1
|
Ma Y, Dong C, Kim JK, Zhu W, Wei L, Wang Y, Kang SM, Wang BZ. Impact of influenza immune imprinting on immune responses to subsequent vaccinations in mice. Vaccine 2024; 46:126670. [PMID: 39731808 DOI: 10.1016/j.vaccine.2024.126670] [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: 08/14/2024] [Revised: 09/23/2024] [Accepted: 12/21/2024] [Indexed: 12/30/2024]
Abstract
The immune memory imprinted during an individual's initial influenza exposure (influenza imprinting) has long-lasting effects on the host's response to subsequent influenza infections and vaccinations. Here, we investigate how different influenza virus imprinting impacts the immune responses to subunit, inactivated virus, and protein-based nanoparticle vaccines in Balb/c mice. Our results indicated a phylogenetic distance-dependent effect of influenza imprinting on subunit hemagglutinin (HA) or formalin-inactivated (FI) virus vaccine immunizations. Aichi (H3N2, group 2) HA (HA3) or FI-Aichi vaccination in mice imprinted with closely related Phili (H3N2) triggered significant Aichi-specific HAI antibody and balanced HA3-specific Th1/Th2 antibody immune responses, resulting in robust protection against Aichi. In contrast, HA3 vaccination in PR8 (H1N1, group 1) imprinted mice (PR8-2HA3) induced Th2-leaning responses comparable to those observed in mice without prior influenza immune imprinting (PBS-2HA3). However, subsequent heterosubtypic infections and vaccinations eliminated such effects on antibody subtype profiles. Nonetheless, initial virus exposure established a long-lasting capacity to produce HAI antibody responses against the imprinting strains. Moreover, Phili imprinting followed by HA3/NP nanocluster vaccination protected mice from Aichi infections and induced enhanced cross-reactive immunity. Our study highlights the significance of considering an individual's influenza exposure history when designing and evaluating the effectiveness of influenza vaccines.
Collapse
Affiliation(s)
- Yao Ma
- Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, 100 Piedmont Ave SE, Atlanta, GA 30303, USA
| | - Chunhong Dong
- Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, 100 Piedmont Ave SE, Atlanta, GA 30303, USA
| | - Joo Kyung Kim
- Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, 100 Piedmont Ave SE, Atlanta, GA 30303, USA
| | - Wandi Zhu
- Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, 100 Piedmont Ave SE, Atlanta, GA 30303, USA
| | - Lai Wei
- Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, 100 Piedmont Ave SE, Atlanta, GA 30303, USA
| | - Ye Wang
- Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, 100 Piedmont Ave SE, Atlanta, GA 30303, USA
| | - Sang-Moo Kang
- Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, 100 Piedmont Ave SE, Atlanta, GA 30303, USA
| | - Bao-Zhong Wang
- Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, 100 Piedmont Ave SE, Atlanta, GA 30303, USA.
| |
Collapse
|
2
|
Li Y, Rodriguez-Otero MR, Champion JA. Self-assembled protein vesicles as vaccine delivery platform to enhance antigen-specific immune responses. Biomaterials 2024; 311:122666. [PMID: 38879893 DOI: 10.1016/j.biomaterials.2024.122666] [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: 09/07/2023] [Revised: 05/18/2024] [Accepted: 06/11/2024] [Indexed: 06/18/2024]
Abstract
Self-assembling protein nanoparticles are beneficial platforms for enhancing the often weak and short-lived immune responses elicited by subunit vaccines. Their benefits include multivalency, similar sizes as pathogens and control of antigen orientation. Previously, the design, preparation, and characterization of self-assembling protein vesicles presenting fluorescent proteins and enzymes on the outer vesicle surface have been reported. Here, a full-size model antigen protein, ovalbumin (OVA), was genetically fused to the recombinant vesicle building blocks and incorporated into protein vesicles via self-assembly. Characterization of OVA protein vesicles showed room temperature stability and tunable size. Immunization of mice with OVA protein vesicles induced strong antigen-specific humoral and cellular immune responses. This work demonstrates the potential of protein vesicles as a modular platform for delivering full-size antigen proteins that can be extended to pathogen antigens to induce antigen specific immune responses.
Collapse
Affiliation(s)
- Yirui Li
- BioEngineering Program, Georgia Institute of Technology, USA
| | - Mariela R Rodriguez-Otero
- BioEngineering Program, Georgia Institute of Technology, USA; School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, USA
| | - Julie A Champion
- BioEngineering Program, Georgia Institute of Technology, USA; School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, USA.
| |
Collapse
|
3
|
Liu DJ, Zhong XQ, Ru YX, Zhao SL, Liu CC, Tang YB, Wu X, Zhang YS, Zhang HH, She JY, Wan MY, Li YW, Zheng HP, Deng L. Disulfide-stabilized trimeric hemagglutinin ectodomains provide enhanced heterologous influenza protection. Emerg Microbes Infect 2024; 13:2389095. [PMID: 39101691 PMCID: PMC11334750 DOI: 10.1080/22221751.2024.2389095] [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: 03/21/2024] [Revised: 07/05/2024] [Accepted: 08/01/2024] [Indexed: 08/06/2024]
Abstract
Influenza virus infection poses a continual menace to public health. Here, we developed soluble trimeric HA ectodomain vaccines by establishing interprotomer disulfide bonds in the stem region, which effectively preserve the native antigenicity of stem epitopes. The stable trimeric H1 ectodomain proteins exhibited higher thermal stabilities in comparison with unmodified HAs and showed strong binding activities towards a panel of anti-stem cross-reactive antibodies that recognize either interprotomer or intraprotomer epitopes. Negative stain transmission electron microscopy (TEM) analysis revealed the stable trimer architecture of the interprotomer disulfide-stapled WA11#5, NC99#2, and FLD#1 proteins as well as the irregular aggregation of unmodified HA molecules. Immunizations of mice with those trimeric HA ectodomain vaccines formulated with incomplete Freund's adjuvant elicited significantly more potent cross-neutralizing antibody responses and offered broader immuno-protection against lethal infections with heterologous influenza strains compared to unmodified HA proteins. Additionally, the findings of our study indicate that elevated levels of HA stem-specific antibody responses correlate with strengthened cross-protections. Our design strategy has proven effective in trimerizing HA ectodomains derived from both influenza A and B viruses, thereby providing a valuable reference for designing future influenza HA immunogens.
Collapse
Affiliation(s)
- De-Jian Liu
- Hunan Provincial Key Laboratory of Medical Virology, Institute of Pathogen Biology and Immunology, College of Biology, Hunan University, Changsha, People’s Republic of China
| | - Xiu-Qin Zhong
- Hunan Provincial Key Laboratory of Medical Virology, Institute of Pathogen Biology and Immunology, College of Biology, Hunan University, Changsha, People’s Republic of China
| | - Yan-Xia Ru
- School of Life Sciences, Southern University of Science and Technology, Shenzhen, People’s Republic of China
| | - Shi-Long Zhao
- Hunan Provincial Key Laboratory of Medical Virology, Institute of Pathogen Biology and Immunology, College of Biology, Hunan University, Changsha, People’s Republic of China
| | - Cui-Cui Liu
- Hunan Provincial Key Laboratory of Medical Virology, Institute of Pathogen Biology and Immunology, College of Biology, Hunan University, Changsha, People’s Republic of China
| | - Yi-Bo Tang
- Hunan Provincial Key Laboratory of Medical Virology, Institute of Pathogen Biology and Immunology, College of Biology, Hunan University, Changsha, People’s Republic of China
| | - Xuan Wu
- Hunan Provincial Key Laboratory of Medical Virology, Institute of Pathogen Biology and Immunology, College of Biology, Hunan University, Changsha, People’s Republic of China
| | - Yi-Shuai Zhang
- Bioinformatics Center, College of Biology, Hunan University, Changsha, People’s Republic of China
| | - Hui-Hui Zhang
- Bioinformatics Center, College of Biology, Hunan University, Changsha, People’s Republic of China
| | - Jia-Yue She
- Hunan Provincial Key Laboratory of Medical Virology, Institute of Pathogen Biology and Immunology, College of Biology, Hunan University, Changsha, People’s Republic of China
| | - Mu-Yang Wan
- Hunan Provincial Key Laboratory of Medical Virology, Institute of Pathogen Biology and Immunology, College of Biology, Hunan University, Changsha, People’s Republic of China
| | - Yao-Wang Li
- School of Life Sciences, Southern University of Science and Technology, Shenzhen, People’s Republic of China
| | - He-Ping Zheng
- Bioinformatics Center, College of Biology, Hunan University, Changsha, People’s Republic of China
| | - Lei Deng
- Hunan Provincial Key Laboratory of Medical Virology, Institute of Pathogen Biology and Immunology, College of Biology, Hunan University, Changsha, People’s Republic of China
- Beijing Weimiao Biotechnology Co., Ltd., Beijing, People’s Republic of China
| |
Collapse
|
4
|
Kim JK, Zhu W, Dong C, Wei L, Ma Y, Denning T, Kang SM, Wang BZ. Double-layered protein nanoparticles conjugated with truncated flagellin induce improved mucosal and systemic immune responses in mice. NANOSCALE HORIZONS 2024; 9:2016-2030. [PMID: 39240547 PMCID: PMC11493517 DOI: 10.1039/d4nh00287c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/07/2024]
Abstract
Influenza viral infection poses a severe risk to global public health. Considering the suboptimal protection provided by current influenza vaccines against circulating influenza A viruses, it is imperative to develop novel vaccine formulations to combat respiratory infections. Here, we report the development of an intranasally-administered, self-adjuvanted double-layered protein nanoparticle consisting of influenza nucleoprotein (NP) cores coated with hemagglutinin (HA) and a truncated form of bacterial flagellin (tFliC). Intranasal vaccination of these nanoparticles notably amplified both antigen-specific humoral and cellular immune responses in the systematic compartments. Elevated antigen-specific IgA and IgG levels in mucosal washes, along with increased lung-resident memory B cell populations, were observed in the respiratory system of the immunized mice. Furthermore, intranasal vaccination of tFliC-adjuvanted nanoparticles enhanced survival rates against homologous and heterologous H3N2 viral challenges. Intriguingly, mucosal slow delivery of the prime dose (by splitting the dose into 5 applications over 8 days) significantly enhanced germinal center reactions and effector T-cell populations in lung draining lymph nodes, therefore promoting the protective efficacy against heterologous influenza viral challenges compared to single-prime immunization. These findings highlight the potential of intranasal immunization with tFliC-adjuvanted protein nanoparticles to bolster mucosal and systemic immune responses, with a slow-delivery strategy offering a promising approach for combating influenza epidemics.
Collapse
Affiliation(s)
- Joo Kyung Kim
- Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, 100 Piedmont Ave SE, Atlanta, Georgia 30303, USA.
| | - Wandi Zhu
- Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, 100 Piedmont Ave SE, Atlanta, Georgia 30303, USA.
| | - Chunhong Dong
- Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, 100 Piedmont Ave SE, Atlanta, Georgia 30303, USA.
| | - Lai Wei
- Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, 100 Piedmont Ave SE, Atlanta, Georgia 30303, USA.
| | - Yao Ma
- Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, 100 Piedmont Ave SE, Atlanta, Georgia 30303, USA.
| | - Timothy Denning
- Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, 100 Piedmont Ave SE, Atlanta, Georgia 30303, USA.
| | - Sang-Moo Kang
- Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, 100 Piedmont Ave SE, Atlanta, Georgia 30303, USA.
| | - Bao-Zhong Wang
- Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, 100 Piedmont Ave SE, Atlanta, Georgia 30303, USA.
| |
Collapse
|
5
|
Shuklina M, Stepanova L, Ozhereleva O, Kovaleva A, Vidyaeva I, Korotkov A, Tsybalova L. Inserting CTL Epitopes of the Viral Nucleoprotein to Improve Immunogenicity and Protective Efficacy of Recombinant Protein against Influenza A Virus. BIOLOGY 2024; 13:801. [PMID: 39452110 PMCID: PMC11505154 DOI: 10.3390/biology13100801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 09/30/2024] [Accepted: 10/03/2024] [Indexed: 10/26/2024]
Abstract
Conserved influenza virus proteins, such as the hemagglutinin stem domain (HA2), nucleoprotein (NP), and matrix protein (M), are the main targets in the development of universal influenza vaccines. Previously, we constructed a recombinant vaccine protein Flg-HA2-2-4M2ehs containing the extracellular domain of the M2 protein (M2e) and the aa76-130 sequence of the second HA subunit as target antigens. It demonstrated immunogenicity and broad protection against influenza A viruses after intranasal and parenteral administration. This study shows that CD8+ epitopes of NP, inserted into a flagellin-fused protein carrying M2e and HA2, affect the post-vaccination immune humoral response to virus antigens without reducing protection. No differences were found between the two proteins in their ability to stimulate the formation of follicular Th in the spleen, which may contribute to a long-lasting antigen-specific humoral response. The data obtained on Balb/c mice suggest that the insertion of CTL NP epitopes into the flagellin-fused protein carrying M2e and HA2 reduces the antibody response to M2e and A/H3N2. In C57Bl6 mice, this stimulates the formation of NP-specific CD8+ Tem and virus-specific mono- and multifunctional CD4+ and CD8+ Tem in the spleen and completely protects mice from influenza virus subtypes A/H1N1pdm09 and A/H3N2.
Collapse
Affiliation(s)
- Marina Shuklina
- Smorodintsev Research Institute of Influenza, Ministry of Health of the Russian Federation, 15/17 Prof. Popova Str., St. Petersburg 197376, Russia
| | - Liudmila Stepanova
- Smorodintsev Research Institute of Influenza, Ministry of Health of the Russian Federation, 15/17 Prof. Popova Str., St. Petersburg 197376, Russia
| | | | | | | | | | | |
Collapse
|
6
|
Jones CH, Hauguel T, Beitelshees M, Davitt M, Welch V, Lindert K, Allen P, True JM, Dolsten M. Deciphering immune responses: a comparative analysis of influenza vaccination platforms. Drug Discov Today 2024; 29:104125. [PMID: 39097221 DOI: 10.1016/j.drudis.2024.104125] [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: 02/15/2024] [Revised: 06/21/2024] [Accepted: 07/29/2024] [Indexed: 08/05/2024]
Abstract
Influenza still poses a significant challenge due to its high mutation rates and the low effectiveness of traditional vaccines. At present, antibodies that neutralize the highly variable hemagglutinin antigen are a major driver of the observed variable protection. To decipher how influenza vaccines can be improved, an analysis of licensed vaccine platforms was conducted, contrasting the strengths and limitations of their different mechanisms of protection. Through this review, it is evident that these vaccines do not elicit the robust cellular immune response critical for protecting high-risk groups. Emerging platforms, such as RNA vaccines, that induce robust cellular responses that may be additive to the recognized mechanism of protection through hemagglutinin inhibition may overcome these constraints to provide broader, protective immunity. By combining both humoral and cellular responses, such platforms could help guide the future influenza vaccine development.
Collapse
Affiliation(s)
| | | | | | | | - Verna Welch
- Pfizer, Hudson Boulevard, New York, NY 10018, USA
| | | | - Pirada Allen
- Pfizer, Hudson Boulevard, New York, NY 10018, USA
| | - Jane M True
- Pfizer, Hudson Boulevard, New York, NY 10018, USA.
| | | |
Collapse
|
7
|
Ji G, Zhang Z, Wang X, Guo Q, Zhang E, Li C. Comprehensive evaluation of the mechanism of human adipose mesenchymal stem cells ameliorating liver fibrosis by transcriptomics and metabolomics analysis. Sci Rep 2024; 14:20035. [PMID: 39198546 PMCID: PMC11358327 DOI: 10.1038/s41598-024-70281-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Accepted: 08/14/2024] [Indexed: 09/01/2024] Open
Abstract
Liver fibrosis is a chronic liver disease with progressive wound healing reaction caused by liver injury. Currently, there is no FDA approved drugs for liver fibrosis. Human adipose mesenchymal stem cells (hADSCs) have shown remarkable therapeutic effects in liver diseases. However, few studies have evaluated the therapeutic role of hADSCs in liver fibrosis, and the detailed mechanism of action is unknown. Here, we investigated the in vitro and in vivo anti-fibrosis efficacy of hADSCs and identified important metabolic changes and detailed mechanisms through transcriptomic and metabolomic analyses. We found that hADSCs could inhibit the proliferation of activated hepatic stellate cells (HSCs), promote their apoptosis, and effectively inhibit the expression of pro-fibrotic protein. It can significantly reduce collagen deposition and liver injury, improve liver function and alleviate liver inflammation in cirrhotic mouse models. In addition, transcriptome analysis revealed that the key mechanism of hADSCs against liver fibrosis is the regulation of AGE-RAGE signaling pathway. Metabolic analysis showed that hADSCs influenced changes of metabolites in lipid metabolism. Therefore, our study shows that hADSCs could reduce the activation of hepatic stellate cells and inhibit the progression of liver fibrosis, which has important potential in the treatment of liver fibrosis as well as other refractory chronic liver diseases.
Collapse
Affiliation(s)
- Guibao Ji
- Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
- Department of Hepatobiliary-Pancreatic and Hernia Surgery, Wuhan Fourth Hospital, Wuhan, Hubei, People's Republic of China
| | - Zilong Zhang
- Department of Hepatobiliary-Pancreatic and Hernia Surgery, Wuhan Fourth Hospital, Wuhan, Hubei, People's Republic of China
| | - Xinze Wang
- Department of Trauma and Orthopedics, Wuhan Fourth Hospital, Wuhan, Hubei, People's Republic of China
| | - Qiuxia Guo
- Department of Gastroenterology Surgery, Wuhan Fourth Hospital, Wuhan, Hubei, People's Republic of China
| | - Erlei Zhang
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Hepatic Surgery Center, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China.
| | - Chuanjiang Li
- Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China.
- Division of Hepatobiliopancreatic Surgery, Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, People's Republic of China.
| |
Collapse
|
8
|
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
|
9
|
Zhou Y, Lin Z, Fang J, Wang Z, Guo J, Li G, Xu Q, Jin M, Chen H, Zou J, Zhou H. The recombinant vaccine of Lactobacillus plantarum elicits immune protection against H1N1 and H9N2 influenza virus infection. Int J Biol Macromol 2024; 275:133453. [PMID: 38942402 DOI: 10.1016/j.ijbiomac.2024.133453] [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: 05/06/2024] [Revised: 06/24/2024] [Accepted: 06/24/2024] [Indexed: 06/30/2024]
Abstract
Influenza A virus (IAV) causes annual epidemics and occasional pandemics, resulting in significant economic losses and numerous fatalities. Current vaccines, typically administered through injection, provide limited protection due to the frequent antigenic shift and drift of IAV strains. Therefore, the development of alternative broad-spectrum vaccine strategies is imperative. Lactic acid bacteria (LAB) represent promising candidates for vaccine engineering due to their low cost, high safety profile, and suitability for oral administration. In this study, we identified a strain of Lactobacillus plantarum (Lp) that is resistant to acid and bile salts and capable of colonizing the intestines of mice. Subsequently, we employed the RecE/T gene editing system to integrate headless hemagglutinins (mini-HA) into the genome of Lp, generating Lp-mini-HA-SP. Remarkably, immunization with Lp-mini-HA-SP elicited serum IgG antibody responses and conferred immune protection against H9N2 and H1N1 influenza virus challenges. Collectively, our findings offer a novel approach for the development of orally administered IAV vaccines and hold significant potential for future drug development endeavors.
Collapse
Affiliation(s)
- Yuanbao Zhou
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, PR China
| | - Zhipeng Lin
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, PR China
| | - Jiaqing Fang
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, PR China
| | - Zhihao Wang
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, PR China
| | - Jinli Guo
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, PR China
| | - Guohong Li
- Wuhan Keqian Biology Co., Ltd, Wuhan, Hubei, PR China
| | - Qiaoxia Xu
- Wuhan Keqian Biology Co., Ltd, Wuhan, Hubei, PR China
| | - Meilin Jin
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, PR China; Hubei Hongshan Laboratory, Wuhan, Hubei, PR China
| | - Huanchun Chen
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, PR China; Frontiers Science Center for Animal Breeding and Sustainable Production, Wuhan, Hubei, PR China; Hubei Hongshan Laboratory, Wuhan, Hubei, PR China
| | - Jiahui Zou
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, PR China.
| | - Hongbo Zhou
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, PR China; Frontiers Science Center for Animal Breeding and Sustainable Production, Wuhan, Hubei, PR China; Hubei Hongshan Laboratory, Wuhan, Hubei, PR China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei, PR China.
| |
Collapse
|
10
|
Tian Y, Deng Z, Chuai Z, Li C, Chang L, Sun F, Cao R, Yu H, Xiao R, Lu S, Xu Y, Yang P. A combination influenza mRNA vaccine candidate provided broad protection against diverse influenza virus challenge. Virology 2024; 596:110125. [PMID: 38805804 DOI: 10.1016/j.virol.2024.110125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 05/19/2024] [Accepted: 05/23/2024] [Indexed: 05/30/2024]
Abstract
Influenza viruses present a significant threat to global health. The production of a universal vaccine is considered essential due to the ineffectiveness of current seasonal influenza vaccines against mutant strains. mRNA technology offers new prospects in vaccinology, with various candidates for different infectious diseases currently in development and testing phases. In this study, we encapsulated a universal influenza mRNA vaccine. The vaccine encoded influenza hemagglutinin (HA), nucleoprotein (NP), and three tandem repeats of matrix protein 2 (3M2e). Twice-vaccinated mice exhibited strong humoral and cell-mediated immune responses in vivo. Notably, these immune responses led to a significant reduction in viral load of the lungs in challenged mice, and also conferred protection against future wild-type H1N1, H3N2, or H5N1 influenza virus challenges. Our findings suggest that this mRNA-universal vaccine strategy for influenza virus may be instrumental in mitigating the impact of future influenza pandemics.
Collapse
MESH Headings
- Animals
- Influenza Vaccines/immunology
- Influenza Vaccines/administration & dosage
- Influenza Vaccines/genetics
- Mice
- Orthomyxoviridae Infections/prevention & control
- Orthomyxoviridae Infections/immunology
- Orthomyxoviridae Infections/virology
- Antibodies, Viral/immunology
- Mice, Inbred BALB C
- mRNA Vaccines/immunology
- Influenza A Virus, H3N2 Subtype/immunology
- Influenza A Virus, H3N2 Subtype/genetics
- Hemagglutinin Glycoproteins, Influenza Virus/immunology
- Hemagglutinin Glycoproteins, Influenza Virus/genetics
- Viral Matrix Proteins/immunology
- Viral Matrix Proteins/genetics
- Female
- Influenza A Virus, H1N1 Subtype/immunology
- Influenza A Virus, H1N1 Subtype/genetics
- Influenza A Virus, H5N1 Subtype/immunology
- Influenza A Virus, H5N1 Subtype/genetics
- Vaccines, Synthetic/immunology
- Vaccines, Synthetic/administration & dosage
- Cross Protection/immunology
- Viral Load
- Lung/virology
- Lung/immunology
- Humans
- Viroporin Proteins
Collapse
Affiliation(s)
- Yuying Tian
- Faculty of Hepato-Pancreato-Biliary Surgery, Institute of Hepatobiliary Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China; School of Basic Medicine, Inner Mongolia Medical University, Hohhot, China
| | - Zhuoya Deng
- Faculty of Hepato-Pancreato-Biliary Surgery, Institute of Hepatobiliary Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Zhengran Chuai
- The Fifth Medical Center, Chinese PLA General Hospital, Beijing, 100039, China
| | - Cong Li
- Faculty of Hepato-Pancreato-Biliary Surgery, Institute of Hepatobiliary Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China; School of Basic Medicine, Inner Mongolia Medical University, Hohhot, China
| | - Liangzheng Chang
- Faculty of Hepato-Pancreato-Biliary Surgery, Institute of Hepatobiliary Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Fang Sun
- The Fifth Medical Center, Chinese PLA General Hospital, Beijing, 100039, China
| | - Rui Cao
- Faculty of Hepato-Pancreato-Biliary Surgery, Institute of Hepatobiliary Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China; School of Basic Medicine, Inner Mongolia Medical University, Hohhot, China
| | - Hongyu Yu
- Faculty of Hepato-Pancreato-Biliary Surgery, Institute of Hepatobiliary Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China; School of Basic Medicine, Inner Mongolia Medical University, Hohhot, China
| | - Ruixue Xiao
- Faculty of Hepato-Pancreato-Biliary Surgery, Institute of Hepatobiliary Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China; School of Basic Medicine, Inner Mongolia Medical University, Hohhot, China
| | - Shuai Lu
- Faculty of Hepato-Pancreato-Biliary Surgery, Institute of Hepatobiliary Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Yan Xu
- Faculty of Hepato-Pancreato-Biliary Surgery, Institute of Hepatobiliary Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Penghui Yang
- Faculty of Hepato-Pancreato-Biliary Surgery, Institute of Hepatobiliary Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China; School of Basic Medicine, Inner Mongolia Medical University, Hohhot, China.
| |
Collapse
|
11
|
Sangeetha Vijayan P, Xavier J, Valappil MP. A review of immune modulators and immunotherapy in infectious diseases. Mol Cell Biochem 2024; 479:1937-1955. [PMID: 37682390 DOI: 10.1007/s11010-023-04825-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 08/05/2023] [Indexed: 09/09/2023]
Abstract
The human immune system responds to harmful foreign invaders frequently encountered by the body and employs defense mechanisms to counteract such assaults. Various exogenous and endogenous factors play a prominent role in maintaining the balanced functioning of the immune system, which can result in immune suppression or immune stimulation. With the advent of different immune-modulatory agents, immune responses can be modulated or regulated to control infections and other health effects. Literature provides evidence on various immunomodulators from different sources and their role in modulating immune responses. Due to the limited efficacy of current drugs and the rise in drug resistance, there is a growing need for new therapies for infectious diseases. In this review, we aim to provide a comprehensive overview of different immune-modulating agents and immune therapies specifically focused on viral infectious diseases.
Collapse
Affiliation(s)
- P Sangeetha Vijayan
- Toxicology Division, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology [Govt. of India], Thiruvananthapuram, 695 012, Kerala, India
| | - Joseph Xavier
- Toxicology Division, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology [Govt. of India], Thiruvananthapuram, 695 012, Kerala, India
| | - Mohanan Parayanthala Valappil
- Toxicology Division, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology [Govt. of India], Thiruvananthapuram, 695 012, Kerala, India.
| |
Collapse
|
12
|
Li X, Xie Z, Wei Y, Li M, Zhang M, Luo S, Xie L. Recombinant Hemagglutinin Protein from H9N2 Avian Influenza Virus Exerts Good Immune Effects in Mice. Microorganisms 2024; 12:1552. [PMID: 39203394 PMCID: PMC11356462 DOI: 10.3390/microorganisms12081552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 07/25/2024] [Accepted: 07/27/2024] [Indexed: 09/03/2024] Open
Abstract
The H9N2 subtype of avian influenza virus (AIV) causes enormous economic losses and poses a significant threat to public health; the development of vaccines against avian influenza is ongoing. To study the immunogenicity of hemagglutinin (HA) protein, we constructed a recombinant pET-32a-HA plasmid, induced HA protein expression with isopropyl β-D-1-thiogalactopyranoside (IPTG), verified it by SDS-PAGE and Western blotting, and determined the sensitivity of the recombinant protein to acid and heat. Subsequently, mice were immunized with the purified HA protein, and the immunization effect was evaluated according to the hemagglutination inhibition (HI) titer, serum IgG antibody titer, and cytokine secretion level of the mice. The results showed that the molecular weight of the HA protein was approximately 84 kDa, and the protein existed in both soluble and insoluble forms; in addition, the HA protein exhibited good acid and thermal stability, the HI antibody titer reached 6 log2-8 log2, and the IgG-binding antibody titer was 1:1,000,000. Moreover, the levels of IL-2, IL-4, and IL-5 in the immunized mouse spleen cells were significantly increased compared with those in the control group. However, the levels of IL-1β, IL-6, IL-13, IFN-γ, IL-18, TNF-α, and GM-CSF were decreased in the immunized group. The recombinant HA protein utilized in this study exhibited good stability and exerted beneficial immune effects, providing a theoretical basis for further research on influenza vaccines.
Collapse
Affiliation(s)
- Xiaofeng Li
- GuangXi Key Laboratory of Veterinary Biotechnology, GuangXi Veterinary Research Institute, Nanning 530000, China; (X.L.); (Y.W.); (M.L.); (S.L.); (L.X.)
- Key Laboratory of China (GuangXi)-ASEAN Cross-Border Animal Disease Prevention and Control, Ministry of Agriculture and Rural Affairs of China, Nanning 530000, China
| | - Zhixun Xie
- GuangXi Key Laboratory of Veterinary Biotechnology, GuangXi Veterinary Research Institute, Nanning 530000, China; (X.L.); (Y.W.); (M.L.); (S.L.); (L.X.)
- Key Laboratory of China (GuangXi)-ASEAN Cross-Border Animal Disease Prevention and Control, Ministry of Agriculture and Rural Affairs of China, Nanning 530000, China
| | - You Wei
- GuangXi Key Laboratory of Veterinary Biotechnology, GuangXi Veterinary Research Institute, Nanning 530000, China; (X.L.); (Y.W.); (M.L.); (S.L.); (L.X.)
- Key Laboratory of China (GuangXi)-ASEAN Cross-Border Animal Disease Prevention and Control, Ministry of Agriculture and Rural Affairs of China, Nanning 530000, China
| | - Meng Li
- GuangXi Key Laboratory of Veterinary Biotechnology, GuangXi Veterinary Research Institute, Nanning 530000, China; (X.L.); (Y.W.); (M.L.); (S.L.); (L.X.)
- Key Laboratory of China (GuangXi)-ASEAN Cross-Border Animal Disease Prevention and Control, Ministry of Agriculture and Rural Affairs of China, Nanning 530000, China
| | - Minxiu Zhang
- GuangXi Key Laboratory of Veterinary Biotechnology, GuangXi Veterinary Research Institute, Nanning 530000, China; (X.L.); (Y.W.); (M.L.); (S.L.); (L.X.)
- Key Laboratory of China (GuangXi)-ASEAN Cross-Border Animal Disease Prevention and Control, Ministry of Agriculture and Rural Affairs of China, Nanning 530000, China
| | - Sisi Luo
- GuangXi Key Laboratory of Veterinary Biotechnology, GuangXi Veterinary Research Institute, Nanning 530000, China; (X.L.); (Y.W.); (M.L.); (S.L.); (L.X.)
- Key Laboratory of China (GuangXi)-ASEAN Cross-Border Animal Disease Prevention and Control, Ministry of Agriculture and Rural Affairs of China, Nanning 530000, China
| | - Liji Xie
- GuangXi Key Laboratory of Veterinary Biotechnology, GuangXi Veterinary Research Institute, Nanning 530000, China; (X.L.); (Y.W.); (M.L.); (S.L.); (L.X.)
- Key Laboratory of China (GuangXi)-ASEAN Cross-Border Animal Disease Prevention and Control, Ministry of Agriculture and Rural Affairs of China, Nanning 530000, China
| |
Collapse
|
13
|
Dong C, Zhu W, Wei L, Kim JK, Ma Y, Kang SM, Wang BZ. Enhancing cross-protection against influenza by heterologous sequential immunization with mRNA LNP and protein nanoparticle vaccines. Nat Commun 2024; 15:5800. [PMID: 38987276 PMCID: PMC11237032 DOI: 10.1038/s41467-024-50087-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 06/27/2024] [Indexed: 07/12/2024] Open
Abstract
Enhancing influenza vaccine cross-protection is imperative to alleviate the significant public health burden of influenza. Heterologous sequential immunization may synergize diverse vaccine formulations and routes to improve vaccine potency and breadth. Here we investigate the effects of immunization strategies on the generation of cross-protective immune responses in female Balb/c mice, utilizing mRNA lipid nanoparticle (LNP) and protein-based PHC nanoparticle vaccines targeting influenza hemagglutinin. Our findings emphasize the crucial role of priming vaccination in shaping Th bias and immunodominance hierarchies. mRNA LNP prime favors Th1-leaning responses, while PHC prime elicits Th2-skewing responses. We demonstrate that cellular and mucosal immune responses are pivotal correlates of cross-protection against influenza. Notably, intranasal PHC immunization outperforms its intramuscular counterpart in inducing mucosal immunity and conferring cross-protection. Sequential mRNA LNP prime and intranasal PHC boost demonstrate optimal cross-protection against antigenically drifted and shifted influenza strains. Our study offers valuable insights into tailoring immunization strategies to optimize influenza vaccine effectiveness.
Collapse
Affiliation(s)
- Chunhong Dong
- Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, 100 Piedmont Ave SE, Atlanta, GA, 30303, USA
| | - Wandi Zhu
- Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, 100 Piedmont Ave SE, Atlanta, GA, 30303, USA
| | - Lai Wei
- Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, 100 Piedmont Ave SE, Atlanta, GA, 30303, USA
| | - Joo Kyung Kim
- Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, 100 Piedmont Ave SE, Atlanta, GA, 30303, USA
| | - Yao Ma
- Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, 100 Piedmont Ave SE, Atlanta, GA, 30303, USA
| | - Sang-Moo Kang
- Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, 100 Piedmont Ave SE, Atlanta, GA, 30303, USA
| | - Bao-Zhong Wang
- Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, 100 Piedmont Ave SE, Atlanta, GA, 30303, USA.
| |
Collapse
|
14
|
Kim KH, Bhatnagar N, Subbiah J, Liu R, Pal SS, Raha JR, Grovenstein P, Shin CH, Wang BZ, Kang SM. Cross-protection against influenza viruses by chimeric M2e-H3 stalk protein or multi-subtype neuraminidase plus M2e virus-like particle vaccine in ferrets. Virology 2024; 595:110097. [PMID: 38685171 PMCID: PMC11110495 DOI: 10.1016/j.virol.2024.110097] [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: 12/26/2023] [Revised: 04/19/2024] [Accepted: 04/23/2024] [Indexed: 05/02/2024]
Abstract
Current influenza vaccine is not effective in providing cross-protection against variants. We evaluated the immunogenicity and efficacy of multi-subtype neuraminidase (NA) and M2 ectodomain virus-like particle (m-cNA-M2e VLP) and chimeric M2e-H3 stalk protein vaccines (M2e-H3 stalk) in ferrets. Our results showed that ferrets with recombinant m-cNA-M2e VLP or M2e-H3 stalk vaccination induced multi-vaccine antigen specific IgG antibodies (M2e, H3 stalk, NA), NA inhibition, antibody-secreting cells, and IFN-γ secreting cell responses. Ferrets immunized with either m-cNA-M2e VLP or M2e-H3 stalk vaccine were protected from H1N1 and H3N2 influenza viruses by lowering viral titers in nasal washes, trachea, and lungs after challenge. Vaccinated ferret antisera conferred broad humoral immunity in naïve mice. Our findings provide evidence that immunity to M2e and HA-stalk or M2e plus multi-subtype NA proteins induces cross-protection in ferrets.
Collapse
Affiliation(s)
- Ki-Hye Kim
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia, USA
| | - Noopur Bhatnagar
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia, USA
| | - Jeeva Subbiah
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia, USA
| | - Rong Liu
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia, USA
| | - Surya Sekhar Pal
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia, USA
| | - Jannatul Ruhan Raha
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia, USA
| | - Phillip Grovenstein
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia, USA
| | - Chong Hyun Shin
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia, USA
| | - Bao-Zhong Wang
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia, USA
| | - Sang-Moo Kang
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia, USA.
| |
Collapse
|
15
|
Nie J, Wang Q, Li C, Zhou Y, Yao X, Xu L, Chang Y, Ding F, Sun L, Zhan L, Zhu L, Xie K, Wang X, Shi Y, Zhao Q, Shan Y. Self-Assembled Multiepitope Nanovaccine Provides Long-Lasting Cross-Protection against Influenza Virus. Adv Healthc Mater 2024; 13:e2303531. [PMID: 37983728 DOI: 10.1002/adhm.202303531] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Indexed: 11/22/2023]
Abstract
Seasonal influenza vaccines typically provide strain-specific protection and are reformulated annually, which is a complex and time-consuming process. Multiepitope vaccines, combining multiple conserved antigenic epitopes from a pathogen, can trigger more robust, diverse, and effective immune responses, providing a potential solution. However, their practical application is hindered by low immunogenicity and short-term effectiveness. In this study, multiple linear epitopes from the conserved stem domain of hemagglutinin and the ectodomain of matrix protein 2 are combined with the Helicobacter pylori ferritin, a stable self-assembled nanoplatform, to develop an influenza multiepitope nanovaccine, named MHF. MHF is prokaryotically expressed in a soluble form and self-assembles into uniform nanoparticles. The subcutaneous immunization of mice with adjuvanted MHF induces cross-reactive neutralizing antibodies, antibody-dependent cell-mediated cytotoxicity, and cellular immunity, offering complete protection against H3N2 as well as partial protection against H1N1. Importantly, the vaccine cargo delivered by ferritin triggers epitope-specific memory B-cell responses, with antibody level persisting for over 6 months post-immunization. These findings indicate that self-assembled multiepitope nanovaccines elicit potent and long-lasting immune responses while significantly reducing the risk of vaccine escape mutants, and offer greater practicality in terms of scalable manufacturing and genetic manipulability, presenting a promising and effective strategy for future vaccine development.
Collapse
Affiliation(s)
- Jiaojiao Nie
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, 2699 Qianjin Street, Changchun, 130012, China
- Cancer Centre, Faculty of Health Sciences, University of Macau, Taipa, Macau, 519000, China
| | - Qingyu Wang
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, 2699 Qianjin Street, Changchun, 130012, China
| | - Chenxi Li
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, 2699 Qianjin Street, Changchun, 130012, China
| | - Yongfei Zhou
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, 2699 Qianjin Street, Changchun, 130012, China
| | - Xin Yao
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, 2699 Qianjin Street, Changchun, 130012, China
| | - Lipeng Xu
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, 2699 Qianjin Street, Changchun, 130012, China
| | - Yaotian Chang
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, 2699 Qianjin Street, Changchun, 130012, China
| | - Fan Ding
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, 2699 Qianjin Street, Changchun, 130012, China
| | - Lulu Sun
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, 2699 Qianjin Street, Changchun, 130012, China
| | - Li Zhan
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, 2699 Qianjin Street, Changchun, 130012, China
| | - Lvzhou Zhu
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, 2699 Qianjin Street, Changchun, 130012, China
| | - Kunpeng Xie
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, 2699 Qianjin Street, Changchun, 130012, China
| | - Xu Wang
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, 2699 Qianjin Street, Changchun, 130012, China
| | - Yuhua Shi
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, 2699 Qianjin Street, Changchun, 130012, China
| | - Qi Zhao
- Cancer Centre, Faculty of Health Sciences, University of Macau, Taipa, Macau, 519000, China
- MoE Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macau SAR, 519000, China
| | - Yaming Shan
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, 2699 Qianjin Street, Changchun, 130012, China
- Key Laboratory for Molecular Enzymology and Engineering, The Ministry of Education, School of Life Sciences, Jilin University, 2699 Qianjin Street, Changchun, 130012, China
| |
Collapse
|
16
|
Lim CML, Komarasamy TV, Adnan NAAB, Radhakrishnan AK, Balasubramaniam VRMT. Recent Advances, Approaches and Challenges in the Development of Universal Influenza Vaccines. Influenza Other Respir Viruses 2024; 18:e13276. [PMID: 38513364 PMCID: PMC10957243 DOI: 10.1111/irv.13276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 02/21/2024] [Accepted: 02/24/2024] [Indexed: 03/23/2024] Open
Abstract
Every year, influenza virus infections cause significant morbidity and mortality worldwide. They pose a substantial burden of disease, in terms of not only health but also the economy. Owing to the ability of influenza viruses to continuously evolve, annual seasonal influenza vaccines are necessary as a prophylaxis. However, current influenza vaccines against seasonal strains have limited effectiveness and require yearly reformulation due to the virus undergoing antigenic drift or shift. Vaccine mismatches are common, conferring suboptimal protection against seasonal outbreaks, and the threat of the next pandemic continues to loom. Therefore, there is a great need to develop a universal influenza vaccine (UIV) capable of providing broad and durable protection against all influenza virus strains. In the quest to develop a UIV that would obviate the need for annual vaccination and formulation, a multitude of strategies is currently underway. Promising approaches include targeting the highly conserved epitopes of haemagglutinin (HA), neuraminidase (NA), M2 extracellular domain (M2e) and internal proteins of the influenza virus. The identification and characterization of broadly neutralizing antibodies (bnAbs) targeting conserved regions of the viral HA protein, in particular, have provided important insight into novel vaccine designs and platforms. This review discusses universal vaccine approaches presently under development, with an emphasis on those targeting the highly conserved stalk of the HA protein, recent technological advancements used and the future prospects of a UIV in terms of its advantages, developmental obstacles and potential shortcomings.
Collapse
Affiliation(s)
- Caryn Myn Li Lim
- Infection and Immunity Research Strength, Jeffrey Cheah School of Medicine & Health SciencesMonash University MalaysiaBandar SunwayMalaysia
| | - Thamil Vaani Komarasamy
- Infection and Immunity Research Strength, Jeffrey Cheah School of Medicine & Health SciencesMonash University MalaysiaBandar SunwayMalaysia
| | - Nur Amelia Azreen Binti Adnan
- Infection and Immunity Research Strength, Jeffrey Cheah School of Medicine & Health SciencesMonash University MalaysiaBandar SunwayMalaysia
| | - Ammu Kutty Radhakrishnan
- Infection and Immunity Research Strength, Jeffrey Cheah School of Medicine & Health SciencesMonash University MalaysiaBandar SunwayMalaysia
| | - Vinod R. M. T. Balasubramaniam
- Infection and Immunity Research Strength, Jeffrey Cheah School of Medicine & Health SciencesMonash University MalaysiaBandar SunwayMalaysia
| |
Collapse
|
17
|
Nie J, Zhou Y, Ding F, Liu X, Yao X, Xu L, Chang Y, Li Z, Wang Q, Zhan L, Zhu L, Xie K, Li C, Shi Y, Zhao Q, Shan Y. Self-adjuvant multiepitope nanovaccine based on ferritin induced long-lasting and effective mucosal immunity against H3N2 and H1N1 viruses in mice. Int J Biol Macromol 2024; 259:129259. [PMID: 38191112 DOI: 10.1016/j.ijbiomac.2024.129259] [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: 01/02/2024] [Accepted: 01/03/2024] [Indexed: 01/10/2024]
Abstract
The influenza A virus (IAV) is a ubiquitous and continuously evolving respiratory pathogen. The intranasal vaccination mimicking natural infections is an attractive strategy for controlling IAVs. Multiepitope vaccines accurately targeting multiple conserved domains have the potential to broaden the protective scope of current seasonal influenza vaccines and reduce the risk of generating escape mutants. Here, multiple linear epitopes from the matrix protein 2 ectodomain (M2e) and the hemagglutinin stem domain (HA2) are fused with the Helicobacter pylori ferritin, a self-assembled nanocarrier and mucosal adjuvant, to develop a multiepitope nanovaccine. Through intranasal delivery, the prokaryotically expressed multiepitope nanovaccine elicits long-lasting mucosal immunity, broad humoral immunity, and robust cellular immunity without any adjuvants, and confers complete protection against H3N2 and H1N1 subtypes of IAV in mice. Importantly, this intranasal multiepitope nanovaccine triggers memory B-cell responses, resulting in secretory immunoglobulin A (sIgA) and serum immunoglobulin G (IgG) levels persisting for more than five months post-immunization. Therefore, this intranasal ferritin-based multiepitope nanovaccine represents a promising approach to combating respiratory pathogens.
Collapse
Affiliation(s)
- Jiaojiao Nie
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, Jilin 130012, China; Cancer Centre, Faculty of Health Sciences, University of Macau, Taipa, Macau
| | - Yongfei Zhou
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, Jilin 130012, China
| | - Fan Ding
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, Jilin 130012, China
| | - Xiaoxi Liu
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, Jilin 130012, China
| | - Xin Yao
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, Jilin 130012, China
| | - Lipeng Xu
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, Jilin 130012, China
| | - Yaotian Chang
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, Jilin 130012, China
| | - Zeyu Li
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, Jilin 130012, China
| | - Qingyu Wang
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, Jilin 130012, China
| | - Li Zhan
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, Jilin 130012, China
| | - Lvzhou Zhu
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, Jilin 130012, China
| | - Kunpeng Xie
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, Jilin 130012, China
| | - Chenxi Li
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, Jilin 130012, China
| | - Yuhua Shi
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, Jilin 130012, China
| | - Qi Zhao
- Cancer Centre, Faculty of Health Sciences, University of Macau, Taipa, Macau; MoE Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macau
| | - Yaming Shan
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, Jilin 130012, China; Key Laboratory for Molecular Enzymology and Engineering, The Ministry of Education, School of Life Sciences, Jilin University, Changchun, Jilin 130012, China.
| |
Collapse
|
18
|
Li R, Chang Z, Liu H, Wang Y, Li M, Chen Y, Fan L, Wang S, Sun X, Liu S, Cheng A, Ding P, Zhang G. Double-layered N-S1 protein nanoparticle immunization elicits robust cellular immune and broad antibody responses against SARS-CoV-2. J Nanobiotechnology 2024; 22:44. [PMID: 38291444 PMCID: PMC10825999 DOI: 10.1186/s12951-024-02293-y] [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: 12/12/2023] [Accepted: 01/02/2024] [Indexed: 02/01/2024] Open
Abstract
BACKGROUND The COVID-19 pandemic is a persistent global threat to public health. As for the emerging variants of SARS-CoV-2, it is necessary to develop vaccines that can induce broader immune responses, particularly vaccines with weak cellular immunity. METHODS In this study, we generated a double-layered N-S1 protein nanoparticle (N-S1 PNp) that was formed by desolvating N protein into a protein nanoparticle as the core and crosslinking S1 protein onto the core surface against SARS-CoV-2. RESULTS Vaccination with N-S1 PNp elicited robust humoral and vigorous cellular immune responses specific to SARS-CoV-2 in mice. Compared to soluble protein groups, the N-S1 PNp induced a higher level of humoral response, as evidenced by the ability of S1-specific antibodies to block hACE2 receptor binding and neutralize pseudovirus. Critically, N-S1 PNp induced Th1-biased, long-lasting, and cross-neutralizing antibodies, which neutralized the variants of SARS-CoV-2 with minimal loss of activity. N-S1 PNp induced strong responses of CD4+ and CD8+ T cells, mDCs, Tfh cells, and GCs B cells in spleens. CONCLUSIONS These results demonstrate that N-S1 PNp vaccination is a practical approach for promoting protection, which has the potential to counteract the waning immune responses against SARS-CoV-2 variants and confer broad efficacy against future new variants. This study provides a new idea for the design of next-generation SARS-CoV-2 vaccines based on the B and T cells response coordination.
Collapse
Affiliation(s)
- Ruiqi Li
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
- School of Advanced Agricultural Sciences , Peking University, Beijing, 100080, China
- Longhu Laboratory, Zhengzhou, 450046, China
- Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou, 450002, China
| | - Zejie Chang
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
- Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou, 450002, China
- College of Animal Medicine, Henan Agricultural University, Zhengzhou, 450046, China
| | - Hongliang Liu
- School of Life Sciences , Zhengzhou University, Zhengzhou, 450001, China
| | - Yanan Wang
- Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou, 450002, China
- College of Animal Medicine, Henan Agricultural University, Zhengzhou, 450046, China
| | - Minghui Li
- Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou, 450002, China
- College of Animal Medicine, Henan Agricultural University, Zhengzhou, 450046, China
| | - Yilan Chen
- Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou, 450002, China
| | - Lu Fan
- Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou, 450002, China
| | - Siqiao Wang
- Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou, 450002, China
| | - Xueke Sun
- Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou, 450002, China
- College of Animal Medicine, Henan Agricultural University, Zhengzhou, 450046, China
| | - Siyuan Liu
- Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou, 450002, China
- College of Animal Medicine, Henan Agricultural University, Zhengzhou, 450046, China
| | - Anchun Cheng
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Peiyang Ding
- School of Life Sciences , Zhengzhou University, Zhengzhou, 450001, China.
| | - Gaiping Zhang
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China.
- School of Advanced Agricultural Sciences , Peking University, Beijing, 100080, China.
- Longhu Laboratory, Zhengzhou, 450046, China.
- Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou, 450002, China.
- College of Animal Medicine, Henan Agricultural University, Zhengzhou, 450046, China.
- School of Life Sciences , Zhengzhou University, Zhengzhou, 450001, China.
| |
Collapse
|
19
|
Dong C, Ma Y, Zhu W, Wang Y, Kim J, Wei L, Gill HS, Kang SM, Wang BZ. Influenza immune imprinting synergizes PEI-HA/CpG nanoparticle vaccine protection against heterosubtypic infection in mice. Vaccine 2024; 42:111-119. [PMID: 38097456 PMCID: PMC10842698 DOI: 10.1016/j.vaccine.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: 08/14/2023] [Revised: 12/09/2023] [Accepted: 12/10/2023] [Indexed: 01/01/2024]
Abstract
The first influenza virus infection (imprinting) can lead to long-term immune memory and influence subsequent vaccinations and infections. Previously, we reported a polyethyleneimine (PEI)-Aichi hemagglutinin (HA)/CpG (PHC) nanoparticle with cross-protective potential against homologous and heterologous influenza strains. Here we studied how influenza immune imprinting influences the antibody responses to the PHC vaccination and the protection against heterosubtypic virus challenges. We found that pre-existing virus immunity can maintain or synergize the vaccine-induced antibody titers, depending on the imprinting virus HA phylogenetic group. The HA group 1 virus (PR8, H1N1)-imprinted mice displayed comparable antigen-specific antibody responses to those without imprinting post-PHC vaccination. In contrast, the group 2 virus (Phi, H3N2)-imprinted mice showed significantly more robust and balanced antibodies post-vaccination, conferring complete protection against body weight loss and lung inflammation upon heterosubtypic reassortant A/Shanghai/2/2013 (rSH, H7N9) virus challenge. Our findings suggest that influenza imprinting from the same HA phylogenetic group can synergize subsequent vaccination, conferring heterosubtypic protection.
Collapse
Affiliation(s)
- Chunhong Dong
- Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, 100 Piedmont Ave SE, Atlanta, GA 30303, USA
| | - Yao Ma
- Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, 100 Piedmont Ave SE, Atlanta, GA 30303, USA
| | - Wandi Zhu
- Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, 100 Piedmont Ave SE, Atlanta, GA 30303, USA
| | - Ye Wang
- Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, 100 Piedmont Ave SE, Atlanta, GA 30303, USA
| | - Joo Kim
- Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, 100 Piedmont Ave SE, Atlanta, GA 30303, USA
| | - Lai Wei
- Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, 100 Piedmont Ave SE, Atlanta, GA 30303, USA
| | - Harvinder Singh Gill
- Department of Chemical Engineering, Texas Tech University, Lubbock, TX 79409, USA
| | - Sang-Moo Kang
- Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, 100 Piedmont Ave SE, Atlanta, GA 30303, USA
| | - Bao-Zhong Wang
- Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, 100 Piedmont Ave SE, Atlanta, GA 30303, USA.
| |
Collapse
|
20
|
Studniski M, Stumvoll K, Kromm M, Ssematimba A, Marusak R, Xing Z, Halvorson D, Culhane M, Cardona C. Vaccination of Poultry Against Influenza. Avian Dis 2024; 67:402-409. [PMID: 38300659 DOI: 10.1637/aviandiseases-d-23-99995] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 08/09/2023] [Indexed: 02/02/2024]
Abstract
The complexity of influenza A virus (IAV) infections in avian hosts leads to equally complex scenarios for the vaccination of poultry. Vaccination against avian influenza strains can be used to prevent infections from sources with a single strain of IAV. It has been used as a part of outbreak control strategies as well as a way to maintain production for both low and high pathogenicity outbreaks. Unlike other viral pathogens of birds, avian influenza vaccination when used against highly pathogenic avian influenza virus, is tied to international trade and thus is not freely available for use without specific permission.
Collapse
Affiliation(s)
| | | | | | - Amos Ssematimba
- Department of Mathematics, Gulu University, Gulu, Uganda
- College of Veterinary Medicine, University of Minnesota, St. Paul, MN 55108
| | - Rosemary Marusak
- College of Veterinary Medicine, University of Minnesota, St. Paul, MN 55108
| | - Zheng Xing
- College of Veterinary Medicine, University of Minnesota, St. Paul, MN 55108
| | - Dave Halvorson
- College of Veterinary Medicine, University of Minnesota, St. Paul, MN 55108
| | - Marie Culhane
- College of Veterinary Medicine, University of Minnesota, St. Paul, MN 55108
| | - Carol Cardona
- College of Veterinary Medicine, University of Minnesota, St. Paul, MN 55108,
| |
Collapse
|
21
|
Park J, Champion JA. Development of Self-Assembled Protein Nanocage Spatially Functionalized with HA Stalk as a Broadly Cross-Reactive Influenza Vaccine Platform. ACS NANO 2023; 17:25045-25060. [PMID: 38084728 PMCID: PMC10753887 DOI: 10.1021/acsnano.3c07669] [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: 08/15/2023] [Revised: 11/29/2023] [Accepted: 12/01/2023] [Indexed: 12/27/2023]
Abstract
There remains a need for the development of a universal influenza vaccine, as current seasonal influenza vaccines exhibit limited protection against mismatched, mutated, or pandemic influenza viruses. A desirable approach to developing an effective universal influenza vaccine is the incorporation of highly conserved antigens in a multivalent scaffold that enhances their immunogenicity. Here, we develop a broadly cross-reactive influenza vaccine by functionalizing self-assembled protein nanocages (SAPNs) with multiple copies of the hemagglutinin stalk on the outer surface and matrix protein 2 ectodomain on the inner surface. SAPNs were generated by engineering short coiled coils, and the design was simulated by MD GROMACS. Due to the short sequences, off-target immune responses against empty SAPN scaffolds were not seen in immunized mice. Vaccination with the multivalent SAPNs induces high levels of broadly cross-reactive antibodies of only external antigens, demonstrating tight spatial control over the designed antigen placement. This work demonstrates the use of SAPNs as a potential influenza vaccine.
Collapse
Affiliation(s)
- Jaeyoung Park
- School of Chemical and Biomolecular
Engineering, Georgia Institute of Technology, 950 Atlantic Dr. NW, Atlanta, Georgia 30332-2000, United States
| | - Julie A. Champion
- School of Chemical and Biomolecular
Engineering, Georgia Institute of Technology, 950 Atlantic Dr. NW, Atlanta, Georgia 30332-2000, United States
| |
Collapse
|
22
|
Park J, Zhang Z, Belinskaya T, Tsoras AN, Chao CC, Jiang L, Champion JA. Dual-Antigen Subunit Vaccine Nanoparticles for Scrub Typhus. Pathogens 2023; 12:1390. [PMID: 38133275 PMCID: PMC10745692 DOI: 10.3390/pathogens12121390] [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: 10/26/2023] [Revised: 11/22/2023] [Accepted: 11/24/2023] [Indexed: 12/23/2023] Open
Abstract
Orientia tsutsugamushi is the causative pathogen of scrub typhus, an acute febrile disease prevalent in the Asia-Pacific region that is spread to people through chigger bites. Despite the emerging threat, there is no currently available vaccine against O. tsutsugamushi. Here, we developed dual-antigen subunit vaccine nanoparticles using recombinant 47 kD and 56 kD proteins, which are immunogenic outer membrane antigens of O. tsutsugamushi. The biocompatible protein vaccine nanoparticles were formed via desolvation of r56 or r47E antigens with acetone, coating with an additional layer of the 56 kD protein, and stabilization with reducible homobifunctional DTSSP and heterobifunctional SDAD crosslinkers. The dual-antigen subunit vaccine nanoparticles significantly improved antigen-specific antibody responses in vaccinated mice. Most importantly, the dual-antigen nanoparticles coated with an additional layer of the 56 kD protein were markedly more immunogenic than soluble antigens or single-antigen nanoparticles in the context of cellular immune responses. Given the significance of cellular immune responses for protection against O. tsutsugamushi, these results demonstrate the potent immunogenicity of dual-layered antigen nanoparticles and their potential as a promising strategy for developing vaccines against scrub typhus.
Collapse
Affiliation(s)
- Jaeyoung Park
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, 950 Atlantic Dr. NW, Atlanta, GA 30332, USA; (J.P.); (A.N.T.)
| | - Zhiwen Zhang
- Henry Jackson Foundation for the Advancement of Military Medicine, 6720A Rockledge Dr, Bethesda, MD 20817, USA; (Z.Z.); (T.B.)
- Naval Medical Research Center, 503 Robert Grant Ave., Silver Spring, MD 20910, USA;
| | - Tatyana Belinskaya
- Henry Jackson Foundation for the Advancement of Military Medicine, 6720A Rockledge Dr, Bethesda, MD 20817, USA; (Z.Z.); (T.B.)
- Naval Medical Research Center, 503 Robert Grant Ave., Silver Spring, MD 20910, USA;
| | - Alexandra N. Tsoras
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, 950 Atlantic Dr. NW, Atlanta, GA 30332, USA; (J.P.); (A.N.T.)
| | - Chien-Chung Chao
- Naval Medical Research Center, 503 Robert Grant Ave., Silver Spring, MD 20910, USA;
| | - Le Jiang
- Henry Jackson Foundation for the Advancement of Military Medicine, 6720A Rockledge Dr, Bethesda, MD 20817, USA; (Z.Z.); (T.B.)
- Naval Medical Research Center, 503 Robert Grant Ave., Silver Spring, MD 20910, USA;
| | - Julie A. Champion
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, 950 Atlantic Dr. NW, Atlanta, GA 30332, USA; (J.P.); (A.N.T.)
| |
Collapse
|
23
|
Hao K, Wang Y, Zhu B, Yu F, Zhao Z, Wang GX. Recombinant surface display vaccine enhances the immersion immune effect against grass carp reovirus in grass carp (Ctenopharyngodon idella). FISH & SHELLFISH IMMUNOLOGY 2023; 142:109160. [PMID: 37858787 DOI: 10.1016/j.fsi.2023.109160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 10/10/2023] [Accepted: 10/10/2023] [Indexed: 10/21/2023]
Abstract
Grass carp (Ctenopharyngodon idella) is subject to a hemorrhagic disease caused by grass carp reovirus (GCRV), which can lead to mass mortality in grass carp culture, causing significant economic loss. Vaccination is the most promising strategy for the prevention of infectious diseases. Immersion vaccination is considered the most effective disease prevention method for juvenile fish because it can be implemented on many fish at once and administered without causing stress. However, immune responses by immersion vaccination are markedly less robust due to the skin barrier and insufficient antigen uptake. The display of heterologous proteins on the cell surface has been explored as a delivery system for viral antigens in veterinary and human vaccine studies. To improve the efficacy of the immersion vaccine, the major capsid protein (VP7) of GCRV was co-displayed with Aeromonas hydrophila outer membrane protein a (OmpA) and major adhesion protein (Mah) on the outer membrane surface of nonpathogenic Escherichia coli BL21 using the anchoring motif of ice-nucleation protein (Inp). The immune responses and protection efficiency against GCRV infection via both the injection and immersion routes were evaluated. The results indicated that the activities of anti-oxidant enzymes (ACP, AKP, SOD and T-AOC), as well as the expression of immune-related genes (TNF-α, IL-1β, MHCI and IgM) and specific VP7 antibody levels, were strongly increased in the grass carp from 7 to 21 days post-injection inoculation in a dose dependent manner. The cumulative mortality rates of injection-vaccinated groups were much lower than those of the control group after the GCRV challenge, and the relative percent survival (RPS) was greater than 80 %. Vitally, the surface co-display of vp7-Mah protein conferred marked protection to grass carp against GCRV infection after immersion administration (RPS >50 %); this was consistent with the production of high level of specific serum antibodies, non-specific immune responses, and the expression of immune-related genes. Moreover, the invasion analysis further showed that surface co-display of the vp7-Mah protein indeed significantly improved the invasion of E. coli BL21 (DE3) in vitro. Altogether, this study demonstrated that surface display GCRV core antigen vaccine system accompanied by invasion component from aquatic pathogenic microorganism is an effective prophylactic against GCRV viral diseases via the immersion administration approach.
Collapse
Affiliation(s)
- Kai Hao
- College of Oceanography, Hohai University, Nanjing, 210098, PR China; College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi Province, 712100, PR China.
| | - Yu Wang
- College of Oceanography, Hohai University, Nanjing, 210098, PR China
| | - Bin Zhu
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi Province, 712100, PR China
| | - Fei Yu
- College of Oceanography, Hohai University, Nanjing, 210098, PR China
| | - Zhe Zhao
- College of Oceanography, Hohai University, Nanjing, 210098, PR China
| | - Gao-Xue Wang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi Province, 712100, PR China.
| |
Collapse
|
24
|
Li M, Chen C, Wang X, Guo P, Feng H, Zhang X, Zhang W, Gu C, Zhu J, Wen G, Feng Y, Xiao L, Peng G, Rao VB, Tao P. T4 bacteriophage nanoparticles engineered through CRISPR provide a versatile platform for rapid development of flu mucosal vaccines. Antiviral Res 2023; 217:105688. [PMID: 37516153 DOI: 10.1016/j.antiviral.2023.105688] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 07/18/2023] [Accepted: 07/25/2023] [Indexed: 07/31/2023]
Abstract
Vaccines that trigger mucosal immune responses at the entry portals of pathogens are highly desired. Here, we showed that antigen-decorated nanoparticle generated through CRISPR engineering of T4 bacteriophage can serve as a universal platform for the rapid development of mucosal vaccines. Insertion of Flu viral M2e into phage T4 genome through fusion to Soc (Small Outer Capsid protein) generated a recombinant phage, and the Soc-M2e proteins self-assembled onto phage capsids to form 3M2e-T4 nanoparticles during propagation of T4 in E. coli. Intranasal administration of 3M2e-T4 nanoparticles maintains antigen persistence in the lungs, resulting in increased uptake and presentation by antigen-presenting cells. M2e-specific secretory IgA, effector (TEM), central (TCM), and tissue-resident memory CD4+ T cells (TRM) were efficiently induced in the local mucosal sites, which mediated protections against divergent influenza viruses. Our studies demonstrated the mechanisms of immune protection following 3M2e-T4 nanoparticles vaccination and provide a versatile T4 platform that can be customized to rapidly develop mucosal vaccines against future emerging epidemics.
Collapse
Affiliation(s)
- Mengling Li
- State Key Laboratory of Agricultural Microbiology, Key Laboratory of Prevention & Control for African Swine Fever and Other Major Pig Diseases, Ministry of Agriculture and Rural Affairs, Cooperative Innovation Center for Sustainable Pig Production, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, 430070, China; Hubei Hongshan Lab, Wuhan, Hubei, 430070, China
| | - Cen Chen
- State Key Laboratory of Agricultural Microbiology, Key Laboratory of Prevention & Control for African Swine Fever and Other Major Pig Diseases, Ministry of Agriculture and Rural Affairs, Cooperative Innovation Center for Sustainable Pig Production, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, 430070, China; Hubei Hongshan Lab, Wuhan, Hubei, 430070, China
| | - Xialin Wang
- State Key Laboratory of Agricultural Microbiology, Key Laboratory of Prevention & Control for African Swine Fever and Other Major Pig Diseases, Ministry of Agriculture and Rural Affairs, Cooperative Innovation Center for Sustainable Pig Production, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, 430070, China; Hubei Hongshan Lab, Wuhan, Hubei, 430070, China
| | - Pengju Guo
- State Key Laboratory of Agricultural Microbiology, Key Laboratory of Prevention & Control for African Swine Fever and Other Major Pig Diseases, Ministry of Agriculture and Rural Affairs, Cooperative Innovation Center for Sustainable Pig Production, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, 430070, China; Hubei Hongshan Lab, Wuhan, Hubei, 430070, China
| | - Helong Feng
- State Key Laboratory of Agricultural Microbiology, Key Laboratory of Prevention & Control for African Swine Fever and Other Major Pig Diseases, Ministry of Agriculture and Rural Affairs, Cooperative Innovation Center for Sustainable Pig Production, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, 430070, China; Institute of Animal Husbandry and Veterinary Sciences, Hubei Academy of Agricultural Sciences, Wuhan, Hubei, 430070, China
| | - Xueqi Zhang
- State Key Laboratory of Agricultural Microbiology, Key Laboratory of Prevention & Control for African Swine Fever and Other Major Pig Diseases, Ministry of Agriculture and Rural Affairs, Cooperative Innovation Center for Sustainable Pig Production, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, 430070, China; Hubei Hongshan Lab, Wuhan, Hubei, 430070, China
| | - Wanpo Zhang
- State Key Laboratory of Agricultural Microbiology, Key Laboratory of Prevention & Control for African Swine Fever and Other Major Pig Diseases, Ministry of Agriculture and Rural Affairs, Cooperative Innovation Center for Sustainable Pig Production, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, 430070, China
| | - Changqin Gu
- State Key Laboratory of Agricultural Microbiology, Key Laboratory of Prevention & Control for African Swine Fever and Other Major Pig Diseases, Ministry of Agriculture and Rural Affairs, Cooperative Innovation Center for Sustainable Pig Production, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, 430070, China
| | - Jingen Zhu
- Bacteriophage Medical Research Center, Department of Biology, The Catholic University of America, Washington, DC, 20064, USA
| | - Guoyuan Wen
- Institute of Animal Husbandry and Veterinary Sciences, Hubei Academy of Agricultural Sciences, Wuhan, Hubei, 430070, China
| | - Yaoyu Feng
- Key Laboratory of Zoonosis of Ministry of Agriculture, College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Lihua Xiao
- Key Laboratory of Zoonosis of Ministry of Agriculture, College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Guiqing Peng
- State Key Laboratory of Agricultural Microbiology, Key Laboratory of Prevention & Control for African Swine Fever and Other Major Pig Diseases, Ministry of Agriculture and Rural Affairs, Cooperative Innovation Center for Sustainable Pig Production, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, 430070, China; Hubei Hongshan Lab, Wuhan, Hubei, 430070, China
| | - Venigalla B Rao
- Bacteriophage Medical Research Center, Department of Biology, The Catholic University of America, Washington, DC, 20064, USA
| | - Pan Tao
- State Key Laboratory of Agricultural Microbiology, Key Laboratory of Prevention & Control for African Swine Fever and Other Major Pig Diseases, Ministry of Agriculture and Rural Affairs, Cooperative Innovation Center for Sustainable Pig Production, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, 430070, China; Hubei Hongshan Lab, Wuhan, Hubei, 430070, China.
| |
Collapse
|
25
|
Batty CJ, Pena ES, Amouzougan EA, Moore KM, Ainslie KM, Bachelder EM. Humoral Response to the Acetalated Dextran M2e Vaccine is Enhanced by Antigen Surface Conjugation. Bioconjug Chem 2023; 34:1447-1458. [PMID: 37458383 PMCID: PMC11654056 DOI: 10.1021/acs.bioconjchem.3c00223] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/17/2023]
Abstract
The influenza A virus causes substantial morbidity and mortality worldwide every year and poses a constant threat of an emergent pandemic. Seasonal influenza vaccination strategies fail to provide complete protection against infection due to antigenic drift and shift. A universal vaccine targeting a conserved influenza epitope could substantially improve current vaccination strategies. The ectodomain of the matrix 2 protein (M2e) of influenza is a highly conserved epitope between virus strains but is also poorly immunogenic. Administration of M2e and the immunostimulatory stimulator of interferon genes (STING) agonist 3'3'-cyclic guanosine-adenosine monophosphate (cGAMP) encapsulated in microparticles made of acetalated dextran (Ace-DEX) has previously been shown to be effective for increasing the immunogenicity of M2e, primarily through T-cell-mediated responses. Here, the immunogenicity of Ace-DEX MPs delivering M2e was further improved by conjugating the M2e peptide to the particle surface in an effort to affect B-cell responses more directly. Conjugated or encapsulated M2e co-administered with Ace-DEX MPs containing cGAMP were used to vaccinate mice, and it was shown that two or three vaccinations could fully protect against a lethal influenza challenge, while only the surface-conjugated antigen constructs could provide some protection against lethal challenge with only one vaccination. Additionally, the use of a reducible linker augmented the T-cell response to the antigen. These results show the utility of conjugating M2e to the surface of a particle carrier to increase its immunogenicity for use as the antigen in a universal influenza vaccine.
Collapse
Affiliation(s)
- Cole J. Batty
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Erik S. Pena
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, NC, USA
| | - Eva A. Amouzougan
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Kathryn M. Moore
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, NC, USA
| | - Kristy M. Ainslie
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, NC, USA
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Eric M. Bachelder
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
26
|
Zhu W, Park J, Pho T, Wei L, Dong C, Kim J, Ma Y, Champion JA, Wang BZ. ISCOMs/MPLA-Adjuvanted SDAD Protein Nanoparticles Induce Improved Mucosal Immune Responses and Cross-Protection in Mice. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2301801. [PMID: 37162451 PMCID: PMC10524461 DOI: 10.1002/smll.202301801] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 04/20/2023] [Indexed: 05/11/2023]
Abstract
The epidemics caused by the influenza virus are a serious threat to public health and the economy. Adding appropriate adjuvants to improve immunogenicity and finding effective mucosal vaccines to combat respiratory infection at the portal of virus entry are important strategies to boost protection. In this study, a novel type of core/shell protein nanoparticle consisting of influenza nucleoprotein (NP) as the core and NA1-M2e or NA2-M2e fusion proteins as the coating antigens by SDAD hetero-bifunctional crosslinking is exploited. Immune-stimulating complexes (ISCOMs)/monophosphoryl lipid A (MPLA) adjuvants further boost the NP/NA-M2e SDAD protein nanoparticle-induced immune responses when administered intramuscularly. The ISCOMs/MPLA-adjuvanted protein nanoparticles are delivered through the intranasal route to validate the application as mucosal vaccines. ISCOMs/MPLA-adjuvanted nanoparticles induce significantly strengthened antigen-specific antibody responses, cytokine-secreting splenocytes in the systemic compartment, and higher levels of antigen-specific IgA and IgG in the local mucosa. Meanwhile, significantly expanded lung resident memory (RM) T and B cells (TRM /BRM ) and alveolar macrophages population are observed in ISCOMs/MPLA-adjuvanted nanoparticle-immunized mice with a 100% survival rate after homogeneous and heterogeneous H3N2 viral challenges. Taken together, ISCOMs/MPLA-adjuvanted protein nanoparticles could improve strong systemic and mucosal immune responses conferring protection in different immunization routes.
Collapse
Affiliation(s)
- Wandi Zhu
- Center for Inflammation, Immunity & Infection, Georgia State University, Atlanta, GA 30303, USA
| | - Jaeyoung Park
- School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Thomas Pho
- School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
- Bioengineering Program, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Lai Wei
- Center for Inflammation, Immunity & Infection, Georgia State University, Atlanta, GA 30303, USA
| | - Chunhong Dong
- Center for Inflammation, Immunity & Infection, Georgia State University, Atlanta, GA 30303, USA
| | - Joo Kim
- Center for Inflammation, Immunity & Infection, Georgia State University, Atlanta, GA 30303, USA
| | - Yao Ma
- Center for Inflammation, Immunity & Infection, Georgia State University, Atlanta, GA 30303, USA
| | - Julie A. Champion
- School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
- Bioengineering Program, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Bao-Zhong Wang
- Center for Inflammation, Immunity & Infection, Georgia State University, Atlanta, GA 30303, USA
| |
Collapse
|
27
|
Park J, Pho T, Champion JA. Chemical and biological conjugation strategies for the development of multivalent protein vaccine nanoparticles. Biopolymers 2023; 114:e23563. [PMID: 37490564 PMCID: PMC10528127 DOI: 10.1002/bip.23563] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Revised: 05/19/2023] [Accepted: 07/03/2023] [Indexed: 07/27/2023]
Abstract
The development of subunit vaccine platforms has been of considerable interest due to their good safety profile and ability to be adapted to new antigens, compared to other vaccine typess. Nevertheless, subunit vaccines often lack sufficient immunogenicity to fully protect against infectious diseases. A wide variety of subunit vaccines have been developed to enhance antigen immunogenicity by increasing antigen multivalency, as well as stability and delivery properties, via presentation of antigens on protein nanoparticles. Increasing multivalency can be an effective approach to provide a potent humoral immune response by more strongly engaging and clustering B cell receptors (BCRs) to induce activation, as well as increased uptake by antigen presenting cells and their subsequent T cell activation. Proper orientation of antigen on protein nanoparticles is also considered a crucial factor for enhanced BCR engagement and subsequent immune responses. Therefore, various strategies have been reported to decorate highly repetitive surfaces of protein nanoparticle scaffolds with multiple copies of antigens, arrange antigens in proper orientation, or combinations thereof. In this review, we describe different chemical bioconjugation methods, approaches for genetic fusion of recombinant antigens, biological affinity tags, and enzymatic conjugation methods to effectively present antigens on the surface of protein nanoparticle vaccine scaffolds.
Collapse
Affiliation(s)
- Jaeyoung Park
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, 950 Atlantic Dr. NW, Atlanta, GA, 30332-2000, USA
| | - Thomas Pho
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, 950 Atlantic Dr. NW, Atlanta, GA, 30332-2000, USA
- BioEngineering Program
| | - Julie A. Champion
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, 950 Atlantic Dr. NW, Atlanta, GA, 30332-2000, USA
- BioEngineering Program
| |
Collapse
|
28
|
Bhatnagar N, Kim KH, Subbiah J, Muhammad-Worsham S, Park BR, Liu R, Grovenstein P, Wang BZ, Kang SM. Heterologous Prime-Boost Vaccination with Inactivated Influenza Viruses Induces More Effective Cross-Protection than Homologous Repeat Vaccination. Vaccines (Basel) 2023; 11:1209. [PMID: 37515025 PMCID: PMC10386405 DOI: 10.3390/vaccines11071209] [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: 04/30/2023] [Revised: 06/24/2023] [Accepted: 06/30/2023] [Indexed: 07/30/2023] Open
Abstract
With concerns about the efficacy of repeat annual influenza vaccination, it is important to better understand the impact of priming vaccine immunity and develop an effective vaccination strategy. Here, we determined the impact of heterologous prime-boost vaccination on inducing broader protective immunity compared to repeat vaccination with the same antigen. The primed mice that were intramuscularly boosted with a heterologous inactivated influenza A virus (H1N1, H3N2, H5N1, H7N9, H9N2) vaccine showed increased strain-specific hemagglutination inhibition titers against prime and boost vaccine strains. Heterologous prime-boost vaccination of mice with inactivated viruses was more effective in inducing high levels of IgG antibodies specific for groups 1 and 2 hemagglutinin stalk domains, as well as cross-protection, compared to homologous vaccination. Both humoral and T cell immunity were found to play a critical role in conferring cross-protection by heterologous prime-boost vaccination. These results support a strategy to enhance cross-protective efficacy by heterologous prime-boost influenza vaccination.
Collapse
Affiliation(s)
- Noopur Bhatnagar
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30302, USA; (N.B.); (K.-H.K.); (J.S.); (S.M.-W.); (B.R.P.); (R.L.); (P.G.); (B.-Z.W.)
| | - Ki-Hye Kim
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30302, USA; (N.B.); (K.-H.K.); (J.S.); (S.M.-W.); (B.R.P.); (R.L.); (P.G.); (B.-Z.W.)
| | - Jeeva Subbiah
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30302, USA; (N.B.); (K.-H.K.); (J.S.); (S.M.-W.); (B.R.P.); (R.L.); (P.G.); (B.-Z.W.)
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA 30329, USA
| | - Sakinah Muhammad-Worsham
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30302, USA; (N.B.); (K.-H.K.); (J.S.); (S.M.-W.); (B.R.P.); (R.L.); (P.G.); (B.-Z.W.)
| | - Bo Ryoung Park
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30302, USA; (N.B.); (K.-H.K.); (J.S.); (S.M.-W.); (B.R.P.); (R.L.); (P.G.); (B.-Z.W.)
| | - Rong Liu
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30302, USA; (N.B.); (K.-H.K.); (J.S.); (S.M.-W.); (B.R.P.); (R.L.); (P.G.); (B.-Z.W.)
| | - Phillip Grovenstein
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30302, USA; (N.B.); (K.-H.K.); (J.S.); (S.M.-W.); (B.R.P.); (R.L.); (P.G.); (B.-Z.W.)
| | - Bao-Zhong Wang
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30302, USA; (N.B.); (K.-H.K.); (J.S.); (S.M.-W.); (B.R.P.); (R.L.); (P.G.); (B.-Z.W.)
| | - Sang-Moo Kang
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30302, USA; (N.B.); (K.-H.K.); (J.S.); (S.M.-W.); (B.R.P.); (R.L.); (P.G.); (B.-Z.W.)
| |
Collapse
|
29
|
Cao F, Peng S, An Y, Xu K, Zheng T, Dai L, Ogino K, Ngai T, Xia Y, Ma G. Inside-out assembly of viral antigens for the enhanced vaccination. Signal Transduct Target Ther 2023; 8:189. [PMID: 37221173 DOI: 10.1038/s41392-023-01414-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 01/13/2023] [Accepted: 03/08/2023] [Indexed: 05/25/2023] Open
Abstract
Current attempts in vaccine delivery systems concentrate on replicating the natural dissemination of live pathogens, but neglect that pathogens evolve to evade the immune system rather than to provoke it. In the case of enveloped RNA viruses, it is the natural dissemination of nucleocapsid protein (NP, core antigen) and surface antigen that delays NP exposure to immune surveillance. Here, we report a multi-layered aluminum hydroxide-stabilized emulsion (MASE) to dictate the delivery sequence of the antigens. In this manner, the receptor-binding domain (RBD, surface antigen) of the spike protein was trapped inside the nanocavity, while NP was absorbed on the outside of the droplets, enabling the burst release of NP before RBD. Compared with the natural packaging strategy, the inside-out strategy induced potent type I interferon-mediated innate immune responses and triggered an immune-potentiated environment in advance, which subsequently boosted CD40+ DC activations and the engagement of the lymph nodes. In both H1N1 influenza and SARS-CoV-2 vaccines, rMASE significantly increased antigen-specific antibody secretion, memory T cell engagement, and Th1-biased immune response, which diminished viral loads after lethal challenge. By simply reversing the delivery sequence of the surface antigen and core antigen, the inside-out strategy may offer major implications for enhanced vaccinations against the enveloped RNA virus.
Collapse
Affiliation(s)
- Fengqiang Cao
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, PR China
- Graduate School of Bio-Applications and Systems Engineering, Tokyo University of Agriculture and Technology, 2-24-16 Nakacho, Koganei, Tokyo, 184-8588, Japan
- Institute of Global Innovation Research, Tokyo University of Agriculture and Technology, 2-24-16 Nakacho, Koganei-shi, Tokyo, 184-8588, Japan
| | - Sha Peng
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, PR China
- Graduate School of Bio-Applications and Systems Engineering, Tokyo University of Agriculture and Technology, 2-24-16 Nakacho, Koganei, Tokyo, 184-8588, Japan
- Institute of Global Innovation Research, Tokyo University of Agriculture and Technology, 2-24-16 Nakacho, Koganei-shi, Tokyo, 184-8588, Japan
| | - Yaling An
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, PR China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, PR China
| | - Kun Xu
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, PR China
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, School of Tropical Medicine and Laboratory Medicine, The First Affiliated Hospital, Hainan Medical University, Hainan, 571199, PR China
| | - Tianyi Zheng
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, PR China
- Zhejiang University School of Medicine, Hangzhou, 310058, PR China
| | - Lianpan Dai
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, PR China
| | - Kenji Ogino
- Graduate School of Bio-Applications and Systems Engineering, Tokyo University of Agriculture and Technology, 2-24-16 Nakacho, Koganei, Tokyo, 184-8588, Japan.
- Institute of Global Innovation Research, Tokyo University of Agriculture and Technology, 2-24-16 Nakacho, Koganei-shi, Tokyo, 184-8588, Japan.
| | - To Ngai
- Department of Chemistry, The Chinese University of Hong Kong, Shatin, N. T., Hong Kong, 999077, PR China
| | - Yufei Xia
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, PR China.
- University of Chinese Academy of Sciences, Beijing, 100049, PR China.
- Innovation Academy for Green Manufacture Chinese Academy of Sciences, Beijing, 100190, PR China.
| | - Guanghui Ma
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, PR China.
- Institute of Global Innovation Research, Tokyo University of Agriculture and Technology, 2-24-16 Nakacho, Koganei-shi, Tokyo, 184-8588, Japan.
- University of Chinese Academy of Sciences, Beijing, 100049, PR China.
- Innovation Academy for Green Manufacture Chinese Academy of Sciences, Beijing, 100190, PR China.
| |
Collapse
|
30
|
Frey SJ, Carreño JM, Bielak D, Arsiwala A, Altomare CG, Varner C, Rosen-Cheriyan T, Bajic G, Krammer F, Kane RS. Nanovaccines Displaying the Influenza Virus Hemagglutinin in an Inverted Orientation Elicit an Enhanced Stalk-Directed Antibody Response. Adv Healthc Mater 2023; 12:e2202729. [PMID: 36689343 PMCID: PMC10386890 DOI: 10.1002/adhm.202202729] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Revised: 01/18/2023] [Indexed: 01/24/2023]
Abstract
Despite the availability of licensed vaccines, influenza causes considerable morbidity and mortality worldwide. Current influenza vaccines elicit an immune response that primarily targets the head domain of the viral glycoprotein hemagglutinin (HA). Influenza viruses, however, readily evade this response by acquiring mutations in the head domain. While vaccines that target the more conserved HA stalk may circumvent this problem, low levels of antistalk antibodies are elicited by vaccination, possibly due to the poor accessibility of the stalk domain to B cell receptors. In this work, it is demonstrated that nanoparticles presenting HA in an inverted orientation generate tenfold higher antistalk antibody titers after a prime immunization and fivefold higher antistalk titers after a boost than nanoparticles displaying HA in its regular orientation. Moreover, nanoparticles presenting HA in an inverted orientation elicit a broader antistalk response that reduces mouse weight loss and improves survival after challenge to a greater extent than nanoparticles displaying HA in a regular orientation. Refocusing the antibody response toward conserved epitopes by controlling antigen orientation may enable the design of broadly protective nanovaccines targeting influenza viruses and other pathogens with pandemic potential.
Collapse
Affiliation(s)
- Steven J Frey
- School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Juan Manuel Carreño
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Dominika Bielak
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Ammar Arsiwala
- School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Clara G Altomare
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Chad Varner
- School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Tania Rosen-Cheriyan
- School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Goran Bajic
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Pathology, Molecular and Cell Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Ravi S Kane
- School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| |
Collapse
|
31
|
Tsai HH, Huang PH, Lin LC, Yao BY, Liao WT, Pai CH, Liu YH, Chen HW, Hu CMJ. Lymph Node Follicle-Targeting STING Agonist Nanoshells Enable Single-Shot M2e Vaccination for Broad and Durable Influenza Protection. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023:e2206521. [PMID: 37092580 DOI: 10.1002/advs.202206521] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 03/14/2023] [Indexed: 05/03/2023]
Abstract
The highly conserved matrix protein 2 ectodomain (M2e) of influenza viruses presents a compelling vaccine antigen candidate for stemming the pandemic threat of the mutation-prone pathogen, yet the low immunogenicity of the diminutive M2e peptide renders vaccine development challenging. A highly potent M2e nanoshell vaccine that confers broad and durable influenza protectivity under a single vaccination is shown. Prepared via asymmetric ionic stabilization for nanoscopic curvature formation, polymeric nanoshells co-encapsulating high densities of M2e peptides and stimulator of interferon genes (STING) agonists are prepared. Robust and long-lasting protectivity against heterotypic influenza viruses is achieved with a single administration of the M2e nanoshells in mice. Mechanistically, molecular adjuvancy by the STING agonist and nanoshell-mediated prolongation of M2e antigen exposure in the lymph node follicles synergistically contribute to the heightened anti-M2e humoral responses. STING agonist-triggered T cell helper functions and extended residence of M2e peptides in the follicular dendritic cell network provide a favorable microenvironment that induces Th1-biased antibody production against the diminutive antigen. These findings highlight a versatile nanoparticulate design that leverages innate immune pathways for enhancing the immunogenicity of weak immunogens. The single-shot nanovaccine further provides a translationally viable platform for pandemic preparedness.
Collapse
Affiliation(s)
- Hsiao-Han Tsai
- Institute of Biomedical Sciences, Academia Sinica, Taipei, 115, Taiwan
- Taiwan International Graduate Program in Molecular Medicine, National Yang Ming Chiao Tung University and Academia Sinica, Taipei, 112, Taiwan
| | - Ping-Han Huang
- Department of Veterinary Medicine, National Taiwan University, Taipei, 10617, Taiwan
| | - Leon Cw Lin
- Institute of Biomedical Sciences, Academia Sinica, Taipei, 115, Taiwan
- Biomedical Translation Research Center, Academia Sinica, Taipei, 115, Taiwan
| | - Bing-Yu Yao
- Institute of Biomedical Sciences, Academia Sinica, Taipei, 115, Taiwan
- Biomedical Translation Research Center, Academia Sinica, Taipei, 115, Taiwan
| | - Wan-Ting Liao
- Department of Veterinary Medicine, National Taiwan University, Taipei, 10617, Taiwan
| | - Chen-Hsueh Pai
- Institute of Biomedical Sciences, Academia Sinica, Taipei, 115, Taiwan
- Biomedical Translation Research Center, Academia Sinica, Taipei, 115, Taiwan
| | - Yu-Han Liu
- Institute of Biomedical Sciences, Academia Sinica, Taipei, 115, Taiwan
| | - Hui-Wen Chen
- Department of Veterinary Medicine, National Taiwan University, Taipei, 10617, Taiwan
| | - Che-Ming J Hu
- Institute of Biomedical Sciences, Academia Sinica, Taipei, 115, Taiwan
- Taiwan International Graduate Program in Molecular Medicine, National Yang Ming Chiao Tung University and Academia Sinica, Taipei, 112, Taiwan
- Biomedical Translation Research Center, Academia Sinica, Taipei, 115, Taiwan
- Center of Applied Nanomedicine, National Cheng Kung University, Tainan, 70101, Taiwan
| |
Collapse
|
32
|
Park J, Champion JA. Effect of Antigen Structure in Subunit Vaccine Nanoparticles on Humoral Immune Responses. ACS Biomater Sci Eng 2023; 9:1296-1306. [PMID: 36848229 PMCID: PMC10015428 DOI: 10.1021/acsbiomaterials.2c01516] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 02/01/2023] [Indexed: 03/01/2023]
Abstract
Subunit vaccines offer numerous attractive features, including good safety profiles and well-defined components with highly characterized properties because they do not contain whole pathogens. However, vaccine platforms based on one or few selected antigens are often poorly immunogenic. Several advances have been made in improving the effectiveness of subunit vaccines, including nanoparticle formulation and/or co-administration with adjuvants. Desolvation of antigens into nanoparticles is one approach that has been successful in eliciting protective immune responses. Despite this advance, damage to the antigen structure by desolvation can compromise the recognition of conformational antigens by B cells and the subsequent humoral response. Here, we used ovalbumin as a model antigen to demonstrate enhanced efficacy of subunit vaccines by preserving antigen structures in nanoparticles. An altered antigen structure due to desolvation was first validated by GROMACS and circular dichroism. Desolvant-free nanoparticles with a stable ovalbumin structure were successfully synthesized by directly cross-linking ovalbumin or using ammonium sulfate to form nanoclusters. Alternatively, desolvated OVA nanoparticles were coated with a layer of OVA after desolvation. Vaccination with salt-precipitated nanoparticles increased OVA-specific IgG titers 4.2- and 22-fold compared to the desolvated and coated nanoparticles, respectively. In addition, enhanced affinity maturation by both salt precipitated and coated nanoparticles was displayed in contrast to desolvated nanoparticles. These results demonstrate both that salt-precipitated antigen nanoparticles are a potential new vaccine platform with significantly improved humoral immunity and a functional value of preserving antigen structures in vaccine nanoparticle design.
Collapse
Affiliation(s)
- Jaeyoung Park
- School of Chemical and Biomolecular
Engineering, Georgia Institute of Technology, 950 Atlantic Dr. NW, Atlanta, Georgia 30332-2000, United States
| | - Julie A. Champion
- School of Chemical and Biomolecular
Engineering, Georgia Institute of Technology, 950 Atlantic Dr. NW, Atlanta, Georgia 30332-2000, United States
| |
Collapse
|
33
|
Deigin V, Premyslova M, Ksenofontova O, Yatskin O, Volpina O. Evaluation of Neuroprotective and Adjuvant Activities of Diketopiperazine‐Based Peptidomimetics. ChemistrySelect 2023. [DOI: 10.1002/slct.202204076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2023]
Affiliation(s)
- Vladislav Deigin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry Russian Academy of Sciences Miklukho- Maklaya St., 16/10 Moscow 117997 Russia
- Immunotech Developments Inc. 2395 Speakman Drive, Suite 2025 Mississauga Ontario L5 K 1B3 Canada
| | - Marina Premyslova
- Immunotech Developments Inc. 2395 Speakman Drive, Suite 2025 Mississauga Ontario L5 K 1B3 Canada
| | - Olga Ksenofontova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry Russian Academy of Sciences Miklukho- Maklaya St., 16/10 Moscow 117997 Russia
| | - Oleg Yatskin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry Russian Academy of Sciences Miklukho- Maklaya St., 16/10 Moscow 117997 Russia
| | - Olga Volpina
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry Russian Academy of Sciences Miklukho- Maklaya St., 16/10 Moscow 117997 Russia
| |
Collapse
|
34
|
Danielsen M, Kempen PJ, Andresen TL, Urquhart AJ. Formulation and characterization of insulin nanoclusters for a controlled release. Int J Biol Macromol 2023; 235:123658. [PMID: 36822285 DOI: 10.1016/j.ijbiomac.2023.123658] [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: 10/27/2022] [Revised: 01/31/2023] [Accepted: 02/09/2023] [Indexed: 02/25/2023]
Abstract
The growing interest in biopharmaceuticals combined with the challenges regarding formulation and delivery continues to encourage the development of new and improved formulations of this class of therapeutics. Nanoclusters (NCs) represent a type of formulation strategy where the biopharmaceutical is clustered in a reversible manner to function as both the therapeutic and the vehicle. In this study, insulin NCs (INCs) were formulated by a new methodology of first crosslinking proteins followed by desolvation. Crosslinking of the protein with the reducible DTSSP crosslinker improved control of the INC synthesis process to give INCs with a mean size of 198 ± 7 nm and a mean zeta potential of -39 ± 1 mV. Crosslinking and clustering of insulin did not induce cytotoxicity or major differences in the biological activity compared to the free unmodified protein. The potency of the crosslinked insulin and the INCs appeared slightly lower than that of the unmodified protein, and significantly higher doses of the INCs compared to the free protein were applied to achieve similar blood sugar lowering effects in vivo. Interestingly, the INCs allowed for high doses to be subcutaneously delivered with prolonged efficacy without being lethal in rats.
Collapse
Affiliation(s)
- Mia Danielsen
- Department of Health Technology, Technical University of Denmark, 2800 Kongens Lyngby, Denmark
| | - Paul Joseph Kempen
- Department of Health Technology, Technical University of Denmark, 2800 Kongens Lyngby, Denmark; National Centre for Nano Fabrication and Characterization, Technical University of Denmark, 2800 Kongens Lyngby, Denmark
| | - Thomas Lars Andresen
- Department of Health Technology, Technical University of Denmark, 2800 Kongens Lyngby, Denmark
| | - Andrew James Urquhart
- Department of Health Technology, Technical University of Denmark, 2800 Kongens Lyngby, Denmark.
| |
Collapse
|
35
|
Zhang Z, Shang J, Yang Q, Dai Z, Liang Y, Lai C, Feng T, Zhong D, Zou H, Sun L, Su Y, Yan S, Chen J, Yao Y, Shi Y, Huang X. Exosomes derived from human adipose mesenchymal stem cells ameliorate hepatic fibrosis by inhibiting PI3K/Akt/mTOR pathway and remodeling choline metabolism. J Nanobiotechnology 2023; 21:29. [PMID: 36698192 PMCID: PMC9878808 DOI: 10.1186/s12951-023-01788-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Accepted: 01/17/2023] [Indexed: 01/26/2023] Open
Abstract
Liver fibrosis is a chronic liver disease with the presence of progressive wound healing response caused by liver injury. Currently, there are no approved therapies for liver fibrosis. Exosomes derived from human adipose mesenchymal stem cells (hADMSCs-Exo) have displayed a prominent therapeutic effect on liver diseases. However, few studies have evaluated therapeutic effect of hADMSCs-Exo in liver fibrosis and cirrhosis, and its precise mechanisms of action remain unclear. Herein, we investigated anti-fibrotic efficacy of hADMSCs-Exo in vitro and in vivo, and identified important metabolic changes and the detailed mechanism through transcriptomic and metabolomic profiling. We found hADMSCs-Exo could inhibit the proliferation of activated hepatic stellate cells through aggravating apoptosis and arresting G1 phase, effectively inhibiting the expression of profibrogenic proteins and epithelial-to-mesenchymal transition (EMT) in vitro. Moreover, it could significantly block collagen deposition and EMT process, improve liver function and reduce liver inflammation in liver cirrhosis mice model. The omics analysis revealed that the key mechanism of hADMSCs-Exo anti-hepatic fibrosis was the inhibition of PI3K/AKT/mTOR signaling pathway and affecting the changes of metabolites in lipid metabolism, and mainly regulating choline metabolism. CHPT1 activated by hADMSCs-Exo facilitated formation and maintenance of vesicular membranes. Thus, our study indicates that hADMSCs-Exo can attenuate hepatic stellate cell activation and suppress the progression of liver fibrosis, which holds the significant potential of hADMSCs-Exo for use as extracellular nanovesicles-based therapeutics in the treatment of liver fibrosis and possibly other intractable chronic liver diseases.
Collapse
Affiliation(s)
- Zilong Zhang
- grid.54549.390000 0004 0369 4060Liver Transplantation Center and HBP Surgery, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Cancer Hospital Affiliate to University of Electronic Science and Technology of China, Chengdu, 610042 Sichuan China ,grid.54549.390000 0004 0369 4060School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610054 Sichuan China
| | - Jin Shang
- grid.54549.390000 0004 0369 4060Liver Transplantation Center and HBP Surgery, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Cancer Hospital Affiliate to University of Electronic Science and Technology of China, Chengdu, 610042 Sichuan China ,grid.54549.390000 0004 0369 4060School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610054 Sichuan China
| | - Qinyan Yang
- grid.54549.390000 0004 0369 4060Liver Transplantation Center and HBP Surgery, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Cancer Hospital Affiliate to University of Electronic Science and Technology of China, Chengdu, 610042 Sichuan China ,grid.54549.390000 0004 0369 4060School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610054 Sichuan China
| | - Zonglin Dai
- grid.54549.390000 0004 0369 4060Liver Transplantation Center and HBP Surgery, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Cancer Hospital Affiliate to University of Electronic Science and Technology of China, Chengdu, 610042 Sichuan China ,grid.54549.390000 0004 0369 4060School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610054 Sichuan China
| | - Yuxin Liang
- grid.54549.390000 0004 0369 4060Liver Transplantation Center and HBP Surgery, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Cancer Hospital Affiliate to University of Electronic Science and Technology of China, Chengdu, 610042 Sichuan China ,grid.54549.390000 0004 0369 4060School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610054 Sichuan China
| | - Chunyou Lai
- grid.54549.390000 0004 0369 4060Liver Transplantation Center and HBP Surgery, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Cancer Hospital Affiliate to University of Electronic Science and Technology of China, Chengdu, 610042 Sichuan China ,grid.54549.390000 0004 0369 4060School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610054 Sichuan China
| | - Tianhang Feng
- grid.54549.390000 0004 0369 4060Liver Transplantation Center and HBP Surgery, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Cancer Hospital Affiliate to University of Electronic Science and Technology of China, Chengdu, 610042 Sichuan China ,grid.54549.390000 0004 0369 4060School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610054 Sichuan China
| | - Deyuan Zhong
- grid.54549.390000 0004 0369 4060Liver Transplantation Center and HBP Surgery, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Cancer Hospital Affiliate to University of Electronic Science and Technology of China, Chengdu, 610042 Sichuan China ,grid.54549.390000 0004 0369 4060School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610054 Sichuan China
| | - Haibo Zou
- grid.54549.390000 0004 0369 4060Liver Transplantation Center and HBP Surgery, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Cancer Hospital Affiliate to University of Electronic Science and Technology of China, Chengdu, 610042 Sichuan China ,grid.54549.390000 0004 0369 4060School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610054 Sichuan China
| | - Lelin Sun
- grid.54549.390000 0004 0369 4060School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610054 Sichuan China
| | - Yuhao Su
- grid.54549.390000 0004 0369 4060Liver Transplantation Center and HBP Surgery, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Cancer Hospital Affiliate to University of Electronic Science and Technology of China, Chengdu, 610042 Sichuan China ,grid.54549.390000 0004 0369 4060School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610054 Sichuan China
| | - Su Yan
- grid.54549.390000 0004 0369 4060Liver Transplantation Center and HBP Surgery, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Cancer Hospital Affiliate to University of Electronic Science and Technology of China, Chengdu, 610042 Sichuan China ,grid.54549.390000 0004 0369 4060School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610054 Sichuan China
| | - Jie Chen
- Department of Core laboratory, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, 610072 Sichuan China
| | - Yutong Yao
- grid.54549.390000 0004 0369 4060Liver Transplantation Center and HBP Surgery, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Cancer Hospital Affiliate to University of Electronic Science and Technology of China, Chengdu, 610042 Sichuan China ,grid.54549.390000 0004 0369 4060School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610054 Sichuan China
| | - Ying Shi
- grid.54549.390000 0004 0369 4060Liver Transplantation Center and HBP Surgery, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Cancer Hospital Affiliate to University of Electronic Science and Technology of China, Chengdu, 610042 Sichuan China ,grid.54549.390000 0004 0369 4060School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610054 Sichuan China
| | - Xiaolun Huang
- grid.54549.390000 0004 0369 4060Liver Transplantation Center and HBP Surgery, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Cancer Hospital Affiliate to University of Electronic Science and Technology of China, Chengdu, 610042 Sichuan China ,grid.54549.390000 0004 0369 4060School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610054 Sichuan China
| |
Collapse
|
36
|
Wang Y, Dong C, Ma Y, Zhu W, Gill HS, Denning TL, Kang SM, Wang BZ. Monophosphoryl lipid A-adjuvanted nucleoprotein-neuraminidase nanoparticles improve immune protection against divergent influenza viruses. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2023; 47:102614. [PMID: 36265560 PMCID: PMC9756393 DOI: 10.1016/j.nano.2022.102614] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 09/29/2022] [Accepted: 10/10/2022] [Indexed: 11/06/2022]
Abstract
Universal influenza vaccines are urgently needed to prevent recurrent influenza epidemics and inevitable pandemics. We generated double-layered protein nanoparticles incorporating two conserved influenza antigens-nucleoprotein and neuraminidase-through a two-step desolvation-crosslinking method. These protein nanoparticles displayed immunostimulatory properties to antigen-presenting cells by promoting inflammatory cytokine (IL-6 and TNF-α) secretion from JAWS II dendric cells. The nanoparticle immunization induced significant antigen-specific humoral and cellular responses, including antigen-binding and neutralizing antibodies, antibody- and cytokine (IFN-γ and IL-4)-secreting cells, and NP147-155 tetramer-specific cytotoxic T lymphocyte (CTL) responses. Co-administration of monophosphoryl lipid A (MPLA, a toll-like receptor 4 agonist) with the protein nanoparticles further improved immune responses and conferred heterologous and heterosubtypic influenza protection. The MPLA-adjuvanted nanoparticles reduced lung inflammation post-infection. The results demonstrated that the combination of MPLA and conserved protein nanoparticles could be developed into an improved universal influenza vaccine strategy.
Collapse
Affiliation(s)
- Ye Wang
- Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, 100 Piedmont Ave SE, Atlanta, GA 30303, USA
| | - Chunhong Dong
- Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, 100 Piedmont Ave SE, Atlanta, GA 30303, USA
| | - Yao Ma
- Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, 100 Piedmont Ave SE, Atlanta, GA 30303, USA
| | - Wandi Zhu
- Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, 100 Piedmont Ave SE, Atlanta, GA 30303, USA
| | - Harvinder Singh Gill
- Department of Chemical Engineering, Texas Tech University, Lubbock, TX 79409, USA
| | - Timothy L Denning
- Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, 100 Piedmont Ave SE, Atlanta, GA 30303, USA
| | - Sang-Moo Kang
- Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, 100 Piedmont Ave SE, Atlanta, GA 30303, USA
| | - Bao-Zhong Wang
- Center for Inflammation, Immunity & Infection, Georgia State University Institute for Biomedical Sciences, 100 Piedmont Ave SE, Atlanta, GA 30303, USA.
| |
Collapse
|
37
|
Brinkhuizen C, Shapman D, Lebon A, Bénard M, Tardivel M, Dubuquoy L, Galas L, Carpentier R. Dipalmitoyl-phosphatidylserine-filled cationic maltodextrin nanoparticles exhibit enhanced efficacy for cell entry and intracellular protein delivery in phagocytic THP-1 cells. Biomol Concepts 2023; 14:bmc-2022-0029. [PMID: 37377352 DOI: 10.1515/bmc-2022-0029] [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: 03/29/2023] [Accepted: 05/08/2023] [Indexed: 06/29/2023] Open
Abstract
Vaccination through the upper respiratory tract is a promising strategy, and particulate antigens, such as antigens associated with nanoparticles, triggered a stronger immune response than the sole antigens. Cationic maltodextrin-based nanoparticles loaded with phosphatidylglycerol (NPPG) are efficient for intranasal vaccination but non-specific to trigger immune cells. Here we focused on phosphatidylserine (PS) receptors, specifically expressed by immune cells including macrophages, to improve nanoparticle targeting through an efferocytosis-like mechanism. Consequently, the lipids associated with NPPG have been substituted by PS to generate cationic maltodextrin-based nanoparticles with dipalmitoyl-phosphatidylserine (NPPS). Both NPPS and NPPG exhibited similar physical characteristics and intracellular distribution in THP-1 macrophages. NPPS cell entry was faster and higher (two times more) than NPPG. Surprisingly, competition of PS receptors with phospho-L-serine did not alter NPPS cell entry and annexin V did not preferentially interact with NPPS. Although the protein association is similar, NPPS delivered more proteins than NPPG in cells. On the contrary, the proportion of mobile nanoparticles (50%), the movement speed of nanoparticles (3 µm/5 min), and protein degradation kinetics in THP-1 were not affected by lipid substitution. Together, the results indicate that NPPS enter cells and deliver protein better than NPPG, suggesting that modification of the lipids of cationic maltodextrin-based nanoparticles may be a useful strategy to enhance nanoparticle efficacy for mucosal vaccination.
Collapse
Affiliation(s)
- Clément Brinkhuizen
- University Lille, Inserm, CHU Lille, U1286 - INFINITE - Institute for Translational Research in Inflammation, F-59000 Lille, France
| | - Damien Shapman
- University of Rouen Normandy, INSERM US 51, CNRS UAR 2026, HeRacLeS-PRIMACEN, Normandie Université, 76000 Rouen, France
| | - Alexis Lebon
- University of Rouen Normandy, INSERM US 51, CNRS UAR 2026, HeRacLeS-PRIMACEN, Normandie Université, 76000 Rouen, France
| | - Magalie Bénard
- University of Rouen Normandy, INSERM US 51, CNRS UAR 2026, HeRacLeS-PRIMACEN, Normandie Université, 76000 Rouen, France
| | - Meryem Tardivel
- University Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, US 41 - UAR 2014 - PLBS, F-59000 Lille, France
| | - Laurent Dubuquoy
- University Lille, Inserm, CHU Lille, U1286 - INFINITE - Institute for Translational Research in Inflammation, F-59000 Lille, France
| | - Ludovic Galas
- University of Rouen Normandy, INSERM US 51, CNRS UAR 2026, HeRacLeS-PRIMACEN, Normandie Université, 76000 Rouen, France
| | - Rodolphe Carpentier
- University Lille, Inserm, CHU Lille, U1286 - INFINITE - Institute for Translational Research in Inflammation, F-59000 Lille, France
| |
Collapse
|
38
|
Deigin V, Koroev D, Volpina O. Peptide ILE-GLU-TRP (Stemokin) Potential Adjuvant Stimulating a Balanced Immune Response. Int J Pept Res Ther 2022; 28:156. [PMID: 36313476 PMCID: PMC9589648 DOI: 10.1007/s10989-022-10461-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/07/2022] [Indexed: 12/03/2022]
Abstract
Vaccines are widely used worldwide to prevent and protect from various infections. A variety of modern approaches to developing prophylactic and therapeutic vaccines is growing. In almost all cases, adjuvants are necessary to obtain an effective immune response.This work investigated the possibility of using the pharmaceutical peptide drug Stemokin as an adjuvant stimulating a balanced Th1/Th2 response.A study was conducted to compare the activity of Stemokin versus the approved adjuvant Alhydrogel in a murine vaccination model with the approved VAXIGRIP® vaccine.The first proof-of-concept experimental study shows that the peptide Ile-Glu-Trp has the adjuvant vaccine properties and anti-HA IgG2a enhancing response, revealing a Th1- favoring balanced Th1/Th2 immunomodulation.
Collapse
|
39
|
Zhang Y, Jiang S, Lin J, Huang P. Antineoplastic Enzyme as Drug Carrier with Activatable Catalytic Activity for Efficient Combined Therapy. Angew Chem Int Ed Engl 2022; 61:e202208583. [DOI: 10.1002/anie.202208583] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Indexed: 01/17/2023]
Affiliation(s)
- Yifan Zhang
- Marshall Laboratory of Biomedical Engineering International Cancer Center Laboratory of Evolutionary Theranostics (LET) School of Biomedical Engineering Shenzhen University Health Science Center Shenzhen 518060 China
| | - Shanshan Jiang
- Marshall Laboratory of Biomedical Engineering International Cancer Center Laboratory of Evolutionary Theranostics (LET) School of Biomedical Engineering Shenzhen University Health Science Center Shenzhen 518060 China
| | - Jing Lin
- Marshall Laboratory of Biomedical Engineering International Cancer Center Laboratory of Evolutionary Theranostics (LET) School of Biomedical Engineering Shenzhen University Health Science Center Shenzhen 518060 China
| | - Peng Huang
- Marshall Laboratory of Biomedical Engineering International Cancer Center Laboratory of Evolutionary Theranostics (LET) School of Biomedical Engineering Shenzhen University Health Science Center Shenzhen 518060 China
| |
Collapse
|
40
|
Chimeric Virus-like Particles Co-Displaying Hemagglutinin Stem and the C-Terminal Fragment of DnaK Confer Heterologous Influenza Protection in Mice. Viruses 2022; 14:v14102109. [PMID: 36298664 PMCID: PMC9610613 DOI: 10.3390/v14102109] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 09/17/2022] [Accepted: 09/20/2022] [Indexed: 11/17/2022] Open
Abstract
Influenza virus hemagglutinin (HA) stem is currently regarded as an extremely promising immunogen for designing universal influenza vaccines. The appropriate antigen-presenting vaccine vector would be conducive to increasing the immunogenicity of the HA stem antigen. In this study, we generated chimeric virus-like particles (cVLPs) co-displaying the truncated C-terminal of DnaK from Escherichia coli and H1 stem or full-length H1 antigen using the baculovirus expression system. Transmission electronic micrography revealed the expression and presentation of H1 stem antigens on the surface of VLPs. Vaccinations of mice with the H1 stem cVLPs induced H1-specific immune responses and provided heterologous immune protection in vivo, which was more effective than vaccinations with VLPs displaying H1 stem alone in protecting mice against weight loss as well as increasing survival rates after lethal influenza viral challenge. The results indicate that the incorporation of the truncated C-terminal of DnaK as an adjuvant protein into the cVLPs significantly enhances the H1-specific immunity and immune protection. We have explicitly identified the VLP platform as an effective way of expressing HA stem antigen and revealed that chimeric VLP is an vaccine vector for developing HA stem-based universal influenza vaccines.
Collapse
|
41
|
Oh J, Subbiah J, Kim KH, Park BR, Bhatnagar N, Garcia KR, Liu R, Jung YJ, Shin CH, Seong BL, Kang SM. Impact of hemagglutination activity and M2e immunity on conferring protection against influenza viruses. Virology 2022; 574:37-46. [PMID: 35914365 PMCID: PMC9978532 DOI: 10.1016/j.virol.2022.07.010] [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: 04/21/2022] [Revised: 07/05/2022] [Accepted: 07/13/2022] [Indexed: 11/17/2022]
Abstract
To improve cross-protection of influenza vaccination, we tested conjugation of conserved M2e epitopes to the surface of inactivated influenza virus (iPR8-M2e*). Treatment of virus with chemical cross-linker led to diminished hemagglutination activity and failure to induce hemagglutination inhibiting antibodies. Conjugated iPR8-M2e* vaccine was less protective against homologous and heterosubtypic viruses, despite the induction of virus-specific binding IgG antibodies. In alternative approaches to enhance cross-protection, we developed a genetically linked chimeric protein (M2e-B stalk) vaccine with M2e of influenza A and hemagglutinin (HA) stalk of influenza B virus. Vaccination of mice with inactivated influenza A virus supplemented with M2e-B stalk effectively induced hemagglutination inhibiting antibodies, humoral and cellular M2e immune responses, and enhanced heterosubtypic protection. This study demonstrates the importance of HA functional integrity in influenza vaccine efficacy and that supplementation of influenza vaccines with M2e-B stalk protein could be a feasible strategy of improving cross-protection against influenza viruses.
Collapse
Affiliation(s)
- Judy Oh
- Center for Inflammation, Immunity, and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, 30303, USA
| | - Jeeva Subbiah
- Center for Inflammation, Immunity, and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, 30303, USA
| | - Ki-Hye Kim
- Center for Inflammation, Immunity, and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, 30303, USA
| | - Bo Ryoung Park
- Center for Inflammation, Immunity, and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, 30303, USA
| | - Noopur Bhatnagar
- Center for Inflammation, Immunity, and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, 30303, USA
| | - Karla Ruiz Garcia
- Center for Inflammation, Immunity, and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, 30303, USA
| | - Rong Liu
- Center for Inflammation, Immunity, and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, 30303, USA
| | - Yu-Jin Jung
- Center for Inflammation, Immunity, and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, 30303, USA
| | - Chong-Hyun Shin
- Center for Inflammation, Immunity, and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, 30303, USA
| | - Baik-Lin Seong
- Department of Microbiology, College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea; Vaccine Innovative Technology ALliance (VITAL)-Korea, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Sang-Moo Kang
- Center for Inflammation, Immunity, and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, 30303, USA.
| |
Collapse
|
42
|
Weiss AM, Hossainy S, Rowan SJ, Hubbell JA, Esser-Kahn AP. Immunostimulatory Polymers as Adjuvants, Immunotherapies, and Delivery Systems. Macromolecules 2022; 55:6913-6937. [PMID: 36034324 PMCID: PMC9404695 DOI: 10.1021/acs.macromol.2c00854] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 07/16/2022] [Indexed: 12/14/2022]
Abstract
![]()
Activating innate immunity in a controlled manner is
necessary
for the development of next-generation therapeutics. Adjuvants, or
molecules that modulate the immune response, are critical components
of vaccines and immunotherapies. While small molecules and biologics
dominate the adjuvant market, emerging evidence supports the use of
immunostimulatory polymers in therapeutics. Such polymers can stabilize
and deliver cargo while stimulating the immune system by functioning
as pattern recognition receptor (PRR) agonists. At the same time,
in designing polymers that engage the immune system, it is important
to consider any unintended initiation of an immune response that results
in adverse immune-related events. Here, we highlight biologically
derived and synthetic polymer scaffolds, as well as polymer–adjuvant
systems and stimuli-responsive polymers loaded with adjuvants, that
can invoke an immune response. We present synthetic considerations
for the design of such immunostimulatory polymers, outline methods
to target their delivery, and discuss their application in therapeutics.
Finally, we conclude with our opinions on the design of next-generation
immunostimulatory polymers, new applications of immunostimulatory
polymers, and the development of improved preclinical immunocompatibility
tests for new polymers.
Collapse
Affiliation(s)
- Adam M. Weiss
- Pritzker School of Molecular Engineering, University of Chicago 5640 S. Ellis Ave., Chicago, Illinois 60637, United States
- Department of Chemistry, University of Chicago 5735 S Ellis Ave., Chicago, Illinois 60637, United States
| | - Samir Hossainy
- Pritzker School of Molecular Engineering, University of Chicago 5640 S. Ellis Ave., Chicago, Illinois 60637, United States
| | - Stuart J. Rowan
- Pritzker School of Molecular Engineering, University of Chicago 5640 S. Ellis Ave., Chicago, Illinois 60637, United States
- Department of Chemistry, University of Chicago 5735 S Ellis Ave., Chicago, Illinois 60637, United States
| | - Jeffrey A. Hubbell
- Pritzker School of Molecular Engineering, University of Chicago 5640 S. Ellis Ave., Chicago, Illinois 60637, United States
| | - Aaron P. Esser-Kahn
- Pritzker School of Molecular Engineering, University of Chicago 5640 S. Ellis Ave., Chicago, Illinois 60637, United States
| |
Collapse
|
43
|
Song Y, Zhu W, Wang Y, Deng L, Ma Y, Dong C, Gonzalez GX, Kim J, Wei L, Kang SM, Wang BZ. Layered protein nanoparticles containing influenza B HA stalk induced sustained cross-protection against viruses spanning both viral lineages. Biomaterials 2022; 287:121664. [PMID: 35810540 PMCID: PMC9822777 DOI: 10.1016/j.biomaterials.2022.121664] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 06/25/2022] [Accepted: 06/28/2022] [Indexed: 01/11/2023]
Abstract
The influenza epidemics pose a significant threat to public health. Of them, type B influenza coincided with several severe flu outbreaks. The efficacy of the current seasonal flu vaccine is limited due to the antigenicity changes of circulating strains. In this study, we generated structure-stabilized HA stalk antigens from influenza B and fabricated double-layered protein nanoparticles as universal influenza B vaccine candidates. In vitro studies found that the resulting protein nanoparticles were effectively taken up to activate dendritic cells. Nanoparticle immunization induced broadly reactive immune responses conferring robust and sustained cross-immune protection against influenza B virus strains of both lineages. The results reveal the potential of layered protein nanoparticles incorporated with structure-stabilized constant antigens as a universal influenza vaccine with improved immune protective potency and breadth.
Collapse
Affiliation(s)
- Yufeng Song
- Center for Inflammation, Immunity & Infection, Georgia State University, Atlanta, GA 30303, USA
| | - Wandi Zhu
- Center for Inflammation, Immunity & Infection, Georgia State University, Atlanta, GA 30303, USA
| | - Ye Wang
- Center for Inflammation, Immunity & Infection, Georgia State University, Atlanta, GA 30303, USA
| | - Lei Deng
- Center for Inflammation, Immunity & Infection, Georgia State University, Atlanta, GA 30303, USA; Hunan Provincial Kay Laboratory of Medical Virology, Institute of Pathogen Biology and Immunology, College of Biology, Hunan University, Changsha, 410082, China
| | - Yao Ma
- Center for Inflammation, Immunity & Infection, Georgia State University, Atlanta, GA 30303, USA
| | - Chunhong Dong
- Center for Inflammation, Immunity & Infection, Georgia State University, Atlanta, GA 30303, USA
| | - Gilbert X Gonzalez
- Center for Inflammation, Immunity & Infection, Georgia State University, Atlanta, GA 30303, USA
| | - Joo Kim
- Center for Inflammation, Immunity & Infection, Georgia State University, Atlanta, GA 30303, USA
| | - Lai Wei
- Center for Inflammation, Immunity & Infection, Georgia State University, Atlanta, GA 30303, USA
| | - Sang-Moo Kang
- Center for Inflammation, Immunity & Infection, Georgia State University, Atlanta, GA 30303, USA
| | - Bao-Zhong Wang
- Center for Inflammation, Immunity & Infection, Georgia State University, Atlanta, GA 30303, USA.
| |
Collapse
|
44
|
Zhang Y, Jiang S, Lin J, Huang P. Antineoplastic Enzyme as Drug Carrier with Activatable Catalytic Activity for Efficient Combined Therapy. Angew Chem Int Ed Engl 2022. [DOI: 10.1002/ange.202208583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Yifan Zhang
- Shenzhen University School of Medicine CHINA
| | | | - Jing Lin
- Shenzhen University School of Medicine CHINA
| | - Peng Huang
- Shenzhen University 3688 Nanhai Ave, Nanshan 518060 Shenzhen CHINA
| |
Collapse
|
45
|
Subbiah J, Oh J, Kim KH, Shin CH, Park BR, Bhatnagar N, Seong BL, Wang BZ, Kang SM. A chimeric thermostable M2e and H3 stalk-based universal influenza A virus vaccine. NPJ Vaccines 2022; 7:68. [PMID: 35768475 PMCID: PMC9243060 DOI: 10.1038/s41541-022-00498-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 05/16/2022] [Indexed: 12/30/2022] Open
Abstract
We developed a new chimeric M2e and H3 hemagglutinin (HA) stalk protein vaccine (M2e-H3 stalk) by genetic engineering of modified H3 stalk domain conjugated with conserved M2e epitopes to overcome the drawbacks of low efficacy by monomeric domain-based universal vaccines. M2e-H3 stalk protein expressed and purified from Escherichia coli was thermostable, displaying native-like antigenic epitopes recognized by antisera of different HA subtype proteins and influenza A virus infections. Adjuvanted M2e-H3 stalk vaccination induced M2e and stalk-specific IgG antibodies recognizing viral antigens on virus particles and on the infected cell surface, CD4+ and CD8+ T-cell responses, and antibody-dependent cytotoxic cell surrogate activity in mice. M2e-H3 stalk was found to confer protection against heterologous and heterosubtypic cross-group subtype viruses (H1N1, H5N1, H9N2, H3N2, H7N9) at similar levels in adult and aged mice. These results provide evidence that M2e-H3 stalk chimeric proteins can be developed as a universal influenza A virus vaccine candidate for young and aged populations.
Collapse
Affiliation(s)
- Jeeva Subbiah
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, 30303, USA
| | - Judy Oh
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, 30303, USA
| | - Ki-Hye Kim
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, 30303, USA
| | - Chong-Hyun Shin
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, 30303, USA
| | - Bo Ryoung Park
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, 30303, USA
| | - Noopur Bhatnagar
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, 30303, USA
| | - Baik-Lin Seong
- Department of Microbiology, College of Medicine, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea.,Vaccine Innovative Technology ALliance (VITAL)-Korea, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Bao-Zhong Wang
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, 30303, USA
| | - Sang-Moo Kang
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, 30303, USA.
| |
Collapse
|
46
|
Mittal D, Ali SA. Use of Nanomaterials for Diagnosis and Treatment: The Advancement of Next-Generation Antiviral Therapy. Microb Drug Resist 2022; 28:670-697. [PMID: 35696335 DOI: 10.1089/mdr.2021.0281] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Globally, viral illness propagation is the leading cause of morbidity and death, causing wreaking havoc on socioeconomic development and health care systems. The rise of infected individuals has outpaced the existing critical care facilities. Early and sophisticated methods are desperately required in this respect to halt the spread of the infection. Therefore, early detection of infectious agents and an early treatment approach may help minimize viral outbreaks. Conventional point-of-care diagnostic techniques such as computed tomography scan, quantitative real time polymerase chain reaction (qRT-PCR), X-ray, and immunoassay are still deemed valuable. However, the labor demanding, low sensitivity, and complex infrastructure needed for these methods preclude their use in distant areas. Nanotechnology has emerged as a potentially transformative technology due to its promise as an effective theranostic platform for diagnosing and treating viral infection, circumventing the limits of traditional techniques. Their unique physical and chemical characteristics make nanoparticles (NPs) advantageous for drug delivery platforms due to their size, encapsulation efficiency, improved bioavailability, effectiveness, immunogenicity, and antiviral response. This study discusses the recent research on nanotechnology-based treatments designed to combat new viruses.
Collapse
Affiliation(s)
- Deepti Mittal
- Nanosafety Lab, Division of Biochemistry, ICAR-NDRI, Karnal, Haryana, India
| | - Syed Azmal Ali
- Cell Biology and Proteomics Lab, Animal Biotechnology Center, ICAR-NDRI, Karnal, Haryana, India
| |
Collapse
|
47
|
Subbbiah J, Oh J, Kim KH, Shin CH, Park BR, Bhatnagar N, Jung YJ, Lee Y, Wang BZ, Seong BL, Kang SM. Thermostable H1 hemagglutinin stem with M2e epitopes provides broad cross-protection against group1 and 2 influenza A viruses. Mol Ther Methods Clin Dev 2022; 26:38-51. [PMID: 35755946 PMCID: PMC9198381 DOI: 10.1016/j.omtm.2022.05.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 05/25/2022] [Indexed: 11/29/2022]
Abstract
Hemagglutinin (HA) stem-based vaccines have limitations in providing broad and effective protection against cross-group influenza viruses, despite being a promising universal vaccine target. To overcome the limited cross-protection and low efficacy by HA stem vaccination, we genetically engineered a chimeric conjugate of thermostable H1 HA stem and highly conserved M2e repeat (M2e-H1stem), which was expressed at high yields in Escherichia coli. M2e-H1stem protein presented native-like epitopes reactive to antisera of live virus infection. M2e-H1stem protein vaccination of mice induced strong M2e- and HA stem-specific immune responses, conferring broadly effective cross-protection against both antigenically distinct group 1 (H1N1, H5N1, and H9N2 subtypes) and group 2 (H3N2 and H7N9 subtypes) seasonal and pandemic potential influenza viruses. M2e-H1stem vaccination generated CD4+ and CD8+ T cell responses and antibody-dependent cytotoxic cellular and humoral immunity, which contributed to enhancing cross-protection. Furthermore, comparable broad cross-group protection was observed in older aged mice after M2e-H1stem vaccination. This study provides evidence warranting further development of chimeric M2e-stem proteins as a promising universal influenza vaccine candidate in adult and aged populations.
Collapse
Affiliation(s)
- Jeeva Subbbiah
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, 100 Piedmont Avenue, PSC 718 P.O. Box 5035, Atlanta, GA 30303, USA
| | - Judy Oh
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, 100 Piedmont Avenue, PSC 718 P.O. Box 5035, Atlanta, GA 30303, USA
| | - Ki-Hye Kim
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, 100 Piedmont Avenue, PSC 718 P.O. Box 5035, Atlanta, GA 30303, USA
| | - Chong Hyun Shin
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, 100 Piedmont Avenue, PSC 718 P.O. Box 5035, Atlanta, GA 30303, USA
| | - Bo Ryoung Park
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, 100 Piedmont Avenue, PSC 718 P.O. Box 5035, Atlanta, GA 30303, USA
| | - Noopur Bhatnagar
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, 100 Piedmont Avenue, PSC 718 P.O. Box 5035, Atlanta, GA 30303, USA
| | - Yu-Jin Jung
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, 100 Piedmont Avenue, PSC 718 P.O. Box 5035, Atlanta, GA 30303, USA
| | - Youri Lee
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, 100 Piedmont Avenue, PSC 718 P.O. Box 5035, Atlanta, GA 30303, USA
| | - Bao-Zhong Wang
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, 100 Piedmont Avenue, PSC 718 P.O. Box 5035, Atlanta, GA 30303, USA
| | - Baik-Lin Seong
- Department of Microbiology, College of Medicine, Yonsei University, Seoul 03722, Republic of Korea
- Vaccine Innovative Technology Alliance (VITAL), Seoul 03722, Republic of Korea
| | - Sang-Moo Kang
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, 100 Piedmont Avenue, PSC 718 P.O. Box 5035, Atlanta, GA 30303, USA
- Corresponding author Sang-Moo Kang, PhD, Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, 100 Piedmont Avenue, PSC 718 P.O. Box 5035, Atlanta, GA 30303, USA.
| |
Collapse
|
48
|
Kar U, Khaleeq S, Garg P, Bhat M, Reddy P, Vignesh VS, Upadhyaya A, Das M, Chakshusmathi G, Pandey S, Dutta S, Varadarajan R. Comparative Immunogenicity of Bacterially Expressed Soluble Trimers and Nanoparticle Displayed Influenza Hemagglutinin Stem Immunogens. Front Immunol 2022; 13:890622. [PMID: 35720346 PMCID: PMC9204493 DOI: 10.3389/fimmu.2022.890622] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Accepted: 04/14/2022] [Indexed: 11/13/2022] Open
Abstract
Current influenza vaccines need to be updated annually due to mutations in the globular head of the viral surface protein, hemagglutinin (HA). To address this, vaccine candidates have been designed based on the relatively conserved HA stem domain and have shown protective efficacy in animal models. Oligomerization of the antigens either by fusion to oligomerization motifs or display on self-assembling nanoparticle scaffolds, can induce more potent immune responses compared to the corresponding monomeric antigen due to multivalent engagement of B-cells. Since nanoparticle display can increase manufacturing complexity, and often involves one or more mammalian cell expressed components, it is important to characterize and compare various display and oligomerization scaffolds. Using a structure guided approach, we successfully displayed multiple copies of a previously designed soluble, trimeric influenza stem domain immunogen, pH1HA10, on the ferritin like protein, MsDps2 (12 copies), Ferritin (24 copies) and Encapsulin (180 copies). All proteins were expressed in Escherichia coli. The nanoparticle fusion immunogens were found to be well folded and bound to the influenza stem directed broadly neutralizing antibodies with high affinity. An 8.5 Å Cryo-EM map of Msdps2-pH1HA10 confirmed the successful design of the nanoparticle fusion immunogen. Mice immunization studies with the soluble trimeric stem and nanoparticle fusion constructs revealed that all of them were immunogenic, and protected mice against homologous (A/Belgium/145-MA/2009) and heterologous (A/Puerto Rico/8/1934) challenge with 10MLD50 mouse adapted virus. Although nanoparticle display conferred a small but statistically significant improvement in protection relative to the soluble trimer in a homologous challenge, heterologous protection was similar in both nanoparticle-stem immunized and trimeric stem immunized groups. Such rapidly producible, bacterially expressed antigens and nanoparticle scaffolds are useful modalities to tackle future influenza pandemics.
Collapse
Affiliation(s)
- Uddipan Kar
- Molecular Biophysics Unit (MBU), Indian Institute of Science, Bengaluru, India
| | - Sara Khaleeq
- Molecular Biophysics Unit (MBU), Indian Institute of Science, Bengaluru, India
| | - Priyanka Garg
- Molecular Biophysics Unit (MBU), Indian Institute of Science, Bengaluru, India
| | - Madhuraj Bhat
- Mynvax Private Limited, ES12, Entrepreneurship Centre, Society for Innovation and Development (SID), Indian Institute of Science, Bengaluru, India
| | - Poorvi Reddy
- Mynvax Private Limited, ES12, Entrepreneurship Centre, Society for Innovation and Development (SID), Indian Institute of Science, Bengaluru, India
| | | | - Aditya Upadhyaya
- Mynvax Private Limited, ES12, Entrepreneurship Centre, Society for Innovation and Development (SID), Indian Institute of Science, Bengaluru, India
| | - Mili Das
- Mynvax Private Limited, ES12, Entrepreneurship Centre, Society for Innovation and Development (SID), Indian Institute of Science, Bengaluru, India
| | - Ghadiyaram Chakshusmathi
- Mynvax Private Limited, ES12, Entrepreneurship Centre, Society for Innovation and Development (SID), Indian Institute of Science, Bengaluru, India
| | - Suman Pandey
- Mynvax Private Limited, ES12, Entrepreneurship Centre, Society for Innovation and Development (SID), Indian Institute of Science, Bengaluru, India
| | - Somnath Dutta
- Molecular Biophysics Unit (MBU), Indian Institute of Science, Bengaluru, India
| | | |
Collapse
|
49
|
Zhao Y, Li Z, Voyer J, Li Y, Chen X. Flagellin/Virus-like Particle Hybrid Platform with High Immunogenicity, Safety, and Versatility for Vaccine Development. ACS APPLIED MATERIALS & INTERFACES 2022; 14:21872-21885. [PMID: 35467839 PMCID: PMC9121874 DOI: 10.1021/acsami.2c01028] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 04/13/2022] [Indexed: 05/07/2023]
Abstract
Hepatitis B core (HBc) virus-like particles (VLPs) and flagellin are highly immunogenic and widely explored vaccine delivery platforms. Yet, HBc VLPs mainly allow the insertion of relatively short antigenic epitopes into the immunodominant c/e1 loop without affecting VLP assembly, and flagellin-based vaccines carry the risk of inducing systemic adverse reactions. This study explored a hybrid flagellin/HBc VLP (FH VLP) platform to present heterologous antigens by replacing the surface-exposed D3 domain of flagellin. FH VLPs were prepared by the insertion of flagellin gene into the c/e1 loop of HBc, followed by E. coli expression, purification, and self-assembly into VLPs. Using the ectodomain of influenza matrix protein 2 (M2e) and ovalbumin (OVA) as models, we found that the D3 domain of flagellin could be replaced with four tandem copies of M2e or the cytotoxic T lymphocyte (CTL) epitope of OVA without interfering with the FH VLP assembly, while the insertion of four tandem copies of M2e into the c/e1 loop of HBc disrupted the VLP assembly. FH VLP-based M2e vaccine elicited potent anti-M2e antibody responses and conferred significant protection against multiple influenza A viral strains, while FljB- or HBc-based M2e vaccine failed to elicit significant protection. FH VLP-based OVA peptide vaccine elicited more potent CTL responses and protection against OVA-expressing lymphoma or melanoma challenges than FljB- or HBc-based OVA peptide vaccine. FH VLP-based vaccines showed a good systemic safety, while flagellin-based vaccines significantly increased serum interleukin 6 and tumor necrosis factor α levels and also rectal temperature at increased doses. We further found that the incorporation of a clinical CpG 1018 adjuvant could enhance the efficacy of FH VLP-based vaccines. Our data support FH VLPs to be a highly immunogenic, safe, and versatile platform for vaccine development to elicit potent humoral and cellular immune responses.
Collapse
Affiliation(s)
- Yiwen Zhao
- Biomedical & Pharmaceutical
Sciences, College of Pharmacy, University
of Rhode Island, 7 Greenhouse Road, Avedisian Hall, Room 480, Kingston, Rhode Island 02881, United States
| | - Zhuofan Li
- Biomedical & Pharmaceutical
Sciences, College of Pharmacy, University
of Rhode Island, 7 Greenhouse Road, Avedisian Hall, Room 480, Kingston, Rhode Island 02881, United States
| | - Jewel Voyer
- Biomedical & Pharmaceutical
Sciences, College of Pharmacy, University
of Rhode Island, 7 Greenhouse Road, Avedisian Hall, Room 480, Kingston, Rhode Island 02881, United States
| | - Yibo Li
- Biomedical & Pharmaceutical
Sciences, College of Pharmacy, University
of Rhode Island, 7 Greenhouse Road, Avedisian Hall, Room 480, Kingston, Rhode Island 02881, United States
| | - Xinyuan Chen
- Biomedical & Pharmaceutical
Sciences, College of Pharmacy, University
of Rhode Island, 7 Greenhouse Road, Avedisian Hall, Room 480, Kingston, Rhode Island 02881, United States
| |
Collapse
|
50
|
Kong D, Chen T, Hu X, Lin S, Gao Y, Ju C, Liao M, Fan H. Supplementation of H7N9 Virus-Like Particle Vaccine With Recombinant Epitope Antigen Confers Full Protection Against Antigenically Divergent H7N9 Virus in Chickens. Front Immunol 2022; 13:785975. [PMID: 35265069 PMCID: PMC8898936 DOI: 10.3389/fimmu.2022.785975] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 01/27/2022] [Indexed: 01/18/2023] Open
Abstract
The continuous evolution of the H7N9 avian influenza virus suggests a potential outbreak of an H7N9 pandemic. Therefore, to prevent a potential epidemic of the H7N9 influenza virus, it is necessary to develop an effective crossprotective influenza vaccine. In this study, we developed H7N9 virus-like particles (VLPs) containing HA, NA, and M1 proteins derived from H7N9/16876 virus and a helper antigen HMN based on influenza conserved epitopes using a baculovirus expression vector system (BEVS). The results showed that the influenza VLP vaccine induced a strong HI antibody response and provided effective protection comparable with the effects of commercial inactivated H7N9 vaccines against homologous H7N9 virus challenge in chickens. Meanwhile, the H7N9 VLP vaccine induced robust crossreactive HI and neutralizing antibody titers against antigenically divergent H7N9 viruses isolated in wave 5 and conferred on chickens complete clinical protection against heterologous H7N9 virus challenge, significantly inhibiting virus shedding in chickens. Importantly, supplemented vaccination with HMN antigen can enhance Th1 immune responses; virus shedding was completely abolished in the vaccinated chickens. Our study also demonstrated that viral receptor-binding avidity should be taken into consideration in evaluating an H7N9 candidate vaccine. These studies suggested that supplementing influenza VLP vaccine with recombinant epitope antigen will be a promising strategy for the development of broad-spectrum influenza vaccines.
Collapse
Affiliation(s)
- Dexin Kong
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China.,Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China.,Key Laboratory of Animal Vaccine Development, Ministry of Agriculture, Guangzhou, China.,National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Taoran Chen
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China.,Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China.,Key Laboratory of Animal Vaccine Development, Ministry of Agriculture, Guangzhou, China.,National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Xiaolong Hu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China.,Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China.,Key Laboratory of Animal Vaccine Development, Ministry of Agriculture, Guangzhou, China.,National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Shaorong Lin
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China.,Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China.,Key Laboratory of Animal Vaccine Development, Ministry of Agriculture, Guangzhou, China.,National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Yinze Gao
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China.,Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China.,Key Laboratory of Animal Vaccine Development, Ministry of Agriculture, Guangzhou, China.,National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Chunmei Ju
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Ming Liao
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China.,Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China.,Key Laboratory of Animal Vaccine Development, Ministry of Agriculture, Guangzhou, China.,National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Huiying Fan
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China.,Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China.,Key Laboratory of Animal Vaccine Development, Ministry of Agriculture, Guangzhou, China.,National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| |
Collapse
|