1
|
Sasaki E, Asanuma H, Momose H, Maeyama JI, Moriyama S, Nagata N, Suzuki T, Hamaguchi I, Hasegawa H, Takahashi Y. Calboxyvinyl polymer adjuvant enhances respiratory iga responses through mucosal and systemic administration. NPJ Vaccines 2025; 10:28. [PMID: 39934182 DOI: 10.1038/s41541-025-01086-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Accepted: 02/04/2025] [Indexed: 02/13/2025] Open
Abstract
Adjuvants play a crucial role in enhancing vaccine efficacy. Although several adjuvants have been approved, there remains a demand for safer and more effective adjuvants for nasal vaccines. Here, we identified calboxyvinyl polymer (CVP) as a superior mucosal vaccine adjuvant from pharmaceutical base materials using our screening systems; single nasal vaccination of the CVP-combined influenza split vaccine-induced antigen-specific IgA and IgG antibodies and provided protection against lethal influenza virus infection. Furthermore, nasal vaccination with CVP-combined severe acute respiratory syndrome coronavirus 2 antigen protected against the virus and stimulated the production of highly cross-reactive IgG antibodies against variants XBB1.5 and JN.1. Intriguingly, intramuscular vaccination of the CVP-combined vaccine also elicited the production of IgA antibodies in both nasal wash and bronchoalveolar lavage fluid in mice and cynomolgus monkeys. CVP therefore offers superior adjuvanticity to existing adjuvants and is anticipated to be a safe and effective adjuvant for mucosal vaccines.
Collapse
Affiliation(s)
- Eita Sasaki
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku, Tokyo, Japan.
- Center for Influenza and Respiratory Virus Research, National Institute of Infectious Diseases, 4-7-1 Gakuen, Musashi-Murayama, Tokyo, Japan.
| | - Hideki Asanuma
- Center for Influenza and Respiratory Virus Research, National Institute of Infectious Diseases, 4-7-1 Gakuen, Musashi-Murayama, Tokyo, Japan
| | - Haruka Momose
- Research Center for Biological Products in the Next Generation, National Institute of Infectious Diseases, 4-7-1 Gakuen, Musashi-Murayama, Tokyo, Japan
| | - Jun-Ichi Maeyama
- Research Center for Biological Products in the Next Generation, National Institute of Infectious Diseases, 4-7-1 Gakuen, Musashi-Murayama, Tokyo, Japan
| | - Saya Moriyama
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku, Tokyo, Japan
| | - Noriyo Nagata
- Department of Pathology, National Institute of Infectious Diseases, 4-7-1 Gakuen, Musashi-Murayama, Tokyo, Japan
| | - Tadaki Suzuki
- Department of Pathology, National Institute of Infectious Diseases, 4-7-1 Gakuen, Musashi-Murayama, Tokyo, Japan
| | - Isao Hamaguchi
- Research Center for Biological Products in the Next Generation, National Institute of Infectious Diseases, 4-7-1 Gakuen, Musashi-Murayama, Tokyo, Japan
- Department of Clinical Laboratory, Subaru Health Insurance Society Ota Memorial Hospital, 455-1, Oshima-cho, Ota, Gumma, Japan
| | - Hideki Hasegawa
- Center for Influenza and Respiratory Virus Research, National Institute of Infectious Diseases, 4-7-1 Gakuen, Musashi-Murayama, Tokyo, Japan
| | - Yoshimasa Takahashi
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku, Tokyo, Japan
| |
Collapse
|
2
|
Li X, Zhang ZW, Zhang FD, Li JH, Lv JL, Zhang LP, Zhai KG, Wang YL, Guo HC, Liu XS, Pan L. Double synergic chitosan-coated poly (lactic-co-glycolic) acid nanospheres loaded with nucleic acids as an intranasally administered vaccine delivery system to control the infection of foot-and-mouth disease virus. Antiviral Res 2024; 226:105900. [PMID: 38705200 DOI: 10.1016/j.antiviral.2024.105900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 03/16/2024] [Accepted: 05/03/2024] [Indexed: 05/07/2024]
Abstract
BACKGROUND & AIMS The spread of foot-and-mouth disease virus (FMDV) through aerosol droplets among cloven-hoofed ungulates in close contact is a major obstacle for successful animal husbandry. Therefore, the development of suitable mucosal vaccines, especially nasal vaccines, to block the virus at the initial site of infection is crucial. PATIENTS AND METHODS Here, we constructed eukaryotic expression plasmids containing the T and B-cell epitopes (pTB) of FMDV in tandem with the molecular mucosal adjuvant Fms-like tyrosine kinase receptor 3 ligand (Flt3 ligand, FL) (pTB-FL). Then, the constructed plasmid was electrostatically attached to mannose-modified chitosan-coated poly(lactic-co-glycolic) acid (PLGA) nanospheres (MCS-PLGA-NPs) to obtain an active nasal vaccine targeting the mannose-receptor on the surface of antigen-presenting cells (APCs). RESULTS The MCS-PLGA-NPs loaded with pTB-FL not only induced a local mucosal immune response, but also induced a systemic immune response in mice. More importantly, the nasal vaccine afforded an 80% protection rate against a highly virulent FMDV strain (AF72) when it was subcutaneously injected into the soles of the feet of guinea pigs. CONCLUSIONS The nasal vaccine prepared in this study can effectively induce a cross-protective immune response against the challenge with FMDV of same serotype in animals and is promising as a potential FMDV vaccine.
Collapse
Affiliation(s)
- Xian Li
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, National Foot-and-Mouth Diseases Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China; Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730046, China
| | - Zhong-Wang Zhang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, National Foot-and-Mouth Diseases Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China; Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730046, China
| | - Fu-Dong Zhang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, National Foot-and-Mouth Diseases Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China; Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730046, China
| | - Jia-Hao Li
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, National Foot-and-Mouth Diseases Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China; Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730046, China
| | - Jian-Liang Lv
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, National Foot-and-Mouth Diseases Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China; Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730046, China
| | - Li-Ping Zhang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, National Foot-and-Mouth Diseases Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China; Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730046, China
| | - Kai-Ge Zhai
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, National Foot-and-Mouth Diseases Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China; Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730046, China
| | - Yong-Lu Wang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, National Foot-and-Mouth Diseases Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China; Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730046, China
| | - Hui-Chen Guo
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, National Foot-and-Mouth Diseases Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China; Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730046, China
| | - Xin-Sheng Liu
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, National Foot-and-Mouth Diseases Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China; Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730046, China.
| | - Li Pan
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, National Foot-and-Mouth Diseases Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China; Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730046, China.
| |
Collapse
|
3
|
Koyanagi K, Kataoka K, Yoshimatsu H, Fujihashi K, Miyake T. Human salivary protein-derived peptides specific-salivary SIgA antibodies enhanced by nasal double DNA adjuvant in mice play an essential role in preventing Porphyromonas gingivalis colonization: an in-vitro study. BMC Oral Health 2023; 23:123. [PMID: 36829152 PMCID: PMC9950703 DOI: 10.1186/s12903-023-02821-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 02/15/2023] [Indexed: 02/26/2023] Open
Abstract
BACKGROUND We previously showed that fimbriae-bore from Poryphyromonas gingivalis (Pg), one of the putative periodontopathogenic bacteria specifically bound to a peptide domain (stat23, prp21) shared on statherin or acidic proline-rich protein 1 (PRP1) molecule of human salivary proteins (HSPs). Here, we investigated whether the nasal administration of DNA plasmid expressing Flt3 ligand (pFL) and CpG oligodeoxynucleotide 1826 as double DNA adjuvant (dDA) with stat23 and prpr21 induces antigen (Ag)-specific salivary secretory IgA (SIgA) antibodies (Abs) in mice. Further, we examined that stat23- and prpr21-specific salivary SIgA Abs induced by dDA have an impact on Pg-binding to human whole saliva-coated hydroxyapatite beads (wsHAPs). MATERIAL AND METHODS C57BL/6N mice were nasally immunized with dDA plus sta23 or/and prp21 peptide as Ag four times at weekly intervals. Saliva was collected one week after the final immunization and was subjected to Ag-specific ELISA. To examine the functional applicability of Ag-specific SIgA Abs, SIgA-enriched saliva samples were subjected to Pg binding inhibition assay to wsHAPs. RESULTS Significantly elevated levels of salivary SIgA Ab to stat23 or prp21 were seen in mice given nasal stat23 or prp21 with dDA compared to those in mice given Ag alone. Of interest, mice nasally given the mixture of stat23 and prp21 as double Ags plus dDA, resulted in both stat23- and prp21-specific salivary SIgA Ab responses, which are mediated through significantly increased numbers of CD11c+ dendritic cell populations and markedly elevated Th1 and Th2 cytokines production by CD4+ T cells in the mucosal inductive and effector tissues. The SIgA Ab-enriched saliva showed significantly reduced numbers of live Pg cells binding to wsHAPs as compared with those in mice given double Ags without dDA or naïve mice. Additionally, saliva from IgA-deficient mice given nasal double Ags plus dDA indicated no decrease of live Pg binding to wsHAPs. CONCLUSION These findings show that HSP-derived peptides-specific salivary SIgA Abs induced by nasal administration of stat23 and prp21 peptides plus dDA, play an essential role in preventing Pg attachment and colonization on the surface of teeth, suggesting a potency that the SIgA may interrupt and mask fimbriae-binding domains in HSPs on the teeth.
Collapse
Affiliation(s)
- Kayo Koyanagi
- Department of Preventive and Community Dentistry, Graduate School of Dentistry, Osaka Dental University, 1-8 Kuzuha Hanazono-Cho, Hirakata-Shi, Osaka, 573-1121, Japan
| | - Kosuke Kataoka
- Department of Preventive and Community Dentistry, Graduate School of Dentistry, Osaka Dental University, 1-8 Kuzuha Hanazono-Cho, Hirakata-Shi, Osaka, 573-1121, Japan. .,Department of Oral Health Science and Social Welfare, Graduate School of Oral Sciences, Tokushima University, 3-18-15 Kuramoto-Cho, Tokushima-Shi, Tokushima, 770-8504, Japan.
| | - Hideki Yoshimatsu
- Department of Preventive and Community Dentistry, Graduate School of Dentistry, Osaka Dental University, 1-8 Kuzuha Hanazono-Cho, Hirakata-Shi, Osaka, 573-1121, Japan
| | - Kohtaro Fujihashi
- Department of Human Mucosal Vaccinology, Chiba University Hospital, Research Institute of Disaster Medicine, Chiba University, 1-8-1 Inohana, Chuo-Ku, Chiba-Shi, Chiba, Japan.,Division of Mucosal Vaccine, International Vaccine Design Center, The Institute of Medical Science, The University of Tokyo, 4-6-1, Shiroganedai, Minato-Ku, Tokyo, 108-8639, Japan.,Department of Pediatric Dentistry, School of Dentistry, The University of Alabama at Birmingham, 1919 7Th Avenue South, Birmingham, AL, 35233, USA
| | - Tatsuro Miyake
- Department of Preventive and Community Dentistry, Graduate School of Dentistry, Osaka Dental University, 1-8 Kuzuha Hanazono-Cho, Hirakata-Shi, Osaka, 573-1121, Japan
| |
Collapse
|
4
|
Nano-Encapsulated Antioxidant: Retinoic Acid as a Natural Mucosal Adjuvant for Intranasal Immunization against Chronic Experimental Toxoplasmosis. Trop Med Infect Dis 2023; 8:tropicalmed8020106. [PMID: 36828522 PMCID: PMC9962073 DOI: 10.3390/tropicalmed8020106] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 01/12/2023] [Accepted: 02/02/2023] [Indexed: 02/10/2023] Open
Abstract
The tight relationship between immunity and retinoid levels provides evidence on the critical role of retinoic acid (RA) in regulating immune activity, especially the mucosal one. Mucosal immune response is the key for determination of the outcome of infection, particularly against intracellular mucosal pathogens such as Toxoplasma gondii, where it plays a crucial role as a sentinel against parasite invasion. Herein, the immunomodulatory adjuvant role of RA was evaluated for prophylactic vaccination against chronic Toxoplasma infection. A quantity of 15 µg of RA pre-encapsulated with lipid-based nanoparticles (SLNs) was intranasally used in three doses, two weeks apart, as an adjuvant to the Toxoplasma lysate antigen (TLA). Afterward, mice were infected with 20 cysts of T. gondii (ME49 strain) and were sacrificed at the 4th week post-infection. Parasitological, immunological, biochemical, and histopathological studies were applied as vaccine efficacy measures. The protective role of the tested vaccine was noted using the statistically marked reduction in brain cyst count, accompanied by remarkable levels of protective IFN-γ and antibodies, with amelioration of infection-induced oxidative stress and brain pathology. Ultimately, this experiment outlined the prospective role of a novel, natural, nano-encapsulated and mucosal vaccine adjuvant RA-SLNs as a propitious candidate against chronic toxoplasmosis.
Collapse
|
5
|
Said DE, Amer EI, Sheta E, Makled S, Diab HE, Arafa FM. Nano-Encapsulated Melatonin: A Promising Mucosal Adjuvant in Intranasal Immunization against Chronic Experimental T. gondii Infection. Trop Med Infect Dis 2022; 7:tropicalmed7120401. [PMID: 36548656 PMCID: PMC9785012 DOI: 10.3390/tropicalmed7120401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 11/17/2022] [Accepted: 11/24/2022] [Indexed: 11/29/2022] Open
Abstract
Melatonin (MLT) is now emerging as one of the universally accepted immunostimulators with broad applications in medicine. It is a biological manipulator of the immune system, including mucosal ones. MLT was encapsulated in solid lipid nanoparticles (SLNs), then 100 mg/kg/dose of MLT-SLNs was used as an adjuvant of Toxoplasma lysate antigen (TLA). Experimental mice were intra-nasally inoculated with three doses of different regimens every two weeks, then challenged with 20 cysts of T. gondii Me49 strain, where they were sacrificed four weeks post-infection. Protective vaccine efficacy was evident via the significant brain cyst count reduction of 58.6%, together with remarkably high levels of humoral systemic and mucosal anti-Toxoplasma antibodies (Ig G, Ig A), supported by a reduced tachyzoites invasion of Vero cells in vitro upon incubation with sera obtained from these vaccinated mice. A cellular immune response was evident through the induction of significant levels of interferon-gamma (IFN γ), associated with morphological deteriorations of cysts harvested from the brains of vaccinated mice. Furthermore, the amelioration of infection-induced oxidative stress (OS) and histopathological changes were evident in mice immunized with TLA/MLT-SLNs. In conclusion, the present study highlighted the promising role of intranasal MLT-SLNs as a novel mucosal adjuvant candidate against chronic toxoplasmosis.
Collapse
Affiliation(s)
- Doaa E. Said
- Department of Medical Parasitology, Faculty of Medicine, Alexandria University, Alexandria 5424041, Egypt
| | - Eglal I. Amer
- Department of Medical Parasitology, Faculty of Medicine, Alexandria University, Alexandria 5424041, Egypt
| | - Eman Sheta
- Department of Pathology, Faculty of Medicine, Alexandria University, Alexandria 5424041, Egypt
| | - Shaimaa Makled
- Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
| | - Hala E. Diab
- Department of Medical Parasitology, Faculty of Medicine, Alexandria University, Alexandria 5424041, Egypt
- Correspondence:
| | - Fadwa M. Arafa
- Department of Medical Parasitology, Faculty of Medicine, Alexandria University, Alexandria 5424041, Egypt
| |
Collapse
|
6
|
Yoshimatsu H, Kataoka K, Fujihashi K, Miyake T, Ono Y. A nasal double DNA adjuvant system induces atheroprotective IgM antibodies via dendritic cell-B-1a B cell interactions. Vaccine 2022; 40:1116-1127. [DOI: 10.1016/j.vaccine.2022.01.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 11/15/2021] [Accepted: 01/13/2022] [Indexed: 11/28/2022]
|
7
|
Hsieh MS, Hsu CW, Tu LL, Chai KM, Yu LL, Wu CC, Chen MY, Chiang CY, Liu SJ, Liao CL, Chen HW. Intranasal Vaccination With Recombinant Antigen-FLIPr Fusion Protein Alone Induces Long-Lasting Systemic Antibody Responses and Broad T Cell Responses. Front Immunol 2021; 12:751883. [PMID: 34707615 PMCID: PMC8543008 DOI: 10.3389/fimmu.2021.751883] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 09/27/2021] [Indexed: 11/29/2022] Open
Abstract
A simple formulation is urgently needed for mucosal vaccine development. We employed formyl peptide receptor-like 1 inhibitory protein (FLIPr), an FcγR antagonist secreted by Staphylococcus aureus, as a vector to target ovalbumin (OVA) to dendritic cells (DCs) via intranasal administration. Our results demonstrate that intranasal administration of recombinant OVA-FLIPr fusion protein (rOVA-FLIPr) alone efficiently delivers OVA to DCs in nasal lymphoid tissue. Subsequently, OVA-specific IgG and IgA antibodies in the circulatory system and IgA antibodies in mucosal tissue were detected. Importantly, activation of OVA-specific CD4+ and CD8+ T cells and induction of a broad-spectrum cytokine secretion profile were detected after intranasal administration of rOVA-FLIPr alone in immunocompetent C57BL/6 mice. Furthermore, we employed immunodeficient AG129 mice as a Zika virus infection model and demonstrated that intranasal administration of recombinant Zika virus envelope protein domain III-FLIPr fusion protein induced protective immune responses against the Zika virus. These results suggest that antigen-FLIPr fusion protein alone via intranasal administration can be applied to mucosal vaccine development.
Collapse
Affiliation(s)
- Ming-Shu Hsieh
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
| | - Chia-Wei Hsu
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
| | - Ling-Ling Tu
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
| | - Kit Man Chai
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
| | - Li-Lu Yu
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
| | - Chiao-Chieh Wu
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
| | - Mei-Yu Chen
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
| | - Chen-Yi Chiang
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
| | - Shih-Jen Liu
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
- Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Ching-Len Liao
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
| | - Hsin-Wei Chen
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
- Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
8
|
Richardson S, Medhavi F, Tanner T, Lundy S, Omosun Y, Igietseme JU, Carroll D, Eko FO. Cellular Basis for the Enhanced Efficacy of the Fms-Like Tyrosine Kinase 3 Ligand (FL) Adjuvanted VCG-Based Chlamydia abortus Vaccine. Front Immunol 2021; 12:698737. [PMID: 34249004 PMCID: PMC8264281 DOI: 10.3389/fimmu.2021.698737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 06/11/2021] [Indexed: 12/02/2022] Open
Abstract
Efficacious vaccines are needed to control genital chlamydial diseases in humans and the veterinary industry. We previously reported a C. abortus (Cab) vaccine comprising recombinant Vibrio cholerae ghosts (rVCG) expressing the conserved and immunogenic N-terminal region of the Cab polymorphic membrane protein D (rVCG-Pmp18.1) protein that protected mice against intravaginal challenge. In this study, we investigated the immunomodulatory effect of the hematopoietic progenitor activator cytokine, Fms-like tyrosine kinase 3-ligand (FL) when co-administered with the rVCG-Pmp18.1 vaccine as a strategy to enhance the protective efficacy and the potential mechanism of immunomodulation. Groups of female C57BL/6J mice were immunized and boosted twice intranasally (IN) with rVCG-PmpD18.1 with and without FL or purified rPmp18.1 or rVCG-gD2 (antigen control) or PBS (medium) per mouse. The results revealed that co-administration of the vaccine with FL enhanced antigen-specific cellular and humoral immune responses and protected against live Cab genital infection. Comparative analysis of immune cell phenotypes infiltrating mucosal and systemic immune inductive tissue sites following immunization revealed that co-administration of rVCG-Pmp18.1 with FL significantly enhanced the number of macrophages, dendritic and NK cells, γδ and NK T cells in the spleen (systemic) and iliac lymph nodes (ILN) draining the genital tract (mucosal) tissues compared to rVCG-Pmp18.1 alone. Furthermore, FL enhanced monocyte infiltration in the ILN, while CD19+ B cells and CD4+ T cells were enhanced in the spleen. These results indicate that the immunomodulatory effect of FL is associated with its ability to mobilize innate immune cells and subsequent activation of robust antigen-specific immune effectors in mucosal and systemic lymphoid tissues.
Collapse
Affiliation(s)
- Shakyra Richardson
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA, United States
| | - Fnu Medhavi
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA, United States
| | - Tayhlor Tanner
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA, United States
| | - Stephanie Lundy
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA, United States
| | - Yusuf Omosun
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA, United States
| | - Joseph U. Igietseme
- National Center for Emerging Zoonotic and Infectious Diseases, Centers for Disease Control and Prevention (CDC), Atlanta, GA, United States
| | - Darin Carroll
- National Center for Emerging Zoonotic and Infectious Diseases, Centers for Disease Control and Prevention (CDC), Atlanta, GA, United States
| | - Francis O. Eko
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA, United States
| |
Collapse
|
9
|
Kataoka K, Kawabata S, Koyanagi K, Hashimoto Y, Miyake T, Fujihashi K. Respiratory FimA-Specific Secretory IgA Antibodies Upregulated by DC-Targeting Nasal Double DNA Adjuvant Are Essential for Elimination of Porphyromonas gingivalis. Front Immunol 2021; 12:634923. [PMID: 33717178 PMCID: PMC7948520 DOI: 10.3389/fimmu.2021.634923] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Accepted: 01/18/2021] [Indexed: 11/13/2022] Open
Abstract
Our previous studies showed that a combination of a DNA plasmid encoding Flt3 ligand (pFL) and CpG oligodeoxynucleotides 1826 (CpG ODN) (FL/CpG) as a nasal adjuvant provoked antigen-specific immune responses. In this study, we investigated the efficacy of a nasal vaccine consisting of FimA as the structural subunit of Porphyromonas gingivalis (P. gingivalis) fimbriae and FL/CpG for the induction of FimA-specific antibody (Ab) responses and their protective roles against nasal and lung infection by P. gingivalis, a keystone pathogen in the etiology of periodontal disease. C57BL/6 mice were nasally immunized with recombinant FimA (rFimA) plus FL/CpG three times at weekly intervals. As a control, mice were given nasal rFimA alone. Nasal washes (NWs) and bronchoalveolar lavage fluid (BALF) of mice given nasal rFimA plus FL/CpG resulted in increased levels of rFimA-specific secretory IgA (SIgA) and IgG Ab responses when compared with those in controls. Significantly increased numbers of CD8- or CD11b-expressing mature-type dendritic cells (DCs) were detected in the respiratory inductive and effector tissues of mice given rFimA plus FL/CpG. Additionally, significantly upregulated Th1/Th2-type cytokine responses by rFimA-stimulated CD4+ T cells were noted in the respiratory effector tissues. When mice were challenged with live P. gingivalis via the nasal route, mice immunized nasally with rFimA plus FL/CpG inhibited P. gingivalis colonization in the nasal cavities and lungs. In contrast, controls failed to show protection. Of interest, when IgA-deficient mice given nasal rFimA plus FL/CpG were challenged with nasal P. gingivalis, the inhibition of bacterial colonization in the respiratory tracts was not seen. Taken together, these results show that nasal FL/CpG effectively enhanced DCs and provided balanced Th1- and Th2-type cytokine response-mediated rFimA-specific IgA protective immunity in the respiratory tract against P. gingivalis. A nasal administration with rFimA and FL/CpG could be a candidate for potent mucosal vaccines for the elimination of inhaled P. gingivalis in periodontal patients.
Collapse
MESH Headings
- Adjuvants, Immunologic/administration & dosage
- Administration, Intranasal
- Animals
- Antibodies, Bacterial/metabolism
- Bacterial Vaccines/administration & dosage
- Bacterial Vaccines/genetics
- Bacterial Vaccines/immunology
- Bacteroidaceae Infections/immunology
- Bacteroidaceae Infections/microbiology
- Bacteroidaceae Infections/prevention & control
- Disease Models, Animal
- Female
- Fimbriae Proteins/administration & dosage
- Fimbriae Proteins/genetics
- Fimbriae Proteins/immunology
- Immunity, Mucosal/drug effects
- Immunization Schedule
- Immunogenicity, Vaccine
- Immunoglobulin A, Secretory/metabolism
- Membrane Proteins/administration & dosage
- Membrane Proteins/genetics
- Membrane Proteins/immunology
- Mice, Inbred C57BL
- Oligodeoxyribonucleotides/administration & dosage
- Oligodeoxyribonucleotides/immunology
- Porphyromonas gingivalis/immunology
- Porphyromonas gingivalis/pathogenicity
- Recombinant Proteins/administration & dosage
- Recombinant Proteins/genetics
- Recombinant Proteins/immunology
- Respiratory System/drug effects
- Respiratory System/immunology
- Respiratory System/metabolism
- Respiratory System/microbiology
- Time Factors
- Vaccines, DNA/administration & dosage
- Vaccines, DNA/immunology
- Mice
Collapse
Affiliation(s)
- Kosuke Kataoka
- Department of Preventive and Community Dentistry, Faculty of Dentistry, Osaka Dental University, Hirakata, Japan
| | - Shigetada Kawabata
- Department of Oral and Molecular Microbiology, Graduate School of Dentistry, Osaka University, Suita, Japan
| | - Kayo Koyanagi
- Department of Preventive and Community Dentistry, Faculty of Dentistry, Osaka Dental University, Hirakata, Japan
| | - Yoshiya Hashimoto
- Department of Biomaterials, Faculty of Dentistry, Osaka Dental University, Hirakata, Japan
| | - Tatsuro Miyake
- Department of Preventive and Community Dentistry, Faculty of Dentistry, Osaka Dental University, Hirakata, Japan
| | - Kohtaro Fujihashi
- Division of Clinical Vaccinology, International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Department of Pediatric Dentistry, School of Dentistry, The University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
10
|
The impact of immuno-aging on SARS-CoV-2 vaccine development. GeroScience 2021; 43:31-51. [PMID: 33569701 PMCID: PMC7875765 DOI: 10.1007/s11357-021-00323-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 01/07/2021] [Indexed: 12/14/2022] Open
Abstract
The SARS-CoV-2 pandemic has almost 56 million confirmed cases resulting in over 1.3 million deaths as of November 2020. This infection has proved more deadly to older adults (those >65 years of age) and those with immunocompromising conditions. The worldwide population aged 65 years and older is increasing, and the total number of aged individuals will outnumber those younger than 65 years by the year 2050. Aging is associated with a decline in immune function and chronic activation of inflammation that contributes to enhanced viral susceptibility and reduced responses to vaccination. Here we briefly review the pathogenicity of the virus, epidemiology and clinical response, and the underlying mechanisms of human aging in improving vaccination. We review current methods to improve vaccination in the older adults using novel vaccine platforms and adjuvant systems. We conclude by summarizing the existing clinical trials for a SARS-CoV-2 vaccine and discussing how to address the unique challenges for vaccine development presented with an aging immune system.
Collapse
|
11
|
Tokuhara D, Hikita N. Cord Blood-Based Approach to Assess Candidate Vaccine Adjuvants Designed for Neonates and Infants. Vaccines (Basel) 2021; 9:vaccines9020095. [PMID: 33514054 PMCID: PMC7911524 DOI: 10.3390/vaccines9020095] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 01/23/2021] [Accepted: 01/25/2021] [Indexed: 12/12/2022] Open
Abstract
Neonates and infants are particularly susceptible to infections, for which outcomes tend to be severe. Vaccination is a key strategy for preventing infectious diseases, but the protective immunity achieved through vaccination typically is weaker in infants than in healthy adults. One possible explanation for the poor acquisition of vaccine-induced immunity in infants is that their innate immune response, represented by toll-like receptors, is immature. The current system for developing pediatric vaccines relies on the confirmation of their safety and effectiveness in studies involving the use of mature animals or adult humans. However, creating vaccines for neonates and infants requires an understanding of their uniquely immature innate immunity. Here we review current knowledge regarding the innate immune system of neonates and infants and challenges in developing vaccine adjuvants for those children through analyses of cord blood.
Collapse
|
12
|
Effect of fatty acid ester structure on cytotoxicity of self-emulsified nanoemulsion and transport of nanoemulsion droplets. Colloids Surf B Biointerfaces 2020; 194:111220. [DOI: 10.1016/j.colsurfb.2020.111220] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 06/09/2020] [Accepted: 06/23/2020] [Indexed: 11/22/2022]
|
13
|
Recombinant E rns-E2 protein vaccine formulated with MF59 and CPG-ODN promotes T cell immunity against bovine viral diarrhea virus infection. Vaccine 2020; 38:3881-3891. [PMID: 32280039 DOI: 10.1016/j.vaccine.2020.03.020] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 03/02/2020] [Accepted: 03/06/2020] [Indexed: 02/07/2023]
Abstract
To obtain an effective vaccine candidate against bovine viral diarrhea virus (BVDV) disease which causes great economical loss in cattle industries, recombinant Erns-E2 protein vaccine containing MF59 and CPG-ODN adjuvants was prepared and assessed in this study. The recombinant plasmid (pET32a-Erns-E2) was constructed and transformed into BL21 (DE3) cells to produce Erns-E2 protein. We immunized mice with the MF59-and CPG-ODN-adjuvanted recombinant Erns-E2 protein, E2 protein, or Erns protein, respectively. To evaluate immunogenicity and efficacy of a vaccine-adjuvant combination, mice were challenged with BVDV BJ175170 strain after immunization. All adjuvanted vaccines elicited detectable humoral and cellular immune responses, the BVDV-specific antibody titers as well as interleukin 4 (IL-4) levels in sera of mice immunized with the recombinant Erns-E2 protein were higher than in those of mice immunized with either the recombinant Erns or E2 protein. Besides, immunization with the Erns-E2 vaccines induced higher percentage of CD4+IFN-γ+, CD8+IFN-γ+ T cells and CD3+TNF-α+ T cells compared with the other vaccines. More protective efficacy against BVDV infection was acquired in the mice treated with the recombinant Erns-E2 protein, as shown by a reduction of viremia and slight pathological changes compared with both the control mice and the other vaccinated mice. Our findings suggest that the use of the recombinant Erns-E2 protein vaccine formulated with MF59 and CPG-ODN adjuvants enhances T cell responses and viral control, which warrants the Erns-E2 protein vaccine-adjuvant combination could be as a vaccine strategy to against BVDV.
Collapse
|
14
|
Kobuchi K, Kataoka K, Taguchi Y, Miyake T, Umeda M. Nasal double DNA adjuvant induces salivary FimA-specific secretory IgA antibodies in young and aging mice and blocks Porphyromonas gingivalis binding to a salivary protein. BMC Oral Health 2019; 19:188. [PMID: 31426773 PMCID: PMC6700810 DOI: 10.1186/s12903-019-0886-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 08/14/2019] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND We previously showed that nasal administration of a combination of dendritic cell (DC) targeted DNA plasmid expressing Flt3 ligand and CpG oligodeoxynucleotides 1826 as a mucosal adjuvant (double adjuvant, DA) provoked protective immunity in the upper respiratory tract of young adult and aging mice. Here, we investigated whether the nasal DA system induces secretory (S)IgA antibodies (Abs) toward recombinant fimbrillin (rFimA) of Porphyromonas gingivalis (P. gingivalis) in the saliva of young adult and aging mice. Further, we examined the functional applicability of rFimA-specific salivary SIgA Abs. METHODS BALB/c mice (8- or 48-week-old) were nasally immunized with rFimA plus DA three times at weekly intervals. Control mice were nasally administered rFimA alone. Saliva samples were collected 1 week after the final immunization, and were subjected to rFimA-specific ELISA. To examine the functional applicability of rFimA-specific SIgA Abs, IgA-enriched saliva samples were subjected to an inhibition assay in order to assess the numbers of P. gingivalis cells bound to the salivary protein statherin. RESULTS The 8- and 48-week-old mice administered nasal rFimA plus DA showed significantly increased levels of rFimA-specific SIgA Abs in saliva and elevated numbers of CD11c+ DCs in sublingual glands (SLGs), periglandular lymph nodes (PGLNs) and submandibular glands (SMGs) as well as nasopharyngeal-associated lymphoid tissues (NALT) compared to mice administered rFimA alone. Further, rFimA-specific SIgA Abs-containing saliva, in which IgG Abs of 8- and 48-week-old mice administered nasal rFimA plus DA were removed, significantly inhibited binding of P. gingivalis to the salivary protein. CONCLUSIONS These findings show that this DA system could be an effective nasal vaccine strategy for the enhancement of P. gingivalis-specific protective immunity in the oral cavity of adolescents and older individuals.
Collapse
Affiliation(s)
- Kenjiro Kobuchi
- Graduate School of Dentistry, Osaka Dental University, Hirakata, Osaka, 573-1121 Japan
- Department of Periodontology, Osaka Dental University, Hirakata, Osaka, 573-1121 Japan
| | - Kosuke Kataoka
- Department of Preventive and Community Dentistry, Osaka Dental University, Hirakata, Osaka, 573-1121 Japan
| | - Yoichiro Taguchi
- Department of Periodontology, Osaka Dental University, Hirakata, Osaka, 573-1121 Japan
| | - Tatsuro Miyake
- Department of Preventive and Community Dentistry, Osaka Dental University, Hirakata, Osaka, 573-1121 Japan
| | - Makoto Umeda
- Department of Periodontology, Osaka Dental University, Hirakata, Osaka, 573-1121 Japan
| |
Collapse
|
15
|
Tateishi K, Fujihashi K, Yamamoto N, Hasegawa H, Ainai A, Sato K, Iho S, Yamamoto S, Maeyama JI, Odagiri T, Asanuma H. CpG ODN G9.1 as a novel nasal ODN adjuvant elicits complete protection from influenza virus infection without causing inflammatory immune responses. Vaccine 2019; 37:5382-5389. [PMID: 31345642 DOI: 10.1016/j.vaccine.2019.07.032] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 07/06/2019] [Accepted: 07/08/2019] [Indexed: 11/26/2022]
Abstract
This study examined the protective efficacy of and immune response to a nasal influenza vaccine combined with a novel mucosal oligodeoxynucleotide (ODN) adjuvant, CpG ODN G9.1 (G9.1), in a model of infection limited to the upper respiratory tract (URT) and a model of infection in the lower respiratory tract (LRT). Mice were nasally primed with an A/California/7/2009 (Cal7) split vaccine (X179A) plus G9.1 and were then nasally given a booster with X179A alone. When mice were challenged with either a large (infection of the LRT) or small (infection limited to the URT) volume of live Cal7 influenza virus, mice nasally given G9.1 combined with X179A had a markedly higher rate of protection against infection limited to the URT. Moreover, this group of mice promptly recovered from an infection of the LRT. When mice were subcutaneously (s.c.) given X179A as a current form of vaccination, they had no protection from an infection limited to the URT but they did recover from an infection of the LRT. The patterns of protection were closely correlated with influenza virus-specific mucosal secretory IgA (SIgA) or serum IgG antibody (Ab) responses. Thus, SIgA Abs responses play an important role in protection from an infection limited to the URT while influenza virus-specific serum IgG Ab responses help to protect from an infection of the LRT. A finding of note is that lungs from mice nasally given G9.1 had low levels of type I IFN-associated protein- and transcription factor-specific mRNA expression. These results suggest that nasal G9.1 can be used as an effective and safe mucosal adjuvant for influenza vaccines since this nasal vaccine system elicits both mucosal SIgA and serum IgG Ab responses that provide complete protection without inducing potent inflammatory responses.
Collapse
Affiliation(s)
- Koichiro Tateishi
- Influenza Virus Research Center, National Institute of Infectious Diseases, Gakuen 4-7-1, Musashi-murayama-shi, Tokyo 208-0011, Japan; Japan Agency for Medical Research and Development, 1-7-1 Otemachi, Chiyoda-ku, Tokyo 100-0004, Japan
| | - Kohtaro Fujihashi
- Division of Clinical Vaccinology, International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, Shirokanedai 4-6-1, Minato-ku, Tokyo 108-8639, Japan; Department of Pediatric Dentistry, The Institute of Oral Health Research, The University of Alabama at Birmingham, Birmingham, AL 35294-0007, USA
| | - Norio Yamamoto
- Influenza Virus Research Center, National Institute of Infectious Diseases, Gakuen 4-7-1, Musashi-murayama-shi, Tokyo 208-0011, Japan; Department of Infection Control Science, Juntendo University, Tokyo, Japan
| | - Hideki Hasegawa
- Department of Pathology, National Institute of Infectious Diseases, Toyama 1-23-1, Shinjuku-ku, Tokyo 162-8640, Japan
| | - Akira Ainai
- Department of Pathology, National Institute of Infectious Diseases, Toyama 1-23-1, Shinjuku-ku, Tokyo 162-8640, Japan
| | - Kayoko Sato
- Influenza Virus Research Center, National Institute of Infectious Diseases, Gakuen 4-7-1, Musashi-murayama-shi, Tokyo 208-0011, Japan
| | - Sumiko Iho
- Division of Medicine, Faculty of Medical Sciences, University of Fukui, Yoshida-gun, Fukui and Department of Bacteriology, Niigata University Graduate School of Medicine, Niigata-shi, Niigata, Japan
| | - Saburo Yamamoto
- Central Laboratory, Japan BCG Laboratory, Kiyose-shi, Tokyo, Japan
| | - Jun-Ichi Maeyama
- Department of Safety Research on Blood and Biological Products, National Institute of Infectious Diseases, Musashi-murayama-shi, Tokyo, Japan
| | - Takato Odagiri
- Influenza Virus Research Center, National Institute of Infectious Diseases, Gakuen 4-7-1, Musashi-murayama-shi, Tokyo 208-0011, Japan
| | - Hideki Asanuma
- Influenza Virus Research Center, National Institute of Infectious Diseases, Gakuen 4-7-1, Musashi-murayama-shi, Tokyo 208-0011, Japan; Department of Pathology, National Institute of Infectious Diseases, Toyama 1-23-1, Shinjuku-ku, Tokyo 162-8640, Japan.
| |
Collapse
|
16
|
Pais R, Omosun Y, Igietseme JU, Fujihashi K, Eko FO. Route of Vaccine Administration Influences the Impact of Fms-Like Tyrosine Kinase 3 Ligand (Flt3L) on Chlamydial-Specific Protective Immune Responses. Front Immunol 2019; 10:1577. [PMID: 31333682 PMCID: PMC6621642 DOI: 10.3389/fimmu.2019.01577] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 06/24/2019] [Indexed: 11/24/2022] Open
Abstract
We tested the hypothesis that the impact of the Fms-like tyrosine kinase 3-ligand (Flt3L; FL) on recombinant Vibrio cholerae ghost (rVCG) vaccine-induced chlamydial immunity is influenced by route of vaccine delivery. Female C57BL/6J mice were immunized rectally (IR) or intramuscularly (IM) with rVCG co-expressing the Chlamydia trachomatis PmpD and PorB proteins (rVCG- PmpD/PorB) with and without FL or glycoprotein D of HSV-2 (rVCG-gD2) as antigen control. Vaccine evaluation was based on measurement of T cell proliferation, Th1/Th2 cytokine, and humoral responses at systemic and mucosal compartments, and protection against intravaginal challenge infection. Results revealed that high levels of CD4+ T cell-mediated and humoral immune responses, were elicited in mice as a function of both IR and IM immunization. Unexpectedly, co-administration of vaccine with FL enhanced specific Th1-type cytokine levels and T cell proliferative responses following IR but not IM immunization. While administration of vaccine with FL enhanced the specific mucosal and systemic IgA antibody responses following both immunization routes, IgG2c responses were not enhanced following IR delivery. The vaccine-induced immune effectors protected mice against live heterologous C. muridarum infection irrespective of route of vaccine administration, with the regimen incorporating FL having a protective advantage. Further evaluation showed that protection afforded by the FL adjuvanted vaccine was facilitated by CD4+ T cells, as indicated by reduction in the intensity and duration of genital chlamydial shedding by naïve mice following adoptive transfer of immune CD4+ T cells. Taken together, the results indicate that comparable protective immunity, which is enhanced by co-delivery with FL, is elicited in the female genital tract against Chlamydia infection after mucosal and systemic administration, highlighting the ability of FL to function as an effective immunostimulator at both mucosal and systemic sites. The differential modulation of humoral and cellular immune responses, and protective immunity afforded by the FL adjuvanted vaccine following IR administration indicates that the immunomodulatory impact of FL on chlamydial-specific immunity is influenced by the route of vaccine administration. Thus, targeting of VCG-based vaccines to antigen presenting cells by co-delivery with FL is a feasible immunization approach for inducing effective chlamydial immunity in the female genital tract.
Collapse
Affiliation(s)
- Roshan Pais
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA, United States
| | - Yusuf Omosun
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA, United States
| | - Joseph U. Igietseme
- Molecular Pathogenesis Laboratory, Centers for Disease Control and Prevention, Atlanta, GA, United States
| | - Kohtaro Fujihashi
- Department of Pediatric Dentistry, Institute of Oral Health Research, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Francis O. Eko
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA, United States
| |
Collapse
|
17
|
Sovran B, Hugenholtz F, Elderman M, Van Beek AA, Graversen K, Huijskes M, Boekschoten MV, Savelkoul HFJ, De Vos P, Dekker J, Wells JM. Age-associated Impairment of the Mucus Barrier Function is Associated with Profound Changes in Microbiota and Immunity. Sci Rep 2019; 9:1437. [PMID: 30723224 PMCID: PMC6363726 DOI: 10.1038/s41598-018-35228-3] [Citation(s) in RCA: 144] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Accepted: 10/16/2018] [Indexed: 02/07/2023] Open
Abstract
Aging significantly increases the vulnerability to gastrointestinal (GI) disorders but there are few studies investigating the key factors in aging that affect the GI tract. To address this knowledge gap, we used 10-week- and 19-month-old litter-mate mice to investigate microbiota and host gene expression changes in association with ageing. In aged mice the thickness of the colonic mucus layer was reduced about 6-fold relative to young mice, and more easily penetrable by luminal bacteria. This was linked to increased apoptosis of goblet cells in the upper part of the crypts. The barrier function of the small intestinal mucus was also compromised and the microbiota were frequently observed in contact with the villus epithelium. Antimicrobial Paneth cell factors Ang4 and lysozyme were expressed in significantly reduced amounts. These barrier defects were accompanied by major changes in the faecal microbiota and significantly decreased abundance of Akkermansia muciniphila which is strongly and negatively affected by old age in humans. Transcriptomics revealed age-associated decreases in the expression of immunity and other genes in intestinal mucosal tissue, including decreased T cell-specific transcripts and T cell signalling pathways. The physiological and immunological changes we observed in the intestine in old age, could have major consequences beyond the gut.
Collapse
Affiliation(s)
- Bruno Sovran
- Top Institute Food and Nutrition, Wageningen, The Netherlands.,Cell Biology and Immunology Group, Wageningen University and Research Center, Wageningen, The Netherlands
| | - Floor Hugenholtz
- Laboratory of Microbiology, Wageningen University and Research Center, Wageningen, The Netherlands
| | - Marlies Elderman
- Top Institute Food and Nutrition, Wageningen, The Netherlands.,University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Adriaan A Van Beek
- Top Institute Food and Nutrition, Wageningen, The Netherlands.,Cell Biology and Immunology Group, Wageningen University and Research Center, Wageningen, The Netherlands
| | - Katrine Graversen
- Host-Microbe Interactomics Group, Wageningen University and Research Center, Wageningen, The Netherlands
| | - Myrte Huijskes
- Host-Microbe Interactomics Group, Wageningen University and Research Center, Wageningen, The Netherlands
| | - Mark V Boekschoten
- Top Institute Food and Nutrition, Wageningen, The Netherlands.,Division of Human Nutrition, Wageningen University and Research Center, Wageningen, The Netherlands
| | - Huub F J Savelkoul
- Top Institute Food and Nutrition, Wageningen, The Netherlands.,Cell Biology and Immunology Group, Wageningen University and Research Center, Wageningen, The Netherlands
| | - Paul De Vos
- Top Institute Food and Nutrition, Wageningen, The Netherlands.,University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Jan Dekker
- Top Institute Food and Nutrition, Wageningen, The Netherlands.,Host-Microbe Interactomics Group, Wageningen University and Research Center, Wageningen, The Netherlands
| | - Jerry M Wells
- Top Institute Food and Nutrition, Wageningen, The Netherlands. .,Host-Microbe Interactomics Group, Wageningen University and Research Center, Wageningen, The Netherlands.
| |
Collapse
|
18
|
Pan Q, Zhang Q, Chu J, Pais R, Liu S, He C, Eko FO. Chlamydia abortus Pmp18.1 Induces IL-1β Secretion by TLR4 Activation through the MyD88, NF-κB, and Caspase-1 Signaling Pathways. Front Cell Infect Microbiol 2017; 7:514. [PMID: 29326885 PMCID: PMC5741698 DOI: 10.3389/fcimb.2017.00514] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Accepted: 11/29/2017] [Indexed: 12/16/2022] Open
Abstract
The polymorphic membrane protein D (Pmp18D) is a 160-kDa outer membrane protein that is conserved and plays an important role in Chlamydia abortus pathogenesis. We have identified an N-terminal fragment of Pmp18D (designated Pmp18.1) as a possible subunit vaccine antigen. In this study, we evaluated the vaccine potential of Pmp18.1 by investigating its ability to induce innate immune responses in dendritic cells and the signaling pathway(s) involved in rPmp18.1-induced IL-1β secretion. We next investigated the immunomodulatory impact of VCG, in comparison with the more established Th1-promoting adjuvants, CpG and FL, on rPmp18.1-mediated innate immune activation. Finally, the effect of siRNA targeting TLR4, MyD88, NF-κB p50, and Caspase-1 mRNA in DCs on IL-1β cytokine secretion was also investigated. Bone marrow-derived dendritic cells (BMDCs) were stimulated with rPmp18.1 in the presence or absence of VCG or CpG or FL and the magnitude of cytokines produced was assessed using a multiplex cytokine ELISA assay. Expression of costimulatory molecules and Toll-like receptors (TLRs) was analyzed by flow cytometry. Quantitation of intracellular levels of myeloid differentiation factor 88 (MyD88), nuclear factor kappa beta (NF-κB p50/p65), and Caspase-1 was evaluated by Western immunoblotting analysis while NF-κB p65 nuclear translocation was assessed by confocal microscopy. The results showed DC stimulation with rPmp18.1 provoked the secretion of proinflammatory cytokines and upregulated expression of TLRs and co-stimulatory molecules associated with DC maturation. These responses were significantly (p ≤ 0.001) enhanced by VCG but not CpG or FL. In addition, rPmp18.1 activated the expression of MyD88, NF-κB p50, and Caspase-1 as well as the nuclear expression of NF-κB p65 in treated DCs. Furthermore, targeting TLR4, MyD88, NF-κB p50, and Caspase-1 mRNA in BMDCs with siRNA significantly reduced their expression levels, resulting in decreased IL-1β cytokine secretion, strongly suggesting their involvement in the rPmp18.1-induced IL-1β cytokine secretion. Taken together, these results indicate that C. abortus Pmp18.1 induces IL-1β secretion by TLR4 activation through the MyD88, NF-κB as well as the Caspase-1 signaling pathways and may be a potential C. abortus vaccine candidate. The vaccine potential of Pmp18.1 will subsequently be evaluated in an appropriate animal model, using VCG as an immunomodulator, following immunization and challenge.
Collapse
Affiliation(s)
- Qing Pan
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA, United States.,Key Lab of Animal Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Qiang Zhang
- Key Lab of Animal Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Jun Chu
- Key Lab of Animal Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Roshan Pais
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA, United States
| | - Shanshan Liu
- Key Lab of Animal Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Cheng He
- Key Lab of Animal Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Francis O Eko
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA, United States
| |
Collapse
|
19
|
Tsuruhara A, Aso K, Tokuhara D, Ohori J, Kawabata M, Kurono Y, McGhee JR, Fujihashi K. Rejuvenation of mucosal immunosenescence by adipose tissue-derived mesenchymal stem cells. Int Immunol 2017; 29:5-10. [PMID: 28391291 DOI: 10.1093/intimm/dxx001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Accepted: 01/05/2017] [Indexed: 02/07/2023] Open
Abstract
Age-associated alterations in the mucosal immune system are generally termed mucosal immunosenescence. The major change seen in the aged mucosa is a failure to elicit an antigen-specific secretory IgA (SIgA) antibody response, which is a central player for host defense from various pathogens at mucosal surfaces. In this regard, it would be a first priority to compensate for mucosal dysregulation in the elderly in order to maintain their health in aging. We have successfully established antigen-specific SIgA antibody responses in aged (2 years old) mice, which provide protective immunity from Streptococcus pneumoniae and influenza virus infections, by using a new adjuvant system consisting of a plasmid encoding Flt3 ligand (pFL) and CpG ODN. In order to explore possible use of current mucosal vaccine strategies for the elderly, we have adoptively transferred adipose tissue-derived mesenchymal stem cells (AMSCs) to aged mice prior to mucosal vaccination. This immune therapy successfully resulted in protective antigen-specific antibody responses in the intestinal mucosa of aged mice that were comparable to those seen in young adult mice. In this regard, we postulate that adoptively transferred AMSCs could augment dendritic cell functions in aged mice. The potential cellular and molecular mechanisms whereby AMSCs restore mucosal immunity in immunosenescence are discussed in this short review. A stem cell transfer system could be an attractive and effective immunologic intervention strategy to reverse mucosal immunosenescence.
Collapse
Affiliation(s)
- Akitoshi Tsuruhara
- Department of Pediatric Dentistry, The University of Alabama at Birmingham, 1919 7th Avenue South, SDB 801 A1, Birmingham, AL 35294-0007, USA
| | - Kazuyoshi Aso
- Department of Pediatric Dentistry, The University of Alabama at Birmingham, 1919 7th Avenue South, SDB 801 A1, Birmingham, AL 35294-0007, USA.,Department of Pediatrics, Graduate School of Medicine, Osaka City University, Asahi-cho 1-5-7, Abeno-ku, Osaka, Osaka 545-0051, Japan
| | - Daisuke Tokuhara
- Department of Pediatrics, Graduate School of Medicine, Osaka City University, Asahi-cho 1-5-7, Abeno-ku, Osaka, Osaka 545-0051, Japan
| | - Junichiro Ohori
- Department of Pediatric Dentistry, The University of Alabama at Birmingham, 1919 7th Avenue South, SDB 801 A1, Birmingham, AL 35294-0007, USA.,Department of Otolaryngology, Kagoshima University Faculty of Medicine, Sakuragaoka 8-35-1, Kagoshima, Kagoshima 890-8520, Japan
| | - Masaki Kawabata
- Department of Pediatric Dentistry, The University of Alabama at Birmingham, 1919 7th Avenue South, SDB 801 A1, Birmingham, AL 35294-0007, USA.,Department of Otolaryngology, Kagoshima University Faculty of Medicine, Sakuragaoka 8-35-1, Kagoshima, Kagoshima 890-8520, Japan
| | - Yuichi Kurono
- Department of Otolaryngology, Kagoshima University Faculty of Medicine, Sakuragaoka 8-35-1, Kagoshima, Kagoshima 890-8520, Japan
| | - Jerry R McGhee
- Department of Pediatric Dentistry, The University of Alabama at Birmingham, 1919 7th Avenue South, SDB 801 A1, Birmingham, AL 35294-0007, USA
| | - Kohtaro Fujihashi
- Department of Pediatric Dentistry, The University of Alabama at Birmingham, 1919 7th Avenue South, SDB 801 A1, Birmingham, AL 35294-0007, USA
| |
Collapse
|
20
|
Suzuki T, Ainai A, Hasegawa H. Functional and structural characteristics of secretory IgA antibodies elicited by mucosal vaccines against influenza virus. Vaccine 2017; 35:5297-5302. [PMID: 28780981 DOI: 10.1016/j.vaccine.2017.07.093] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Accepted: 07/14/2017] [Indexed: 02/04/2023]
Abstract
Mucosal tissues are major targets for pathogens. The secretions covering mucosal surfaces contain several types of molecules that protect the host from infection. Among these, mucosal immunoglobulins, including secretory IgA (S-IgA) antibodies, are the major contributor to pathogen-specific immune responses. IgA is the primary antibody class found in many external secretions and has unique structural and functional features not observed in other antibody classes. Recently, extensive efforts have been made to develop novel vaccines that induce immunity via the mucosal route. S-IgA is a key molecule that underpins the mechanism of action of these mucosal vaccines. Thus, precise characterization of S-IgA induced by mucosal vaccines is important, if the latter are to be used successfully in a clinical setting. Intensive studies identified the fundamental characteristics of S-IgA, which was first discovered almost half a century ago. However, S-IgA itself has not gained much attention of late, despite its importance to mucosal immunity; therefore, some important questions remain. This review summarizes the current understanding of the molecular characteristics of S-IgA and its role in intranasal mucosal vaccines against influenza virus infection.
Collapse
Affiliation(s)
- Tadaki Suzuki
- Department of Pathology, National Institute of Infectious Diseases, Tokyo 162-8640, Japan.
| | - Akira Ainai
- Department of Pathology, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| | - Hideki Hasegawa
- Department of Pathology, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| |
Collapse
|
21
|
Ainai A, Suzuki T, Tamura SI, Hasegawa H. Intranasal Administration of Whole Inactivated Influenza Virus Vaccine as a Promising Influenza Vaccine Candidate. Viral Immunol 2017. [PMID: 28650274 DOI: 10.1089/vim.2017.0022] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
The effect of the current influenza vaccine, an inactivated virus vaccine administered by subcutaneous/intramuscular injection, is limited to reducing the morbidity and mortality associated with seasonal influenza outbreaks. Intranasal vaccination, by contrast, mimics natural infection and induces not only systemic IgG antibodies but also local secretory IgA (S-IgA) antibodies found on the surface of the mucosal epithelium in the upper respiratory tract. S-IgA antibodies are highly effective at preventing virus infection. Although the live attenuated influenza vaccine (LAIV) administered intranasally can induce local antibodies, this vaccine is restricted to healthy populations aged 2-49 years because of safety concerns associated with using live viruses in a vaccine. Instead of LAIV, an intranasal vaccine made with inactivated virus could be applied to high-risk populations, including infants and elderly adults. Normally, a mucosal adjuvant would be required to enhance the effect of intranasal vaccination with an inactivated influenza vaccine. However, we found that intranasal administration of a concentrated, whole inactivated influenza virus vaccine without any mucosal adjuvant was enough to induce local neutralizing S-IgA antibodies in the nasal epithelium of healthy individuals with some immunological memory for seasonal influenza viruses. This intranasal vaccine is a novel candidate that could improve on the current injectable vaccine or the LAIV for the prevention of seasonal influenza epidemics.
Collapse
Affiliation(s)
- Akira Ainai
- Department of Pathology, National Institute of Infectious Diseases , Tokyo, Japan
| | - Tadaki Suzuki
- Department of Pathology, National Institute of Infectious Diseases , Tokyo, Japan
| | - Shin-Ichi Tamura
- Department of Pathology, National Institute of Infectious Diseases , Tokyo, Japan
| | - Hideki Hasegawa
- Department of Pathology, National Institute of Infectious Diseases , Tokyo, Japan
| |
Collapse
|
22
|
Falkeborn T, Hinkula J, Olliver M, Lindberg A, Maltais AK. The intranasal adjuvant Endocine™ enhances both systemic and mucosal immune responses in aged mice immunized with influenza antigen. Virol J 2017; 14:44. [PMID: 28253901 PMCID: PMC5335733 DOI: 10.1186/s12985-017-0698-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Accepted: 02/02/2017] [Indexed: 01/17/2023] Open
Abstract
Despite availability of annual influenza vaccines, influenza causes significant morbidity and mortality in the elderly. This is at least in part a result of immunosenescence; the age-dependent decrease in immunological competence that results in greater susceptibility to infections and reduced responses to vaccination. To improve protective immune responses in this age group, new vaccines strategies, such as the use of adjuvants, are needed. Here, we evaluated the mucosal vaccine adjuvant Endocine™, formulated with split influenza antigen and administered intranasally in aged (20-month old) mice. Humoral immune responses were assessed and compared to unadjuvanted intranasal and subcutaneous vaccines. We show that formulation with Endocine™ significantly enhances hemagglutination inhibition (HI) titers, as well as serum IgG and mucosal IgA antibody titers, compared to both types of unadjuvanted vaccines. Thus, our results indicate that intranasal vaccination with Endocine™ is a possible approach for the development of mucosal influenza vaccines for the elderly.
Collapse
Affiliation(s)
- Tina Falkeborn
- Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Jorma Hinkula
- Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Marie Olliver
- Eurocine Vaccines AB, Karolinska Institutet Science Park, Solna, Sweden
| | - Alf Lindberg
- Eurocine Vaccines AB, Karolinska Institutet Science Park, Solna, Sweden
| | | |
Collapse
|
23
|
Matos MN, Cazorla SI, Schulze K, Ebensen T, Guzmán CA, Malchiodi EL. Immunization with Tc52 or its amino terminal domain adjuvanted with c-di-AMP induces Th17+Th1 specific immune responses and confers protection against Trypanosoma cruzi. PLoS Negl Trop Dis 2017; 11:e0005300. [PMID: 28234897 PMCID: PMC5342303 DOI: 10.1371/journal.pntd.0005300] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Revised: 03/08/2017] [Accepted: 01/03/2017] [Indexed: 12/21/2022] Open
Abstract
The development of new adjuvants enables fine modulation of the elicited immune responses. Ideally, the use of one or more adjuvants should result in the induction of a protective immune response against the specific pathogen. We have evaluated the immune response and protection against Trypanosoma cruzi infection in mice vaccinated with recombinant Tc52 or its N- and C-terminal domains (NTc52 and CTc52) adjuvanted either with the STING (Stimulator of Interferon Genes) agonist cyclic di-AMP (c-di-AMP), a pegylated derivative of α-galactosylceramide (αGC-PEG), or oligodeoxynucleotides containing unmethylated CpG motifs (ODN-CpG). All groups immunized with the recombinant proteins plus adjuvant: Tc52+c-di-AMP, NTc52+c-di-AMP, CTc52+c-di-AMP, NTc52+c-di-AMP+αGC-PEG, NTc52+CpG, developed significantly higher anti-Tc52 IgG titers than controls. Groups immunized with c-di-AMP and Tc52, NTc52 or CTc52 showed the highest Tc52-specific IgA titers in nasal lavages. All groups immunized with the recombinant proteins plus adjuvant developed a strong specific cellular immune response in splenocytes and lymph node cells with significant differences for groups immunized with c-di-AMP and Tc52, NTc52 or CTc52. These groups also showed high levels of Tc52-specific IL-17 and IFN-γ producing cells, while NTc52+CpG group only showed significant difference with control in IFN-γ producing cells. Groups immunized with c-di-AMP and Tc52, NTc52 or CTc52 developed predominantly a Th17 and Th1immune response, whereas for NTc52+CpG it was a dominant Th1 response. It was previously described that αGC-PEG inhibits Th17 differentiation by activating NKT cells. Thus, in this work we have also included a group immunized with both adjuvants (NTc52+c-di-AMP+αGC-PEG) with the aim to modulate the Th17 response induced by c-di-AMP. This group showed a significant reduction in the number of Tc52-specific IL-17 producing splenocytes, as compared to the group NTc52+c-di-AMP, which has in turn correlated with a reduction in protection against infection. These results suggest that the Th17 immune response developed after immunizing with NTc52+c-di-AMP could have a protective role against T. cruzi infection. Groups NTc52+c-di-AMP, Tc52+c-di-AMP and NTc52PB, were the ones that showed better protection against infection with lower parasitemia and weight loss, and higher survival. Chagas disease is a parasitic disease caused by a protozoan parasite (Trypanosoma cruzi) which has a complex life cycle including insect vector and mammalians. In Latin America, 7–10 million people are infected, 100 million people are at risk of infection, and about 56,000 new infection cases and 12,000 deaths are registered annually. Migration spread the geographic distribution of the disease to North America and Europe. The infection in humans has an initial acute stage followed by a chronic stage where up to 30% of patients develop cardiac alterations and 10% develop digestive, neurological or mixed alterations. The acute infection is hardly detected and there is not drug to treat the chronic infection. Thus, there is an urgent need for prophylactic and therapeutic vaccines development. Several attempts to find a vaccine antigen has been made and the protein Tc52 is a good candidate. In a vaccine composition, as important as the antigen is the adjuvants, which are substances able to increase, improve or modified the immune response. This research provides information about the immune response and protection against Trypanosoma cruzi infection elicited by Tc52 or portions of this molecule using different adjuvants.
Collapse
Affiliation(s)
- Marina N. Matos
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Cátedra de Inmunología and Instituto de Estudios de la Inmunidad Humoral (IDEHU), UBA-CONICET, Buenos Aires, Argentina
- Universidad de Buenos Aires, Facultad de Medicina, Departamento de Microbiología, Parasitología e Inmunología and Instituto de Microbiología y Parasitología Médica (IMPaM), UBA-CONICET, Buenos Aires, Argentina
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Silvia I. Cazorla
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Cátedra de Inmunología and Instituto de Estudios de la Inmunidad Humoral (IDEHU), UBA-CONICET, Buenos Aires, Argentina
- Universidad de Buenos Aires, Facultad de Medicina, Departamento de Microbiología, Parasitología e Inmunología and Instituto de Microbiología y Parasitología Médica (IMPaM), UBA-CONICET, Buenos Aires, Argentina
| | - Kai Schulze
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Thomas Ebensen
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Carlos A. Guzmán
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Emilio L. Malchiodi
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Cátedra de Inmunología and Instituto de Estudios de la Inmunidad Humoral (IDEHU), UBA-CONICET, Buenos Aires, Argentina
- Universidad de Buenos Aires, Facultad de Medicina, Departamento de Microbiología, Parasitología e Inmunología and Instituto de Microbiología y Parasitología Médica (IMPaM), UBA-CONICET, Buenos Aires, Argentina
- * E-mail:
| |
Collapse
|
24
|
Liu Z, Yin L, Li Y, Yuan F, Zhang X, Ma J, Liu H, Wang Y, Zheng K, Cao J. Intranasal immunization with recombinant Toxoplasma gondii actin depolymerizing factor confers protective efficacy against toxoplasmosis in mice. BMC Immunol 2016; 17:37. [PMID: 27716047 PMCID: PMC5053087 DOI: 10.1186/s12865-016-0173-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Accepted: 09/20/2016] [Indexed: 11/29/2022] Open
Abstract
Background Toxoplasma gondii is an opportunistic protozoan closely associated with AIDS and vertical transmission. T. gondii actin depolymerizing factor (TgADF) plays an important role in actin cytoskeleton remodeling, and it is required to invade host cells. TgADF was a promising vaccine candidate. To observe the immunological changes and protective efficacy of recombinant TgADF protein (rTgADF) against T. gondii infection, we optimized the intranasal immunization dose of rTgADF and analyzed the survival rate and tachyzoite loads in mouse tissues after oral challenge with T. gondii tachyzoites. Results rTgADF was prepared, purified, and combined with mouse anti-His antibody and rabbit anti-T. gondii serum. After intranasal immunization with 10 μg, 20 μg, 30 μg, or 40 μg of rTgADF, the 30-μg group elicited high levels of secretory IgA (sIgA) in nasal, intestinal, and vesical washes, raised IgG titres in the sera, strong proliferation of splenocytes, and increased secretion of IL-2 and IFN-γ when compared with the control group. When the mice were orally challenged with T. gondii, an increase in the survival rate (36.36 %) and a decrease in the tachyzoite loads in the liver (67.77 %) and brain (51.01 %) were observed. Conclusions Our findings demonstrate that intranasal immunization with rTgADF can simultaneously trigger mucosal and systemic immune responses and protect the mice against T. gondii infection.
Collapse
Affiliation(s)
- Zhuanzhuan Liu
- Department of Pathogenic Biology and Immunology, Laboratory of Infection and Immunity, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, People's Republic of China
| | - Litian Yin
- Department of Physiology, Shanxi Medical University, Taiyuan, Shanxi, 030001, People's Republic of China
| | - Yaqing Li
- Department of Physiology, Shanxi Medical University, Taiyuan, Shanxi, 030001, People's Republic of China
| | - Fei Yuan
- Department of Pathogenic Biology and Immunology, Laboratory of Infection and Immunity, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, People's Republic of China
| | - Xiaofan Zhang
- School of Public Health and Tropical Medicine, Southern Medical University, Guangzhou, Guangdong, 510000, People's Republic of China
| | - Jiazhi Ma
- School of Public Health and Tropical Medicine, Southern Medical University, Guangzhou, Guangdong, 510000, People's Republic of China
| | - Hongmei Liu
- School of Public Health and Tropical Medicine, Southern Medical University, Guangzhou, Guangdong, 510000, People's Republic of China
| | - Yanjuan Wang
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention; Key Laboratory of Parasite and Vector Biology, MOH, China; National Center for International Research on Tropical Diseases, China; WHO Collaborating Center for Tropical Diseases, Shanghai, 200025, People's Republic of China
| | - Kuiyang Zheng
- Department of Pathogenic Biology and Immunology, Laboratory of Infection and Immunity, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, People's Republic of China.
| | - Jianping Cao
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention; Key Laboratory of Parasite and Vector Biology, MOH, China; National Center for International Research on Tropical Diseases, China; WHO Collaborating Center for Tropical Diseases, Shanghai, 200025, People's Republic of China.
| |
Collapse
|
25
|
Kusakabe T, Ozasa K, Kobari S, Momota M, Kishishita N, Kobiyama K, Kuroda E, Ishii KJ. Intranasal hydroxypropyl-β-cyclodextrin-adjuvanted influenza vaccine protects against sub-heterologous virus infection. Vaccine 2016; 34:3191-3198. [PMID: 27160037 DOI: 10.1016/j.vaccine.2016.04.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2015] [Revised: 03/26/2016] [Accepted: 04/01/2016] [Indexed: 12/31/2022]
Abstract
Intranasal vaccination with inactivated influenza viral antigens is an attractive and valid alternative to currently available influenza (flu) vaccines; many of which seem to need efficient and safe adjuvant, however. In this study, we examined whether hydroxypropyl-β-cyclodextrin (HP-β-CD), a widely used pharmaceutical excipient to improve solubility and drug delivery, can act as a mucosal adjuvant for intranasal flu vaccines. We found that intranasal immunization of mice with hemagglutinin split- as well as inactivated whole-virion influenza vaccine with HP-β-CD resulted in secretion of antigen-specific IgA and IgGs in the airway mucosa and the serum as well. As a result, both HP-β-CD adjuvanted-flu intranasal vaccine protected mice against lethal challenge with influenza virus, equivalent to those induced by experimental cholera toxin-adjuvanted ones. Of note, intranasal use of HP-β-CD as an adjuvant induced significantly lower antigen-specific IgE responses than that induced by aluminum salt adjuvant. These results suggest that HP-β-CD may be a potent mucosal adjuvant for seasonal and pandemic influenza vaccine.
Collapse
Affiliation(s)
- Takato Kusakabe
- Laboratory of Adjuvant Innovation, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka 567-0085, Japan; Laboratory of Vaccine Science, WPI Immunology Frontier Research Center (IFReC), Osaka University, Osaka 565-0871, Japan
| | - Koji Ozasa
- Laboratory of Adjuvant Innovation, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka 567-0085, Japan
| | - Shingo Kobari
- Laboratory of Adjuvant Innovation, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka 567-0085, Japan
| | - Masatoshi Momota
- Laboratory of Adjuvant Innovation, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka 567-0085, Japan; Laboratory of Vaccine Science, WPI Immunology Frontier Research Center (IFReC), Osaka University, Osaka 565-0871, Japan
| | - Natsuko Kishishita
- Laboratory of Adjuvant Innovation, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka 567-0085, Japan
| | - Kouji Kobiyama
- Laboratory of Adjuvant Innovation, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka 567-0085, Japan; Laboratory of Vaccine Science, WPI Immunology Frontier Research Center (IFReC), Osaka University, Osaka 565-0871, Japan
| | - Etsushi Kuroda
- Laboratory of Vaccine Science, WPI Immunology Frontier Research Center (IFReC), Osaka University, Osaka 565-0871, Japan
| | - Ken J Ishii
- Laboratory of Adjuvant Innovation, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka 567-0085, Japan; Laboratory of Vaccine Science, WPI Immunology Frontier Research Center (IFReC), Osaka University, Osaka 565-0871, Japan.
| |
Collapse
|
26
|
Ross K, Adams J, Loyd H, Ahmed S, Sambol A, Broderick S, Rajan K, Kohut M, Bronich T, Wannemuehler MJ, Carpenter S, Mallapragada S, Narasimhan B. Combination Nanovaccine Demonstrates Synergistic Enhancement in Efficacy against Influenza. ACS Biomater Sci Eng 2016; 2:368-374. [PMID: 33429541 DOI: 10.1021/acsbiomaterials.5b00477] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
H5N1 influenza virus has the potential to become a significant global health threat, and next generation vaccine technologies are needed. In this work, the combined efficacy of two nanoadjuvant platforms (polyanhydride nanoparticles and pentablock copolymer-based hydrogels) to induce protective immunity against H5N1 influenza virus was examined. Mice received two subcutaneous vaccinations (day 0 and 21) containing 10 μg of H5 hemagglutinin trimer alone or in combination with the nanovaccine platforms. Nanovaccine immunization induced high neutralizing antibody titers that were sustained through 70 days postimmunization. Finally, mice were intranasally challenged with A/H5N1 VNH5N1-PR8CDC-RG virus and monitored for 14 days. Animals receiving the combination nanovaccine had lower viral loads in the lung and weight loss after challenge in comparison to animals vaccinated with each platform alone. These data demonstrate the synergy between polyanhydride nanoparticles and pentablock copolymer-based hydrogels as adjuvants in the design of a more efficacious influenza vaccine.
Collapse
Affiliation(s)
| | | | | | | | | | - Scott Broderick
- Materials Design and Innovation, University at Buffalo-The State University of New York, Buffalo, New York 14260, United States
| | - Krishna Rajan
- Materials Design and Innovation, University at Buffalo-The State University of New York, Buffalo, New York 14260, United States
| | | | | | | | | | | | | |
Collapse
|
27
|
Aso K, Tsuruhara A, Takagaki K, Oki K, Ota M, Nose Y, Tanemura H, Urushihata N, Sasanuma J, Sano M, Hirano A, Aso R, McGhee JR, Fujihashi K. Adipose-Derived Mesenchymal Stem Cells Restore Impaired Mucosal Immune Responses in Aged Mice. PLoS One 2016; 11:e0148185. [PMID: 26840058 PMCID: PMC4740412 DOI: 10.1371/journal.pone.0148185] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Accepted: 01/14/2016] [Indexed: 12/29/2022] Open
Abstract
It has been shown that adipose-derived mesenchymal stem cells (AMSCs) can differentiate into adipocytes, chondrocytes and osteoblasts. Several clinical trials have shown the ability of AMSCs to regenerate these differentiated cell types. Age-associated dysregulation of the gastrointestinal (GI) immune system has been well documented. Our previous studies showed that impaired mucosal immunity in the GI tract occurs earlier during agingthan is seen in the systemic compartment. In this study, we examined the potential of AMSCs to restore the GI mucosal immune system in aged mice. Aged (>18 mo old) mice were adoptively transferred with AMSCs. Two weeks later, mice were orally immunized with ovalbumin (OVA) plus cholera toxin (CT) three times at weekly intervals. Seven days after the final immunization, when fecal extract samples and plasma were subjected to OVA- and CT-B-specific ELISA, elevated levels of mucosal secretory IgA (SIgA) and plasma IgG antibody (Ab) responses were noted in aged mouse recipients. Similar results were also seen aged mice which received AMSCs at one year of age. When cytokine production was examined, OVA-stimulated Peyer's patch CD4+ T cells produced increased levels of IL-4. Further, CD4+ T cells from the lamina propria revealed elevated levels of IL-4 and IFN-γ production. In contrast, aged mice without AMSC transfer showed essentially no OVA- or CT-B-specific mucosal SIgA or plasma IgG Ab or cytokine responses. Of importance, fecal extracts from AMSC transferred aged mice showed neutralization activity to CT intoxication. These results suggest that AMSCs can restore impaired mucosal immunity in the GI tract of aged mice.
Collapse
Affiliation(s)
- Kazuyoshi Aso
- Department of Pediatric Dentistry, The University of Alabama at Birmingham, Birmingham, AL, United States of America
| | - Akitoshi Tsuruhara
- Department of Pediatric Dentistry, The University of Alabama at Birmingham, Birmingham, AL, United States of America
| | | | | | - Megumi Ota
- BioMimetics Sympathies Inc., Tokyo, Japan
| | | | | | | | - Jinichi Sasanuma
- Department of Neurosurgery, Shinyurigaoka General Hospital, Kawasaki, Japan
| | | | | | - Rio Aso
- Department of Pediatric Dentistry, The University of Alabama at Birmingham, Birmingham, AL, United States of America
| | - Jerry R. McGhee
- Department of Pediatric Dentistry, The University of Alabama at Birmingham, Birmingham, AL, United States of America
| | - Kohtaro Fujihashi
- Department of Pediatric Dentistry, The University of Alabama at Birmingham, Birmingham, AL, United States of America
- * E-mail:
| |
Collapse
|
28
|
Pabst R. Mucosal vaccination by the intranasal route. Nose-associated lymphoid tissue (NALT)-Structure, function and species differences. Vaccine 2015. [PMID: 26196324 DOI: 10.1016/j.vaccine.2015.07.022] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The advantage of mucosal vaccination in viral and bacterial infections in different age groups is of enormous clinical relevance. The advantages and potential hazards of intranasal vaccination have always to be considered. The intranasal route for vaccination is very successful for some antigens. Specific adjuvants are necessary. In the nose of rodents there is a structured lymphoid tissue (nose-associated lymphoid tissue (NALT)). This abbreviation should not be used for nasopharynx-associated lymphoid tissue, as this includes parts of the tonsils. In children lymphoid tissue is more dispersed in the nose and not concentrated at the bottom of the dorsal nose ducts as in rodents. There are no data on organized lymphoid tissue in the nose of adults. In NALT of rodents there is a unique structure of adhesion molecule expression; the postnatal development and the different composition of T and B lymphocytes in comparison with Peyer's patches document the uniqueness of this lymphoid organ. There is also a mucosa in the nose with antigen-presenting dendritic cells. Thus, it is often unclear whether intranasal vaccination is initiated via NALT or the diffuse nasal mucosa. There are still many open questions e. g., which adjuvant is necessary for a specific virus, bacterium or other allergen, how many doses are critical for an effective nasal vaccination. Species differences are of major importance when extrapolating results from rodents to humans.
Collapse
Affiliation(s)
- Reinhard Pabst
- Institute of Immunomorphology Centre of Anatomy Medical School, Hannover, Germany.
| |
Collapse
|
29
|
Pan Q, Pais R, Ohandjo A, He C, He Q, Omosun Y, Igietseme JU, Eko FO. Comparative evaluation of the protective efficacy of two formulations of a recombinant Chlamydia abortus subunit candidate vaccine in a mouse model. Vaccine 2015; 33:1865-72. [PMID: 25698486 DOI: 10.1016/j.vaccine.2015.02.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Revised: 01/22/2015] [Accepted: 02/04/2015] [Indexed: 10/24/2022]
Abstract
Chlamydia abortus (C. abortus) is the causative agent of ovine enzootic abortion (OEA) and poses a zoonotic risk to pregnant women. Current live attenuated 1B vaccines are efficacious but cause disease in vaccinated animals and inactivated vaccines are only marginally protective. We tested the ability of a new C. abortus subunit vaccine candidate based on the conserved and immunogenic polymorphic membrane protein D (Pmp18D) formulated in CpG1826+FL (Fms-like tyrosine kinase 3 Ligand; Flt3L) or Vibrio cholerae ghosts (VCG) to induce innate and cross protective immunity against genital C. abortus infection. We found that delivery of rPmp18D with VCG was more effective than with CpG+FL in up-regulating the expression of molecules critically involved in T cell activation and differentiation, including MHC II, CD40, CD80, and CD86, activation of TLRs and NLRP3 inflammasome engagement, and secretion of IL-1β and TNF-α but not IL-10 and IL-4. rVCG-Pmp18D-immunized mice elicited more robust antigen-specific IFN-γ, IgA and IgG2c antibody responses compared to CpG+FL-delivered rPmp18D. Based on the number of mice with positive vaginal cultures, length of vaginal shedding, and number of inclusion forming units recovered following challenge with the heterologous C. abortus strain B577, vaccine delivery with VCG induced superior protective immunity than delivery with a combination of CpG1826 and FL, a nasal DC-targeting adjuvant. These results demonstrate that the ability of VCG to enhance protective immunity against genital C. abortus infection is superior to that of CpG+FL adjuvants.
Collapse
Affiliation(s)
- Qing Pan
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA, USA; College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Roshan Pais
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA, USA
| | - Adaugo Ohandjo
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA, USA
| | - Cheng He
- College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Qing He
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA, USA
| | - Yusuf Omosun
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA, USA
| | - J U Igietseme
- Centers for Disease Control and Prevention (CDC), Atlanta, GA, USA
| | - F O Eko
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
30
|
Sato S, Kiyono H, Fujihashi K. Mucosal Immunosenescence in the Gastrointestinal Tract: A Mini-Review. Gerontology 2014; 61:336-42. [PMID: 25531743 DOI: 10.1159/000368897] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Accepted: 10/07/2014] [Indexed: 01/15/2023] Open
Abstract
It has been shown that pathogen-specific secretory IgA (SIgA) antibody (Ab) is the major player at mucosal surfaces for host defense. However, alterations in the mucosal immune system occur in advanced aging, which results in a failure of induction of SIgA Abs for the protection from infectious diseases. Signs of mucosal senescence first appear in the gut immune system. Further, changes in the intestinal microbiota most likely influence mucosal immunity. To overcome the immunological aging decline in mucosal immunity, several adjuvant systems including mucosal dendritic cell targeting have been shown to be attractive and effective immunological strategies. Similarly, microfold (M) cells involved in the antigen (Ag) uptake are ideal targets for facilitating Ag-specific mucosal immune responses. However, the numbers of M cells are reduced in aged mice. In this regard, Spi-B, an essential transcription factor for the functional and structural differentiation of M cells, could be a potent strategy for the induction of effective mucosal immunity in aging.
Collapse
Affiliation(s)
- Shintaro Sato
- Division of Mucosal Immunology, Department of Microbiology and Immunology, The University of Tokyo, Tokyo, Japan
| | | | | |
Collapse
|
31
|
Wang Y, Yamamoto Y, Shigemori S, Watanabe T, Oshiro K, Wang X, Wang P, Sato T, Yonekura S, Tanaka S, Kitazawa H, Shimosato T. Inhibitory/suppressive oligodeoxynucleotide nanocapsules as simple oral delivery devices for preventing atopic dermatitis in mice. Mol Ther 2014; 23:297-309. [PMID: 25502904 DOI: 10.1038/mt.2014.239] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2014] [Accepted: 12/05/2014] [Indexed: 12/19/2022] Open
Abstract
Here, we report a simple and low-cost oral oligodeoxynucleotide (ODN) delivery system targeted to the gut Peyer's patches (PPs). This system requires only Dulbecco's modified eagle's medium, calcium chloride, ODNs, and basic laboratory equipment. ODN nanocapsules (ODNcaps) were directly delivered to the PPs through oral administration and were taken up by macrophages in the PPs, where they induced an immune response. Long-term continuous oral dosing with inhibitory/suppressive ODNcaps (iODNcaps, "iSG3caps" in this study) was evaluated using an atopic dermatitis mouse model to visually monitor disease course. Administration of iSG3caps improved skin lesions and decreased epidermal thickness. Underlying this effect is the ability of iSG3 to bind to and prevent phosphorylation of signal transducer and activator of transcription 6, thereby blocking the interleukin-4 signaling cascade mediated by binding of allergens to type 2 helper T cells. The results of our iSG3cap oral delivery experiments suggest that iSG3 may be useful for treating allergic diseases.
Collapse
Affiliation(s)
- Yeqin Wang
- Interdisciplinary Graduate School of Science and Technology, Shinshu University, Kamiina, Japan
| | | | - Suguru Shigemori
- 1] Interdisciplinary Graduate School of Science and Technology, Shinshu University, Kamiina, Japan [2] Research Fellow of the Japan Society for the Promotion of Science (JSPS), Chiyoda-ku, Japan
| | | | - Kazushi Oshiro
- Graduate School of Agriculture, Shinshu University, Kamiina, Japan
| | - Xinyu Wang
- Graduate School of Agriculture, Shinshu University, Kamiina, Japan
| | - Pengfei Wang
- Graduate School of Agriculture, Shinshu University, Kamiina, Japan
| | - Takashi Sato
- Department of Internal Medicine and Clinical Immunology, Graduate School of Medicine, Yokohama City University, Yokohama, Japan
| | - Shinichi Yonekura
- Department of Interdisciplinary Genome Sciences and Cell Metabolism, Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research (ICCER), Shinshu University, Kamiina, Japan
| | - Sachi Tanaka
- Frontier Agriscience and Technology Center (FAST), Shinshu University, Kamiina, Japan
| | - Haruki Kitazawa
- Food and Feed Immunology Group, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Takeshi Shimosato
- 1] Interdisciplinary Graduate School of Science and Technology, Shinshu University, Kamiina, Japan [2] Graduate School of Agriculture, Shinshu University, Kamiina, Japan [3] Department of Interdisciplinary Genome Sciences and Cell Metabolism, Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research (ICCER), Shinshu University, Kamiina, Japan
| |
Collapse
|
32
|
EL-Malky MA, Al-Harthi SA, Mohamed RT, EL Bali MA, Saudy NS. Vaccination with Toxoplasma lysate antigen and CpG oligodeoxynucleotides: comparison of immune responses in intranasal versus intramuscular administrations. Parasitol Res 2014; 113:2277-84. [PMID: 24728558 DOI: 10.1007/s00436-014-3882-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2014] [Accepted: 03/24/2014] [Indexed: 10/25/2022]
Abstract
Toxoplasma gondii (T. gondii) is one of the most successful intracellular protozoan parasites on earth and highly prevalent in most warm-blooded vertebrates. There are no drugs that target the chronic cyst stage of this infection; therefore, development of an effective vaccine would be an important advance in disease control. Oligodeoxynucleotides (ODN) which contain immunostimulatory CG motifs (CpG ODN) can promote T-helper 1 (Th1) responses, an adjuvant activity that is desirable for vaccination against intracellular pathogen. In this study, we compare the immune responses of Toxoplasma susceptible C57BL/6 mice following intranasal and intramuscular vaccination with Toxoplasma lysate antigen (TLA) with or without CpG ODN as adjuvant. Immunized and control non-immunized mice were challenged with 85 cyst of the moderately virulent Beverley strain of T. gondii. Intranasal vaccination gave significantly a higher protection compared to other groups as indicated by prolonged survival and significantly reduced brain cyst burden (P < 0.01). Intranasal vaccination stimulated cellular immunity towards Th1 response characterized by significant INF-γ production (P < 0.01). Furthermore, fecal IgA antibody levels as an indicator of mucosal immune responses were significantly higher (P < 0.05) in intranasal vaccinated group before the challenge compared to all other groups. Intranasal vaccination was not able to upgrade the Th1 humoral arm. In contrast, intramuscular vaccination enhanced humoral immunity towards a type Th1 pattern characterized by a significant increase of specific IgG and Ig2a. Our results suggest that intranasal administration of CpG/TLA would provide a stable, pronounced, and effective vaccine against toxoplasmosis through stimulation of Th1 cellular immunity and mucosal IgA.
Collapse
Affiliation(s)
- Mohamed A EL-Malky
- Department of Medical Parasitology, Faculty of Medicine, Umm AL-Qura University, Makkah, Kingdom of Saudi Arabia,
| | | | | | | | | |
Collapse
|
33
|
Fujihashi K, Sato S, Kiyono H. Mucosal adjuvants for vaccines to control upper respiratory infections in the elderly. Exp Gerontol 2014; 54:21-6. [PMID: 24440991 DOI: 10.1016/j.exger.2014.01.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2013] [Revised: 01/02/2014] [Accepted: 01/06/2014] [Indexed: 11/19/2022]
Abstract
Influenza virus and Streptococcus pneumoniae are two major pathogens that lead to significant morbidity and mortality in the elderly. Since both pathogens enter the host via the mucosa, especially the upper respiratory tract (URT), it is essential to elicit pathogen-specific secretory IgA (SIgA) antibody (Ab) responses at mucosal surfaces for defense of the elderly. However, as aging occurs, alterations in the mucosal immune system of older individuals result in a failure to induce SIgA Abs for protection from these infections. To overcome mucosal immunosenescence, we have developed a mucosal dendritic cell targeting, novel double adjuvant system which we show to be an attractive and effective immunological modulator. This system induces a more balanced Th1- and Th2-type cytokine response which supports both mucosal SIgA and systemic IgG1 and IgG2a Ab responses. Thus, adaptation of this adjuvant system to nasal vaccines for influenza virus and S. pneumoniae could successfully provide protection by supporting pathogen-specific SIgA Ab responses in the URT in the mouse model of aging. In summary, a double adjuvant system is considered to be an attractive and potentially important strategy for the future development of mucosal vaccines for the elderly.
Collapse
Affiliation(s)
- Kohtaro Fujihashi
- Department of Pediatric Dentistry, The Immunobiology Vaccine Center, The Institute of Oral Health Research, The School of Dentistry, The University of Alabama at Birmingham, Birmingham, AL 35294-0007, USA; Department of Microbiology, The Immunobiology Vaccine Center, The Institute of Oral Health Research, The School of Dentistry, The University of Alabama at Birmingham, Birmingham, AL 35294-0007, USA.
| | - Shintaro Sato
- Division of Mucosal Immunology, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo, Japan; International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo, Japan
| | - Hiroshi Kiyono
- Department of Pediatric Dentistry, The Immunobiology Vaccine Center, The Institute of Oral Health Research, The School of Dentistry, The University of Alabama at Birmingham, Birmingham, AL 35294-0007, USA; Department of Microbiology, The Immunobiology Vaccine Center, The Institute of Oral Health Research, The School of Dentistry, The University of Alabama at Birmingham, Birmingham, AL 35294-0007, USA; Division of Mucosal Immunology, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo, Japan; International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo, Japan
| |
Collapse
|
34
|
Liang J, Fu J, Kang H, Lin J, Yu Q, Yang Q. Comparison of 3 kinds of Toll-like receptor ligands for inactivated avian H5N1 influenza virus intranasal immunization in chicken. Poult Sci 2013; 92:2651-60. [PMID: 24046412 DOI: 10.3382/ps.2013-03193] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
To evaluate the effects of co-administration of inactivated avian influenza H5N1 virus (IAIV) and different Toll-like receptor (TLR) ligands in chickens, 10-d-old chickens were immunized intranasally with IAIV and TLR ligand [Bacillus subtilis spores, polyinosinic-polycytidylic acid, and CpG oligodeoxynucleotides (CpG-ODN), respectively]. The results showed that both anti-avian influenza virus (AIV) specific secretory IgA level in respiratory tract and anti-AIV specific IgG level in serum significantly increased, as well as the expressions of IL-12, interferon-γ, IL-6, and TLR in the nasal cavity and trachea after intranasal immunization with IAIV and TLR ligand. Among the used TLR ligands, B. subtilis spores as the adjuvant for nasal IAIV had the strongest effect on the expression of IL-6 and IL-12 (P < 0.01), whereas the CpG-ODN could present an advantageous effect on the induction of anti-AIV specific IgG and neutralization antibodies (P < 0.01). The chickens that were previously co-administrated with IAIV and B. subtilis spores could survive at an improved rate upon challenge by live AIV H5N1 virus. Our study suggested that B. subtilis spores, polyinosinic-polycytidylic acid, or CpG-ODN all could effectively enhance the local and systemic immune responses to IAIV in chickens. Considering of the effects and cost of these TLR ligands, we prospected that B. subtilis spores might serve as a more affordable and efficacious mucosal adjuvant for nasal IAIV in chickens.
Collapse
Affiliation(s)
- Jinfeng Liang
- Key Lab of Animal Physiology and Biochemistry, Ministry of Agriculture, Nanjing Agricultural University, Weigang 1, Nanjing, Jiangsu, 210095, China
| | | | | | | | | | | |
Collapse
|
35
|
Shaw AC, Goldstein DR, Montgomery RR. Age-dependent dysregulation of innate immunity. Nat Rev Immunol 2013; 13:875-87. [PMID: 24157572 DOI: 10.1038/nri3547] [Citation(s) in RCA: 754] [Impact Index Per Article: 62.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
As we age, the innate immune system becomes dysregulated and is characterized by persistent inflammatory responses that involve multiple immune and non-immune cell types and that vary depending on the cell activation state and tissue context. This ageing-associated basal inflammation, particularly in humans, is thought to be induced by several factors, including the reactivation of latent viral infections and the release of endogenous damage-associated ligands of pattern recognition receptors (PRRs). Innate immune cell functions that are required to respond to pathogens or vaccines, such as cell migration and PRR signalling, are also impaired in aged individuals. This immune dysregulation may affect conditions associated with chronic inflammation, such as atherosclerosis and Alzheimer's disease.
Collapse
Affiliation(s)
- Albert C Shaw
- Section of Infectious Diseases, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut 06520, USA
| | | | | |
Collapse
|
36
|
Dehghan S, Kheiri MT, Tabatabaiean M, Darzi S, Tafaghodi M. Dry-powder form of chitosan nanospheres containing influenza virus and adjuvants for nasal immunization. Arch Pharm Res 2013; 36:981-92. [DOI: 10.1007/s12272-013-0043-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2012] [Accepted: 02/04/2013] [Indexed: 12/12/2022]
|
37
|
Abstract
Infectious diseases are responsible for an overwhelming number of deaths worldwide and their clinical management is often hampered by the emergence of multi-drug-resistant strains. Therefore, prevention through vaccination currently represents the best course of action to combat them. However, immune escape and evasion by pathogens often render vaccine development difficult. Furthermore, most currently available vaccines were empirically designed. In this review, we discuss why rational design of vaccines is not only desirable but also necessary. We introduce recent developments towards specifically tailored antigens, adjuvants, and delivery systems, and discuss the methodological gaps and lack of knowledge still hampering true rational vaccine design. Finally, we address the potential and limitations of different strategies and technologies for advancing vaccine development.
Collapse
Affiliation(s)
- Christine Rueckert
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Carlos A. Guzmán
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, Braunschweig, Germany
- * E-mail:
| |
Collapse
|
38
|
NS1-truncated live attenuated virus vaccine provides robust protection to aged mice from viral challenge. J Virol 2012; 86:10293-301. [PMID: 22787224 DOI: 10.1128/jvi.01131-12] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Immunological changes associated with age contribute to the high rates of influenza virus morbidity and mortality in the elderly. Compounding this problem, aged individuals do not respond to vaccination as well as younger, healthy adults. Efforts to increase protection to this demographic group are of utmost importance, as the proportion of the population above the age of 65 is projected to increase in the coming decade. Using a live influenza virus with a truncated nonstructural protein 1 (NS1), we are able to stimulate cellular and humoral immune responses of aged mice comparable to levels seen in young mice. Impressively, a single vaccination provided protection following stringent lethal challenge in aged mice.
Collapse
|
39
|
O'Keeffe M. Conventional dendritic cells may be ideal targets for vaccine strategies in the aged. Immunol Cell Biol 2012; 90:665-6. [DOI: 10.1038/icb.2012.16] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Meredith O'Keeffe
- Centre for Immunology, Burnet Institute, 85 Commercial Road Melbourne 3004 Victoria Australia
| |
Collapse
|