1
|
Mo H, Chen Q, Zhang Z, Lin G, Wang Y, Mo L, Liang W, He J, Su B, Li J, Hu Z, Li H, Pei N, Wu Y, Su H. Development of a blocking ELISA for evaluating neutralizing antibodies in human and canine serum based on rabies virus glycoprotein epitope I. Int J Biol Macromol 2025; 301:140275. [PMID: 39863206 DOI: 10.1016/j.ijbiomac.2025.140275] [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: 10/08/2024] [Revised: 01/18/2025] [Accepted: 01/22/2025] [Indexed: 01/27/2025]
Abstract
Rabies virus (RABV) is extremely hazardous to both humans and animals, causing up to 100 % death. Accurate and easy-to-use serological evaluation of vaccine potency following immunization is crucial for rabies control. In this study, recombinant RABV glycoprotein (rG) was designed and produced in 293FT cells. Subsequently, a monoclonal antibody (S049), against the antigenic epitope I of RABV glycoprotein, was screened. Using the recombinant RABV glycoprotein and S049, a blocking enzyme-linked immunosorbent assay (bELISA) was developed. The rG-encapsulated antigen was optimized to a concentration of 100 ng. Experimental conditions were refined, and the receiver operator characteristic (ROC) curve analysis demonstrated a maximal Youden index of 0.9978 for the canine serum detection, with a critical bELISA value of 23.21 %, specificity of 99.15 %, and sensitivity of 97.06 %. For human serum, the maximum Youden index was 0.9903, with a critical bELISA value of 30.60 %, specificity of 100 %, and sensitivity of 95.65 %. These findings indicate that the blocking ELISA exhibits comparable sensitivity and specificity to the fluorescent antibody virus neutralization test. In conclusion, the present study developed a robust blocking ELISA for post-immunization RABV detection, offering a promising tool for high-throughput sample assessment and surveillance of herd immunity, especially in resource-limited settings.
Collapse
Affiliation(s)
- Haifeng Mo
- School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Qian Chen
- School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Zhigao Zhang
- School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Guanfeng Lin
- School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Yuyan Wang
- Department of Geriatrics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Lijun Mo
- Department of Clinical Laboratory, Dermatology Hospital, Southern Medical University, Guangzhou, China
| | | | - Jiaming He
- School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Bijia Su
- Shunde Hospital, Southern Medical University, Foshan, China
| | - Jinlong Li
- School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Zhiming Hu
- School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Hongwei Li
- School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Nana Pei
- Department of Pathology, The First Affiliated Hospital of Jinan University, Guangzhou, China.
| | - Yingsong Wu
- School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China.
| | - Hailong Su
- Department of Immunology, Zunyi Medical University, Guizhou, China; School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China.
| |
Collapse
|
2
|
Khalifa ME, Munir M. Protocol for constructing and characterizing recombinant vectored vaccines for rabies virus. STAR Protoc 2024; 5:103392. [PMID: 39423123 PMCID: PMC11513535 DOI: 10.1016/j.xpro.2024.103392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Revised: 06/30/2024] [Accepted: 09/24/2024] [Indexed: 10/21/2024] Open
Abstract
Recombinant vectored vaccines provide economical, rapidly engineered, and tailored immunization strategies. Here, we present a protocol for constructing and characterizing recombinant vectored vaccines for rabies virus (RV). We describe steps for generating replication-competent vesicular stomatitis virus (rcVSV) by replacing vesicular stomatitis virus (VSV) glycoprotein (G) with RV G gene and for recovering rcVSV by co-transfecting baby hamster kidney (BHK-21) cells with the antigenomic VSV cDNA plasmid (pVSV). We then detail procedures for the functional, structural, and molecular characterization of generated rcVSV-RV-Gs.
Collapse
Affiliation(s)
- Manar E Khalifa
- Division of Biomedical and Life Sciences, Faculty of Health and Medicine, Lancaster University, Lancaster LA1 4YG, UK.
| | - Muhammad Munir
- Division of Biomedical and Life Sciences, Faculty of Health and Medicine, Lancaster University, Lancaster LA1 4YG, UK.
| |
Collapse
|
3
|
Zhang HQ, Zhang YN, Deng CL, Zhu QX, Zhang ZR, Li XD, Yuan ZM, Zhang B. Rational design of self-amplifying virus-like vesicles with Ebola virus glycoprotein as vaccines. Mol Ther 2024; 32:3695-3711. [PMID: 39217415 PMCID: PMC11489537 DOI: 10.1016/j.ymthe.2024.08.026] [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: 12/18/2023] [Revised: 07/13/2024] [Accepted: 08/29/2024] [Indexed: 09/04/2024] Open
Abstract
As emerging and re-emerging pathogens, filoviruses, especially Ebola virus (EBOV), pose a great threat to public health and require sustained attention and ongoing surveillance. More vaccines and antiviral drugs are imperative to be developed and stockpiled to respond to unpredictable outbreaks. Virus-like vesicles, generated by alphavirus replicons expressing homogeneous or heterogeneous glycoproteins (GPs), have demonstrated the capacity of self-propagation and shown great potential in vaccine development. Here, we describe a novel class of EBOV-like vesicles (eVLVs) incorporating both EBOV GP and VP40. The eVLVs exhibited similar antigenicity as EBOV. In murine models, eVLVs were highly attenuated and elicited robust GP-specific antibodies with neutralizing activities. Importantly, a single dose of eVLVs conferred complete protection in a surrogate EBOV lethal mouse model. Furthermore, our VLVs strategy was also successfully applied to Marburg virus (MARV), the representative member of the genus Marburgvirus. Taken together, our findings indicate the feasibility of an alphavirus-derived VLVs strategy in combating infection of filoviruses represented by EBOV and MARV, which provides further evidence of the potential of this platform for universal live-attenuated vaccine development.
Collapse
Affiliation(s)
- Hong-Qing Zhang
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430062, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ya-Nan Zhang
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430062, China; Hubei Jiangxia Laboratory, Wuhan 430200, China
| | - Cheng-Lin Deng
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430062, China
| | - Qin-Xuan Zhu
- Hunan Normal University, School of Medicine, Changsha 410081, China
| | - Zhe-Rui Zhang
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430062, China
| | - Xiao-Dan Li
- Hunan Normal University, School of Medicine, Changsha 410081, China
| | - Zhi-Ming Yuan
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430062, China; University of Chinese Academy of Sciences, Beijing 100049, China; Hubei Jiangxia Laboratory, Wuhan 430200, China
| | - Bo Zhang
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430062, China; University of Chinese Academy of Sciences, Beijing 100049, China; Hubei Jiangxia Laboratory, Wuhan 430200, China.
| |
Collapse
|
4
|
Wang Y, Su M, Huang Y, Ren J, Niu S, Zhao Y, Yan F, Yan Y, Tian WX. Development of a novel PCV2 and PCV3 vaccine using virus-like vesicles incorporating Venezuelan equine encephalomyelitis virus-containing vesicular stomatitis virus glycoprotein. Front Vet Sci 2024; 11:1359421. [PMID: 38840631 PMCID: PMC11150706 DOI: 10.3389/fvets.2024.1359421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 05/06/2024] [Indexed: 06/07/2024] Open
Abstract
Porcine circovirus disease (PCV) causes substantial economic losses in the pig industry, primarily from porcine circovirus type 2 (PCV2) and porcine circovirus type 3 (PCV3). Novel vaccines are necessary to prevent and control PCV infections. PCV coat proteins are crucial for eliciting immunogenic proteins that induce the production of antibodies and immune responses. A vaccine platform utilizing Semliki Forest virus RNA replicons expressing vesicular stomatitis virus glycoprotein (VSV-G), was recently developed. This platform generates virus-like vesicles (VLVs) containing VSV-G exclusively, excluding other viral structural proteins. In our study, we developed a novel virus-like vesicle vaccine by constructing recombinant virus-like vesicles (rVLVs) that also express EGFP. These rVLVs were created using the RNA replicon of Venezuelan equine encephalomyelitis (VEEV) and New Jersey serotype VSV-G. The rVLVs underwent characterization and safety evaluation in vitro. Subsequently, rVLVs expressing PCV2d-Cap and PCV3-Cap proteins were constructed. Immunization of C57 mice with these rVLVs led to a significant increase in anti-porcine circovirus type 2 and type 3 capsid protein antibodies in mouse serum. Additionally, a cellular immune response was induced, as evidenced by high production of IFN-γ and IL-4 cytokines. Overall, this study demonstrates the feasibility of developing a novel porcine circovirus disease vaccine based on rVLVs.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Yi Yan
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China
| | - Wen-xia Tian
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, China
| |
Collapse
|
5
|
Wan J, Wang Z, Wang L, Wu L, Zhang C, Zhou M, Fu ZF, Zhao L. Circular RNA vaccines with long-term lymph node-targeting delivery stability after lyophilization induce potent and persistent immune responses. mBio 2024; 15:e0177523. [PMID: 38078742 PMCID: PMC10790773 DOI: 10.1128/mbio.01775-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Accepted: 10/26/2023] [Indexed: 01/17/2024] Open
Abstract
IMPORTANCE messenger RNA (mRNA) vaccines are a key technology in combating existing and emerging infectious diseases. However, the inherent instability of mRNA and the nonspecificity of lipid nanoparticle-encapsulated (LNP) delivery systems result in the need for cold storage and a relatively short-duration immune response to mRNA vaccines. Herein, we develop a novel vaccine in the form of circRNAs encapsulated in LNPs, and the circular structure of the circRNAs enhances their stability. Lyophilization is considered the most effective method for the long-term preservation of RNA vaccines. However, this process may result in irreversible damage to the nanoparticles, particularly the potential disruption of targeting modifications on LNPs. During the selection of lymph node-targeting ligands, we found that LNPs modified with mannose maintained their physical properties almost unchanged after lyophilization. Additionally, the targeting specificity and immunogenicity remained unaffected. In contrast, even with the addition of cryoprotectants such as sucrose, the physical properties of LNPs were impaired, leading to an obvious decrease in immunogenicity. This may be attributed to the protective role of mannose on the surface of LNPs during lyophilization. Freshly prepared and lyophilized mLNP-circRNA vaccines elicited comparable immune responses in both the rabies virus model and the SARS-CoV-2 model. Our data demonstrated that mLNP-circRNA vaccines elicit robust immune responses while improving stability after lyophilization, with no compromise in tissue targeting specificity. Therefore, mannose-modified LNP-circRNA vaccines represent a promising vaccine design strategy.
Collapse
Affiliation(s)
- Jiawu Wan
- National Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- Hubei Hongshan Laboratory, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Zongmei Wang
- National Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- Hubei Hongshan Laboratory, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Lingli Wang
- National Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- Hubei Hongshan Laboratory, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Liqin Wu
- National Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- Hubei Hongshan Laboratory, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Chengguang Zhang
- National Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- Hubei Hongshan Laboratory, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Ming Zhou
- National Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- Hubei Hongshan Laboratory, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Zhen F. Fu
- National Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- Hubei Hongshan Laboratory, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Ling Zhao
- National Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- Hubei Hongshan Laboratory, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| |
Collapse
|
6
|
Zhang HQ, Li N, Zhang ZR, Deng CL, Xia H, Ye HQ, Yuan ZM, Zhang B. A Chimeric Classical Insect-Specific Flavivirus Provides Complete Protection Against West Nile Virus Lethal Challenge in Mice. J Infect Dis 2024; 229:43-53. [PMID: 37368353 DOI: 10.1093/infdis/jiad238] [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: 02/15/2023] [Revised: 06/15/2023] [Accepted: 06/23/2023] [Indexed: 06/28/2023] Open
Abstract
West Nile virus (WNV), an arthropod-borne flavivirus, can cause severe symptoms, including encephalitis, and death, posing a threat to public health and the economy. However, there is still no approved treatment or vaccine available for humans. Here, we developed a novel vaccine platform based on a classical insect-specific flavivirus (cISF) YN15-283-02, which was derived from Culicoides. The cISF-WNV chimera was constructed by replacing prME structural genes of the infectious YN15-283-02 cDNA clone with those of WNV and successfully rescued in Aedes albopictus cells. cISF-WNV was nonreplicable in vertebrate cells and nonpathogenic in type I interferon receptor (IFNAR)-deficient mice. A single-dose immunization of cISF-WNV elicited considerable Th1-biased antibody responses in C57BL/6 mice, which was sufficient to offer complete protection against lethal WNV challenge with no symptoms. Our studies demonstrated the potential of the insect-specific cISF-WNV as a prophylactic vaccine candidate to prevent infection with WNV.
Collapse
Affiliation(s)
- Hong-Qing Zhang
- Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Na Li
- Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China
| | - Zhe-Rui Zhang
- Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China
| | - Cheng-Lin Deng
- Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China
| | - Han Xia
- Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China
- University of Chinese Academy of Sciences, Beijing, China
- Hubei Jiangxia Laboratory, Wuhan, China
| | - Han-Qing Ye
- Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Zhi-Ming Yuan
- Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Bo Zhang
- Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China
- University of Chinese Academy of Sciences, Beijing, China
- Hubei Jiangxia Laboratory, Wuhan, China
| |
Collapse
|
7
|
Wan J, Yang J, Wang Z, Shen R, Zhang C, Wu Y, Zhou M, Chen H, Fu ZF, Sun H, Yi Y, Shen H, Li H, Zhao L. A single immunization with core-shell structured lipopolyplex mRNA vaccine against rabies induces potent humoral immunity in mice and dogs. Emerg Microbes Infect 2023; 12:2270081. [PMID: 37819147 PMCID: PMC10768744 DOI: 10.1080/22221751.2023.2270081] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Accepted: 10/05/2023] [Indexed: 10/13/2023]
Abstract
The persistence and clinical consequences of rabies virus (RABV) infection have prompted global efforts to develop a safe and effective vaccines against rabies. mRNA vaccines represent a promising option against emerging and re-emerging infectious diseases, gaining particular interest since the outbreak of COVID-19. Herein, we report the development of a highly efficacious rabies mRNA vaccine composed of sequence-modified mRNA encoding RABV glycoprotein (RABV-G) packaged in core-shell structured lipopolyplex (LPP) nanoparticles, named LPP-mRNA-G. The bilayer structure of LPP improves protection and delivery of RABV-G mRNA and allows gradual release of mRNA molecules as the polymer degrades. The unique core-shell structured nanoparticle of LPP-mRNA-G facilitates vaccine uptake and demonstrates a desirable biodistribution pattern with low liver targeting upon intramuscular immunization. Single administration of low-dose LPP-mRNA-G in mice elicited potent humoral immune response and provided complete protection against intracerebral challenge with lethal RABV. Similarly, single immunization of low-dose LPP-mRNA-G induced high levels of virus-neutralizing antibody titers in dogs. Collectively, our data demonstrate the potential of LPP-mRNA-G as a promising next-generation rabies vaccine used in human and companion animals.
Collapse
Affiliation(s)
- Jiawu Wan
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, People’s Republic of China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, People’s Republic of China
- Hubei Hongshan Laboratory, Wuhan, People’s Republic of China
- Frontiers Science Center for Animal Breeding and Sustainable Production, Wuhan, People’s Republic of China
| | - Jianmei Yang
- Stemirna Therapeutics, Shanghai, People’s Republic of China
| | - Zongmei Wang
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, People’s Republic of China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, People’s Republic of China
- Hubei Hongshan Laboratory, Wuhan, People’s Republic of China
- Frontiers Science Center for Animal Breeding and Sustainable Production, Wuhan, People’s Republic of China
| | - Ruizhong Shen
- Stemirna Therapeutics, Shanghai, People’s Republic of China
| | - Chengguang Zhang
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, People’s Republic of China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, People’s Republic of China
- Hubei Hongshan Laboratory, Wuhan, People’s Republic of China
- Frontiers Science Center for Animal Breeding and Sustainable Production, Wuhan, People’s Republic of China
| | - Yuntao Wu
- Stemirna Therapeutics, Shanghai, People’s Republic of China
| | - Ming Zhou
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, People’s Republic of China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, People’s Republic of China
- Hubei Hongshan Laboratory, Wuhan, People’s Republic of China
- Frontiers Science Center for Animal Breeding and Sustainable Production, Wuhan, People’s Republic of China
| | - Huanchun Chen
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, People’s Republic of China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, People’s Republic of China
- Hubei Hongshan Laboratory, Wuhan, People’s Republic of China
- Frontiers Science Center for Animal Breeding and Sustainable Production, Wuhan, People’s Republic of China
| | - Zhen F. Fu
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, People’s Republic of China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, People’s Republic of China
- Hubei Hongshan Laboratory, Wuhan, People’s Republic of China
- Frontiers Science Center for Animal Breeding and Sustainable Production, Wuhan, People’s Republic of China
| | - Haiwei Sun
- Stemirna Therapeutics, Shanghai, People’s Republic of China
| | - Yinglei Yi
- Stemirna Therapeutics, Shanghai, People’s Republic of China
| | - Haifa Shen
- Stemirna Therapeutics, Shanghai, People’s Republic of China
| | - Hangwen Li
- Stemirna Therapeutics, Shanghai, People’s Republic of China
| | - Ling Zhao
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, People’s Republic of China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, People’s Republic of China
- Hubei Hongshan Laboratory, Wuhan, People’s Republic of China
- Frontiers Science Center for Animal Breeding and Sustainable Production, Wuhan, People’s Republic of China
| |
Collapse
|
8
|
Dominguez F, Palchevska O, Frolova EI, Frolov I. Alphavirus-based replicons demonstrate different interactions with host cells and can be optimized to increase protein expression. J Virol 2023; 97:e0122523. [PMID: 37877718 PMCID: PMC10688356 DOI: 10.1128/jvi.01225-23] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 09/18/2023] [Indexed: 10/26/2023] Open
Abstract
IMPORTANCE Alphavirus replicons are being developed as self-amplifying RNAs aimed at improving the efficacy of mRNA vaccines. These replicons are convenient for genetic manipulations and can express heterologous genetic information more efficiently and for a longer time than standard mRNAs. However, replicons mimic many aspects of viral replication in terms of induction of innate immune response, modification of cellular transcription and translation, and expression of nonstructural viral genes. Moreover, all replicons used in this study demonstrated expression of heterologous genes in cell- and replicon's origin-specific modes. Thus, many aspects of the interactions between replicons and the host remain insufficiently investigated, and further studies are needed to understand the biology of the replicons and their applicability for designing a new generation of mRNA vaccines. On the other hand, our data show that replicons are very flexible expression systems, and additional modifications may have strong positive impacts on protein expression.
Collapse
Affiliation(s)
- Francisco Dominguez
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Oksana Palchevska
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Elena I. Frolova
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Ilya Frolov
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
9
|
Comes JDG, Pijlman GP, Hick TAH. Rise of the RNA machines - self-amplification in mRNA vaccine design. Trends Biotechnol 2023; 41:1417-1429. [PMID: 37328401 PMCID: PMC10266560 DOI: 10.1016/j.tibtech.2023.05.007] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 05/09/2023] [Accepted: 05/18/2023] [Indexed: 06/18/2023]
Abstract
mRNA vaccines have won the race for early COVID-19 vaccine approval, yet improvements are necessary to retain this leading role in combating infectious diseases. A next generation of self-amplifying mRNAs, also known as replicons, form an ideal vaccine platform. Replicons induce potent humoral and cellular responses with few adverse effects upon a minimal, single-dose immunization. Delivery of replicons is achieved with virus-like replicon particles (VRPs), or in nonviral vehicles such as liposomes or lipid nanoparticles. Here, we discuss innovative advances, including multivalent, mucosal, and therapeutic replicon vaccines, and highlight novelties in replicon design. As soon as essential safety evaluations have been resolved, this promising vaccine concept can transform into a widely applied clinical platform technology taking center stage in pandemic preparedness.
Collapse
Affiliation(s)
- Jerome D G Comes
- Wageningen University and Research, Laboratory of Virology, Droevendaalsesteeg 1, 6708PB, Wageningen, The Netherlands
| | - Gorben P Pijlman
- Wageningen University and Research, Laboratory of Virology, Droevendaalsesteeg 1, 6708PB, Wageningen, The Netherlands.
| | - Tessy A H Hick
- Wageningen University and Research, Laboratory of Virology, Droevendaalsesteeg 1, 6708PB, Wageningen, The Netherlands
| |
Collapse
|
10
|
Han L, Song S, Feng H, Ma J, Wei W, Si F. A roadmap for developing Venezuelan equine encephalitis virus (VEEV) vaccines: Lessons from the past, strategies for the future. Int J Biol Macromol 2023:125514. [PMID: 37353130 DOI: 10.1016/j.ijbiomac.2023.125514] [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: 04/11/2023] [Revised: 06/16/2023] [Accepted: 06/20/2023] [Indexed: 06/25/2023]
Abstract
Venezuelan equine encephalitis (VEE) is a zoonotic infectious disease caused by the Venezuelan equine encephalitis virus (VEEV), which can lead to severe central nervous system infections in both humans and animals. At present, the medical community does not possess a viable means of addressing VEE, rendering the prevention of the virus a matter of paramount importance. Regarding the prevention and control of VEEV, the implementation of a vaccination program has been recognized as the most efficient strategy. Nevertheless, there are currently no licensed vaccines or drugs available for human use against VEEV. This imperative has led to a surge of interest in vaccine research, with VEEV being a prime focus for researchers in the field. In this paper, we initially present a comprehensive overview of the current taxonomic classification of VEEV and the cellular infection mechanism of the virus. Subsequently, we provide a detailed introduction of the prominent VEEV vaccine types presently available, including inactivated vaccines, live attenuated vaccines, genetic, and virus-like particle vaccines. Moreover, we emphasize the challenges that current VEEV vaccine development faces and suggest urgent measures that must be taken to overcome these obstacles. Notably, based on our latest research, we propose the feasibility of incorporation codon usage bias strategies to create the novel VEEV vaccine. Finally, we prose several areas that future VEEV vaccine development should focus on. Our objective is to encourage collaboration between the medical and veterinary communities, expedite the translation of existing vaccines from laboratory to clinical applications, while also preparing for future outbreaks of new VEEV variants.
Collapse
Affiliation(s)
- Lulu Han
- Institute of Animal Science and Veterinary Medicine, Shanghai Academy of Agricultural Sciences, Shanghai Key Laboratory of Agricultural Genetics and Breeding, Shanghai Engineering Research Center of Breeding Pig, Shanghai 201106, China; Huaihe Hospital of Henan University, Clinical Medical College of Henan University, Kai Feng 475000, China
| | - Shuai Song
- Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Key Laboratory of Livestock Disease Prevention of Guangdong Province, Scientific Observation and Experiment Station of Veterinary Drugs and Diagnostic Techniques of Guangdong Province, Ministry of Agriculture and Rural Affairs, Guangzhou 510640, PR China
| | - Huilin Feng
- Kaifeng Key Laboratory of Infection and Biological Safety, School of Basic Medical Sciences of Henan University, Kai Feng 475000, China
| | - Jing Ma
- Huaihe Hospital of Henan University, Clinical Medical College of Henan University, Kai Feng 475000, China
| | - Wenqiang Wei
- Kaifeng Key Laboratory of Infection and Biological Safety, School of Basic Medical Sciences of Henan University, Kai Feng 475000, China.
| | - Fusheng Si
- Institute of Animal Science and Veterinary Medicine, Shanghai Academy of Agricultural Sciences, Shanghai Key Laboratory of Agricultural Genetics and Breeding, Shanghai Engineering Research Center of Breeding Pig, Shanghai 201106, China.
| |
Collapse
|
11
|
Bai S, Yang T, Zhu C, Feng M, Zhang L, Zhang Z, Wang X, Yu R, Pan X, Zhao C, Xu J, Zhang X. A single vaccination of nucleoside-modified Rabies mRNA vaccine induces prolonged highly protective immune responses in mice. Front Immunol 2023; 13:1099991. [PMID: 36761167 PMCID: PMC9907168 DOI: 10.3389/fimmu.2022.1099991] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 12/28/2022] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND Rabies is a lethal zoonotic disease that kills approximately 60,000 people each year. Although inactivated rabies vaccines are available, multiple-dose regimensare recommended for pre-exposure prophylaxis or post-exposure prophylaxis,which cuts down the cost- and time-effectiveness, especially in low- and middle incomecountries. METHODS We developed a nucleoside-modified Rabies mRNA-lipid nanoparticle vaccine (RABV-G mRNA-LNP) encoding codon-optimized viral glycoprotein and assessed the immunogenicity and protective efficacy of this vaccine in mice comparing to a commercially available inactivated vaccine. RESULTS We first showed that, when evaluated in mice, a single vaccination of RABV-G mRNA with a moderate or high dose induces more potent humoral and T-cell immune responses than that elicited by three inoculations of the inactivated vaccine. Importantly, mice receiving a single immunization of RABV-G mRNA, even at low doses, showed full protection against the lethal rabies challenge. We further demonstrated that the humoral immune response induced by single RABV-G mRNA vaccination in mice could last for at least 25 weeks, while a two-dose strategy could extend the duration of the highly protective response to one year or even longer. In contrast, the three-dose regimen of inactivated vaccine failed to do so. CONCLUSION Our study confirmed that it is worth developing a single-dose nucleoside-modified Rabies mRNA-LNP vaccine, which could confer much prolonged and more effective protection.
Collapse
Affiliation(s)
- Shimeng Bai
- Shanghai Public Health Clinical Center & Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Tianhan Yang
- Shanghai Public Health Clinical Center & Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Cuisong Zhu
- Shanghai Public Health Clinical Center & Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Meiqi Feng
- Shanghai Public Health Clinical Center & Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Li Zhang
- Shanghai Public Health Clinical Center & Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Ziling Zhang
- Shanghai Public Health Clinical Center & Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Xiang Wang
- Shanghai Public Health Clinical Center & Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Rui Yu
- Shanghai Public Health Clinical Center & Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Xinghao Pan
- Shanghai Public Health Clinical Center & Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Chen Zhao
- Shanghai Public Health Clinical Center & Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Jianqing Xu
- Shanghai Public Health Clinical Center & Institutes of Biomedical Sciences, Fudan University, Shanghai, China
- Clinical Center of Biotherapy, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiaoyan Zhang
- Shanghai Public Health Clinical Center & Institutes of Biomedical Sciences, Fudan University, Shanghai, China
- Clinical Center of Biotherapy, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
12
|
Li N, Chen XL, Li Q, Zhang ZR, Deng CL, Zhang B, Li XD, Ye HQ. A new screening system for entry inhibitors based on cell-to-cell transmitted syncytia formation mediated by self-propagating hybrid VEEV-SARS-CoV-2 replicon. Emerg Microbes Infect 2022; 11:465-476. [PMID: 35034586 PMCID: PMC8820800 DOI: 10.1080/22221751.2022.2030198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The extremely high transmission rate of SARS-CoV-2 and severe cases of COVID-19 pose the two critical challenges in the battle against COVID-19. Increasing evidence has shown that the viral spike (S) protein-driven syncytia may be responsible for these two events. Intensive attention has thus been devoted to seeking S-guided syncytium inhibitors. However, the current screening campaigns mainly rely on either live virus-based or plasmid-based method, which are always greatly limited by the shortage of high-level biosafety BSL-3 facilities or too much labour-intensive work. Here, we constructed a new hybrid VEEV-SARS-CoV-2-S-eGFP reporter vector through replacement of the structural genes of Venezuelan equine encephalitis virus (VEEV) with the S protein of SARS-CoV-2 as the single structural protein. VEEV-SARS-CoV-2-S-eGFP can propagate steadily through cell-to-cell transmission pathway in S- and ACE2-dependent manner, forming GFP positive syncytia. In addition, a significant dose-dependent decay in GFP signals was observed in VEEV-SARS-CoV-2-S-eGFP replicating cells upon treatment with SARS-CoV-2 antiserum or entry inhibitors, providing further evidence that VEEV-SARS-CoV-2-S-eGFP system is highly sensitive to characterize the anti-syncytium-formation activity of antiviral agents. More importantly, the assay is able to be performed in a BSL-2 laboratory without manipulation of live SARS-CoV-2. Taken together, our work establishes a more convenient and efficient VEEV-SARS-CoV-2-S-eGFP replicating cells-based method for rapid screening of inhibitors blocking syncytium formation.
Collapse
Affiliation(s)
- Na Li
- Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiao-Ling Chen
- Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qi Li
- College of Pharmacy and Drug Discovery Center for Infectious Diseases, Nankai University, Tianjin, 300350, China
| | - Zhe-Rui Zhang
- Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Cheng-Lin Deng
- Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Bo Zhang
- Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Xiao-Dan Li
- Hunan Normal University, School of Medicine, Changsha, 410081, China
| | - Han-Qing Ye
- Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China
| |
Collapse
|
13
|
Research Advances on the Interactions between Rabies Virus Structural Proteins and Host Target Cells: Accrued Knowledge from the Application of Reverse Genetics Systems. Viruses 2021; 13:v13112288. [PMID: 34835093 PMCID: PMC8617671 DOI: 10.3390/v13112288] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 11/07/2021] [Accepted: 11/12/2021] [Indexed: 11/17/2022] Open
Abstract
Rabies is a lethal zoonotic disease caused by lyssaviruses, such as rabies virus (RABV), that results in nearly 100% mortality once clinical symptoms appear. There are no curable drugs available yet. RABV contains five structural proteins that play an important role in viral replication, transcription, infection, and immune escape mechanisms. In the past decade, progress has been made in research on the pathogenicity of RABV, which plays an important role in the creation of new recombinant RABV vaccines by reverse genetic manipulation. Here, we review the latest advances on the interaction between RABV proteins in the infected host and the applied development of rabies vaccines by using a fully operational RABV reverse genetics system. This article provides a background for more in-depth research on the pathogenic mechanism of RABV and the development of therapeutic drugs and new biologics.
Collapse
|
14
|
Wang Z, Yuan Y, Chen C, Zhang C, Huang F, Zhou M, Chen H, Fu ZF, Zhao L. Colloidal Manganese Salt Improves the Efficacy of Rabies Vaccines in Mice, Cats, and Dogs. J Virol 2021; 95:e0141421. [PMID: 34495701 PMCID: PMC8577392 DOI: 10.1128/jvi.01414-21] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 08/30/2021] [Indexed: 12/14/2022] Open
Abstract
Rabies, caused by rabies virus (RABV), remains a serious threat to public health in most countries worldwide. At present, the administration of rabies vaccines has been the most effective strategy to control rabies. Herein, we evaluate the effect of colloidal manganese salt (Mn jelly [MnJ]) as an adjuvant of rabies vaccine in mice, cats, and dogs. The results showed that MnJ promoted type I interferon (IFN-I) and cytokine production in vitro and the maturation of dendritic cells (DCs) in vitro and in vivo. Besides, MnJ serving as an adjuvant for rabies vaccines could significantly facilitate the generation of T follicular helper (Tfh) cells, germinal center (GC) B cells, plasma cells (PCs), and RABV-specific antibody-secreting cells (ASCs), consequently improve the immunogenicity of rabies vaccines, and provide better protection against virulent RABV challenge. Similarly, MnJ enhanced the humoral immune response in cats and dogs as well. Collectively, our results suggest that MnJ can facilitate the maturation of DCs during rabies vaccination, which can be a promising adjuvant candidate for rabies vaccines. IMPORTANCE Extending the humoral immune response by using adjuvants is an important strategy for vaccine development. In this study, a novel adjuvant, MnJ, supplemented in rabies vaccines was evaluated in mice, cats, and dogs. Our results in the mouse model revealed that MnJ increased the numbers of mature DCs, Tfh cells, GC B cells, PCs, and RABV-specific ASCs, resulting in enhanced immunogenicity and protection rate of rabies vaccines. We further found that MnJ had the same stimulative effect in cats and dogs. Our study provides the first evidence that MnJ serving as a novel adjuvant of rabies vaccines can boost the immune response in both a mouse and pet model.
Collapse
Affiliation(s)
- Zongmei Wang
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Yueming Yuan
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Chen Chen
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Chengguang Zhang
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Fei Huang
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Ming Zhou
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Huanchun Chen
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Zhen F. Fu
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Ling Zhao
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| |
Collapse
|
15
|
Virus-like vesicles based on SFV-containing rabies virus glycoprotein make a safe and efficacious rabies vaccine candidate in a mouse model. J Virol 2021; 95:e0079021. [PMID: 34346765 DOI: 10.1128/jvi.00790-21] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Rabies is a fatal zoonosis causing encephalitis in mammals, and vaccination is the most effective method to control and eliminate rabies. Virus-like vesicles (VLVs), which are characterized as infectious, self-propagating membrane-enveloped particles composed of only Semliki Forest virus (SFV) replicase and vesicular stomatitis virus glycoprotein (VSV-G), have been proven safe and efficient as vaccine candidates. However, previous studies showed that VLVs containing rabies virus glycoprotein (RABV-G) grew at relatively low titers in cells, impeding their potential use as a rabies vaccine. In this study, we constructed novel VLVs by transfection of a mutant SFV RNA replicon encoding RABV-G. We found these VLVs could self-propagate efficiently in cell culture and could evolve to high titers (approximately 108 FFU/ml) by extensive passaging 25 times in BHK-21 cells. Furthermore, we found that the evolved amino acid change in SFV nsP1 at positions 470 and 482 was critical for this high-titer phenotype. Remarkably, VLVs could induce robust type I IFN expression in BV2 cells and were highly sensitive to IFN-α. We found that direct inoculation of VLVs into the mouse brain caused lesser body weight loss, mortality and neuroinflammation compared with RABV vaccine strain. Finally, it could induce increased generation of germinal centre (GC) B cells, plasma cells (PCs) and virus-neutralizing antibodies (VNAs), as well as provide protection against virulent RABV challenge in immunized mice. This study demonstrated that VLVs containing RABV-G could proliferate in cells and were highly evolvable, revealing the feasibility of developing an economic, safe and efficacious rabies vaccine. IMPORTANCE VLVs have been shown to represent a more versatile and superior vaccine platform. In previous studies, VLVs containing the Semliki Forest Virus replicase (SFV nsP1-4) and rabies virus glycoprotein (RABV-G) grew to relatively low titers in cells. In our study, we not only succeeded in generating VLVs that proliferate in cells and stably express RABV-G, the VLVs that evolved grew to higher titers reaching 108 FFU/ml. We also found that nucleic acid changes at positions 470 and 482 in nsP1 were vital for this high-titer phenotype. Moreover, the VLVs that evolved in our studies were highly attenuated in mice, induced potent immunity and protected mice from lethal RABV infection. Collectively, our study showed that high titers of VLVs containing RABV-G were achieved demonstrating that these VLVs could be an economical, safe, and efficacious rabies vaccine candidate.
Collapse
|
16
|
Krasnopolsky Y. BIOTECHNOLOGICAL RESEARCH IN THE CREATION AND PRODUCTION OF ANTIRABIC VACCINES. BIOTECHNOLOGIA ACTA 2021. [DOI: 10.15407/biotech14.04.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Rabies is a neurological disease of a viral nature, leading to death. Rabies virus is an RNA virus that invades the central nervous system, leading to neuronal dysfunction. Timely vaccination can prevent the diseases development. Aim. The article is devoted to immunobiotechnological research aimed at creating antirabic vaccines. Results. The history of the antirabic vaccines creation from the first inactivated vaccines obtained from nervous tissue to the cultivation of the virus on animal cell cultures is considered. The article presents commercially available anti-rabies vaccines: their composition, the used rabies virus strains, cell cultures, the methods of inactivation and purification. The technology of producing an anti-rabies vaccine based on a Pitman Moore virus strain and a chicken fibroblast cell culture is presented. The advantages of different vaccine types are considered: live attenuated, peptide, liposomal, RNA vaccines, vaccines based on viral vectors, transgenic plants and reverse genetics methods. Conclusions. The development of biotechnology, immunology and virology makes it possible to improve constantly vaccine preparations, including those against rabies, increasing their effectiveness and safety.
Collapse
|