drug molecules with powerful anti-tumor activities.
Collapse
Affiliation(s)
- Kang Liang
- College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Rui Zhang
- College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Haiyan Luo
- College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Jinlong Zhang
- College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Zhenyuan Tian
- College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Xiaofen Zhang
- College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Yulin Zhang
- College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Md Kaisar Ali
- College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Qingke Kong
- College of Veterinary Medicine, Southwest University, Chongqing, China
| |
Collapse
|
31
|
Mandal RK, Jiang T, Kwon YM. Genetic Determinants in Salmonella enterica Serotype Typhimurium Required for Overcoming In Vitro Stressors in the Mimicking Host Environment. Microbiol Spectr 2021; 9:e0015521. [PMID: 34878334 PMCID: PMC8653844 DOI: 10.1128/spectrum.00155-21] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 10/29/2021] [Indexed: 11/20/2022] Open
Abstract
Salmonella enterica serotype Typhimurium, a nontyphoidal Salmonella (NTS), results in a range of enteric diseases, representing a major disease burden worldwide. There is still a significant portion of Salmonella genes whose mechanistic basis to overcome host innate defense mechanisms largely remains unknown. Here, we have applied transposon insertion sequencing (Tn-seq) method to unveil the genetic factors required for the growth or survival of S. Typhimurium under various host stressors simulated in vitro. A highly saturating Tn5 library of S. Typhimurium 14028s was subjected to selection during growth in the presence of short-chain fatty acid (100 mM propionate), osmotic stress (3% NaCl), or oxidative stress (1 mM H2O2) or survival in extreme acidic pH (30 min in pH 3) or starvation (12 days in 1× phosphate-buffered saline [PBS]). We have identified a total of 339 conditionally essential genes (CEGs) required to overcome at least one of these conditions mimicking host insults. Interestingly, all eight genes encoding FoF1-ATP synthase subunit proteins were required for fitness in all five stresses. Intriguingly, a total of 88 genes in Salmonella pathogenicity islands (SPI), including SPI-1, SPI-2, SPI-3, SPI-5, SPI-6, and SPI-11, are also required for fitness under the in vitro conditions. Additionally, by comparative analysis of the genes identified in this study and the genes previously shown to be required for in vivo fitness, we identified novel genes (marBCT, envF, barA, hscA, rfaQ, rfbI, and the genes encoding putative proteins STM14_1138, STM14_3334, STM14_4825, and STM_5184) that have compelling potential for the development of vaccines and antibacterial drugs to curb Salmonella infection. IMPORTANCE Salmonella enterica serotype Typhimurium is a major human bacterial pathogen that enters the food chain through meat animals asymptomatically carrying this pathogen. Despite the rich genome sequence data, a significant portion of Salmonella genes remain to be characterized for their potential contributions to virulence. In this study, we used transposon insertion sequencing (Tn-seq) to elucidate the genetic factors required for growth or survival under various host stressors, including short-chain fatty acids, osmotic stress, oxidative stress, extreme acid, and starvation. Among the total of 339 conditionally essential genes (CEGs) that are required under at least one of these five stress conditions were 221 previously known virulence genes required for in vivo fitness during infection in at least one of four animal species, including mice, chickens, pigs, and cattle. This comprehensive map of virulence phenotype-genotype in S. Typhimurium provides a roadmap for further interrogation of the biological functions encoded by the genome of this important human pathogen to survive in hostile host environments.
Collapse
Affiliation(s)
- Rabindra K. Mandal
- Center of Excellence for Poultry Science, University of Arkansas System Division of Agriculture, Fayetteville, Arkansas, USA
| | - Tieshan Jiang
- Center of Excellence for Poultry Science, University of Arkansas System Division of Agriculture, Fayetteville, Arkansas, USA
| | - Young Min Kwon
- Center of Excellence for Poultry Science, University of Arkansas System Division of Agriculture, Fayetteville, Arkansas, USA
- Cell and Molecular Biology Program, University of Arkansas, Fayetteville, Arkansas, USA
| |
Collapse
|
32
|
Adler BA, Kazakov AE, Zhong C, Liu H, Kutter E, Lui LM, Nielsen TN, Carion H, Deutschbauer AM, Mutalik VK, Arkin AP. The genetic basis of phage susceptibility, cross-resistance and host-range in Salmonella. MICROBIOLOGY (READING, ENGLAND) 2021; 167. [PMID: 34910616 PMCID: PMC8744999 DOI: 10.1099/mic.0.001126] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Though bacteriophages (phages) are known to play a crucial role in bacterial fitness and virulence, our knowledge about the genetic basis of their interaction, cross-resistance and host-range is sparse. Here, we employed genome-wide screens in Salmonella enterica serovar Typhimurium to discover host determinants involved in resistance to eleven diverse lytic phages including four new phages isolated from a therapeutic phage cocktail. We uncovered 301 diverse host factors essential in phage infection, many of which are shared between multiple phages demonstrating potential cross-resistance mechanisms. We validate many of these novel findings and uncover the intricate interplay between RpoS, the virulence-associated general stress response sigma factor and RpoN, the nitrogen starvation sigma factor in phage cross-resistance. Finally, the infectivity pattern of eleven phages across a panel of 23 genome sequenced Salmonella strains indicates that additional constraints and interactions beyond the host factors uncovered here define the phage host range.
Collapse
Affiliation(s)
- Benjamin A Adler
- The UC Berkeley-UCSF Graduate Program in Bioengineering, Berkeley, California, USA.,Department of Bioengineering, University of California, Berkeley, Berkeley, California, USA.,Innovative Genomics Institute, University of California, Berkeley, California, USA
| | - Alexey E Kazakov
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, California, USA
| | - Crystal Zhong
- Department of Bioengineering, University of California, Berkeley, Berkeley, California, USA
| | - Hualan Liu
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, California, USA
| | | | - Lauren M Lui
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, California, USA
| | - Torben N Nielsen
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, California, USA
| | - Heloise Carion
- Department of Bioengineering, University of California, Berkeley, Berkeley, California, USA
| | - Adam M Deutschbauer
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, California, USA.,Department of Plant and Microbial Biology, University of California, Berkeley, Berkeley, California, USA
| | - Vivek K Mutalik
- Innovative Genomics Institute, University of California, Berkeley, California, USA.,Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, California, USA
| | - Adam P Arkin
- Department of Bioengineering, University of California, Berkeley, Berkeley, California, USA.,Innovative Genomics Institute, University of California, Berkeley, California, USA.,Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, California, USA
| |
Collapse
|
33
|
Kang X, Yang Y, Meng C, Wang X, Liu B, Geng S, Jiao X, Pan Z. Safety and protective efficacy of Salmonella Pullorum spiC and rfaH deletion rough mutant as a live attenuated DIVA vaccine candidate. Poult Sci 2021; 101:101655. [PMID: 34991038 PMCID: PMC8743217 DOI: 10.1016/j.psj.2021.101655] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 11/09/2021] [Accepted: 11/09/2021] [Indexed: 11/19/2022] Open
Abstract
Salmonella enterica serovar Pullorum (S. Pullorum) causes pullorum disease (PD), which is an acute systemic disease, in chickens, and leads to serious economic losses in many developing countries because of its high morbidity and mortality rate in young chicks. The live-attenuated vaccine is considered to be an effective measure to control the Salmonella infection. In addition, the DIVA (differentiation of infected and vaccinated animals) feature without the interference of serological monitoring of Salmonella infection is an important consideration in the development of the Salmonella vaccine. In this study, we evaluated the immunogenicity and protective efficacy of a S. Pullorum rough mutant S06004ΔspiCΔrfaH as a live attenuated DIVA vaccine candidate in chickens. The S06004ΔspiCΔrfaH exhibited a significant rough lipopolysaccharides (LPS) phenotype which was agglutinated with the acriflavine, not with the O9 mono antibody. Compared to the wild-type, 50% lethal dose (LD50) of the rough mutant increased 100-fold confirmed its attenuation. The mutant strain also showed a decreased bacterial colonization in the spleen and liver. The immunization with the mutant strain had no effect on the body weight and no tissue lesions were observed in the liver and spleen. The high level of the S. Pullorum-specific IgG titers in the serum indicated that significant humoral immune responses were induced in the immunization group. The cellular immune responses were also elicited from the analysis of lymphocyte proliferation and expression of cytokines in the spleen. In addition, the S06004ΔspiCΔrfaH immunized group exhibited a negative response for the serological test, while the wild-type S06004 infection group was strongly positive for the serological test showing a DIVA capability. The survival rates in the vaccinated chickens were 87% after intramuscular challenge with wild-type S. Pullorum, while the survival rates were 20% in the control groups. Overall, these results have demonstrated that the rough mutant S06004ΔspiCΔrfaH strain can be developed as an efficient live attenuated DIVA vaccine candidate to control the systemic S. Pullorum infection without the interference of salmonellosis monitoring program in poultry.
Collapse
Affiliation(s)
- Xilong Kang
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, Jiangsu, China; Jiangsu Co-innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, Jiangsu, China; Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, MOA, Yangzhou University, Yangzhou, Jiangsu, China; Joint International Research Laboratory of Agriculture and Agri-product Safety of the Ministry of Education, Yangzhou University, Yangzhou, China
| | - Yang Yang
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, Jiangsu, China; Jiangsu Co-innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, Jiangsu, China; Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, MOA, Yangzhou University, Yangzhou, Jiangsu, China; Joint International Research Laboratory of Agriculture and Agri-product Safety of the Ministry of Education, Yangzhou University, Yangzhou, China
| | - Chuang Meng
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, Jiangsu, China; Jiangsu Co-innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, Jiangsu, China; Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, MOA, Yangzhou University, Yangzhou, Jiangsu, China; Joint International Research Laboratory of Agriculture and Agri-product Safety of the Ministry of Education, Yangzhou University, Yangzhou, China
| | - Xinwei Wang
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, Jiangsu, China; Jiangsu Co-innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, Jiangsu, China; Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, MOA, Yangzhou University, Yangzhou, Jiangsu, China; Joint International Research Laboratory of Agriculture and Agri-product Safety of the Ministry of Education, Yangzhou University, Yangzhou, China
| | - Bowen Liu
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, Jiangsu, China; Jiangsu Co-innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, Jiangsu, China; Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, MOA, Yangzhou University, Yangzhou, Jiangsu, China; Joint International Research Laboratory of Agriculture and Agri-product Safety of the Ministry of Education, Yangzhou University, Yangzhou, China
| | - Shizhong Geng
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, Jiangsu, China; Jiangsu Co-innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, Jiangsu, China; Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, MOA, Yangzhou University, Yangzhou, Jiangsu, China; Joint International Research Laboratory of Agriculture and Agri-product Safety of the Ministry of Education, Yangzhou University, Yangzhou, China
| | - Xinan Jiao
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, Jiangsu, China; Jiangsu Co-innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, Jiangsu, China; Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, MOA, Yangzhou University, Yangzhou, Jiangsu, China; Joint International Research Laboratory of Agriculture and Agri-product Safety of the Ministry of Education, Yangzhou University, Yangzhou, China
| | - Zhiming Pan
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, Jiangsu, China; Jiangsu Co-innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, Jiangsu, China; Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, MOA, Yangzhou University, Yangzhou, Jiangsu, China; Joint International Research Laboratory of Agriculture and Agri-product Safety of the Ministry of Education, Yangzhou University, Yangzhou, China.
| |
Collapse
|
34
|
Zhang JF, Shang K, Wei B, Lee YJ, Park JY, Jang HK, Cha SY, Kang M. Evaluation of Safety and Protective Efficacy of a waaJ and spiC Double Deletion Korean Epidemic Strain of Salmonella enterica Serovar Gallinarum. Front Vet Sci 2021; 8:756123. [PMID: 34869728 PMCID: PMC8635151 DOI: 10.3389/fvets.2021.756123] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 10/11/2021] [Indexed: 11/16/2022] Open
Abstract
With an aim to develop a highly attenuated and strongly immunogenic distinguishable vaccine candidate, a waaJ (a gene involved in the synthesis of lipopolysaccharide) and spiC (a virulence gene) double deletion Korean epidemic strain of S. enterica ser. Gallinarum (SG005) was constructed. Our results showed that the growth and biochemical characteristics were not altered by this double deletion. The double deletion strain contained dual markers. One was a bacteriological marker (rough phenotype) and the other was a serological marker helping distinguish infected chickens from vaccinated chickens. The double deletion strain showed good genetic stability and reduced resistance to environmental stresses in vitro; furthermore, it was extremely safe and highly avirulent in broilers. Single intramuscular or oral immunization of 7-day-old broilers with the double deletion strain could stimulate the body to produce antibody levels similar to the conventional vaccine strain SG9R. In addition, against a lethal wild-type challenge, it conferred effective protection that was comparable to that seen in the group vaccinated with SG9R. In conclusion, this double deletion strain may be an effective vaccine candidate for controlling S. enterica ser. Gallinarum infection in broilers.
Collapse
Affiliation(s)
- Jun-Feng Zhang
- Department of Veterinary Infectious Diseases and Avian Diseases, College of Veterinary Medicine and Center for Poultry Diseases Control, Jeonbuk National University, Iksan, South Korea
| | - Ke Shang
- Department of Veterinary Infectious Diseases and Avian Diseases, College of Veterinary Medicine and Center for Poultry Diseases Control, Jeonbuk National University, Iksan, South Korea
| | - Bai Wei
- Department of Veterinary Infectious Diseases and Avian Diseases, College of Veterinary Medicine and Center for Poultry Diseases Control, Jeonbuk National University, Iksan, South Korea
| | - Yea-Jin Lee
- Department of Veterinary Infectious Diseases and Avian Diseases, College of Veterinary Medicine and Center for Poultry Diseases Control, Jeonbuk National University, Iksan, South Korea
| | - Jong-Yeol Park
- Department of Veterinary Infectious Diseases and Avian Diseases, College of Veterinary Medicine and Center for Poultry Diseases Control, Jeonbuk National University, Iksan, South Korea
| | - Hyung-Kwan Jang
- Department of Veterinary Infectious Diseases and Avian Diseases, College of Veterinary Medicine and Center for Poultry Diseases Control, Jeonbuk National University, Iksan, South Korea
| | - Se-Yeoun Cha
- Department of Veterinary Infectious Diseases and Avian Diseases, College of Veterinary Medicine and Center for Poultry Diseases Control, Jeonbuk National University, Iksan, South Korea
| | - Min Kang
- Department of Veterinary Infectious Diseases and Avian Diseases, College of Veterinary Medicine and Center for Poultry Diseases Control, Jeonbuk National University, Iksan, South Korea
| |
Collapse
|
35
|
Rahimi-Midani A, Kim MJ, Choi TJ. Identification of a Cupin Protein Gene Responsible for Pathogenicity, Phage Susceptibility and LPS Synthesis of Acidovorax citrulli. THE PLANT PATHOLOGY JOURNAL 2021; 37:555-565. [PMID: 34897248 PMCID: PMC8666233 DOI: 10.5423/ppj.oa.08.2021.0134] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 10/12/2021] [Indexed: 05/12/2023]
Abstract
Bacteriophages infecting Acidovorax citrulli, the causal agent of bacterial fruit blotch, have been proven to be effective for the prevention and control of this disease. However, the occurrence of bacteriophage-resistant bacteria is one of hurdles in phage biocontrol and the understanding of phage resistance in this bacterium is an essential step. In this study, we aim to investigate possible phage resistance of A. citrulli and relationship between phage resistance and pathogenicity, and to isolate and characterize the genes involved in these phenomena. A phage-resistant and less-virulent mutant named as AC-17-G1 was isolated among 3,264 A. citrulli Tn5 mutants through serial spot assays and plaque assays followed by pathogenicity test using seed coating method. The mutant has the integrated Tn5 in the middle of a cupin protein gene. This mutant recovered its pathogenicity and phage sensitivity by complementation with corresponding wild-type gene. Site-directed mutation of this gene from wild-type by CRISPR/Cas9 system resulted in the loss of pathogenicity and acquisition of phage resistance. The growth of AC-17-G1 in King's B medium was much less than the wild-type, but the growth turned into normal in the medium supplemented with D-mannose 6-phosphate or D-fructose 6-phosphate indicating the cupin protein functions as a phosphomannos isomerase. Sodium dodecyl sulfa analysis of lipopolysaccharide (LPS) extracted from the mutant was smaller than that from wild-type. All these data suggest that the cupin protein is a phosphomannos isomerase involved in LPS synthesis, and LPS is an important determinant of pathogenicity and phage susceptibility of A. citrulli.
Collapse
Affiliation(s)
| | - Min-Jung Kim
- Department of Microbiology, Pukyong National University, Busan 48513,
Korea
| | - Tae-Jin Choi
- Department of Microbiology, Pukyong National University, Busan 48513,
Korea
- Division of Marine Biosciences, Pukyong National University, Busan 48513,
Korea
- Corresponding author: Phone) +82-51-620-6367, FAX) +82-51-611-6358, E-mail)
| |
Collapse
|
36
|
Nothaft H, Bian X, Shajahan A, Miller WG, Bolick DT, Guerrant RL, Azadi P, Ng KKS, Szymanski CM. Detecting Glucose Fluctuations in the Campylobacter jejuni N-Glycan Structure. ACS Chem Biol 2021; 16:2690-2701. [PMID: 34726367 DOI: 10.1021/acschembio.1c00498] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Campylobacter jejuni is a significant cause of human gastroenteritis worldwide, and all strains express an N-glycan that is added to at least 80 different proteins. We characterized 98 C. jejuni isolates from infants from 7 low- and middle-income countries and identified 4 isolates unreactive with our N-glycan-specific antiserum that was raised against the C. jejuni heptasaccharide composed of GalNAc-GalNAc-GalNAc(Glc)-GalNAc-GalNAc-diNAcBac. Mass spectrometric analyses indicated these isolates express a hexasaccharide lacking the glucose branch. Although all 4 strains encode the PglI glucosyltransferase (GlcTF), one aspartate in the DXDD motif was missing, an alteration also present in ∼4% of all available PglI sequences. Deleting this residue from an active PglI resulted in a nonfunctional GlcTF when the protein glycosylation system was reconstituted in E. coli, while replacement with Glu/Ala was not deleterious. Molecular modeling proposed a mechanism for how the DXDD residues and the structure/length beyond the motif influence activity. Mouse vaccination with an E. coli strain expressing the full-length heptasaccharide produced N-glycan-specific antibodies and a corresponding reduction in Campylobacter colonization and weight loss following challenge. However, the antibodies did not recognize the hexasaccharide and were unable to opsonize C. jejuni isolates lacking glucose, suggesting this should be considered when designing N-glycan-based vaccines to prevent campylobacteriosis.
Collapse
Affiliation(s)
- Harald Nothaft
- Department of Medical Microbiology and Immunology, University of Alberta, Katz Group Centre, Edmonton, Alberta T6G 2E9, Canada
| | - Xiaoming Bian
- Department of Microbiology, University of Georgia, 527 Biological Sciences Building, Athens, Georgia 30602, United States
- Complex Carbohydrate Research Center, University of Georgia, 315 Riverbend Road, Athens, Georgia 30602, United States
| | - Asif Shajahan
- Complex Carbohydrate Research Center, University of Georgia, 315 Riverbend Road, Athens, Georgia 30602, United States
| | - William G. Miller
- Produce Safety and Microbiology Research Unit, Agricultural Research Service, United States Department of Agriculture, 800 Buchanan Street, Albany, California 94710, United States
| | - David T. Bolick
- Center for Global Health Equity, Division of Infectious Diseases and International Health, University of Virginia School of Medicine, Charlottesville, Virginia 22908, United States
| | - Richard L. Guerrant
- Center for Global Health Equity, Division of Infectious Diseases and International Health, University of Virginia School of Medicine, Charlottesville, Virginia 22908, United States
| | - Parastoo Azadi
- Complex Carbohydrate Research Center, University of Georgia, 315 Riverbend Road, Athens, Georgia 30602, United States
| | - Kenneth K. S. Ng
- Department of Chemistry and Biochemistry, University of Windsor, 401 Sunset Avenue, Windsor, Ontario N9B 3P4, Canada
| | - Christine M. Szymanski
- Department of Medical Microbiology and Immunology, University of Alberta, Katz Group Centre, Edmonton, Alberta T6G 2E9, Canada
- Department of Microbiology, University of Georgia, 527 Biological Sciences Building, Athens, Georgia 30602, United States
- Complex Carbohydrate Research Center, University of Georgia, 315 Riverbend Road, Athens, Georgia 30602, United States
| |
Collapse
|
37
|
A triple-sugar regulated Salmonella vaccine protects against Clostridium perfringens-induced necrotic enteritis in broiler chickens. Poult Sci 2021; 101:101592. [PMID: 34922043 PMCID: PMC8686071 DOI: 10.1016/j.psj.2021.101592] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 11/02/2021] [Accepted: 11/04/2021] [Indexed: 02/08/2023] Open
Abstract
Gram-positive Clostridium perfringens type G, the causative agent of necrotic enteritis (NE), has gained more attention in the poultry industry due to governmental restrictions on the use of growth-promoting antibiotics in poultry feed. Our previous work has proved that regulated delayed lysis Salmonella vaccines delivering a plasmid encoding an operon fusion of the nontoxic C-terminal adhesive part of alpha toxin and a GST-NetB toxin fusion were able to elicit significant protective immunity in broilers against C. perfringens challenge. We recently improved our S. Typhimurium antigen delivery vaccine strain by integrating a rhamnose-regulated O-antigen synthesis gene enabling a triple-sugar regulation system to control virulence, antigen-synthesis and lysis in vivo traits. The strain also includes a ΔsifA mutation that was previously shown to increase the immunogenicity of and level of protective immunity induced by Salmonella vectored influenza and Eimeria antigens. The new antigen-delivery vaccine vector system confers on the vaccine strain a safe profile and improved protection against C. perfringens challenge. The strain with the triple-sugar regulation system delivering a regulated lysis plasmid pG8R220 encoding the PlcC and GST-NetB antigens protected chickens at a similar level observed in antibiotic-treated chickens. Feed conversion and growth performance were also similar to antibiotic-treated chickens. These studies made use of a severe C. perfringens challenge with lesion formation and mortality enhanced by pre-exposure to Eimeria maxima oocysts. The vaccine achieved effectiveness through three different immunization routes, oral, spray and in drinking water. The vaccine has a potential for application in commercial hatcher and broiler-rearing conditions.
Collapse
|
38
|
Lobato-Márquez D, Xu J, Güler GÖ, Ojiakor A, Pilhofer M, Mostowy S. Mechanistic insight into bacterial entrapment by septin cage reconstitution. Nat Commun 2021; 12:4511. [PMID: 34301939 PMCID: PMC8302635 DOI: 10.1038/s41467-021-24721-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 07/01/2021] [Indexed: 11/22/2022] Open
Abstract
Septins are cytoskeletal proteins that assemble into hetero-oligomeric complexes and sense micron-scale membrane curvature. During infection with Shigella flexneri, an invasive enteropathogen, septins restrict actin tail formation by entrapping bacteria in cage-like structures. Here, we reconstitute septin cages in vitro using purified recombinant septin complexes (SEPT2-SEPT6-SEPT7), and study how these recognize bacterial cells and assemble on their surface. We show that septin complexes recognize the pole of growing Shigella cells. An amphipathic helix domain in human SEPT6 enables septins to sense positively curved membranes and entrap bacterial cells. Shigella strains lacking lipopolysaccharide components are more efficiently entrapped in septin cages. Finally, cryo-electron tomography of in vitro cages reveals how septins assemble as filaments on the bacterial cell surface. Septins are cytoskeletal proteins that assemble into complexes and contribute to immunity by entrapping intracellular bacteria in cage-like structures. Here, Lobato-Márquez et al. reconstitute septin cages in vitro using purified recombinant complexes, and study how these recognize bacterial cells and assemble as filaments on their surface.
Collapse
Affiliation(s)
- Damián Lobato-Márquez
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, UK.
| | - Jingwei Xu
- Department of Biology, Institute of Molecular Biology and Biophysics, Eidgenössische Technische Hochschule Zürich, Zürich, Switzerland
| | - Gizem Özbaykal Güler
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, UK
| | - Adaobi Ojiakor
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, UK
| | - Martin Pilhofer
- Department of Biology, Institute of Molecular Biology and Biophysics, Eidgenössische Technische Hochschule Zürich, Zürich, Switzerland
| | - Serge Mostowy
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, UK.
| |
Collapse
|
39
|
Gaudet RG, Zhu S, Halder A, Kim BH, Bradfield CJ, Huang S, Xu D, Mamiñska A, Nguyen TN, Lazarou M, Karatekin E, Gupta K, MacMicking JD. A human apolipoprotein L with detergent-like activity kills intracellular pathogens. Science 2021; 373:eabf8113. [PMID: 34437126 PMCID: PMC8422858 DOI: 10.1126/science.abf8113] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 04/29/2021] [Accepted: 06/03/2021] [Indexed: 12/11/2022]
Abstract
Activation of cell-autonomous defense by the immune cytokine interferon-γ (IFN-γ) is critical to the control of life-threatening infections in humans. IFN-γ induces the expression of hundreds of host proteins in all nucleated cells and tissues, yet many of these proteins remain uncharacterized. We screened 19,050 human genes by CRISPR-Cas9 mutagenesis and identified IFN-γ-induced apolipoprotein L3 (APOL3) as a potent bactericidal agent protecting multiple non-immune barrier cell types against infection. Canonical apolipoproteins typically solubilize mammalian lipids for extracellular transport; APOL3 instead targeted cytosol-invasive bacteria to dissolve their anionic membranes into human-bacterial lipoprotein nanodiscs detected by native mass spectrometry and visualized by single-particle cryo-electron microscopy. Thus, humans have harnessed the detergent-like properties of extracellular apolipoproteins to fashion an intracellular lysin, thereby endowing resident nonimmune cells with a mechanism to achieve sterilizing immunity.
Collapse
Affiliation(s)
- Ryan G Gaudet
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT 06510, USA
- Yale Systems Biology Institute, West Haven, CT 06477, USA
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06510, USA
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Shiwei Zhu
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT 06510, USA
- Yale Systems Biology Institute, West Haven, CT 06477, USA
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06510, USA
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Anushka Halder
- Yale Nanobiology Institute, West Haven, CT 06477, USA
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Bae-Hoon Kim
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT 06510, USA
- Yale Systems Biology Institute, West Haven, CT 06477, USA
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06510, USA
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Clinton J Bradfield
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT 06510, USA
- Yale Systems Biology Institute, West Haven, CT 06477, USA
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06510, USA
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Shuai Huang
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT 06510, USA
- Yale Systems Biology Institute, West Haven, CT 06477, USA
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06510, USA
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Dijin Xu
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT 06510, USA
- Yale Systems Biology Institute, West Haven, CT 06477, USA
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06510, USA
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Agnieszka Mamiñska
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT 06510, USA
- Yale Systems Biology Institute, West Haven, CT 06477, USA
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06510, USA
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Thanh Ngoc Nguyen
- Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Melbourne 3800, Australia
| | - Michael Lazarou
- Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Melbourne 3800, Australia
| | - Erdem Karatekin
- Yale Nanobiology Institute, West Haven, CT 06477, USA
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT 06510, USA
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06510, USA
- Saints-Pères Paris Institute for the Neurosciences, Centre National de la Recherche Scientifique (CNRS), Université de Paris, F-75006 Paris, France
| | - Kallol Gupta
- Yale Nanobiology Institute, West Haven, CT 06477, USA
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06510, USA
| | - John D MacMicking
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT 06510, USA.
- Yale Systems Biology Institute, West Haven, CT 06477, USA
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06510, USA
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT 06510, USA
| |
Collapse
|
40
|
Diard M, Bakkeren E, Lentsch V, Rocker A, Bekele NA, Hoces D, Aslani S, Arnoldini M, Böhi F, Schumann-Moor K, Adamcik J, Piccoli L, Lanzavecchia A, Stadtmueller BM, Donohue N, van der Woude MW, Hockenberry A, Viollier PH, Falquet L, Wüthrich D, Bonfiglio F, Loverdo C, Egli A, Zandomeneghi G, Mezzenga R, Holst O, Meier BH, Hardt WD, Slack E. A rationally designed oral vaccine induces immunoglobulin A in the murine gut that directs the evolution of attenuated Salmonella variants. Nat Microbiol 2021; 6:830-841. [PMID: 34045711 PMCID: PMC7611113 DOI: 10.1038/s41564-021-00911-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Accepted: 04/14/2021] [Indexed: 12/12/2022]
Abstract
The ability of gut bacterial pathogens to escape immunity by antigenic variation-particularly via changes to surface-exposed antigens-is a major barrier to immune clearance1. However, not all variants are equally fit in all environments2,3. It should therefore be possible to exploit such immune escape mechanisms to direct an evolutionary trade-off. Here, we demonstrate this phenomenon using Salmonella enterica subspecies enterica serovar Typhimurium (S.Tm). A dominant surface antigen of S.Tm is its O-antigen: a long, repetitive glycan that can be rapidly varied by mutations in biosynthetic pathways or by phase variation4,5. We quantified the selective advantage of O-antigen variants in the presence and absence of O-antigen-specific immunoglobulin A and identified a set of evolutionary trajectories allowing immune escape without an associated fitness cost in naive mice. Through the use of rationally designed oral vaccines, we induced immunoglobulin A responses blocking all of these trajectories. This selected for Salmonella mutants carrying deletions of the O-antigen polymerase gene wzyB. Due to their short O-antigen, these evolved mutants were more susceptible to environmental stressors (detergents or complement) and predation (bacteriophages) and were impaired in gut colonization and virulence in mice. Therefore, a rationally induced cocktail of intestinal antibodies can direct an evolutionary trade-off in S.Tm. This lays the foundations for the exploration of mucosal vaccines capable of setting evolutionary traps as a prophylactic strategy.
Collapse
Affiliation(s)
- Médéric Diard
- Biozentrum, University of Basel, Basel, Switzerland.
| | - Erik Bakkeren
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland.,Department of Zoology, University of Oxford, Oxford, UK
| | - Verena Lentsch
- Institute of Food, Nutrition and Health, D-HEST, ETH Zürich, Zürich, Switzerland
| | | | | | - Daniel Hoces
- Institute of Food, Nutrition and Health, D-HEST, ETH Zürich, Zürich, Switzerland
| | - Selma Aslani
- Institute of Food, Nutrition and Health, D-HEST, ETH Zürich, Zürich, Switzerland
| | - Markus Arnoldini
- Institute of Food, Nutrition and Health, D-HEST, ETH Zürich, Zürich, Switzerland
| | - Flurina Böhi
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland.,Department of Molecular Mechanisms of Disease, University of Zürich, Zürich, Switzerland
| | - Kathrin Schumann-Moor
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland.,Division of Surgical Research, University Hospital of Zürich, Zürich, Switzerland
| | - Jozef Adamcik
- Institute of Food, Nutrition and Health, D-HEST, ETH Zürich, Zürich, Switzerland
| | - Luca Piccoli
- Institute for Research in Biomedicine, Università della Svizzera Italiana, Bellinzona, Switzerland
| | - Antonio Lanzavecchia
- Institute for Research in Biomedicine, Università della Svizzera Italiana, Bellinzona, Switzerland
| | - Beth M Stadtmueller
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Nicholas Donohue
- York Biomedical Research Institute, Hull York Medical School, University of York, York, UK.,Department of Orthopedics and Trauma, Medical University of Graz, Graz, Austria
| | - Marjan W van der Woude
- York Biomedical Research Institute, Hull York Medical School, University of York, York, UK
| | - Alyson Hockenberry
- Department of Environmental Microbiology, Eawag, Dubendorf, Switzerland.,Department of Environmental Sciences, ETH Zürich, Zürich, Switzerland
| | - Patrick H Viollier
- Microbiology and Molecular Medicine, University of Geneva, Geneva, Switzerland
| | - Laurent Falquet
- Department of Biology, University of Fribourg, Fribourg, Switzerland.,Swiss Institute of Bioinformatics, Fribourg, Switzerland
| | - Daniel Wüthrich
- Infection Biology, University Hospital of Basel, Basel, Switzerland
| | | | - Claude Loverdo
- Department of Materials, ETH Zürich, Zürich, Switzerland
| | - Adrian Egli
- Infection Biology, University Hospital of Basel, Basel, Switzerland.,Department of Biomedicine, University of Basel, Basel, Switzerland
| | | | - Raffaele Mezzenga
- Institute of Food, Nutrition and Health, D-HEST, ETH Zürich, Zürich, Switzerland.,Department of Materials, ETH Zürich, Zürich, Switzerland
| | - Otto Holst
- Forschungszentrum Borstel, Borstel, Germany
| | - Beat H Meier
- Institute for Physical Chemistry, ETH Zürich, Zürich, Switzerland
| | - Wolf-Dietrich Hardt
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland.
| | - Emma Slack
- Institute of Microbiology, Department of Biology, ETH Zürich, Zürich, Switzerland. .,Institute of Food, Nutrition and Health, D-HEST, ETH Zürich, Zürich, Switzerland.
| |
Collapse
|
41
|
Cobb J, Rawson J, Gonzalez N, Hensel M, Kandeel F, Husseiny MI. Oral Salmonella msbB Mutant as a Carrier for a Salmonella-Based Vaccine for Prevention and Reversal of Type 1 Diabetes. Front Immunol 2021; 12:667897. [PMID: 34108968 PMCID: PMC8181392 DOI: 10.3389/fimmu.2021.667897] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 05/06/2021] [Indexed: 11/18/2022] Open
Abstract
A therapy that includes an oral vaccine for type 1 diabetes (T1D) using live attenuated Salmonella MvP728 (ΔhtrA/ΔpurD), cytokines (IL10 and TGFβ) and preproinsulin (PPI) antigen in combination with a sub-therapeutic dose of anti-CD3 mAb was developed by our team. The vaccine combination therapy reduced insulitis and prevented and reversed diabetes in non-obese diabetic (NOD) mice. Here, we show the effectiveness of an alternative Salmonella mutant (ΔmsbB) as a carrier strain, which is anticipated to have lower risks of an inflammatory response and septicemia as a result of modification in the lipopolysaccharide (LPS) via detoxification of lipid A. This mutant strain proved to have highly reduced pathogenic side effects. Salmonella strain ΔmsbB expressed autoantigens and in combination with cytokines and anti-CD3 mAb, successfully prevented and reversed T1D to levels comparable to the previously used carrier strain ΔhtrA/ΔpurD. Additionally, the Salmonella msbB mutant resulted in higher rates of host cell infection. These results further demonstrate the potential of an oral Salmonella-based combined therapy in the treatment of early T1D.
Collapse
Affiliation(s)
- Jacob Cobb
- Department of Translational Research & Cellular Therapeutics, Arthur Riggs Diabetes & Metabolism Research Institute, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, United States
| | - Jeffrey Rawson
- Department of Translational Research & Cellular Therapeutics, Arthur Riggs Diabetes & Metabolism Research Institute, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, United States
| | - Nelson Gonzalez
- Department of Translational Research & Cellular Therapeutics, Arthur Riggs Diabetes & Metabolism Research Institute, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, United States
| | - Michael Hensel
- Division of Microbiology, University of Osnabrück, Osnabrück, Germany
| | - Fouad Kandeel
- Department of Translational Research & Cellular Therapeutics, Arthur Riggs Diabetes & Metabolism Research Institute, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, United States
| | - Mohamed I Husseiny
- Department of Translational Research & Cellular Therapeutics, Arthur Riggs Diabetes & Metabolism Research Institute, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, United States.,Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| |
Collapse
|
42
|
Synthesis and delivery of Streptococcus pneumoniae capsular polysaccharides by recombinant attenuated Salmonella vaccines. Proc Natl Acad Sci U S A 2021; 118:2013350118. [PMID: 33380455 PMCID: PMC7812815 DOI: 10.1073/pnas.2013350118] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Pneumococcal infection-caused diseases are responsible for substantial morbidity and mortality worldwide. Traditional pneumococcal vaccines are developed based on purified capsular polysaccharides (CPS) or CPS conjugated to a protein carrier. Production processes of the traditional vaccines are laborious, and thereby increase the vaccine cost and limit their use in developing nations. A cost-effective pneumococcal vaccine using the recombinant attenuated Salmonella vaccine (RASV) was developed in this study. We cloned and expressed genes for seven serotypes of CPSs in the RASV strain. The RASV-delivered CPSs induced robust humoral and cell-mediated responses and mediated efficient protection of mice against pneumococcal infection. Our work provides an innovative strategy for mass producing low-cost bioconjugated polysaccharide vaccines for needle-free mucosal delivery against pneumococcal infections. Streptococcus pneumoniae capsular polysaccharides (CPSs) are major determinants of bacterial pathogenicity. CPSs of different serotypes form the main components of the pneumococcal vaccines Pneumovax, Prevnar7, and Prevnar13, which substantially reduced the S. pneumoniae disease burden in developed countries. However, the laborious production processes of traditional polysaccharide-based vaccines have raised the cost of the vaccines and limited their impact in developing countries. The aim of this study is to develop a kind of low-cost live vaccine based on using the recombinant attenuated Salmonella vaccine (RASV) system to protect against pneumococcal infections. We cloned genes for seven different serotypes of CPSs to be expressed by the RASV strain. Oral immunization of mice with the RASV-CPS strains elicited robust Th1 biased adaptive immune responses. All the CPS-specific antisera mediated opsonophagocytic killing of the corresponding serotype of S. pneumoniae in vitro. The RASV-CPS2 and RASV-CPS3 strains provided efficient protection of mice against challenge infections with either S. pneumoniae strain D39 or WU2. Synthesis and delivery of S. pneumoniae CPSs using the RASV strains provide an innovative strategy for low-cost pneumococcal vaccine development, production, and use.
Collapse
|
43
|
Genome-Wide Analyses of the Temperature-Responsive Genetic Loci of the Pectinolytic Plant Pathogenic Pectobacterium atrosepticum. Int J Mol Sci 2021; 22:ijms22094839. [PMID: 34063632 PMCID: PMC8125463 DOI: 10.3390/ijms22094839] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/23/2021] [Accepted: 04/27/2021] [Indexed: 12/12/2022] Open
Abstract
Temperature is one of the critical factors affecting gene expression in bacteria. Despite the general interest in the link between bacterial phenotypes and environmental temperature, little is known about temperature-dependent gene expression in plant pathogenic Pectobacterium atrosepticum, a causative agent of potato blackleg and tuber soft rot worldwide. In this study, twenty-nine P. atrosepticum SCRI1043 thermoregulated genes were identified using Tn5-based transposon mutagenesis coupled with an inducible promotorless gusA gene as a reporter. From the pool of 29 genes, 14 were up-regulated at 18 °C, whereas 15 other genes were up-regulated at 28 °C. Among the thermoregulated loci, genes involved in primary bacterial metabolism, membrane-related proteins, fitness-corresponding factors, and several hypothetical proteins were found. The Tn5 mutants were tested for their pathogenicity in planta and for features that are likely to remain important for the pathogen to succeed in the (plant) environment. Five Tn5 mutants expressed visible phenotypes differentiating these mutants from the phenotype of the SCRI1043 wild-type strain. The gene disruptions in the Tn5 transposon mutants caused alterations in bacterial generation time, ability to form a biofilm, production of lipopolysaccharides, and virulence on potato tuber slices. The consequences of environmental temperature on the ability of P. atrosepticum to cause disease symptoms in potato are discussed.
Collapse
|
44
|
Outer Membrane Structural Defects in Salmonella enterica Serovar Typhimurium Affect Neutrophil Chemokinesis but Not Chemotaxis. mSphere 2021; 6:6/1/e01012-20. [PMID: 33627508 PMCID: PMC8544890 DOI: 10.1128/msphere.01012-20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Neutrophils, the first line of defense against pathogens, are critical in the host response to acute and chronic infections. In Gram-negative pathogens, the bacterial outer membrane (OM) is a key mediator of pathogen detection; nonetheless, the effects of variations in its molecular structure on the neutrophil migratory response to bacteria remain largely unknown. Here, we developed a quantitative microfluidic assay that precludes physical contact between bacteria and neutrophils while maintaining chemical communication, thus allowing investigation of both transient and steady-state responses of neutrophils to a library of Salmonella enterica serovar Typhimurium OM-related mutants at single-cell resolution. Using single-cell quantitative metrics, we found that transient neutrophil chemokinesis is highly gradated based upon OM structure, while transient and steady-state chemotaxis responses differ little between mutants. Based on our finding of a lack of correlation between chemokinesis and chemotaxis, we define "stimulation score" as a metric that comprehensively describes the neutrophil response to pathogens. Complemented with a killing assay, our results provide insight into how OM modifications affect neutrophil recruitment and pathogen survival. Altogether, our platform enables the discovery of transient and steady-state migratory responses and provides a new path for quantitative interrogation of cell decision-making processes in a variety of host-pathogen interactions.IMPORTANCE Our findings provide insights into the previously unexplored effects of Salmonella envelope defects on fundamental innate immune cell behavior, which advance the knowledge in pathogen-host cell biology and potentially inspire the rational design of attenuated strains for vaccines or immunotherapeutic strains for cancer therapy. Furthermore, the microfluidic assay platform and analytical tools reported herein enable high-throughput, sensitive, and quantitative screening of microbial strains' immunogenicity in vitro This approach could be particularly beneficial for rapid in vitro screening of engineered microbial strains (e.g., vaccine candidates) as the quantitative ranking of the overall strength of the neutrophil response, reported by "stimulation score," agrees with in vivo cytokine response trends reported in the literature.
Collapse
|
45
|
Wang B, Artsimovitch I. NusG, an Ancient Yet Rapidly Evolving Transcription Factor. Front Microbiol 2021; 11:619618. [PMID: 33488562 PMCID: PMC7819879 DOI: 10.3389/fmicb.2020.619618] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 12/07/2020] [Indexed: 12/19/2022] Open
Abstract
Timely and accurate RNA synthesis depends on accessory proteins that instruct RNA polymerase (RNAP) where and when to start and stop transcription. Among thousands of transcription factors, NusG/Spt5 stand out as the only universally conserved family of regulators. These proteins interact with RNAP to promote uninterrupted RNA synthesis and with diverse cellular partners to couple transcription to RNA processing, modification or translation, or to trigger premature termination of aberrant transcription. NusG homologs are present in all cells that utilize bacterial-type RNAP, from endosymbionts to plants, underscoring their ancient and essential function. Yet, in stark contrast to other core RNAP components, NusG family is actively evolving: horizontal gene transfer and sub-functionalization drive emergence of NusG paralogs, such as bacterial LoaP, RfaH, and UpxY. These specialized regulators activate a few (or just one) operons required for expression of antibiotics, capsules, secretion systems, toxins, and other niche-specific macromolecules. Despite their common origin and binding site on the RNAP, NusG homologs differ in their target selection, interacting partners and effects on RNA synthesis. Even among housekeeping NusGs from diverse bacteria, some factors promote pause-free transcription while others slow the RNAP down. Here, we discuss structure, function, and evolution of NusG proteins, focusing on unique mechanisms that determine their effects on gene expression and enable bacterial adaptation to diverse ecological niches.
Collapse
Affiliation(s)
- Bing Wang
- Department of Microbiology and the Center for RNA Biology, The Ohio State University, Columbus, OH, United States
| | - Irina Artsimovitch
- Department of Microbiology and the Center for RNA Biology, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
46
|
Senevirathne A, Hewawaduge C, Park S, Park JY, Kirthika P, Lee JH. O-antigen-deficient, live, attenuated Salmonella typhimurium confers efficient uptake, reduced cytotoxicity, and rapid clearance in chicken macrophages and lymphoid organs and induces significantly high protective immune responses that protect chickens against Salmonella infection. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2020; 111:103745. [PMID: 32470560 DOI: 10.1016/j.dci.2020.103745] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 04/26/2020] [Accepted: 05/09/2020] [Indexed: 06/11/2023]
Abstract
In the present study, we developed an O-antigen-deficient, live, attenuated Salmonella Typhimurium (ST) strain (JOL2377) and assessed its safety, macrophage toxicity, invasion into lymphoid tissues, immunogenicity, and protection against Salmonella infection in chickens. The JOL2377 induced significantly lower cytotoxicity and higher level of cytokine response in IL-2, IL-10, IL-4, and IFN- γ than the WT strain upon macrophage uptake. It did not persist in macrophages or in chicken organs and rapidly cleared without systemic infection. None of the chicken were found to secrete Salmonella in feces into the environment exacerbating its attenuation. Interestingly JOL2377 successfully arrived in immunological hot-spots such as spleen, liver and bursa of Fabricius for an efficient antigen presentation and immune stimulation. Mucosal and parenteral immunization with JOL2377 significantly elicit antigen-specific humoral (IgY) and cell mediated responses marked by peripheral blood mononuclear cell proliferation, cytokine induction, increase in T-cell responses than non-immunized control. JOL2377 did not generate significant levels of LPS specific antibodies as compared to the WT strain due to the lack of immunogenic O-antigen component from its LPS structure. Upon virulent challenge, route dependent efficacy differences were leaving the intramuscular route is superior to the oral route on reducing splenic and liver colonization of the challenge ST. The least cytotoxicity, virulence, and superior immunogenicity of JL2377 that effectively engage both humoral and IFN- γ mediated CMI responses present an ideal scenario in host immune modulation to fight against intracellular pathogen Salmonella.
Collapse
Affiliation(s)
- Amal Senevirathne
- College of Veterinary Medicine, Jeonbuk National University, Iksan Campus, 54596, Iksan, Republic of Korea
| | - Chamith Hewawaduge
- College of Veterinary Medicine, Jeonbuk National University, Iksan Campus, 54596, Iksan, Republic of Korea
| | - Sungwoo Park
- College of Veterinary Medicine, Jeonbuk National University, Iksan Campus, 54596, Iksan, Republic of Korea
| | - Ji-Young Park
- College of Veterinary Medicine, Jeonbuk National University, Iksan Campus, 54596, Iksan, Republic of Korea
| | - Perumalraja Kirthika
- College of Veterinary Medicine, Jeonbuk National University, Iksan Campus, 54596, Iksan, Republic of Korea
| | - John Hwa Lee
- College of Veterinary Medicine, Jeonbuk National University, Iksan Campus, 54596, Iksan, Republic of Korea.
| |
Collapse
|
47
|
Augustin LB, Milbauer L, Hastings SE, Leonard AS, Saltzman DA, Schottel JL. Salmonella enterica Typhimurium engineered for nontoxic systemic colonization of autochthonous tumors. J Drug Target 2020; 29:294-299. [PMID: 32886538 DOI: 10.1080/1061186x.2020.1818759] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Much of the bacterial anticancer therapy being developed relies on the ability of bacteria to specifically colonise tumours. Initial attempts to translate promising Salmonella enterica Typhimurium (S. Typhimurium) preclinical results to the clinical setting failed, primarily due to lack of tumour colonisation and the significant toxicities from systemically administered Gram-negative bacteria. To address the difference in results between preclinical experiments performed in mice with transplant tumours and clinical trials in human volunteers with autochthonous tumours, a genetically engineered mouse model of breast cancer (BALB-neuT) was utilised to develop a strain of virulence-attenuated S. Typhimurium capable of robust colonisation of autochthonous tumours. Several genes that code for bacterial surface molecules, responsible for signalling a toxic immune response against the bacteria, were mutated. The resulting S. Typhimurium strain, BCT2, allowed non-toxic intravenous administration of 3 × 106 colony forming units of bacteria in tumour-burdened mice when combined with a vascular disruption agent to induce intratumoral necrotic space and facilitate bacterial colonisation.
Collapse
Affiliation(s)
- Lance B Augustin
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - Liming Milbauer
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - Sara E Hastings
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - Arnold S Leonard
- Department of Surgery, University of Minnesota, Minneapolis, MN, USA
| | - Daniel A Saltzman
- Department of Surgery, University of Minnesota, Minneapolis, MN, USA
| | - Janet L Schottel
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
48
|
Splichal I, Rychlik I, Splichalova I, Karasova D, Splichalova A. Toll-Like Receptor 4 Signaling in the Ileum and Colon of Gnotobiotic Piglets Infected with Salmonella Typhimurium or Its Isogenic ∆ rfa Mutants. Toxins (Basel) 2020; 12:toxins12090545. [PMID: 32842482 PMCID: PMC7551901 DOI: 10.3390/toxins12090545] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 08/16/2020] [Accepted: 08/20/2020] [Indexed: 02/06/2023] Open
Abstract
Salmonella Typhimurium is a Gram-negative bacterium that causes enterocolitis in humans and pigs. Lipopolysaccharide (LPS) is a component of the outer leaflet of Gram-negative bacteria that provokes endotoxin shock. LPS can be synthesized completely or incompletely and creates S (smooth) or R (rough) chemotypes. Toll-like receptors (TLR) 2, 4, and 9 initiate an inflammatory reaction to combat bacterial infections. We associated/challenged one-week-old gnotobiotic piglets with wild-type S. Typhimurium with S chemotype or its isogenic ∆rfa mutants with R chemotype LPS. The wild-type S. Typhimurium induced TLR2 and TLR4 mRNA expression but not TLR9 mRNA expression in the ileum and colon of one-week-old gnotobiotic piglets 24 h after challenge. The TLR2 and TLR4 stimulatory effects of the S. Typhimurium ∆rfa mutants were related to the completeness of their LPS chain. The transcription of IL-12/23 p40, IFN-γ, and IL-6 in the intestine and the intestinal and plasmatic levels of IL-12/23 p40 and IL-6 but not IFN-γ were related to the activation of TLR2 and TLR4 signaling pathways. The avirulent S. Typhimurium ∆rfa mutants are potentially useful for modulation of the TLR2 and TLR4 signaling pathways to protect the immunocompromised gnotobiotic piglets against subsequent infection with the virulent S. Typhimurium.
Collapse
Affiliation(s)
- Igor Splichal
- Laboratory of Gnotobiology, Institute of Microbiology, Czech Academy of Sciences, 549 22 Novy Hradek, Czech Republic;
| | - Ivan Rychlik
- Department of Immunology, Veterinary Research Institute, 621 00 Brno, Czech Republic; (I.R.); (D.K.)
| | - Iva Splichalova
- Laboratory of Immunobiology, Institute of Molecular Genetics, Czech Academy of Sciences, 142 20 Prague 4-Krc, Czech Republic;
| | - Daniela Karasova
- Department of Immunology, Veterinary Research Institute, 621 00 Brno, Czech Republic; (I.R.); (D.K.)
| | - Alla Splichalova
- Laboratory of Gnotobiology, Institute of Microbiology, Czech Academy of Sciences, 549 22 Novy Hradek, Czech Republic;
- Correspondence: ; Tel.: +420-491-418-539
| |
Collapse
|
49
|
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
|
50
|
Liu Q, Shen X, Bian X, Kong Q. Effect of deletion of gene cluster involved in synthesis of Enterobacterial common antigen on virulence and immunogenicity of live attenuated Salmonella vaccine when delivering heterologous Streptococcus pneumoniae antigen PspA. BMC Microbiol 2020; 20:150. [PMID: 32513100 PMCID: PMC7278252 DOI: 10.1186/s12866-020-01837-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 06/01/2020] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Enterobacterial common antigen (ECA) is a family-specific surface antigen shared by all members of the Enterobacteriaceae family. Previous studies showed that the loss of ECA results in Salmonella attenuation, indicating its usefulness as a vaccine candidate for Salmonella infection, but no studies have shown whether the mutation resulting from the deletion of the ECA operon in conjunction with other mutations could be used as an antigen vehicle for heterologous protein antigen delivery. RESULTS In this study, we introduced a nonpolar, defined ECA operon deletion into wild-type S. Typhimurium χ3761 and an attenuated vaccine strain χ9241, obtaining two isogenic ECA operon mutants, namely, χ12357 and χ12358, respectively. A number of in vitro and in vivo properties of the mutants were analyzed. We found that the loss of ECA did not affect the growth, lipopolysaccharide (LPS) production and motility of S. Typhimurium wild type strain χ3761 and its attenuated vaccine strain χ9241 but significantly affected the virulence when administered orally to BALB/c mice. Furthermore, the effects of the ECA mutation on the immunogenicity of a recombinant S. Typhimurium vaccine strain χ9241 when delivering the pneumococcal antigen PspA were determined. The result showed that the total anti-PspA IgG level of χ12358 (pYA4088) was slightly lower than that of χ9241 (pYA4088), but the protection rate was not compromised. CONCLUSIONS ECA affects virulence and benefits the Th2 immunity of Salmonella Typhimurium, therefore, it is feasible to use a reversible ECA mutant mode to design future Salmonella vaccine strains for heterologous protective antigens.
Collapse
Affiliation(s)
- Qing Liu
- College of Animal Science and Technology, Southwest University, Chongqing, China
- Chongqing Engineering Research Center for Herbivores Resource Protection and Utilization, Chongqing, China
| | - Xuegang Shen
- College of Animal Science and Technology, Southwest University, Chongqing, China
| | - Xiaoping Bian
- College of Animal Science and Technology, Southwest University, Chongqing, China
| | - Qingke Kong
- College of Animal Science and Technology, Southwest University, Chongqing, China.
| |
Collapse
|