1
|
Inflammasome-Mediated Immunogenicity of Clinical and Experimental Vaccine Adjuvants. Vaccines (Basel) 2020; 8:vaccines8030554. [PMID: 32971761 PMCID: PMC7565252 DOI: 10.3390/vaccines8030554] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 09/16/2020] [Accepted: 09/18/2020] [Indexed: 02/06/2023] Open
Abstract
In modern vaccines, adjuvants can be sophisticated immunological tools to promote robust and long-lasting protection against prevalent diseases. However, there is an urgent need to improve immunogenicity of vaccines in order to protect mankind from life-threatening diseases such as AIDS, malaria or, most recently, COVID-19. Therefore, it is important to understand the cellular and molecular mechanisms of action of vaccine adjuvants, which generally trigger the innate immune system to enhance signal transition to adaptive immunity, resulting in pathogen-specific protection. Thus, improved understanding of vaccine adjuvant mechanisms may aid in the design of “intelligent” vaccines to provide robust protection from pathogens. Various commonly used clinical adjuvants, such as aluminium salts, saponins or emulsions, have been identified as activators of inflammasomes - multiprotein signalling platforms that drive activation of inflammatory caspases, resulting in secretion of pro-inflammatory cytokines of the IL-1 family. Importantly, these cytokines affect the cellular and humoral arms of adaptive immunity, which indicates that inflammasomes represent a valuable target of vaccine adjuvants. In this review, we highlight the impact of different inflammasomes on vaccine adjuvant-induced immune responses regarding their mechanisms and immunogenicity. In this context, we focus on clinically relevant adjuvants that have been shown to activate the NLRP3 inflammasome and also present various experimental adjuvants that activate the NLRP3-, NLRC4-, AIM2-, pyrin-, or non-canonical inflammasomes and could have the potential to improve future vaccines. Together, we provide a comprehensive overview on vaccine adjuvants that are known, or suggested, to promote immunogenicity through inflammasome-mediated signalling.
Collapse
|
2
|
Yazdani Z, Rafiei A, Yazdani M, Valadan R. Design an Efficient Multi-Epitope Peptide Vaccine Candidate Against SARS-CoV-2: An in silico Analysis. Infect Drug Resist 2020; 13:3007-3022. [PMID: 32943888 PMCID: PMC7459237 DOI: 10.2147/idr.s264573] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 07/28/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND To date, no specific vaccine or drug has been proven to be effective against SARS-CoV-2 infection. Therefore, we implemented an immunoinformatic approach to design an efficient multi-epitopes vaccine against SARS-CoV-2. RESULTS The designed-vaccine construct consists of several immunodominant epitopes from structural proteins of spike, nucleocapsid, membrane, and envelope. These peptides promote cellular and humoral immunity and interferon-gamma responses. Also, these epitopes have a high antigenic capacity and are not likely to cause allergies. To enhance the vaccine immunogenicity, we used three potent adjuvants: Flagellin of Salmonella enterica subsp. enterica serovar Dublin, a driven peptide from high mobility group box 1 as HP-91, and human beta-defensin 3 protein. The physicochemical and immunological properties of the vaccine structure were evaluated. The tertiary structure of the vaccine protein was predicted and refined by Phyre2 and Galaxi refine and validated using RAMPAGE and ERRAT. Results of ElliPro showed 246 sresidues from vaccine might be conformational B-cell epitopes. Docking of the vaccine with toll-like receptors (TLR) 3, 5, 8, and angiotensin-converting enzyme 2 approved an appropriate interaction between the vaccine and receptors. Prediction of mRNA secondary structure and in silico cloning demonstrated that the vaccine can be efficiently expressed in Escherichia coli. CONCLUSION Our results demonstrated that the multi-epitope vaccine might be potentially antigenic and induce humoral and cellular immune responses against SARS-CoV-2. This vaccine can interact appropriately with the TLR3, 5, and 8. Also, it has a high-quality structure and suitable characteristics such as high stability and potential for expression in Escherichia coli .
Collapse
Affiliation(s)
- Zahra Yazdani
- Department of Immunology, Molecular and Cell Biology Research Center, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Alireza Rafiei
- Department of Immunology, Molecular and Cell Biology Research Center, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Mohammadreza Yazdani
- Department of Chemistry, Isfahan University of Technology, Isfahan84156-83111, Iran
| | - Reza Valadan
- Department of Immunology, Molecular and Cell Biology Research Center, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| |
Collapse
|
3
|
Zhang Y, Wu Q, Zhou M, Luo Z, Lv L, Pei J, Wang C, Chai B, Sui B, Huang F, Fu ZF, Zhao L. Composition of the murine gut microbiome impacts humoral immunity induced by rabies vaccines. Clin Transl Med 2020; 10:e161. [PMID: 32898335 PMCID: PMC7443138 DOI: 10.1002/ctm2.161] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 08/12/2020] [Accepted: 08/12/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Gut microbiome plays a crucial role in modulating human and animal immune responses. Rabies is a fatal zoonosis causing encephalitis in mammals and vaccination is the most effective method to control and eliminate rabies. The relationship between the gut microbiome and humoral immunity post rabies vaccination has not been investigated yet. METHODS Mice orally administrated with a cocktail of broad-spectrum antibiotics were inoculated with rabies vaccines, and humoral immune response was analyzed at indicated time points. The 16S ribosomal RNA (16S rRNA) gene sequencing was performed on fecal samples from groups in vancomycin-treated and untreated mice. Mice were immunized with rabies vaccines and virus-neutralizing antibody (VNA) levels were measured, resulting in VNA high (H) and low (L) groups. Then 16S rRNA gene sequencing was performed on fecal samples from H and L group mice. RESULTS After antibiotic (Abx) treatment, mice had decreased levels of rabies virus (RABV)-specific IgM, IgG, and virus-neutralizing antibody compared with untreated mice. Abx-treated mice had fewer T follicular helper cells, germinal center B cells, and antibody secreting cells (ASCs) in lymph nodes than did untreated mice. Gut microbiome facilitated secondary immune responses by increasing the generation of ASCs. Treatment with vancomycin alone had a similarly impaired effect on the humoral immune responses compared with Abx-treated mice. From the natural population group of mice received rabies vaccines, VNA titers vary significantly and the abundance of Clostridiales and Lachnospiraceae was positively associated with the antibody titers in mice. CONCLUSIONS Our results provide the evidence that the gut microbiome impacts humoral immunity post rabies vaccination, and further investigation of the mechanism will help the development of novel adjuvants and vaccines.
Collapse
Affiliation(s)
- Yachun Zhang
- State Key Laboratory of Agricultural MicrobiologyHuazhong Agricultural UniversityWuhanChina
- Key Laboratory of Preventive Veterinary Medicine of Hubei ProvinceCollege of Veterinary MedicineHuazhong Agricultural UniversityWuhanChina
| | - Qiong Wu
- State Key Laboratory of Agricultural MicrobiologyHuazhong Agricultural UniversityWuhanChina
- Key Laboratory of Preventive Veterinary Medicine of Hubei ProvinceCollege of Veterinary MedicineHuazhong Agricultural UniversityWuhanChina
| | - Ming Zhou
- State Key Laboratory of Agricultural MicrobiologyHuazhong Agricultural UniversityWuhanChina
- Key Laboratory of Preventive Veterinary Medicine of Hubei ProvinceCollege of Veterinary MedicineHuazhong Agricultural UniversityWuhanChina
| | - Zhaochen Luo
- State Key Laboratory of Agricultural MicrobiologyHuazhong Agricultural UniversityWuhanChina
- Key Laboratory of Preventive Veterinary Medicine of Hubei ProvinceCollege of Veterinary MedicineHuazhong Agricultural UniversityWuhanChina
| | - Lei Lv
- State Key Laboratory of Agricultural MicrobiologyHuazhong Agricultural UniversityWuhanChina
- Key Laboratory of Preventive Veterinary Medicine of Hubei ProvinceCollege of Veterinary MedicineHuazhong Agricultural UniversityWuhanChina
| | - Jie Pei
- State Key Laboratory of Agricultural MicrobiologyHuazhong Agricultural UniversityWuhanChina
- Key Laboratory of Preventive Veterinary Medicine of Hubei ProvinceCollege of Veterinary MedicineHuazhong Agricultural UniversityWuhanChina
| | - Caiqian Wang
- State Key Laboratory of Agricultural MicrobiologyHuazhong Agricultural UniversityWuhanChina
- Key Laboratory of Preventive Veterinary Medicine of Hubei ProvinceCollege of Veterinary MedicineHuazhong Agricultural UniversityWuhanChina
| | - Benjie Chai
- State Key Laboratory of Agricultural MicrobiologyHuazhong Agricultural UniversityWuhanChina
- Key Laboratory of Preventive Veterinary Medicine of Hubei ProvinceCollege of Veterinary MedicineHuazhong Agricultural UniversityWuhanChina
| | - Baokun Sui
- State Key Laboratory of Agricultural MicrobiologyHuazhong Agricultural UniversityWuhanChina
- Key Laboratory of Preventive Veterinary Medicine of Hubei ProvinceCollege of Veterinary MedicineHuazhong Agricultural UniversityWuhanChina
| | - Fei Huang
- State Key Laboratory of Agricultural MicrobiologyHuazhong Agricultural UniversityWuhanChina
- Key Laboratory of Preventive Veterinary Medicine of Hubei ProvinceCollege of Veterinary MedicineHuazhong Agricultural UniversityWuhanChina
| | - Zhen F. Fu
- State Key Laboratory of Agricultural MicrobiologyHuazhong Agricultural UniversityWuhanChina
- Key Laboratory of Preventive Veterinary Medicine of Hubei ProvinceCollege of Veterinary MedicineHuazhong Agricultural UniversityWuhanChina
| | - Ling Zhao
- State Key Laboratory of Agricultural MicrobiologyHuazhong Agricultural UniversityWuhanChina
- Key Laboratory of Preventive Veterinary Medicine of Hubei ProvinceCollege of Veterinary MedicineHuazhong Agricultural UniversityWuhanChina
| |
Collapse
|
4
|
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
|
5
|
Tsybalova LM, Stepanova LA, Shuklina MA, Mardanova ES, Kotlyarov RY, Potapchuk MV, Petrov SA, Blokhina EA, Ravin NV. Combination of M2e peptide with stalk HA epitopes of influenza A virus enhances protective properties of recombinant vaccine. PLoS One 2018; 13:e0201429. [PMID: 30138320 PMCID: PMC6107133 DOI: 10.1371/journal.pone.0201429] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Accepted: 07/16/2018] [Indexed: 12/14/2022] Open
Abstract
Background Influenza infection could be more effectively controlled if a multi-purpose vaccine with the ability to induce responses against most, or all, influenza A subtypes could be generated. Conserved viral proteins are a promising basis for the creation of a broadly protective vaccine. In the present study, the immunogenicity and protective properties of three recombinant proteins (vaccine candidates), comprising conserved viral proteins fused with bacterial flagellin, were compared. Methods Balb/c mice were immunized intranasally with recombinant proteins comprising either one viral protein (the ectodomain of the M2 protein, ‘M2e’) or two viral proteins (M2e and the hemagglutinin second subunit ‘HA2’ epitope) genetically fused with flagellin. Further, two different consensus variants of HA2 were used. Therefore, three experimental positives were used in addition to the negative control (Flg-his). The mucosal, humoral, and T-cell immune responses to these constructs were evaluated. Result We have demonstrated that insertion of the HA2 consensus polypeptide (aa 76–130), derived from either the first (HA2-1) or second (HA2-2) virus phylogenetic group, into the recombinant Flg4M2e protein significantly enhanced its immunogenicity and protective properties. Intranasal administration of the vaccine candidates (Flg-HA2-2-4M2e or Flg-HA2-1-4M2e) induced considerable mucosal and systemic responses directed at both the M2e-protein and, in general, the influenza A virus. However, the immune response elicited by the Flg-HA2-1-4M2e protein was weaker than the one generated by Flg-HA2-2-4M2e. These recombinant proteins containing both viral peptides provide complete protection from lethal challenge with various influenza viruses: A/H3N2; A/H2N2; and A/H5N1. Conclusion This study demonstrates that the intranasal administration of Flg-HA2-2-4M2e recombinant protein induces a strong immune response which provides broad protection against various influenza viruses. This construct is therefore a strong candidate for development as a universal vaccine.
Collapse
Affiliation(s)
- Liudmila M. Tsybalova
- Department of Vaccinology, Smorodintsev Research Institute of Influenza, Ministry of Health of the Russian Federation, St. Petersburg, Russia
- * E-mail:
| | - Liudmila A. Stepanova
- Department of Vaccinology, Smorodintsev Research Institute of Influenza, Ministry of Health of the Russian Federation, St. Petersburg, Russia
| | - Marina A. Shuklina
- Department of Vaccinology, Smorodintsev Research Institute of Influenza, Ministry of Health of the Russian Federation, St. Petersburg, Russia
| | - Eugenia S. Mardanova
- Institute of Bioengineering, Research Center of Biotechnology, Russian Academy of Sciences, Moscow, Russia
| | - Roman Y. Kotlyarov
- Institute of Bioengineering, Research Center of Biotechnology, Russian Academy of Sciences, Moscow, Russia
| | - Marina V. Potapchuk
- Department of Vaccinology, Smorodintsev Research Institute of Influenza, Ministry of Health of the Russian Federation, St. Petersburg, Russia
| | - Sergei A. Petrov
- Department of Vaccinology, Smorodintsev Research Institute of Influenza, Ministry of Health of the Russian Federation, St. Petersburg, Russia
| | - Elena A. Blokhina
- Institute of Bioengineering, Research Center of Biotechnology, Russian Academy of Sciences, Moscow, Russia
| | - Nikolai V. Ravin
- Institute of Bioengineering, Research Center of Biotechnology, Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
6
|
Abstract
INTRODUCTION Bacterial flagellin, as a pathogen-associated molecular pattern (PAMP), can activate both innate and adaptive immunity. Its unique structural characteristics endow an effective and flexible adjuvant activity, which allow the design of different types of vaccine strategies to prevent various diseases. This review will discuss recent progress in the mechanism of action of flagellin and its prospects for use as a vaccine adjuvant. AREAS COVERED Herein we summarize various types of information related to flagellin adjuvants from PubMed, including structures, signaling pathways, natural immunity, and extensive applications in vaccines, and it discusses the immunogenicity, safety, and efficacy of flagellin-adjuvanted vaccines in clinical trials. EXPERT COMMENTARY It is widely accepted that as an adjuvant, flagellin can induce an enhanced antigen-specific immune response. Flagellin adjuvants will allow more effective flagellin-based vaccines to enter clinical trials. Furthermore, vaccine formulations containing PAMPs are crucial to exert the maximum potential of vaccine antigens. Therefore, combinations of flagellin-adjuvanted vaccines with other adjuvants that act in a synergistic manner, particularly TLR ligands, represent a promising method for tailoring targeted vaccines to meet specific requirements.
Collapse
Affiliation(s)
- Baofeng Cui
- a State Key Laboratory of Veterinary Etiological Biology, OIE/National Foot and Mouth Disease Reference Laboratory, Key Laboratory of Animal Virology of Ministry of Agriculture , Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences , Lanzhou , China.,b Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses , Yangzhou , China
| | - Xinsheng Liu
- a State Key Laboratory of Veterinary Etiological Biology, OIE/National Foot and Mouth Disease Reference Laboratory, Key Laboratory of Animal Virology of Ministry of Agriculture , Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences , Lanzhou , China.,b Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses , Yangzhou , China
| | - Yuzhen Fang
- a State Key Laboratory of Veterinary Etiological Biology, OIE/National Foot and Mouth Disease Reference Laboratory, Key Laboratory of Animal Virology of Ministry of Agriculture , Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences , Lanzhou , China.,b Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses , Yangzhou , China
| | - Peng Zhou
- a State Key Laboratory of Veterinary Etiological Biology, OIE/National Foot and Mouth Disease Reference Laboratory, Key Laboratory of Animal Virology of Ministry of Agriculture , Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences , Lanzhou , China.,b Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses , Yangzhou , China
| | - Yongguang Zhang
- a State Key Laboratory of Veterinary Etiological Biology, OIE/National Foot and Mouth Disease Reference Laboratory, Key Laboratory of Animal Virology of Ministry of Agriculture , Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences , Lanzhou , China.,b Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses , Yangzhou , China
| | - Yonglu Wang
- a State Key Laboratory of Veterinary Etiological Biology, OIE/National Foot and Mouth Disease Reference Laboratory, Key Laboratory of Animal Virology of Ministry of Agriculture , Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences , Lanzhou , China.,b Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses , Yangzhou , China
| |
Collapse
|
7
|
Schülke S, Kuttich K, Wolfheimer S, Duschek N, Wangorsch A, Reuter A, Briza P, Pablos I, Gadermaier G, Ferreira F, Vieths S, Toda M, Scheurer S. Conjugation of wildtype and hypoallergenic mugwort allergen Art v 1 to flagellin induces IL-10-DC and suppresses allergen-specific TH2-responses in vivo. Sci Rep 2017; 7:11782. [PMID: 28924222 PMCID: PMC5603567 DOI: 10.1038/s41598-017-11972-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Accepted: 08/16/2017] [Indexed: 11/09/2022] Open
Abstract
Allergies to weed pollen including members of the Compositae family, such as mugwort, ragweed, and feverfew are spreading worldwide. To efficiently treat these newly arising allergies, allergen specific immunotherapy needs to be improved. Therefore, we generated novel vaccine candidates consisting of the TLR5-ligand Flagellin A from Listeria and the major mugwort allergen Art v 1 including either the wild type Art v 1 sequence (rFlaA:Artv1) or a hypoallergenic variant (rFlaA:Artv1hyp) with reduced IgE-binding capacity. Immune modulating capacity of these constructs and respective controls was evaluated in vitro and in vivo. Incorporation of hypoallergenic Art v 1 derivative did not interfere with the resulting fusion proteins’ immune stimulatory capacity. Both rFlaA:Artv1 and rFlaA:Artv1hyp induced a prominent, mTOR-dependent, IL-10 secretion from murine dendritic cells, and suppressed allergen-specific TH2-cytokine secretion in vitro and in vivo. Both conjugates retained the capacity to induce rFlaA-specific antibody responses while efficiently inducing production of Art v 1-specific IgG1 and IgG2a antibodies in mice. Interestingly, only the suppression of TH2-cytokine secretion by rFlaA:Artv1 (but not rFlaA:Artv1hyp) was paralleled by a strong secretion of IFN-γ. In summary, we provided evidence that incorporating hypoallergens into flagellin:allergen fusion proteins is a suitable strategy to further improve these promising vaccine candidates.
Collapse
Affiliation(s)
- Stefan Schülke
- Section Molecular Allergology, Paul-Ehrlich-Institut, Langen, Hessen, Germany.
| | - Kirsten Kuttich
- Section Molecular Allergology, Paul-Ehrlich-Institut, Langen, Hessen, Germany
| | - Sonja Wolfheimer
- Section Molecular Allergology, Paul-Ehrlich-Institut, Langen, Hessen, Germany
| | - Nadine Duschek
- Section Molecular Allergology, Paul-Ehrlich-Institut, Langen, Hessen, Germany
| | - Andrea Wangorsch
- Section Molecular Allergology, Paul-Ehrlich-Institut, Langen, Hessen, Germany
| | - Andreas Reuter
- Division of Allergology, Paul-Ehrlich-Institut, Langen, Hessen, Germany
| | - Peter Briza
- Department of Molecular Biology, Division of Allergy and Immunology, University of Salzburg, Salzburg, Austria
| | - Isabel Pablos
- Department of Molecular Biology, Division of Allergy and Immunology, University of Salzburg, Salzburg, Austria
| | - Gabriele Gadermaier
- Department of Molecular Biology, Division of Allergy and Immunology, University of Salzburg, Salzburg, Austria
| | - Fatima Ferreira
- Department of Molecular Biology, Division of Allergy and Immunology, University of Salzburg, Salzburg, Austria
| | - Stefan Vieths
- Section Molecular Allergology, Paul-Ehrlich-Institut, Langen, Hessen, Germany
| | - Masako Toda
- Section Molecular Allergology, Paul-Ehrlich-Institut, Langen, Hessen, Germany
| | - Stephan Scheurer
- Section Molecular Allergology, Paul-Ehrlich-Institut, Langen, Hessen, Germany
| |
Collapse
|
8
|
Iwasa S, Ota H, Nishio K, Ohtsu M, Kusunoki M, Gojoubori T, Shirakawa T, Asano M. Functional expression of TLR5 in murine salivary gland epithelial cells. J Oral Sci 2017; 58:317-23. [PMID: 27665969 DOI: 10.2334/josnusd.15-0588] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
Abstract
Toll-like receptors (TLR) recognize microbe-associated molecular patterns and induce the innate immune response. Among them, TLR5 recognizes the Gram-negative bacterial component flagellin. The aim of this study was to examine the expression of TLR5 in mouse salivary gland (SG). The SG was excised from 8- to 10-week-old female C57BL/6 mice. Salivary gland epithelial cells (SGECs) were purified and subjected to reverse transcription polymerase chain reaction (RT-PCR). Western blotting was performed to detect TLR5 expression at the protein level in several organs. The localization of TLR5 in SG was examined using immunohistochemical staining. The responsiveness of SGECs to flagellin was further examined by evaluating the induction of CXCL1 by real-time PCR and immunoprecipitation followed by Western blotting. TLR5 expression in SG was confirmed at the gene and protein levels. Immunohistochemical staining detected TLR5 in both acinic and ductal cells of the sublingual gland, but not in serous acinic cells of the submandibular gland. Although TLR5 was detected throughout the cytoplasm in ductal cells, positive staining was observed on the basal side of the mucous acinic cells. The purified SGECs responded to flagellin and induced the production of CXCL1. These findings suggest that TLR5 is functionally expressed in the SG and responds to its cognate ligand flagellin. (J Oral Sci 58, 317-323, 2016).
Collapse
Affiliation(s)
- Satoko Iwasa
- Department of Pediatric Dentistry, Nihon University School of Dentistry
| | | | | | | | | | | | | | | |
Collapse
|
9
|
Verma V, Tan W, Puth S, Cho KO, Lee SE, Rhee JH. Norovirus (NoV) specific protective immune responses induced by recombinant P dimer vaccine are enhanced by the mucosal adjuvant FlaB. J Transl Med 2016; 14:135. [PMID: 27184355 PMCID: PMC4869196 DOI: 10.1186/s12967-016-0899-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Accepted: 05/07/2016] [Indexed: 12/12/2022] Open
Abstract
Background Noroviruses (NoVs) are a major cause of childhood gastroenteritis and foodborne diseases worldwide. Lack of appropriate animal models or cell-based culture systems makes the development and evaluation of NoV-specific vaccines a daunting task. VP1 is the major capsid protein of the NoVs that acts as a binding motif to human histo-blood group antigens (HBGAs) through its protruding 2 (P2) domain and can serve as a protective antigen candidate for vaccine development. Methods Recombinantly produced NoV specific P domain (Pd) vaccine was inoculated into groups of mice either alone or in conjugation with mucosal adjuvant FlaB, the flagellar protein from Vibrio vulnificus. Antigen specific humoral and cell mediated immune responses were assessed by enzyme linked immunosorbent assay (ELISA) or fluorescent activated cell sorting (FACS). A comparative analysis of various routes of vaccination viz. intranasal, sublingual and subcutaneous, was also done. Results In this study, we show that a recombinant Pd-vaccine administered through intranasal route induced a robust TH2-dependent humoral immune response and that the combination of vaccine with FlaB significantly enhanced the antibody response. Interestingly, FlaB induced a mixed TH1/TH2 type of immune response with a significant induction of IgG1 as well as IgG2a antibodies. FlaB also induced strong IgA responses in serum and feces. FlaB mediated antibody responses were toll like receptor 5 (TLR5) dependent, since the FlaB adjuvanticity was lost in TLR5−/− mice. Further, though the Pd-vaccine by itself failed to induce a cell mediated immune response, the Pd-FlaB combination stimulated a robust CD4+IFNγ+ and CD8+IFNγ+ T cell response in spleen and mesenteric lymph nodes. We also compared the adjuvant effects of FlaB with that of alum and complete Freund’s adjuvant (CFA). We found that subcutaneously inoculated FlaB induced more significant levels of IgG and IgA in both serum and feces compared to alum or CFA in respective samples. Conclusion We validate the use of TLR5 agonist as a strong mucosal adjuvant that would facilitate the development of NoV specific vaccines for humans and veterinary use. This study also highlights the importance of route of immunization in inducing the appropriate immune responses in mucosal compartments.
Collapse
Affiliation(s)
- Vivek Verma
- Clinical Vaccine R&D Center, Chonnam National University Medical School, Gwangju, South Korea.,Department of Microbiology, Chonnam National University Medical School, Gwangju, South Korea.,Georgia Cancer Center, Augusta University, 1410 Laney Walker Blvd, Augusta, 30912, Georgia, USA
| | - Wenzhi Tan
- Clinical Vaccine R&D Center, Chonnam National University Medical School, Gwangju, South Korea
| | - Sao Puth
- Clinical Vaccine R&D Center, Chonnam National University Medical School, Gwangju, South Korea
| | - Kyoung-Oh Cho
- Laboratory of Veterinary Pathology, College of Veterinary Medicine, Chonnam National University, Gwangju, South Korea
| | - Shee Eun Lee
- Clinical Vaccine R&D Center, Chonnam National University Medical School, Gwangju, South Korea.,Department of Pharmacology and Dental Therapeutics, School of Dentistry, Chonnam National University, Gwangju, South Korea
| | - Joon Haeng Rhee
- Clinical Vaccine R&D Center, Chonnam National University Medical School, Gwangju, South Korea. .,Department of Microbiology, Chonnam National University Medical School, Gwangju, South Korea.
| |
Collapse
|
10
|
Immunity Elicited by an Experimental Vaccine Based on Recombinant Flagellin-Porcine Circovirus Type 2 Cap Fusion Protein in Piglets. PLoS One 2016; 11:e0147432. [PMID: 26848967 PMCID: PMC4746120 DOI: 10.1371/journal.pone.0147432] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Accepted: 01/03/2016] [Indexed: 01/09/2023] Open
Abstract
In a recent study, we reported that a recombinant protein from fusion expression of flagellin to porcine circovirus type 2 (PCV2) Cap induced robust humoral and cell-mediated immunity that afforded full protection for PCV2 infection using BALB/c mice. Here, we further evaluated the immunogenicity and protection of the recombinant protein using specific pathogen free (SPF) pigs. Twenty-five 3-week-old piglets without passively acquired immunity were divided into 5 groups. All piglets except negative controls were challenged with a virulent PCV2 at 21 days after booster vaccination and necropsied at 21 days post-challenge. Vaccination of piglets with the recombinant protein without adjuvant induced strong humoral and cellular immune responses as observed by high levels of PCV2-specific IgG antibodies and neutralizing antibodies, as well as frequencies of PCV2-specific IFN-γ-secreting cells that conferred good protection against PCV2 challenge, with significant reduced PCV2 viremia, mild lesions, low PCV2 antigen-positive cells, as well as improved body weight gain, comparable to piglets vaccinated with a commercial PCV2 subunit vaccine. These results further demonstrated that the recombinant flagellin-Cap fusion protein is capable of inducing solid protective humoral and cellular immunity when administered to pigs, thereby becoming an effective PCV2 vaccine candidate for control of PCV2 infection.
Collapse
|
11
|
Deb R, Dey S, Madhan Mohan C, Gaikwad S, Kamble N, Khulape SA, Gupta SK, Maity HK, Pathak DC. Development and evaluation of a Salmonella typhimurium flagellin based chimeric DNA vaccine against infectious bursal disease of poultry. Res Vet Sci 2015; 102:7-14. [PMID: 26412511 DOI: 10.1016/j.rvsc.2015.07.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2015] [Revised: 06/27/2015] [Accepted: 07/05/2015] [Indexed: 02/07/2023]
Abstract
Infectious bursal disease (IBD) is an acute immunosuppressive disease of young chicks, caused by a double-stranded RNA virus. VP2 being the major capsid protein of the virus is an ideal vaccine candidate possessing the neutralizing epitopes. The present study involves the use of flagellin (fliC) as a genetic adjuvant to improve the immune response of VP2 based DNA vaccine against IBD. Our findings revealed that birds immunized with plasmid pCIVP2fliC showed robust immune response than pCIVP2 immunized groups. Further, challenge study proved that genetic fusion of fliC and VP2 can provide a comparatively higher level of protection against vvIBDV challenge in chickens than VP2 alone. These results thus indicate that Salmonella flagellin could enhance the immune responses and protection efficacy of a DNA vaccine candidate against IBDV infection in chickens, highlighting the potential of flagellin as a genetic adjuvant in the prevention of vvIBDV infection.
Collapse
Affiliation(s)
- Rajib Deb
- Recombinant DNA Laboratory, Division of Veterinary Biotechnology, Indian Veterinary Research Institute, Izatnagar, Bareilly 243122, UP, India.
| | - Sohini Dey
- Recombinant DNA Laboratory, Division of Veterinary Biotechnology, Indian Veterinary Research Institute, Izatnagar, Bareilly 243122, UP, India
| | - C Madhan Mohan
- Recombinant DNA Laboratory, Division of Veterinary Biotechnology, Indian Veterinary Research Institute, Izatnagar, Bareilly 243122, UP, India
| | - Satish Gaikwad
- Recombinant DNA Laboratory, Division of Veterinary Biotechnology, Indian Veterinary Research Institute, Izatnagar, Bareilly 243122, UP, India
| | - Nitin Kamble
- Recombinant DNA Laboratory, Division of Veterinary Biotechnology, Indian Veterinary Research Institute, Izatnagar, Bareilly 243122, UP, India
| | - Sagar A Khulape
- Recombinant DNA Laboratory, Division of Veterinary Biotechnology, Indian Veterinary Research Institute, Izatnagar, Bareilly 243122, UP, India
| | - Shishir Kumar Gupta
- Recombinant DNA Laboratory, Division of Veterinary Biotechnology, Indian Veterinary Research Institute, Izatnagar, Bareilly 243122, UP, India
| | - Hemanta Kumar Maity
- Recombinant DNA Laboratory, Division of Veterinary Biotechnology, Indian Veterinary Research Institute, Izatnagar, Bareilly 243122, UP, India
| | - Dinesh Chandra Pathak
- Recombinant DNA Laboratory, Division of Veterinary Biotechnology, Indian Veterinary Research Institute, Izatnagar, Bareilly 243122, UP, India
| |
Collapse
|
12
|
Zhang C, Zhu S, Wei L, Yan X, Wang J, Quan R, She R, Hu F, Liu J. Recombinant Flagellin-Porcine Circovirus Type 2 Cap Fusion Protein Promotes Protective Immune Responses in Mice. PLoS One 2015; 10:e0129617. [PMID: 26070075 PMCID: PMC4466504 DOI: 10.1371/journal.pone.0129617] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Accepted: 05/11/2015] [Indexed: 01/19/2023] Open
Abstract
The Cap protein of porcine circovirus type 2 (PCV2) that serves as a major host-protective immunogen was used to develop recombinant vaccines for control of PCV2-associated diseases. Growing research data have demonstrated the high effectiveness of flagellin as an adjuvant for humoral and cellular immune responses. Here, a recombinant protein was designed by fusing a modified version of bacterial flagellin to PCV2 Cap protein and expressed in a baculovirus system. When administered without adjuvant to BALB/c mice, the flagellin-Cap fusion protein elicited stronger PCV2-specific IgG antibody response, higher neutralizing antibody levels, milder histopathological changes and lower viremia, as well as higher secretion of cytokines such as TNF-α and IFN-γ that conferred better protection against virus challenge than those in the recombinant Cap alone-inoculated mice. These results suggest that the recombinant Cap protein when fused to flagellin could elicit better humoral and cellular immune responses against PCV2 infection in a mouse model, thereby acting as an attractive candidate vaccine for control of the PCV2-associated diseases.
Collapse
Affiliation(s)
- Chunyan Zhang
- Beijing Key Laboratory for Prevention and Control of Infectious Diseases in Livestock and Poultry, Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, No. 9 Shuguang Garden Middle Road, Haidian District, Beijing 100097, People’s Republic of China
| | - Shanshan Zhu
- Beijing Key Laboratory for Prevention and Control of Infectious Diseases in Livestock and Poultry, Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, No. 9 Shuguang Garden Middle Road, Haidian District, Beijing 100097, People’s Republic of China
| | - Li Wei
- Beijing Key Laboratory for Prevention and Control of Infectious Diseases in Livestock and Poultry, Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, No. 9 Shuguang Garden Middle Road, Haidian District, Beijing 100097, People’s Republic of China
| | - Xu Yan
- Beijing Key Laboratory for Prevention and Control of Infectious Diseases in Livestock and Poultry, Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, No. 9 Shuguang Garden Middle Road, Haidian District, Beijing 100097, People’s Republic of China
| | - Jing Wang
- Beijing Key Laboratory for Prevention and Control of Infectious Diseases in Livestock and Poultry, Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, No. 9 Shuguang Garden Middle Road, Haidian District, Beijing 100097, People’s Republic of China
| | - Rong Quan
- College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Haidian District, Beijing 100197, People’s Republic of China
| | - Ruiping She
- College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Haidian District, Beijing 100197, People’s Republic of China
| | - Fengjiao Hu
- College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Haidian District, Beijing 100197, People’s Republic of China
| | - Jue Liu
- Beijing Key Laboratory for Prevention and Control of Infectious Diseases in Livestock and Poultry, Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, No. 9 Shuguang Garden Middle Road, Haidian District, Beijing 100097, People’s Republic of China
- * E-mail:
| |
Collapse
|
13
|
|
14
|
Girard A, Roques E, Massie B, Archambault D. Flagellin in fusion with human rotavirus structural proteins exerts an adjuvant effect when delivered with replicating but non-disseminating adenovectors through the intrarectal route. Mol Biotechnol 2014; 56:394-407. [PMID: 24271565 DOI: 10.1007/s12033-013-9723-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Human rotavirus (HRV) is the worldwide leading cause of gastroenteritis in young children. Two live attenuated HRV vaccines have been approved since 2006. However, these live vaccines still have potential risks including reversion of virulence. Adenoviruses are suitable vectors for mucosal administration of subunit vaccines. In addition to the adjuvant effect of certain adenovirus components, the use of an adjuvant like flagellin is also another means to increase the immune response to the immunogen. The aim of this study was to determine whether flagellin in fusion with HRV structural proteins stimulates the innate immune response and enhances the HRV-specific immune response when delivered through the intrarectal route with replicating but non-disseminating adenovector (R-AdV). Salmonella typhimurium flagellin B (FljB) in fusion with HRV VP4Δ::VP7 protein induced IL-1β production in J774A.1 macrophages exposed to the R-AdV. Intrarectal administration of R-AdVs expressing either VP4Δ::VP7 or VP4Δ::VP7::FljB in BALB/c mice resulted in HRV-specific mixed Th1/Th2 immune responses. The HRV-specific antibody response elicited with the use of R-AdV expressing VP4Δ::VP7::FljB was higher than that with R-AdV expressing VP4Δ::VP7. The results also show that the replication capability of R-AdVs contributed to enhance the HRV-specific immune response as compared with that obtained with non-replicative AdVs. This work lays the foundation for using the R-AdV system and FljB-adjuvanted formulation to elicit a mucosal immune response specific to HRV.
Collapse
Affiliation(s)
- Aurélie Girard
- Department of Biological Sciences, University of Québec at Montréal, P.O. Box 8888, Succursale Centre-Ville, Montreal, QC, H3C 3P8, Canada
| | | | | | | |
Collapse
|
15
|
Taherkhani R, Farshadpour F, Makvandi M, Samarbafzadeh AR. Cloning of fliC Gene From Salmonella typhimurium in the Expression Vector pVAX1 and Evaluation of its Expression in Eukaryotic Cells. Jundishapur J Microbiol 2014; 7:e12351. [PMID: 25774273 PMCID: PMC4332235 DOI: 10.5812/jjm.12351] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2013] [Revised: 09/22/2013] [Accepted: 10/05/2013] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Flagellin is the main structural protein of the flagella of many pathogens including Salmonella typhimurium. It is a potent trigger of innate immune responses that enhance adaptive immune responses to a variety of protein antigens. Flagellin has intrinsic adjuvant activity mediated through toll-like receptor (TLR) 5 and is an attractive candidate for highly effective vaccine adjuvant conferring enhanced antibody and cellular immune responses to proteins or peptides. In the present study, we cloned the fliC gene from S. enterica typhimurium in eukaryote vector pVAX1 and evaluated its expression in eukaryotic cells. OBJECTIVES The main aim of the present study was to construct a DNA vaccine expressing fliC as an adjuvant. MATERIALS AND METHODS The fliC gene of S. typhimurium (ATCC 14028) was amplified by PCR with specific primers and cloned into the pPrime cloning vector and successfully subcloned into expression vector pVAX1. The recombinant plasmid pVAX-fliC was finally expressed in eukaryotic cells. RESULTS Cloning and subcloning of the fliC gene were confirmed by colony PCR, restriction enzymes digestion and DNA sequencing of the recombinant plasmids pPrime-fliC and pVAX-fliC. The expression of flagellin protein in eukaryotic cells was approved by immunofluorescence assay (IFA), western blotting analysis and the reverse transcriptase polymerase chain reaction (RT-PCR) method. CONCLUSIONS The results of this study demonstrated that the fliC gene in recombinant plasmid pVAX-fliC was successfully expressed in eukaryotic cells and produced flagellin protein, which could be used as an effective adjuvant for DNA vaccine research.
Collapse
Affiliation(s)
- Reza Taherkhani
- Health Research Institute, Infectious and Tropical Disease Research Center, Ahvaz Jundishapur University of Medical Science, Ahvaz, IR Iran
| | - Fatemeh Farshadpour
- Health Research Institute, Infectious and Tropical Disease Research Center, Ahvaz Jundishapur University of Medical Science, Ahvaz, IR Iran
| | - Manoochehr Makvandi
- Health Research Institute, Infectious and Tropical Disease Research Center, Ahvaz Jundishapur University of Medical Science, Ahvaz, IR Iran
| | - Ali Reza Samarbafzadeh
- Health Research Institute, Infectious and Tropical Disease Research Center, Ahvaz Jundishapur University of Medical Science, Ahvaz, IR Iran
| |
Collapse
|
16
|
Cunningham AL, Dang KM, Yu JJ, Guentzel MN, Heidner HW, Klose KE, Arulanandam BP. Enhancement of vaccine efficacy by expression of a TLR5 ligand in the defined live attenuated Francisella tularensis subsp. novicida strain U112ΔiglB::fljB. Vaccine 2014; 32:5234-40. [PMID: 25050972 DOI: 10.1016/j.vaccine.2014.07.038] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2014] [Revised: 06/11/2014] [Accepted: 07/09/2014] [Indexed: 01/13/2023]
Abstract
Oral vaccination with the defined live attenuated Francisella novicida vaccine strain U112ΔiglB has been demonstrated to induce protective immunity against pulmonary challenge with the highly human virulent Francisella tularensis strain SCHU S4. However, this vaccination regimen requires a booster dose in mice and Exhibits 50% protective efficacy in the Fischer 344 rat model. To enhance the efficacy of this vaccine strain, we engineered U112ΔiglB to express the Salmonella typhimurium FljB flagellin D1 domain, a TLR5 agonist. The U112ΔiglB::fljB strain was highly attenuated for intracellular macrophage replication, and although the FljB protein was expressed within the cytosol, it exhibited TLR5 activation in a TLR5-expressing HEK cell line. Additionally, infection of splenocytes and lymphocytes with U112ΔiglB::fljB induced significantly greater TNF-α production than infection with U112ΔiglB. Oral vaccination with U112ΔiglB::fljB also induced significantly greater protection than U112ΔiglB against pulmonary SCHU S4 challenge in rats. The enhanced protection was accompanied by higher IgG2a production and serum-mediated reduction of Francisella infectivity. Thus, the U112ΔiglB::fljB strain may serve as a potential vaccine candidate against pneumonic tularemia.
Collapse
Affiliation(s)
- Aimee L Cunningham
- South Texas Center for Emerging Infectious Disease and the Center for Excellence in Infection Genomics, University of Texas at San Antonio, San Antonio, TX 78249, United States; Department of Microbiology and Immunology, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, United States
| | - Kim Minh Dang
- South Texas Center for Emerging Infectious Disease and the Center for Excellence in Infection Genomics, University of Texas at San Antonio, San Antonio, TX 78249, United States
| | - Jieh-Juen Yu
- South Texas Center for Emerging Infectious Disease and the Center for Excellence in Infection Genomics, University of Texas at San Antonio, San Antonio, TX 78249, United States
| | - M Neal Guentzel
- South Texas Center for Emerging Infectious Disease and the Center for Excellence in Infection Genomics, University of Texas at San Antonio, San Antonio, TX 78249, United States
| | - Hans W Heidner
- South Texas Center for Emerging Infectious Disease and the Center for Excellence in Infection Genomics, University of Texas at San Antonio, San Antonio, TX 78249, United States
| | - Karl E Klose
- South Texas Center for Emerging Infectious Disease and the Center for Excellence in Infection Genomics, University of Texas at San Antonio, San Antonio, TX 78249, United States
| | - Bernard P Arulanandam
- South Texas Center for Emerging Infectious Disease and the Center for Excellence in Infection Genomics, University of Texas at San Antonio, San Antonio, TX 78249, United States.
| |
Collapse
|
17
|
Airway structural cells regulate TLR5-mediated mucosal adjuvant activity. Mucosal Immunol 2014; 7:489-500. [PMID: 24064672 DOI: 10.1038/mi.2013.66] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2013] [Revised: 07/16/2013] [Accepted: 08/13/2013] [Indexed: 02/04/2023]
Abstract
Antigen-presenting cell (APC) activation is enhanced by vaccine adjuvants. Most vaccines are based on the assumption that adjuvant activity of Toll-like receptor (TLR) agonists depends on direct, functional activation of APCs. Here, we sought to establish whether TLR stimulation in non-hematopoietic cells contributes to flagellin's mucosal adjuvant activity. Nasal administration of flagellin enhanced T-cell-mediated immunity, and systemic and secretory antibody responses to coadministered antigens in a TLR5-dependent manner. Mucosal adjuvant activity was not affected by either abrogation of TLR5 signaling in hematopoietic cells or the presence of flagellin-specific, circulating neutralizing antibodies. We found that flagellin is rapidly degraded in conducting airways, does not translocate into lung parenchyma and stimulates an early immune response, suggesting that TLR5 signaling is regionalized. The flagellin-specific early response of lung was regulated by radioresistant cells expressing TLR5 (particularly the airway epithelial cells). Flagellin stimulated the epithelial production of a small set of mediators that included the chemokine CCL20, which is known to promote APC recruitment in mucosal tissues. Our data suggest that (i) the adjuvant activity of TLR agonists in mucosal vaccination may require TLR stimulation of structural cells and (ii) harnessing the effect of adjuvants on epithelial cells can improve mucosal vaccines.
Collapse
|
18
|
Hajam IA, Dar PA, Chandrasekar S, Nanda RK, Kishore S, Bhanuprakash V, Ganesh K. Co-administration of flagellin augments immune responses to inactivated foot-and-mouth disease virus (FMDV) antigen. Res Vet Sci 2013; 95:936-41. [PMID: 23941960 DOI: 10.1016/j.rvsc.2013.07.021] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2013] [Revised: 07/13/2013] [Accepted: 07/16/2013] [Indexed: 10/26/2022]
Abstract
Foot-and-mouth disease virus (FMDV) is one of the most contagious animal virus known that affects livestock health and production. This study aimed to investigate the effect of flagellin, a toll-like receptor 5 agonist, on the immune responses to inactivated FMDV antigen in guinea pig model. Our results showed that the co-administration of flagellin with FMDV antigen through intradermal route induces earlier and higher anti-FMDV neutralizing antibody responses as compared to FMDV antigen alone. Both IgG1 and IgG2 antibody-isotype responses were enhanced, but the IgG1/IgG2 ratios were relatively low, indicative of TH1 type of immune activation. On live viral challenge, flagellin+FMDV immunized guinea pigs showed 70% (7 out of 10) protection rate as compared to 40% (4 out of 10) in FMDV alone immunized guinea pigs. The results demonstrate that the co-administration of flagellin augments immune responses (preferably TH1 type) and protective efficacy against FMDV in guinea pigs.
Collapse
Affiliation(s)
- Irshad A Hajam
- FMD Research Centre, Indian Veterinary Research Institute, Bangalore 560024, India
| | | | | | | | | | | | | |
Collapse
|
19
|
Recombinant rabies viruses expressing GM-CSF or flagellin are effective vaccines for both intramuscular and oral immunizations. PLoS One 2013; 8:e63384. [PMID: 23700422 PMCID: PMC3658976 DOI: 10.1371/journal.pone.0063384] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2013] [Accepted: 04/02/2013] [Indexed: 12/23/2022] Open
Abstract
Our previous studies indicated that recombinant rabies viruses (rRABV) expressing chemokines or cytokines (including GM-CSF) could enhance the immunogenicity by recruiting and/or activating dendritic cells (DC). In this study, bacterial flagellin was cloned into the RABV genome and recombinant virus LBNSE-Flagellin was rescued. To compare the immunogenicity of LBNSE-Flagellin with recombinant virus expressing GMCSF (LBNSE-GMCSF), mice were immunized with each of these rRABVs by intramuscular (i.m.) or oral route. The parent virus (LBNSE) without expression of any foreign molecules was included for comparison. The i.m.-immunized mice were bled at three weeks after the immunization for the measurement of virus neutralizing antibody (VNA) and then challenged with 50 LD50 challenge virus standard (CVS-24). Orally immunized mice were boosted after three weeks and then bled and challenged one week after the booster immunization. It was found that both LBNSE-GMCSF and LBNSE-Flagellin recruited/activated more DCs and B cells in the periphery, stimulated higher levels of adaptive immune responses (VNA), and protected more mice against challenge infection than the parent virus LBNSE in both the i.m. and the orally immunized groups. Together, these studies suggest that recombinant RABV expressing GM-CSF or flagellin are more immunogenic than the parent virus in both i.m. and oral immunizations.
Collapse
|
20
|
Asadi Karam MR, Oloomi M, Mahdavi M, Habibi M, Bouzari S. Vaccination with recombinant FimH fused with flagellin enhances cellular and humoral immunity against urinary tract infection in mice. Vaccine 2013; 31:1210-6. [PMID: 23306365 DOI: 10.1016/j.vaccine.2012.12.059] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2012] [Revised: 11/23/2012] [Accepted: 12/21/2012] [Indexed: 01/01/2023]
Abstract
Urinary tract infection (UTI) caused by Uropathogenic Escherichia coli (UPEC) is one of the most common infections in the world. Despite extensive efforts, a vaccine that confers protection against UTIs in human is currently lacking. In this study, the ability of flagellin (FliC), a Toll-like receptor 5 (TLR5) agonist of UPEC strain, and the conventional adjuvant Montanide ISA 206 to enhance the protective immune responses of FimH against urinary tract infection have been compared. Mice immunized with the fused FimH.FliC protein induced significantly higher humoral (IgG1 and IgG2a) and cellular (IFN-γ and IL-4) immune responses than with FimH alone or FimH admixed with FliC. The immune responses of Montanide formulations were comparable to that of the fusion protein and were significantly higher than that of FimH alone. Our results showed that based on the IgG1/IgG2a ratios, FliC directed the anti-FimH responses preferentially toward Th2 and Montanide toward Th1. The FimH.FliC fusion and FimH admixed with FliC and Montanide formulations gave the best results in protection of bladder colonization, compared to the control mice. The results propose new promising vaccine formulation based on the adjuvant properties of FliC and Montanide against UTI caused by UPEC strains.
Collapse
|
21
|
Mori J, Vranac T, Smrekar B, Cernilec M, Serbec VČ, Horvat S, Ihan A, Benčina M, Jerala R. Chimeric flagellin as the self-adjuvanting antigen for the activation of immune response against Helicobacter pylori. Vaccine 2012; 30:5856-63. [PMID: 22819990 DOI: 10.1016/j.vaccine.2012.07.011] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2012] [Revised: 06/19/2012] [Accepted: 07/08/2012] [Indexed: 12/11/2022]
Abstract
Helicobacter pylori infection can cause gastritis, peptic ulcer and can lead to gastric cancer. Lengthy antibiotic therapy does not protect the host against reinfection. H. pylori evolved to evade the recognition of the immune response by modifying several of its components whose orthologous proteins from other bacteria activate the innate immune response. Flagella are essential for the H. pylori effective colonization of human duodenum and stomach. TLR5, a member of the Toll-like receptor family, recognizes flagellin of most bacteria, such as Escherichia coli, but does not recognize the flagellin FlaA of H. pylori. We restored the ability of FlaA for the recognition by TLR5 by engineering a chimeric flagellin, in which both terminal segments of H. pylori flagellin were replaced by the corresponding segments from TLR5-activating E. coli flagellin. Recombinant chimeric flagellin folded correctly and was able to activate TLR5. Significantly increased serum IgG and IgA antibody responses were determined in mice vaccinated with chimeric flagellin in comparison to mice vaccinated with a control protein (FlaA) or negative control. Antibody titers remained high even 8 months after the last immunization. Antibodies were able to bind native flagellin from H. pylori lysate. Vaccination with chimeric flagellin provided mice with significant protection against H. pylori. The approach of chimeric flagellin can therefore generate effective immunogens that enable activation of innate and adaptive immune response and can be used to construct efficient vaccines against H. pylori or other flagellated bacteria that evade TLR5 recognition.
Collapse
Affiliation(s)
- Jerneja Mori
- Department of Biotechnology, National Institute of Chemistry, Ljubljana, Slovenia
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Construction and immunological evaluation of dual cell surface display of HIV-1 gag and Salmonella enterica serovar Typhimurium FliC in Lactobacillus acidophilus for vaccine delivery. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2012; 19:1374-81. [PMID: 22761297 DOI: 10.1128/cvi.00049-12] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Oral vaccines that elicit a mucosal immune response may be effective against human immunodeficiency virus type 1 (HIV-1) because its transmission occurs mainly at the mucosa. The aim of this study was to construct recombinant Lactobacillus for oral delivery of oral vaccines against HIV-1 and to evaluate their immunogenicity. A recombinant Lactobacillus acidophilus strain expressing the HIV-1 Gag on the bacterial cell surface was established by fusion with the signal peptide and anchor motif of a mucus binding protein (Mub) from L. acidophilus with or without coexpression of Salmonella enterica serovar Typhimurium flagellin (FliC) fused to a different Mub signal peptide and anchor. Using HEK293 cells engineered to express Toll-like receptor 5 (TLR5), the biological activity of FliC on the bacterial cell surfaces was determined. The surface-exposed flagellin retained its TLR5-stimulating activity, suggesting that the recombinant strain with Gag and FliC dual display might provide a different immunopotency than the strain expressing only Gag. The immunological properties of the recombinant strains were assessed by coculture with human myeloid dendritic cells (DCs). The heterologous antigens on the cell surface affected maturation and cytokine responses of DCs. Acquired immune responses were also investigated by intragastric immunization of mice. The enzyme-linked immunosorbent spot assay showed induction of gamma interferon-producing cells at local mucosa after immunization of mice with the Gag-producing strain. Meanwhile, the immunization with L. acidophilus displaying both Gag and FliC resulted in an increase of Gag-specific IgA-secreting cells. These results suggested that the Gag-displaying L. acidophilus elicited specific immune responses and the coexistence of FliC conferred an adjuvant effect on local IgA production.
Collapse
|
23
|
Chaung HC, Cheng LT, Hung LH, Tsai PC, Skountzou I, Wang B, Compans RW, Lien YY. Salmonella flagellin enhances mucosal immunity of avian influenza vaccine in chickens. Vet Microbiol 2012; 157:69-77. [DOI: 10.1016/j.vetmic.2011.12.014] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2011] [Revised: 11/30/2011] [Accepted: 12/08/2011] [Indexed: 10/25/2022]
|
24
|
Girard A, Saron W, Bergeron-Sandoval LP, Sarhan F, Archambault D. Flagellin produced in plants is a potent adjuvant for oral immunization. Vaccine 2011; 29:6695-703. [PMID: 21745522 DOI: 10.1016/j.vaccine.2011.06.092] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2011] [Revised: 06/14/2011] [Accepted: 06/24/2011] [Indexed: 11/24/2022]
Abstract
The aim of this study was to produce adjuvant with high biosafety, efficacy and low cost. Towards this goal, the plant Nicotiana benthamiana transient expression system was successfully used to express Salmonella typhimurium's flagellin (FljB). The yield of the expressed FljB was 280 mg per kg of fresh weight (FW) leaves. The lyophilized plant powder containing plant expressing FljB was mixed with ovalbumin (OVA) and used for oral immunization of BALB/c mice. The ELISA analysis showed higher and accelerated OVA-specific serum antibody responses in mice given the mixture when compared to animals receiving OVA alone. Furthermore, FljB elicited a mixed Th1/Th2 response as shown by the presence of specific anti-OVA IgG1, IgG2a and IgG2b isotypes. OVA-specific IgAs were also detected in mice given the mixture. Cell-mediated immune response to OVA was induced by FljB as determined by a spleen lymphocyte specific proliferation test. No immune response was generated against FljB. In conclusion, our results showed for the first time the production of FljB in plants and the efficient use of the crude lyophilized extract as an adjuvant for oral immunization.
Collapse
Affiliation(s)
- Aurélie Girard
- University of Québec at Montréal, Department of Biological Sciences, PO Box 8888, Succursale Centre-Ville, Montréal, Québec, Canada H3C 3P8
| | | | | | | | | |
Collapse
|
25
|
Efficacy of a conjugate vaccine containing polymannuronic acid and flagellin against experimental Pseudomonas aeruginosa lung infection in mice. Infect Immun 2011; 79:3455-64. [PMID: 21628521 DOI: 10.1128/iai.00157-11] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Vaccines that could effectively prevent Pseudomonas aeruginosa pulmonary infections in the settings of cystic fibrosis (CF) and nosocomial pneumonia could be exceedingly useful, but to date no effective immunotherapy targeting this pathogen has been successfully developed for routine use in humans. Evaluations using animals and limited human trials of vaccines and their associated immune effectors against different P. aeruginosa antigens have suggested that antibody to the conserved surface polysaccharide alginate, as well as the flagellar proteins, often give high levels of protection. However, alginate itself does not elicit protective antibody in humans, and flagellar vaccines containing the two predominant serotypes of this antigen may not provide sufficient coverage against variant flagellar types. To evaluate if combining these antigens in a conjugate vaccine would be potentially efficacious, we conjugated polymannuronic acid (PMA), containing the blocks of mannuronic acid conserved in all P. aeruginosa alginates, to type a flagellin (FLA) and evaluated immunogenicity, opsonic killing activity, and passive protective efficacy in mice. The PMA-FLA conjugate was highly immunogenic in mice and rabbits and elicited opsonic antibodies against mucoid but not nonmucoid P. aeruginosa, but nonetheless rabbit antibody to PMA-FLA showed evidence of protective efficacy against both types of this organism in a mouse lung infection model. Importantly, the PMA-FLA conjugate vaccine did not elicit antibodies that neutralized the Toll-like receptor 5 (TLR5)-activating activity of flagellin, an important part of innate immunity to flagellated microbial pathogens. Conjugation of PMA to FLA appears to be a promising path for developing a broadly protective vaccine against P. aeruginosa.
Collapse
|
26
|
Malaria Vaccine Development: Are Bacterial Flagellin Fusion Proteins the Bridge between Mouse and Humans? J Parasitol Res 2011; 2011:965369. [PMID: 21603205 PMCID: PMC3095412 DOI: 10.1155/2011/965369] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2010] [Accepted: 01/18/2011] [Indexed: 12/25/2022] Open
Abstract
In the past 25 years, the development of an effective malaria vaccine has become one of the biggest riddles in the biomedical sciences. Experimental data using animal infection models demonstrated that it is possible to induce protective immunity against different stages of malaria parasites. Nonetheless, the vast body of knowledge has generated disappointments when submitted to clinical conditions and presently a single antigen formulation has progressed to the point where it may be translated into a human vaccine. In parallel, new means to increase the protective effects of antigens in general have been pursued and depicted, such as the use of bacterial flagellins as carriers/adjuvants. Flagellins activate pathways in the innate immune system of both mice and humans. The recent report of the first Phase I clinical trial of a vaccine containing a Salmonella flagellin as carrier/adjuvant may fuel the use of these proteins in vaccine formulations. Herein, we review the studies on the use of recombinant flagellins as vaccine adjuvants with malarial antigens in the light of the current state of the art of malaria vaccine development. The available information indicates that bacterial flagellins should be seriously considered for malaria vaccine formulations to the development of effective human vaccines.
Collapse
|
27
|
Mizel SB, Bates JT. Flagellin as an adjuvant: cellular mechanisms and potential. THE JOURNAL OF IMMUNOLOGY 2010; 185:5677-82. [PMID: 21048152 DOI: 10.4049/jimmunol.1002156] [Citation(s) in RCA: 293] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Flagellin is a potent activator of a broad range of cell types involved in innate and adaptive immunity. An increasing number of studies have demonstrated the effectiveness of flagellin as an adjuvant, as well as its ability to promote cytokine production by a range of innate cell types, trigger a generalized recruitment of T and B lymphocytes to secondary lymphoid sites, and activate TLR5(+)CD11c(+) cells and T lymphocytes in a manner that is distinct from cognate Ag recognition. The plasticity of flagellin has allowed for the generation of a range of flagellin-Ag fusion proteins that have proven to be effective vaccines in animal models. This review summarizes the state of our current understanding of the adjuvant effect of flagellin and addresses important areas of current and future research interest.
Collapse
Affiliation(s)
- Steven B Mizel
- Department of Microbiology and Immunology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA.
| | | |
Collapse
|
28
|
Subbarayan P, Qin H, Pillai S, Lee JJ, Pfendt AP, Willing G, Miller ME, Dennis VA, Singh SR. Expression and characterization of a multivalent human respiratory syncytial virus protein. Mol Biol 2010. [DOI: 10.1134/s0026893310030106] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
29
|
Immunogenic properties of a recombinant fusion protein containing the C-terminal 19 kDa of Plasmodium falciparum merozoite surface protein-1 and the innate immunity agonist FliC flagellin of Salmonella typhimurium. Vaccine 2010; 28:2818-26. [PMID: 20170765 DOI: 10.1016/j.vaccine.2010.02.004] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2009] [Revised: 01/30/2010] [Accepted: 02/03/2010] [Indexed: 12/17/2022]
Abstract
In a recent study, we demonstrated the immunogenic properties of a new malaria vaccine polypeptide based on a 19 kDa C-terminal fragment of the merozoite surface protein-1 (MSP1(19)) from Plasmodium vivax and an innate immunity agonist, the Salmonella enterica serovar Typhimurium flagellin (FliC). Herein, we tested whether the same strategy, based on the MSP1(19) component of the deadly malaria parasite Plasmodium falciparum, could also generate a fusion polypeptide with enhanced immunogenicity. The His(6)FliC-MSP1(19) fusion protein was expressed from a recombinant Escherichia coli and showed preserved in vitro TLR5-binding activity. In contrast to animals injected with His(6)MSP1(19), mice subcutaneously immunised with the recombinant His(6)FliC-MSP1(19) developed strong MSP1(19)-specific systemic antibody responses with a prevailing IgG1 subclass. Incorporation of other adjuvants, such as CpG ODN 1826, complete and incomplete Freund's adjuvants or Quil-A, improved the IgG responses after the second, but not the third, immunising dose. It also resulted in a more balanced IgG subclass response, as evaluated by the IgG1/IgG2c ratio, and higher cell-mediated immune response, as determined by the detection of antigen-specific interferon-gamma secretion by immune spleen cells. MSP1(19)-specific antibodies recognised not only the recombinant protein, but also the native protein expressed on the surface of P. falciparum parasites. Finally, sera from rabbits immunised with the fusion protein alone inhibited the in vitro growth of three different P. falciparum strains. In summary, these results extend our previous observations and further demonstrate that fusion of the innate immunity agonist FliC to Plasmodium antigens is a promising alternative to improve their immunogenicity.
Collapse
|
30
|
Salmonella enterica serovar Typhimurium vaccine strains expressing a nontoxic Shiga-like toxin 2 derivative induce partial protective immunity to the toxin expressed by enterohemorrhagic Escherichia coli. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2010; 17:529-36. [PMID: 20147499 DOI: 10.1128/cvi.00495-09] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Shiga-like toxin 2 (Stx2)-producing enterohemorrhagic Escherichia coli (referred to as EHEC or STEC) strains are the primary etiologic agents of hemolytic-uremic syndrome (HUS), which leads to renal failure and high mortality rates. Expression of Stx2 is the most relevant virulence-associated factor of EHEC strains, and toxin neutralization by antigen-specific serum antibodies represents the main target for both preventive and therapeutic anti-HUS approaches. In the present report, we describe two Salmonella enterica serovar Typhimurium aroA vaccine strains expressing a nontoxic plasmid-encoded derivative of Stx2 (Stx2DeltaAB) containing the complete nontoxic A2 subunit and the receptor binding B subunit. The two S. Typhimurium strains differ in the expression of flagellin, the structural subunit of the flagellar shaft, which exerts strong adjuvant effects. The vaccine strains expressed Stx2DeltaAB, either cell bound or secreted into the extracellular environment, and showed enhanced mouse gut colonization and high plasmid stability under both in vitro and in vivo conditions. Oral immunization of mice with three doses of the S. Typhimurium vaccine strains elicited serum anti-Stx2B (IgG) antibodies that neutralized the toxic effects of the native toxin under in vitro conditions (Vero cells) and conferred partial protection under in vivo conditions. No significant differences with respect to gut colonization or the induction of antigen-specific antibody responses were detected in mice vaccinated with flagellated versus nonflagellated bacterial strains. The present results indicate that expression of Stx2DeltaAB by attenuated S. Typhimurium strains is an alternative vaccine approach for HUS control, but additional improvements in the immunogenicity of Stx2 toxoids are still required.
Collapse
|
31
|
Braga CJM, Massis LM, Sbrogio-Almeida ME, Alencar BCG, Bargieri DY, Boscardin SB, Rodrigues MM, Ferreira LCS. CD8+ T cell adjuvant effects of Salmonella FliCd flagellin in live vaccine vectors or as purified protein. Vaccine 2009; 28:1373-82. [PMID: 19932669 DOI: 10.1016/j.vaccine.2009.11.003] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2009] [Revised: 11/01/2009] [Accepted: 11/03/2009] [Indexed: 12/21/2022]
Abstract
Salmonella flagellin, the flagellum structural subunit, has received particular interest as a vaccine adjuvant conferring enhanced immunogenity to soluble proteins or peptides, both for activation of antibody and cellular immune responses. In the present study, we evaluated the Salmonella enterica FliCd flagellin as a T cell vaccine adjuvant using as model the 9-mer (SYVPSAEQI) synthetic H2(d)-restricted CD8(+) T cell-specific epitope (CS(280-288)) derived from the Plasmodium yoelii circumsporozoite (CS) protein. The FliCd adjuvant effects were determined under two different conditions: (i) as recombinant flagella, expressed by orally delivered live S. Dublin vaccine strains expressing the target CS(280-288) peptide fused at the central hypervariable domain, and (ii) as purified protein in acellular vaccines in which flagellin was administered to mice either as a recombinant protein fused or admixed with the target CS(280-288) peptide. The results showed that CS(280-288)-specific cytotoxic CD8(+) T cells were primed when BALB/c mice were orally inoculated with the expressing the CS(280-288) epitope S. Dublin vaccine strain. In contrast, mice immunized with purified FliCd admixed with the CS(280-288) peptide and, to a lesser extent, fused with the target peptide developed specific cytotoxic CD8(+) T cell responses without the need of a heterologous booster immunization. The CD8(+) T cell adjuvant effects of flagellin, either fused or not with the target peptide, correlated with the in vivo activation of CD11c(+) dendritic cells. Taken together, the present results demonstrate that Salmonella flagellins are flexible adjuvant and induce adaptative immune responses when administered by different routes or vaccine formulations.
Collapse
Affiliation(s)
- Catarina J M Braga
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, Av. Prof. Lineu Prestes, 1374, São Paulo, SP 05008-000, Brazil
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Weimer ET, Ervin SE, Wozniak DJ, Mizel SB. Immunization of young African green monkeys with OprF epitope 8-OprI-type A- and B-flagellin fusion proteins promotes the production of protective antibodies against nonmucoid Pseudomonas aeruginosa. Vaccine 2009; 27:6762-9. [PMID: 19744586 DOI: 10.1016/j.vaccine.2009.08.080] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2009] [Revised: 08/18/2009] [Accepted: 08/22/2009] [Indexed: 02/01/2023]
Abstract
There is currently no approved vaccine against Pseudomonas aeruginosa, the major cause of morbidity and mortality in cystic fibrosis (CF) patients and a major pathogen in ventilated and burn patients. In a previous study, we demonstrated the immunization of mice with OprF(311-341)-OprI-type A- and B-flagellin fusion proteins dramatically enhanced clearance of nonmucoid P. aeruginosa. The goal of the current study was to evaluate the ability of OprF(311-341)-OprI-flagellins to elicit the production of protective IgG in young (4-6 months old) African green monkeys. Intramuscular immunization of African green monkeys with 1, 3, 10, or 30mug of OprF(311-341)-OprI-flagellins generated robust antigen-specific IgG responses. In addition, immunization with OprF(311-341)-OprI-flagellins elicited high-affinity anti-flagellins, OprI, and OprF IgG that individually promoted extensive deposition of complement component C3 on P. aeruginosa and synergized to facilitate maximal C3 deposition. Passive immunization of mice with plasma from OprF(311-341)-OprI-flagellins immunized monkeys significantly reduced lung bacterial burden three days post-challenge compared to mice that received pre-immunization plasma. Based on our results, OprF(311-341)-OprI-A- and B-flagellin fusion proteins are highly effective in mice and nonhuman primates and thus merit additional development as a potential vaccine for use in humans.
Collapse
Affiliation(s)
- Eric T Weimer
- Department of Microbiology & Immunology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | | | | | | |
Collapse
|
33
|
Skountzou I, Martin MDP, Wang B, Ye L, Koutsonanos D, Weldon W, Jacob J, Compans RW. Salmonella flagellins are potent adjuvants for intranasally administered whole inactivated influenza vaccine. Vaccine 2009; 28:4103-12. [PMID: 19654062 DOI: 10.1016/j.vaccine.2009.07.058] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2009] [Revised: 07/01/2009] [Accepted: 07/19/2009] [Indexed: 12/24/2022]
Abstract
Bacterial flagellins are potent inducers of innate immune responses in the mouse lung because they bind to TLR5 expressed on the apical surfaces of airway epithelial cells. TLR engagement leads to the initiation of a signaling cascade that results in a pro-inflammatory response with subsequent up-regulation of several cytokines and leads to adaptive immune responses. We examined the ability of two soluble flagellins, a monomeric flagellin expressed in Escherichia coli and a highly purified polymeric flagellin directly isolated from Salmonella, to enhance the efficacy of influenza vaccines in mice. Here we demonstrate that both flagellins co-administered intranasally with inactivated A/PR/8/34 (PR8) virus induced robust increases of systemic influenza-specific IgA and IgG titers and resulted in a more comprehensive humoral response as indicated by the increase of IgG2a and IgG2b subclass responses. Groups immunized with the adjuvanted vaccines were fully protected against high dose lethal challenge by homologous virus whereas inactivated PR8 alone conferred only partial protection. Finally we show that shortly after immunization the adjuvanted vaccines induced a dramatic increase in pro-inflammatory cytokines in the lung, resulting in extensive lung infiltration by granulocytes and monocytes/macrophages. Our results reveal a promising perspective for the use of both soluble monomeric and polymeric flagellin as mucosal vaccine adjuvants to improve protection against influenza epidemics as well as a range of other infectious diseases.
Collapse
Affiliation(s)
- Ioanna Skountzou
- Department of Microbiology and Immunology and Emory Vaccine Center, Emory University School of Medicine, 1510 Clifton Rd, Atlanta, GA 30322, USA
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Bates JT, Uematsu S, Akira S, Mizel SB. Direct stimulation of tlr5+/+ CD11c+ cells is necessary for the adjuvant activity of flagellin. THE JOURNAL OF IMMUNOLOGY 2009; 182:7539-47. [PMID: 19494277 DOI: 10.4049/jimmunol.0804225] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Flagellin is a highly effective adjuvant, but the cellular mechanism underlying this activity remains uncertain. More specifically, no consensus exists as to whether flagellin activates dendritic cells (DC) directly or indirectly. Intramuscular immunization with flagellin-OVA fusion protein resulted in enhanced in vivo T cell clustering in draining lymph nodes and IL-2 production by OVA-specific CD4(+) T cells. Immunization with flagellin-OVA also triggered greater levels of Ag-specific CD4(+) T cell proliferation than immunization with flagellin and OVA as separate proteins. To determine whether flagellin, in the context of a fusion protein with OVA, was acting directly on DC, we used a combination of CD4(+) T cell adoptive transfers and bone marrow chimera mice in which the presence or absence of potential tlr5(+/+) CD11c(+) cells was controlled by injection of diphtheria toxin. The Ag-specific CD4(+) T cell response in mice with CD11c(+) cells from a tlr5(-/-) background and mixed populations of all other hematopoietic cells was dramatically reduced in comparison to mice that had DC from tlr5(-/-) and wild-type backgrounds. Immunization of MyD88(-/-)tlr5(+/+) mice revealed that the enhanced response following immunization with flagellin-OVA is dependent on signaling via the TLR5-MyD88 pathway as well as enhanced Ag uptake and processing resulting from Ag targeting via TLR5. In summary, our data are consistent with the conclusion that direct stimulation of tlr5(+/+) CD11c(+) cells is necessary for the adjuvant activity of a flagellin fusion protein and that this adjuvant effect requires signaling through TLR5.
Collapse
Affiliation(s)
- John T Bates
- Department of Microbiology and Immunology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | | | | | | |
Collapse
|
35
|
Janot L, Sirard JC, Secher T, Noulin N, Fick L, Akira S, Uematsu S, Didierlaurent A, Hussell T, Ryffel B, Erard F. Radioresistant cells expressing TLR5 control the respiratory epithelium's innate immune responses to flagellin. Eur J Immunol 2009; 39:1587-96. [PMID: 19424969 DOI: 10.1002/eji.200838907] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Bacterial products (such as endotoxins and flagellin) trigger innate immune responses through TLRs. Flagellin-induced signalling involves TLR5 and MyD88 and, according to some reports, TLR4. Whereas epithelial and dendritic cells are stimulated by flagellin in vitro, the cell contribution to the in vivo response is still unclear. Here, we studied the respective roles of radioresistant and radiosensitive cells in flagellin-induced airway inflammation in mice. We found that i.n. delivery of flagellin elicits a transient change in respiratory function and an acute, pro-inflammatory response in the lungs, characterized by TLR5- and MyD88-dependent chemokine secretion and neutrophil recruitment. In contrast, TLR4, CD14 and TRIF were not essential for flagellin-mediated responses, indicating that TLR4 does not cooperate with TLR5 in the lungs. Respiratory function, chemokine secretion and airway infiltration by neutrophils were dependent on radioresistant, TLR5-expressing cells. Furthermore, lung haematopoietic cells also responded to flagellin by activating TNF-alpha production. We suggest that the radioresistant lung epithelial cells are essential for initiating early, TLR5-dependent signalling in response to flagellin and thus triggering the lung's innate immune responses.
Collapse
Affiliation(s)
- Laure Janot
- University of Orleans and CNRS, Molecular Immunology and Embryology, UMR 6218, Institut de Transgénose, Orléans, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
A fusion protein vaccine containing OprF epitope 8, OprI, and type A and B flagellins promotes enhanced clearance of nonmucoid Pseudomonas aeruginosa. Infect Immun 2009; 77:2356-66. [PMID: 19349426 DOI: 10.1128/iai.00054-09] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Although chronic Pseudomonas aeruginosa infection is the major cause of morbidity and mortality in cystic fibrosis (CF) patients, there is no approved vaccine for human use against P. aeruginosa. The goal of this study was to establish whether a multivalent vaccine containing P. aeruginosa type A and B flagellins as well as the outer membrane proteins OprF and OprI would promote enhanced clearance of P. aeruginosa. Intramuscular immunization with flagellins and OprI (separate) or OprI-flagellin fusion proteins generated significant antiflagellin immunoglobulin G (IgG) responses. However, only the fusions of OprI with type A and type B flagellins generated OprI-specific IgG. Immunization with a combination of OprF epitope 8 (OprF(311-341)), OprI, and flagellins elicited high-affinity IgG antibodies specific to flagellins, OprI, and OprF that individually promoted extensive deposition of C3 on P. aeruginosa. Although these antibodies exhibited potent antibody-dependent complement-mediated killing of nonmucoid bacteria, they were significantly less effective with mucoid isolates. Mice immunized with the OprF(311-341)-OprI-flagellin fusion had a significantly lower bacterial burden three days postchallenge and cleared the infection significantly faster than control mice. In addition, mice immunized with the OprF(311-341)-OprI-flagellin fusion had significantly less inflammation and lung damage throughout the infection than OprF-OprI-immunized mice. Based on our results, OprF(311-341)-OprI-flagellin fusion proteins have substantial potential as components of a vaccine against nonmucoid P. aeruginosa, which appears to be the phenotype of the bacterium that initially colonizes CF patients.
Collapse
|
37
|
Paracoccidioides brasiliensis vaccine formulations based on the gp43-derived P10 sequence and the Salmonella enterica FliC flagellin. Infect Immun 2009; 77:1700-7. [PMID: 19204092 DOI: 10.1128/iai.01470-08] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Paracoccidioidomycosis (PCM) is a systemic granulomatous disease caused by the dimorphic fungus Paracoccidioides brasiliensis. Anti-PCM vaccine formulations based on the secreted fungal cell wall protein (gp43) or the derived P10 sequence containing a CD4(+) T-cell-specific epitope have shown promising results. In the present study, we evaluated new anti-PCM vaccine formulations based on the intranasal administration of P. brasiliensis gp43 or the P10 peptide in combination with the Salmonella enterica FliC flagellin, an innate immunity agonist binding specifically to the Toll-like receptor 5, in a murine model. BALB/c mice immunized with gp43 developed high-specific-serum immunoglobulin G1 responses and enhanced interleukin-4 (IL-4) and IL-10 levels. On the other hand, mice immunized with recombinant purified flagellins genetically fused with P10 at the central hypervariable domain, either flanked or not by two lysine residues, or the synthetic P10 peptide admixed with purified FliC elicited a prevailing Th1-type immune response based on lung cell-secreted type 1 cytokines. Mice immunized with gp43 and FliC and intratracheally challenged with P. brasiliensis yeast cells had increased fungal proliferation and lung tissue damage. In contrast, mice immunized with the chimeric flagellins and particularly those immunized with P10 admixed with FliC reduced P. brasiliensis growth and lung damage. Altogether, these results indicate that S. enterica FliC flagellin modulates the immune response to P. brasiliensis P10 antigen and represents a promising alternative for the generation of anti-PCM vaccines.
Collapse
|
38
|
Flagellin-F1-V fusion protein is an effective plague vaccine in mice and two species of nonhuman primates. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2008; 16:21-8. [PMID: 18987167 DOI: 10.1128/cvi.00333-08] [Citation(s) in RCA: 111] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
A number of studies have clearly demonstrated that flagellin is a potent adjuvant that promotes robust immune responses when it is given with a protein antigen. In view of the potential biological and practical benefits of a recombinant protein vaccine composed of a single fusion protein containing flagellin and antigen, we have evaluated the efficacy of a fusion protein composed of flagellin and two protective antigens of Yersinia pestis (F1 and V) in eliciting protection against respiratory challenge with Y. pestis. Flagellin-F1-V was produced and purified in high yield under good manufacturing practices conditions. The fusion protein retains full Toll-like receptor 5-stimulating activity in vitro. Using a prime-boost immunization protocol, we found that flagellin-F1-V elicits robust antigen-specific humoral immunity in mice and two species of nonhuman primates. Immune mice were fully protected against intranasal challenge with 150 mean tolerated doses of Y. pestis CO92. In immune mice, the bacteria were completely cleared within 3 days after challenge. Flagellin-F1-V exhibited full stability for at least 297 days at 4 degrees C and at least 168 days at 25 degrees C. At between 29 and 84 days at 37 degrees C, the protein exhibited a loss of biological activity that appeared to be associated with a substantial change in protein diameter, possibly due to oligomerization. On the basis of our results, we believe that flagellin-F1-V is an outstanding candidate for evaluation in studies with humans.
Collapse
|
39
|
New malaria vaccine candidates based on the Plasmodium vivax Merozoite Surface Protein-1 and the TLR-5 agonist Salmonella Typhimurium FliC flagellin. Vaccine 2008; 26:6132-42. [DOI: 10.1016/j.vaccine.2008.08.070] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2008] [Revised: 07/31/2008] [Accepted: 08/31/2008] [Indexed: 11/21/2022]
|
40
|
Dearman RJ, Cumberbatch M, Maxwell G, Basketter DA, Kimber I. Toll-like receptor ligand activation of murine bone marrow-derived dendritic cells. Immunology 2008; 126:475-84. [PMID: 18778283 DOI: 10.1111/j.1365-2567.2008.02922.x] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Dendritic cells (DCs) are required for the initiation of primary immune responses. The pattern of Toll-like receptor (TLR) expression on various subsets of these cells has been shown to differ, suggestive of distinct roles in influencing immune responses. We have examined here the responses of immature DCs derived from murine bone marrow (BMDCs) to a range of TLR ligands. BMDCs cultured for 6 days in the presence of granulocyte-macrophage colony-stimulating factor were stimulated for 24 hr with ligands to TLR1-2 [Pam(3)Cys-Ser-(Lys)(4) (PAM)], TLR2-6 (macrophage-activating lipopeptide-2 (MALP-2); zymosan or peptidoglycan (PG)], TLR3 (polyinosinic-polycytidylic acid), TLR4 [lipopolysaccharide R515 (LPS)], TLR5 (flagellin), TLR7 (polyuridylic acid) and TLR9 [CpG ODN2395 (CpG)]. DC activation was monitored using membrane marker expression and analysis of culture supernatants for cytokine/chemokine release. Ligands to TLR3 and TLR7 failed to activate BMDCs. All other TLR ligands caused elevated expression of membrane markers. PAM, MALP-2 and LPS induced high-level expression of proinflammatory cytokines and chemokines. Treatment with CpG was associated with a preferential type 1 cytokine and chemokine profile. Zymosan and PG were proinflammatory but also skewed towards a type 2 pattern of cytokines and chemokines. In contrast, flagellin did not cause marked secretion by BMDCs of cytokines or chemokines. These data for BMDCs are largely consistent with the reported TLR repertoire of freshly isolated murine Langerhans cells. In addition, murine BMDCs show selective responses to TLR ligands with respect to general activation, with differentiated cytokine patterns suggestive of potential priming for divergent immune responses.
Collapse
|
41
|
Nempont C, Cayet D, Rumbo M, Bompard C, Villeret V, Sirard JC. Deletion of flagellin's hypervariable region abrogates antibody-mediated neutralization and systemic activation of TLR5-dependent immunity. THE JOURNAL OF IMMUNOLOGY 2008; 181:2036-43. [PMID: 18641341 DOI: 10.4049/jimmunol.181.3.2036] [Citation(s) in RCA: 106] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
TLRs trigger immunity by detecting microbe-associated molecular patterns (MAMPs). Flagellin is a unique MAMP because it harbors 1) an antigenic hypervariable region and 2) a conserved domain involved in TLR5-dependent systemic and mucosal proinflammatory and adjuvant activities. In this study, the contribution of the flagellin domains in TLR5 activation was investigated. We showed that TLR5 signaling can be neutralized in vivo by flagellin-specific Abs, which target the conserved domain. However, deletions of flagellin's hypervariable region abrogated the protein's intrinsic ability to trigger the production of neutralizing Abs. The fact that MAMP-specific Abs block TLR-mediated responses shows that this type of neutralization is a novel mechanism for down-regulating innate immunity. The stimulation of mucosal innate immunity and adjuvancy to foreign Ag was not altered by the hypervariable domain deletions. In contrast, this domain is essential to trigger systemic innate immunity, suggesting that there are distinct mechanisms for TLR5 activation in systemic and mucosal compartments. In summary, specific MAMP determinants control the production of neutralizing Abs and the compartmentalization of innate responses.
Collapse
Affiliation(s)
- Clément Nempont
- Institut National de la Santé et de la Recherche Médicale, Unité 801, Institut Pasteur de Lille, Université de Lille 2, Lille, France
| | | | | | | | | | | |
Collapse
|
42
|
Braga CJM, Massis LM, Alencar BCG, Rodrigues MM, Sbrogio-Almeida ME, Ferreira LCS. Cytotoxic T cell adjuvant effects of three Salmonella enterica flagellins. Braz J Microbiol 2008; 39:44-9. [PMID: 24031176 PMCID: PMC3768349 DOI: 10.1590/s1517-838220080001000011] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2007] [Revised: 10/17/2007] [Accepted: 01/21/2008] [Indexed: 02/02/2023] Open
Abstract
Bacterial flagellins are important virulence-associated factors and strong inducers of inflammatory responses in mammalian hosts. Flagellins have also been investigated as potential vaccine adjuvants, either for induction of humoral or cellular immune responses, to different target antigens. In this study we investigated the adjuvant properties of three Salmonella enterica flagellins types (FliCd, FliCi and FljB) to an ovalbumin-derived CD8+ T cell-restricted epitope (OVA257–264). Although mice immunized with the three tested flagellins elicited antigen-specific activated CD8+ T cells, only animals immunized with FliCi and FliCd flagellins admixed with ovalbumin mounted specific in vivo cytotoxic responses to peptide-pulsed target cells. The present results indicate that Salmonella flagellins are endowed with type-specific adjuvant effects toward murine CD8+ T cells, a feature that may impact their use as adjuvants for prophylatic or therapeutic vaccines.
Collapse
Affiliation(s)
- Catarina J M Braga
- Departmento de Microbiologia, Instituto de Ciências Biomédicas, Universidade de São Paulo , São Paulo, SP , Brasil
| | | | | | | | | | | |
Collapse
|
43
|
Bates JT, Honko AN, Graff AH, Kock ND, Mizel SB. Mucosal adjuvant activity of flagellin in aged mice. Mech Ageing Dev 2008; 129:271-81. [PMID: 18367233 DOI: 10.1016/j.mad.2008.01.009] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2007] [Revised: 12/11/2007] [Accepted: 01/25/2008] [Indexed: 01/22/2023]
Abstract
We evaluated the ability of flagellin, a highly effective mucosal adjuvant in mice and non-human primates, to promote mucosal innate and adaptive immunity in aged mice. We found that intratracheal instillation of flagellin induced a stronger respiratory innate response in aged mice than in young mice, and that intranasal instillation of flagellin was equally effective at triggering recruitment of T and B lymphocytes to the draining lymph nodes of young and aged mice. Intranasal immunization of aged mice with flagellin and the Yersinia pestis protein F1 promoted specific IgG and IgA production, but at lower levels and lower avidities than in young mice. Although intranasal instillation of flagellin and F1 antigen increased germinal center formation and size in young mice, it did not do so in aged mice. Our findings are consistent with the conclusion that flagellin can promote adaptive immune responses in aged mice, but at a less robust level than in young mice.
Collapse
Affiliation(s)
- John T Bates
- Department of Microbiology and Immunology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | | | | | | | | |
Collapse
|
44
|
|
45
|
Rochon M, Römling U. Flagellin in combination with curli fimbriae elicits an immune response in the gastrointestinal epithelial cell line HT-29. Microbes Infect 2006; 8:2027-33. [PMID: 16807036 DOI: 10.1016/j.micinf.2006.03.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2005] [Revised: 01/31/2006] [Accepted: 03/02/2006] [Indexed: 11/28/2022]
Abstract
Flagellin is the major cytokine-releasing factor when Salmonella enterica serovar Typhimurium (S. Typhimurium) infects intestinal epithelial cells. In this work it is shown that curli, an adhesive proteinaceous surface component of Enterobacteriaceae involved in biofilm formation of S. Typhimurium and Escherichia coli strains can bind flagellin and thus elicit an immune response by the intestinal epithelial cell line HT-29.
Collapse
Affiliation(s)
- Maike Rochon
- Microbiology and Tumor Biology Center, Karolinska Institutet, Box 280, SE-171 77 Stockholm, Sweden
| | | |
Collapse
|
46
|
Rumbo M, Nempont C, Kraehenbuhl JP, Sirard JC. Mucosal interplay among commensal and pathogenic bacteria: lessons from flagellin and Toll-like receptor 5. FEBS Lett 2006; 580:2976-84. [PMID: 16650409 DOI: 10.1016/j.febslet.2006.04.036] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2006] [Accepted: 04/08/2006] [Indexed: 12/19/2022]
Abstract
Toll-like receptors (TLR) detect pathogen-associated molecular patterns (PAMP) and play a crucial role in triggering immunity. Due to their large surfaces in direct contact with the environment, mucosal tissues are the major sites of PAMP-TLR signalling. How innate and adaptive immunity are triggered through flagellin-TLR5 interaction is the main focus of the review. In view of recent reports on genetic polymorphism, we will summarize the impact of TLR5 on the susceptibility to mucosal infections and on various immuno-pathologies. Finally, the contribution of TLRs in the induction and maintenance of mucosal homeostasis and commensal discrimination is discussed.
Collapse
Affiliation(s)
- Martin Rumbo
- UNLP, Laboratorio de Investigaciones en el Sistema Inmune, Facultad de Ciencias Exactas, La Plata, Argentina
| | | | | | | |
Collapse
|