1
|
Chai S, Wang L, Du H, Jiang H. Achievement and Challenges in Orthohantavirus Vaccines. Vaccines (Basel) 2025; 13:198. [PMID: 40006744 PMCID: PMC11861054 DOI: 10.3390/vaccines13020198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 02/08/2025] [Accepted: 02/12/2025] [Indexed: 02/27/2025] Open
Abstract
Orthohantaviruses (also known as hantaviruses) are pathogens that cause two distinct, yet related forms of severe human disease: hemorrhagic fever with renal syndrome (HFRS) and hantavirus pulmonary syndrome (HPS). These diseases pose a significant threat to global public health due to their high case fatality rates, which can range from 1% to 50%. In recent years, an increasing number of countries and regions have reported human cases, underscoring the urgent need for improved understanding, prevention, and treatment strategies. Given the severity of these diseases and the lack of specific post-exposure antiviral treatments, preventive measures are critical. For several decades, substantial efforts have been dedicated to developing orthohantavirus vaccines, leading to significant advancements. The first large-scale deployment involved inactivated vaccines, which played a crucial role in reducing HFRS incidence in South Korea and China. Subunit vaccines, viral vector vaccines, and virus-like particle (VLP) vaccines have also been extensively researched. Nucleic acid vaccines, including both mRNA and DNA vaccines, hold the greatest potential for future development due to their rapid design and production cycles, ability to elicit robust immune responses, ease of storage and transportation, and adaptable production platforms. Ongoing advancements in computer technology and artificial intelligence promise to further enhance the development of more effective orthohantavirus vaccines.
Collapse
Affiliation(s)
- Shiqi Chai
- Center for Diagnosis and Treatment of Infectious Diseases, TangDu Hospital, The Fourth Military Medical University, Xi’an 710038, China;
| | - Limei Wang
- Department of Microbiology and Pathogenic Biology, School of Basic Medicine, The Fourth Military Medical University, Xi’an 710032, China;
| | - Hong Du
- Center for Diagnosis and Treatment of Infectious Diseases, TangDu Hospital, The Fourth Military Medical University, Xi’an 710038, China;
| | - Hong Jiang
- Center for Diagnosis and Treatment of Infectious Diseases, TangDu Hospital, The Fourth Military Medical University, Xi’an 710038, China;
| |
Collapse
|
2
|
Song BH, Frank JC, Yun SI, Julander JG, Mason JB, Polejaeva IA, Davies CJ, White KL, Dai X, Lee YM. Comparison of Three Chimeric Zika Vaccine Prototypes Developed on the Genetic Background of the Clinically Proven Live-Attenuated Japanese Encephalitis Vaccine SA 14-14-2. Int J Mol Sci 2024; 26:195. [PMID: 39796052 PMCID: PMC11720029 DOI: 10.3390/ijms26010195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 12/17/2024] [Accepted: 12/24/2024] [Indexed: 01/13/2025] Open
Abstract
Zika virus (ZIKV) is a medically important mosquito-borne orthoflavivirus, but no vaccines are currently available to prevent ZIKV-associated disease. In this study, we compared three recombinant chimeric viruses developed as candidate vaccine prototypes (rJEV/ZIKVMR-766, rJEV/ZIKVP6-740, and rJEV/ZIKVPRVABC-59), in which the two neutralizing antibody-inducing prM and E genes from each of three genetically distinct ZIKV strains were used to replace the corresponding genes of the clinically proven live-attenuated Japanese encephalitis virus vaccine SA14-14-2 (rJEV). In WHO-certified Vero cells (a cell line suitable for vaccine production), rJEV/ZIKVP6-740 exhibited the slowest viral growth, formed the smallest plaques, and displayed a unique protein expression profile with the highest ratio of prM to cleaved M when compared to the other two chimeric viruses, rJEV/ZIKVMR-766 and rJEV/ZIKVPRVABC-59, as well as their vector, rJEV. In IFNAR-/- mice, an animal model of ZIKV infection, subcutaneous inoculation of rJEV/ZIKVP6-740 caused a low-level localized infection limited to the spleen, with no clinical signs of infection, weight loss, or mortality; in contrast, the other two chimeric viruses and their vector caused high-level systemic infections involving multiple organs, consistently leading to clear clinical signs of infection, rapid weight loss, and 100% mortality. Subsequently, subcutaneous immunization with rJEV/ZIKVP6-740 proved highly effective, offering complete protection against a lethal intramuscular ZIKV challenge 28 days after a single-dose immunization. This protection was specific to ZIKV prM/E and likely mediated by neutralizing antibodies targeting ZIKV prM/E. Therefore, our data indicate that the chimeric virus rJEV/ZIKVP6-740 is a highly promising vaccine prototype for developing a safe and effective vaccine for inducing neutralizing antibody-mediated protective immunity against ZIKV.
Collapse
Affiliation(s)
- Byung-Hak Song
- Department of Animal Dairy and Veterinary Sciences, College of Agriculture and Applied Sciences, Utah State University, Logan, UT 84322, USA; (B.-H.S.); (J.C.F.); (S.-I.Y.); (J.G.J.); (I.A.P.); (C.J.D.); (K.L.W.)
| | - Jordan C. Frank
- Department of Animal Dairy and Veterinary Sciences, College of Agriculture and Applied Sciences, Utah State University, Logan, UT 84322, USA; (B.-H.S.); (J.C.F.); (S.-I.Y.); (J.G.J.); (I.A.P.); (C.J.D.); (K.L.W.)
| | - Sang-Im Yun
- Department of Animal Dairy and Veterinary Sciences, College of Agriculture and Applied Sciences, Utah State University, Logan, UT 84322, USA; (B.-H.S.); (J.C.F.); (S.-I.Y.); (J.G.J.); (I.A.P.); (C.J.D.); (K.L.W.)
| | - Justin G. Julander
- Department of Animal Dairy and Veterinary Sciences, College of Agriculture and Applied Sciences, Utah State University, Logan, UT 84322, USA; (B.-H.S.); (J.C.F.); (S.-I.Y.); (J.G.J.); (I.A.P.); (C.J.D.); (K.L.W.)
- Institute for Antiviral Research, Utah State University, Logan, UT 84322, USA
| | - Jeffrey B. Mason
- Department of Veterinary Clinical and Life Sciences, College of Veterinary Medicine, Center for Integrated BioSystems, Utah State University, Logan, UT 84322, USA;
| | - Irina A. Polejaeva
- Department of Animal Dairy and Veterinary Sciences, College of Agriculture and Applied Sciences, Utah State University, Logan, UT 84322, USA; (B.-H.S.); (J.C.F.); (S.-I.Y.); (J.G.J.); (I.A.P.); (C.J.D.); (K.L.W.)
| | - Christopher J. Davies
- Department of Animal Dairy and Veterinary Sciences, College of Agriculture and Applied Sciences, Utah State University, Logan, UT 84322, USA; (B.-H.S.); (J.C.F.); (S.-I.Y.); (J.G.J.); (I.A.P.); (C.J.D.); (K.L.W.)
| | - Kenneth L. White
- Department of Animal Dairy and Veterinary Sciences, College of Agriculture and Applied Sciences, Utah State University, Logan, UT 84322, USA; (B.-H.S.); (J.C.F.); (S.-I.Y.); (J.G.J.); (I.A.P.); (C.J.D.); (K.L.W.)
| | - Xin Dai
- Utah Agricultural Experiment Station, Utah State University, Logan, UT 84322, USA;
| | - Young-Min Lee
- Department of Animal Dairy and Veterinary Sciences, College of Agriculture and Applied Sciences, Utah State University, Logan, UT 84322, USA; (B.-H.S.); (J.C.F.); (S.-I.Y.); (J.G.J.); (I.A.P.); (C.J.D.); (K.L.W.)
| |
Collapse
|
3
|
Zhao C, Chen J, Liu Z, Liang H, Chen X, Cheng L, Xie S, Lin Z, Wu R, Zhao Q, Xue Y, Lai X, Jin X, Xu JF, Su X. Activation of nicotinic acetylcholine receptor α7 subunit limits Zika viral infection via promoting autophagy and ferroptosis. Mol Ther 2024; 32:2641-2661. [PMID: 38822526 PMCID: PMC11405161 DOI: 10.1016/j.ymthe.2024.05.037] [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: 12/04/2023] [Revised: 04/13/2024] [Accepted: 05/29/2024] [Indexed: 06/03/2024] Open
Abstract
Vagus nerve regulates viral infection and inflammation via the alpha 7 nicotinic acetylcholine receptor (α7 nAChR); however, the role of α7 nAChR in ZIKA virus (ZIKV) infection, which can cause severe neurological diseases such as microcephaly and Guillain-Barré syndrome, remains unknown. Here, we first examined the role of α7 nAChR in ZIKV infection in vitro. A broad effect of α7 nAChR activation was identified in limiting ZIKV infection in multiple cell lines. Combined with transcriptomics analysis, we further demonstrated that α7 nAChR activation promoted autophagy and ferroptosis pathways to limit cellular ZIKV viral loads. Additionally, activation of α7 nAChR prevented ZIKV-induced p62 nucleus accumulation, which mediated an enhanced autophagy pathway. By regulating proteasome complex and an E3 ligase NEDD4, activation of α7 nAChR resulted in increased amount of cellular p62, which further enhanced the ferroptosis pathway to reduce ZIKV infection. Moreover, utilizing in vivo neonatal mouse models, we showed that α7 nAChR is essential in controlling the disease severity of ZIKV infection. Taken together, our findings identify an α7 nAChR-mediated effect that critically contributes to limiting ZIKV infection, and α7 nAChR activation offers a novel strategy for combating ZIKV infection and its complications.
Collapse
Affiliation(s)
- Caiqi Zhao
- Unit of Respiratory Infection and Immunity, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai 200032, China; Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200000, China; University of Chinese Academy of Sciences, Beijing 100190, China
| | - Jie Chen
- Unit of Respiratory Infection and Immunity, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai 200032, China; University of Chinese Academy of Sciences, Beijing 100190, China; Shanghai Key Laboratory of Lung Inflammation and Injury, Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Zhihua Liu
- University of Chinese Academy of Sciences, Beijing 100190, China; Vaccine Center, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai 200032, China
| | - Huabin Liang
- University of Chinese Academy of Sciences, Beijing 100190, China; Vaccine Center, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai 200032, China
| | - Xiaoyan Chen
- Shanghai Key Laboratory of Lung Inflammation and Injury, Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Lianping Cheng
- Unit of Respiratory Infection and Immunity, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai 200032, China
| | - Shitao Xie
- Unit of Respiratory Infection and Immunity, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai 200032, China
| | - Zhekai Lin
- Unit of Respiratory Infection and Immunity, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai 200032, China
| | - Renlan Wu
- Unit of Respiratory Infection and Immunity, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai 200032, China
| | - Qi Zhao
- Unit of Respiratory Infection and Immunity, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai 200032, China
| | - Yue Xue
- Unit of Respiratory Infection and Immunity, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai 200032, China
| | - Xiaoyun Lai
- Unit of Respiratory Infection and Immunity, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai 200032, China
| | - Xia Jin
- University of Chinese Academy of Sciences, Beijing 100190, China; Vaccine Center, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai 200032, China.
| | - Jin-Fu Xu
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200000, China.
| | - Xiao Su
- Unit of Respiratory Infection and Immunity, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai 200032, China; Vaccine Center, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai 200032, China; Shanghai Key Laboratory of Lung Inflammation and Injury, Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China.
| |
Collapse
|
4
|
Miranda-López A, González-Ortega O, Govea-Alonso DO, Betancourt-Mendiola L, Comas-García M, Rosales-Mendoza S. Rational design and production of a chimeric antigen targeting Zika virus that induces neutralizing antibodies in mice. Vaccine 2024; 42:3674-3683. [PMID: 38749821 DOI: 10.1016/j.vaccine.2024.04.080] [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: 12/14/2023] [Revised: 03/14/2024] [Accepted: 04/25/2024] [Indexed: 06/07/2024]
Abstract
The Zika virus (ZIKV) is considered a public health problem worldwide due to its association with the development of microcephaly and the Guillain-Barré syndrome. Currently, there is no specific treatment or vaccine approved to combat this disease, and thus, developing safe and effective vaccines is a relevant goal. In this study, a multi-epitope protein called rpZDIII was designed based on a series of ZIKV antigenic sequences, a bacterial carrier, and linkers. The analysis of the predicted 3D structure of the rpZDIII chimeric antigen was performed on the AlphaFold 2 server, and it was produced in E. coli and purified from inclusion bodies, followed by solubilization and refolding processes. The yield achieved for rpZDIII was 11 mg/L in terms of pure soluble recombinant protein per liter of fermentation. rpZDIII was deemed immunogenic since it induced serum IgG and IgM responses in mice upon subcutaneous immunization in a three-dose scheme. Moreover, sera from mice immunized with rpZDIII showed neutralizing activity against ZIKV. Therefore, this study reveals rpZDIII as a promising immunogen for the development of a rationally designed multi-epitope vaccine against ZIKV, and completion of its preclinical evaluation is guaranteed.
Collapse
Affiliation(s)
- Arleth Miranda-López
- Facultad de Ciencias Químicas, Universidad Autónoma de San Luis Potosí, Av. Dr. Manuel Nava 6, SLP 78210, México
| | - Omar González-Ortega
- Facultad de Ciencias Químicas, Universidad Autónoma de San Luis Potosí, Av. Dr. Manuel Nava 6, SLP 78210, México; Sección de Biotecnología, Centro de Investigación en Ciencias de la Salud y Biomedicina, Universidad Autónoma de San Luis Potosí, Av. Sierra Leona 550, Lomas 2ª. Sección, San Luis Potosí 78210 México
| | - Dania O Govea-Alonso
- Sección de Biotecnología, Centro de Investigación en Ciencias de la Salud y Biomedicina, Universidad Autónoma de San Luis Potosí, Av. Sierra Leona 550, Lomas 2ª. Sección, San Luis Potosí 78210 México
| | - Lourdes Betancourt-Mendiola
- Facultad de Ciencias Químicas, Universidad Autónoma de San Luis Potosí, Av. Dr. Manuel Nava 6, SLP 78210, México; Sección de Biotecnología, Centro de Investigación en Ciencias de la Salud y Biomedicina, Universidad Autónoma de San Luis Potosí, Av. Sierra Leona 550, Lomas 2ª. Sección, San Luis Potosí 78210 México
| | - Mauricio Comas-García
- Sección de Microscopía de Alta Resolución, Centro de Investigación en Ciencias de la Salud y Biomedicina, Universidad Autónoma de San Luis Potosí, Av. Sierra Leona 550, Lomas 2ª. Sección, San Luis Potosí 78210, México; Facultad de Ciencias, Universidad Autónoma de San Luis Potosí, Av. Parque Chapultepec 1570, San Luis, S.L.P., San Luis Potosí 78210, México.
| | - Sergio Rosales-Mendoza
- Facultad de Ciencias Químicas, Universidad Autónoma de San Luis Potosí, Av. Dr. Manuel Nava 6, SLP 78210, México; Sección de Biotecnología, Centro de Investigación en Ciencias de la Salud y Biomedicina, Universidad Autónoma de San Luis Potosí, Av. Sierra Leona 550, Lomas 2ª. Sección, San Luis Potosí 78210 México.
| |
Collapse
|
5
|
da Costa HHM, Bielavsky M, Orts DJB, Araujo S, Adriani PP, Nogueira JS, Astray RM, Pandey RP, Lancellotti M, Cunha-Junior JP, Prudencio CR. Production of Recombinant Zika Virus Envelope Protein by Airlift Bioreactor as a New Subunit Vaccine Platform. Int J Mol Sci 2023; 24:13955. [PMID: 37762254 PMCID: PMC10531330 DOI: 10.3390/ijms241813955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 03/23/2023] [Accepted: 04/04/2023] [Indexed: 09/29/2023] Open
Abstract
The Zika Virus (ZIKV) is an emerging arbovirus of great public health concern, particularly in the Americas after its last outbreak in 2015. There are still major challenges regarding disease control, and there is no ZIKV vaccine currently approved for human use. Among many different vaccine platforms currently under study, the recombinant envelope protein from Zika Virus (rEZIKV) constitutes an alternative option for vaccine development and has great potential for monitoring ZIKV infection and antibody response. This study describes a method to obtain a bioactive and functional rEZIKV using an E. coli expression system, with the aid of a 5-L airlift bioreactor and following an automated fast protein liquid chromatography (FPLC) protocol, capable of obtaining high yields of approximately 20 mg of recombinant protein per liter of bacterium cultures. The purified rEZIKV presented preserved antigenicity and immunogenicity. Our results show that the use of an airlift bioreactor for the production of rEZIKV is ideal for establishing protocols and further research on ZIKV vaccines bioprocess, representing a promising system for the production of a ZIKV envelope recombinant protein-based vaccine candidate.
Collapse
Affiliation(s)
- Hernan H. M. da Costa
- Immunology Center, Adolfo Lutz Institute, São Paulo 01246-902, Brazil
- Interunits Graduate Program in Biotechnology, University of São Paulo, São Paulo 05508-000, Brazil
| | - Monica Bielavsky
- Immunology Center, Adolfo Lutz Institute, São Paulo 01246-902, Brazil
| | - Diego J. B. Orts
- Immunology Center, Adolfo Lutz Institute, São Paulo 01246-902, Brazil
- Laboratory of Cardiobiology, Department of Biophysics, Paulista School of Medicine, Federal University of Sao Paulo, São Paulo 04023-062, Brazil
| | - Sergio Araujo
- Immunology Center, Adolfo Lutz Institute, São Paulo 01246-902, Brazil
| | - Patrícia P. Adriani
- Skinzymes Biotechnology Ltd., São Paulo 05441-040, Brazil
- Laboratory of Nanopharmaceuticals and Delivery Systems, Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil
| | | | - Renato M. Astray
- Interunits Graduate Program in Biotechnology, University of São Paulo, São Paulo 05508-000, Brazil
- Multi-Purpose Laboratory Butantan Institute, São Paulo 05503-900, Brazil
| | - Ramendra P. Pandey
- School of Health Sciences and Technology, UPES University, Dehradun 248007, Uttarakhand, India
| | - Marcelo Lancellotti
- Faculty of Pharmaceutical Sciences—FCF, University of Campinas—UNICAMP, Campinas 13083-871, Brazil
| | - Jair P. Cunha-Junior
- Laboratory of Immunochemistry and Immunotechnology, Department of Immunology, Federal University of Uberlândia, Uberlândia 38405-317, Brazil
| | - Carlos R. Prudencio
- Immunology Center, Adolfo Lutz Institute, São Paulo 01246-902, Brazil
- Interunits Graduate Program in Biotechnology, University of São Paulo, São Paulo 05508-000, Brazil
| |
Collapse
|
6
|
Liu R, Lv Y, Sun W, Li M, Ge N, Zhu C, Ding Y, Liu Z, Ma R, Huang Y, Hou S, Ying Q, Gu T, Wang F, Nie L, Wang Y, Huang W, Shu J, Wu X. Investigation of a subunit protein vaccine for HFRS based on a consensus sequence between envelope glycoproteins of HTNV and SEOV. Virus Res 2023; 334:199149. [PMID: 37329903 PMCID: PMC10410520 DOI: 10.1016/j.virusres.2023.199149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 06/01/2023] [Accepted: 06/07/2023] [Indexed: 06/19/2023]
Abstract
Due to the global resurgence of hemorrhagic fever with renal syndrome (HFRS), more attention is being focused on this dangerous illness. In China and Korea, the only vaccines available are the virus-inactivated vaccine against Hantaan virus (HTNV) or Seoul virus (SEOV), but their efficacy and safety are inadequate. Therefore, it is important to develop new vaccines that are safer and more efficient to neutralize and regulate areas with a high prevalence of HFRS. We employed bioinformatics methods to design a recombinant protein vaccine based on conserved regions of protein consensus sequences in HTNV and SEOV membranes. The S2 Drosophila expression system was utilized to enhance protein expression, solubility and immunogenicity. After the Gn and Gc proteins of HTNV and SEOV were successfully expressed, mice were immunized, and the humoral immunity, cellular immunity, and in vivo protection of the HFRS universal subunit vaccine were systematically evaluated in mouse models. These results indicated that the HFRS subunit vaccine generated elevated levels of binding and neutralizing antibodies, particularly IgG1, compared to that of the traditional inactivated HFRS vaccine. Additionally, the spleen cells of immunized mice secreted IFN-r and IL-4 cytokines effectively. Moreover, the HTNV-Gc protein vaccine successfully protected suckling mice from HTNV infection and stimulated GC responses. In this research, a new scientific approach is investigated to develop a universal HFRS subunit protein vaccine that is capable of producing effective humoral and cellular immunity in mice. The results suggest that this vaccine could be a promising candidate for preventing HFRS in humans.
Collapse
Affiliation(s)
- Rongrong Liu
- Department of Microbiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Yunhua Lv
- Department of Microbiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Wenjie Sun
- Department of Microbiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China; Northwest University, Xi'an, China
| | - Min Li
- Institute Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
| | - Ningning Ge
- Institute Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
| | - Cheng Zhu
- Tianjin Key Laboratory of Function and Application of Biological Macromolecular Structures, School of Life Sciences, Tianjin University, Tianjin, China
| | - Yaxin Ding
- Department of Microbiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China; Northwest University, Xi'an, China
| | - Ziyu Liu
- Department of Microbiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Ruixue Ma
- Department of Microbiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Yuxiao Huang
- Department of Microbiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Shiyuan Hou
- Department of Microbiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Qikang Ying
- Department of Microbiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Tianle Gu
- Department of Microbiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Fang Wang
- Department of Microbiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Lingling Nie
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC) and WHO Collaborating Center for Standardization and Evaluation of Biologicals, Beijing, China
| | - Youchun Wang
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC) and WHO Collaborating Center for Standardization and Evaluation of Biologicals, Beijing, China
| | - Weijin Huang
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC) and WHO Collaborating Center for Standardization and Evaluation of Biologicals, Beijing, China.
| | - Jiayi Shu
- Clinical Center for Biotherapy, Zhongshan Hospital & Zhongshan Hospital (Xiamen), Fudan University, Shanghai, China.
| | - Xingan Wu
- Department of Microbiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China.
| |
Collapse
|
7
|
Lunardelli VAS, Almeida BDS, Apostolico JDS, Rezende T, Yamamoto MM, Pereira SS, Bueno MFC, Pereira LR, Carvalho KI, Slhessarenko RD, de Souza Ferreira LC, Boscardin SB, Rosa DS. Diagnostic and vaccine potential of Zika virus envelope protein (E) derivates produced in bacterial and insect cells. Front Immunol 2023; 14:1071041. [PMID: 37006270 PMCID: PMC10060818 DOI: 10.3389/fimmu.2023.1071041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Accepted: 03/03/2023] [Indexed: 03/18/2023] Open
Abstract
IntroductionIn the present study we evaluated the features of different recombinant forms of Zika virus (ZIKV) proteins produced in either bacterial (Eschericha coli) or insect cells (Drosophila melanogaster). The ZIKV-envelope glycoprotein (EZIKV) is responsible for virus entry into host cells, is the main target of neutralizing antibodies and has been used as a target antigen either for serological tests or for the development of subunit vaccines. The EZIKV is composed of three structural and functional domains (EDI, EDII, and EDIII), which share extensive sequence conservation with the corresponding counterparts expressed by other flaviviruses, particularly the different dengue virus (DENV) subtypes.MethodsIn this study, we carried out a systematic comparison of the antigenicity and immunogenicity of recombinant EZIKV, EDI/IIZIKV and EDIIIZIKV produced in E. coli BL21 and Drosophila S2 cells. For the antigenicity analysis we collected 88 serum samples from ZIKV-infected participants and 57 serum samples from DENV-infected. For immunogenicity, C57BL/6 mice were immunized with two doses of EZIKV, EDI/IIZIKV and EDIIIZIKV produced in E. coli BL21 and Drosophila S2 cells to evaluate humoral and cellular immune response. In addition, AG129 mice were immunized with EZIKV and then challenge with ZIKV.ResultsTesting of samples collected from ZIKV-infected and DENV-infected participants demonstrated that the EZIKV and EDIIIZIKV produced in BL21 cells presented better sensitivity and specificity compared to proteins produced in S2 cells. In vivo analyses were carried out with C57BL/6 mice and the results indicated that, despite similar immunogenicity, antigens produced in S2 cells, particularly EZIKV and EDIIIZIKV, induced higher ZIKV-neutralizing antibody levels in vaccinated mice. In addition, immunization with EZIKV expressed in S2 cells delayed the onset of symptoms and increased survival rates in immunocompromised mice. All recombinant antigens, either produced in bacteria or insect cells, induced antigen-specific CD4+ and CD8+ T cell responses.ConclusionIn conclusion, the present study highlights the differences in antigenicity and immunogenicity of recombinant ZIKV antigens produced in two heterologous protein expression systems.
Collapse
Affiliation(s)
- Victória Alves Santos Lunardelli
- Departmento de Microbiologia, Imunologia e Parasitologia, Universidade Federal de São Paulo- Escola Paulista de Medicina (UNIFESP/EPM), São Paulo, Brazil
| | - Bianca da Silva Almeida
- Departmento de Parasitologia, Instituto de Ciências Biomédicas, Universidade de São Paulo (USP), São Paulo, Brazil
| | - Juliana de Souza Apostolico
- Departmento de Microbiologia, Imunologia e Parasitologia, Universidade Federal de São Paulo- Escola Paulista de Medicina (UNIFESP/EPM), São Paulo, Brazil
| | - Thais Rezende
- Departmento de Microbiologia, Imunologia e Parasitologia, Universidade Federal de São Paulo- Escola Paulista de Medicina (UNIFESP/EPM), São Paulo, Brazil
| | - Marcio Massao Yamamoto
- Departmento de Parasitologia, Instituto de Ciências Biomédicas, Universidade de São Paulo (USP), São Paulo, Brazil
| | - Samuel Santos Pereira
- Departmento de Microbiologia, Instituto de Ciências Biomédicas, Universidade de São Paulo (USP), São Paulo, Brazil
| | - Maria Fernanda Campagnari Bueno
- Departmento de Microbiologia, Imunologia e Parasitologia, Universidade Federal de São Paulo- Escola Paulista de Medicina (UNIFESP/EPM), São Paulo, Brazil
| | - Lennon Ramos Pereira
- Departmento de Microbiologia, Instituto de Ciências Biomédicas, Universidade de São Paulo (USP), São Paulo, Brazil
| | - Karina Inacio Carvalho
- Hospital Israelita Albert Einstein, São Paulo, Brazil
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, United States
| | | | - Luis Carlos de Souza Ferreira
- Departmento de Microbiologia, Instituto de Ciências Biomédicas, Universidade de São Paulo (USP), São Paulo, Brazil
- Plataforma Científica Pasteur- Universidade de São Paulo, São Paulo, Brazil
| | - Silvia Beatriz Boscardin
- Departmento de Parasitologia, Instituto de Ciências Biomédicas, Universidade de São Paulo (USP), São Paulo, Brazil
- Instituto Nacional de Ciência e Tecnologia (INCT) de Investigação em Imunologia (iii), São Paulo, Brazil
| | - Daniela Santoro Rosa
- Departmento de Microbiologia, Imunologia e Parasitologia, Universidade Federal de São Paulo- Escola Paulista de Medicina (UNIFESP/EPM), São Paulo, Brazil
- Instituto Nacional de Ciência e Tecnologia (INCT) de Investigação em Imunologia (iii), São Paulo, Brazil
- *Correspondence: Daniela Santoro Rosa,
| |
Collapse
|
8
|
Pereira GC. An Automated Strategy to Handle Antigenic Variability in Immunisation Protocols, Part II: In Vitro Transcribed mRNA Vector Design for Inoculation Against Infectious Agent Variants. Methods Mol Biol 2023; 2575:323-340. [PMID: 36301484 DOI: 10.1007/978-1-0716-2716-7_17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
A fully automated strategy to handle antigenic variability in immunisation protocols is here presented. The method comprises of (1) nanopore sequencing of infectious agent variants, with focus on the SARS-CoV-2 and its variants, followed by (2) in-vitro transcribed mRNA vector design for immunotherapy. This chapter introduces the mRNA vector design protocol and Chapter 16 presents the nano-pore sequencing step.
Collapse
|
9
|
Shin M, Kim K, Lee HJ, Jung YJ, Park J, Hahn TW. Vaccination with a Zika virus envelope domain III protein induces neutralizing antibodies and partial protection against Asian genotype in immunocompetent mice. Trop Med Health 2022; 50:91. [PMID: 36471432 PMCID: PMC9721077 DOI: 10.1186/s41182-022-00485-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 11/24/2022] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Zika virus (ZIKV) is a mosquito-borne flavivirus classified in Flaviviridae family such as dengue (DENV), yellow fever, and West Nile virus. An outbreak of ZIKV infection can pose a major public health risk because the contagion is unpredictable and induces severe pathology such as Guillan-Barre syndrome and neonatal microcephaly. However, an authorized ZIKV vaccine is not yet available, while several vaccine candidates are under development. METHODS In this study, we constructed a recombinant ZIKV vaccine (Z_EDIII) that includes ZIKV envelope protein domain III using E. coli expression system. Then both humoral and cellular immunity were examined in C57BL/6 (female, 8-weeks-old) mice via Indirect ELISA assay, PRNT, ELISpot and cytokine detection for IFN-γ, TNF-α, and IL-12. In addition, the cross protection against DENV was evaluated in pups from Z_EDIII vaccinated and infected dam. RESULTS Mice immunized by Z_EDIII produced a significant amount of ZIKV EDIII-specific and neutralizing antibodies. Together with antibodies, effector cytokines, such as IFN-γ, TNF-α, and IL-12 were induced. Moreover, vaccinated females delivered the adaptive immunity to neonates who are protective against ZIKV and DENV challenge. CONCLUSIONS This study observed Z-EDIII-induced humoral and cellular immunity that protected hosts from both ZIKV and DENV challenges. The result suggests that our ZIKV EDIII recombinant vaccine has potential to provide a new preventive strategy against ZIKV infection.
Collapse
Affiliation(s)
- Minna Shin
- INNOVAC, Chuncheon, 24341 Republic of Korea
| | - Kiju Kim
- INNOVAC, Chuncheon, 24341 Republic of Korea ,grid.412010.60000 0001 0707 9039College of Veterinary Medicine & Institute of Veterinary Science, Kangwon National University, Chuncheon, 24341 Republic of Korea
| | - Hyo-Ji Lee
- grid.412010.60000 0001 0707 9039College of Biological Sciences, Kangwon National University, Chuncheon, 24341 Republic of Korea
| | - Yu-Jin Jung
- grid.412010.60000 0001 0707 9039College of Biological Sciences, Kangwon National University, Chuncheon, 24341 Republic of Korea
| | - Jeongho Park
- grid.412010.60000 0001 0707 9039College of Veterinary Medicine & Institute of Veterinary Science, Kangwon National University, Chuncheon, 24341 Republic of Korea
| | - Tae-Wook Hahn
- INNOVAC, Chuncheon, 24341 Republic of Korea ,grid.412010.60000 0001 0707 9039College of Veterinary Medicine & Institute of Veterinary Science, Kangwon National University, Chuncheon, 24341 Republic of Korea
| |
Collapse
|
10
|
Wang Y, Ling L, Zhang Z, Marin-Lopez A. Current Advances in Zika Vaccine Development. Vaccines (Basel) 2022; 10:vaccines10111816. [PMID: 36366325 PMCID: PMC9694033 DOI: 10.3390/vaccines10111816] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 10/19/2022] [Accepted: 10/25/2022] [Indexed: 11/16/2022] Open
Abstract
Zika virus (ZIKV), an emerging arthropod-borne flavivirus, was first isolated in Uganda in 1947 from monkeys and first detected in humans in Nigeria in 1952; it has been associated with a dramatic burden worldwide. Since then, interventions to reduce the burden of ZIKV infection have been mainly restricted to mosquito control, which in the end proved to be insufficient by itself. Hence, the situation prompted scientists to increase research on antivirals and vaccines against the virus. These efforts are still ongoing as the pathogenesis and immune evasion mechanisms of ZIKV have not yet been fully elucidated. Understanding the viral disease mechanism will provide a better landscape to develop prophylactic and therapeutic strategies against ZIKV. Currently, no specific vaccines or drugs have been approved for ZIKV. However, some are undergoing clinical trials. Notably, different platforms have been evaluated for the design of vaccines, including DNA, mRNA, viral vectors, virus-like particles (VLPs), inactivated virus, live attenuated virus, peptide and protein-based vaccines, passive immunizations by using monoclonal antibodies (MAbs), and vaccines that target vector-derived antigens. These vaccines have been shown to induce specific humoral and cellular immune responses and reduce viremia and viral RNA titers, both in vitro and in vivo. This review provides a comprehensive summary of current advancements in the development of vaccines against Zika virus.
Collapse
Affiliation(s)
- Yuchen Wang
- Department of Inspection and Quarantine Technology Communication, Shanghai Customs College, Shanghai 201204, China
- Correspondence:
| | - Lin Ling
- Department of Inspection and Quarantine Technology Communication, Shanghai Customs College, Shanghai 201204, China
| | - Zilei Zhang
- Department of Inspection and Quarantine Technology Communication, Shanghai Customs College, Shanghai 201204, China
| | - Alejandro Marin-Lopez
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06420, USA
| |
Collapse
|
11
|
Cheong HC, Cheok YY, Chan YT, Sulaiman S, Looi CY, Alshanon AF, Hassan J, Abubakar S, Wong WF. Zika Virus Vaccine: The Current State of Affairs and Challenges Posed by Antibody-Dependent Enhancement Reaction. Viral Immunol 2022; 35:586-596. [PMID: 36301533 DOI: 10.1089/vim.2022.0082] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Affiliation(s)
- Heng Choon Cheong
- Department of Medical Microbiology and Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Yi Ying Cheok
- Department of Medical Microbiology and Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Yee Teng Chan
- Department of Medical Microbiology and Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Sofiah Sulaiman
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Chung Yeng Looi
- School of Biosciences, Faculty of Health & Medical Sciences, Taylor's University, Subang Jaya, Malaysia
| | - Ahmed F. Alshanon
- Center of Biotechnology Researches, University of Al-Nahrain, Baghdad, Iraq
| | - Jamiyah Hassan
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Sazaly Abubakar
- Department of Medical Microbiology and Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
- Tropical Infectious Diseases Research and Educational Center (TIDREC), University of Malaya, Kuala Lumpur, Malaysia
| | - Won Fen Wong
- Department of Medical Microbiology and Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
12
|
Selection and Characterization of Single-Stranded DNA Aptamers of Diagnostic Potential against the Whole Zika Virus. Viruses 2022; 14:v14091867. [PMID: 36146678 PMCID: PMC9504500 DOI: 10.3390/v14091867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 08/17/2022] [Accepted: 08/22/2022] [Indexed: 11/29/2022] Open
Abstract
Zika virus became a major public health problem in early 2015, when cases of Guillain–Barré syndrome and microcephaly were associated with viral infection. Currently, ZIKV is endemic in all tropical areas of the world, and the chance for future Zika epidemics remains very real and accurate diagnosis is crucial. The aim of this work was to select specific ssDNA aptamers that bind to the entire Zika virus and can be used to compose specific diagnostics, without cross-reactivity with other flaviviruses. Zika virus was cultivated in Vero cells and used as a target for aptamer selection. Aptamers specific for the ZIKV were selected using whole-virus SELEX, with counterselection for other flavivirus. Secondary and tertiary structures were evaluated and the molecular anchoring between the aptamers and target were simulated by the HDOCK server. Aptamer interaction was evaluated by ELISA/ELASA and the dissociation constant (Kd) was calculated by thermophoresis. Four ZIKV-specific aptamers were selected. The best two were further characterized and proved to be specific for ZIKV. Aptamers are capable of binding specifically to the ZIKV and differentiate from Dengue virus. The aptamers selected in this work can be used as capture agents in the composition of diagnostic tests to specifically detect ZIKV infection.
Collapse
|
13
|
Ke Q, Sun P, Wang T, Mi T, Xu H, Wu J, Liu B. Non-glycosylated SARS-CoV-2 RBD elicited a robust neutralizing antibody response in mice. J Immunol Methods 2022; 506:113279. [PMID: 35533747 PMCID: PMC9075978 DOI: 10.1016/j.jim.2022.113279] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 05/04/2022] [Indexed: 12/16/2022]
Abstract
The glycosylated receptor-binding domain (glycoRBD) of SARS-CoV-2 can induce protective neutralizing antibodies to function as a vaccine. However, it is unclear whether vaccines using non-glycosylated RBD (non-glycoRBD) can induce protective immunity. Here, we report the efficacy of a SARS-CoV-2 non-glycoRBD vaccine produced by prokaryotic system in mice. The recombinant non-glycoRBD protein was overexpressed in Escherichia coli in the form of inclusion bodies, and was obtained after renaturation and three-step purification. From HPLC analysis, the purity of the RBD was 99%. Additionally, angiotensin converting enzyme 2 (ACE2)-binding assays revealed that E.coli-derived non-glycoRBD had binding activity consistent with glycoRBD. The RBD was formulated with CpG ODN and Al(OH)3 adjuvants and the obtained RBD candidate vaccine elicited potent antibody responses and neutralized SARS-CoV-2 wild-type, Delta, and Omicron pseudoviruses. In summary, our data showed that a non-glycoRBD candidate vaccine produced by E.coli provided a robust immune response and had pseudovirus neutralizing activity, making it a solid candidate vaccine for protection against SARS-CoV-2.
Collapse
Affiliation(s)
- Qian Ke
- Institutes of Physical Science and Information Technology, Anhui University, Hefei 230601, China,Department of Microorganism Engineering, Beijing Institute of Biotechnology, Beijing 100071, China
| | - Peng Sun
- Department of Microorganism Engineering, Beijing Institute of Biotechnology, Beijing 100071, China
| | - Tiantian Wang
- Department of Microorganism Engineering, Beijing Institute of Biotechnology, Beijing 100071, China
| | - Taotao Mi
- Department of Microorganism Engineering, Beijing Institute of Biotechnology, Beijing 100071, China,College of Life Sciences, Shihezi University, Shihezi 832003, China
| | - Huifang Xu
- Department of Microorganism Engineering, Beijing Institute of Biotechnology, Beijing 100071, China
| | - Jun Wu
- Department of Microorganism Engineering, Beijing Institute of Biotechnology, Beijing 100071, China,Corresponding authors
| | - Bo Liu
- Department of Microorganism Engineering, Beijing Institute of Biotechnology, Beijing 100071, China,Corresponding authors
| |
Collapse
|
14
|
Shin M, Kim K, Lee HJ, Lee R, Jung YJ, Park J, Hahn TW. Zika virus baculovirus-expressed envelope protein elicited humoral and cellular immunity in immunocompetent mice. Sci Rep 2022; 12:660. [PMID: 35027643 PMCID: PMC8758750 DOI: 10.1038/s41598-021-04713-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 09/24/2021] [Indexed: 12/05/2022] Open
Abstract
Zika virus (ZIKV) is a mosquito-borne virus that has a high risk of inducing Guillain-Barré syndrome and microcephaly in newborns. Because vaccination is considered the most effective strategy against ZIKV infection, we designed a recombinant vaccine utilizing the baculovirus expression system with two strains of ZIKV envelope protein (MR766, Env_M; ZBRX6, Env_Z). Animals inoculated with Env_M and Env_Z produced ZIKV-specific antibodies and secreted effector cytokines such as interferon-γ, tumor necrosis factor-α, and interleukin-12. Moreover, the progeny of immunized females had detectable maternal antibodies that protected them against two ZIKV strains (MR766 and PRVABC59) and a Dengue virus strain. We propose that the baculovirus expression system ZIKV envelope protein recombinant provides a safe and effective vaccine strategy.
Collapse
Affiliation(s)
- Minna Shin
- College of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Kiju Kim
- College of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Hyo-Ji Lee
- College of Biological Sciences, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Rangyeon Lee
- College of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Yu-Jin Jung
- College of Biological Sciences, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Jeongho Park
- College of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University, Chuncheon, 24341, Republic of Korea.
| | - Tae-Wook Hahn
- College of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University, Chuncheon, 24341, Republic of Korea.
| |
Collapse
|
15
|
T-cell epitope-based vaccine designing against Orthohantavirus: a causative agent of deadly cardio-pulmonary disease. NETWORK MODELING AND ANALYSIS IN HEALTH INFORMATICS AND BIOINFORMATICS 2021; 11:2. [PMID: 34900515 PMCID: PMC8649322 DOI: 10.1007/s13721-021-00339-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 09/26/2021] [Accepted: 09/28/2021] [Indexed: 12/20/2022]
Abstract
Orthohantavirus, a zoonotic virus responsible for causing human cardio-pulmonary disease, is proven to be a fatal disease. Due to the paucity of regimens to cure the disease and efficient management to eradicate this deadly virus, there is a constant need to expand in-silico approaches belonging to immunology domain to formulate best feasible peptide-based vaccine against it. In lieu of that, we have predicted and validated an epitope of nine-residue-long sequence “MIGLLSSRI”. The predicted epitope has shown best interactions with HLA alleles of MHC Class II proteins, namely HLA DRB1_0101, DRB1_0401, DRB1_0405, DRB1_0701, DRB1_0901, DRB1_1302, and DRB1_1501. The structure of the epitope was modeled by deploying PEPFOLD 3.5 and verified by Ramachandran plot analysis. Molecular docking and simulation studies reveal that this epitope has satisfactory binding scores, ACE value and global energies for docked complexes along with selectable range of RMSD and RMSF values. Also, the predicted epitope “MIGLLSSRI” exhibits population coverage of more than 62% in world population and maximum of 70% in the United States of America. In this intensive study, we have used many tools like AllergenFP, NETMHCII 3.2, VaxiJen, ToxinPred, PEPFOLD 3.5, DINC, IEDB-Population coverage, MHCPred and JCat server. Most of these tools are based on modern innovative statistical algorithms like HMM, ANN, ML, etc. that help in better predictions of putative candidates for vaccine crafting. This innovative methodology is facile, cost-effective and time-efficient, which could facilitate designing of a vaccine against this virus.
Collapse
|
16
|
Núñez-Muñoz L, Marcelino-Pérez G, Calderón-Pérez B, Pérez-Saldívar M, Acosta-Virgen K, González-Conchillos H, Vargas-Hernández B, Olivares-Martínez A, Ruiz-Medrano R, Roa-Velázquez D, Morales-Ríos E, Ramos-Flores J, Torres-Franco G, Peláez-González D, Fernández-Hernández J, Espinosa-Cantellano M, Tapia-Sidas D, Ramírez-Pool JA, Padilla-Viveros A, Xoconostle-Cázares B. Recombinant Antigens Based on Non-Glycosylated Regions from RBD SARS-CoV-2 as Potential Vaccine Candidates against COVID-19. Vaccines (Basel) 2021; 9:928. [PMID: 34452053 PMCID: PMC8402574 DOI: 10.3390/vaccines9080928] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 08/17/2021] [Accepted: 08/18/2021] [Indexed: 01/22/2023] Open
Abstract
The Receptor-Binding Domain (RBD) of the Spike (S) protein from Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) has glycosylation sites which can limit the production of reliable antigens expressed in prokaryotic platforms, due to glycan-mediated evasion of the host immune response. However, protein regions without glycosylated residues capable of inducing neutralizing antibodies could be useful for antigen production in systems that do not carry the glycosylation machinery. To test this hypothesis, the potential antigens NG06 and NG19, located within the non-glycosylated S-RBD region, were selected and expressed in Escherichia coli, purified by FPLC and employed to determine their immunogenic potential through detection of antibodies in serum from immunized rabbits, mice, and COVID-19 patients. IgG antibodies from sera of COVID-19-recovered patients detected the recombinant antigens NG06 and NG19 (A450 nm = 0.80 ± 0.33; 1.13 ± 0.33; and 0.11 ± 0.08 for and negatives controls, respectively). Also, the purified antigens were able to raise polyclonal antibodies in animal models evoking a strong immune response with neutralizing activity in mice model. This research highlights the usefulness of antigens based on the non-N-glycosylated region of RBD from SARS-CoV-2 for candidate vaccine development.
Collapse
Affiliation(s)
- Leandro Núñez-Muñoz
- Department of Biotechnology and Bioengineering, Centro de Investigación y de Estudios Avanzados (CINVESTAV), Av. Instituto Politécnico Nacional 2508, México City 07360, Mexico; (L.N.-M.); (G.M.-P.); (B.C.-P.); (B.V.-H.); (A.O.-M.); (R.R.-M.); (D.T.-S.); (J.A.R.-P.)
| | - Gabriel Marcelino-Pérez
- Department of Biotechnology and Bioengineering, Centro de Investigación y de Estudios Avanzados (CINVESTAV), Av. Instituto Politécnico Nacional 2508, México City 07360, Mexico; (L.N.-M.); (G.M.-P.); (B.C.-P.); (B.V.-H.); (A.O.-M.); (R.R.-M.); (D.T.-S.); (J.A.R.-P.)
- Doctoral Program in Nanosciences and Nanotechnology, Centro de Investigación y de Estudios Avanzados (CINVESTAV), Av. Instituto Politécnico Nacional 2508, México City 07360, Mexico;
| | - Berenice Calderón-Pérez
- Department of Biotechnology and Bioengineering, Centro de Investigación y de Estudios Avanzados (CINVESTAV), Av. Instituto Politécnico Nacional 2508, México City 07360, Mexico; (L.N.-M.); (G.M.-P.); (B.C.-P.); (B.V.-H.); (A.O.-M.); (R.R.-M.); (D.T.-S.); (J.A.R.-P.)
| | - Miriam Pérez-Saldívar
- Department of Infectomics and Molecular Pathogenesis, Centro de Investigación y de Estudios Avanzados (CINVESTAV), Av. Instituto Politécnico Nacional 2508, México City 07360, Mexico; (M.P.-S.); (K.A.-V.); (H.G.-C.); (M.E.-C.)
| | - Karla Acosta-Virgen
- Department of Infectomics and Molecular Pathogenesis, Centro de Investigación y de Estudios Avanzados (CINVESTAV), Av. Instituto Politécnico Nacional 2508, México City 07360, Mexico; (M.P.-S.); (K.A.-V.); (H.G.-C.); (M.E.-C.)
| | - Hugo González-Conchillos
- Department of Infectomics and Molecular Pathogenesis, Centro de Investigación y de Estudios Avanzados (CINVESTAV), Av. Instituto Politécnico Nacional 2508, México City 07360, Mexico; (M.P.-S.); (K.A.-V.); (H.G.-C.); (M.E.-C.)
| | - Brenda Vargas-Hernández
- Department of Biotechnology and Bioengineering, Centro de Investigación y de Estudios Avanzados (CINVESTAV), Av. Instituto Politécnico Nacional 2508, México City 07360, Mexico; (L.N.-M.); (G.M.-P.); (B.C.-P.); (B.V.-H.); (A.O.-M.); (R.R.-M.); (D.T.-S.); (J.A.R.-P.)
| | - Ana Olivares-Martínez
- Department of Biotechnology and Bioengineering, Centro de Investigación y de Estudios Avanzados (CINVESTAV), Av. Instituto Politécnico Nacional 2508, México City 07360, Mexico; (L.N.-M.); (G.M.-P.); (B.C.-P.); (B.V.-H.); (A.O.-M.); (R.R.-M.); (D.T.-S.); (J.A.R.-P.)
| | - Roberto Ruiz-Medrano
- Department of Biotechnology and Bioengineering, Centro de Investigación y de Estudios Avanzados (CINVESTAV), Av. Instituto Politécnico Nacional 2508, México City 07360, Mexico; (L.N.-M.); (G.M.-P.); (B.C.-P.); (B.V.-H.); (A.O.-M.); (R.R.-M.); (D.T.-S.); (J.A.R.-P.)
| | - Daniela Roa-Velázquez
- Doctoral Program in Nanosciences and Nanotechnology, Centro de Investigación y de Estudios Avanzados (CINVESTAV), Av. Instituto Politécnico Nacional 2508, México City 07360, Mexico;
- Department of Biochemistry, Centro de Investigación y de Estudios Avanzados (CINVESTAV), Av. Instituto Politécnico Nacional 2508, México City 07360, Mexico;
| | - Edgar Morales-Ríos
- Department of Biochemistry, Centro de Investigación y de Estudios Avanzados (CINVESTAV), Av. Instituto Politécnico Nacional 2508, México City 07360, Mexico;
| | - Jorge Ramos-Flores
- Laboratory Animal Production and Experimentation Unit, Centro de Investigación y de Estudios Avanzados (CINVESTAV), Av. Instituto Politécnico Nacional 2508, México City 07360, Mexico; (J.R.-F.); (G.T.-F.); (D.P.-G.); (J.F.-H.)
| | - Gustavo Torres-Franco
- Laboratory Animal Production and Experimentation Unit, Centro de Investigación y de Estudios Avanzados (CINVESTAV), Av. Instituto Politécnico Nacional 2508, México City 07360, Mexico; (J.R.-F.); (G.T.-F.); (D.P.-G.); (J.F.-H.)
| | - Diana Peláez-González
- Laboratory Animal Production and Experimentation Unit, Centro de Investigación y de Estudios Avanzados (CINVESTAV), Av. Instituto Politécnico Nacional 2508, México City 07360, Mexico; (J.R.-F.); (G.T.-F.); (D.P.-G.); (J.F.-H.)
| | - Jorge Fernández-Hernández
- Laboratory Animal Production and Experimentation Unit, Centro de Investigación y de Estudios Avanzados (CINVESTAV), Av. Instituto Politécnico Nacional 2508, México City 07360, Mexico; (J.R.-F.); (G.T.-F.); (D.P.-G.); (J.F.-H.)
| | - Martha Espinosa-Cantellano
- Department of Infectomics and Molecular Pathogenesis, Centro de Investigación y de Estudios Avanzados (CINVESTAV), Av. Instituto Politécnico Nacional 2508, México City 07360, Mexico; (M.P.-S.); (K.A.-V.); (H.G.-C.); (M.E.-C.)
| | - Diana Tapia-Sidas
- Department of Biotechnology and Bioengineering, Centro de Investigación y de Estudios Avanzados (CINVESTAV), Av. Instituto Politécnico Nacional 2508, México City 07360, Mexico; (L.N.-M.); (G.M.-P.); (B.C.-P.); (B.V.-H.); (A.O.-M.); (R.R.-M.); (D.T.-S.); (J.A.R.-P.)
| | - José Abrahan Ramírez-Pool
- Department of Biotechnology and Bioengineering, Centro de Investigación y de Estudios Avanzados (CINVESTAV), Av. Instituto Politécnico Nacional 2508, México City 07360, Mexico; (L.N.-M.); (G.M.-P.); (B.C.-P.); (B.V.-H.); (A.O.-M.); (R.R.-M.); (D.T.-S.); (J.A.R.-P.)
| | - América Padilla-Viveros
- Transdisciplinary Doctoral Program in Scientific and Technological Development for Society, Centro de Investigación y de Estudios Avanzados (CINVESTAV), Av. Instituto Politécnico Nacional 2508, México City 07360, Mexico;
| | - Beatriz Xoconostle-Cázares
- Department of Biotechnology and Bioengineering, Centro de Investigación y de Estudios Avanzados (CINVESTAV), Av. Instituto Politécnico Nacional 2508, México City 07360, Mexico; (L.N.-M.); (G.M.-P.); (B.C.-P.); (B.V.-H.); (A.O.-M.); (R.R.-M.); (D.T.-S.); (J.A.R.-P.)
| |
Collapse
|
17
|
Sun J, Zheng Z, Li M, Liu Z, Su X, Jin X. Development of a novel ZIKV vaccine comprised of immunodominant CD4+ and CD8+ T cell epitopes identified through comprehensive epitope mapping in Zika virus infected mice. Vaccine 2021; 39:5173-5186. [PMID: 34353682 DOI: 10.1016/j.vaccine.2021.07.036] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 07/10/2021] [Accepted: 07/13/2021] [Indexed: 11/28/2022]
Abstract
Zika virus (ZIKV) caused over two million human infections in more than 80 countries around 2015-2016. Current vaccines under development are mostly focused on inducing antibodies that despite capable of inhibiting the virus, may have the potential to trigger antibody dependent enhancement (ADE). T cell vaccines that do not induce antibodies targeting viral surface will unlikely cause ADE, but be capable of potentiating the effectiveness of an antibody-inducing vaccine. To develop such a protective T cell vaccine, we first examined the repertoire of antigen-specific T cells in immunocompetent mice that have been transiently infected by ZIKV. Through epitope mapping using 427 overlapping peptides spanning the entire length of ZIKV polyprotein, we discovered 27 immunodominant epitopes scattered throughout the virus on C, E, NS1-NS5 proteins. Among them, 8 were confirmed as CD4+ T cell epitopes, and 16 as CD8+ T cell epitopes, while 3 for both T cell subsets. From these 27 newly identified epitopes, the top 10 epitopes were selected to formulate three T cell vaccines comprised of either CD4+ T cell epitopes, or CD8+ T cell epitopes, or a mixture of both. Immunization with these T cell epitopes induced T cell-mediated cytotoxicity and cytokine production, and conferred varying degrees of protection against ZIKV challenge. Moreover, these new T cell vaccines also improved the protective efficacy of a neutralizing antibody-inducing recombinant E80 protein vaccine. Together, our results provided additional evidence in support of the protective role of ZIKV-specific CD4+ and CD8+ T cells, and laid foundation for future development of T cell vaccines for ZIKV.
Collapse
Affiliation(s)
- Jin Sun
- The Center for Microbes, Development and Health, CAS Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Zhihang Zheng
- Shanghai Public Health Clinical Center, Fudan University, Shanghai 201058, China
| | - Min Li
- Shanghai Public Health Clinical Center, Fudan University, Shanghai 201058, China
| | - Zhihua Liu
- Shanghai Public Health Clinical Center, Fudan University, Shanghai 201058, China
| | - Xiao Su
- CAS Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Xia Jin
- Shanghai Public Health Clinical Center, Fudan University, Shanghai 201058, China.
| |
Collapse
|
18
|
Prior Heterologous Flavivirus Exposure Results in Reduced Pathogenesis in a Mouse Model of Zika Virus Infection. J Virol 2021; 95:e0057321. [PMID: 34076486 PMCID: PMC8312874 DOI: 10.1128/jvi.00573-21] [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] [Indexed: 01/24/2023] Open
Abstract
The 2015/2016 Zika virus epidemic in South and Central America left the scientific community urgently trying to understand the factors that contribute to Zika virus pathogenesis. Because multiple other flaviviruses are endemic in areas where Zika virus emerged, it is hypothesized that a key to understanding Zika virus disease severity is to study Zika virus infection in the context of prior flavivirus exposure. Human and animal studies have highlighted the idea that having been previously exposed to a different flavivirus may modulate the immune response to Zika virus. However, it is still unclear how prior flavivirus exposure impacts Zika viral burden and disease. In this murine study, we longitudinally examine multiple factors involved in Zika disease, linking viral burden with increased neurological disease severity, weight loss, and inflammation. We show that prior heterologous flavivirus exposure with dengue virus type 2 or 3 or the vaccine strain of yellow fever provides protection from mortality in a lethal Zika virus challenge. However, reduction in viral burden and Zika disease varies depending on the infecting primary flavivirus; with primary Zika virus infection being most protective from Zika virus challenge, followed by dengue virus 2, with yellow fever and dengue virus 3 protecting against mortality but showing more severe disease. This study demonstrates the variation in protective effects of prior flavivirus exposure on Zika virus pathogenesis and identifies distinct relationships between primary flavivirus infection and the potential for Zika virus disease. IMPORTANCE The emergence and reemergence of various vector-borne diseases in recent years highlights the need to understand the mechanisms of protection for each pathogen. In this study, we investigated the impact of prior exposure to Zika virus, dengue virus serotypes 2 or 3, or the vaccine strain of yellow fever on pathogenesis and disease outcomes in a mouse model of Zika virus infection. We found that prior exposure to a heterologous flavivirus was protective from mortality, and to varying degrees, prior flavivirus exposure was protective against neurological disease, weight loss, and severe viral burden during a lethal Zika challenge. Using a longitudinal and cross-sectional study design, we were able to link multiple disease parameters, including viral burden, with neurological disease severity, weight loss, and inflammatory response in the context of flavivirus infection. This study demonstrates a measurable but varied impact of prior flavivirus exposure in modulating flavivirus pathophysiology. Given the cyclic nature of most flavivirus outbreaks, this work will contribute to the forecasting of disease severity for future outbreaks.
Collapse
|
19
|
Yang R, Liu Q, Pang W, Gao F, Liang H, Zhang W, Lin Y, Li M, Liu Z, Gao GF, Zhang L, Xiao H, Zheng Y, Huang Z, Jin X. Two immunogenic recombinant protein vaccine candidates showed disparate protective efficacy against Zika virus infection in rhesus macaques. Vaccine 2021; 39:915-925. [PMID: 33451779 DOI: 10.1016/j.vaccine.2020.12.077] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Revised: 12/22/2020] [Accepted: 12/28/2020] [Indexed: 12/27/2022]
Abstract
Zika virus (ZIKV) infection has caused major public health problems recently. To develop subunit vaccines for ZIKV, we have previously constructed recombinant ZIKV envelope protein domain III (EDIII), and the entire ectodomain (E80, which comprises EDI, EDII and EDIII), as vaccine candidates and showed both of them being immunogenic and protective in murine models. In this follow-up study, we compared these vaccine candidates in non-human primates. Both of them elicited neutralizing antibody responses, but only E80 immunization inhibited ZIKV infection in both peripheral blood and monkey tissues, whereas EDIII increased blood ZIKV RNA through possibly antibody-dependent enhancement. Further investigations revealed that the virion-binding antibody response in E80 immunized monkeys persisted longer and stronger than in EDIII immunized monkeys. These results demonstrate that E80 is superior to EDIII as a vaccine candidate, and that the magnitude, quality and durability of virion-binding neutralizing antibodies are correlates of protection.
Collapse
Affiliation(s)
- Ruoheng Yang
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, China; Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
| | - Qingwei Liu
- Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
| | - Wei Pang
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Fei Gao
- Comprehensive AIDS Research Center, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China
| | - Huabin Liang
- Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
| | - Wei Zhang
- Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
| | - Yalong Lin
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Min Li
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, China; Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
| | - Zhihua Liu
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, China; Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
| | - George F Gao
- Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Linqi Zhang
- Comprehensive AIDS Research Center, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China
| | - Hui Xiao
- Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
| | - Yongtang Zheng
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Zhong Huang
- Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China.
| | - Xia Jin
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, China.
| |
Collapse
|
20
|
Zika Virus-Like Particles Bearing a Covalent Dimer of Envelope Protein Protect Mice from Lethal Challenge. J Virol 2020; 95:JVI.01415-20. [PMID: 33028720 PMCID: PMC7737734 DOI: 10.1128/jvi.01415-20] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 10/03/2020] [Indexed: 12/17/2022] Open
Abstract
Zika virus (ZIKV) envelope (E) protein is the major target of neutralizing antibodies in infected hosts and thus represents a candidate of interest for vaccine design. However, a major concern in the development of vaccines against ZIKV and the related dengue virus is the induction of cross-reactive poorly neutralizing antibodies that can cause antibody-dependent enhancement (ADE) of infection. This risk necessitates particular care in vaccine design. Specifically, the engineered immunogens should have their cross-reactive epitopes masked, and they should be optimized for eliciting virus-specific strongly neutralizing antibodies upon vaccination. Here, we developed ZIKV subunit- and virus-like particle (VLP)-based vaccines displaying E in its wild-type form or E locked in a covalently linked dimeric (cvD) conformation to enhance the exposure of E dimers to the immune system. Compared with their wild-type derivatives, cvD immunogens elicited antibodies with a higher capacity to neutralize virus infection in cultured cells. More importantly, these immunogens protected animals from lethal challenge with both the African and Asian lineages of ZIKV, impairing virus dissemination to brain and sexual organs. Moreover, the locked conformation of E reduced the exposure of epitopes recognized by cross-reactive antibodies and therefore showed a lower potential to induce ADE in vitro Our data demonstrated a higher efficacy of the VLPs in comparison with that of the soluble dimer and support VLP-cvD as a promising ZIKV vaccine.IMPORTANCE Infection with Zika virus (ZIKV) leads to the production by the host of antibodies that target the viral surface envelope (E) protein. A subset of these antibodies can inhibit virus infection, thus making E a suitable candidate for the development of vaccine against the virus. However, the anti-ZIKV E antibodies can cross-react with the E protein of the related dengue virus on account of the high level of similarity exhibited by the two viral proteins. Such a scenario may lead to severe dengue disease. Therefore, the design of a ZIKV vaccine requires particular care. Here, we tested two candidate vaccines containing a recombinant form of the ZIKV E protein that is forced in a covalently stable dimeric conformation (cvD). They were generated with an explicit aim to reduce the exposure of the cross-reactive epitopes. One vaccine is composed of a soluble form of the E protein (sE-cvD), the other is a more complex virus-like particle (VLP-cvD). We used the two candidate vaccines to immunize mice and later infected them with ZIKV. The animals produced a high level of inhibitory antibodies and were protected from the infection. The VLP-cvD was the most effective, and we believe it represents a promising ZIKV vaccine candidate.
Collapse
|
21
|
Berneck BS, Rockstroh A, Fertey J, Grunwald T, Ulbert S. A Recombinant Zika Virus Envelope Protein with Mutations in the Conserved Fusion Loop Leads to Reduced Antibody Cross-Reactivity upon Vaccination. Vaccines (Basel) 2020; 8:E603. [PMID: 33066262 PMCID: PMC7712286 DOI: 10.3390/vaccines8040603] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 10/07/2020] [Accepted: 10/09/2020] [Indexed: 12/18/2022] Open
Abstract
Zika virus (ZIKV) is a zoonotic, human pathogenic, and mosquito-borne flavivirus. Its distribution is rapidly growing worldwide. Several attempts to develop vaccines for ZIKV are currently ongoing. Central to most vaccination approaches against flavivirus infections is the envelope (E) protein, which is the major target of neutralizing antibodies. Insect-cell derived, recombinantly expressed variants of E from the flaviviruses West Nile and Dengue virus have entered clinical trials in humans. Also for ZIKV, these antigens are promising vaccine candidates. Due to the structural similarity of flaviviruses, cross-reactive antibodies are induced by flavivirus antigens and have been linked to the phenomenon of antibody-dependent enhancement of infection (ADE). Especially the highly conserved fusion loop domain (FL) in the E protein is a target of such cross-reactive antibodies. In areas where different flaviviruses co-circulate and heterologous infections cannot be ruled out, this is of concern. To exclude the possibility that recombinant E proteins of ZIKV might induce ADE in infections with related flaviviruses, we performed an immunization study with an insect-cell derived E protein containing four mutations in and near the FL. Our data show that this mutant antigen elicits antibodies with equal neutralizing capacity as the wildtype equivalent. However, it induces much less serological cross-reactivity and does not cause ADE in vitro. These results indicate that mutated variants of the E protein might lead to ZIKV and other flavivirus vaccines with increased safety profiles.
Collapse
Affiliation(s)
| | | | | | | | - Sebastian Ulbert
- Fraunhofer-Institute for Cell Therapy and Immunology, 04103 Leipzig, Germany; (B.S.B.); (A.R.); (J.F.); (T.G.)
| |
Collapse
|
22
|
Araujo SC, Pereira LR, Alves RPS, Andreata-Santos R, Kanno AI, Ferreira LCS, Gonçalves VM. Anti-Flavivirus Vaccines: Review of the Present Situation and Perspectives of Subunit Vaccines Produced in Escherichia coli. Vaccines (Basel) 2020; 8:vaccines8030492. [PMID: 32878023 PMCID: PMC7564369 DOI: 10.3390/vaccines8030492] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 08/22/2020] [Accepted: 08/23/2020] [Indexed: 12/14/2022] Open
Abstract
This article aims to review the present status of anti-flavivirus subunit vaccines, both those at the experimental stage and those already available for clinical use. Aspects regarding development of vaccines to Yellow Fever virus, (YFV), Dengue virus (DENV), West Nile virus (WNV), Zika virus (ZIKV), and Japanese encephalitis virus (JEV) are highlighted, with particular emphasis on purified recombinant proteins generated in bacterial cells. Currently licensed anti-flavivirus vaccines are based on inactivated, attenuated, or virus-vector vaccines. However, technological advances in the generation of recombinant antigens with preserved structural and immunological determinants reveal new possibilities for the development of recombinant protein-based vaccine formulations for clinical testing. Furthermore, novel proposals for multi-epitope vaccines and the discovery of new adjuvants and delivery systems that enhance and/or modulate immune responses can pave the way for the development of successful subunit vaccines. Nonetheless, advances in this field require high investments that will probably not raise interest from private pharmaceutical companies and, therefore, will require support by international philanthropic organizations and governments of the countries more severely stricken by these viruses.
Collapse
Affiliation(s)
- Sergio C. Araujo
- Laboratory of Vaccine Development, Instituto Butantan, São Paulo–SP 05503-900, Brazil; (S.C.A.); (A.I.K.)
| | - Lennon R. Pereira
- Laboratory of Vaccine Development, Institute of Biomedical Sciences, Universidade de São Paulo, São Paulo–SP 05508-000, Brazil; (L.R.P.); (R.P.S.A.); (R.A.-S.)
| | - Rubens P. S. Alves
- Laboratory of Vaccine Development, Institute of Biomedical Sciences, Universidade de São Paulo, São Paulo–SP 05508-000, Brazil; (L.R.P.); (R.P.S.A.); (R.A.-S.)
| | - Robert Andreata-Santos
- Laboratory of Vaccine Development, Institute of Biomedical Sciences, Universidade de São Paulo, São Paulo–SP 05508-000, Brazil; (L.R.P.); (R.P.S.A.); (R.A.-S.)
| | - Alex I. Kanno
- Laboratory of Vaccine Development, Instituto Butantan, São Paulo–SP 05503-900, Brazil; (S.C.A.); (A.I.K.)
| | - Luis Carlos S. Ferreira
- Laboratory of Vaccine Development, Institute of Biomedical Sciences, Universidade de São Paulo, São Paulo–SP 05508-000, Brazil; (L.R.P.); (R.P.S.A.); (R.A.-S.)
- Correspondence: (L.C.S.F.); (V.M.G.)
| | - Viviane M. Gonçalves
- Laboratory of Vaccine Development, Instituto Butantan, São Paulo–SP 05503-900, Brazil; (S.C.A.); (A.I.K.)
- Correspondence: (L.C.S.F.); (V.M.G.)
| |
Collapse
|
23
|
Zheng Z, Li M, Liu Z, Jin X, Sun J. Establishment of Murine Infection Models with Biological Clones of Dengue Viruses Derived from a Single Clinical Viral Isolate. Virol Sin 2020; 35:626-636. [PMID: 32451883 PMCID: PMC7246292 DOI: 10.1007/s12250-020-00229-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2019] [Accepted: 03/11/2020] [Indexed: 01/25/2023] Open
Abstract
Dengue virus (DENV) is a single-stranded RNA virus transmitted by mosquitoes in tropical and subtropical regions. It causes dengue fever, dengue hemorrhagic fever and dengue shock syndrome in patients. Each year, 390 million people are estimated to be infected by four serotypes of dengue virus, creating a great burden on global public health and local economy. So far, no antiviral drug is available for dengue disease, and the newly licensed vaccine is far from satisfactory. One large obstacle for dengue vaccine and drug development is the lack of suitable small animal models. Although some DENV infection models have been developed, only a small number of viral strains can infect immunodeficient mice. In this study, with biologically cloned viruses from a single clinical isolate, we have established two mouse models of DENV infection, one is severe lethal infection in immunocompromised mice, and the other resembles self-limited disease manifestations in Balb/c mice with transient blockage of type I IFN responses. This study not only offers new small animal models of dengue viral infection, but also provides new viral variants for further investigations on dengue viral pathogenesis.
Collapse
Affiliation(s)
- Zhihang Zheng
- CAS Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, 200031, Shanghai, China.,Chinese Academy of Sciences, University of Chinese Academy of Sciences, 100049, Beijing, China.,Shanghai Public Health Clinical Center, Fudan University, 201508, Shanghai, China
| | - Min Li
- CAS Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, 200031, Shanghai, China
| | - Zhihua Liu
- CAS Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, 200031, Shanghai, China.,Chinese Academy of Sciences, University of Chinese Academy of Sciences, 100049, Beijing, China.,Shanghai Public Health Clinical Center, Fudan University, 201508, Shanghai, China
| | - Xia Jin
- CAS Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, 200031, Shanghai, China. .,Shanghai Public Health Clinical Center, Fudan University, 201508, Shanghai, China.
| | - Jin Sun
- CAS Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, 200031, Shanghai, China.
| |
Collapse
|
24
|
Amaral MP, Apostolico JDS, Tomita N, Coirada FC, Lunardelli VAS, Fernandes ER, Souza HFS, Astray RM, Boscardin SB, Rosa DS. Homologous prime-boost with Zika virus envelope protein and poly (I:C) induces robust specific humoral and cellular immune responses. Vaccine 2020; 38:3653-3664. [PMID: 32247567 DOI: 10.1016/j.vaccine.2020.03.037] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2019] [Revised: 03/18/2020] [Accepted: 03/19/2020] [Indexed: 01/18/2023]
Abstract
The recent outbreaks of Zika virus (ZIKV) infection and the potential association with Guillain-Barré syndrome in adults and with congenital abnormalities have highlighted the urgency for an effective vaccine. The ZIKV Envelope glycoprotein (EZIKV) is the most abundant protein on the virus surface, and has been evaluated together with the pre-membrane protein (prM) of the viral coat as a vaccine candidate in clinical trials. In this study, we performed a head-to-head comparison of the immune response induced by different EZIKV-based vaccine candidates in mice. We compared different platforms (DNA, recombinant protein), adjuvants (poly (I:C), CpG ODN 1826) and immunization strategies (homologous, heterologous). The hierarchy of adjuvant potency showed that poly (I:C) was a superior adjuvant than CpG ODN. While poly (I:C) assisted immunization reached a plateau in antibody titers after two doses, the CpG ODN group required an extra immunization dose. Besides, the administration of poly (I:C) induced higher EZIKV-specific cellular immune responses than CpG ODN. We also show that immunization with homologous prime-boost EZIKV protein + poly (I:C) regimen induced a more robust humoral response than homologous DNA (pVAX-EZIKV) or heterologous regimens (DNA/protein or protein/DNA). A detailed analysis of cellular immune responses revealed that homologous (EZIKV + poly (I:C)) and heterologous (pVAX-EZIKV/EZIKV + poly (I:C)) prime-boost regimens induced the highest magnitude of IFN-γ secreting cells and cytokine-producing CD4+ T cells. Overall, our data demonstrate that homologous EZIKV + poly (I:C) prime-boost immunization is sufficient to induce more robust specific-EZIKV humoral and cellular immune responses than the other strategies that contemplate homologous DNA (pVAX-EZIKV) or heterologous (pVAX-EZIKV/EZIKV + poly (I:C), and vice-versa) immunizations.
Collapse
Affiliation(s)
- Marcelo Pires Amaral
- Department of Microbiology, Immunology and Parasitology, Federal University of São Paulo (UNIFESP/EPM), São Paulo, Brazil
| | - Juliana de Souza Apostolico
- Department of Microbiology, Immunology and Parasitology, Federal University of São Paulo (UNIFESP/EPM), São Paulo, Brazil; Institute for Investigation in Immunology (iii), INCT, São Paulo, Brazil
| | - Nádia Tomita
- Department of Microbiology, Immunology and Parasitology, Federal University of São Paulo (UNIFESP/EPM), São Paulo, Brazil
| | - Fernanda Caroline Coirada
- Department of Microbiology, Immunology and Parasitology, Federal University of São Paulo (UNIFESP/EPM), São Paulo, Brazil
| | - Victória Alves Santos Lunardelli
- Department of Microbiology, Immunology and Parasitology, Federal University of São Paulo (UNIFESP/EPM), São Paulo, Brazil; Institute for Investigation in Immunology (iii), INCT, São Paulo, Brazil
| | - Edgar Ruz Fernandes
- Department of Microbiology, Immunology and Parasitology, Federal University of São Paulo (UNIFESP/EPM), São Paulo, Brazil
| | - Higo Fernando Santos Souza
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | | | - Silvia Beatriz Boscardin
- Institute for Investigation in Immunology (iii), INCT, São Paulo, Brazil; Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Daniela Santoro Rosa
- Department of Microbiology, Immunology and Parasitology, Federal University of São Paulo (UNIFESP/EPM), São Paulo, Brazil; Institute for Investigation in Immunology (iii), INCT, São Paulo, Brazil.
| |
Collapse
|
25
|
Sun J, Du S, Zheng Z, Cheng G, Jin X. Defeat Dengue and Zika Viruses With a One-Two Punch of Vaccine and Vector Blockade. Front Microbiol 2020; 11:362. [PMID: 32265852 PMCID: PMC7100368 DOI: 10.3389/fmicb.2020.00362] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2019] [Accepted: 02/18/2020] [Indexed: 01/07/2023] Open
Abstract
Dengue virus (DENV) and Zika virus (ZIKV) are two mosquito-borne flaviviruses afflicting nearly half of the world population. Human infection by these viruses can either be asymptomatic or manifest as clinical diseases from mild to severe. Despite more cases are presented as self-limiting febrile illness, severe dengue disease can be manifested as hemorrhagic fever and hemorrhagic shock syndrome, and ZIKV infection has been linked to increased incidence of peripheral neuropathy Guillain-Barre syndrome and central neural disease such as microcephaly. The current prevention and treatment of these infectious diseases are either non-satisfactory or entirely lacking. Because DENV and ZIKV have much similarities in genomic and structural features, almost identical mode of mosquito-mediated transmission, and probably the same pattern of host innate and adaptive immunity toward them, it is reasonable and often desirable to investigate these two viruses side-by-side, and thereby devise common countermeasures against both. Here, we review the existing knowledge on DENV and ZIKV regarding epidemiology, molecular virology, protective immunity and vaccine development, discuss recent new discoveries on the functions of flavivirus NS1 protein in viral pathogenesis and transmission, and propose a one-two punch strategy using vaccine and vector blockade to overcome antibody-dependent enhancement and defeat Dengue and Zika viruses.
Collapse
Affiliation(s)
- Jin Sun
- Viral Disease and Vaccine Translational Research Unit, CAS Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
| | - Senyan Du
- Tsinghua-Peking Center for Life Sciences, School of Medicine, Tsinghua University, Beijing, China
| | - Zhihang Zheng
- Viral Disease and Vaccine Translational Research Unit, CAS Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China,Institut Pasteur of Shanghai, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Gong Cheng
- Tsinghua-Peking Center for Life Sciences, School of Medicine, Tsinghua University, Beijing, China
| | - Xia Jin
- Viral Disease and Vaccine Translational Research Unit, CAS Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China,Shanghai Public Health Clinical Center, Fudan University, Shanghai, China,*Correspondence: Xia Jin, ;
| |
Collapse
|
26
|
Liu R, Ma H, Shu J, Zhang Q, Han M, Liu Z, Jin X, Zhang F, Wu X. Vaccines and Therapeutics Against Hantaviruses. Front Microbiol 2020; 10:2989. [PMID: 32082263 PMCID: PMC7002362 DOI: 10.3389/fmicb.2019.02989] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Accepted: 12/10/2019] [Indexed: 12/16/2022] Open
Abstract
Hantaviruses (HVs) are rodent-transmitted viruses that can cause hantavirus cardiopulmonary syndrome (HCPS) in the Americas and hemorrhagic fever with renal syndrome (HFRS) in Eurasia. Together, these viruses have annually caused approximately 200,000 human infections worldwide in recent years, with a case fatality rate of 5–15% for HFRS and up to 40% for HCPS. There is currently no effective treatment available for either HFRS or HCPS. Only whole virus inactivated vaccines against HTNV or SEOV are licensed for use in the Republic of Korea and China, but the protective efficacies of these vaccines are uncertain. To a large extent, the immune correlates of protection against hantavirus are not known. In this review, we summarized the epidemiology, virology, and pathogenesis of four HFRS-causing viruses, HTNV, SEOV, PUUV, and DOBV, and two HCPS-causing viruses, ANDV and SNV, and then discussed the existing knowledge on vaccines and therapeutics against these diseases. We think that this information will shed light on the rational development of new vaccines and treatments.
Collapse
Affiliation(s)
- Rongrong Liu
- Department of Microbiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Hongwei Ma
- Department of Microbiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Jiayi Shu
- Scientific Research Center, Shanghai Public Health Clinical Center & Institutes of Biomedical Sciences, Key Laboratory of Medical Molecular Virology of Ministry of Education & Health, Shanghai Medical College, Fudan University, Shanghai, China.,Viral Disease and Vaccine Translational Research Unit, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
| | - Qiang Zhang
- School of Biology and Basic Medical Sciences, Soochow University, Suzhou, China
| | - Mingwei Han
- Cadet Brigade, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Ziyu Liu
- Department of Microbiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Xia Jin
- Scientific Research Center, Shanghai Public Health Clinical Center & Institutes of Biomedical Sciences, Key Laboratory of Medical Molecular Virology of Ministry of Education & Health, Shanghai Medical College, Fudan University, Shanghai, China
| | - Fanglin Zhang
- Department of Microbiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Xingan Wu
- Department of Microbiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
27
|
Genetic Diversity of Collaborative Cross Mice Controls Viral Replication, Clinical Severity, and Brain Pathology Induced by Zika Virus Infection, Independently of Oas1b. J Virol 2020; 94:JVI.01034-19. [PMID: 31694939 DOI: 10.1128/jvi.01034-19] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Accepted: 11/03/2019] [Indexed: 12/11/2022] Open
Abstract
The explosive spread of Zika virus (ZIKV) has been associated with major variations in severe disease and congenital afflictions among infected populations, suggesting an influence of host genes. We investigated how genome-wide variants could impact susceptibility to ZIKV infection in mice. We first describe that the susceptibility of Ifnar1-knockout mice is largely influenced by their genetic background. We then show that Collaborative Cross (CC) mice, which exhibit a broad genetic diversity, in which the type I interferon receptor (IFNAR) was blocked by an anti-IFNAR antibody expressed phenotypes ranging from complete resistance to severe symptoms and death, with large variations in the peak and the rate of decrease in the plasma viral load, in the brain viral load, in brain histopathology, and in the viral replication rate in infected cells. The differences in susceptibility to ZIKV between CC strains correlated with the differences in susceptibility to dengue and West Nile viruses between the strains. We identified highly susceptible and resistant mouse strains as new models to investigate the mechanisms of human ZIKV disease and other flavivirus infections. Genetic analyses revealed that phenotypic variations are driven by multiple genes with small effects, reflecting the complexity of ZIKV disease susceptibility in the human population. Notably, our results rule out the possibility of a role of the Oas1b gene in the susceptibility to ZIKV. Altogether, the findings of this study emphasize the role of host genes in the pathogeny of ZIKV infection and lay the foundation for further genetic and mechanistic studies.IMPORTANCE In recent outbreaks, ZIKV has infected millions of people and induced rare but potentially severe complications, including Guillain-Barré syndrome and encephalitis in adults. While several viral sequence variants were proposed to enhance the pathogenicity of ZIKV, the influence of host genetic variants in mediating the clinical heterogeneity remains mostly unexplored. We addressed this question using a mouse panel which models the genetic diversity of the human population and a ZIKV strain from a recent clinical isolate. Through a combination of in vitro and in vivo approaches, we demonstrate that multiple host genetic variants determine viral replication in infected cells and the clinical severity, the kinetics of blood viral load, and brain pathology in mice. We describe new mouse models expressing high degrees of susceptibility or resistance to ZIKV and to other flaviviruses. These models will facilitate the identification and mechanistic characterization of host genes that influence ZIKV pathogenesis.
Collapse
|
28
|
Mwale PF, Lee CH, Lin LT, Leu SJ, Huang YJ, Chiang LC, Mao YC, Yang YY. Expression, Purification, and Characterization of Anti- Zika virus Envelope Protein: Polyclonal and Chicken-Derived Single Chain Variable Fragment Antibodies. Int J Mol Sci 2020; 21:ijms21020492. [PMID: 31940993 PMCID: PMC7014089 DOI: 10.3390/ijms21020492] [Citation(s) in RCA: 10] [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: 12/29/2019] [Accepted: 01/08/2020] [Indexed: 12/14/2022] Open
Abstract
Zika virus (ZIKV) is a new and emerging virus that has caused outbreaks worldwide. The virus has been linked to congenital neurological malformations in neonates and Guillain-Barré syndrome in adults. Currently there are no effective vaccines available. As a result, there is a great need for ZIKV treatment. In this study, we developed single chain variable fragment (scFv) antibodies that target the ZIKV envelope protein using phage display technology. We first induced an immune response in white leghorn laying hens against the ZIKV envelope (E) protein. Chickens were immunized and polyclonal immunoglobulin yolk (IgY) antibodies were extracted from egg yolks. A high-level titer of anti-ZIKV_E IgY antibodies was detected using enzyme-linked immunosorbent assay (ELISA) after the third immunization. The titer persisted for at least 9 weeks. We constructed two antibody libraries that contained 5.3 × 106 and 4.5 × 106 transformants. After biopanning, an ELISA phage assay confirmed the enrichment of specific clones. We randomly selected 26 clones that expressed ZIKV scFv antibodies and classified them into two groups, short-linker and long-linker. Of these, four showed specific binding activities toward ZIKV_E proteins. These data suggest that the polyclonal and monoclonal scFv antibodies have the diagnostic or therapeutic potential for ZIKV.
Collapse
Affiliation(s)
- Pharaoh Fellow Mwale
- Ph.D. Program in Medical Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan; (P.F.M.); (C.-H.L.)
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan;
| | - Chi-Hsin Lee
- Ph.D. Program in Medical Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan; (P.F.M.); (C.-H.L.)
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan;
| | - Liang-Tzung Lin
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; (L.-T.L.); (S.-J.L.)
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Sy-Jye Leu
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; (L.-T.L.); (S.-J.L.)
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Yun-Ju Huang
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan;
| | - Liao-Chun Chiang
- College of Life Sciences, National Tsing Hua University, Hsinchu 30013, Taiwan;
| | - Yan-Chiao Mao
- Division of Clinical Toxicology, Department of Emergency Medicine, Taichung Veterans General Hospital, Taichung 40705, Taiwan;
| | - Yi-Yuan Yang
- Ph.D. Program in Medical Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan; (P.F.M.); (C.-H.L.)
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan;
- Core Laboratory of Antibody Generation and Research, Taipei Medical University, Taipei 11031, Taiwan
- Correspondence: ; Tel.: +886-2-273-616-61 (ext. 3325); Fax: +886-2-273-245-10
| |
Collapse
|
29
|
Maness NJ, Schouest B, Singapuri A, Dennis M, Gilbert MH, Bohm RP, Schiro F, Aye PP, Baker K, Van Rompay KKA, Lackner AA, Bonaldo MC, Blair RV, Permar SR, Coffey LL, Panganiban AT, Magnani D. Postnatal Zika virus infection of nonhuman primate infants born to mothers infected with homologous Brazilian Zika virus. Sci Rep 2019; 9:12802. [PMID: 31488856 PMCID: PMC6728326 DOI: 10.1038/s41598-019-49209-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Accepted: 08/15/2019] [Indexed: 02/07/2023] Open
Abstract
Recent data in a nonhuman primate model showed that infants postnatally infected with Zika virus (ZIKV) were acutely susceptible to high viremia and neurological damage, suggesting the window of vulnerability extends beyond gestation. In this pilot study, we addressed the susceptibility of two infant rhesus macaques born healthy to dams infected with Zika virus during pregnancy. Passively acquired neutralizing antibody titers dropped below detection limits between 2 and 3 months of age, while binding antibodies remained detectable until viral infection at 5 months. Acute serum viremia was comparatively lower than adults infected with the same Brazilian isolate of ZIKV (n = 11 pregnant females, 4 males, and 4 non-pregnant females). Virus was never detected in cerebrospinal fluid nor in neural tissues at necropsy two weeks after infection. However, viral RNA was detected in lymph nodes, confirming some tissue dissemination. Though protection was not absolute and our study lacks an important comparison with postnatally infected infants born to naïve dams, our data suggest infants born healthy to infected mothers may harbor a modest but important level of protection from postnatally acquired ZIKV for several months after birth, an encouraging result given the potentially severe infection outcomes of this population.
Collapse
Affiliation(s)
- Nicholas J Maness
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana, USA.
- Tulane National Primate Research Center, Tulane University, Covington, Louisiana, USA.
| | - Blake Schouest
- Tulane National Primate Research Center, Tulane University, Covington, Louisiana, USA
- Biomedical Sciences Training Program, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Anil Singapuri
- Department of Pathology, Microbiology and Immunology, University of California, Davis, CA, USA
| | - Maria Dennis
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, North Carolina, USA
| | - Margaret H Gilbert
- Tulane National Primate Research Center, Tulane University, Covington, Louisiana, USA
| | - Rudolf P Bohm
- Tulane National Primate Research Center, Tulane University, Covington, Louisiana, USA
| | - Faith Schiro
- Tulane National Primate Research Center, Tulane University, Covington, Louisiana, USA
| | - Pyone P Aye
- Tulane National Primate Research Center, Tulane University, Covington, Louisiana, USA
| | - Kate Baker
- Tulane National Primate Research Center, Tulane University, Covington, Louisiana, USA
| | - Koen K A Van Rompay
- Department of Pathology, Microbiology and Immunology, University of California, Davis, CA, USA
- California National Primate Research Center, University of California, Davis, California, USA
| | - Andrew A Lackner
- Tulane National Primate Research Center, Tulane University, Covington, Louisiana, USA
| | - Myrna C Bonaldo
- Laboratório de Biologia Molecular de Flavivírus, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro, RJ, Brazil
| | - Robert V Blair
- Tulane National Primate Research Center, Tulane University, Covington, Louisiana, USA
| | - Sallie R Permar
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, North Carolina, USA
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, North Carolina, USA
| | - Lark L Coffey
- Department of Pathology, Microbiology and Immunology, University of California, Davis, CA, USA
| | - Antonito T Panganiban
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana, USA
- Tulane National Primate Research Center, Tulane University, Covington, Louisiana, USA
| | - Diogo Magnani
- MassBiologics of the University of Massachusetts Medical School, Boston, Massachusetts, USA
| |
Collapse
|
30
|
Therapeutic Advances Against ZIKV: A Quick Response, a Long Way to Go. Pharmaceuticals (Basel) 2019; 12:ph12030127. [PMID: 31480297 PMCID: PMC6789873 DOI: 10.3390/ph12030127] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 08/15/2019] [Accepted: 08/16/2019] [Indexed: 01/07/2023] Open
Abstract
Zika virus (ZIKV) is a mosquito-borne flavivirus that spread throughout the American continent in 2015 causing considerable worldwide social and health alarm due to its association with ocular lesions and microcephaly in newborns, and Guillain-Barré syndrome (GBS) cases in adults. Nowadays, no licensed vaccines or antivirals are available against ZIKV, and thus, in this very short time, the scientific community has conducted enormous efforts to develop vaccines and antivirals. So that, different platforms (purified inactivated and live attenuated viruses, DNA and RNA nucleic acid based candidates, virus-like particles, subunit elements, and recombinant viruses) have been evaluated as vaccine candidates. Overall, these vaccines have shown the induction of vigorous humoral and cellular responses, the decrease of viremia and viral RNA levels in natural target organs, the prevention of vertical and sexual transmission, as well as that of ZIKV-associated malformations, and the protection of experimental animal models. Some of these vaccine candidates have already been assayed in clinical trials. Likewise, the search for antivirals have also been the focus of recent investigations, with dozens of compounds tested in cell culture and a few in animal models. Both direct acting antivirals (DAAs), directed to viral structural proteins and enzymes, and host acting antivirals (HAAs), directed to cellular factors affecting all steps of the viral life cycle (binding, entry, fusion, transcription, translation, replication, maturation, and egress), have been evaluated. It is expected that this huge collaborative effort will produce affordable and effective therapeutic and prophylactic tools to combat ZIKV and other related still unknown or nowadays neglected flaviviruses. Here, a comprehensive overview of the advances made in the development of therapeutic measures against ZIKV and the questions that still have to be faced are summarized.
Collapse
|
31
|
ZIKV infection induces robust Th1-like Tfh cell and long-term protective antibody responses in immunocompetent mice. Nat Commun 2019; 10:3859. [PMID: 31455769 PMCID: PMC6712032 DOI: 10.1038/s41467-019-11754-0] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 08/01/2019] [Indexed: 01/07/2023] Open
Abstract
Induction of long-lived antibody responses during infection or vaccination is often essential for subsequent protection, but the relative contributions of T follicular helper (Tfh) cells and T helper 1 (Th1) cells for induction of antigen specific antibody responses to viruses are unclear. Here, we establish an acute Zika virus (ZIKV) infection model in immunocompetent mice, and show that ZIKV infection elicits robust Th1-like Tfh cell and protective antibody responses. While these Th1-like Tfh cells share phenotypic and transcriptomic profiles with both Tfh and Th1 cells, they also have unique surface markers and gene expression characteristics, and are dependent on T-bet for their development. Th1-like Tfh cells, but not Th1 cells, are essential for class switching of ZIKV-specific IgG2c antibodies and maintenance of long-term neutralizing antibody responses. Our study suggests that specific modulation of the Th1-like Tfh cell response during infection or vaccination may augment the induction of antiviral antibody response to ZIKV and other viruses.
Collapse
|
32
|
Frumence E, Viranaicken W, Bos S, Alvarez-Martinez MT, Roche M, Arnaud JD, Gadea G, Desprès P. A Chimeric Zika Virus between Viral Strains MR766 and BeH819015 Highlights a Role for E-glycan Loop in Antibody-mediated Virus Neutralization. Vaccines (Basel) 2019; 7:vaccines7020055. [PMID: 31238493 PMCID: PMC6630725 DOI: 10.3390/vaccines7020055] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 06/15/2019] [Accepted: 06/20/2019] [Indexed: 12/25/2022] Open
Abstract
Zika virus (ZIKV) is an emerging mosquito-borne flavivirus which is of major public health concern. ZIKV infection is recognized as the cause of congenital Zika disease and other neurological defects, with no specific prophylactic or therapeutic treatments. As the humoral immune response is an essential component of protective immunity, there is an urgent need for effective vaccines that confer protection against ZIKV infection. In the present study, we evaluate the immunogenicity of chimeric viral clone ZIKBeHMR-2, in which the region encoding the structural proteins of the African strain MR766 backbone was replaced with its counterpart from the epidemic strain BeH819015. Three amino-acid substitutions I152T, T156I, and H158Y were introduced in the glycan loop of the E protein (E-GL) making ZIKBeHMR-2 a non-glycosylated virus. Adult BALB/c mice inoculated intraperitoneally with ZIKBeHMR-2 developed anti-ZIKV antibodies directed against viral proteins E and NS1 and a booster dose increased antibody titers. Immunization with ZIKBeHMR-2 resulted in a rapid production of neutralizing anti-ZIKV antibodies. Antibody-mediated ZIKV neutralization was effective against viral strain MR766, whereas epidemic ZIKV strains were poorly sensitive to neutralization by anti-ZIKBeHMR-2 immune sera. From our data, we propose that the three E-GL residues at positions E-152, E-156, and E-158 greatly influence the accessibility of neutralizing antibody epitopes on ZIKV.
Collapse
Affiliation(s)
- Etienne Frumence
- Université de La Réunion, INSERM U1187, CNRS UMR 9192, IRD UMR 249, Unité Mixte Processus Infectieux en Milieu Insulaire Tropical, Plateforme Technologique CYROI, 97491 Sainte-Clotilde, La Réunion, France.
| | - Wildriss Viranaicken
- Université de La Réunion, INSERM U1187, CNRS UMR 9192, IRD UMR 249, Unité Mixte Processus Infectieux en Milieu Insulaire Tropical, Plateforme Technologique CYROI, 97491 Sainte-Clotilde, La Réunion, France.
| | - Sandra Bos
- Université de La Réunion, INSERM U1187, CNRS UMR 9192, IRD UMR 249, Unité Mixte Processus Infectieux en Milieu Insulaire Tropical, Plateforme Technologique CYROI, 97491 Sainte-Clotilde, La Réunion, France.
| | | | - Marjolaine Roche
- Université de La Réunion, INSERM U1187, CNRS UMR 9192, IRD UMR 249, Unité Mixte Processus Infectieux en Milieu Insulaire Tropical, Plateforme Technologique CYROI, 97491 Sainte-Clotilde, La Réunion, France.
| | | | - Gilles Gadea
- Université de La Réunion, INSERM U1187, CNRS UMR 9192, IRD UMR 249, Unité Mixte Processus Infectieux en Milieu Insulaire Tropical, Plateforme Technologique CYROI, 97491 Sainte-Clotilde, La Réunion, France.
| | - Philippe Desprès
- Université de La Réunion, INSERM U1187, CNRS UMR 9192, IRD UMR 249, Unité Mixte Processus Infectieux en Milieu Insulaire Tropical, Plateforme Technologique CYROI, 97491 Sainte-Clotilde, La Réunion, France.
| |
Collapse
|
33
|
Ghaffar KA, Ng LFP, Renia L. Fast Tracks and Roadblocks for Zika Vaccines. Vaccines (Basel) 2018; 6:vaccines6040077. [PMID: 30469444 PMCID: PMC6313897 DOI: 10.3390/vaccines6040077] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2018] [Revised: 11/16/2018] [Accepted: 11/20/2018] [Indexed: 01/07/2023] Open
Abstract
In early 2014, a relatively obscure virus, the Zika virus, made headlines worldwide following an increase in the number of congenital malformations. Since then, research on Zika virus, treatment and vaccines have progressed swiftly with various drugs being repurposed and vaccines heading into clinical trials. Nonetheless, the need for a vaccine is crucial in order to eradicate this re-emerging arthropod-borne virus which remained silent since its first discovery in 1947. In this review, we focused on how the inconspicuous virus managed to spread, the key immunological factors required for a vaccine and the various vaccine platforms that are currently being studied.
Collapse
Affiliation(s)
- Khairunnisa Abdul Ghaffar
- Singapore Immunology Network, Agency for Science, Technology and Research (A*STAR), Singapore 138648, Singapore.
| | - Lisa F P Ng
- Singapore Immunology Network, Agency for Science, Technology and Research (A*STAR), Singapore 138648, Singapore.
- Institute of Infection and Global Health, University of Liverpool, Liverpool L69 7BE, UK.
| | - Laurent Renia
- Singapore Immunology Network, Agency for Science, Technology and Research (A*STAR), Singapore 138648, Singapore.
| |
Collapse
|
34
|
das Neves Almeida R, Racine T, Magalhães KG, Kobinger GP. Zika Virus Vaccines: Challenges and Perspectives. Vaccines (Basel) 2018; 6:vaccines6030062. [PMID: 30217027 PMCID: PMC6161012 DOI: 10.3390/vaccines6030062] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 09/08/2018] [Accepted: 09/11/2018] [Indexed: 01/07/2023] Open
Abstract
Zika virus is an arbovirus that has rapidly spread within the Americas since 2014, presenting a variety of clinical manifestations and neurological complications resulting in congenital malformation, microcephaly, and possibly, in male infertility. These significant clinical manifestations have led investigators to develop several candidate vaccines specific to Zika virus. In this review we describe relevant targets for the development of vaccines specific for Zika virus, the development status of various vaccine candidates and their different platforms, as well as their clinical progression.
Collapse
Affiliation(s)
| | - Trina Racine
- Centre de Recherche en Infectiologie du CHU de Québec-Université Laval, Québec, QC G1V 4G2, Canada.
- Department of Medical Microbiology, University of Manitoba, Winnipeg, MB R3E 0J9, Canada.
| | - Kelly G Magalhães
- Laboratory of Immunology and Inflammation, University of Brasilia, Brasilia 70910-900, Brazil.
| | - Gary P Kobinger
- Centre de Recherche en Infectiologie du CHU de Québec-Université Laval, Québec, QC G1V 4G2, Canada.
- Department of Medical Microbiology, University of Manitoba, Winnipeg, MB R3E 0J9, Canada.
- Department of Immunology, University of Manitoba, Winnipeg, MB R3E 0T5, Canada.
- Départment de Microbiologie-Infectiologie et D'immunologie, Université Laval, Québc, QC G1V 0A6, Canada.
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA 19104-4238, USA.
| |
Collapse
|
35
|
Emanuel J, Callison J, Dowd KA, Pierson TC, Feldmann H, Marzi A. A VSV-based Zika virus vaccine protects mice from lethal challenge. Sci Rep 2018; 8:11043. [PMID: 30038228 PMCID: PMC6056530 DOI: 10.1038/s41598-018-29401-x] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Accepted: 07/05/2018] [Indexed: 01/25/2023] Open
Abstract
Infection with Zika virus (ZIKV) is commonly mild in humans but has been associated with alarming negative health outcomes including Guillain-Barré syndrome in adults and microcephaly in fetuses. As such, developing a vaccine for ZIKV is a global public health priority. Recombinant vesicular stomatitis virus (VSV) expressing the Ebola virus (EBOV) glycoprotein (GP) has been successfully used as a vaccine platform in the past. In this study, two novel VSV-ZIKV vaccines were generated utilizing the favorable immune targeting of the existing VSV-EBOV vector. In addition to the EBOV GP, these new vaccines express the full-length pre-membrane and envelope proteins or pre-membrane and truncated soluble envelope proteins as antigens. Efficacy testing of both of the VSV vectors against ZIKV was conducted in IFNAR−/− mice and resulted in uniform protection when a single dose was administered 28 days prior to lethal challenge. Furthermore, this vaccine is fast-acting and can uniformly protect mice from lethal disease when administered as late as 3 days prior to ZIKV challenge. Thus, VSV-ZIKV vectors are promising vaccine candidates and should move forward along the licensure pathway.
Collapse
Affiliation(s)
- Jackson Emanuel
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Julie Callison
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Kimberly A Dowd
- Laboratory of Viral Diseases, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Theodore C Pierson
- Laboratory of Viral Diseases, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Heinz Feldmann
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Andrea Marzi
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA.
| |
Collapse
|
36
|
Beaver JT, Lelutiu N, Habib R, Skountzou I. Evolution of Two Major Zika Virus Lineages: Implications for Pathology, Immune Response, and Vaccine Development. Front Immunol 2018; 9:1640. [PMID: 30072993 PMCID: PMC6058022 DOI: 10.3389/fimmu.2018.01640] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 07/03/2018] [Indexed: 12/13/2022] Open
Abstract
Zika virus (ZIKV) became a public health emergency of global concern in 2015 due to its rapid expansion from French Polynesia to Brazil, spreading quickly throughout the Americas. Its unexpected correlation to neurological impairments and defects, now known as congenital Zika syndrome, brought on an urgency to characterize the pathology and develop safe, effective vaccines. ZIKV genetic analyses have identified two major lineages, Asian and African, which have undergone substantial changes during the past 50 years. Although ZIKV infections have been circulating throughout Africa and Asia for the later part of the 20th century, the symptoms were mild and not associated with serious pathology until now. ZIKV evolution also took the form of novel modes of transmission, including maternal-fetal transmission, sexual transmission, and transmission through the eye. The African and Asian lineages have demonstrated differential pathogenesis and molecular responses in vitro and in vivo. The limited number of human infections prior to the 21st century restricted ZIKV research to in vitro studies, but current animal studies utilize mice deficient in type I interferon (IFN) signaling in order to invoke enhanced viral pathogenesis. This review examines ZIKV strain differences from an evolutionary perspective, discussing how these differentially impact pathogenesis via host immune responses that modulate IFN signaling, and how these differential effects dictate the future of ZIKV vaccine candidates.
Collapse
Affiliation(s)
| | | | | | - Ioanna Skountzou
- Department of Microbiology and Immunology, Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, United States
| |
Collapse
|