1
|
Abil OZ, Liu S, Yeh YW, Wu Y, Sen Chaudhuri A, Li NS, Deng C, Xiang Z. A mucosal vaccine formulation against tuberculosis by exploiting the adjuvant activity of S100A4-A damage-associated molecular pattern molecule. Vaccine 2024; 42:126151. [PMID: 39089961 DOI: 10.1016/j.vaccine.2024.07.052] [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: 03/11/2024] [Revised: 06/28/2024] [Accepted: 07/15/2024] [Indexed: 08/04/2024]
Abstract
Mycobacterium tuberculosis (Mtb), the causative agent of tuberculosis (TB), remains one of the top three causes of death. Currently, the only licensed vaccine against TB is the bacillus Calmette-Guerin (BCG), which lacks efficacy in preventing and controlling pulmonary TB in adults. We aimed to evaluate a nasal TB vaccine formulation composed of the Mtb-specific vaccine antigen ESAT-6, an Mtb-associated protein that can trigger protective immune responses, and S100A4, a recently characterized novel mucosal adjuvant. Mice were intranasally given recombinant ESAT-6 in the presence or absence of S100A4 as an adjuvant. We have provided experimental evidence demonstrating that S100A4 admixed to ESAT-6 could induce Mtb-specific adaptive immune responses after intranasal immunization. S100A4 remarkably augmented the levels of anti-ESAT-6 IgG in serum and IgA in mucosal sites, including lung exudates, bronchoalveolar lavage fluid (BALF) and nasal lavage. Furthermore, in both lung and spleen tissues, S100A4 strongly promoted ESAT-6-specific expansion of CD4 T cells. Both CD4 and CD8 T cells from these tissues expressed increased levels of IFN-γ, TNF-α, and IL-17, cytokines critical for antimicrobial activity. Antigen-reencounter-induced T cell proliferative responses, a key vaccine performance indicator, were augmented in the spleen of S100A4-adjuvanted mice. Furthermore, CD8 T cells from the spleen and lung tissues of these mice expressed higher levels of granzyme B upon antigen re-stimulation. S100A4-adjuvanted immunization may predict good mucosal protection against TB.
Collapse
Affiliation(s)
- Olifan Zewdie Abil
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, China
| | - Shuwei Liu
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, China
| | - Yu-Wen Yeh
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, China
| | - Yuxuan Wu
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, China
| | - Arka Sen Chaudhuri
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, China
| | - Nga Shan Li
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, China
| | - Chujun Deng
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, China
| | - Zou Xiang
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, China.
| |
Collapse
|
2
|
Oguro-Igashira E, Murakami M, Mori R, Kuwahara R, Kihara T, Kohara M, Fujiwara M, Motooka D, Okuzaki D, Arase M, Toyota H, Peng S, Ogino T, Kitabatake Y, Morii E, Hirota S, Ikeuchi H, Umemoto E, Kumanogoh A, Takeda K. The pyruvate-GPR31 axis promotes transepithelial dendrite formation in human intestinal dendritic cells. Proc Natl Acad Sci U S A 2024; 121:e2318767121. [PMID: 39432783 DOI: 10.1073/pnas.2318767121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 08/30/2024] [Indexed: 10/23/2024] Open
Abstract
The intestinal lumen is rich in gut microbial metabolites that serve as signaling molecules for gut immune cells. G-protein-coupled receptors (GPCRs) sense metabolites and can act as key mediators that translate gut luminal signals into host immune responses. However, the impacts of gut microbe-GPCR interactions on human physiology have not been fully elucidated. Here, we show that GPR31, which is activated by the gut bacterial metabolite pyruvate, is specifically expressed on type 1 conventional dendritic cells (cDC1s) in the lamina propria of the human intestine. Using human induced pluripotent stem cell-derived cDC1s and a monolayer human gut organoid coculture system, we show that cDC1s extend their dendrites toward pyruvate on the luminal side, forming transepithelial dendrites (TED). Accordingly, GPR31 activation via pyruvate enhances the fundamental function of cDC1 by allowing efficient uptake of gut luminal antigens, such as dietary compounds and bacterial particles through TED formation. Our results highlight the role of GPCRs in tuning the human gut immune system according to local metabolic cues.
Collapse
Affiliation(s)
- Eri Oguro-Igashira
- Department of Microbiology and Immunology, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
- World Premier International Research Center Initiative (WPI) Immunology Frontier Research Center, Osaka University, Osaka 565-0871, Japan
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
| | - Mari Murakami
- Department of Microbiology and Immunology, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
- World Premier International Research Center Initiative (WPI) Immunology Frontier Research Center, Osaka University, Osaka 565-0871, Japan
| | - Ryota Mori
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
| | - Ryuichi Kuwahara
- Department of Gastroenterological Surgery, Division of Inflammatory Bowel Disease Surgery, Hyogo Medical University, Hyogo 663-8501, Japan
| | - Takako Kihara
- Department of Surgical Pathology, Hyogo Medical University, Hyogo 663-8501, Japan
| | - Masaharu Kohara
- Department of Pathology, Osaka University Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
| | - Makoto Fujiwara
- Department of Pediatrics, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
| | - Daisuke Motooka
- World Premier International Research Center Initiative (WPI) Immunology Frontier Research Center, Osaka University, Osaka 565-0871, Japan
- Department of Infection Metagenomics, Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Osaka 565-0871, Japan
| | - Daisuke Okuzaki
- World Premier International Research Center Initiative (WPI) Immunology Frontier Research Center, Osaka University, Osaka 565-0871, Japan
- Department of Infection Metagenomics, Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Osaka 565-0871, Japan
- Division of Microbiology and Immunology, Center for Infectious Disease Education and Research, Osaka University, Osaka 565-0871, Japan
- Center for Advanced Modalities and Drug Delivery System, Osaka University, Osaka 565-0871, Japan
| | - Mitsuru Arase
- Department of Microbiology and Immunology, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
- World Premier International Research Center Initiative (WPI) Immunology Frontier Research Center, Osaka University, Osaka 565-0871, Japan
| | - Hironobu Toyota
- Department of Microbiology and Immunology, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
| | - Siyun Peng
- Department of Microbiology and Immunology, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
- World Premier International Research Center Initiative (WPI) Immunology Frontier Research Center, Osaka University, Osaka 565-0871, Japan
| | - Takayuki Ogino
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
| | - Yasuji Kitabatake
- Department of Pediatrics, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
| | - Eiichi Morii
- Department of Pathology, Osaka University Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
| | - Seiichi Hirota
- Department of Surgical Pathology, Hyogo Medical University, Hyogo 663-8501, Japan
| | - Hiroki Ikeuchi
- Department of Gastroenterological Surgery, Division of Inflammatory Bowel Disease Surgery, Hyogo Medical University, Hyogo 663-8501, Japan
| | - Eiji Umemoto
- Laboratory of Microbiology and Immunology, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka 422-8526, Japan
| | - Atsushi Kumanogoh
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Osaka 565-0871, Japan
- Division of Microbiology and Immunology, Center for Infectious Disease Education and Research, Osaka University, Osaka 565-0871, Japan
- Center for Advanced Modalities and Drug Delivery System, Osaka University, Osaka 565-0871, Japan
- Department of Immunopathology, World Premier International Research Center Initiative (WPI) Immunology Frontier Research Center, Osaka University, Osaka 565-0871, Japan
| | - Kiyoshi Takeda
- Department of Microbiology and Immunology, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
- World Premier International Research Center Initiative (WPI) Immunology Frontier Research Center, Osaka University, Osaka 565-0871, Japan
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Osaka 565-0871, Japan
- Division of Microbiology and Immunology, Center for Infectious Disease Education and Research, Osaka University, Osaka 565-0871, Japan
| |
Collapse
|
3
|
Sun M, Liu Y, Ni X, Tan R, Wang Y, Jiang Y, Ke D, Du H, Guo G, Liu K. Intranasal immunization with poly I:C and CpG ODN adjuvants enhances the protective efficacy against Helicobacter pylori infection in mice. Microbes Infect 2024:105433. [PMID: 39461584 DOI: 10.1016/j.micinf.2024.105433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 10/17/2024] [Accepted: 10/19/2024] [Indexed: 10/29/2024]
Abstract
Helicobacter pylori (H. pylori) infection is a serious public health issue, and development of vaccines is a desirable preventive strategy for H. pylori. Toll-like receptor (TLR) ligands have shown potential as vaccine adjuvants that induce immune responses, but polyinosinic-polycytidylic acid (poly I:C), a nucleic acid-based TLR9 ligand, is less well studied in H. pylori vaccine research. Here, we evaluated the effects of poly I:C and CpG oligodeoxynucleotide (CpG ODN), a nucleic acid TLR3 ligand, as adjuvants in combination with the H. pylori recombinant proteins LpoB and UreA to protect against H. pylori infection. For analysis of specific immune responses, the levels of specific antibodies and splenic cytokines were measured in the immunized mice. Compared with CpG ODN, poly I:C could induce mucosal sIgA antibody responses and reduce H. pylori colonization. Additionally, the combination of poly I:C and CpG ODN caused greater immunoprotection and significantly reduced gastritis, exerting synergistic effects. Analysis of splenic cytokines revealed that poly I:C mainly triggered a mixed Th1/Th2/Th17 immune response, whereas the combination of CpG ODN and poly I:C induced a Th1/Th17 immune response. Our findings indicated that increased levels of mucosal sIgA antibodies and a robust splenic Th1/Th17 immune response were associated with reduced H. pylori colonization in vaccinated mice. This study identified a potential TLR ligand adjuvant for developing more effective H. pylori vaccines.
Collapse
Affiliation(s)
- Min Sun
- Biopharmaceutical Research Institute, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yu Liu
- Biopharmaceutical Research Institute, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xiumei Ni
- Biopharmaceutical Research Institute, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Runqing Tan
- Biopharmaceutical Research Institute, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yi Wang
- Biopharmaceutical Research Institute, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yajun Jiang
- Biopharmaceutical Research Institute, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Dingxin Ke
- Biopharmaceutical Research Institute, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Han Du
- Biopharmaceutical Research Institute, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Gang Guo
- Biopharmaceutical Research Institute, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Kaiyun Liu
- Biopharmaceutical Research Institute, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
4
|
Reddiar SB, Xie Y, Abdallah M, Han S, Hu L, Feeney OM, Gracia G, Anshabo A, Lu Z, Farooq MA, Styles IK, Phillips ARJ, Windsor JA, Porter CJH, Cao E, Trevaskis NL. Intestinal Lymphatic Biology, Drug Delivery, and Therapeutics: Current Status and Future Directions. Pharmacol Rev 2024; 76:1326-1398. [PMID: 39179383 DOI: 10.1124/pharmrev.123.001159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 07/29/2024] [Accepted: 08/14/2024] [Indexed: 08/26/2024] Open
Abstract
Historically, the intestinal lymphatics were considered passive conduits for fluids, immune cells, dietary lipids, lipid soluble vitamins, and lipophilic drugs. Studies of intestinal lymphatic drug delivery in the late 20th century focused primarily on the drugs' physicochemical properties, especially high lipophilicity, that resulted in intestinal lymphatic transport. More recent discoveries have changed our traditional view by demonstrating that the lymphatics are active, plastic, and tissue-specific players in a range of biological and pathological processes, including within the intestine. These findings have, in turn, inspired exploration of lymph-specific therapies for a range of diseases, as well as the development of more sophisticated strategies to actively deliver drugs or vaccines to the intestinal lymph, including a range of nanotechnologies, lipid prodrugs, and lipid-conjugated materials that "hitchhike" onto lymphatic transport pathways. With the increasing development of novel therapeutics such as biologics, there has been interest in whether these therapeutics are absorbed and transported through intestinal lymph after oral administration. Here we review the current state of understanding of the anatomy and physiology of the gastrointestinal lymphatic system in health and disease, with a focus on aspects relevant to drug delivery. We summarize the current state-of-the-art approaches to deliver drugs and quantify their uptake into the intestinal lymphatic system. Finally, and excitingly, we discuss recent examples of significant pharmacokinetic and therapeutic benefits achieved via intestinal lymphatic drug delivery. We also propose approaches to advance the development and clinical application of intestinal lymphatic delivery strategies in the future. SIGNIFICANCE STATEMENT: This comprehensive review details the understanding of the anatomy and physiology of the intestinal lymphatic system in health and disease, with a focus on aspects relevant to drug delivery. It highlights current state-of-the-art approaches to deliver drugs to the intestinal lymphatics and the shift toward the use of these strategies to achieve pharmacokinetic and therapeutic benefits for patients.
Collapse
Affiliation(s)
- Sanjeevini Babu Reddiar
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| | - Yining Xie
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| | - Mohammad Abdallah
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| | - Sifei Han
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| | - Luojuan Hu
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| | - Orlagh M Feeney
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| | - Gracia Gracia
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| | - Abel Anshabo
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| | - Zijun Lu
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| | - Muhammad Asim Farooq
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| | - Ian K Styles
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| | - Anthony R J Phillips
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| | - John A Windsor
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| | - Christopher J H Porter
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| | - Enyuan Cao
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| | - Natalie L Trevaskis
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| |
Collapse
|
5
|
Grego E, Kelly SM, McGill JL, Wannemuehler M, Narasimhan B. Bovine Respiratory Syncytial Virus Nanovaccine Induces Long-Lasting Humoral Immunity in Mice. ACS Pharmacol Transl Sci 2024; 7:3205-3215. [PMID: 39421663 PMCID: PMC11480889 DOI: 10.1021/acsptsci.4c00375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 08/28/2024] [Accepted: 08/30/2024] [Indexed: 10/19/2024]
Abstract
With limited therapies and vaccines available, human respiratory syncytial virus (HRSV) has a significant negative health impact on all age groups but particularly on infants, young children, and older adults. Bovine respiratory syncytial virus (BRSV) is pathogenically and antigenically similar to HRSV. Building upon previous studies using a BRSV nanovaccine coencapsulating multiple proteins, this work demonstrates the development and comparative evaluation of a coencapsulated nanovaccine to a cocktail nanovaccine formulation composed of polyanhydride nanoparticles encapsulating BRSV postfusion (F) glycoprotein and CpG ODN 1668 coadjuvant delivered simultaneously with nanoparticles encapsulating BRSV attachment glycoprotein (G) and CpG ODN 1668. These nanovaccine formulations were administered to C57BL/6 mice by one of two prime-boost regimens (i.e., intranasal/intranasal or intranasal/subcutaneous) followed by assessment of humoral immunity. The cocktail nanovaccine induced sustained anti-F and anti-G serum IgG antibody responses for 12 weeks postprimary immunization. Using polyanhydride particles to deliver G protein in a prime-boost regime also significantly induced serum anti-G antibodies compared to protein and coadjuvant alone. Serum IgG induced by the nanovaccine demonstrated virus-neutralizing capability from 42 to 119 days postprimary immunization. Further, anti-F IgG antibodies were detected in the bronchoalveolar lavage fluid of vaccinated animals. Finally, the nanovaccine induced long-lived anti-F antibody secreting plasma cells that were detectable in the bone marrow 205 days postprimary immunization. Overall, the BRSV nanovaccine(s) successfully induced long-lived humoral immune responses capable of virus neutralization, making this a promising vaccine candidate for further evaluation in other relevant animal models.
Collapse
Affiliation(s)
- Elizabeth Grego
- Chemical
& Biological Engineering, Iowa State
University, Ames, Iowa 50011, United States
- Nanovaccine
Institute, Ames, Iowa 50011, United States
| | - Sean M. Kelly
- Chemical
& Biological Engineering, Iowa State
University, Ames, Iowa 50011, United States
- Nanovaccine
Institute, Ames, Iowa 50011, United States
| | - Jodi L. McGill
- Nanovaccine
Institute, Ames, Iowa 50011, United States
- Veterinary
Microbiology & Preventive Medicine, Iowa State University, Ames, Iowa 50011, United States
| | - Michael Wannemuehler
- Nanovaccine
Institute, Ames, Iowa 50011, United States
- Veterinary
Microbiology & Preventive Medicine, Iowa State University, Ames, Iowa 50011, United States
| | - Balaji Narasimhan
- Chemical
& Biological Engineering, Iowa State
University, Ames, Iowa 50011, United States
- Nanovaccine
Institute, Ames, Iowa 50011, United States
| |
Collapse
|
6
|
Lokras AG, Bobak TR, Baghel SS, Sebastiani F, Foged C. Advances in the design and delivery of RNA vaccines for infectious diseases. Adv Drug Deliv Rev 2024; 213:115419. [PMID: 39111358 DOI: 10.1016/j.addr.2024.115419] [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: 03/01/2024] [Revised: 07/19/2024] [Accepted: 07/30/2024] [Indexed: 08/23/2024]
Abstract
RNA medicines represent a paradigm shift in treatment and prevention of critical diseases of global significance, e.g., infectious diseases. The highly successful messenger RNA (mRNA) vaccines against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) were developed at record speed during the coronavirus disease 2019 pandemic. A consequence of this is exceptionally shortened vaccine development times, which in combination with adaptability makes the RNA vaccine technology highly attractive against infectious diseases and for pandemic preparedness. Here, we review state of the art in the design and delivery of RNA vaccines for infectious diseases based on different RNA modalities, including linear mRNA, self-amplifying RNA, trans-amplifying RNA, and circular RNA. We provide an overview of the clinical pipeline of RNA vaccines for infectious diseases, and present analytical procedures, which are paramount for characterizing quality attributes and guaranteeing their quality, and we discuss future perspectives for using RNA vaccines to combat pathogens beyond SARS-CoV-2.
Collapse
Affiliation(s)
- Abhijeet Girish Lokras
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen Ø, Denmark
| | - Thomas Rønnemoes Bobak
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen Ø, Denmark
| | - Saahil Sandeep Baghel
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen Ø, Denmark
| | - Federica Sebastiani
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen Ø, Denmark; Division of Physical Chemistry, Department of Chemistry, Lund University, 22100, Lund, Sweden
| | - Camilla Foged
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen Ø, Denmark.
| |
Collapse
|
7
|
Lei T, Dai T, Zhuang L, Liu Y, Li X, Huang C, Zheng X. Enhanced Systemic and Mucosal Immune Responses to Haemophilus parasuis by Intranasal Administration of Lactic-Co-Glycolic Acid Microspheres. Vaccines (Basel) 2024; 12:1103. [PMID: 39460270 PMCID: PMC11511020 DOI: 10.3390/vaccines12101103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 09/18/2024] [Accepted: 09/24/2024] [Indexed: 10/28/2024] Open
Abstract
Swine Glasser's disease, instigated by Haemophilus parasuis (H. parasuis), is a significant bacterial infection that causes substantial economic losses in pig farming operations. The role of mucosal immunity is pivotal in defending against H. parasuis. This study focused on the construction of PLGA microspheres that encapsulate the outer membrane protein OMP16 from H. parasuis (PLGA-OMP16) and evaluated their immunological effectiveness in a mouse model. After being intranasally immunized twice, the PLGA-OMP16 microspheres effectively induced IgAs in saliva and nasal and lung fluids. The PLGA-OMP16 microspheres also significantly increased the number of anti H. parasuis IgGs in serum. Furthermore, the PLGA-OMP16 microspheres triggered elevated levels of IL-2, IL-4, and IFN-γ. The mice vaccinated with PLGA-OMP16 showed a significant reduction in H. parasuis burden in the spleen and lungs following bacterial challenge. These results indicate that intranasal immunization using PLGA microspheres is a promising adjuvant delivery system for vaccines targeting H. parasuis.
Collapse
Affiliation(s)
- Tianyu Lei
- College of Life Sciences, Longyan University, Longyan 364000, China
| | - Tingting Dai
- College of Life Sciences, Longyan University, Longyan 364000, China
| | - Liyun Zhuang
- College of Life Sciences, Longyan University, Longyan 364000, China
| | - Yiting Liu
- College of Life Sciences, Longyan University, Longyan 364000, China
| | - Xiaohua Li
- College of Life Sciences, Longyan University, Longyan 364000, China
- Fujian Provincial Key Laboratory of Preventive Veterinary Medicine and Veterinary Biotechnology, Longyan 364000, China
| | - Cuiqin Huang
- College of Life Sciences, Longyan University, Longyan 364000, China
- Fujian Provincial Key Laboratory of Preventive Veterinary Medicine and Veterinary Biotechnology, Longyan 364000, China
| | - Xintian Zheng
- College of Life Sciences, Longyan University, Longyan 364000, China
- Fujian Provincial Key Laboratory of Preventive Veterinary Medicine and Veterinary Biotechnology, Longyan 364000, China
| |
Collapse
|
8
|
Sefat KMSR, Kumar M, Kehl S, Kulkarni R, Leekha A, Paniagua MM, Ackart DF, Jones N, Spencer C, Podell BK, Ouellet H, Varadarajan N. An intranasal nanoparticle vaccine elicits protective immunity against Mycobacterium tuberculosis. Vaccine 2024; 42:125909. [PMID: 38704256 DOI: 10.1016/j.vaccine.2024.04.055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 04/15/2024] [Accepted: 04/18/2024] [Indexed: 05/06/2024]
Abstract
Mucosal vaccines have the potential to elicit protective immune responses at the point of entry of respiratory pathogens, thus preventing even the initial seed infection. Unlike licensed injectable vaccines, mucosal vaccines comprising protein subunits are only in development. One of the primary challenges associated with mucosal vaccines has been identifying and characterizing safe yet effective mucosal adjuvants that can effectively prime multi-factorial mucosal immunity. In this study, we tested NanoSTING, a liposomal formulation of the endogenous activator of the stimulator of interferon genes (STING) pathway, cyclic guanosine adenosine monophosphate (cGAMP), as a mucosal adjuvant. We formulated a vaccine based on the H1 antigen (fusion protein of Ag85b and ESAT-6) adjuvanted with NanoSTING. Intranasal immunization of NanoSTING-H1 elicited a strong T-cell response in the lung of vaccinated animals characterized by (a) CXCR3+ KLRG1- lung resident T cells that are known to be essential for controlling bacterial infection, (b) IFNγ-secreting CD4+ T cells which is necessary for intracellular bactericidal activity, and (c) IL17-secreting CD4+ T cells that can confer protective immunity against multiple clinically relevant strains of Mtb. Upon challenge with aerosolized Mycobacterium tuberculosis Erdman strain, intranasal NanoSTING-H1 provides protection comparable to subcutaneous administration of the live attenuated Mycobacterium bovis vaccine strain Bacille-Calmette-Guérin (BCG). Our results indicate that NanoSTING adjuvanted protein vaccines can elicit a multi-factorial immune response that protects from infection by M. tuberculosis.
Collapse
Affiliation(s)
- K M Samiur Rahman Sefat
- Department of Chemical and Biomolecular Engineering, University of Houston, Houston, TX 77054, USA
| | - Monish Kumar
- Department of Chemical and Biomolecular Engineering, University of Houston, Houston, TX 77054, USA
| | - Stephanie Kehl
- Department of Biological Sciences, University of Texas, El Paso, TX 79968, USA
| | - Rohan Kulkarni
- Department of Chemical and Biomolecular Engineering, University of Houston, Houston, TX 77054, USA
| | - Ankita Leekha
- Department of Chemical and Biomolecular Engineering, University of Houston, Houston, TX 77054, USA
| | - Melisa-Martinez Paniagua
- Department of Chemical and Biomolecular Engineering, University of Houston, Houston, TX 77054, USA
| | - David F Ackart
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO 80523, USA
| | - Nicole Jones
- Department of Biological Sciences, University of Texas, El Paso, TX 79968, USA
| | - Charles Spencer
- Department of Biological Sciences, University of Texas, El Paso, TX 79968, USA
| | - Brendan K Podell
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO 80523, USA
| | - Hugues Ouellet
- Department of Biological Sciences, University of Texas, El Paso, TX 79968, USA
| | - Navin Varadarajan
- Department of Chemical and Biomolecular Engineering, University of Houston, Houston, TX 77054, USA.
| |
Collapse
|
9
|
Seefeld ML, Templeton EL, Lehtinen JM, Sinclair N, Yadav D, Hartwell BL. Harnessing the potential of the NALT and BALT as targets for immunomodulation using engineering strategies to enhance mucosal uptake. Front Immunol 2024; 15:1419527. [PMID: 39286244 PMCID: PMC11403286 DOI: 10.3389/fimmu.2024.1419527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 08/08/2024] [Indexed: 09/19/2024] Open
Abstract
Mucosal barrier tissues and their mucosal associated lymphoid tissues (MALT) are attractive targets for vaccines and immunotherapies due to their roles in both priming and regulating adaptive immune responses. The upper and lower respiratory mucosae, in particular, possess unique properties: a vast surface area responsible for frontline protection against inhaled pathogens but also simultaneous tight regulation of homeostasis against a continuous backdrop of non-pathogenic antigen exposure. Within the upper and lower respiratory tract, the nasal and bronchial associated lymphoid tissues (NALT and BALT, respectively) are key sites where antigen-specific immune responses are orchestrated against inhaled antigens, serving as critical training grounds for adaptive immunity. Many infectious diseases are transmitted via respiratory mucosal sites, highlighting the need for vaccines that can activate resident frontline immune protection in these tissues to block infection. While traditional parenteral vaccines that are injected tend to elicit weak immunity in mucosal tissues, mucosal vaccines (i.e., that are administered intranasally) are capable of eliciting both systemic and mucosal immunity in tandem by initiating immune responses in the MALT. In contrast, administering antigen to mucosal tissues in the absence of adjuvant or costimulatory signals can instead induce antigen-specific tolerance by exploiting regulatory mechanisms inherent to MALT, holding potential for mucosal immunotherapies to treat autoimmunity. Yet despite being well motivated by mucosal biology, development of both mucosal subunit vaccines and immunotherapies has historically been plagued by poor drug delivery across mucosal barriers, resulting in weak efficacy, short-lived responses, and to-date a lack of clinical translation. Development of engineering strategies that can overcome barriers to mucosal delivery are thus critical for translation of mucosal subunit vaccines and immunotherapies. This review covers engineering strategies to enhance mucosal uptake via active targeting and passive transport mechanisms, with a parallel focus on mechanisms of immune activation and regulation in the respiratory mucosa. By combining engineering strategies for enhanced mucosal delivery with a better understanding of immune mechanisms in the NALT and BALT, we hope to illustrate the potential of these mucosal sites as targets for immunomodulation.
Collapse
Affiliation(s)
- Madison L Seefeld
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, United States
| | - Erin L Templeton
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, United States
| | - Justin M Lehtinen
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, United States
| | - Noah Sinclair
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, United States
| | - Daman Yadav
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, United States
| | - Brittany L Hartwell
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, United States
- Center for Immunology, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
10
|
Shi Q, Wang Q, Shen Y, Chen S, Gan S, Lin T, Song F, Ma Y. Escherichia coli LTB26 mutant enhances immune responses to rotavirus antigen VP8 in a mouse model. Mol Immunol 2024; 173:10-19. [PMID: 39004021 DOI: 10.1016/j.molimm.2024.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Revised: 06/03/2024] [Accepted: 07/05/2024] [Indexed: 07/16/2024]
Abstract
Adjuvant is a major supplementary component of vaccines to boost adaptive immune responses. To select an efficient adjuvant from the heat-labile toxin B subunit (LTB) of E. coli, four LTB mutants (numbered LTB26, LTB34, LTB57, and LTB85) were generated by multi-amino acid random replacement. Mice have been intranasally vaccinated with human rotavirus VP8 admixed. Among the four mutants, enzyme-linked immunosorbent assay (ELISA) revealed that LTB26 had enhanced mucosal immune adjuvanticity compared to LTB, showing significantly enhanced immune responses in both serum IgG and mucosal sIgA levels. The 3D modeling analysis suggested that the enhanced immune adjuvanticity of LTB26 might be due to the change of the first LTB α-helix to a β-sheet. The molecular mechanism was studied using transcriptomic and flow cytometric (FCM) analysis. The transcriptomic data demonstrated that LTB26 enhanced immune response by enhancing B cell receptor (BCR) and major histocompatibility complex (MHC) II+-related pathways. Furthermore, LTB26 promoted Th1 and Th2-type immune responses which were confirmed by detecting IFN-γ and IL-4 expression levels. Immunohistochemical analysis demonstrated that LTB26 enhanced both Th1 and Th2 type immunity. Therefore, LTB26 was a potent mucosal immune adjuvant meeting the requirement for use in human clinics in the future.
Collapse
Affiliation(s)
- Qinlin Shi
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Qiujuan Wang
- Department of Biochemistry and Molecular Biology, Basic Medical College, Molecular Medicine & Cancer Research Center, Chongqing Medical University, Yuzhong District, Yi XueYuan Road, No 1, Chongqing 400016, China
| | - Yanxi Shen
- Department of Biochemistry and Molecular Biology, Basic Medical College, Molecular Medicine & Cancer Research Center, Chongqing Medical University, Yuzhong District, Yi XueYuan Road, No 1, Chongqing 400016, China
| | - Sijing Chen
- Department of Biochemistry and Molecular Biology, Basic Medical College, Molecular Medicine & Cancer Research Center, Chongqing Medical University, Yuzhong District, Yi XueYuan Road, No 1, Chongqing 400016, China
| | - Sijie Gan
- Department of Biochemistry and Molecular Biology, Basic Medical College, Molecular Medicine & Cancer Research Center, Chongqing Medical University, Yuzhong District, Yi XueYuan Road, No 1, Chongqing 400016, China
| | - Tao Lin
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Fangzhou Song
- Department of Biochemistry and Molecular Biology, Basic Medical College, Molecular Medicine & Cancer Research Center, Chongqing Medical University, Yuzhong District, Yi XueYuan Road, No 1, Chongqing 400016, China
| | - Yongping Ma
- Department of Biochemistry and Molecular Biology, Basic Medical College, Molecular Medicine & Cancer Research Center, Chongqing Medical University, Yuzhong District, Yi XueYuan Road, No 1, Chongqing 400016, China.
| |
Collapse
|
11
|
Chen Y, Wang Y, Li Z, Jiang H, Pan W, Liu M, Jiang W, Zhang X, Wang F. Preparation and immunological activity evaluation of an intranasal protein subunit vaccine against ancestral and mutant SARS-CoV-2 with curdlan sulfate/O-linked quaternized chitosan nanoparticles as carrier and adjuvant. Int J Biol Macromol 2024; 276:133733. [PMID: 39002905 DOI: 10.1016/j.ijbiomac.2024.133733] [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: 11/28/2023] [Revised: 06/07/2024] [Accepted: 07/06/2024] [Indexed: 07/15/2024]
Abstract
Chitosan and its derivatives are ideal nasal vaccine adjuvant to deliver antigens to immune cells. Previously, we successfully used a chitosan derivative, O-(2-Hydroxyl) propyl-3-trimethyl ammonium chitosan chloride (O-HTCC), and a β-glucan derivative, curdlan sulfate (CS), to prepare a nanoparticle adjuvant CS/O-HTCC which could deliver ovalbumin to antigen presenting cells (APCs) through nasal inhalation. In this article, we used SARS-CoV-2 spike receptor binding domain (S-RBD) as the antigen and CS/O-HTCC nanoparticles as the adjuvant to develop a nasal mucosal protein subunit vaccine, CS/S-RBD/O-HTCC. The humoral immunity, cell-mediated immunity and mucosal immunity induced by vaccines were evaluated. The results showed that CS/S-RBD/O-HTCC could induce desirable immunization with single or bivalent antigen through nasal inoculation, giving one booster vaccination with mutated S-RBD (beta) could bring about a broad cross reaction with ancestral and different mutated S-RBD, and vaccination of the BALB/c mice with CS/S-RBD/O-HTCC containing S-RBD mix antigens (ancestral and omicron) could induce the production of binding and neutralizing antibodies against both of the two antigens. Our results indicate that CS/O-HTCC is a promising nasal mucosal adjuvant to prepare protein subunit vaccine for both primary and booster immunization, and the adjuvant is suitable for loading more than one antigen for preparing multivalent vaccines.
Collapse
MESH Headings
- Chitosan/chemistry
- Animals
- Nanoparticles/chemistry
- beta-Glucans/chemistry
- beta-Glucans/immunology
- SARS-CoV-2/immunology
- Vaccines, Subunit/immunology
- Mice
- Administration, Intranasal
- Spike Glycoprotein, Coronavirus/immunology
- Spike Glycoprotein, Coronavirus/chemistry
- Spike Glycoprotein, Coronavirus/genetics
- Adjuvants, Immunologic/pharmacology
- Mice, Inbred BALB C
- COVID-19/prevention & control
- COVID-19/immunology
- Female
- COVID-19 Vaccines/immunology
- COVID-19 Vaccines/chemistry
- Antibodies, Viral/immunology
- Immunity, Mucosal/drug effects
- Mutation
- Antibodies, Neutralizing/immunology
- Drug Carriers/chemistry
- Adjuvants, Vaccine/chemistry
- Humans
Collapse
Affiliation(s)
- Yipan Chen
- Key Laboratory of Chemical Biology of Natural Products, Ministry of education, Institute of Biochemical and Biotechnological Drug, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, Shandong, China
| | - Yan Wang
- Key Laboratory of Chemical Biology of Natural Products, Ministry of education, Institute of Biochemical and Biotechnological Drug, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, Shandong, China
| | - Zuyi Li
- Key Laboratory of Chemical Biology of Natural Products, Ministry of education, Institute of Biochemical and Biotechnological Drug, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, Shandong, China
| | - Honglei Jiang
- Key Laboratory of Chemical Biology of Natural Products, Ministry of education, Institute of Biochemical and Biotechnological Drug, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, Shandong, China
| | - Wei Pan
- Key Laboratory of Chemical Biology of Natural Products, Ministry of education, Institute of Biochemical and Biotechnological Drug, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, Shandong, China
| | - Minghui Liu
- Key Laboratory of Chemical Biology of Natural Products, Ministry of education, Institute of Biochemical and Biotechnological Drug, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, Shandong, China
| | - Wenjie Jiang
- Key Laboratory of Chemical Biology of Natural Products, Ministry of education, Institute of Biochemical and Biotechnological Drug, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, Shandong, China.
| | - Xinke Zhang
- Department of Pharmacology, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China.
| | - Fengshan Wang
- Key Laboratory of Chemical Biology of Natural Products, Ministry of education, Institute of Biochemical and Biotechnological Drug, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, Shandong, China; NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate-based Medicine, National Glycoengineering Research Center, Shandong University, Jinan 250012, Shandong, China.
| |
Collapse
|
12
|
Huang L, Tang W, He L, Li M, Lin X, Hu A, Huang X, Wu Z, Wu Z, Chen S, Hu Y. Engineered probiotic Escherichia coli elicits immediate and long-term protection against influenza A virus in mice. Nat Commun 2024; 15:6802. [PMID: 39122688 PMCID: PMC11315933 DOI: 10.1038/s41467-024-51182-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 07/31/2024] [Indexed: 08/12/2024] Open
Abstract
Influenza virus infection remains a major global health problem and requires a universal vaccine with broad protection against different subtypes as well as a rapid-response vaccine to provide immediate protection in the event of an epidemic outbreak. Here, we show that intranasal administration of probiotic Escherichia coli Nissle 1917 activates innate immunity in the respiratory tract and provides immediate protection against influenza virus infection within 1 day. Based on this vehicle, a recombinant strain is engineered to express and secret five tandem repeats of the extracellular domain of matrix protein 2 from different influenza virus subtypes. Intranasal vaccination with this strain induces durable humoral and mucosal responses in the respiratory tract, and provides broad protection against the lethal challenge of divergent influenza viruses in female BALB/c mice. Our findings highlight a promising delivery platform for developing mucosal vaccines that provide immediate and sustained protection against respiratory pathogens.
Collapse
Affiliation(s)
- Ling Huang
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Wei Tang
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Lina He
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Mengke Li
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Xian Lin
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
- Hubei JiangXia Laboratory, Wuhan, 430071, China
| | - Ao Hu
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Xindi Huang
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Zhouyu Wu
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Zhiyong Wu
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Shiyun Chen
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China.
| | - Yangbo Hu
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China.
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China.
- Hubei JiangXia Laboratory, Wuhan, 430071, China.
| |
Collapse
|
13
|
Fathi Kisomi M, Yadegar A, Shekari T, Amin M, Llopis-Lorente A, Liu C, Haririan I, Aghdaei HA, Shokrgozar MA, Zali MR, Rad-Malekshahi M, Miri AH, Hamblin MR, Wacker MG. Unveiling the potential role of micro/nano biomaterials in the treatment of Helicobacter pylori infection. Expert Rev Anti Infect Ther 2024; 22:613-630. [PMID: 39210553 DOI: 10.1080/14787210.2024.2391910] [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: 05/30/2024] [Accepted: 08/09/2024] [Indexed: 09/04/2024]
Abstract
INTRODUCTION Helicobacter pylori causes stubborn infections and leads to a variety of stomach disorders, such as peptic ulcer, chronic atrophic gastritis, and gastric cancer. Although antibiotic-based approaches have been widely used against H. pylori, some challenges such as antibiotic resistance are increasing in severity. Therefore, simpler but more effective strategies are needed. AREAS COVERED In this review, basic information on functionalized and non-functionalized micro/nano biomaterials and routes of administration for H. pylori inhibition are provided in an easy-to-understand format. Afterward, in vitro and in vivo studies of some promising bio-platforms including metal-based biomaterials, biopolymers, small-molecule saccharides, and vaccines for H. pylori inhibition are discussed in a holistic manner. EXPERT OPINION Functionalized or non-functionalized micro/nano biomaterials loaded with anti-H. pylori agents can show efficient bactericidal activity with no/slight negative influence on the host gastrointestinal microbiota. However, this claim needs to be substantiated with hard data such as assessment of the biopharmaceutical parameters of anti-H. pylori systems and the measurement of diversity/abundance of bacterial genera in the host gastric/gut microbiota before and after H. pylori eradication.
Collapse
Affiliation(s)
- Misagh Fathi Kisomi
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Abbas Yadegar
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Tara Shekari
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohsen Amin
- Department of Drug and Food Control, Faculty of Pharmacy, and the Institute of Pharmaceutical Sciences, Tehran University of Medical Sciences, Tehran, Iran
| | - Antoni Llopis-Lorente
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Universitat Politècnica de València, Universitat de València, Valencia, Spain
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Chenguang Liu
- College of Marine Life Science, Ocean University of China, Qingdao, P.R. China
| | - Ismaeil Haririan
- Department of Pharmaceutical Biomaterials and Medical Biomaterials Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Hamid Asadzadeh Aghdaei
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Mohammad Reza Zali
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mazda Rad-Malekshahi
- Department of Pharmaceutical Biomaterials and Medical Biomaterials Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Amir Hossein Miri
- Department of Pharmaceutical Biomaterials and Medical Biomaterials Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Michael R Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein, South Africa
| | - Matthias G Wacker
- Department of Pharmacy, Faculty of Science, National University of Singapore, 4 Science Drive 2, Singapore 117545, Singapore
| |
Collapse
|
14
|
Gribonika I, Strömberg A, Chandode RK, Schön K, Lahl K, Bemark M, Lycke N. Migratory CD103 +CD11b + cDC2s in Peyer's patches are critical for gut IgA responses following oral immunization. Mucosal Immunol 2024; 17:509-523. [PMID: 38492746 DOI: 10.1016/j.mucimm.2024.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 03/06/2024] [Accepted: 03/07/2024] [Indexed: 03/18/2024]
Abstract
Induction and regulation of specific intestinal immunoglobulin (Ig)A responses critically depend on dendritic cell (DC) subsets and the T cells they activate in the Peyer's patches (PP). We found that oral immunization with cholera toxin (CT) as an adjuvant resulted in migration-dependent changes in the composition and localization of PP DC subsets with increased numbers of cluster of differentiation (CD)103- conventional DC (cDC)2s and lysozyme-expressing DC (LysoDCs) in the subepithelial dome and of CD103+ cDC2s that expressed CD101 in the T cell zones, while oral ovalbumin (OVA) tolerization was instead associated with greater accumulation of cDC1s and peripherally induced regulatory T cells (pTregs) in this area. Decreased IgA responses were observed after CT-adjuvanted immunization in huCD207DTA mice lacking CD103+ cDC2s, while oral OVA tolerization was inefficient in cDC1-deficient Batf3-/- mice. Using OVA transgenic T cell receptor CD4 T cell adoptive transfer models, we found that co-transferred endogenous wildtype CD4 T cells can hinder the induction of OVA-specific IgA responses through secretion of interleukin-10. CT could overcome this blocking effect, apparently through a modulating effect on pTregs while promoting an expansion of follicular helper T cells. The data support a model where cDC1-induced pTreg normally suppresses PP responses for any given antigen and where CT's oral adjuvanticity effect is dependent on promoting follicular helper T cell responses through induction of CD103+ cDC2s.
Collapse
Affiliation(s)
- Inta Gribonika
- Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden.
| | - Anneli Strömberg
- Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Rakesh K Chandode
- Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Karin Schön
- Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Katharina Lahl
- Immunology Section, Lund University, Lund, Sweden; Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada; Department of Microbiology, Immunology, and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; Section for Experimental and Translational Immunology, Institute for Health Technology, Technical University of Denmark (DTU), Kongens Lyngby, Denmark
| | - Mats Bemark
- Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden; Department of Clinical Immunology and Transfusion Medicine, Sahlgrenska University Hospital, Gothenburg, Sweden; Department of Translational Medicine - Human Immunology, Lund University, Malmö, Sweden.
| | - Nils Lycke
- Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
15
|
Mandviwala AS, Huckriede ALW, Arankalle VA, Patil HP. Mucosal delivery of a prefusogenic-F, glycoprotein, and matrix proteins-based virus-like particle respiratory syncytial virus vaccine induces protective immunity as evidenced by challenge studies in mice. Virology 2024; 598:110194. [PMID: 39096774 DOI: 10.1016/j.virol.2024.110194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 07/18/2024] [Accepted: 07/28/2024] [Indexed: 08/05/2024]
Abstract
RSV infection remains a serious threat to the children all over the world, especially, in the low-middle income countries. Vaccine delivery via the mucosa holds great potential for inducing local immune responses in the respiratory tract. Previously, we reported the development of highly immunogenic RSV virus-like-particles (RSV-VLPs) based on the conformationally stable prefusogenic-F protein (preFg), glycoprotein and matrix protein. Here, to explore whether mucosal delivery of RSV-VLPs is an effective strategy to induce RSV-specific mucosal and systemic immunity, RSV-VLPs were administered via the nasal, sublingual and pulmonary routes to BALB/c mice. The results demonstrate that immunization with the VLPs via the mucosal routes induced minimal mucosal response and yet facilitated modest levels of serum IgG antibodies, enhanced T cell responses and the expression of the lung-homing marker CXCR3 on splenocytes. Immunization with VLPs via all three mucosal routes provided protection against RSV challenge with no signs of RSV induced pathology.
Collapse
Affiliation(s)
- Ahmedali S Mandviwala
- Department of Communicable Diseases, Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to be University), Pune, India
| | - Anke L W Huckriede
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Vidya A Arankalle
- Department of Communicable Diseases, Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to be University), Pune, India
| | - Harshad P Patil
- Department of Communicable Diseases, Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to be University), Pune, India.
| |
Collapse
|
16
|
Teshigahara A, Banba Y, Yoshida H, Kaji M, Zhou Z, Koyama N, Sakai Y, Karrow NA, Ogasawara K, Hirakawa R, Islam J, Furukawa M, Nochi T. Formation of the junctions between lymph follicles in the Peyer's patches even before postweaning activation. Sci Rep 2024; 14:15783. [PMID: 38982122 PMCID: PMC11233632 DOI: 10.1038/s41598-024-65984-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 06/26/2024] [Indexed: 07/11/2024] Open
Abstract
Peyer's patches (PPs), which contain an abundance of B and T cells, play a key role in inducing pivotal immune responses in the intestinal tract. PPs are defined as aggregated lymph follicles, which consist of multiple lymph follicles (LFs) that may interact with each other in a synergistic manner. LFs are thought to be spherical in shape; however, the characteristics of their structure are not fully understood. To elucidate changes in the structure of PPs as individuals grow, we generated serial 2D sections from entire PPs harvested from mice at 2, 4, and 10 weeks of age and performed a 3D analysis using a software, Amira. Although the number of LFs in PPs was not changed throughout the experiment, the volume and surface area of LFs increased significantly, indicating that LFs in PPs develop continuously by recruiting immune cells, even after weaning. In response to the dramatic changes in the intestinal environment after weaning, the development of germinal centers (GCs) in LFs was observed at 4 and 10 weeks (but not 2 weeks) of age. In addition, GCs gradually began to form away from the center of LFs and close to the muscle layer where export lymphatic vessels develop. Importantly, each LF was joined to the adjacent LF; this feature was observed even in preweaning nonactivated PPs. These results suggest that PPs may have a unique organization and structure that enhance immune functions, allowing cells in LFs to have free access to adjacent LFs and egress smoothly from PPs to the periphery upon stimulation after weaning.
Collapse
Affiliation(s)
- Anri Teshigahara
- International Education and Research Center for Food and Agricultural Immunology, Graduate School of Agricultural Science, Tohoku University, Miyagi, 980-8572, Japan
- Laboratory of Animal Functional Morphology, Graduate School of Agricultural Science, Tohoku University, Miyagi, 980-8572, Japan
| | - Yuri Banba
- International Education and Research Center for Food and Agricultural Immunology, Graduate School of Agricultural Science, Tohoku University, Miyagi, 980-8572, Japan
- Laboratory of Animal Functional Morphology, Graduate School of Agricultural Science, Tohoku University, Miyagi, 980-8572, Japan
| | - Hiromi Yoshida
- Institute of Development, Aging and Cancer, Tohoku University, Miyagi, 980-8575, Japan
| | - Mitsuji Kaji
- Institute of Development, Aging and Cancer, Tohoku University, Miyagi, 980-8575, Japan
| | - Zhou Zhou
- International Education and Research Center for Food and Agricultural Immunology, Graduate School of Agricultural Science, Tohoku University, Miyagi, 980-8572, Japan
- Laboratory of Animal Functional Morphology, Graduate School of Agricultural Science, Tohoku University, Miyagi, 980-8572, Japan
| | - Nao Koyama
- International Education and Research Center for Food and Agricultural Immunology, Graduate School of Agricultural Science, Tohoku University, Miyagi, 980-8572, Japan
- Laboratory of Animal Functional Morphology, Graduate School of Agricultural Science, Tohoku University, Miyagi, 980-8572, Japan
| | - Yoshifumi Sakai
- International Education and Research Center for Food and Agricultural Immunology, Graduate School of Agricultural Science, Tohoku University, Miyagi, 980-8572, Japan
| | - Niel A Karrow
- Department of Animal Biosciences, University of Guelph, Ontario, N1G 2W1, Canada
| | - Kouetsu Ogasawara
- Institute of Development, Aging and Cancer, Tohoku University, Miyagi, 980-8575, Japan
| | - Ryota Hirakawa
- International Education and Research Center for Food and Agricultural Immunology, Graduate School of Agricultural Science, Tohoku University, Miyagi, 980-8572, Japan
- Laboratory of Animal Functional Morphology, Graduate School of Agricultural Science, Tohoku University, Miyagi, 980-8572, Japan
- Laboratory of Animal Mucosal Immunology, Graduate School of Agricultural Science, Tohoku University, Miyagi, 980-8572, Japan
| | - Jahidul Islam
- International Education and Research Center for Food and Agricultural Immunology, Graduate School of Agricultural Science, Tohoku University, Miyagi, 980-8572, Japan
- Laboratory of Animal Functional Morphology, Graduate School of Agricultural Science, Tohoku University, Miyagi, 980-8572, Japan
| | - Mutsumi Furukawa
- International Education and Research Center for Food and Agricultural Immunology, Graduate School of Agricultural Science, Tohoku University, Miyagi, 980-8572, Japan
- Laboratory of Animal Functional Morphology, Graduate School of Agricultural Science, Tohoku University, Miyagi, 980-8572, Japan
- Laboratory of Animal Mucosal Immunology, Graduate School of Agricultural Science, Tohoku University, Miyagi, 980-8572, Japan
| | - Tomonori Nochi
- International Education and Research Center for Food and Agricultural Immunology, Graduate School of Agricultural Science, Tohoku University, Miyagi, 980-8572, Japan.
- Laboratory of Animal Functional Morphology, Graduate School of Agricultural Science, Tohoku University, Miyagi, 980-8572, Japan.
- Department of Animal Biosciences, University of Guelph, Ontario, N1G 2W1, Canada.
- Laboratory of Animal Mucosal Immunology, Graduate School of Agricultural Science, Tohoku University, Miyagi, 980-8572, Japan.
- Division of Mucosal Vaccines, International Vaccine Design Center, The Institute of Medical Science, The University of Tokyo, Tokyo, 108-8639, Japan.
- Center for Professional Development, Institute for Excellence in Higher Education, Tohoku University, Miyagi, 980-8576, Japan.
| |
Collapse
|
17
|
Silvestre D, Moreno G, Argüelles MH, Tomás Fariña J, Biedma ME, Peri Ibáñez ES, Mandile MG, Glikmann G, Rumbo M, Castello AA, Temprana CF. Display of FliC131 on the Surface of Lactococcus lactis as a Strategy to Increase its Adjuvanticity for Mucosal Immunization. J Pharm Sci 2024; 113:1794-1803. [PMID: 38522753 DOI: 10.1016/j.xphs.2024.03.013] [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: 11/30/2023] [Revised: 03/15/2024] [Accepted: 03/15/2024] [Indexed: 03/26/2024]
Abstract
Research on innovative mucosal adjuvants is essential to develop new vaccines for safe mucosal application. In this work, we propose the development of a Lactococcus lactis that expresses a variant of flagellin on its surface (FliC131*), to increase the adjuvanticity of the living cell and cell wall-derived particles (CWDP). We optimized the expression of FliC131*, and confirmed its identity and localization by Western blot and flow cytometry. We also generated CWDP containing FliC131* (CDWP-FliC131*) and evaluated their storage stability. Lastly, we measured the human TLR5 stimulating activity in vitro and assessed the adjuvanticity in vivo using ovalbumin (OVA) as a model antigen. As a result, we generated L. lactis/pCWA-FliC131*, that expresses and displays FliC131* on its surface, obtained the corresponding CWDP-FliC131*, and showed that both activated hTLR5 in vitro in a dose-dependent manner. Furthermore, CWDP-FliC131* retained this biological activity after being lyophilized and stored for a year. Finally, intranasal immunization of mice with OVA plus live L. lactis/pCWA-FliC131* or CWDP-FliC131* induced OVA-specific IgG and IgA in serum, intestinal lavages, and bronchoalveolar lavages. Our work demonstrates the potential of this recombinant L. lactis with an enhanced adjuvant effect, prompting its further evaluation for the design of novel mucosal vaccines.
Collapse
Affiliation(s)
- Dalila Silvestre
- Laboratorio de Inmunología y Virología, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes, Roque Sáenz Peña 352, Bernal, 1876, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Godoy Cruz 2290, Ciudad Autónoma de Buenos Aires, 1425, Argentina
| | - Griselda Moreno
- Instituto de Estudios Inmunológicos y Fisiopatológicos (IIFP), Facultad de Ciencias Exactas, Universidad Nacional de La Plata - Consejo Nacional de Investigaciones Científicas y Técnicas (UNLP-CONICET), Boulevard 120 1489, La Plata, 1900, Argentina
| | - Marcelo H Argüelles
- Laboratorio de Inmunología y Virología, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes, Roque Sáenz Peña 352, Bernal, 1876, Argentina
| | - Julieta Tomás Fariña
- Laboratorio de Inmunología y Virología, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes, Roque Sáenz Peña 352, Bernal, 1876, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Godoy Cruz 2290, Ciudad Autónoma de Buenos Aires, 1425, Argentina
| | - Marina E Biedma
- Instituto de Estudios Inmunológicos y Fisiopatológicos (IIFP), Facultad de Ciencias Exactas, Universidad Nacional de La Plata - Consejo Nacional de Investigaciones Científicas y Técnicas (UNLP-CONICET), Boulevard 120 1489, La Plata, 1900, Argentina
| | - Estefanía S Peri Ibáñez
- Laboratorio de Inmunología y Virología, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes, Roque Sáenz Peña 352, Bernal, 1876, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Godoy Cruz 2290, Ciudad Autónoma de Buenos Aires, 1425, Argentina
| | - Marcelo G Mandile
- Laboratorio de Inmunología y Virología, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes, Roque Sáenz Peña 352, Bernal, 1876, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Godoy Cruz 2290, Ciudad Autónoma de Buenos Aires, 1425, Argentina
| | - Graciela Glikmann
- Laboratorio de Inmunología y Virología, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes, Roque Sáenz Peña 352, Bernal, 1876, Argentina
| | - Martín Rumbo
- Instituto de Estudios Inmunológicos y Fisiopatológicos (IIFP), Facultad de Ciencias Exactas, Universidad Nacional de La Plata - Consejo Nacional de Investigaciones Científicas y Técnicas (UNLP-CONICET), Boulevard 120 1489, La Plata, 1900, Argentina
| | - Alejandro A Castello
- Laboratorio de Inmunología y Virología, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes, Roque Sáenz Peña 352, Bernal, 1876, Argentina; Instituto de Ciencias de la Salud, Universidad Nacional Arturo Jauretche, Av. Calchaquí 6200, Florencio Varela, 1888, Buenos Aires, Argentina
| | - C Facundo Temprana
- Laboratorio de Inmunología y Virología, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes, Roque Sáenz Peña 352, Bernal, 1876, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Godoy Cruz 2290, Ciudad Autónoma de Buenos Aires, 1425, Argentina.
| |
Collapse
|
18
|
Shetty S, Alvarado PC, Pettie D, Collier JH. Next-Generation Vaccine Development with Nanomaterials: Recent Advances, Possibilities, and Challenges. Annu Rev Biomed Eng 2024; 26:273-306. [PMID: 38959389 DOI: 10.1146/annurev-bioeng-110122-124359] [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] [Indexed: 07/05/2024]
Abstract
Nanomaterials are becoming important tools for vaccine development owing to their tunable and adaptable nature. Unique properties of nanomaterials afford opportunities to modulate trafficking through various tissues, complement or augment adjuvant activities, and specify antigen valency and display. This versatility has enabled recent work designing nanomaterial vaccines for a broad range of diseases, including cancer, inflammatory diseases, and various infectious diseases. Recent successes of nanoparticle vaccines during the coronavirus disease 2019 (COVID-19) pandemic have fueled enthusiasm further. In this review, the most recent developments in nanovaccines for infectious disease, cancer, inflammatory diseases, allergic diseases, and nanoadjuvants are summarized. Additionally, challenges and opportunities for clinical translation of this unique class of materials are discussed.
Collapse
Affiliation(s)
- Shamitha Shetty
- Department of Biomedical Engineering, Duke University, Durham, North Carolina, USA; , , ,
| | - Pablo Cordero Alvarado
- Department of Biomedical Engineering, Duke University, Durham, North Carolina, USA; , , ,
| | - Deleah Pettie
- Department of Biomedical Engineering, Duke University, Durham, North Carolina, USA; , , ,
| | - Joel H Collier
- Department of Biomedical Engineering, Duke University, Durham, North Carolina, USA; , , ,
| |
Collapse
|
19
|
Vilander AC, Burak J, Gilfillan D, Dean GA, Abdo Z. Host Functional Response to a Prototypic Orally Delivered Self-Replicating Vaccine Platform. Vaccines (Basel) 2024; 12:701. [PMID: 39066339 PMCID: PMC11281611 DOI: 10.3390/vaccines12070701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 06/14/2024] [Accepted: 06/17/2024] [Indexed: 07/28/2024] Open
Abstract
The development of mucosal vaccines has been limited and could be aided by a systems vaccinology approach to identify platforms and adjuvant strategies that induce protective immune responses. The induction of local immune responses by mucosal-delivered vaccines has been difficult to evaluate from peripheral samples, as systemic responses often do not correlate with the mucosal response. Here, we utilized transcriptomics in combination with Gene Set Enrichment Analysis (GSEA) to assess innate immune activation by an oral probiotic Lactobacillus acidophilus-based vaccine platform in mice. The goal was to explore the earliest immune responses elicited after oral immunization at the Peyer's patch. Twenty-four hours after oral delivery of the L. acidophilus vaccine platform, we found an abundance of L. acidophilus at Peyer's patches and detected expression of the vaccine viral proteins and adjuvants, confirming in vivo vaccine delivery. Compared to mice orally dosed with buffer or wild-type L. acidophilus, we identified enhanced responses in immune pathways related to cytokine and gene signaling, T and B cell activation, phagocytosis, and humoral responses. While more work is needed to correlate these pathways with protection from infection and/or disease, they indicate this method's potential to evaluate and aid in the iterative development of next-generation mucosal vaccines.
Collapse
Affiliation(s)
- Allison C. Vilander
- Department of Microbiology, Immunology and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO 80521, USA; (A.C.V.); (D.G.)
| | - Julia Burak
- Department of Clinical Science, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO 80521, USA;
| | - Darby Gilfillan
- Department of Microbiology, Immunology and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO 80521, USA; (A.C.V.); (D.G.)
| | - Gregg A. Dean
- Department of Microbiology, Immunology and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO 80521, USA; (A.C.V.); (D.G.)
| | - Zaid Abdo
- Department of Microbiology, Immunology and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO 80521, USA; (A.C.V.); (D.G.)
| |
Collapse
|
20
|
Wang C, Tang H, Duan Y, Zhang Q, Shan W, Wang X, Ren L. Oral biomimetic virus vaccine hydrogel for robust abscopal antitumour efficacy. J Colloid Interface Sci 2024; 674:92-107. [PMID: 38917715 DOI: 10.1016/j.jcis.2024.06.102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 06/07/2024] [Accepted: 06/11/2024] [Indexed: 06/27/2024]
Abstract
Remarkable progress has been made in tumour immunotherapy in recent decades. However, the clinical outcomes of therapeutic interventions remain unpredictable, largely because of inefficient immune responses. To address this challenge and optimise immune stimulation, we present a novel administration route for enhancing the bioavailability of immunotherapeutic drugs. Our approach involves the development of an oral tumour vaccine utilising virus-like particles derived from the Hepatitis B virus core (HBc) antigen. The external surfaces of these particles are engineered to display the model tumour antigen OVA, whereas the interiors are loaded with cytosine phosphoguanosine oligodeoxynucleotide (CpG ODN), resulting in a construct called CpG@OVAHBc with enhanced antigenicity and immune response. For oral delivery, CpG@OVAHBc is encapsulated in a crosslinked dextran hydrogel called CpG@OVAHBc@Dex. The external hydrogel shield safeguards the biomimetic virus particles from degradation by gastric acid and proteases. Upon exposure to intestinal flora, the hydrogel disintegrates, releasing CpG@OVAHBc at the intestinal mucosal site. Owing to its virus-like structure, CpG@OVAHBc exhibits enhanced adhesion to the mucosal surface, facilitating uptake by microfold cells (M cells) and subsequent transmission to antigen-presenting cells. The enzyme-triggered release of this oral hydrogel ensures the integrity of the tumour vaccine within the digestive tract, allowing targeted release and significantly improving bioavailability. Beyond its efficacy, this oral hydrogel vaccine streamlines drug administration, alleviates patient discomfort, and enhances treatment compliance without the need for specialised injection methods. Consequently, our approach expands the horizons of vaccine development in the field of oral drug administration.
Collapse
Affiliation(s)
- Chufan Wang
- Key Laboratory of Biomedical Engineering of Fujian Province University/Research Center of Biomedical Engineering of Xiamen, Department of Biomaterials, College of Materials, Xiamen University, Xiamen 361005, PR China
| | - Haobo Tang
- Key Laboratory of Biomedical Engineering of Fujian Province University/Research Center of Biomedical Engineering of Xiamen, Department of Biomaterials, College of Materials, Xiamen University, Xiamen 361005, PR China
| | - Yufei Duan
- Key Laboratory of Biomedical Engineering of Fujian Province University/Research Center of Biomedical Engineering of Xiamen, Department of Biomaterials, College of Materials, Xiamen University, Xiamen 361005, PR China
| | - Qiang Zhang
- Key Laboratory of Biomedical Engineering of Fujian Province University/Research Center of Biomedical Engineering of Xiamen, Department of Biomaterials, College of Materials, Xiamen University, Xiamen 361005, PR China
| | - Wenjun Shan
- Department of Pharmacology, College of Pharmacy, Army Medical University (Third Military Medical University), Chongqing 400038, PR China.
| | - Xiumin Wang
- School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, PR China.
| | - Lei Ren
- Key Laboratory of Biomedical Engineering of Fujian Province University/Research Center of Biomedical Engineering of Xiamen, Department of Biomaterials, College of Materials, Xiamen University, Xiamen 361005, PR China; State Key Lab of Physical Chemistry of Solid Surfaces, Xiamen University, Xiamen 361005, PR China.
| |
Collapse
|
21
|
Weng S, Li Q, Zhang T, Lin T, He Y, Yang G, Wang H, Xu Y. Enhanced Glycosylation Caused by Overexpression of Rv1002c in a Recombinant BCG Promotes Immune Response and Protects against Mycobacterium tuberculosis Infection. Vaccines (Basel) 2024; 12:622. [PMID: 38932351 PMCID: PMC11209282 DOI: 10.3390/vaccines12060622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 05/18/2024] [Accepted: 05/21/2024] [Indexed: 06/28/2024] Open
Abstract
Tuberculosis (TB) is a major global health threat despite its virtual elimination in developed countries. Issues such as drug accessibility, emergence of multidrug-resistant strains, and limitations of the current BCG vaccine highlight the urgent need for more effective TB control measures. This study constructed BCG strains overexpressing Rv1002c and found that the rBCG-Rv1002c strain secreted more glycosylated proteins, significantly enhancing macrophage activation and immune protection against Mycobacterium tuberculosis (M. tb). These results indicate that Rv1002c overexpression promotes elevated levels of O-glycosylation in BCG bacteriophages, enhancing their phagocytic and antigenic presentation functions. Moreover, rBCG-Rv1002c significantly upregulated immune regulatory molecules on the macrophage surface, activated the NF-κB pathway, and facilitated the release of large amounts of NO and H2O2, thereby enhancing bacterial control. In mice, rBCG-Rv1002c immunization induced greater innate and adaptive immune responses, including increased production of multifunctional and long-term memory T cells. Furthermore, rBCG-Rv1002c-immunized mice exhibited reduced lung bacterial load and histological damage upon M. tb infection. This result shows that it has the potential to be an excellent candidate for a preventive vaccine against TB.
Collapse
Affiliation(s)
- Shufeng Weng
- Department of Infectious Diseases, National Medical Center for Infectious Diseases, State Key Laboratory of Genetic Engineering, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, School of Life Sciences, Huashan Hospital, Fudan University, Shanghai 200437, China; (S.W.); (Q.L.); (T.Z.); (T.L.); (Y.H.); (G.Y.); (H.W.)
- Shanghai Sci-Tech Inno Center for Infection & Immunity, Shanghai 200052, China
| | - Qingchun Li
- Department of Infectious Diseases, National Medical Center for Infectious Diseases, State Key Laboratory of Genetic Engineering, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, School of Life Sciences, Huashan Hospital, Fudan University, Shanghai 200437, China; (S.W.); (Q.L.); (T.Z.); (T.L.); (Y.H.); (G.Y.); (H.W.)
| | - Tianran Zhang
- Department of Infectious Diseases, National Medical Center for Infectious Diseases, State Key Laboratory of Genetic Engineering, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, School of Life Sciences, Huashan Hospital, Fudan University, Shanghai 200437, China; (S.W.); (Q.L.); (T.Z.); (T.L.); (Y.H.); (G.Y.); (H.W.)
| | - Taiyue Lin
- Department of Infectious Diseases, National Medical Center for Infectious Diseases, State Key Laboratory of Genetic Engineering, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, School of Life Sciences, Huashan Hospital, Fudan University, Shanghai 200437, China; (S.W.); (Q.L.); (T.Z.); (T.L.); (Y.H.); (G.Y.); (H.W.)
| | - Yumo He
- Department of Infectious Diseases, National Medical Center for Infectious Diseases, State Key Laboratory of Genetic Engineering, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, School of Life Sciences, Huashan Hospital, Fudan University, Shanghai 200437, China; (S.W.); (Q.L.); (T.Z.); (T.L.); (Y.H.); (G.Y.); (H.W.)
| | - Guang Yang
- Department of Infectious Diseases, National Medical Center for Infectious Diseases, State Key Laboratory of Genetic Engineering, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, School of Life Sciences, Huashan Hospital, Fudan University, Shanghai 200437, China; (S.W.); (Q.L.); (T.Z.); (T.L.); (Y.H.); (G.Y.); (H.W.)
| | - Honghai Wang
- Department of Infectious Diseases, National Medical Center for Infectious Diseases, State Key Laboratory of Genetic Engineering, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, School of Life Sciences, Huashan Hospital, Fudan University, Shanghai 200437, China; (S.W.); (Q.L.); (T.Z.); (T.L.); (Y.H.); (G.Y.); (H.W.)
| | - Ying Xu
- Department of Infectious Diseases, National Medical Center for Infectious Diseases, State Key Laboratory of Genetic Engineering, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, School of Life Sciences, Huashan Hospital, Fudan University, Shanghai 200437, China; (S.W.); (Q.L.); (T.Z.); (T.L.); (Y.H.); (G.Y.); (H.W.)
- Shanghai Sci-Tech Inno Center for Infection & Immunity, Shanghai 200052, China
| |
Collapse
|
22
|
Stylianou E, Satti I. Inhaled aerosol viral-vectored vaccines against tuberculosis. Curr Opin Virol 2024; 66:101408. [PMID: 38574628 DOI: 10.1016/j.coviro.2024.101408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 03/08/2024] [Accepted: 03/08/2024] [Indexed: 04/06/2024]
Abstract
Bacille Calmette-Guérin (BCG) remains the sole licensed vaccine against tuberculosis (TB), despite its variable efficacy in protecting against pulmonary TB. The development of effective TB vaccines faces significant challenges, marked by the absence of validated correlates of protection and predictive animal models. Strategic approaches to enhance TB vaccines and augment BCG efficacy include utilising prime-boost strategies with viral-vectored vaccines and exploring innovative delivery techniques, such as mucosal vaccine administration. Viral vectors offer numerous advantages, including the capacity to accommodate genes encoding extensive antigenic fragments and the induction of robust immune responses. Aerosol delivery aligns with the route of Mycobacterium tuberculosis infection and holds the potential to enhance protective mucosal immunity. Aerosolised viral-vectored vaccines overcome anti-vector immunity, facilitating repeated aerosol deliveries.
Collapse
Affiliation(s)
- Elena Stylianou
- The Jenner Institute, Old Road Roosevelt Drive, Oxford OX3 7DQ, UK.
| | - Iman Satti
- The Jenner Institute, Old Road Roosevelt Drive, Oxford OX3 7DQ, UK.
| |
Collapse
|
23
|
Li W, Li Y, Li J, Meng J, Jiang Z, Yang C, Wen Y, Liu S, Cheng X, Mi S, zhao Y, Miao L, Lu X. All-Trans-Retinoic Acid-Adjuvanted mRNA Vaccine Induces Mucosal Anti-Tumor Immune Responses for Treating Colorectal Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2309770. [PMID: 38528670 PMCID: PMC11165559 DOI: 10.1002/advs.202309770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Indexed: 03/27/2024]
Abstract
Messenger RNA (mRNA) cancer vaccines are a new class of immunotherapies that can activate the immune system to recognize and destroy cancer cells. However, their effectiveness in treating colorectal cancer located on the mucosal surface of the gut is limited due to the insufficient activation of mucosal immune response and inadequate infiltration of cytotoxic T cells into tumors. To address this issue, a new mRNA cancer vaccine is developed that can stimulate mucosal immune responses in the gut by co-delivering all-trans-retinoic acid (ATRA) and mRNA using lipid nanoparticle (LNP). The incorporation of ATRA has not only improved the mRNA transfection efficiency of LNP but also induced high expression of gut-homing receptors on vaccine-activated T cells. Additionally, the use of LNP improves the aqueous solubility of ATRA, eliminating the need for toxic solvents to administer ATRA. Upon intramuscular injections, ATRA-adjuvanted mRNA-LNP significantly increase the infiltration of antigen-specific, cytotoxic T cells in the lamina propria of the intestine, mesenteric lymph nodes, and orthotopic colorectal tumors, resulting in significantly improved tumor inhibition and prolonged animal survival compared to conventional mRNA-LNP without ATRA. Overall, this study provides a promising approach for improving the therapeutic efficacy of mRNA cancer vaccines against colorectal cancer.
Collapse
Affiliation(s)
- Wei Li
- Beijing National Laboratory for Molecular SciencesCAS Key Laboratory of ColloidInterface and Chemical ThermodynamicsInstitute of ChemistryChinese Academy of SciencesBeijing100190China
- University of Chinese Academy of SciencesBeijing100049China
| | - Yijia Li
- State Key Laboratory of Natural and Biomimetic DrugsSchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery SystemSchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
| | - Jingjiao Li
- Beijing National Laboratory for Molecular SciencesCAS Key Laboratory of ColloidInterface and Chemical ThermodynamicsInstitute of ChemistryChinese Academy of SciencesBeijing100190China
- University of Chinese Academy of SciencesBeijing100049China
| | - Junli Meng
- Beijing National Laboratory for Molecular SciencesCAS Key Laboratory of ColloidInterface and Chemical ThermodynamicsInstitute of ChemistryChinese Academy of SciencesBeijing100190China
- University of Chinese Academy of SciencesBeijing100049China
| | - Ziqiong Jiang
- State Key Laboratory of Natural and Biomimetic DrugsSchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery SystemSchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
| | - Chen Yang
- Beijing National Laboratory for Molecular SciencesCAS Key Laboratory of ColloidInterface and Chemical ThermodynamicsInstitute of ChemistryChinese Academy of SciencesBeijing100190China
- University of Chinese Academy of SciencesBeijing100049China
| | - Yixing Wen
- Beijing National Laboratory for Molecular SciencesCAS Key Laboratory of ColloidInterface and Chemical ThermodynamicsInstitute of ChemistryChinese Academy of SciencesBeijing100190China
- University of Chinese Academy of SciencesBeijing100049China
| | - Shuai Liu
- Beijing National Laboratory for Molecular SciencesCAS Key Laboratory of ColloidInterface and Chemical ThermodynamicsInstitute of ChemistryChinese Academy of SciencesBeijing100190China
| | - Xingdi Cheng
- Beijing National Laboratory for Molecular SciencesCAS Key Laboratory of ColloidInterface and Chemical ThermodynamicsInstitute of ChemistryChinese Academy of SciencesBeijing100190China
| | - Shiwei Mi
- Beijing National Laboratory for Molecular SciencesCAS Key Laboratory of ColloidInterface and Chemical ThermodynamicsInstitute of ChemistryChinese Academy of SciencesBeijing100190China
- University of Chinese Academy of SciencesBeijing100049China
| | - Yuanyuan zhao
- Beijing National Laboratory for Molecular SciencesCAS Key Laboratory of ColloidInterface and Chemical ThermodynamicsInstitute of ChemistryChinese Academy of SciencesBeijing100190China
- University of Chinese Academy of SciencesBeijing100049China
| | - Lei Miao
- State Key Laboratory of Natural and Biomimetic DrugsSchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery SystemSchool of Pharmaceutical SciencesPeking UniversityBeijing100191China
| | - Xueguang Lu
- Beijing National Laboratory for Molecular SciencesCAS Key Laboratory of ColloidInterface and Chemical ThermodynamicsInstitute of ChemistryChinese Academy of SciencesBeijing100190China
- University of Chinese Academy of SciencesBeijing100049China
| |
Collapse
|
24
|
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
|
25
|
Wang J, Wang H, Zhang D, Liu F, Li X, Gao M, Cheng M, Bao H, Zhan J, Zeng Y, Wang C, Cao X. Lactiplantibacillus plantarum surface-displayed VP6 (PoRV) protein can prevent PoRV infection in piglets. Int Immunopharmacol 2024; 133:112079. [PMID: 38615376 DOI: 10.1016/j.intimp.2024.112079] [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: 03/04/2024] [Revised: 04/09/2024] [Accepted: 04/09/2024] [Indexed: 04/16/2024]
Abstract
Porcine rotavirus (PoRV) poses a threat to the development of animal husbandry and human health, leading to substantial economic losses. VP6 protein is the most abundant component in virus particles and also the core structural protein of the virus. Firstly, this study developed an antibiotic-resistance-free, environmentally friendly expression vector, named asd-araC-PBAD-alr (AAPA). Then Recombinant Lactiplantibacillus plantarum (L. plantarum) strains induced by arabinose to express VP6 and VP6-pFc fusion proteins was constructed. Subsequently, This paper discovered that NC8/Δalr-pCXa-VP6-S and NC8/Δalr-pCXa-VP6-pFc-S could enhance host immunity and prevent rotavirus infection in neonatal mice and piglets. The novel recombinant L. plantarum strains constructed in this study can serve as oral vaccines to boost host immunity, offering a new strategy to prevent PoRV infection.
Collapse
Affiliation(s)
- Junhong Wang
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, 130118, China
| | - Haixu Wang
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, 130118, China
| | - Dongliang Zhang
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, 130118, China
| | - Fangyuan Liu
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, 130118, China
| | - Xiaoxu Li
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, 130118, China
| | - Ming Gao
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, 130118, China
| | - Mingyang Cheng
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, 130118, China
| | - Hongyu Bao
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, 130118, China
| | - Jiaxing Zhan
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, 130118, China
| | - Yan Zeng
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, 130118, China.
| | - Chunfeng Wang
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, 130118, China.
| | - Xin Cao
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, 130118, China.
| |
Collapse
|
26
|
Sheng S, Zhang H, Li X, Chen J, Wang P, Liang Y, Li C, Li H, Pan N, Bao X, Liu M, Zhao L, Li X, Guan P, Wang X. Probiotic-derived amphiphilic exopolysaccharide self-assembling adjuvant delivery platform for enhancing immune responses. J Nanobiotechnology 2024; 22:267. [PMID: 38764014 PMCID: PMC11103965 DOI: 10.1186/s12951-024-02528-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 05/03/2024] [Indexed: 05/21/2024] Open
Abstract
Enhancing immune response activation through the synergy of effective antigen delivery and immune enhancement using natural, biodegradable materials with immune-adjuvant capabilities is challenging. Here, we present NAPSL.p that can activate the Toll-like receptor 4 (TLR4) pathway, an amphiphilic exopolysaccharide, as a potential self-assembly adjuvant delivery platform. Its molecular structure and unique properties exhibited remarkable self-assembly, forming a homogeneous nanovaccine with ovalbumin (OVA) as the model antigen. When used as an adjuvant, NAPSL.p significantly increased OVA uptake by dendritic cells. In vivo imaging revealed prolonged pharmacokinetics of NAPSL. p-delivered OVA compared to OVA alone. Notably, NAPSL.p induced elevated levels of specific serum IgG and isotype titers, enhancing rejection of B16-OVA melanoma xenografts in vaccinated mice. Additionally, NAPSL.p formulation improved therapeutic effects, inhibiting tumor growth, and increasing animal survival rates. The nanovaccine elicited CD4+ and CD8+ T cell-based immune responses, demonstrating the potential for melanoma prevention. Furthermore, NAPSL.p-based vaccination showed stronger protective effects against influenza compared to Al (OH)3 adjuvant. Our findings suggest NAPSL.p as a promising, natural self-adjuvanting delivery platform to enhance vaccine design across applications.
Collapse
Affiliation(s)
- Shouxin Sheng
- State Key Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, School of Life Science, Inner Mongolia University, Hohhot, 010021, P.R. China
| | - Haochi Zhang
- State Key Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, School of Life Science, Inner Mongolia University, Hohhot, 010021, P.R. China
| | - Xinyu Li
- State Key Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, School of Life Science, Inner Mongolia University, Hohhot, 010021, P.R. China
| | - Jian Chen
- State Key Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, School of Life Science, Inner Mongolia University, Hohhot, 010021, P.R. China
- JinYuBaoLing Biopharmaceutical Co. Ltd, Inner Mongolia, 010000, Hohhot, P.R. China
| | - Pu Wang
- State Key Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, School of Life Science, Inner Mongolia University, Hohhot, 010021, P.R. China
| | - Yanchen Liang
- State Key Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, School of Life Science, Inner Mongolia University, Hohhot, 010021, P.R. China
| | - Chunhe Li
- State Key Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, School of Life Science, Inner Mongolia University, Hohhot, 010021, P.R. China
| | - Haotian Li
- State Key Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, School of Life Science, Inner Mongolia University, Hohhot, 010021, P.R. China
| | - Na Pan
- State Key Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, School of Life Science, Inner Mongolia University, Hohhot, 010021, P.R. China
| | - Xuemei Bao
- State Key Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, School of Life Science, Inner Mongolia University, Hohhot, 010021, P.R. China
| | - Mengnan Liu
- State Key Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, School of Life Science, Inner Mongolia University, Hohhot, 010021, P.R. China
| | - Lixia Zhao
- JinYuBaoLing Biopharmaceutical Co. Ltd, Inner Mongolia, 010000, Hohhot, P.R. China
| | - Xiaoyan Li
- JinYuBaoLing Biopharmaceutical Co. Ltd, Inner Mongolia, 010000, Hohhot, P.R. China
| | - Pingyuan Guan
- JinYuBaoLing Biopharmaceutical Co. Ltd, Inner Mongolia, 010000, Hohhot, P.R. China
| | - Xiao Wang
- State Key Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, School of Life Science, Inner Mongolia University, Hohhot, 010021, P.R. China.
| |
Collapse
|
27
|
Amimo JO, Michael H, Chepngeno J, Jung K, Raev SA, Paim FC, Lee MV, Damtie D, Vlasova AN, Saif LJ. Maternal immunization and vitamin A sufficiency impact sow primary adaptive immunity and passive protection to nursing piglets against porcine epidemic diarrhea virus infection. Front Immunol 2024; 15:1397118. [PMID: 38812505 PMCID: PMC11133611 DOI: 10.3389/fimmu.2024.1397118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 04/24/2024] [Indexed: 05/31/2024] Open
Abstract
Porcine epidemic diarrhea virus (PEDV) causes a highly contagious enteric disease with major economic losses to swine production worldwide. Due to the immaturity of the neonatal piglet immune system and given the high virulence of PEDV, improving passive lactogenic immunity is the best approach to protect suckling piglets against the lethal infection. We tested whether oral vitamin A (VA) supplementation and PEDV exposure of gestating and lactating VA-deficient (VAD) sows would enhance their primary immune responses and boost passive lactogenic protection against the PEDV challenge of their piglets. We demonstrated that PEDV inoculation of pregnant VAD sows in the third trimester provided higher levels of lactogenic protection of piglets as demonstrated by >87% survival rates of their litters compared with <10% in mock litters and that VA supplementation to VAD sows further improved the piglets' survival rates to >98%. We observed significantly elevated PEDV IgA and IgG antibody (Ab) titers and Ab-secreting cells (ASCs) in VA-sufficient (VAS)+PEDV and VAD+VA+PEDV sows, with the latter maintaining higher Ab titers in blood prior to parturition and in blood and milk throughout lactation. The litters of VAD+VA+PEDV sows also had the highest serum PEDV-neutralizing Ab titers at piglet post-challenge days (PCD) 0 and 7, coinciding with higher PEDV IgA ASCs and Ab titers in the blood and milk of their sows, suggesting an immunomodulatory role of VA in sows. Thus, sows that delivered sufficient lactogenic immunity to their piglets provided the highest passive protection against the PEDV challenge. Maternal immunization during pregnancy (± VA) and VA sufficiency enhanced the sow primary immune responses, expression of gut-mammary gland trafficking molecules, and passive protection of their offspring. Our findings are relevant to understanding the role of VA in the Ab responses to oral attenuated vaccines that are critical for successful maternal vaccination programs against enteric infections in infants and young animals.
Collapse
Affiliation(s)
- Joshua O. Amimo
- Center for Food Animal Health, Department of Animal Sciences, College of Food, Agricultural and Environmental Sciences, Ohio Agricultural Research and Development Center (OARDC), The Ohio State University, Wooster, OH, United States
- Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, OH, United States
- Department of Animal Production, Faculty of Veterinary Medicine, University of Nairobi, Nairobi, Kenya
| | - Husheem Michael
- Center for Food Animal Health, Department of Animal Sciences, College of Food, Agricultural and Environmental Sciences, Ohio Agricultural Research and Development Center (OARDC), The Ohio State University, Wooster, OH, United States
- Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, OH, United States
| | - Juliet Chepngeno
- Center for Food Animal Health, Department of Animal Sciences, College of Food, Agricultural and Environmental Sciences, Ohio Agricultural Research and Development Center (OARDC), The Ohio State University, Wooster, OH, United States
- Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, OH, United States
| | - Kwonil Jung
- Center for Food Animal Health, Department of Animal Sciences, College of Food, Agricultural and Environmental Sciences, Ohio Agricultural Research and Development Center (OARDC), The Ohio State University, Wooster, OH, United States
- Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, OH, United States
| | - Sergei A. Raev
- Center for Food Animal Health, Department of Animal Sciences, College of Food, Agricultural and Environmental Sciences, Ohio Agricultural Research and Development Center (OARDC), The Ohio State University, Wooster, OH, United States
- Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, OH, United States
| | - Francine C. Paim
- Center for Food Animal Health, Department of Animal Sciences, College of Food, Agricultural and Environmental Sciences, Ohio Agricultural Research and Development Center (OARDC), The Ohio State University, Wooster, OH, United States
- Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, OH, United States
| | - Marcia V. Lee
- Center for Food Animal Health, Department of Animal Sciences, College of Food, Agricultural and Environmental Sciences, Ohio Agricultural Research and Development Center (OARDC), The Ohio State University, Wooster, OH, United States
- Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, OH, United States
| | - Debasu Damtie
- Department of Immunology and Molecular Biology, School of Biomedical and Laboratory Sciences, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Anastasia N. Vlasova
- Center for Food Animal Health, Department of Animal Sciences, College of Food, Agricultural and Environmental Sciences, Ohio Agricultural Research and Development Center (OARDC), The Ohio State University, Wooster, OH, United States
- Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, OH, United States
| | - Linda J. Saif
- Center for Food Animal Health, Department of Animal Sciences, College of Food, Agricultural and Environmental Sciences, Ohio Agricultural Research and Development Center (OARDC), The Ohio State University, Wooster, OH, United States
- Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
28
|
Xing Y, Clark JR, Chang JD, Zulk JJ, Chirman DM, Piedra FA, Vaughan EE, Hernandez Santos HJ, Patras KA, Maresso AW. Progress toward a vaccine for extraintestinal pathogenic E. coli (ExPEC) II: efficacy of a toxin-autotransporter dual antigen approach. Infect Immun 2024; 92:e0044023. [PMID: 38591882 PMCID: PMC11075464 DOI: 10.1128/iai.00440-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 03/18/2024] [Indexed: 04/10/2024] Open
Abstract
Extraintestinal pathogenic Escherichia coli (ExPEC) is a leading cause of worldwide morbidity and mortality, the top cause of antimicrobial-resistant (AMR) infections, and the most frequent cause of life-threatening sepsis and urinary tract infections (UTI) in adults. The development of an effective and universal vaccine is complicated by this pathogen's pan-genome, its ability to mix and match virulence factors and AMR genes via horizontal gene transfer, an inability to decipher commensal from pathogens, and its intimate association and co-evolution with mammals. Using a pan virulome analysis of >20,000 sequenced E. coli strains, we identified the secreted cytolysin α-hemolysin (HlyA) as a high priority target for vaccine exploration studies. We demonstrate that a catalytically inactive pure form of HlyA, expressed in an autologous host using its own secretion system, is highly immunogenic in a murine host, protects against several forms of ExPEC infection (including lethal bacteremia), and significantly lowers bacterial burdens in multiple organ systems. Interestingly, the combination of a previously reported autotransporter (SinH) with HlyA was notably effective, inducing near complete protection against lethal challenge, including commonly used infection strains ST73 (CFT073) and ST95 (UTI89), as well as a mixture of 10 of the most highly virulent sequence types and strains from our clinical collection. Both HlyA and HlyA-SinH combinations also afforded some protection against UTI89 colonization in a murine UTI model. These findings suggest recombinant, inactive hemolysin and/or its combination with SinH warrant investigation in the development of an E. coli vaccine against invasive disease.
Collapse
Affiliation(s)
- Yikun Xing
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
- TAILOR Labs, Vaccine Development Group, Baylor College of Medicine, Houston, Texas, USA
| | - Justin R. Clark
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
- TAILOR Labs, Vaccine Development Group, Baylor College of Medicine, Houston, Texas, USA
| | - James D. Chang
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
- TAILOR Labs, Vaccine Development Group, Baylor College of Medicine, Houston, Texas, USA
| | - Jacob J. Zulk
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
- TAILOR Labs, Vaccine Development Group, Baylor College of Medicine, Houston, Texas, USA
| | - Dylan M. Chirman
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
- TAILOR Labs, Vaccine Development Group, Baylor College of Medicine, Houston, Texas, USA
| | - Felipe-Andres Piedra
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | - Ellen E. Vaughan
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | - Haroldo J. Hernandez Santos
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
- TAILOR Labs, Vaccine Development Group, Baylor College of Medicine, Houston, Texas, USA
| | - Kathryn A. Patras
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
- Alkek Center for Metagenomics and Microbiome Research, Baylor College of Medicine, Houston, Texas, USA
| | - Anthony W. Maresso
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
- TAILOR Labs, Vaccine Development Group, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
29
|
Dowaidar M. Uptake pathways of cell-penetrating peptides in the context of drug delivery, gene therapy, and vaccine development. Cell Signal 2024; 117:111116. [PMID: 38408550 DOI: 10.1016/j.cellsig.2024.111116] [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: 01/03/2024] [Revised: 02/19/2024] [Accepted: 02/21/2024] [Indexed: 02/28/2024]
Abstract
Cell-penetrating peptides have been extensively utilized for the purpose of facilitating the intracellular delivery of cargo that is impermeable to the cell membrane. The researchers have exhibited proficient delivery capabilities for oligonucleotides, thereby establishing cell-penetrating peptides as a potent instrument in the field of gene therapy. Furthermore, they have demonstrated a high level of efficiency in delivering several additional payloads. Cell penetrating peptides (CPPs) possess the capability to efficiently transport therapeutic molecules to specific cells, hence offering potential remedies for many illnesses. Hence, their utilization is imperative for the improvement of therapeutic vaccines. In contemporary studies, a plethora of cell-penetrating peptides have been unveiled, each characterized by its own distinct structural attributes and associated mechanisms. Although it is widely acknowledged that there are multiple pathways through which particles might be internalized, a comprehensive understanding of the specific mechanisms by which these particles enter cells has to be fully elucidated. The absorption of cell-penetrating peptides can occur through either direct translocation or endocytosis. However, it is worth noting that categories of cell-penetrating peptides are not commonly linked to specific entrance mechanisms. Furthermore, research has demonstrated that cell-penetrating peptides (CPPs) possess the capacity to enhance antigen uptake by cells and facilitate the traversal of various biological barriers. The primary objective of this work is to examine the mechanisms by which cell-penetrating peptides are internalized by cells and their significance in facilitating the administration of drugs, particularly in the context of gene therapy and vaccine development. The current study investigates the immunostimulatory properties of numerous vaccine components administered using different cell-penetrating peptides (CPPs). This study encompassed a comprehensive discussion on various topics, including the uptake pathways and mechanisms of cell-penetrating peptides (CPPs), the utilization of CPPs as innovative vectors for gene therapy, the role of CPPs in vaccine development, and the potential of CPPs for antigen delivery in the context of vaccine development.
Collapse
Affiliation(s)
- Moataz Dowaidar
- Bioengineering Department, King Fahd University of Petroleum and Minerals (KFUPM), Dhahran 31261, Saudi Arabia; Interdisciplinary Research Center for Hydrogen Technologies and Carbon Management, King Fahd University of Petroleum and Minerals (KFUPM), Dhahran 31261, Saudi Arabia; Biosystems and Machines Research Center, King Fahd University of Petroleum and Minerals (KFUPM), Dhahran 31261, Saudi Arabia.
| |
Collapse
|
30
|
Ma B, Tao M, Li Z, Zheng Q, Wu H, Chen P. Mucosal vaccines for viral diseases: Status and prospects. Virology 2024; 593:110026. [PMID: 38373360 DOI: 10.1016/j.virol.2024.110026] [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: 09/19/2023] [Revised: 02/08/2024] [Accepted: 02/12/2024] [Indexed: 02/21/2024]
Abstract
Virus-associated infectious diseases are highly detrimental to human health and animal husbandry. Among all countermeasures against infectious diseases, prophylactic vaccines, which developed through traditional or novel approaches, offer potential benefits. More recently, mucosal vaccines attract attention for their extraordinary characteristics compared to conventional parenteral vaccines, particularly for mucosal-related pathogens. Representatively, coronavirus disease 2019 (COVID-19), a respiratory disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), further accelerated the research and development efforts for mucosal vaccines by thoroughly investigating existing strategies or involving novel techniques. While several vaccine candidates achieved positive progresses, thus far, part of the current COVID-19 mucosal vaccines have shown poor performance, which underline the need for next-generation mucosal vaccines and corresponding platforms. In this review, we summarized the typical mucosal vaccines approved for humans or animals and sought to elucidate the underlying mechanisms of these successful cases. In addition, mucosal vaccines against COVID-19 that are in human clinical trials were reviewed in detail since this public health event mobilized all advanced technologies for possible solutions. Finally, the gaps in developing mucosal vaccines, potential solutions and prospects were discussed. Overall, rational application of mucosal vaccines would facilitate the establishing of mucosal immunity and block the transmission of viral diseases.
Collapse
Affiliation(s)
- Bingjie Ma
- College of Animal Science and Technology, Xinyang Agriculture and Forestry University, Xinyang, China
| | - Mengxiao Tao
- College of Animal Science and Technology, Xinyang Agriculture and Forestry University, Xinyang, China
| | - Zhili Li
- College of Animal Science and Technology, Xinyang Agriculture and Forestry University, Xinyang, China
| | - Quanfang Zheng
- College of Animal Science and Technology, Xinyang Agriculture and Forestry University, Xinyang, China
| | - Haigang Wu
- College of Animal Science and Technology, Xinyang Agriculture and Forestry University, Xinyang, China
| | - Peirong Chen
- College of Animal Science and Technology, Xinyang Agriculture and Forestry University, Xinyang, China.
| |
Collapse
|
31
|
Eberlein V, Rosencrantz S, Finkensieper J, Besecke JK, Mansuroglu Y, Kamp JC, Lange F, Dressman J, Schopf S, Hesse C, Thoma M, Fertey J, Ulbert S, Grunwald T. Mucosal immunization with a low-energy electron inactivated respiratory syncytial virus vaccine protects mice without Th2 immune bias. Front Immunol 2024; 15:1382318. [PMID: 38646538 PMCID: PMC11026718 DOI: 10.3389/fimmu.2024.1382318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 03/18/2024] [Indexed: 04/23/2024] Open
Abstract
The respiratory syncytial virus (RSV) is a leading cause of acute lower respiratory tract infections associated with numerous hospitalizations. Recently, intramuscular (i.m.) vaccines against RSV have been approved for elderly and pregnant women. Noninvasive mucosal vaccination, e.g., by inhalation, offers an alternative against respiratory pathogens like RSV. Effective mucosal vaccines induce local immune responses, potentially resulting in the efficient and fast elimination of respiratory viruses after natural infection. To investigate this immune response to an RSV challenge, low-energy electron inactivated RSV (LEEI-RSV) was formulated with phosphatidylcholine-liposomes (PC-LEEI-RSV) or 1,2-dioleoyl-3-trimethylammonium-propane and 1,2-dioleoyl-sn-glycero-3-phosphoethanolamine (DD-LEEI-RSV) for vaccination of mice intranasally. As controls, LEEI-RSV and formalin-inactivated-RSV (FI-RSV) were used via i.m. vaccination. The RSV-specific immunogenicity of the different vaccines and their protective efficacy were analyzed. RSV-specific IgA antibodies and a statistically significant reduction in viral load upon challenge were detected in mucosal DD-LEEI-RSV-vaccinated animals. Alhydrogel-adjuvanted LEEI-RSV i.m. showed a Th2-bias with enhanced IgE, eosinophils, and lung histopathology comparable to FI-RSV. These effects were absent when applying the mucosal vaccines highlighting the potential of DD-LEEI-RSV as an RSV vaccine candidate and the improved performance of this mucosal vaccine candidate.
Collapse
Affiliation(s)
- Valentina Eberlein
- Fraunhofer Institute for Cell Therapy and Immunology IZI, Leipzig, Germany
- Fraunhofer Cluster of Excellence Immune-Mediated Diseases (CIMD), Frankfurt am Main, Germany
| | - Sophia Rosencrantz
- Fraunhofer Cluster of Excellence Immune-Mediated Diseases (CIMD), Frankfurt am Main, Germany
- Fraunhofer Institute for Applied Polymer Research (IAP), Potsdam, Germany
| | - Julia Finkensieper
- Fraunhofer Institute for Cell Therapy and Immunology IZI, Leipzig, Germany
- Fraunhofer Cluster of Excellence Immune-Mediated Diseases (CIMD), Frankfurt am Main, Germany
| | - Joana Kira Besecke
- Fraunhofer Cluster of Excellence Immune-Mediated Diseases (CIMD), Frankfurt am Main, Germany
- Fraunhofer Institute for Organic Electronics, Electron Beam and Plasma Technology (FEP), Dresden, Germany
| | - Yaser Mansuroglu
- Fraunhofer Cluster of Excellence Immune-Mediated Diseases (CIMD), Frankfurt am Main, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Frankfurt, Germany
| | - Jan-Christopher Kamp
- Department of Respiratory Medicine and Infectious Diseases, Hannover Medical School, Hannover, Germany
- Member of the German Center for Lung Research (DZL), Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Hannover, Germany
| | - Franziska Lange
- Fraunhofer Institute for Cell Therapy and Immunology IZI, Leipzig, Germany
- Fraunhofer Cluster of Excellence Immune-Mediated Diseases (CIMD), Frankfurt am Main, Germany
| | - Jennifer Dressman
- Fraunhofer Cluster of Excellence Immune-Mediated Diseases (CIMD), Frankfurt am Main, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Frankfurt, Germany
| | - Simone Schopf
- Fraunhofer Cluster of Excellence Immune-Mediated Diseases (CIMD), Frankfurt am Main, Germany
- Fraunhofer Institute for Organic Electronics, Electron Beam and Plasma Technology (FEP), Dresden, Germany
| | - Christina Hesse
- Fraunhofer Cluster of Excellence Immune-Mediated Diseases (CIMD), Frankfurt am Main, Germany
- Member of the German Center for Lung Research (DZL), Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Hannover, Germany
- Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM), Hannover, Germany
| | - Martin Thoma
- Fraunhofer Cluster of Excellence Immune-Mediated Diseases (CIMD), Frankfurt am Main, Germany
- Fraunhofer Institute for Manufacturing Engineering and Automation (IPA), Stuttgart, Germany
| | - Jasmin Fertey
- Fraunhofer Institute for Cell Therapy and Immunology IZI, Leipzig, Germany
- Fraunhofer Cluster of Excellence Immune-Mediated Diseases (CIMD), Frankfurt am Main, Germany
| | - Sebastian Ulbert
- Fraunhofer Institute for Cell Therapy and Immunology IZI, Leipzig, Germany
- Fraunhofer Cluster of Excellence Immune-Mediated Diseases (CIMD), Frankfurt am Main, Germany
| | - Thomas Grunwald
- Fraunhofer Institute for Cell Therapy and Immunology IZI, Leipzig, Germany
- Fraunhofer Cluster of Excellence Immune-Mediated Diseases (CIMD), Frankfurt am Main, Germany
| |
Collapse
|
32
|
Airola C, Andaloro S, Gasbarrini A, Ponziani FR. Vaccine Responses in Patients with Liver Cirrhosis: From the Immune System to the Gut Microbiota. Vaccines (Basel) 2024; 12:349. [PMID: 38675732 PMCID: PMC11054513 DOI: 10.3390/vaccines12040349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 03/11/2024] [Accepted: 03/21/2024] [Indexed: 04/28/2024] Open
Abstract
Vaccines prevent a significant number of deaths annually. However, certain populations do not respond adequately to vaccination due to impaired immune systems. Cirrhosis, a condition marked by a profound disruption of immunity, impairs the normal immunization process. Critical vaccines for cirrhotic patients, such as the hepatitis A virus (HAV), hepatitis B virus (HBV), influenza, pneumococcal, and coronavirus disease 19 (COVID-19), often elicit suboptimal responses in these individuals. The humoral response, essential for immunization, is less effective in cirrhosis due to a decline in B memory cells and an increase in plasma blasts, which interfere with the creation of a long-lasting response to antigen vaccination. Additionally, some T cell subtypes exhibit reduced activation in cirrhosis. Nonetheless, the persistence of memory T cell activity, while not preventing infections, may help to attenuate the severity of diseases in these patients. Alongside that, the impairment of innate immunity, particularly in dendritic cells (DCs), prevents the normal priming of adaptive immunity, interrupting the immunization process at its onset. Furthermore, cirrhosis disrupts the gut-liver axis balance, causing dysbiosis, reduced production of short-chain fatty acids (SCFAs), increased intestinal permeability, and bacterial translocation. Undermining the physiological activity of the immune system, these alterations could impact the vaccine response. Enhancing the understanding of the molecular and cellular factors contributing to impaired vaccination responses in cirrhotic patients is crucial for improving vaccine efficacy in this population and developing better prevention strategies.
Collapse
Affiliation(s)
- Carlo Airola
- Liver Unit, CEMAD Centro Malattie dell’Apparato Digerente, Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (C.A.); (S.A.); (A.G.)
| | - Silvia Andaloro
- Liver Unit, CEMAD Centro Malattie dell’Apparato Digerente, Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (C.A.); (S.A.); (A.G.)
| | - Antonio Gasbarrini
- Liver Unit, CEMAD Centro Malattie dell’Apparato Digerente, Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (C.A.); (S.A.); (A.G.)
- Department of Translational Medicine and Surgery, Catholic University, 00168 Rome, Italy
| | - Francesca Romana Ponziani
- Liver Unit, CEMAD Centro Malattie dell’Apparato Digerente, Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (C.A.); (S.A.); (A.G.)
- Department of Translational Medicine and Surgery, Catholic University, 00168 Rome, Italy
| |
Collapse
|
33
|
Nie L, Huang Y, Cheng Z, Luo H, Zhan Y, Dou K, Ma C, Yu C, Luo C, Liu Z, Liu S, Zhu Y. An intranasal influenza virus vector vaccine protects against Helicobacter pylori in mice. J Virol 2024; 98:e0192323. [PMID: 38358289 PMCID: PMC10949480 DOI: 10.1128/jvi.01923-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Accepted: 01/22/2024] [Indexed: 02/16/2024] Open
Abstract
Helicobacter pylori is a human pathogen that infects almost half of the population. Antibiotic resistance in H. pylori threatens health and increases the demand for prophylactic and therapeutic vaccines. Traditional oral vaccine research faces considerable challenges because of the epithelial barrier, potential enterotoxicity of adjuvants, and the challenging conditions of the gastric environment. We developed an intranasal influenza A virus (IAV) vector vaccine based on two live attenuated influenza viruses with modified acidic polymerase protein (PA) genes encoding the A subunit of H. pylori neutrophil-activating protein (NapA), named IAV-NapA, including influenza virus A/WSN/33 (WSN)-NapA and A/Puerto Rico/8/34 (PR8)-NapA. These recombinant influenza viruses were highly attenuated and exhibited strong immunogenicity in mice. Vaccination with IAV-NapA induced antigen-specific humoral and mucosal immune responses while stimulating robust Th1 and Th17 cell immune responses in mice. Our findings suggest that prophylactic and therapeutic vaccination with influenza virus vector vaccines significantly reduces colonization of H. pylori and inflammation in the stomach of mice.IMPORTANCEHelicobacter pylori is the most common cause of chronic gastritis and leads to severe gastroduodenal pathology in some patients. Many studies have shown that Th1 and Th17 cellular and gastric mucosal immune responses are critical in reducing H. pylori load. IAV vector vaccines can stimulate these immune responses while overcoming potential adjuvant toxicity and antigen dosing issues. To date, no studies have demonstrated the role of live attenuated IAV vector vaccines in preventing and treating H. pylori infection. Our work indicates that vaccination with IAV-NapA induces antigen-specific humoral, cellular, and mucosal immunity, producing a protective and therapeutic effect against H. pylori infection in BALB/c mice. This undescribed H. pylori vaccination approach may provide valuable information for developing vaccines against H. pylori infection.
Collapse
Affiliation(s)
- Longyu Nie
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China
| | - Yu Huang
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China
| | - Zhikui Cheng
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China
| | - Hao Luo
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China
| | - Yuxin Zhan
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China
| | - Kaiwen Dou
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China
| | - Caijiao Ma
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China
| | - Chen Yu
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China
| | - Chuanjin Luo
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China
| | - Zhiqiang Liu
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China
| | - Shi Liu
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China
| | - Ying Zhu
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China
| |
Collapse
|
34
|
Sinha D, Yaugel-Novoa M, Waeckel L, Paul S, Longet S. Unmasking the potential of secretory IgA and its pivotal role in protection from respiratory viruses. Antiviral Res 2024; 223:105823. [PMID: 38331200 DOI: 10.1016/j.antiviral.2024.105823] [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: 12/11/2023] [Revised: 01/22/2024] [Accepted: 01/25/2024] [Indexed: 02/10/2024]
Abstract
Mucosal immunity has regained its spotlight amidst the ongoing Coronavirus disease 19 (COVID-19) pandemic, with numerous studies highlighting the crucial role of mucosal secretory IgA (SIgA) in protection against Severe acute respiratory syndrome coronavirus-2 or SARS-CoV-2 infections. The observed limitations in the efficacy of currently authorized COVID-19 vaccines in inducing effective mucosal immune responses remind us of the limitations of systemic vaccination in promoting protective mucosal immunity. This resurgence of interest has motivated the development of vaccine platforms capable of enhancing mucosal responses, specifically the SIgA response, and the development of IgA-based therapeutics. Recognizing viral respiratory infections as a global threat, we would like to comprehensively review the existing knowledge on mucosal immunity, with a particular emphasis on SIgA, in the context of SARS-CoV-2, influenza, and Respiratory Syncytial Virus (RSV) infections. This review aims to describe the structural and functional specificities of SIgA, along with its nuanced role in combating influenza, RSV, and SARS-CoV-2 infections. Subsequent sections further elaborate promising vaccine strategies, including mucosal vaccines against Influenza, RSV, and SARS-CoV-2 respiratory viruses, currently undergoing preclinical and clinical development. Additionally, we address the challenges associated with mucosal vaccine development, concluding with a discussion on IgA-based therapeutics as a promising platform for the treatment of viral respiratory infections. This comprehensive review not only synthesizes current insights into mucosal immunity but also identifies critical knowledge gaps, strengthening the way for further advancements in our current understanding and approaches to combat respiratory viral threats.
Collapse
Affiliation(s)
- Divya Sinha
- CIRI - Centre International de Recherche en Infectiologie, Team GIMAP, Univ Lyon, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR5308, CIC 1408 Vaccinology, F42023, Saint-Etienne, France
| | - Melyssa Yaugel-Novoa
- CIRI - Centre International de Recherche en Infectiologie, Team GIMAP, Univ Lyon, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR5308, CIC 1408 Vaccinology, F42023, Saint-Etienne, France
| | - Louis Waeckel
- CIRI - Centre International de Recherche en Infectiologie, Team GIMAP, Univ Lyon, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR5308, CIC 1408 Vaccinology, F42023, Saint-Etienne, France; Immunology Department, University Hospital of Saint-Etienne, F42055, Saint-Etienne, France
| | - Stéphane Paul
- CIRI - Centre International de Recherche en Infectiologie, Team GIMAP, Univ Lyon, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR5308, CIC 1408 Vaccinology, F42023, Saint-Etienne, France; Immunology Department, University Hospital of Saint-Etienne, F42055, Saint-Etienne, France; CIC 1408 Inserm Vaccinology, University Hospital of Saint-Etienne, F42055, Saint-Etienne, France.
| | - Stéphanie Longet
- CIRI - Centre International de Recherche en Infectiologie, Team GIMAP, Univ Lyon, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR5308, CIC 1408 Vaccinology, F42023, Saint-Etienne, France.
| |
Collapse
|
35
|
Zhang F, Ni L, Zhang Z, Luo X, Wang X, Zhou W, Chen J, Liu J, Qu Y, Liu K, Guo L. Recombinant L. lactis vaccine LL-plSAM-WAE targeting four virulence factors provides mucosal immunity against H. pylori infection. Microb Cell Fact 2024; 23:61. [PMID: 38402145 PMCID: PMC10893618 DOI: 10.1186/s12934-024-02321-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 01/29/2024] [Indexed: 02/26/2024] Open
Abstract
BACKGROUND Helicobacter pylori (H. pylori) causes chronic gastric disease. An efficient oral vaccine would be mucosa-targeted and offer defense against colonization of invasive infection in the digestive system. Proteolytic enzymes and acidic environment in the gastrointestinal tract (GT) can, however, reduce the effectiveness of oral vaccinations. For the creation of an edible vaccine, L. lactis has been proposed as a means of delivering vaccine antigens. RESULTS We developed a plSAM (pNZ8148-SAM) that expresses a multiepitope vaccine antigen SAM-WAE containing Urease, HpaA, HSP60, and NAP extracellularly (named LL-plSAM-WAE) to increase the efficacy of oral vaccinations. We then investigated the immunogenicity of LL-plSAM-WAE in Balb/c mice. Mice that received LL-plSAM-WAE or SAM-WAE with adjuvant showed increased levels of antibodies against H. pylori, including IgG and sIgA, and resulted in significant reductions in H. pylori colonization. Furthermore, we show that SAM-WAE and LL-plSAM-WAE improved the capacity to target the vaccine to M cells. CONCLUSIONS These findings suggest that recombinant L. lactis could be a promising oral mucosa vaccination for preventing H. pylori infection.
Collapse
Affiliation(s)
- Furui Zhang
- College of First Clinical Medicine, Ningxia Medical University, Yinchuan, 750004, China
- College of Laboratory Medicine , Ningxia Medical University, Yinchuan, 750004, China
| | - Linhan Ni
- College of Pharmacy, Ningxia Medical University, Yinchuan, 750004, China
| | - Zhen Zhang
- Department of Geriatrics and Special Needs Medicine, General Hospital of Ningxia Medical University, Yinchuan, 750004, China
| | - Xuegang Luo
- Key Laboratory of Industrial Fermentation Microbiology of the Ministry of Education, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, 300457, China
| | - Xuequan Wang
- Key Laboratory of Radiation Oncology of Taizhou, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Taizhou, 317000, China
| | - Wenmiao Zhou
- College of First Clinical Medicine, Ningxia Medical University, Yinchuan, 750004, China
| | - Jiale Chen
- College of First Clinical Medicine, Ningxia Medical University, Yinchuan, 750004, China
| | - Jing Liu
- College of Laboratory Medicine , Ningxia Medical University, Yinchuan, 750004, China
| | - Yuliang Qu
- College of Laboratory Medicine , Ningxia Medical University, Yinchuan, 750004, China.
| | - Kunmei Liu
- College of Pharmacy, Ningxia Medical University, Yinchuan, 750004, China.
- Ningxia Key Laboratory of Cerebrocranial Diseases, Ningxia Medical University, Yinchuan, 750004, China.
| | - Le Guo
- College of First Clinical Medicine, Ningxia Medical University, Yinchuan, 750004, China.
- College of Laboratory Medicine , Ningxia Medical University, Yinchuan, 750004, China.
- Ningxia Key Laboratory of Cerebrocranial Diseases, Ningxia Medical University, Yinchuan, 750004, China.
- Ningxia Key Laboratory of Clinical and Pathogenic Microbiology, General Hospital of Ningxia Medical University, Yinchuan, 750004, China.
| |
Collapse
|
36
|
Zhang F, Shi T, Zhang Z, Wang S, Liu J, Li Y, Wang X, Liu K, Guo L. An M cell-targeting recombinant L. lactis vaccine against four H. pylori adhesins. Appl Microbiol Biotechnol 2024; 108:231. [PMID: 38396242 PMCID: PMC10891252 DOI: 10.1007/s00253-024-13070-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 01/23/2024] [Accepted: 02/08/2024] [Indexed: 02/25/2024]
Abstract
The acidic environment and enzyme degradation lead to oral vaccines often having little immune effect. Therefore, it is an attractive strategy to study an effective and safe oral vaccine delivery system that can promote gastrointestinal mucosal immune responses and inhibit antigen degradation. Moreover, the antigens uptake by microfold cells (M cells) is the determining step in initiating efficient immune responses. Therefore, M cell-targeting is one promising approach for enhancing oral vaccine potency. In the present study, an M cell-targeting L. lactis surface display system (plSAM) was built to favor the multivalent epitope vaccine antigen (FAdE) to achieve effective gastrointestinal mucosal immunity against Helicobacter pylori. Therefore, a recombinant Lactococcus lactic acid vaccine (LL-plSAM-FAdE) was successfully prepared, and its immunological properties and protective efficacy were analyzed. The results showed that LL-plSAM-FAdE can secretively express the recombinant proteins SAM-FAdE and display the SAM-FAdE on the bacterial cell surface. More importantly, LL-plSAM-FAdE effectively promoted the phagocytosis and transport of vaccine antigen by M cells in the gastrointestinal tract of mice, and simulated high levels of cellular and humoral immune responses against four key H. pylori adhesins (Urease, CagL, HpaA, and Lpp20) in the gastrointestinal tract, thus enabling effective prevention of H. pylori infection and to some extent eliminating H. pylori already present in the gastrointestinal tract. KEY POINTS: • M-cell-targeting L. lactis surface display system LL- plSAM was designed • This system displays H. pylori vaccine-promoted phagocytosis and transport of M cell • A promising vaccine candidate for controlling H. pylori infection was verified.
Collapse
Affiliation(s)
- Furui Zhang
- School of Laboratory, Ningxia Medical University, Yinchuan, 750004, China
| | - Tianyi Shi
- School of Pharmacy, Ningxia Medical University, Yinchuan, 750004, China
| | - Zhen Zhang
- Department of Geriatrics and Special Needs Medicine, General Hospital of Ningxia Medical University, Yinchuan, 750004, China
| | - Shue Wang
- School of Laboratory, Ningxia Medical University, Yinchuan, 750004, China
| | - Jing Liu
- School of Laboratory, Ningxia Medical University, Yinchuan, 750004, China
| | - Yonghong Li
- School of Public Health and Management, Ningxia Medical University, Yinchuan, 750004, China
| | - Xuequan Wang
- Key Laboratory of Radiation Oncology of Taizhou, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Taizhou, 317000, China.
| | - Kunmei Liu
- School of Pharmacy, Ningxia Medical University, Yinchuan, 750004, China.
- Ningxia Key Laboratory of Cerebrocranial Diseases, Ningxia Medical University, Yinchuan, 750004, China.
| | - Le Guo
- School of Laboratory, Ningxia Medical University, Yinchuan, 750004, China.
- Key Laboratory of Radiation Oncology of Taizhou, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Taizhou, 317000, China.
- Ningxia Key Laboratory of Clinical and Pathogenic Microbiology, General Hospital of Ningxia Medical University, Yinchuan, 750004, China.
| |
Collapse
|
37
|
Hsieh MS, Hsu CW, Liao HC, Lin CL, Chiang CY, Chen MY, Liu SJ, Liao CL, Chen HW. SARS-CoV-2 spike-FLIPr fusion protein plus lipidated FLIPr protects against various SARS-CoV-2 variants in hamsters. J Virol 2024; 98:e0154623. [PMID: 38299865 PMCID: PMC10878263 DOI: 10.1128/jvi.01546-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 12/22/2023] [Indexed: 02/02/2024] Open
Abstract
Vaccine-induced mucosal immunity and broad protective capacity against various severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants remain inadequate. Formyl peptide receptor-like 1 inhibitory protein (FLIPr), produced by Staphylococcus aureus, can bind to various Fcγ receptor subclasses. Recombinant lipidated FLIPr (rLF) was previously found to be an effective adjuvant. In this study, we developed a vaccine candidate, the recombinant Delta SARS-CoV-2 spike (rDS)-FLIPr fusion protein (rDS-F), which employs the property of FLIPr binding to various Fcγ receptors. Our study shows that rDS-F plus rLF promotes rDS capture by dendritic cells. Intranasal vaccination of mice with rDS-F plus rLF increases persistent systemic and mucosal antibody responses and CD4/CD8 T-cell responses. Importantly, antibodies induced by rDS-F plus rLF vaccination neutralize Delta, Wuhan, Alpha, Beta, and Omicron strains. Additionally, rDS-F plus rLF provides protective effects against various SARS-CoV-2 variants in hamsters by reducing inflammation and viral loads in the lung. Therefore, rDS-F plus rLF is a potential vaccine candidate to induce broad protective responses against various SARS-CoV-2 variants.IMPORTANCEMucosal immunity is vital for combating pathogens, especially in the context of respiratory diseases like COVID-19. Despite this, most approved vaccines are administered via injection, providing systemic but limited mucosal protection. Developing vaccines that stimulate both mucosal and systemic immunity to address future coronavirus mutations is a growing trend. However, eliciting strong mucosal immune responses without adjuvants remains a challenge. In our study, we have demonstrated that using a recombinant severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike-formyl peptide receptor-like 1 inhibitory protein (FLIPr) fusion protein as an antigen, in combination with recombinant lipidated FLIPr as an effective adjuvant, induced simultaneous systemic and mucosal immune responses through intranasal immunization in mice and hamster models. This approach offered protection against various SARS-CoV-2 strains, making it a promising vaccine candidate for broad protection. This finding is pivotal for future broad-spectrum 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
| | - Hung-Chun Liao
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
| | - Chang-Ling Lin
- 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
| | - Mei-Yu Chen
- 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
|
38
|
Koirala P, Shalash AO, Chen SPR, Faruck MO, Wang J, Hussein WM, Khalil ZG, Capon RJ, Monteiro MJ, Toth I, Skwarczynski M. Polymeric Nanoparticles as Oral and Intranasal Peptide Vaccine Delivery Systems: The Role of Shape and Conjugation. Vaccines (Basel) 2024; 12:198. [PMID: 38400181 PMCID: PMC10893271 DOI: 10.3390/vaccines12020198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 02/09/2024] [Accepted: 02/13/2024] [Indexed: 02/25/2024] Open
Abstract
Mucosal vaccines are highly attractive due to high patient compliance and their suitability for mass immunizations. However, all currently licensed mucosal vaccines are composed of attenuated/inactive whole microbes, which are associated with a variety of safety concerns. In contrast, modern subunit vaccines use minimal pathogenic components (antigens) that are safe but typically poorly immunogenic when delivered via mucosal administration. In this study, we demonstrated the utility of various functional polymer-based nanostructures as vaccine carriers. A Group A Streptococcus (GAS)-derived peptide antigen (PJ8) was selected in light of the recent global spread of invasive GAS infection. The vaccine candidates were prepared by either conjugation or physical mixing of PJ8 with rod-, sphere-, worm-, and tadpole-shaped polymeric nanoparticles. The roles of nanoparticle shape and antigen conjugation in vaccine immunogenicity were demonstrated through the comparison of three distinct immunization pathways (subcutaneous, intranasal, and oral). No additional adjuvant or carrier was required to induce bactericidal immune responses even upon oral vaccine administration.
Collapse
Affiliation(s)
- Prashamsa Koirala
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD 4072, Australia; (P.K.); (A.O.S.); (M.O.F.); (J.W.); (W.M.H.)
| | - Ahmed O. Shalash
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD 4072, Australia; (P.K.); (A.O.S.); (M.O.F.); (J.W.); (W.M.H.)
| | - Sung-Po R. Chen
- Australian Institute of Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia; (S.-P.R.C.); (M.J.M.)
| | - Mohammad O. Faruck
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD 4072, Australia; (P.K.); (A.O.S.); (M.O.F.); (J.W.); (W.M.H.)
| | - Jingwen Wang
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD 4072, Australia; (P.K.); (A.O.S.); (M.O.F.); (J.W.); (W.M.H.)
| | - Waleed M. Hussein
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD 4072, Australia; (P.K.); (A.O.S.); (M.O.F.); (J.W.); (W.M.H.)
| | - Zeinab G. Khalil
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD 4072, Australia; (Z.G.K.); (R.J.C.)
| | - Robert J. Capon
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD 4072, Australia; (Z.G.K.); (R.J.C.)
| | - Michael J. Monteiro
- Australian Institute of Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia; (S.-P.R.C.); (M.J.M.)
| | - Istvan Toth
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD 4072, Australia; (P.K.); (A.O.S.); (M.O.F.); (J.W.); (W.M.H.)
- School of Pharmacy, The University of Queensland, Woolloongabba, QLD 4102, Australia
| | - Mariusz Skwarczynski
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD 4072, Australia; (P.K.); (A.O.S.); (M.O.F.); (J.W.); (W.M.H.)
| |
Collapse
|
39
|
Song Y, Mehl F, Zeichner SL. Vaccine Strategies to Elicit Mucosal Immunity. Vaccines (Basel) 2024; 12:191. [PMID: 38400174 PMCID: PMC10892965 DOI: 10.3390/vaccines12020191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 01/29/2024] [Accepted: 02/08/2024] [Indexed: 02/25/2024] Open
Abstract
Vaccines are essential tools to prevent infection and control transmission of infectious diseases that threaten public health. Most infectious agents enter their hosts across mucosal surfaces, which make up key first lines of host defense against pathogens. Mucosal immune responses play critical roles in host immune defense to provide durable and better recall responses. Substantial attention has been focused on developing effective mucosal vaccines to elicit robust localized and systemic immune responses by administration via mucosal routes. Mucosal vaccines that elicit effective immune responses yield protection superior to parenterally delivered vaccines. Beyond their valuable immunogenicity, mucosal vaccines can be less expensive and easier to administer without a need for injection materials and more highly trained personnel. However, developing effective mucosal vaccines faces many challenges, and much effort has been directed at their development. In this article, we review the history of mucosal vaccine development and present an overview of mucosal compartment biology and the roles that mucosal immunity plays in defending against infection, knowledge that has helped inform mucosal vaccine development. We explore new progress in mucosal vaccine design and optimization and novel approaches created to improve the efficacy and safety of mucosal vaccines.
Collapse
Affiliation(s)
- Yufeng Song
- Department of Pediatrics, University of Virginia, Charlottesville, VA 22908, USA; (Y.S.)
| | - Frances Mehl
- Department of Pediatrics, University of Virginia, Charlottesville, VA 22908, USA; (Y.S.)
| | - Steven L. Zeichner
- Department of Pediatrics, University of Virginia, Charlottesville, VA 22908, USA; (Y.S.)
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA 22908, USA
| |
Collapse
|
40
|
Liu X, Lin X, Hong H, Wang J, Tao Y, Huai Y, Pang H, Liu M, Li J, Bo R. Polysaccharide from Atractylodes macrocephala Koidz Binding with Zinc Oxide Nanoparticles as a Novel Mucosal Immune Adjuvant for H9N2 Inactivated Vaccine. Int J Mol Sci 2024; 25:2132. [PMID: 38396809 PMCID: PMC10889192 DOI: 10.3390/ijms25042132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 01/31/2024] [Accepted: 02/07/2024] [Indexed: 02/25/2024] Open
Abstract
H9N2 avian influenza poses a significant public health risk, necessitating effective vaccines for mass immunization. Oral inactivated vaccines offer advantages like the ease of administration, but their efficacy often requires enhancement through mucosal adjuvants. In a previous study, we established a novel complex of polysaccharide from Atractylodes macrocephala Koidz binding with zinc oxide nanoparticles (AMP-ZnONPs) and preliminarily demonstrated its immune-enhancing function. This work aimed to evaluate the efficacy of AMP-ZnONPs as adjuvants in an oral H9N2-inactivated vaccine and the vaccine's impact on intestinal mucosal immunity. In this study, mice were orally vaccinated on days 0 and 14 after adapting to the environment. AMP-ZnONPs significantly improved HI titers, the levels of specific IgG, IgG1 and IgG2a in serum and sIgA in intestinal lavage fluid; increased the number of B-1 and B-2 cells and dendritic cell populations; and enhanced the mRNA expression of intestinal homing factors and immune-related cytokines. Interestingly, AMP-ZnONPs were more likely to affect B-1 cells than B-2 cells. AMP-ZnONPs showed mucosal immune enhancement that was comparable to positive control (cholera toxin, CT), but not to the side effect of weight loss caused by CT. Compared to the whole-inactivated H9N2 virus (WIV) group, the WIV + AMP-ZnONP and WIV + CT groups exhibited opposite shifts in gut microbial abundance. AMP-ZnONPs serve as an effective and safe mucosal adjuvant for oral WIV, improving cellular, humoral and mucosal immunity and microbiota in the gastrointestinal tract, avoiding the related undesired effects of CT.
Collapse
Affiliation(s)
- Xiaopan Liu
- College of Veterinary Medicine, Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China; (X.L.); (X.L.); (H.H.); (J.W.); (Y.T.); (Y.H.); (M.L.)
| | - Xinyi Lin
- College of Veterinary Medicine, Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China; (X.L.); (X.L.); (H.H.); (J.W.); (Y.T.); (Y.H.); (M.L.)
| | - Hailong Hong
- College of Veterinary Medicine, Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China; (X.L.); (X.L.); (H.H.); (J.W.); (Y.T.); (Y.H.); (M.L.)
| | - Jing Wang
- College of Veterinary Medicine, Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China; (X.L.); (X.L.); (H.H.); (J.W.); (Y.T.); (Y.H.); (M.L.)
| | - Ya Tao
- College of Veterinary Medicine, Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China; (X.L.); (X.L.); (H.H.); (J.W.); (Y.T.); (Y.H.); (M.L.)
| | - Yuying Huai
- College of Veterinary Medicine, Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China; (X.L.); (X.L.); (H.H.); (J.W.); (Y.T.); (Y.H.); (M.L.)
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Huan Pang
- School of Chemistry and Chemical Engineering, Yangzhou University, Yangzhou 225009, China;
| | - Mingjiang Liu
- College of Veterinary Medicine, Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China; (X.L.); (X.L.); (H.H.); (J.W.); (Y.T.); (Y.H.); (M.L.)
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Jingui Li
- College of Veterinary Medicine, Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China; (X.L.); (X.L.); (H.H.); (J.W.); (Y.T.); (Y.H.); (M.L.)
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Ruonan Bo
- College of Veterinary Medicine, Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China; (X.L.); (X.L.); (H.H.); (J.W.); (Y.T.); (Y.H.); (M.L.)
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| |
Collapse
|
41
|
Macedo BG, Masuda MY, Borges da Silva H. Location versus ID: what matters to lung-resident memory T cells? Front Immunol 2024; 15:1355910. [PMID: 38375476 PMCID: PMC10875077 DOI: 10.3389/fimmu.2024.1355910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 01/16/2024] [Indexed: 02/21/2024] Open
Abstract
Tissue-resident memory T cells (TRM cells) are vital for the promotion of barrier immunity. The lung, a tissue constantly exposed to foreign pathogenic or non-pathogenic antigens, is not devoid of these cells. Lung TRM cells have been considered major players in either the protection against respiratory viral infections or the pathogenesis of lung allergies. Establishment of lung TRM cells rely on intrinsic and extrinsic factors. Among the extrinsic regulators of lung TRM cells, the magnitude of the impact of factors such as the route of antigen entry or the antigen natural tropism for the lung is not entirely clear. In this perspective, we provide a summary of the literature covering this subject and present some preliminary results on this potential dichotomy between antigen location versus antigen type. Finally, we propose a hypothesis to synthesize the potential contributions of these two variables for lung TRM cell development.
Collapse
|
42
|
Hoover AR, More S, Liu K, West CL, Valerio TI, Furrer CL, Adams JP, Yu N, Villalva C, Kumar A, Alleruzzo L, Lam SSK, Hode T, Papin JF, Chen WR. N-dihydrogalactochitosan serves as an effective mucosal adjuvant for intranasal vaccine in combination with recombinant viral proteins against respiratory infection. Acta Biomater 2024; 175:279-292. [PMID: 38160856 DOI: 10.1016/j.actbio.2023.12.039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 12/21/2023] [Accepted: 12/22/2023] [Indexed: 01/03/2024]
Abstract
Mucosal vaccinations for respiratory pathogens provide effective protection as they stimulate localized cellular and humoral immunities at the site of infection. Currently, the major limitation of intranasal vaccination is using effective adjuvants capable of withstanding the harsh environment imposed by the mucosa. Herein, we describe the efficacy of using a unique biopolymer, N-dihydrogalactochitosan (GC), as a nasal mucosal vaccine adjuvant against respiratory infections. Specifically, we mixed GC with recombinant SARS-CoV-2 trimeric spike (S) and nucleocapsid (NC) proteins to intranasally vaccinate K18-hACE2 transgenic mice, in comparison with Addavax (AV), an MF-59 equivalent. In contrast to AV, intranasal application of GC induces a robust, systemic antigen-specific antibody response and increases the number of T cells in the cervical lymph nodes. Moreover, GC+S+NC-vaccinated animals were largely resistant to the lethal SARS-CoV-2 challenge and experienced drastically reduced morbidity and mortality, with animal weights and behavior returning to normal 22 days post-infection. In contrast, animals intranasally vaccinated with AV+S+NC experienced severe weight loss, mortality, and respiratory distress, with none surviving beyond 6 days post-infection. Our findings demonstrate that GC can serve as a potent mucosal vaccine adjuvant against SARS-CoV-2 and potentially other respiratory viruses. STATEMENT OF SIGNIFICANCE: We demonstrated that a unique biopolymer, N-dihydrogalactochitosan (GC), was an effective nasal mucosal vaccine adjuvant against respiratory infections. Specifically, we mixed GC with recombinant SARS-CoV-2 trimeric spike (S) and nucleocapsid (NC) proteins to intranasally vaccinate K18-hACE2 transgenic mice, in comparison with Addavax (AV). In contrast to AV, GC induces a robust, systemic antigen-specific antibody response and increases the number of T cells in the cervical lymph nodes. About 90 % of the GC+S+NC-vaccinated animals survived the lethal SARS-CoV-2 challenge and remained healthy 22 days post-infection, while the AV+S+NC-vaccinated animals experienced severe weight loss and respiratory distress, and all died within 6 days post-infection. Our findings demonstrate that GC is a potent mucosal vaccine adjuvant against SARS-CoV-2 and potentially other respiratory viruses.
Collapse
Affiliation(s)
- Ashley R Hoover
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, USA; Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Sunil More
- Veterinary Pathobiology, College of Veterinary Medicine, Oklahoma State University, Stillwater, OK USA
| | - Kaili Liu
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, USA
| | - Connor L West
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, USA
| | - Trisha I Valerio
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, USA
| | - Coline L Furrer
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, USA
| | - Jacob P Adams
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, USA
| | - Ningli Yu
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, USA
| | - Crystal Villalva
- Veterinary Pathobiology, College of Veterinary Medicine, Oklahoma State University, Stillwater, OK USA
| | - Amit Kumar
- Biogen Inc., 225 Bnney Street, Cambridge, MA, USA
| | - Lu Alleruzzo
- Immunophotonics, Inc., 4340 Duncan Avenue, Suite 212, Saint Louis, MO, USA
| | - Samuel S K Lam
- Immunophotonics, Inc., 4340 Duncan Avenue, Suite 212, Saint Louis, MO, USA
| | - Tomas Hode
- Immunophotonics, Inc., 4340 Duncan Avenue, Suite 212, Saint Louis, MO, USA
| | - James F Papin
- Department Pathology and Division of Comparative Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, USA
| | - Wei R Chen
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, USA.
| |
Collapse
|
43
|
Park SI, Park S, Lee K, Kwak HW, Kim YK, Park HJ, Bang YJ, Kim JY, Kim D, Seo KW, Lee SJ, Kim H, Kim Y, Kim DH, Park HJ, Jung SY, Ga E, Hwang J, Na W, Hong SH, Lee SM, Nam JH. Intranasal immunization with the recombinant measles virus encoding the spike protein of SARS-CoV-2 confers protective immunity against COVID-19 in hamsters. Vaccine 2024; 42:69-74. [PMID: 38097457 DOI: 10.1016/j.vaccine.2023.12.011] [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: 04/22/2023] [Revised: 10/12/2023] [Accepted: 12/02/2023] [Indexed: 01/01/2024]
Abstract
BACKGROUND As the nasal mucosa is the initial site of infection for COVID-19, intranasal vaccines are more favorable than conventional vaccines. In recent clinical studies, intranasal immunization has been shown to generate higher neutralizing antibodies; however, there is a lack of evidence on sterilizing immunity in the upper airway. Previously, we developed a recombinant measles virus encoding the spike protein of SARS-CoV-2 (rMeV-S), eliciting humoral and cellular immune responses against SARS-CoV-2. OBJECTIVES In this study, we aim to provide an experiment on nasal vaccines focusing on a measles virus platform as well as injection routes. STUDY DESIGN Recombinant measles viruses expressing rMeV-S were prepared, and 5 × 105 PFUs of rMeV-S were administered to Syrian golden hamsters via intramuscular or intranasal injection. Subsequently, the hamsters were challenged with inoculations of 1 × 105 PFUs of SARS-CoV-2 and euthanized 4 days post-infection. Neutralizing antibodies and RBD-specific IgG in the serum and RBD-specific IgA in the bronchoalveolar lavage fluid (BALF) were measured, and SARS-CoV-2 clearance capacity was determined via quantitative reverse-transcription PCR (qRT-PCR) analysis and viral titer measurement in the upper respiratory tract and lungs. Immunohistochemistry and histopathological examinations of lung samples from experimental hamsters were conducted. RESULTS The intranasal immunization of rMeV-S elicits protective immune responses and alleviates virus-induced pathophysiology, such as body weight reduction and lung weight increase in hamsters. Furthermore, lung immunohistochemistry demonstrated that intranasal rMeV-S immunization induces effective SARS-CoV-2 clearance that correlates with viral RNA content, as determined by qRT-PCR, in the lung and nasal wash samples, SARS-CoV-2 viral titers in lung, nasal wash, BALF samples, serum RBD-specific IgG concentration, and RBD-specific IgA concentration in the BALF. CONCLUSION An intranasal vaccine based on the measles virus platform is a promising strategy owing to the typical route of infection of the virus, the ease of administration of the vaccine, and the strong immune response it elicits.
Collapse
Affiliation(s)
| | - Sohyun Park
- Chungbuk National University, Cheongju, Republic of Korea
| | - Kunse Lee
- SK Bioscience, Seongnam, Republic of Korea
| | - Hye Won Kwak
- SML Biopharm, Gwangmyeong, Republic of Korea; The Catholic University of Korea, Bucheon, Republic of Korea
| | | | - Hyeong-Jun Park
- SML Biopharm, Gwangmyeong, Republic of Korea; The Catholic University of Korea, Bucheon, Republic of Korea
| | - Yoo-Jin Bang
- The Catholic University of Korea, Bucheon, Republic of Korea
| | - Jae-Yong Kim
- The Catholic University of Korea, Bucheon, Republic of Korea
| | - Daegeun Kim
- SML Biopharm, Gwangmyeong, Republic of Korea
| | | | | | - Hun Kim
- SK Bioscience, Seongnam, Republic of Korea
| | - Yeonhwa Kim
- Chungbuk National University, Cheongju, Republic of Korea
| | - Do-Hyung Kim
- SML Biopharm, Gwangmyeong, Republic of Korea; The Catholic University of Korea, Bucheon, Republic of Korea
| | - Hyo-Jung Park
- The Catholic University of Korea, Bucheon, Republic of Korea
| | | | - Eulhae Ga
- Chonnam National University, Gwangju, Republic of Korea
| | - Jaehyun Hwang
- Chonnam National University, Gwangju, Republic of Korea
| | - Woonsung Na
- Chonnam National University, Gwangju, Republic of Korea
| | - So-Hee Hong
- Ewha Womans University, Seoul, Republic of Korea
| | | | - Jae-Hwan Nam
- The Catholic University of Korea, Bucheon, Republic of Korea.
| |
Collapse
|
44
|
Zhang C, Zhao Z, Jia YJ, Zhang PQ, Sun Y, Zhou YC, Wang GX, Zhu B. Rationally Designed Self-Assembling Nanovaccines Elicit Robust Mucosal and Systemic Immunity against Rhabdovirus. ACS APPLIED MATERIALS & INTERFACES 2024; 16:228-244. [PMID: 38055273 DOI: 10.1021/acsami.3c14305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/07/2023]
Abstract
Viral diseases have constantly caused great threats to global public health, resulting in an urgent need for effective vaccines. However, the current viral vaccines often show low immunogenicity. To counter this, we report a smart strategy of a well-designed modular nanoparticle (LSG-TDH) that recapitulates the dominant antigen SG, low-molecular-weight protamine, and tetralysine-modified H-chain apoferritin (TDH). The constructed LSG-TDH nanovaccine could self-assemble into a nanocage structure, which confers excellent mucus-penetrating, cellular affinity, and uptake ability. Studies demonstrate that the LSG-TDH nanovaccine could strongly activate both mucosal and systemic immune responses. Importantly, by immunizing wild-type and TLR2 knockout (TLR2-KO) zebrafish, we found that TLR2 could mediate LSG-TDH-induced adaptive mucosal and systemic immune responses by activating antigen-presenting cells. Collectively, our findings offer new insights into rational viral vaccine design and provide additional evidence of the vital role of TLR2 in regulating adaptive immunity.
Collapse
Affiliation(s)
- Chen Zhang
- Collaborative Innovation Center of Marine Science and Technology, Hainan Provincial Key Laboratory for Tropical Hydrobiology and Biotechnology, College of Marine Science, Hainan University, Haikou 570228, P. R. China
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, Shaanxi, P. R. China
| | - Zhao Zhao
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, Shaanxi, P. R. China
| | - Yi-Jun Jia
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, Shaanxi, P. R. China
| | - Peng-Qi Zhang
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, Shaanxi, P. R. China
| | - Yun Sun
- Collaborative Innovation Center of Marine Science and Technology, Hainan Provincial Key Laboratory for Tropical Hydrobiology and Biotechnology, College of Marine Science, Hainan University, Haikou 570228, P. R. China
| | - Yong-Can Zhou
- Collaborative Innovation Center of Marine Science and Technology, Hainan Provincial Key Laboratory for Tropical Hydrobiology and Biotechnology, College of Marine Science, Hainan University, Haikou 570228, P. R. China
| | - Gao-Xue Wang
- College of Animal Science and Technology, Key Laboratory of Livestock Biology, Northwest A&F University, Yangling 712100, Shaanxi, P. R. China
| | - Bin Zhu
- College of Animal Science and Technology, Key Laboratory of Livestock Biology, Northwest A&F University, Yangling 712100, Shaanxi, P. R. China
| |
Collapse
|
45
|
Song H, Abdullah SW, Pei C, Shi X, Chen X, Ma Y, Yin S, Sun S, Huang Y, Guo H. Self-Assembling E2-Based Nanoparticles Improve Vaccine Thermostability and Protective Immunity against CSFV. Int J Mol Sci 2024; 25:596. [PMID: 38203765 PMCID: PMC10778992 DOI: 10.3390/ijms25010596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 12/23/2023] [Accepted: 12/25/2023] [Indexed: 01/12/2024] Open
Abstract
Classical swine fever virus (CSFV) is a highly contagious pathogen causing significant economic losses in the swine industry. Conventional inactivated or attenuated live vaccines for classical swine fever (CSF) are effective but face biosafety concerns and cannot distinguish vaccinated animals from those infected with the field virus, complicating CSF eradication efforts. It is noteworthy that nanoparticle (NP)-based vaccines resemble natural viruses in size and antigen structure, and offer an alternative tool to circumvent these limitations. In this study, we developed an innovative vaccine delivery scaffold utilizing self-assembled mi3 NPs, which form stable structures carrying the CSFV E2 glycoprotein. The expressed yeast E2-fused protein (E2-mi3 NPs) exhibited robust thermostability (25 to 70 °C) and long-term storage stability at room temperature (25 °C). Interestingly, E2-mi3 NPs made with this technology elicited enhanced antigen uptake by RAW264.7 cells. In a rabbit model, the E2-mi3 NP vaccine against CSFV markedly increased CSFV-specific neutralizing antibody titers. Importantly, it conferred complete protection in rabbits challenged with the C-strain of CSFV. Furthermore, we also found that the E2-mi3 NP vaccines triggered stronger cellular (T-lymphocyte proliferation, CD8+ T-lymphocytes, IFN-γ, IL-2, and IL-12p70) and humoral (CSFV-specific neutralizing antibodies, CD4+ T-lymphocytes, and IL-4) immune responses in pigs than the E2 vaccines. To sum up, these structure-based, self-assembled mi3 NPs provide valuable insights for novel antiviral strategies against the constantly infectious agents.
Collapse
Affiliation(s)
- Hetao Song
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China;
| | - Sahibzada Waheed Abdullah
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China; (S.W.A.); (X.S.); (X.C.); (Y.M.); (S.Y.); (S.S.)
| | - Chenchen Pei
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China; (S.W.A.); (X.S.); (X.C.); (Y.M.); (S.Y.); (S.S.)
| | - Xiaoni Shi
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China; (S.W.A.); (X.S.); (X.C.); (Y.M.); (S.Y.); (S.S.)
| | - Xiangyang Chen
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China; (S.W.A.); (X.S.); (X.C.); (Y.M.); (S.Y.); (S.S.)
| | - Yuqing Ma
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China; (S.W.A.); (X.S.); (X.C.); (Y.M.); (S.Y.); (S.S.)
| | - Shuanghui Yin
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China; (S.W.A.); (X.S.); (X.C.); (Y.M.); (S.Y.); (S.S.)
| | - Shiqi Sun
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China; (S.W.A.); (X.S.); (X.C.); (Y.M.); (S.Y.); (S.S.)
| | - Yong Huang
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China;
| | - Huichen Guo
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China; (S.W.A.); (X.S.); (X.C.); (Y.M.); (S.Y.); (S.S.)
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou 730046, China
| |
Collapse
|
46
|
Heck K, Farris E, Pannier AK. Formulation of Chitosan-Zein Nano-in-Microparticles for Oral DNA Delivery. Methods Mol Biol 2024; 2720:165-176. [PMID: 37775665 DOI: 10.1007/978-1-0716-3469-1_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/01/2023]
Abstract
Gene delivery via the oral route offers a promising strategy for improving DNA vaccination and gene-based therapy outcomes. The noninvasive nature of oral delivery lends to ease of dosing, which can facilitate convenience and patient compliance. Moreover, oral administration allows for both local and systemic production of therapeutic genes or, in the case of DNA vaccination, mucosal and systemic immunity. Here, we describe the methods to produce a dual biomaterial, oral DNA delivery system composed of chitosan (CS) and zein (ZN). In this system, CS serves to encapsulate and deliver DNA cargo to intestinal cells in the form of CS-DNA nanoparticles (CS-DNA NPs), while ZN is used to form a protective matrix around the CS-DNA NPs that prevent degradation during gastric transit but then degrades to release the CS-DNA NPs for transfection upon entry into the intestines. These particles have demonstrated the ability to effectively protect cargo DNA from simulated gastric degradation in vitro and mediate transgene production in vivo, making them an effective oral gene delivery system.
Collapse
Affiliation(s)
- Kari Heck
- Department of Biological Systems Engineering, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Eric Farris
- Adjuvance Technologies Inc., Lincoln, NE, USA
| | - Angela K Pannier
- Department of Biological Systems Engineering, University of Nebraska-Lincoln, Lincoln, NE, USA.
| |
Collapse
|
47
|
Liu Q, Liu Z, Huang B, Teng Y, Li M, Peng S, Guo H, Wang M, Liang J, Zhang Y. Global trends in poliomyelitis research over the past 20 years: A bibliometric analysis. Hum Vaccin Immunother 2023; 19:2173905. [PMID: 36803526 PMCID: PMC10038019 DOI: 10.1080/21645515.2023.2173905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2023] Open
Abstract
Poliomyelitis is an acute infectious disease caused by poliovirus. This bibliometric analysis aims to examine the status of poliomyelitis research in the past 20 years. Information regarding polio research was obtained from the Web of Science Core Collection database. CiteSpace, VOSviewer, and Excel were used to perform visual and bibliometric analysis with respect to countries/regions, institutions, authors, journals and keywords. A total of 5,335 publications on poliomyelitis were published from 2002 to 2021. The USA was the county with the majority of publications. Additionally, the most productive institution was the Centers for Disease Control and Prevention. Sutter, RW produced the most papers and had the most co-citations. Vaccine was the journal with the most polio-related publications and citations. The most common keywords were mainly about polio immunology research ("polio," "immunization," "children," "eradication" and "vaccine"). Our study is helpful for identifying research hotspots and providing direction for future research on poliomyelitis.
Collapse
Affiliation(s)
- Qi Liu
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ze Liu
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Biling Huang
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yuanyuan Teng
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Mingliu Li
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Shuqin Peng
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Hongbin Guo
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Min Wang
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jieyu Liang
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yi Zhang
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
48
|
Tong T, Qi Y, Rollins D, Bussiere LD, Dhar D, Miller CL, Yu C, Wang Q. Rational design of oral drugs targeting mucosa delivery with gut organoid platforms. Bioact Mater 2023; 30:116-128. [PMID: 37560199 PMCID: PMC10406959 DOI: 10.1016/j.bioactmat.2023.07.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 07/16/2023] [Accepted: 07/17/2023] [Indexed: 08/11/2023] Open
Abstract
Effective oral drugs and vaccines require high delivery efficiency across the gastrointestinal epithelia and protection of medically effective payloads (i.e., immunogens) against gastric damage. In this study, hollowed nanocarriers (NCs: silica nanospheres and gold nanocages) with poly-l-lysine (PLL) coating and mammalian orthoreovirus cell attachment protein σ1 functionalization (NC-PLL-σ1) were explored as functional oral drug delivery vehicles (ODDVs). The transport of these ODDVs to mucosal lymphoid tissues could be facilitated by microfold cells (M-cells) mediated transcytosis (via σ1-α2-3-linked sialic acids adherence) across gastrointestinal epithelia. PLL coating provided protection and slow-release of rhodamine 6 G (R6G), a model payload. The transport effectiveness of these ODDVs was tested on intestinal organoid monolayers in vitro. When compared with other experimental groups, the fully functionalized ODDV system (with PLL-σ1) demonstrated two significant advantages: a significantly higher transport efficiency (198% over blank control at 48 h); and protection of payloads which led to both better transport efficiency and extended-release of payloads (61% over uncoated carriers at 48 h). In addition, it was shown that the M cell presence in intestinal organoid monolayers (modulated by Rank L stimulation) was a determining factor on the transport efficiency of the ODDVs: more M-cells (induced by higher Rank L) in the organoid monolayers led to higher transport efficiency for ODDV-delivered model payload (R6G). The fully functionalized ODDVs showed great potential as effective oral delivery vehicles for drugs and vaccines.
Collapse
Affiliation(s)
- Tianjian Tong
- Department of Agricultural Biosystem and Engineering, Iowa State University, Ames, IA, USA
| | - Yijun Qi
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA, USA
| | - Derrick Rollins
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA, USA
- Department of Statistics, Iowa State University, Ames, IA, USA
| | - Luke D. Bussiere
- Department of Veterinary Microbiology and Preventive Medicine, College of Veterinary Medicine, Iowa State University, Ames, IA, USA
- Interdepartmental Microbiology Program, Iowa State University, Ames, IA, USA
| | - Debarpan Dhar
- Department of Veterinary Microbiology and Preventive Medicine, College of Veterinary Medicine, Iowa State University, Ames, IA, USA
- Interdepartmental Microbiology Program, Iowa State University, Ames, IA, USA
| | - Cathy L. Miller
- Department of Veterinary Microbiology and Preventive Medicine, College of Veterinary Medicine, Iowa State University, Ames, IA, USA
- Interdepartmental Microbiology Program, Iowa State University, Ames, IA, USA
| | - Chenxu Yu
- Department of Agricultural Biosystem and Engineering, Iowa State University, Ames, IA, USA
| | - Qun Wang
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA, USA
| |
Collapse
|
49
|
Wang S, Cui H, Zhang C, Li W, Wang W, He W, Feng N, Zhao Y, Wang T, Tang X, Yan F, Xia X. Oral delivery of a chitosan adjuvanted COVID-19 vaccine provides long-lasting and broad-spectrum protection against SARS-CoV-2 variants of concern in golden hamsters. Antiviral Res 2023; 220:105765. [PMID: 38036065 DOI: 10.1016/j.antiviral.2023.105765] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 10/27/2023] [Accepted: 11/22/2023] [Indexed: 12/02/2023]
Abstract
Coronavirus disease 2019 (COVID-19) seriously threatens public health safety and the global economy, which warrant effective prophylactic and therapeutic approaches. Currently, vaccination and establishment of immunity have significantly reduced the severity and mortality of COVID-19. However, in regard to COVID-19 vaccines, the broad-spectrum protective efficacy against SARS-CoV-2 variants and the blocking of virus transmission need to be further improved. In this study, an optimum oral COVID-19 vaccine candidate, rVSVΔG-Sdelta, was selected from a panel of vesicular stomatitis virus (VSV)-based constructs bearing spike proteins from different SARS-CoV-2 strains. After chitosan modification, rVSVΔG-Sdelta induced both local and peripheral antibody response, particularly, broad-spectrum and long-lasting neutralizing antibodies against SARS-CoV-2 persisted for 1 year. Cross-protection against SARS-CoV-2 WT, Beta, Delta, BA.1, and BA.2 strains was achieved in golden hamsters, which presented as significantly reduced viral replication in the respiratory tract and alleviated pulmonary pathology post SARS-CoV-2 challenge. Overall, this study provides a convenient, oral-delivered, and effective oral mucosal vaccine against COVID-19, which would supplement pools and facilitate the distribution of COVID-19 vaccines.
Collapse
Affiliation(s)
- Shen Wang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130000, China
| | - Huan Cui
- College of Veterinary Medicine, Hebei Agricultural University, 2596 Lucky South Street, Baoding, 071000, China
| | - Cheng Zhang
- College of Veterinary Medicine, Hebei Agricultural University, 2596 Lucky South Street, Baoding, 071000, China
| | - Wujian Li
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130000, China; College of Veterinary Medicine, Jilin University, Changchun, 130062, Jilin, China
| | - Weiqi Wang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130000, China; College of Veterinary Medicine, Jilin University, Changchun, 130062, Jilin, China
| | - Wenwen He
- The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 42100, China
| | - Na Feng
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130000, China
| | - Yongkun Zhao
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130000, China
| | - Tiecheng Wang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130000, China
| | - Xiaoqing Tang
- The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 42100, China.
| | - Feihu Yan
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130000, China.
| | - Xianzhu Xia
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130000, China.
| |
Collapse
|
50
|
Gilfillan D, Vilander AC, Pan M, Goh YJ, O’Flaherty S, Feng N, Fox BE, Lang C, Greenberg HB, Abdo Z, Barrangou R, Dean GA. Lactobacillus acidophilus Expressing Murine Rotavirus VP8 and Mucosal Adjuvants Induce Virus-Specific Immune Responses. Vaccines (Basel) 2023; 11:1774. [PMID: 38140179 PMCID: PMC10747613 DOI: 10.3390/vaccines11121774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 11/14/2023] [Accepted: 11/22/2023] [Indexed: 12/24/2023] Open
Abstract
Rotavirus diarrhea-associated illness remains a major cause of global death in children under five, attributable in part to discrepancies in vaccine performance between high- and low-middle-income countries. Next-generation probiotic vaccines could help bridge this efficacy gap. We developed a novel recombinant Lactobacillus acidophilus (rLA) vaccine expressing rotavirus antigens of the VP8* domain from the rotavirus EDIM VP4 capsid protein along with the adjuvants FimH and FliC. The upp-based counterselective gene-replacement system was used to chromosomally integrate FimH, VP8Pep (10 amino acid epitope), and VP8-1 (206 amino acid protein) into the L. acidophilus genome, with FliC expressed from a plasmid. VP8 antigen and adjuvant expression were confirmed by flow cytometry and Western blot. Rotavirus naïve adult BALB/cJ mice were orally immunized followed by murine rotavirus strain ECWT viral challenge. Antirotavirus serum IgG and antigen-specific antibody-secreting cell responses were detected in rLA-vaccinated mice. A day after the oral rotavirus challenge, fecal antigen shedding was significantly decreased in the rLA group. These results indicate that novel rLA constructs expressing VP8 can be successfully constructed and used to generate modest homotypic protection from rotavirus challenge in an adult murine model, indicating the potential for a probiotic next-generation vaccine construct against human rotavirus.
Collapse
Affiliation(s)
- Darby Gilfillan
- Department of Microbiology, Immunology and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA; (D.G.); (A.C.V.); (B.E.F.); (C.L.); (Z.A.)
| | - Allison C. Vilander
- Department of Microbiology, Immunology and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA; (D.G.); (A.C.V.); (B.E.F.); (C.L.); (Z.A.)
| | - Meichen Pan
- Department of Food, Bioprocessing and Nutrition Sciences, North Carolina State University, Raleigh, NC 27695, USA; (M.P.); (Y.J.G.); (S.O.); (R.B.)
| | - Yong Jun Goh
- Department of Food, Bioprocessing and Nutrition Sciences, North Carolina State University, Raleigh, NC 27695, USA; (M.P.); (Y.J.G.); (S.O.); (R.B.)
| | - Sarah O’Flaherty
- Department of Food, Bioprocessing and Nutrition Sciences, North Carolina State University, Raleigh, NC 27695, USA; (M.P.); (Y.J.G.); (S.O.); (R.B.)
| | - Ningguo Feng
- Departments of Medicine and Microbiology and Immunology, School of Medicine, Stanford University, Stanford, CA 94305, USA (H.B.G.)
- VA Palo Alto Health Care System, Department of Veterans Affairs, Palo Alto, CA 94304, USA
| | - Bridget E. Fox
- Department of Microbiology, Immunology and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA; (D.G.); (A.C.V.); (B.E.F.); (C.L.); (Z.A.)
| | - Callie Lang
- Department of Microbiology, Immunology and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA; (D.G.); (A.C.V.); (B.E.F.); (C.L.); (Z.A.)
| | - Harry B. Greenberg
- Departments of Medicine and Microbiology and Immunology, School of Medicine, Stanford University, Stanford, CA 94305, USA (H.B.G.)
- VA Palo Alto Health Care System, Department of Veterans Affairs, Palo Alto, CA 94304, USA
| | - Zaid Abdo
- Department of Microbiology, Immunology and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA; (D.G.); (A.C.V.); (B.E.F.); (C.L.); (Z.A.)
| | - Rodolphe Barrangou
- Department of Food, Bioprocessing and Nutrition Sciences, North Carolina State University, Raleigh, NC 27695, USA; (M.P.); (Y.J.G.); (S.O.); (R.B.)
| | - Gregg A. Dean
- Department of Microbiology, Immunology and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA; (D.G.); (A.C.V.); (B.E.F.); (C.L.); (Z.A.)
| |
Collapse
|