1
|
Bi J, Wang H, Han Q, Pei H, Wang H, Jin H, Jin S, Chi H, Yang S, Zhao Y, Yan F, Ge L, Xia X. A rabies virus-vectored vaccine expressing two copies of the Marburg virus glycoprotein gene induced neutralizing antibodies against Marburg virus in humanized mice. Emerg Microbes Infect 2023; 12:2149351. [PMID: 36453198 PMCID: PMC9809360 DOI: 10.1080/22221751.2022.2149351] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
Abstract
Marburg virus disease (MVD) is a lethal viral haemorrhagic fever caused by Marburg virus (MARV) with a case fatality rate as high as 88%. There is currently no vaccine or antiviral therapy approved for MVD. Due to high variation among MARV isolates, vaccines developed against one strain fail to protect against other strains. Here we report that three recombinant rabies virus (RABV) vector vaccines encoding two copies of GPs covering both MARV lineages induced pseudovirus neutralizing antibodies in BALB/c mice. Furthermore, high-affinity human neutralizing antibodies were isolated from a humanized mouse model. The three vaccines produced a Th1-biased serological response similar to that of human patients. Adequate sequential immunization enhanced the production of neutralizing antibodies. Virtual docking suggested that neutralizing antibodies induced by the Angola strain seemed to be able to hydrogen bond to the receptor-binding site (RBS) in the GP of the Ravn strain through hypervariable regions 2 (CDR2) and CDR3 of the VH region. These findings demonstrate that three inactivated vaccines are promising candidates against different strains of MARV, and a novel fully humanized neutralizing antibody against MARV was isolated.
Collapse
Affiliation(s)
- Jinhao Bi
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, People’s Republic of China,Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, People’s Republic of China
| | - Haojie Wang
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, People’s Republic of China
| | - Qiuxue Han
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, People’s Republic of China,Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College (PUMC), Beijing, People’s Republic of China
| | - Hongyan Pei
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, People’s Republic of China,College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, People’s Republic of China
| | - Hualei Wang
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, People’s Republic of China
| | - Hongli Jin
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, People’s Republic of China
| | - Song Jin
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, People’s Republic of China,Ruminant Disease Research Center, College of Life Sciences, Shandong Normal University, Jinan, People’s Republic of China
| | - Hang Chi
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, People’s Republic of China
| | - Songtao Yang
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, People’s Republic of China
| | - Yongkun Zhao
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, People’s Republic of China
| | - Feihu Yan
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, People’s Republic of China, Feihu Yan ; Liangpeng Ge ; Xianzhu Xia
| | - Liangpeng Ge
- Chongqing Academy of Animal Sciences, Chongqing, People’s Republic of China, Feihu Yan ; Liangpeng Ge ; Xianzhu Xia
| | - Xianzhu Xia
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, People’s Republic of China,Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, People’s Republic of China,Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College (PUMC), Beijing, People’s Republic of China, Feihu Yan ; Liangpeng Ge ; Xianzhu Xia
| |
Collapse
|
2
|
Wang S, Liang B, Wang W, Li L, Feng N, Zhao Y, Wang T, Yan F, Yang S, Xia X. Viral vectored vaccines: design, development, preventive and therapeutic applications in human diseases. Signal Transduct Target Ther 2023; 8:149. [PMID: 37029123 PMCID: PMC10081433 DOI: 10.1038/s41392-023-01408-5] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 03/06/2023] [Accepted: 03/14/2023] [Indexed: 04/09/2023] Open
Abstract
Human diseases, particularly infectious diseases and cancers, pose unprecedented challenges to public health security and the global economy. The development and distribution of novel prophylactic and therapeutic vaccines are the prioritized countermeasures of human disease. Among all vaccine platforms, viral vector vaccines offer distinguished advantages and represent prominent choices for pathogens that have hampered control efforts based on conventional vaccine approaches. Currently, viral vector vaccines remain one of the best strategies for induction of robust humoral and cellular immunity against human diseases. Numerous viruses of different families and origins, including vesicular stomatitis virus, rabies virus, parainfluenza virus, measles virus, Newcastle disease virus, influenza virus, adenovirus and poxvirus, are deemed to be prominent viral vectors that differ in structural characteristics, design strategy, antigen presentation capability, immunogenicity and protective efficacy. This review summarized the overall profile of the design strategies, progress in advance and steps taken to address barriers to the deployment of these viral vector vaccines, simultaneously highlighting their potential for mucosal delivery, therapeutic application in cancer as well as other key aspects concerning the rational application of these viral vector vaccines. Appropriate and accurate technological advances in viral vector vaccines would consolidate their position as a leading approach to accelerate breakthroughs in novel vaccines and facilitate a rapid response to public health emergencies.
Collapse
Affiliation(s)
- Shen Wang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Bo Liang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Weiqi Wang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
- College of Veterinary Medicine, Jilin University, Changchun, China
| | - Ling Li
- China National Research Center for Exotic Animal Diseases, China Animal Health and Epidemiology Center, Qingdao, China
| | - Na Feng
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Yongkun Zhao
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Tiecheng Wang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Feihu Yan
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China.
| | - Songtao Yang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China.
| | - Xianzhu Xia
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China.
| |
Collapse
|
3
|
Viral Vectors in Gene Therapy: Where Do We Stand in 2023? Viruses 2023; 15:v15030698. [PMID: 36992407 PMCID: PMC10059137 DOI: 10.3390/v15030698] [Citation(s) in RCA: 58] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 02/23/2023] [Accepted: 03/02/2023] [Indexed: 03/11/2023] Open
Abstract
Viral vectors have been used for a broad spectrum of gene therapy for both acute and chronic diseases. In the context of cancer gene therapy, viral vectors expressing anti-tumor, toxic, suicide and immunostimulatory genes, such as cytokines and chemokines, have been applied. Oncolytic viruses, which specifically replicate in and kill tumor cells, have provided tumor eradication, and even cure of cancers in animal models. In a broader meaning, vaccine development against infectious diseases and various cancers has been considered as a type of gene therapy. Especially in the case of COVID-19 vaccines, adenovirus-based vaccines such as ChAdOx1 nCoV-19 and Ad26.COV2.S have demonstrated excellent safety and vaccine efficacy in clinical trials, leading to Emergency Use Authorization in many countries. Viral vectors have shown great promise in the treatment of chronic diseases such as severe combined immunodeficiency (SCID), muscular dystrophy, hemophilia, β-thalassemia, and sickle cell disease (SCD). Proof-of-concept has been established in preclinical studies in various animal models. Clinical gene therapy trials have confirmed good safety, tolerability, and therapeutic efficacy. Viral-based drugs have been approved for cancer, hematological, metabolic, neurological, and ophthalmological diseases as well as for vaccines. For example, the adenovirus-based drug Gendicine® for non-small-cell lung cancer, the reovirus-based drug Reolysin® for ovarian cancer, the oncolytic HSV T-VEC for melanoma, lentivirus-based treatment of ADA-SCID disease, and the rhabdovirus-based vaccine Ervebo against Ebola virus disease have been approved for human use.
Collapse
|
4
|
Lundstrom K. Gene Therapy Cargoes Based on Viral Vector Delivery. Curr Gene Ther 2023; 23:111-134. [PMID: 36154608 DOI: 10.2174/1566523222666220921112753] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Revised: 07/13/2022] [Accepted: 08/05/2022] [Indexed: 11/22/2022]
Abstract
Viral vectors have been proven useful in a broad spectrum of gene therapy applications due to their possibility to accommodate foreign genetic material for both local and systemic delivery. The wide range of viral vectors has enabled gene therapy applications for both acute and chronic diseases. Cancer gene therapy has been addressed by the delivery of viral vectors expressing anti-tumor, toxic, and suicide genes for the destruction of tumors. Delivery of immunostimulatory genes such as cytokines and chemokines has also been applied for cancer therapy. Moreover, oncolytic viruses specifically replicating in and killing tumor cells have been used as such for tumor eradication or in combination with tumor killing or immunostimulatory genes. In a broad meaning, vaccines against infectious diseases and various cancers can be considered gene therapy, which has been highly successful, not the least for the development of effective COVID-19 vaccines. Viral vector-based gene therapy has also demonstrated encouraging and promising results for chronic diseases such as severe combined immunodeficiency (SCID), muscular dystrophy, and hemophilia. Preclinical gene therapy studies in animal models have demonstrated proof-of-concept for a wide range of disease indications. Clinical evaluation of drugs and vaccines in humans has showed high safety levels, good tolerance, and therapeutic efficacy. Several gene therapy drugs such as the adenovirus-based drug Gendicine® for non-small-cell lung cancer, the reovirus-based drug Reolysin® for ovarian cancer, lentivirus-based treatment of SCID-X1 disease, and the rhabdovirus-based vaccine Ervebo against Ebola virus disease, and adenovirus-based vaccines against COVID-19 have been developed.
Collapse
|
5
|
Zhang Y, Zhang M, Liao X, Yu Y, Liu Q, Luo Y, Luo J, Guo X. Interleukin-25 enhances humoral immune responses caused by the rabies virus. Virulence 2022; 13:1446-1454. [PMID: 35999776 PMCID: PMC9423819 DOI: 10.1080/21505594.2022.2116146] [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: 10/26/2022] Open
Abstract
Rabies is an important zoonotic disease caused by the rabies virus (RABV). Currently, no effective treatment is available for this condition. The prevention and control of rabies mainly depend on effective vaccination. Therefore, it is crucial to enhance the immune responses induced by the rabies vaccine. Virus neutralizing antibodies (VNA) induced by rabies vaccines are important for the clearance of RABV. Interleukin-25 (IL-25) has been demonstrated to activate T helper type 2 cells that contribute to humoral immune responses. The IL-25 gene was inserted into the genome of RABV, and the immunogenicity of recombinant RABV with IL-25 gene was investigated to develop more efficient rabies vaccines. Here, we found that the expression of IL-25 did not affect RABV production in vitro and pathogenicity in vivo. However, recombinant RABV expression of IL-25 induced a better VNA level than the parental virus in mice. In addition, expression of IL-25 enhanced the IgG1 level induced by RABV. Furthermore, mice immunized with recombinant RABV showed a higher survival rate and milder clinical signs than those immunized with the parent strain after challenge with CVS-11. Thus, these results showed that IL-25 could enhance the humoral immune responses induced by RABV, suggesting that IL-25 can be used as a viral vaccine adjuvant.
Collapse
Affiliation(s)
- Yue Zhang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Mengwei Zhang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Xilan Liao
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Yunsong Yu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Qing Liu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Yongwen Luo
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Jun Luo
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Xiaofeng Guo
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| |
Collapse
|
6
|
Rabies Vaccine: Recent Update and Comprehensive Review of in vitro and in vivo Studies. Process Biochem 2022. [DOI: 10.1016/j.procbio.2022.11.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
7
|
Chailangkarn T, Tanwattana N, Jaemthaworn T, Sriswasdi S, Wanasen N, Tangphatsornruang S, Leetanasaksakul K, Jantraphakorn Y, Nawae W, Chankeeree P, Lekcharoensuk P, Lumlertdacha B, Kaewborisuth C. Establishment of Human-Induced Pluripotent Stem Cell-Derived Neurons-A Promising In Vitro Model for a Molecular Study of Rabies Virus and Host Interaction. Int J Mol Sci 2021; 22:ijms222111986. [PMID: 34769416 PMCID: PMC8584829 DOI: 10.3390/ijms222111986] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 10/24/2021] [Accepted: 11/02/2021] [Indexed: 12/15/2022] Open
Abstract
Rabies is a deadly viral disease caused by the rabies virus (RABV), transmitted through a bite of an infected host, resulting in irreversible neurological symptoms and a 100% fatality rate in humans. Despite many aspects describing rabies neuropathogenesis, numerous hypotheses remain unanswered and concealed. Observations obtained from infected primary neurons or mouse brain samples are more relevant to human clinical rabies than permissive cell lines; however, limitations regarding the ethical issue and sample accessibility become a hurdle for discovering new insights into virus-host interplays. To better understand RABV pathogenesis in humans, we generated human-induced pluripotent stem cell (hiPSC)-derived neurons to offer the opportunity for an inimitable study of RABV infection at a molecular level in a pathologically relevant cell type. This study describes the characteristics and detailed proteomic changes of hiPSC-derived neurons in response to RABV infection using LC-MS/MS quantitative analysis. Gene ontology (GO) enrichment of differentially expressed proteins (DEPs) reveals temporal changes of proteins related to metabolic process, immune response, neurotransmitter transport/synaptic vesicle cycle, cytoskeleton organization, and cell stress response, demonstrating fundamental underlying mechanisms of neuropathogenesis in a time-course dependence. Lastly, we highlighted plausible functions of heat shock cognate protein 70 (HSC70 or HSPA8) that might play a pivotal role in regulating RABV replication and pathogenesis. Our findings acquired from this hiPSC-derived neuron platform help to define novel cellular mechanisms during RABV infection, which could be applicable to further studies to widen views of RABV-host interaction.
Collapse
Affiliation(s)
- Thanathom Chailangkarn
- Virology and Cell Technology Research Team, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), Pathum Thani 12120, Thailand; (N.W.); (Y.J.)
- Correspondence: (T.C.); (C.K.)
| | - Nathiphat Tanwattana
- Interdisciplinary Program in Genetic Engineering and Bioinformatics, Graduate School, Kasetsart University, Bangkok 10900, Thailand;
| | - Thanakorn Jaemthaworn
- Computational Molecular Biology Group, Chulalongkorn University, Pathum Wan, Bangkok 10330, Thailand; (T.J.); (S.S.)
| | - Sira Sriswasdi
- Computational Molecular Biology Group, Chulalongkorn University, Pathum Wan, Bangkok 10330, Thailand; (T.J.); (S.S.)
- Research Affairs, Faculty of Medicine, Chulalongkorn University, Pathum Wan, Bangkok 10330, Thailand
| | - Nanchaya Wanasen
- Virology and Cell Technology Research Team, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), Pathum Thani 12120, Thailand; (N.W.); (Y.J.)
| | - Sithichoke Tangphatsornruang
- National Omics Center, National Science and Technology Development Agency (NSTDA), Pathum Thani 12120, Thailand; (S.T.); (W.N.)
| | - Kantinan Leetanasaksakul
- Functional Proteomics Technology, Functional Ingredients and Food Innovation Research Group, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), Pathum Thani 12120, Thailand;
| | - Yuparat Jantraphakorn
- Virology and Cell Technology Research Team, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), Pathum Thani 12120, Thailand; (N.W.); (Y.J.)
| | - Wanapinun Nawae
- National Omics Center, National Science and Technology Development Agency (NSTDA), Pathum Thani 12120, Thailand; (S.T.); (W.N.)
| | - Penpicha Chankeeree
- Department of Microbiology and Immunology, Faculty of Veterinary Medicine, Kasetsart University, Bangkok 10900, Thailand; (P.C.); (P.L.)
| | - Porntippa Lekcharoensuk
- Department of Microbiology and Immunology, Faculty of Veterinary Medicine, Kasetsart University, Bangkok 10900, Thailand; (P.C.); (P.L.)
- Center for Advance Studies in Agriculture and Food, KU Institute Studies, Kasetsart University, Bangkok 10900, Thailand
| | - Boonlert Lumlertdacha
- Queen Saovabha Memorial Institute, Thai Red Cross Society, WHO Collaborating Center for Research and Training Prophylaxis on Rabies, 1871 Rama 4 Road, Pathumwan, Bangkok 10330, Thailand;
| | - Challika Kaewborisuth
- Virology and Cell Technology Research Team, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), Pathum Thani 12120, Thailand; (N.W.); (Y.J.)
- Correspondence: (T.C.); (C.K.)
| |
Collapse
|
8
|
Jakhmola S, Jha HC. Glial cell response to Epstein-Barr Virus infection: A plausible contribution to virus-associated inflammatory reactions in the brain. Virology 2021; 559:182-195. [PMID: 33964684 DOI: 10.1016/j.virol.2021.04.005] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 04/09/2021] [Accepted: 04/16/2021] [Indexed: 02/07/2023]
Abstract
Epstein-Barr Virus (EBV) is clinically related to various neurological ailments. The manipulation of neural homeostasis through altered glial cells functions is enigmatic. We investigated EBV mediated nuances in glial cells through direct infection (group-1) or by supplementing them with EBV-infected lymphocytes (PBMCs) supernatant (group-3). Also, the cells were co-cultured with infected PBMCs (group-2). Upon confirmation of infection in U-87 MG through qRT-PCR, the gene expression of crucial molecules was analysed. We reported enhanced expression of IL6 in group-1 and 3 unlike group-2. PBMCs migrated and invaded the matrigel significantly when exposed to group-1 and 3 conditions. Thus, EBV may aid neuroinflammatory reactions through PBMCs infiltration. Also, the exposure of neurons to conditioned supernatant from group-2 caused reduced neuronal healing. Additionally, group-1 milieu contained chemical modulators that induced glial cells death and reduced NF-κB. Conclusively, the three modes of EBV infection can influence glial cells' functions to maneuver the microenvironment distinctly.
Collapse
Affiliation(s)
- Shweta Jakhmola
- Infection Bio-engineering Group, Discipline of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, India
| | - Hem Chandra Jha
- Infection Bio-engineering Group, Discipline of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, India.
| |
Collapse
|
9
|
Luo J, Zhang Y, Wang Y, Liu Q, Li J, He H, Luo Y, Huang S, Guo X. Artesunate and Dihydroartemisinin Inhibit Rabies Virus Replication. Virol Sin 2021; 36:721-729. [PMID: 33661488 PMCID: PMC7930525 DOI: 10.1007/s12250-021-00349-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 11/16/2020] [Indexed: 12/25/2022] Open
Abstract
Rabies is caused by infection of rabies virus (RABV) and remains a serious threat to the global public health. Except for the requirement for cold chain and high cost of human rabies immune globulin, no small molecule drugs are currently available for clinical treatment of rabies. So, it is of great importance to identify novel compounds that can effectively inhibit RABV infection. Artesunate (ART) and dihydroartemisinin (DHA), two derivatives of artemisinin, are widely used for treatment of malaria in adults and children, showing high safety. In this study, we found that both ART and DHA were able to inhibit RABV replication in host cells at a low concentration (0.1 μmol/L). The antiviral effects of ART and DHA were independent of viral strains and cell lines. Pre-treatment with ART or DHA for 2 h in vitro did not affect the viral replication in host cells, implying that ART and DHA neither reduced the viability of RABV directly nor inhibited the binding and entrance of the virus to host cells. Further studies revealed that ART and DHA inhibited RABV genomic RNA synthesis and viral gene transcription. Treatment with ART or DHA (5 mg/kg) by intramuscular injection improved, to some extent, the survival rate of RABV-challenged mice. Combination treatment with derivatives of artemisinin and mannitol significantly improved the survival rate of RABV-challenged mice. The results suggest that ART and DHA have a great potential to be explored as new anti-rabies agents for treatment of rabies.
Collapse
Affiliation(s)
- Jun Luo
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Yue Zhang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Yang Wang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Qing Liu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Jiesen Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Hongling He
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Yongwen Luo
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Shile Huang
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, LA, 71130-3932, USA. .,Feist-Weiller Cancer Center, Louisiana State University Health Sciences Center, Shreveport, LA, 71130-3932, USA.
| | - Xiaofeng Guo
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China.
| |
Collapse
|
10
|
Yang DK, Kim HH, Park YR, Yoo JY, Park Y, Park J, Hyun BH. Generation of a recombinant rabies virus expressing green fluorescent protein for a virus neutralization antibody assay. J Vet Sci 2021; 22:e56. [PMID: 34313041 PMCID: PMC8318786 DOI: 10.4142/jvs.2021.22.e56] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 04/27/2021] [Accepted: 06/29/2021] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND Fluorescent antibody virus neutralization (FAVN) test is a standard assay for quantifying rabies virus-neutralizing antibody (VNA) in serum. However, a safer rabies virus (RABV) should be used in the FAVN assay. There is a need for a new method that is economical and time-saving by eliminating the immunostaining step. OBJECTIVES We aimed to improve the traditional FAVN method by rescuing and characterizing a new recombinant RABV expressing green fluorescent protein (GFP). METHODS A new recombinant RABV expressing GFP designated as ERAGS-GFP was rescued using a reverse genetic system. Immuno-fluorescence assay, peroxidase-linked assay, electron microscopy and reverse transcription polymerase chain reaction were performed to confirm the recombinant ERAGS-GFP virus as a RABV expressing the GFP gene. The safety of ERAGS-GFP was evaluated in 4-week-old mice. The rabies VNA titers were measured and compared with conventional FAVN and FAVN-GFP tests using VERO cells. RESULTS The virus propagated in VERO cells was confirmed as RABV expressing GFP. The ERAGS-GFP showed the highest titer (108.0 TCID50/mL) in VERO cells at 5 days post-inoculation, and GFP expression persisted until passage 30. The body weight of 4-week-old mice inoculated intracranially with ERAGS-GFP continued to increase and the survival rate was 100%. In 62 dog sera, the FAVN-GFP result was significantly correlated with that of conventional FAVN (r = 0.95). CONCLUSIONS We constructed ERAGS-GFP, which could replace the challenge virus standard-11 strain used in FAVN test.
Collapse
Affiliation(s)
- Dong Kun Yang
- Viral Disease Division, Animal and Plant Quarantine Agency, Ministry of Agriculture, Food and Rural Affairs, Gimcheon 39660, Korea.
| | - Ha Hyun Kim
- Viral Disease Division, Animal and Plant Quarantine Agency, Ministry of Agriculture, Food and Rural Affairs, Gimcheon 39660, Korea
| | - Yu Ri Park
- Viral Disease Division, Animal and Plant Quarantine Agency, Ministry of Agriculture, Food and Rural Affairs, Gimcheon 39660, Korea
| | - Jae Young Yoo
- Viral Disease Division, Animal and Plant Quarantine Agency, Ministry of Agriculture, Food and Rural Affairs, Gimcheon 39660, Korea
| | - Yeseul Park
- Viral Disease Division, Animal and Plant Quarantine Agency, Ministry of Agriculture, Food and Rural Affairs, Gimcheon 39660, Korea
| | - Jungwon Park
- Viral Disease Division, Animal and Plant Quarantine Agency, Ministry of Agriculture, Food and Rural Affairs, Gimcheon 39660, Korea
| | - Bang Hun Hyun
- Viral Disease Division, Animal and Plant Quarantine Agency, Ministry of Agriculture, Food and Rural Affairs, Gimcheon 39660, Korea
| |
Collapse
|
11
|
Luo J, Zhang Y, Wang Y, Liu Q, Chen L, Zhang B, Luo Y, Huang S, Guo X. Rhabdovirus Infection Is Dependent on Serine/Threonine Kinase AP2-Associated Kinase 1. Life (Basel) 2020; 10:E170. [PMID: 32872567 PMCID: PMC7554979 DOI: 10.3390/life10090170] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 08/24/2020] [Accepted: 08/28/2020] [Indexed: 01/01/2023] Open
Abstract
Rabies virus (RABV) causes a fatal neurological disease in both humans and animals. Understanding the mechanism of RABV infection is vital for prevention and therapy of virulent rabies infection. Our previous proteomics analysis based on isobaric tags for relative and absolute quantitation to identify factors revealed that RABV infection enhanced AP-2-associated protein kinase 1 (AAK1) in N2a cells. In this study, to further confirm the role of AAK1, we showed that RABV infection increased the transcription and expression of AAK1 in N2a cells. AAK1 knockdown significantly decreased RABV infection in both N2a and BHK-21 cells. AAK1 knockout inhibited RABV infection in N2a cells. Furthermore, inhibition of AAK1 kinase activity using sunitinib decreased RABV infection. However, AAK1 overexpression did not change RABV infection in vitro. Therapeutic administration of sunitinib did not significantly improve the survival rate of mice following lethal RABV challenge. In addition, AAK1 knockdown decreased infection in N2a cells by vesicular stomatitis virus, which is another rhabdovirus. These results indicate that rhabdovirus infection is dependent on AAK1 and inhibition of AAK1 is a potential strategy for the prevention and therapy of rabies.
Collapse
Affiliation(s)
- Jun Luo
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; (J.L.); (Y.Z.); (Y.W.); (Q.L.); (L.C.); (B.Z.); (Y.L.)
| | - Yue Zhang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; (J.L.); (Y.Z.); (Y.W.); (Q.L.); (L.C.); (B.Z.); (Y.L.)
| | - Yang Wang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; (J.L.); (Y.Z.); (Y.W.); (Q.L.); (L.C.); (B.Z.); (Y.L.)
| | - Qing Liu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; (J.L.); (Y.Z.); (Y.W.); (Q.L.); (L.C.); (B.Z.); (Y.L.)
| | - Luman Chen
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; (J.L.); (Y.Z.); (Y.W.); (Q.L.); (L.C.); (B.Z.); (Y.L.)
| | - Boyue Zhang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; (J.L.); (Y.Z.); (Y.W.); (Q.L.); (L.C.); (B.Z.); (Y.L.)
| | - Yongwen Luo
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; (J.L.); (Y.Z.); (Y.W.); (Q.L.); (L.C.); (B.Z.); (Y.L.)
| | - Shile Huang
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, LA 71130-3932, USA;
- Feist-Weiller Cancer Center, Louisiana State University Health Sciences Center, Shreveport, LA 71130-3932, USA
| | - Xiaofeng Guo
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; (J.L.); (Y.Z.); (Y.W.); (Q.L.); (L.C.); (B.Z.); (Y.L.)
| |
Collapse
|
12
|
Luo J, Zhang B, Lyu Z, Wu Y, Zhang Y, Guo X. Single amino acid change at position 255 in rabies virus glycoprotein decreases viral pathogenicity. FASEB J 2020; 34:9650-9663. [PMID: 32469133 DOI: 10.1096/fj.201902577r] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 02/27/2020] [Accepted: 05/11/2020] [Indexed: 12/22/2022]
Abstract
Previous studies have indicated that the amino acid at position 333 in the glycoprotein (G) is closely related to rabies virus (RABV) pathogenicity. However, whether there are other amino acid residues in G that relate to pathogenicity remain unclear. The aim of this study is to find new amino acid residues in G that could strongly reduce RABV pathogenicity. The present study found that the pathogenicity of a virulent strain was strongly attenuated when the amino acid glycine (Gly) replaced the aspartic acid (Asp) at position 255 in G (D255G) as intracranial (i.c.) infection with this D255G mutant virus did not cause death in adult mice. The indexes of neurotropism of the D255G mutant strain and the parent GD-SH-01 are 0.72 and 10.0, respectively, which indicate that the D255G mutation decreased the neurotropism of RABV. In addition, the D255G mutation significantly decreased RABV replication in the mouse brain. Furthermore, the D255G mutation enhanced the immune response in mice, which contributed to the clearance of RABV after infection. The Asp255 → Gly255 mutation was genetically stable in vitro and in vivo. In this study, we describe a new referenced amino acid site in G that relates to the pathogenicity of RABV.
Collapse
Affiliation(s)
- Jun Luo
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Boyue Zhang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Ziyu Lyu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Yuting Wu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Yue Zhang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Xiaofeng Guo
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| |
Collapse
|
13
|
Luo J, Zhang Y, Zhang Q, Wu Y, Zhang B, Mo M, Tian Q, Zhao J, Mei M, Guo X. The Deoptimization of Rabies Virus Matrix Protein Impacts Viral Transcription and Replication. Viruses 2019; 12:v12010004. [PMID: 31861477 PMCID: PMC7019236 DOI: 10.3390/v12010004] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 12/06/2019] [Accepted: 12/16/2019] [Indexed: 12/17/2022] Open
Abstract
Rabies virus (RABV) matrix (M) protein plays several important roles during RABV infection. Although previous studies have assessed the functions of M through gene rearrangements, this interferes with the position of other viral proteins. In this study, we attenuated M expression through deoptimizing its codon usage based on codon pair bias in RABV. This strategy more objectively clarifies the role of M during virus infection. Codon-deoptimized M inhibited RABV replication during the early stages of infection, but enhanced viral titers at later stages. Codon-deoptimized M also inhibited genome synthesis at early stage of infection and increased the RABV transcription rates. Attenuated M through codon deoptimization enhanced RABV glycoprotein expression following RABV infection in neuronal cells, but had no influence on the cell-to-cell spread of RABV. In addition, codon-deoptimized M virus induced higher levels of apoptosis compared to the parental RABV. These results indicate that codon-deoptimized M increases glycoprotein expression, providing a foundation for further investigation of the role of M during RABV infection.
Collapse
|
14
|
Mei M, Long T, Zhang Q, Zhao J, Tian Q, Peng J, Luo J, Jiang H, Lin Y, Lin Z, Guo X. Phenotypic Consequence of Rearranging the N Gene of RABV HEP-Flury. Viruses 2019; 11:v11050402. [PMID: 31035728 PMCID: PMC6563252 DOI: 10.3390/v11050402] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 04/20/2019] [Accepted: 04/25/2019] [Indexed: 02/08/2023] Open
Abstract
Nucleoprotein (N) is a key element in rabies virus (RABV) replication. To further investigate the effect of N on RABV, we manipulated an infectious cDNA clone of the RABV HEP-Flury to rearrange the N gene from its wild-type position of 1 (N-P-M-G-L) to 2 (P-N-M-G-L), 3 (P-M-N-G-L), or 4 (P-M-G-N-L), using an approach that left the viral nucleotide sequence unaltered. Subsequently, viable viruses were recovered from each of the rearranged cDNA and examined for their gene expression levels, growth kinetics in cell culture, pathogenicity in suckling mice and protection in mice. The results showed that gene rearrangement decreased N mRNA transcription and vRNA replication. As a result, all viruses with rearranged genomes showed worse replication than that of rHEP-Flury in NA cells at a MOI of 0.01, but equivalent or slightly better replication levels at a MOI of 3. Consequently, the lethality in suckling mice infected with N4 was clearly attenuated compared with rHEP-Flury. However, the protection to mice was not enhanced. This study not only gives us insight into the understanding of the phenotype of RABV N gene rearrangement, but also helps with rabies vaccine candidate construction.
Collapse
Affiliation(s)
- Mingzhu Mei
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China.
- Guangdong Inspection and Quarantine Technology Center, Guangzhou 510623, China.
| | - Teng Long
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China.
| | - Qiong Zhang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China.
| | - Jing Zhao
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China.
| | - Qin Tian
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China.
| | - Jiaojiao Peng
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China.
| | - Jun Luo
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China.
| | - He Jiang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China.
| | - Yingyi Lin
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China.
| | - Zhixiong Lin
- Guangdong Inspection and Quarantine Technology Center, Guangzhou 510623, China.
| | - Xiaofeng Guo
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China.
| |
Collapse
|
15
|
Qin S, Volokhov D, Rodionova E, Wirblich C, Schnell MJ, Chizhikov V, Dabrazhynetskaya A. A new recombinant rabies virus expressing a green fluorescent protein: A novel and fast approach to quantify virus neutralizing antibodies. Biologicals 2019; 59:56-61. [PMID: 30898479 DOI: 10.1016/j.biologicals.2019.03.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 02/27/2019] [Accepted: 03/02/2019] [Indexed: 12/25/2022] Open
Abstract
The Rapid Fluorescent Focus Inhibition Test (RFFIT) is a standard assay used to detect and assess the titers of rabies virus neutralizing antibodies (RVNA) in blood sera. To simplify the multistep RFFIT procedure by eliminating the immunostaining step, we generated a new recombinant RV expressing a green fluorescent protein (rRV-GFP) and assess its suitability for quantifying RVNA. We rescued the rRV-GFP virus from plasmid DNA carrying a full-length genome of the CVS-N2c strain of RV in which the eGFP gene was inserted between the glycoprotein and RNA-polymerase genes. The recombinant virus was genetically stable and grew efficiently in appropriate cells expressing sufficient GFP fluorescence to detect directly 20 h post infection (hpi). We evaluated the feasibility of using rRV-GFP in RFFIT by comparing RVNA titers in 27 serum samples measured by conventional RFFIT and RFFIT-GFP. A linear regression analysis of the data demonstrated a good agreement between these two methods (r = 0.9776) including results with samples having RVNA titers close to the minimally acceptable vaccine potency threshold (0.5 IU/ml). Study results showed that the rRV-GFP virus could replace the CVS-11 challenge virus currently used in the conventional RFFIT and enabling more rapid, simpler, and less expensive detection and quantitation of RVNA.
Collapse
Affiliation(s)
- Shuyun Qin
- Laboratory of Method Development, Division of Viral Products, Center for Biologics Evaluation, Food and Drug Administration, Silver Spring, MD, USA
| | - Dmitriy Volokhov
- Laboratory of Method Development, Division of Viral Products, Center for Biologics Evaluation, Food and Drug Administration, Silver Spring, MD, USA
| | - Elvira Rodionova
- Laboratory of Method Development, Division of Viral Products, Center for Biologics Evaluation, Food and Drug Administration, Silver Spring, MD, USA
| | - Christoph Wirblich
- Department of Microbiology and Immunology, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA, USA
| | - Matthias J Schnell
- Department of Microbiology and Immunology, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA, USA; Jefferson Vaccine Center at Thomas Jefferson University, Philadelphia, PA, USA
| | - Vladimir Chizhikov
- Laboratory of Method Development, Division of Viral Products, Center for Biologics Evaluation, Food and Drug Administration, Silver Spring, MD, USA
| | - Alena Dabrazhynetskaya
- Laboratory of Method Development, Division of Viral Products, Center for Biologics Evaluation, Food and Drug Administration, Silver Spring, MD, USA.
| |
Collapse
|
16
|
Recombinant rabies virus expressing interleukin-6 enhances the immune response in mouse brain. Arch Virol 2018; 163:1889-1895. [PMID: 29594364 DOI: 10.1007/s00705-018-3808-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Accepted: 02/15/2018] [Indexed: 12/25/2022]
Abstract
Rabies, which is caused by the rabies virus (RABV), is an ancient zoonosis that has a high mortality rate. Previous studies have indicated that recombinant RABV expressing canine interleukin-6 (rHEP-CaIL6), induced more virus-neutralizing antibodies than parental RABV in mice following intramuscular immunization. To investigate the immune response induced in the CNS by rHEP-CaIL6 after intranasal or intracranial administration in mice, the permeability of the blood-brain barrier (BBB), the infiltration of CD3 T cells, and innate immune response-related effector molecules in the CNS were examined. It was observed that infection of rHEP-CaIL6 led to enhanced BBB permeability following intranasal infection. More CD3 T cells infiltrated into the central nervous system (CNS) in mice infected with rHEP-CaIL6 than in those infected with the HEP-Flury strain. Furthermore, rHEP-CaIL6 induced an increased expression of innate immune response-related effector molecules, compared with the parental HEP-Flury strain, within the CNS. Taken together, these findings suggest that rHEP-CaIL6 induced stronger immune responses in mice brains, which is more beneficial for virus clearance. These results may also partly illustrate the role of IL6 in RABV infection.
Collapse
|
17
|
Isomura M, Yamada K, Noguchi K, Nishizono A. Near-infrared fluorescent protein iRFP720 is optimal for in vivo fluorescence imaging of rabies virus infection. J Gen Virol 2017; 98:2689-2698. [PMID: 29039733 DOI: 10.1099/jgv.0.000950] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
In vivo imaging is a noninvasive method that enables real-time monitoring of viral infection dynamics in a small animal, which allows a better understanding of viral pathogenesis. In vivo bioluminescence imaging of virus infection is widely used but, despite its advantage over bioluminescence that no substrate administration is required, fluorescence imaging is not used because of severe autofluorescence. Recently, several far-red and near-infrared (NIR) fluorescent proteins (FPs) have been developed and shown to be useful for whole-body fluorescence imaging. Here, we report comparative testing of far-red and NIR FPs in the imaging of rabies virus (RABV) infection. Using the highly neuroinvasive 1088 strain, we generated recombinant RABV that expressed FPs such as Katushka2S, E2-Crimson, iRFP670 or iRFP720. After intracerebral inoculation to nude mice, the 1088 strain expressing iRFP720, the most red-shifted FP, was detected the earliest with the highest signal-to-noise ratio using a filter set for >700 nm, in which the background signal level was very low. Furthermore, we could also track viral dissemination from the spinal cord to the brain in nude mice after intramuscular inoculation of iRFP720-expressing 1088 into the hind limb. Hence, we conclude that the NIR FP iRFP720 used with a filter set for >700 nm is useful for in vivo fluorescence imaging not only for RABV infection but also for other virus infections. Our findings will also be useful for developing dual-optical imaging of virus-host interaction dynamics using bioluminescence reporter mice for inflammation imaging.
Collapse
Affiliation(s)
- Minori Isomura
- Department of Microbiology, Faculty of Medicine, Oita University, Yufu city, Oita, Japan
| | - Kentaro Yamada
- Department of Microbiology, Faculty of Medicine, Oita University, Yufu city, Oita, Japan.,Research Promotion Institute, Oita University, Yufu city, Oita, Japan
| | - Kazuko Noguchi
- Department of Microbiology, Faculty of Medicine, Oita University, Yufu city, Oita, Japan.,Present address: Department of Food Science and Technology, Minami Kyusyu University, Miyazaki city, Miyazaki, Japan
| | - Akira Nishizono
- Department of Microbiology, Faculty of Medicine, Oita University, Yufu city, Oita, Japan
| |
Collapse
|
18
|
Rescue of a wild-type rabies virus from cloned cDNA and assessment of the proliferative capacity of recombinant viruses. Virus Genes 2017; 53:573-583. [DOI: 10.1007/s11262-017-1458-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Accepted: 04/18/2017] [Indexed: 01/07/2023]
|
19
|
Mei M, Long T, Zhang Q, Zhao J, Tian Q, Peng J, Luo J, Wang Y, Lin Y, Guo X. Phenotypic Consequences In vivo and In vitro of Rearranging the P Gene of RABV HEP-Flury. Front Microbiol 2017; 8:120. [PMID: 28217116 PMCID: PMC5289960 DOI: 10.3389/fmicb.2017.00120] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Accepted: 01/17/2017] [Indexed: 12/24/2022] Open
Abstract
Phosphoprotein (P) of the Rabies virus (RABV) is critically required for viral replication and pathogenicity. Here we manipulated infectious cDNA clones of the RABV HEP-Flury to translocate the P gene from its wild-type position 2 to 1, 3, or 4 in gene order, using an approach which left the viral nucleotide sequence unaltered. The recovered viruses were evaluated for the levels of gene expression, growth kinetics in cell culture, lethality in suckling mice and protection of mice. The results showed that viral replication was affected by the absolute value of N protein which was regulated by P protein. Viral lethality in suckling mice was consistent with the ratio of P mRNA in one complete transcription. The protection of mice induced by viruses was related to the antibody titer 5 weeks post-infection which might be regulated by G protein. However, the ability to induce cell apoptosis and viral spread were not only related to the viral replication but also to the ratio of related gene which affected by the gene position. These findings might not only improve the understanding of phenotype of RABV and P gene rearrangement, but also help rabies vaccine candidate construction.
Collapse
Affiliation(s)
- Mingzhu Mei
- College of Veterinary Medicine, South China Agricultural UniversityGuangzhou, China; Key Laboratory of Zoonosis Prevention and Control of Guangdong ProvinceGuangzhou, China
| | - Teng Long
- College of Veterinary Medicine, South China Agricultural UniversityGuangzhou, China; Key Laboratory of Zoonosis Prevention and Control of Guangdong ProvinceGuangzhou, China
| | - Qiong Zhang
- College of Veterinary Medicine, South China Agricultural UniversityGuangzhou, China; Key Laboratory of Zoonosis Prevention and Control of Guangdong ProvinceGuangzhou, China
| | - Jing Zhao
- College of Veterinary Medicine, South China Agricultural UniversityGuangzhou, China; Key Laboratory of Zoonosis Prevention and Control of Guangdong ProvinceGuangzhou, China
| | - Qin Tian
- College of Veterinary Medicine, South China Agricultural UniversityGuangzhou, China; Key Laboratory of Zoonosis Prevention and Control of Guangdong ProvinceGuangzhou, China
| | - Jiaojiao Peng
- College of Veterinary Medicine, South China Agricultural UniversityGuangzhou, China; Key Laboratory of Zoonosis Prevention and Control of Guangdong ProvinceGuangzhou, China
| | - Jun Luo
- College of Veterinary Medicine, South China Agricultural UniversityGuangzhou, China; Key Laboratory of Zoonosis Prevention and Control of Guangdong ProvinceGuangzhou, China
| | - Yifei Wang
- College of Veterinary Medicine, South China Agricultural UniversityGuangzhou, China; Key Laboratory of Zoonosis Prevention and Control of Guangdong ProvinceGuangzhou, China
| | - Yingyi Lin
- College of Veterinary Medicine, South China Agricultural UniversityGuangzhou, China; Key Laboratory of Zoonosis Prevention and Control of Guangdong ProvinceGuangzhou, China
| | - Xiaofeng Guo
- College of Veterinary Medicine, South China Agricultural UniversityGuangzhou, China; Key Laboratory of Zoonosis Prevention and Control of Guangdong ProvinceGuangzhou, China
| |
Collapse
|
20
|
Expression of interleukin-6 by a recombinant rabies virus enhances its immunogenicity as a potential vaccine. Vaccine 2017; 35:938-944. [DOI: 10.1016/j.vaccine.2016.12.069] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Revised: 12/20/2016] [Accepted: 12/20/2016] [Indexed: 11/13/2022]
|
21
|
Two potential recombinant rabies vaccines expressing canine parvovirus virion protein 2 induce immunogenicity to canine parvovirus and rabies virus. Vaccine 2016; 34:4392-8. [PMID: 27449079 DOI: 10.1016/j.vaccine.2016.07.020] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Revised: 07/12/2016] [Accepted: 07/14/2016] [Indexed: 12/25/2022]
Abstract
Both rabies virus (RABV) and canine parvovirus (CPV) cause lethal diseases in dogs. In this study, both high egg passage Flury (HEP-Flury) strains of RABV and recombinant RABV carrying double RABV glycoprotein (G) gene were used to express the CPV virion protein 2 (VP2) gene, and were designated rHEP-VP2 and, rHEP-dG-VP2 respectively. The two recombinant RABVs maintained optimal virus titration according to their viral growth kinetics assay compared with the parental strain HEP-Flury. Western blotting indicated that G protein and VP2 were expressed in vitro. The expression of VP2 in Crandell feline kidney cells post-infection by rHEP-VP2 and rHEP-dG-VP2 was confirmed by indirect immunofluorescence assay with antibody against VP2. Immunogenicity of recombinant rabies viruses was tested in Kunming mice. Both rHEP-VP2 and rHEP-dG-VP2 induced high levels of rabies antibody compared with HEP-Flury. Mice immunized with rHEP-VP2 and rHEP-dG-VP2 both had a high level of antibodies against VP2, which can protect against CPV infection. A challenge experiment indicated that more than 80% mice immunized with recombinant RABVs survived after infection of challenge virus standard 24 (CVS-24). Together, this study showed that recombinant RABVs expressing VP2 induced protective immune responses to RABV and CPV. Therefore, rHEP-VP2 and rHEP-dG-VP2 might be potential combined vaccines for RABV and CPV.
Collapse
|