1
|
Jafari Najaf Abadi MH, Abyaneh FA, Zare N, Zamani J, Abdoli A, Aslanbeigi F, Hamblin MR, Tarrahimofrad H, Rahimi M, Hashemian SM, Mirzaei H. In silico design and immunoinformatics analysis of a chimeric vaccine construct based on Salmonella pathogenesis factors. Microb Pathog 2023; 180:106130. [PMID: 37121524 DOI: 10.1016/j.micpath.2023.106130] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 04/26/2023] [Accepted: 04/27/2023] [Indexed: 05/02/2023]
Abstract
Currently, there are two vaccines based on killed and/or weakened Salmonella bacteria, but no recombinant vaccine is available for preventing or treating the disease. We used an in silico approach to design a multi-epitope vaccine against Salmonella using OmpA, OmpS, SopB, SseB, SthA and FilC antigens. We predicted helper T lymphocyte, cytotoxic T lymphocyte, and IFN-γ epitopes. The FilC sequence was used as a bovine TLR5 agonist, and the linkers KK, AAY, GPGPG and EAAAK were used to connect epitopes. The final sequence consisted of 747 amino acid residues, and the expressed soluble protein (∼79.6 kDa) was predicted to be both non-allergenic and antigenic. The tertiary structure of modeled protein was refined and validated, and the interactions of vaccine 3D structure were evaluated using molecular docking, and molecular dynamics simulation (RMSD, RMSF and Gyration). This structurally stable protein could interact with human TLR5. The C-ImmSim server predicted that this proposed vaccine likely induces an immune response by stimulating T and B cells, making it a potential candidate for further evaluation for the prevention and treatment of Salmonella infection.
Collapse
Affiliation(s)
| | | | - Noushid Zare
- Faculty of Pharmacy, International Campus, Tehran University of Medical Science, Tehran, Iran
| | - Javad Zamani
- Department of Animal Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran
| | - Amirhossein Abdoli
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran; Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Fatemeh Aslanbeigi
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran; Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Michael R Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein, 2028, South Africa
| | - Hossein Tarrahimofrad
- Department of Animal Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran.
| | - Mohammadreza Rahimi
- Infectious Diseases Research Center, Faculty of Medicine, Kashan University of Medical Sciences, Kashan, Iran; Department of Microbiology and Immunology, Faculty of Medicine, Kashan University of Medical Sciences, Kashan, Iran.
| | - Seyed Mohammadreza Hashemian
- Chronic Respiratory Diseases Research Center, National Research Institute of Tuberculosis and Lung Disease, Shahid Beheshti University of Medical Sciences, Tehran, 1983535511, Iran.
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
2
|
Niu L, Cai W, Cheng X, Li Z, Ruan J, Li F, Qi K, Tu J. Fur Protein Regulates the Motility of Avian Pathogenic Escherichia coli AE17 Through Promoter Regions of the Flagella Key Genes flhD. Front Vet Sci 2022; 9:854916. [PMID: 35518642 PMCID: PMC9062578 DOI: 10.3389/fvets.2022.854916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 03/21/2022] [Indexed: 12/01/2022] Open
Abstract
Avian pathogenic Escherichia coli (APEC) is an important pathogen causing several diseases in birds. It is responsible for local and systemic infections in poultry, seriously impeding the development of the poultry industry, and poses a potential risk to public health. The iron absorption regulatory protein Fur and the noncoding RNA, RyhB, that it negatively regulates are important factors in bacterial iron uptake, but the regulation of bacterial virulence genes varies greatly among different bacteria. We found that Fur is very important for the mobility of APEC. The expression of fur and RyhB is extensively regulated in APEC, and RyhB expression is also negatively regulated by Fur. A transcriptomic analysis showed that the genes significantly differentially regulated by Fur are related to cell movement, including pilus- or flagellum-dependent cell motility. To verify these results, we examined the effects of fur knockdown on cell movement by measuring the diameter of the bacteria colonies. Consistent with the RNA sequencing results, the mobility of AE17Δfur was significantly reduced compared with that of the wild type, and it had almost lost its ability to move. Using an electrophoretic mobility assay, we confirmed that the Fur protein directly binds to the promoter region of the key flagellum-related gene flhD, thereby affecting the assembly and synthesis of the APEC flagellum. This study extends our understanding of gene regulation in APEC.
Collapse
Affiliation(s)
- Lulu Niu
- Anhui Province Key Laboratory of Veterinary Pathobiology and Disease Control, College of Animal Science and Technology, Anhui Agricultural University, Hefei, China
| | - Weizhen Cai
- Anhui Province Key Laboratory of Veterinary Pathobiology and Disease Control, College of Animal Science and Technology, Anhui Agricultural University, Hefei, China
| | - Xi Cheng
- Anhui Province Key Laboratory of Veterinary Pathobiology and Disease Control, College of Animal Science and Technology, Anhui Agricultural University, Hefei, China
| | - Zhe Li
- Anhui Province Key Laboratory of Veterinary Pathobiology and Disease Control, College of Animal Science and Technology, Anhui Agricultural University, Hefei, China
| | - Jianming Ruan
- Anhui Province Key Laboratory of Veterinary Pathobiology and Disease Control, College of Animal Science and Technology, Anhui Agricultural University, Hefei, China
| | - Fangguo Li
- Anhui Province Key Laboratory of Veterinary Pathobiology and Disease Control, College of Animal Science and Technology, Anhui Agricultural University, Hefei, China
| | - Kezong Qi
- Anhui Province Key Laboratory of Veterinary Pathobiology and Disease Control, College of Animal Science and Technology, Anhui Agricultural University, Hefei, China
| | - Jian Tu
- Anhui Province Key Laboratory of Veterinary Pathobiology and Disease Control, College of Animal Science and Technology, Anhui Agricultural University, Hefei, China
| |
Collapse
|
3
|
Abstract
Polysaccharides are often the most abundant antigens found on the extracellular surfaces of bacterial cells. These polysaccharides play key roles in interactions with the outside world, and for many bacterial pathogens, they represent what is presented to the human immune system. As a result, many vaccines have been or currently are being developed against carbohydrate antigens. In this review, we explore the diversity of capsular polysaccharides (CPS) in Salmonella and other selected bacterial species and explain the classification and function of CPS as vaccine antigens. Despite many vaccines being developed using carbohydrate antigens, the low immunogenicity and the diversity of infecting strains and serovars present an antigen formulation challenge to manufacturers. Vaccines tend to focus on common serovars or have changing formulations over time, reflecting the trends in human infection, which can be costly and time-consuming. We summarize the approaches to generate carbohydrate-based vaccines for Salmonella, describe vaccines that are in development and emphasize the need for an effective vaccine against non-typhoidal Salmonella strains.
Collapse
|
4
|
Soulier A, Prevosto C, Chol M, Deban L, Cranenburgh RM. Engineering a Novel Bivalent Oral Vaccine against Enteric Fever. Int J Mol Sci 2021; 22:ijms22063287. [PMID: 33807097 PMCID: PMC8005139 DOI: 10.3390/ijms22063287] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 03/18/2021] [Accepted: 03/21/2021] [Indexed: 11/25/2022] Open
Abstract
Enteric fever is a major global healthcare issue caused largely by Salmonella enterica serovars Typhi and Paratyphi A. The objective of this study was to develop a novel, bivalent oral vaccine capable of protecting against both serovars. Our approach centred on genetically engineering the attenuated S. Typhi ZH9 strain, which has an excellent safety record in clinical trials, to introduce two S. Paratyphi A immunogenic elements: flagellin H:a and lipopolysaccharide (LPS) O:2. We first replaced the native S. Typhi fliC gene encoding flagellin with the highly homologous fliC gene from S. Paratyphi A using Xer-cise technology. Next, we replaced the S. Typhi rfbE gene encoding tyvelose epimerase with a spacer sequence to enable the sustained expression of O:2 LPS and prevent its conversion to O:9 through tyvelose epimerase activity. The resulting new strain, ZH9PA, incorporated these two genetic changes and exhibited comparable growth kinetics to the parental ZH9 strain. A formulation containing both ZH9 and ZH9PA strains together constitutes a new bivalent vaccine candidate that targets both S. Typhi and S. Paratyphi A antigens to address a major global healthcare gap for enteric fever prophylaxis. This vaccine is now being tested in a Phase I clinical trial (NCT04349553).
Collapse
|
5
|
Khani MH, Bagheri M, Zahmatkesh A, Aghaiypour K, Mirjalili A. Effect of flagellin on inhibition of infectious mechanisms by activating opsonization and salmonella flagellum disruption. Microb Pathog 2020; 142:104057. [PMID: 32058025 DOI: 10.1016/j.micpath.2020.104057] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 02/03/2020] [Accepted: 02/10/2020] [Indexed: 01/31/2023]
Abstract
Some serovars of salmonella cause huge global diseases such as enteric fever and invasive non typhoidal Salmonella disease. Flagellin as a key antigenic component of salmonella, can induce humoral and cellular immunity responses. In this research, we performed an opsonophagocytic killing assay (OPKA) as an important mechanism of the host-defense system, for salmonella to study the activity of anti-sera of native FliC, truncated modified recombinant FliC (tmFliC) and full length recombinant FliC proteins (flFliC). Also, the potency of antibodies for inhibiting bacterial movement was evaluated by traditional and newly-designed motility inhibition assay methods. Results showed both recombinant FliC anti-sera and native FliC (nFliC) anti-serum had the ability to opsonize Salmonella typhimurim, which led to bacterial clearance by mice macrophages. Also, inhibition of bacterial motility was observed for all anti-sera. Anti-nFliC and anti-flFliC sera showed higher effects on Salmonella typhimurim motility than that of tmFliC. In traditional method, about 88%, 86% and 80% inhibition were observed by using 5% nFliC, anti-flFliC and anti-tmFliC sera, respectively. In the newly-designed method using SIM (Sulfide indole motility) medium, results confirmed the traditional method for motility inhibition. Our findings suggest that salmonella fliC as a protective antigen may disrupt the flagellum apparatus activity.
Collapse
Affiliation(s)
- Mohammad-Hosein Khani
- Department of Genomics and Genetic Engineering, Razi Vaccine and Serum Research Institute (RVSRI), Agricultural Research, Education and Extension Organization (AREEO), Karaj, Iran
| | - Masoumeh Bagheri
- Department of Genomics and Genetic Engineering, Razi Vaccine and Serum Research Institute (RVSRI), Agricultural Research, Education and Extension Organization (AREEO), Karaj, Iran.
| | - Azadeh Zahmatkesh
- Department of Genomics and Genetic Engineering, Razi Vaccine and Serum Research Institute (RVSRI), Agricultural Research, Education and Extension Organization (AREEO), Karaj, Iran
| | - Khosrow Aghaiypour
- Department of Genomics and Genetic Engineering, Razi Vaccine and Serum Research Institute (RVSRI), Agricultural Research, Education and Extension Organization (AREEO), Karaj, Iran
| | - Ali Mirjalili
- Department of Genomics and Genetic Engineering, Razi Vaccine and Serum Research Institute (RVSRI), Agricultural Research, Education and Extension Organization (AREEO), Karaj, Iran
| |
Collapse
|
6
|
Felgner S, Spöring I, Pawar V, Kocijancic D, Preusse M, Falk C, Rohde M, Häussler S, Weiss S, Erhardt M. The immunogenic potential of bacterial flagella for Salmonella-mediated tumor therapy. Int J Cancer 2020; 147:448-460. [PMID: 31755108 DOI: 10.1002/ijc.32807] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 11/07/2019] [Accepted: 11/12/2019] [Indexed: 01/07/2023]
Abstract
Genetically engineered Salmonella Typhimurium are potent vectors for prophylactic and therapeutic measures against pathogens as well as cancer. This is based on the potent adjuvanticity that supports strong immune responses. The physiology of Salmonella is well understood. It simplifies engineering of both enhanced immune-stimulatory properties as well as safety features, thus, resulting in an appropriate balance between attenuation and efficacy for clinical applications. A major virulence factor of Salmonella is the flagellum. It is also a strong pathogen-associated molecular pattern recognized by extracellular and intracellular receptors of immune cells of the host. At the same time, it represents a serious metabolic burden. Accordingly, the bacteria evolved tight regulatory mechanisms that control flagella synthesis in vivo. Here, we systematically investigated the immunogenicity and adjuvant properties of various flagella mutants of Salmonella in vitro and in a mouse cancer model in vivo. We found that mutants lacking the flagellum-specific ATPase FliHIJ or the inner membrane ring FliF displayed the greatest stimulatory capacity and strongest antitumor effects, while remaining safe in vivo. Scanning electron microscopy revealed the presence of outer membrane vesicles in the ΔfliF and ΔfliHIJ mutants. Finally, the combination of the ΔfliF and ΔfliHIJ mutations with our previously described attenuated and immunogenic background strain SF102 displayed strong efficacy against the highly resistant cancer cell line RenCa. We thus conclude that manipulating flagella biosynthesis has great potential for the construction of highly efficacious and versatile Salmonella vector strains.
Collapse
Affiliation(s)
- Sebastian Felgner
- Infection Biology of Salmonella, Helmholtz Centre for Infection Research, Braunschweig, Germany.,Department of Molecular Bacteriology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Imke Spöring
- Infection Biology of Salmonella, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Vinay Pawar
- Department of Molecular Bacteriology, Helmholtz Centre for Infection Research, Braunschweig, Germany.,Central Facilities for Microscopy, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Dino Kocijancic
- Institute of Immunology, Medical School Hannover, Hannover, Germany
| | - Matthias Preusse
- Department of Molecular Bacteriology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Christine Falk
- Institute of Transplant Immunology, Medical School Hannover, Hannover, Germany
| | - Manfred Rohde
- Central Facilities for Microscopy, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Susanne Häussler
- Department of Molecular Bacteriology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Siegfried Weiss
- Institute of Immunology, Medical School Hannover, Hannover, Germany
| | - Marc Erhardt
- Infection Biology of Salmonella, Helmholtz Centre for Infection Research, Braunschweig, Germany.,Institute for Biology, Humboldt-Universität zu Berlin, Berlin, Germany
| |
Collapse
|
7
|
Acharya D, Sullivan MJ, Duell BL, Eveno T, Schembri MA, Ulett GC. Physical Extraction and Fast Protein Liquid Chromatography for Purifying Flagella Filament From Uropathogenic Escherichia coli for Immune Assay. Front Cell Infect Microbiol 2019; 9:118. [PMID: 31069177 PMCID: PMC6491459 DOI: 10.3389/fcimb.2019.00118] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Accepted: 04/03/2019] [Indexed: 01/17/2023] Open
Abstract
Flagella are expressed on the surface of a wide range of bacteria, conferring motility and contributing to virulence and innate immune stimulation. Host-pathogen interaction studies of the roles of flagella in infection, including due to uropathogenic Escherichia coli (UPEC), have used various methods to purify and examine the biology of the major flagella subunit protein, FliC. These studies have offered insight into the ways in which flagella proteins interact with host cells. However, previous methods used to extract and purify FliC, such as mechanical shearing, ultracentrifugation, heterologous expression in laboratory E. coli strains, and precipitation-inducing chemical treatments have various limitations; as a result, there are few observations based on highly purified, non-denatured FliC in the literature. This is especially relevant to host-pathogen interaction studies such as immune assays that are designed to parallel, as closely as possible, naturally-occurring interactions between host cells and flagella. In this study, we sought to establish a new, carefully optimized method to extract and purify non-denatured, native FliC from the reference UPEC strain CFT073 to be suitable for immune assays. To achieve purification of FliC to homogeneity, we used a mutant CFT073 strain containing deletions in four major chaperone-usher fimbriae operons (type 1, F1C and two P fimbrial gene clusters; CFT073Δ4). A sequential flagella extraction method based on mechanical shearing, ultracentrifugation, size exclusion chromatography, protein concentration and endotoxin removal was applied to CFT073Δ4. Protein purity and integrity was assessed using SDS-PAGE, Western blots with anti-flagellin antisera, and native-PAGE. We also generated a fliC-deficient strain, CFT073Δ4ΔfliC, to enable the concurrent preparation of a suitable carrier control to be applied in downstream assays. Innate immune stimulation was examined by exposing J774A.1 macrophages to 0.05-1 μg of purified FliC for 5 h; the supernatants were analyzed for cytokines known to be induced by flagella, including TNF-α, IL-6, and IL-12; the results were assessed in the context of prior literature. Macrophage responses to purified FliC encompassed significant levels of several cytokines consistent with prior literature reports. The purification method described here establishes a new approach to examine highly purified FliC in the context of host-pathogen interaction model systems.
Collapse
Affiliation(s)
- Dhruba Acharya
- School of Medical Science, Griffith University, Southport, QLD, Australia.,Menzies Health Institute Queensland, Griffith University, Southport, QLD, Australia
| | - Matthew J Sullivan
- School of Medical Science, Griffith University, Southport, QLD, Australia.,Menzies Health Institute Queensland, Griffith University, Southport, QLD, Australia
| | - Benjamin L Duell
- School of Medical Science, Griffith University, Southport, QLD, Australia.,Menzies Health Institute Queensland, Griffith University, Southport, QLD, Australia
| | - Tanguy Eveno
- Institute for Glycomics, Griffith University, Southport, QLD, Australia
| | - Mark A Schembri
- School of Chemistry and Molecular Biosciences, University of Queensland, St. Lucia, QLD, Australia
| | - Glen C Ulett
- School of Medical Science, Griffith University, Southport, QLD, Australia.,Menzies Health Institute Queensland, Griffith University, Southport, QLD, Australia
| |
Collapse
|
8
|
Stedman A, Maluquer de Motes C, Lesellier S, Dalley D, Chambers M, Gutierrez-Merino J. Lactic acid Bacteria isolated from European badgers (Meles meles) reduce the viability and survival of Bacillus Calmette-Guerin (BCG) vaccine and influence the immune response to BCG in a human macrophage model. BMC Microbiol 2018; 18:74. [PMID: 30005620 PMCID: PMC6044090 DOI: 10.1186/s12866-018-1210-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 06/27/2018] [Indexed: 02/08/2023] Open
Abstract
Background Bovine tuberculosis (bTB) caused by Mycobacterium bovis is the most serious endemic disease affecting livestock in the UK. The European badger (Meles meles) is the most important wildlife reservoir of bTB transmission to cattle, making eradication particularly difficult. In this respect, oral vaccination with the attenuated M. bovis vaccine Bacillus Calmette-Guerin (BCG) has been suggested as a wide-scale intervention to reduce bTB infection in badgers. However, experimental studies show variable protection. Among the possibilities for this variation is that the resident gut bacteria may influence the success of oral vaccination in badgers; either through competitive exclusion and/or inhibition, or via effects on the host immune system. In order to explore this possibility, we have tested whether typical gut commensals such as Lactic Acid Bacteria (LAB) have the capacity to impact on the viability and survival rate of BCG and to modulate the immune response to BCG using an in vitro model. Results Twelve LAB isolated from badger faeces displayed inhibitory activity to BCG that was species-dependent. Weissella had a bacteriostatic effect, whereas isolates of enterococci, lactobacilli and pediococci had a more bactericidal activity. Furthermore, BCG-induced activation of the pro-inflammatory transcription factor NF-κB in human THP-1 macrophages was modulated by LAB in a strain-dependent manner. Most pediococci enhanced NF-κB activation but one strain had the opposite effect. Interestingly, isolates of enterococci, lactobacilli and weissella had different effects as immunomodulators of BCG-induced macrophage responses as some had no significant influence on NF-κB activation, but others increased it significantly. Conclusions Our in vitro results show that LAB isolated from badgers exhibit significant inhibitory activity against BCG and influence the immune activation mediated by BCG in a human macrophage assay. These findings suggest that gut commensal bacteria could play a role in influencing the outcome of oral BCG vaccination. Inactivated cells of LAB, or LAB that are bacteriostatic but have a synergistic immunostimulatory effect with BCG, could be potential adjuvants to be used for oral vaccination in badgers. Further work is needed to take into account the complex nature of the gut microbiome, specific immunity of the badger and the in vivo context. Electronic supplementary material The online version of this article (10.1186/s12866-018-1210-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Anna Stedman
- School of Biosciences and Medicine, University of Surrey, Guildford, GU2 7XH, UK.,The Pirbright Institute, Woking, GU24 0NF, UK
| | | | - Sandrine Lesellier
- Bacteriology Department, Animal and Plant Health Agency, Addlestone, KT15 3NB, UK
| | - Deanna Dalley
- Bacteriology Department, Animal and Plant Health Agency, Addlestone, KT15 3NB, UK
| | - Mark Chambers
- Bacteriology Department, Animal and Plant Health Agency, Addlestone, KT15 3NB, UK.,School of Veterinary Medicine, University of Surrey, Guildford, GU2 7AL, UK
| | | |
Collapse
|
9
|
Felgner S, Kocijancic D, Frahm M, Heise U, Rohde M, Zimmermann K, Falk C, Erhardt M, Weiss S. Engineered Salmonella enterica serovar Typhimurium overcomes limitations of anti-bacterial immunity in bacteria-mediated tumor therapy. Oncoimmunology 2017; 7:e1382791. [PMID: 29308303 PMCID: PMC5749626 DOI: 10.1080/2162402x.2017.1382791] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 09/14/2017] [Accepted: 09/16/2017] [Indexed: 01/19/2023] Open
Abstract
Cancer is one of the leading causes of death in the industrialized world and represents a tremendous social and economic burden. As conventional therapies fail to provide a sustainable cure for most cancer patients, the emerging unique immune therapeutic approach of bacteria-mediated tumor therapy (BMTT) is marching towards a feasible solution. Although promising results have been obtained with BMTT using various preclinical tumor models, for advancement a major concern is immunity against the bacterial vector itself. Pre-exposure to the therapeutic agent under field conditions is a reasonable expectation and may limit the therapeutic efficacy of BMTT. In the present study, we investigated the therapeutic potential of Salmonella and E. coli vector strains in naïve and immunized tumor bearing mice. Pre-exposure to the therapeutic agent caused a significant aberrant phenotype of the microenvironment of colonized tumors and limited the in vivo efficacy of established BMTT vector strains Salmonella SL7207 and E. coli Symbioflor-2. Using targeted genetic engineering, we generated the optimized auxotrophic Salmonella vector strain SF200 (ΔlpxR9 ΔpagL7 ΔpagP8 ΔaroA ΔydiV ΔfliF) harboring modifications in Lipid A and flagella synthesis. This combination of mutations resulted in an increased immune-stimulatory capacity and as such the strain was able to overcome the efficacy-limiting effects of pre-exposure. Thus, we conclude that any limitations of BMTT concerning anti-bacterial immunity may be countered by strategies that optimize the immune-stimulatory capacity of the attenuated vector strains.
Collapse
Affiliation(s)
- Sebastian Felgner
- Department of Molecular Immunology, Helmholtz Centre for Infection Research, Braunschweig, Lower Saxony, Germany
- Infection Biology of Salmonella, Helmholtz Centre for Infection Research, Braunschweig, Lower Saxony, Germany
| | - Dino Kocijancic
- Department of Molecular Immunology, Helmholtz Centre for Infection Research, Braunschweig, Lower Saxony, Germany
| | - Michael Frahm
- Department of Molecular Immunology, Helmholtz Centre for Infection Research, Braunschweig, Lower Saxony, Germany
| | - Ulrike Heise
- Mouse-Pathology Service Unit, Helmholtz Centre for Infection Research, Braunschweig, Lower Saxony, Germany
| | - Manfred Rohde
- Central Facility for Microscopy, Helmholtz Centre for Infection Research, Braunschweig, Lower Saxony, Germany
| | | | - Christine Falk
- Institute of Transplant Immunology, Medical School Hannover, Hannover, Hessia, Germany
| | - Marc Erhardt
- Infection Biology of Salmonella, Helmholtz Centre for Infection Research, Braunschweig, Lower Saxony, Germany
| | - Siegfried Weiss
- Department of Molecular Immunology, Helmholtz Centre for Infection Research, Braunschweig, Lower Saxony, Germany
- Institute of Immunology, Medical School Hannover, Hannover, Lower Saxony, Germany
| |
Collapse
|
10
|
Abstract
Salmonella enterica subspecies enterica includes several serovars infecting both humans and other animals and leading to typhoid fever or gastroenteritis. The high prevalence of associated morbidity and mortality, together with an increased emergence of multidrug-resistant strains, is a current global health issue that has prompted the development of vaccination strategies that confer protection against most serovars. Currently available systemic vaccine approaches have major limitations, including a reduced effectiveness in young children and a lack of cross-protection among different strains. Having studied host-pathogen interactions, microbiologists and immunologists argue in favor of topical gastrointestinal administration for improvement in vaccine efficacy. Here, recent advances in this field are summarized, including mechanisms of bacterial uptake at the intestinal epithelium, the assessment of protective host immunity, and improved animal models that closely mimic infection in humans. The pros and cons of existing vaccines are presented, along with recent progress made with novel formulations. Finally, new candidate antigens and their relevance in the refined design of anti-Salmonella vaccines are discussed, along with antigen vectorization strategies such as nanoparticles or secretory immunoglobulins, with a focus on potentiating mucosal vaccine efficacy.
Collapse
|
11
|
aroA-Deficient Salmonella enterica Serovar Typhimurium Is More Than a Metabolically Attenuated Mutant. mBio 2016; 7:mBio.01220-16. [PMID: 27601574 PMCID: PMC5013297 DOI: 10.1128/mbio.01220-16] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Recombinant attenuated Salmonella enterica serovar Typhimurium strains are believed to act as powerful live vaccine carriers that are able to elicit protection against various pathogens. Auxotrophic mutations, such as a deletion of aroA, are commonly introduced into such bacteria for attenuation without incapacitating immunostimulation. In this study, we describe the surprising finding that deletion of aroA dramatically increased the virulence of attenuated Salmonella in mouse models. Mutant bacteria lacking aroA elicited increased levels of the proinflammatory cytokine tumor necrosis factor alpha (TNF-α) after systemic application. A detailed genetic and phenotypic characterization in combination with transcriptomic and metabolic profiling demonstrated that ΔaroA mutants display pleiotropic alterations in cellular physiology and lipid and amino acid metabolism, as well as increased sensitivity to penicillin, complement, and phagocytic uptake. In concert with other immunomodulating mutations, deletion of aroA affected flagellin phase variation and gene expression of the virulence-associated genes arnT and ansB. Finally, ΔaroA strains displayed significantly improved tumor therapeutic activity. These results highlight the importance of a functional shikimate pathway to control homeostatic bacterial physiology. They further highlight the great potential of ΔaroA-attenuated Salmonella for the development of vaccines and cancer therapies with important implications for host-pathogen interactions and translational medicine. Recombinant attenuated bacterial vector systems based on genetically engineered Salmonella have been developed as highly potent vaccines. Due to the pathogenic properties of Salmonella, efficient attenuation is required for clinical applications. Since the hallmark study by Hoiseth and Stocker in 1981 (S. K. Hoiseth and B. A. D. Stocker, Nature 291:238–239, 1981, http://dx.doi.org/10.1038/291238a0), the auxotrophic ΔaroA mutation has been generally considered safe and universally used to attenuate bacterial strains. Here, we are presenting the remarkable finding that a deletion of aroA leads to pronounced alterations of gene expression, metabolism, and cellular physiology, which resulted in increased immunogenicity, virulence, and adjuvant potential of Salmonella. These results suggest that the enhanced immunogenicity of aroA-deficient Salmonella strains might be advantageous for optimizing bacterial vaccine carriers and immunotherapy. Accordingly, we demonstrate a superior performance of ΔaroA Salmonella in bacterium-mediated tumor therapy. In addition, the present study highlights the importance of a functional shikimate pathway to sustain bacterial physiology and metabolism.
Collapse
|
12
|
Porta A, Petrone AM, Morello S, Granata I, Rizzo F, Memoli D, Weisz A, Maresca B. Design and expression of peptides with antimicrobial activity againstSalmonellatyphimurium. Cell Microbiol 2016; 19. [DOI: 10.1111/cmi.12645] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Accepted: 07/06/2016] [Indexed: 02/02/2023]
Affiliation(s)
- Amalia Porta
- Department of Pharmacy; University of Salerno; Fisciano (SA) Italy
| | - Anna Maria Petrone
- Department of Pharmacy; University of Salerno; Fisciano (SA) Italy
- PhD Program in Drug Discovery and Development; University of Salerno; Fisciano SA Italy
| | - Silvana Morello
- Department of Pharmacy; University of Salerno; Fisciano (SA) Italy
| | - Ilaria Granata
- Department of Pharmacy; University of Salerno; Fisciano (SA) Italy
| | - Francesca Rizzo
- Laboratory of Molecular Medicine and Genomics, Department of Medicine and Surgery; University of Salerno; Baronissi SA Italy
| | - Domenico Memoli
- Laboratory of Molecular Medicine and Genomics, Department of Medicine and Surgery; University of Salerno; Baronissi SA Italy
| | - Alessandro Weisz
- Laboratory of Molecular Medicine and Genomics, Department of Medicine and Surgery; University of Salerno; Baronissi SA Italy
| | - Bruno Maresca
- Department of Pharmacy; University of Salerno; Fisciano (SA) Italy
| |
Collapse
|
13
|
Felgner S, Kocijancic D, Frahm M, Weiss S. Bacteria in Cancer Therapy: Renaissance of an Old Concept. Int J Microbiol 2016; 2016:8451728. [PMID: 27051423 PMCID: PMC4802035 DOI: 10.1155/2016/8451728] [Citation(s) in RCA: 104] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Revised: 02/03/2016] [Accepted: 02/11/2016] [Indexed: 01/20/2023] Open
Abstract
The rising incidence of cancer cases worldwide generates an urgent need of novel treatment options. Applying bacteria may represent a valuable therapeutic variant that is intensively investigated nowadays. Interestingly, the idea to apply bacteria wittingly or unwittingly dates back to ancient times and was revived in the 19th century mainly by the pioneer William Coley. This review summarizes and compares the results of the past 150 years in bacteria mediated tumor therapy from preclinical to clinical studies. Lessons we have learned from the past provide a solid foundation on which to base future efforts. In this regard, several perspectives are discussed by which bacteria in addition to their intrinsic antitumor effect can be used as vector systems that shuttle therapeutic compounds into the tumor. Strategic solutions like these provide a sound and more apt exploitation of bacteria that may overcome limitations of conventional therapies.
Collapse
Affiliation(s)
- Sebastian Felgner
- Department of Molecular Immunology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
| | - Dino Kocijancic
- Department of Molecular Immunology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
| | - Michael Frahm
- Department of Molecular Immunology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
| | - Siegfried Weiss
- Department of Molecular Immunology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
- Institute of Immunology, Hannover Medical School, 30625 Hannover, Germany
| |
Collapse
|
14
|
Wang G, Shi B, Li T, Zuo T, Wang B, Si W, Xin J, Yang K, Shi X, Liu S, Liu H. Linear antigenic mapping of flagellin (FliC) from Salmonella enterica serovar Enteritidis with yeast surface expression system. Vet Microbiol 2016; 184:20-6. [PMID: 26854340 DOI: 10.1016/j.vetmic.2016.01.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Revised: 12/01/2015] [Accepted: 01/01/2016] [Indexed: 12/21/2022]
Abstract
Salmonella enterica serovar Enteritidis (S. Enteritidis) is a major cause of food-borne illness around the world and can have significant health implications in humans, poultry and other animals. Flagellin (FliC) is the primary component of bacterial flagella. It has been shown that the FliC of S. Enteritidis is a significant antigenic structure and can elicit strong humoral responses against S. Enteritidis infection in chickens. Here, we constructed a FliC antigen library using a yeast surface expression system. Yeast cells expressing FliC peptide antigens were labeled with chicken sera against S. Enteritidis and sorted using FACS. The analyses of FliC peptides revealed that the FliC linear antigenicity in chickens resided on three domains which were able to elicit strong humoral responses in vivo. Animal experiments further revealed that the antibodies elicited by these antigenic domains were able to significantly inhibit the invasion of S. Enteritidis into the liver and spleen of chickens. These findings will facilitate our better understanding of the humoral responses elicited by FliC in chickens upon infection by S. Enteritidis.
Collapse
Affiliation(s)
- Gaoling Wang
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin, China
| | - Bingtian Shi
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin, China
| | - Tao Li
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin, China; College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Teng Zuo
- Comprehensive AIDS Research Center, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Bin Wang
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin, China; College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Wei Si
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin, China
| | - Jiuqing Xin
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin, China
| | - Kongbin Yang
- Neurosurgery Department of the First Affiliated Hospital, Harbin Medical University, China
| | - Xuanlin Shi
- Comprehensive AIDS Research Center, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Siguo Liu
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin, China.
| | - Henggui Liu
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin, China.
| |
Collapse
|
15
|
Goh YS, Armour KL, Clark MR, Grant AJ, Mastroeni P. Igg Subclasses Targeting the Flagella of Salmonella enterica Serovar Typhimurium Can Mediate Phagocytosis and Bacterial Killing. ACTA ACUST UNITED AC 2016; 7. [PMID: 27366588 DOI: 10.4172/2157-7560.1000322] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Invasive non-typhoidal Salmonella are a common cause of invasive disease in immuno-compromised individuals and in children. Multi-drug resistance poses challenges to disease control, with a critical need for effective vaccines. Flagellin is an attractive vaccine candidate due to surface exposure and high epitope copy number, but its potential as a target for opsonophacytic antibodies is unclear. We examined the effect of targeting flagella with different classes of IgG on the interaction between Salmonella Typhimurium and a human phagocyte-like cell line, THP-1. We tagged the FliC flagellar protein with a foreign CD52 mimotope (TSSPSAD) and bacteria were opsonized with a panel of humanised CD52 antibodies with the same antigen-binding V-region, but different constant regions. We found that IgG binding to flagella increases bacterial phagocytosis and reduces viable intracellular bacterial numbers. Opsonisation with IgG3, followed by IgG1, IgG4, and IgG2, resulted in the highest level of bacterial uptake and in the highest reduction in the intracellular load of viable bacteria. Taken together, our data provide proof-of-principle evidence that targeting flagella with antibodies can increase the antibacterial function of host cells, with IgG3 being the most potent subclass. These data will assist the rational design of urgently needed, optimised vaccines against iNTS disease.
Collapse
Affiliation(s)
- Yun Shan Goh
- Department of Veterinary Medicine, University of Cambridge, Madingley Road, Cambridge CB3 0ES, United Kingdom
| | - Kathryn L Armour
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QP, United Kingdom
| | - Michael R Clark
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QP, United Kingdom
| | - Andrew J Grant
- Department of Veterinary Medicine, University of Cambridge, Madingley Road, Cambridge CB3 0ES, United Kingdom
| | - Pietro Mastroeni
- Department of Veterinary Medicine, University of Cambridge, Madingley Road, Cambridge CB3 0ES, United Kingdom
| |
Collapse
|
16
|
Delavari S, Sohrabi M, Ardestani MS, Faezi S, Tebianian M, Taghizadeh M, Shajiei A, Hosseini SY, Moghaddampour M, Mahdavi M. Pseudomonas aeruginosa flagellin as an adjuvant: superiority of a conjugated form of flagellin versus a mixture with a human immunodeficiency virus type 1 vaccine candidate in the induction of immune responses. J Med Microbiol 2015; 64:1361-1368. [PMID: 26404477 DOI: 10.1099/jmm.0.000174] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
In the present study, the adjuvant activity of flagellin was compared, in the conjugated and mixed forms, against a peptide vaccine from human immunodeficiency virus type 1 (HIV-1) p24-Nef vaccine candidate. Mice were immunized with the HIV-1 p24-Nef peptide with flagellin in both conjugated and mixed forms. Lymphocyte proliferation, CTL activity, and IL-4 and IFN-γ cytokines were evaluated by bromodeoxyuridine, carboxyfluorescein succinimidyl ester and ELISA methods, respectively. At the same time, the frequency of IFN-γ-producing T-lymphocytes, as well as total and isotype-specific antibodies, were assessed by ELISPOT and indirect ELISA, respectively. Our experimental results showed that the immunized mice with the HIV-1 p24-Nef conjugated or mixed forms of flagellin led to increases in the proliferative responses and Th1 cytokine pattern. The conjugated form of vaccine led to increased CTL activity and a Th1 cytokine pattern of immune responses, as well as an IgM isotype of humoral responses in comparison with the mixed form. The flagellin-conjugated vaccine seems to be more potent in increasing vaccine immunogenicity.
Collapse
Affiliation(s)
- Safura Delavari
- Department of Biology, Faculty of Sciences, Qom branch of Islamic Azad University, Qom, Iran
| | - Mojtaba Sohrabi
- Department of Biology, Faculty of Sciences, Qom branch of Islamic Azad University, Qom, Iran
| | - Mehdi Shafiee Ardestani
- Department of Radiopharmacy, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Sobhan Faezi
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran
| | - Majid Tebianian
- Department of Biotechnology, Razi Vaccine and Serum Research Institute, Karaj, Iran
| | - Morteza Taghizadeh
- Department of Medical Virology, Razi Vaccine and Serum Research Institute, Karaj, Iran
| | - Arezoo Shajiei
- Molecular Pathology Laboratory, Cancer Molecular Pathology Research Center, Ghaem Hospital, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Younes Hosseini
- Department of Bacteriology and Virology, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Masoud Moghaddampour
- Department of Medical Virology, Razi Vaccine and Serum Research Institute, Karaj, Iran.,Department of Virology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mehdi Mahdavi
- Department of Immunology, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
17
|
Matsui H, Fukiya S, Kodama-Akaboshi C, Eguchi M, Yamamoto T. Mouse models for assessing the cross-protective efficacy of oral non-typhoidal Salmonella vaccine candidates harbouring in-frame deletions of the ATP-dependent protease lon and other genes. J Med Microbiol 2015; 64:295-302. [PMID: 25589672 DOI: 10.1099/jmm.0.000014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
In BALB/c mouse models of Salmonella enterica serovar Typhimurium infection, a single oral immunization with a mutant strain with an insertion of the chloramphenicol resistance gene into the ATP-dependent protease clpP or lon gene decreased the number of salmonellae in each tissue sample 5 days after oral challenge with virulent S. Typhimurium at weeks 26 and 54 post-immunization. These data suggested that an oral immunization with the ClpP- or Lon-disrupted S. Typhimurium strain could provide long-term protection against oral challenge with virulent S. Typhimurium. Accordingly, recombinant oral non-typhoidal Salmonella (NTS) vaccines were constructed by incorporating mutants of both S. Typhimurium and S. enterica serovar Enteritidis harbouring stable in-frame markerless deletions of the clpP-lon-sulA (suppressor of lon), lon-sulA or lon-msbB (acyltransferase) genes. Amongst these orally administered vaccine candidates, those with the lon-sulA gene deletion mutants of S. Typhimurium and S. Enteritidis protected BALB/c and C57BL/6J mice against oral challenge with both virulent S. Typhimurium and virulent S. Enteritidis. Therefore, the in-frame markerless lon-sulA gene deletion mutant of S. Typhimurium or S. Enteritidis could be a promising cross-protective NTS live vaccine candidate for practical use in humans.
Collapse
Affiliation(s)
- Hidenori Matsui
- Kitasato Institute for Life Sciences and Graduate School of Infection Control Sciences, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641, Japan
| | - Satoru Fukiya
- Kitasato Institute for Life Sciences and Graduate School of Infection Control Sciences, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641, Japan
| | - Chie Kodama-Akaboshi
- Kitasato Institute for Life Sciences and Graduate School of Infection Control Sciences, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641, Japan
| | - Masahiro Eguchi
- National Institute of Animal Health, National Agriculture and Food Research Organization, 3-1-5 Kannondai, Tsukuba, Ibaraki 305-0856, Japan
| | - Tomoko Yamamoto
- Graduate School of Pharmaceutical Sciences, Chiba University, Yayoi-cho, Inage-ku, Chiba 263-8522, Japan
| |
Collapse
|
18
|
MacLennan CA, Martin LB, Micoli F. Vaccines against invasive Salmonella disease: current status and future directions. Hum Vaccin Immunother 2014; 10:1478-93. [PMID: 24804797 PMCID: PMC4185946 DOI: 10.4161/hv.29054] [Citation(s) in RCA: 156] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Though primarily enteric pathogens, Salmonellae are responsible for a considerable yet under-appreciated global burden of invasive disease. In South and South-East Asia, this manifests as enteric fever caused by serovars Typhi and Paratyphi A. In sub-Saharan Africa, a similar disease burden results from invasive nontyphoidal Salmonellae, principally serovars Typhimurium and Enteritidis. The existing Ty21a live-attenuated and Vi capsular polysaccharide vaccines target S. Typhi and are not effective in young children where the burden of invasive Salmonella disease is highest. After years of lack of investment in new Salmonella vaccines, recent times have seen increased interest in the area led by emerging-market manufacturers, global health vaccine institutes and academic partners. New glycoconjugate vaccines against S. Typhi are becoming available with similar vaccines against other invasive serovars in development. With other new vaccines under investigation, including live-attenuated, protein-based and GMMA vaccines, now is an exciting time for the Salmonella vaccine field.
Collapse
Affiliation(s)
- Calman A MacLennan
- Novartis Vaccines Institute for Global Health; Siena, Italy; Medical Research Council Centre for Immune Regulation and Clinical Immunology Service; Institute of Biomedical Research, School of Immunity and Infection; College of Medicine and Dental Sciences; University of Birmingham; Birmingham, UK
| | - Laura B Martin
- Novartis Vaccines Institute for Global Health; Siena, Italy
| | | |
Collapse
|
19
|
Cong H, Yuan Q, Zhao Q, Zhao L, Yin H, Zhou H, He S, Wang Z. Comparative efficacy of a multi-epitope DNA vaccine via intranasal, peroral, and intramuscular delivery against lethal Toxoplasma gondii infection in mice. Parasit Vectors 2014; 7:145. [PMID: 24685150 PMCID: PMC4229990 DOI: 10.1186/1756-3305-7-145] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2013] [Accepted: 03/21/2014] [Indexed: 12/03/2022] Open
Abstract
Background Toxoplasmosis is an important zoonosis, being a cause of congenital disease and abortion in animals and humans. DNA vaccination as a promising vaccine remains a challenge for an improved delivery system. Methods In this study, attenuated Salmonella typhimurium BRD509 was used to deliver a DNA vaccine encoding several epitopes, derived from the tachyzoite proteins SAG1, GRA1, ROP2, GRA4 and bradyzoite proteins SAG2C, SAG2X of Toxoplasma gondii and A2/B subunit of cholera toxin. The recombinant plasmids were electroporated into attenuated Salmonella typhimurium. Humoral and cellular immune responses were evaluated for BALB/c mice administered with this attenuated recombinant Salmonella vaccine via the oral and nasal route or by intramuscular injection with DNA plasmid directly. Results High IgG levels were present in the mice immunized intramuscularly, while IgA levels were higher in the oral and nasal immunization groups. Furthermore, cellular immunity was activated in oral immunization groups with 60% survival rate following challenge with high virulent RH strain. Conclusions The results from this study indicate that a DNA vaccine encoding multi-epitopes of T. gondii delivered by attenuated Salmonella is promising.
Collapse
Affiliation(s)
- Hua Cong
- Department of human parasitology, Medical school, Shandong University, No,44 Wenhuaxi Road, Jinan, Shandong 250012, P, R, China.
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Chin'ombe N, Ruhanya V. Recombinant Salmonella Bacteria Vectoring HIV/AIDS Vaccines. Open Virol J 2013; 7:121-6. [PMID: 24478808 PMCID: PMC3905348 DOI: 10.2174/1874357901307010121] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2013] [Revised: 11/13/2013] [Accepted: 11/18/2013] [Indexed: 11/29/2022] Open
Abstract
HIV/AIDS is an important public health problem globally. An affordable, easy-to-deliver and protective HIV
vaccine is therefore required to curb the pandemic from spreading further. Recombinant Salmonella bacteria can be
harnessed to vector HIV antigens or DNA vaccines to the immune system for induction of specific protective immunity.
These are capable of activating the innate, humoral and cellular immune responses at both mucosal and systemic
compartments. Several studies have already demonstrated the utility of live recombinant Salmonella in delivering
expressed foreign antigens as well as DNA vaccines to the host immune system. This review gives an overview of the
studies in which recombinant Salmonella bacteria were used to vector HIV/AIDS antigens and DNA vaccines. Most of
the recombinant Salmonella-based HIV/AIDS vaccines developed so far have only been tested in animals (mainly mice)
and are yet to reach human trials.
Collapse
Affiliation(s)
- Nyasha Chin'ombe
- Department of Medical Microbiology, University of Zimbabwe, Harare, Zimbabwe ; Division of Medical Virology, University of Cape Town, Cape Town, South Africa
| | - Vurayai Ruhanya
- Department of Medical Microbiology, University of Zimbabwe, Harare, Zimbabwe
| |
Collapse
|