1
|
Lara-Ramírez D, Santacruz-Tinoco CE, Ramón-Gallegos E, Muñoz-Medina JE. In silico design of Ebola virus Glycoprotein antigenic peptides as vaccine candidates. PLoS One 2025; 20:e0319496. [PMID: 40153397 PMCID: PMC11952221 DOI: 10.1371/journal.pone.0319496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 02/03/2025] [Indexed: 03/30/2025] Open
Abstract
Ebola virus (EBOV) is a filovirus that causes severe hemorrhagic fever and has a fatality rate between 50 and 90%. The vaccines were developed against the Ebola Zaire species; therefore, it is necessary to develop vaccines against other species to control future outbreaks. The objective of this work was to obtain vaccine candidate peptides against different EBOV species through the use of bioinformatics programs and servers that allow glycoprotein (GP) to be analyzed. GP sequences of various EBOV species that did not present gaps or unspecified amino acids or that were repeated (same year, region and laboratory) were downloaded from the NCBI database. A consensus sequence was generated and used to determine vaccine candidate peptides, which were evaluated, through a combination of servers and molecular dynamics, for their ability to interact with B and T lymphocytes, toxicity, allergenicity, solvent exposure, glycosylation, antigenicity, and presence in mature GP. Five vaccine candidate peptides were identified, of which PEP4 had the best characteristics evaluated in this study. PEP4 may be a potential candidate for the development of an EBOV vaccine.
Collapse
Affiliation(s)
- David Lara-Ramírez
- Environmental Cytopathology Laboratory, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico CityMexico
- División de Laboratorios Especializados. Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | | | - Eva Ramón-Gallegos
- Environmental Cytopathology Laboratory, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico CityMexico
| | | |
Collapse
|
2
|
Michita RT, Tran LB, Bark SJ, Kumar D, Toner SA, Jose J, Mysorekar IU, Narayanan A. Zika virus NS1 drives tunneling nanotube formation for mitochondrial transfer and stealth transmission in trophoblasts. Nat Commun 2025; 16:1803. [PMID: 39979240 PMCID: PMC11842757 DOI: 10.1038/s41467-025-56927-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 02/05/2025] [Indexed: 02/22/2025] Open
Abstract
Zika virus (ZIKV) is unique among orthoflaviviruses in its vertical transmission capacity in humans, yet the underlying mechanisms remain incompletely understood. Here, we show that ZIKV induces tunneling nanotubes (TNTs) in placental trophoblasts which facilitate transfer of viral particles, proteins, mitochondria, and RNA to neighboring uninfected cells. TNT formation is driven exclusively via ZIKV non-structural protein 1 (NS1). Specifically, the N-terminal 1-50 amino acids of membrane-bound ZIKV NS1 are necessary for triggering TNT formation in host cells. Trophoblasts infected with TNT-deficient ZIKVΔTNT mutant virus elicited a robust antiviral IFN-λ 1/2/3 response relative to WT ZIKV, suggesting TNT-mediated trafficking allows ZIKV cell-to-cell transmission camouflaged from host defenses. Using affinity purification-mass spectrometry of cells expressing wild-type NS1 or non-TNT forming NS1, we found mitochondrial proteins are dominant NS1-interacting partners. We demonstrate that ZIKV infection or NS1 expression induces elevated mitochondria levels in trophoblasts and that mitochondria are siphoned via TNTs from healthy to ZIKV-infected cells. Together our findings identify a stealth mechanism that ZIKV employs for intercellular spread among placental trophoblasts, evasion of antiviral interferon response, and the hijacking of mitochondria to augment its propagation and survival and offers a basis for novel therapeutic developments targeting these interactions to limit ZIKV dissemination.
Collapse
Affiliation(s)
- Rafael T Michita
- Department of Medicine, Section of Infectious Diseases, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Long B Tran
- Department of Medicine, Section of Infectious Diseases, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Steven J Bark
- Department of Medicine, Section of Infectious Diseases, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Deepak Kumar
- Department of Medicine, Section of Infectious Diseases, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Shay A Toner
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, Pennsylvania, PA, 16802, USA
| | - Joyce Jose
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, Pennsylvania, PA, 16802, USA
- The Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, Pennsylvania, PA, 16802, USA
| | - Indira U Mysorekar
- Department of Medicine, Section of Infectious Diseases, Baylor College of Medicine, Houston, TX, 77030, USA.
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, 77030, USA.
- Huffington Centre on Aging, Baylor College of Medicine, Houston, TX, 77030, USA.
| | - Anoop Narayanan
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, Pennsylvania, PA, 16802, USA.
| |
Collapse
|
3
|
Bello MB, Alsaadi A, Naeem A, Almahboub SA, Bosaeed M, Aljedani SS. Development of nucleic acid-based vaccines against dengue and other mosquito-borne flaviviruses: the past, present, and future. Front Immunol 2025; 15:1475886. [PMID: 39840044 PMCID: PMC11747009 DOI: 10.3389/fimmu.2024.1475886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Accepted: 12/06/2024] [Indexed: 01/23/2025] Open
Abstract
Due to their widespread geographic distribution and frequent outbreaks, mosquito-borne flaviviruses, such as DENV (DENV), Zika virus (ZIKV), Japanese encephalitis virus (JEV), yellow fever virus (YFV), and West Nile virus (WNV), are considered significant global public health threats and contribute to dramatic socioeconomic imbalances worldwide. The global prevalence of these viruses is largely driven by extensive international travels and ecological disruptions that create favorable conditions for the breeding of Aedes and Culex species, the mosquito vectors responsible for the spread of these pathogens. Currently, vaccines are available for only DENV, YFV, and JEV, but these face several challenges, including safety concerns, lengthy production processes, and logistical difficulties in distribution, especially in resource-limited regions, highlighting the urgent need for innovative vaccine approaches. Nucleic acid-based platforms, including DNA and mRNA vaccines, have emerged as promising alternatives due to their ability to elicit strong immune responses, facilitate rapid development, and support scalable manufacturing. This review provides a comprehensive update on the progress of DNA and mRNA vaccine development against mosquito-borne flaviviruses, detailing early efforts and current strategies that have produced candidates with remarkable protective efficacy and strong immunogenicity in preclinical models. Furthermore, we explore future directions for advancing nucleic acid vaccine candidates, which hold transformative potential for enhancing global public health.
Collapse
Affiliation(s)
- Muhammad Bashir Bello
- Infectious Disease Research Department, King Abdullah International Medical Research Center, King Saud bin Abdulaziz University of Health Sciences, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| | - Ahlam Alsaadi
- Infectious Disease Research Department, King Abdullah International Medical Research Center, King Saud bin Abdulaziz University of Health Sciences, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| | - Asif Naeem
- Infectious Disease Research Department, King Abdullah International Medical Research Center, King Saud bin Abdulaziz University of Health Sciences, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| | - Sarah A. Almahboub
- Infectious Disease Research Department, King Abdullah International Medical Research Center, King Saud bin Abdulaziz University of Health Sciences, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| | - Mohammad Bosaeed
- Infectious Disease Research Department, King Abdullah International Medical Research Center, King Saud bin Abdulaziz University of Health Sciences, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
- Department of Medicine, King Abdulaziz Medical City, Riyadh, Saudi Arabia
| | - Safia S. Aljedani
- Infectious Disease Research Department, King Abdullah International Medical Research Center, King Saud bin Abdulaziz University of Health Sciences, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| |
Collapse
|
4
|
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
|
5
|
Wahaab A, Mustafa BE, Hameed M, Batool H, Tran Nguyen Minh H, Tawaab A, Shoaib A, Wei J, Rasgon JL. An Overview of Zika Virus and Zika Virus Induced Neuropathies. Int J Mol Sci 2024; 26:47. [PMID: 39795906 PMCID: PMC11719530 DOI: 10.3390/ijms26010047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Revised: 12/19/2024] [Accepted: 12/21/2024] [Indexed: 01/13/2025] Open
Abstract
Flaviviruses pose a major public health concern across the globe. Among them, Zika virus (ZIKV) is an emerging and reemerging arthropod-borne flavivirus that has become a major international public health problem following multiple large outbreaks over the past two decades. The majority of infections caused by ZIKV exhibit mild symptoms. However, the virus has been found to be associated with a variety of congenital neural abnormalities, including microcephaly in children and Guillain-Barre syndrome in adults. The exact prediction of the potential of ZIKV transmission is still enigmatic and underlines the significance of routine detection of the virus in suspected areas. ZIKV transmission from mother to fetus (including fetal abnormalities), viral presence in immune-privileged areas, and sexual transmission demonstrate the challenges in understanding the factors governing viral persistence and pathogenesis. This review illustrates the transmission patterns, epidemiology, control strategies (through vaccines, antivirals, and vectors), oncolytic aspects, molecular insights into neuro-immunopathogenesis, and other neuropathies caused by ZIKV. Additionally, we summarize in vivo and in vitro models that could provide an important platform to study ZIKV pathogenesis and the underlying governing cellular and molecular mechanisms.
Collapse
Affiliation(s)
- Abdul Wahaab
- Department of Entomology, Pennsylvania State University, University Park, PA 16802, USA; (A.W.); (H.T.N.M.)
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA 16802, USA
- The Center for Infectious Disease Dynamics, Pennsylvania State University, University Park, PA 16802, USA
- The Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA 16802, USA
| | - Bahar E Mustafa
- School of Veterinary Science, Faculty of Science, The University of Melbourne, Melbourne, VIC 3030, Australia;
- Sub Campus Toba Tek Singh, University of Agriculture, Faisalabad 36050, Pakistan;
| | - Muddassar Hameed
- Department of Biomedical Sciences and Pathobiology, VA-MD Regional College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA 24060, USA;
- Center for Zoonotic and Arthropod-Borne Pathogens, Virginia Polytechnic Institute and State University, Blacksburg, VA 24060, USA
- Department of Otolaryngology-Head and Neck Surgery, Department of Pathology and Immunology, Alvin J. Siteman Cancer Center, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Hira Batool
- Chughtai Lab, Head Office, 7-Jail Road, Main Gulberg, Lahore 54000, Pakistan;
| | - Hieu Tran Nguyen Minh
- Department of Entomology, Pennsylvania State University, University Park, PA 16802, USA; (A.W.); (H.T.N.M.)
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA 16802, USA
- The Center for Infectious Disease Dynamics, Pennsylvania State University, University Park, PA 16802, USA
- The Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA 16802, USA
| | - Abdul Tawaab
- Sub Campus Toba Tek Singh, University of Agriculture, Faisalabad 36050, Pakistan;
| | - Anam Shoaib
- School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX 75080, USA;
| | - Jianchao Wei
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China;
| | - Jason L. Rasgon
- Department of Entomology, Pennsylvania State University, University Park, PA 16802, USA; (A.W.); (H.T.N.M.)
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA 16802, USA
- The Center for Infectious Disease Dynamics, Pennsylvania State University, University Park, PA 16802, USA
- The Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA 16802, USA
| |
Collapse
|
6
|
Liu T, Li M, Tian Y, Dong Y, Liu N, Wang Z, Zhang H, Zheng A, Cui C. Immunogenicity and safety of a self-assembling ZIKV nanoparticle vaccine in mice. Int J Pharm 2024; 660:124320. [PMID: 38866086 DOI: 10.1016/j.ijpharm.2024.124320] [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: 02/14/2024] [Revised: 05/07/2024] [Accepted: 06/06/2024] [Indexed: 06/14/2024]
Abstract
Zika virus (ZIKV) is a mosquito-borne flavivirus that highly susceptibly causes Guillain-Barré syndrome and microcephaly in newborns. Vaccination is one of the most effective measures for preventing infectious diseases. However, there is currently no approved vaccine to prevent ZIKV infection. Here, we developed nanoparticle (NP) vaccines by covalently conjugating self-assembled 24-subunit ferritin to the envelope structural protein subunit of ZIKV to achieve antigen polyaggregation. The immunogenicityof the NP vaccine was evaluated in mice. Compared to monomer vaccines, the NP vaccine achieved effective antigen presentation, promoted the differentiation of follicular T helper cells in lymph nodes, and induced significantly greater antigen-specific humoral and cellular immune responses. Moreover, the NP vaccine enhanced high-affinity antigen-specific IgG antibody levels, increased secretion of the cytokines IL-4 and IFN-γ by splenocytes, significantly activated T/B lymphocytes, and improved the generation of memory T/B cells. In addition, no significant adverse reactions occurred when NP vaccine was combined with adjuvants. Overall, ferritin-based NP vaccines are safe and effective ZIKV vaccine candidates.
Collapse
Affiliation(s)
- Ting Liu
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Capital Medical University, Beijing 100069, China; Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Beijing 100069, China; Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing 100069, China; Beijing Laboratory of Biomedical Materials, Beijing 100069, China
| | - Meng Li
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Yang Tian
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Capital Medical University, Beijing 100069, China; Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Beijing 100069, China; Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing 100069, China; Beijing Laboratory of Biomedical Materials, Beijing 100069, China
| | - Yuhan Dong
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Nan Liu
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Zengming Wang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Hui Zhang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Aiping Zheng
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Chunying Cui
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Capital Medical University, Beijing 100069, China; Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Beijing 100069, China; Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing 100069, China; Beijing Laboratory of Biomedical Materials, Beijing 100069, China.
| |
Collapse
|
7
|
Zhang Y, Chamblee M, Xu J, Qu P, Shamseldin MM, Yoo SJ, Misny J, Thongpan I, Kc M, Hall JM, Gupta YA, Evans JP, Lu M, Ye C, Hsu CC, Liang X, Martinez-Sobrido L, Yount JS, Boyaka PN, Liu SL, Dubey P, Peeples ME, Li J. Three SARS-CoV-2 spike protein variants delivered intranasally by measles and mumps vaccines are broadly protective. Nat Commun 2024; 15:5589. [PMID: 38961063 PMCID: PMC11222507 DOI: 10.1038/s41467-024-49443-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 05/29/2024] [Indexed: 07/05/2024] Open
Abstract
As the new SARS-CoV-2 Omicron variants and subvariants emerge, there is an urgency to develop intranasal, broadly protective vaccines. Here, we developed highly efficacious, intranasal trivalent SARS-CoV-2 vaccine candidates (TVC) based on three components of the MMR vaccine: measles virus (MeV), mumps virus (MuV) Jeryl Lynn (JL1) strain, and MuV JL2 strain. Specifically, MeV, MuV-JL1, and MuV-JL2 vaccine strains, each expressing prefusion spike (preS-6P) from a different variant of concern (VoC), were combined to generate TVCs. Intranasal immunization of IFNAR1-/- mice and female hamsters with TVCs generated high levels of S-specific serum IgG antibodies, broad neutralizing antibodies, and mucosal IgA antibodies as well as tissue-resident memory T cells in the lungs. The immunized female hamsters were protected from challenge with SARS-CoV-2 original WA1, B.1.617.2, and B.1.1.529 strains. The preexisting MeV and MuV immunity does not significantly interfere with the efficacy of TVC. Thus, the trivalent platform is a promising next-generation SARS-CoV-2 vaccine candidate.
Collapse
Affiliation(s)
- Yuexiu Zhang
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, USA
| | - Michelle Chamblee
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, USA
| | - Jiayu Xu
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, USA
| | - Panke Qu
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, USA
| | - Mohamed M Shamseldin
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH, USA
- Department of Microbiology, The Ohio State University, Columbus, OH, USA
- Department of Microbiology and Immunology, Faculty of Pharmacy, Helwan University, Ain Helwan, Helwan, Egypt
| | - Sung J Yoo
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, USA
| | - Jack Misny
- Center for Vaccines and Immunity, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Ilada Thongpan
- Center for Vaccines and Immunity, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Mahesh Kc
- Center for Vaccines and Immunity, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Jesse M Hall
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Yash A Gupta
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - John P Evans
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, USA
| | - Mijia Lu
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, USA
| | - Chengjin Ye
- Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Cheng Chih Hsu
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, USA
| | - Xueya Liang
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, USA
| | | | - Jacob S Yount
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH, USA
- Infectious Disease Institute, The Ohio State University, Columbus, OH, USA
| | - Prosper N Boyaka
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, USA
- Infectious Disease Institute, The Ohio State University, Columbus, OH, USA
| | - Shan-Lu Liu
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, USA
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH, USA
- Infectious Disease Institute, The Ohio State University, Columbus, OH, USA
- Center for Retrovirus Research, The Ohio State University, Columbus, OH, USA
| | - Purnima Dubey
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH, USA
- Infectious Disease Institute, The Ohio State University, Columbus, OH, USA
| | - Mark E Peeples
- Center for Vaccines and Immunity, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
- Infectious Disease Institute, The Ohio State University, Columbus, OH, USA
- Department of Pediatrics, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Jianrong Li
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, USA.
- Infectious Disease Institute, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
8
|
Kubinski M, Beicht J, Gerlach T, Aregay A, Osterhaus ADME, Tscherne A, Sutter G, Prajeeth CK, Rimmelzwaan GF. Immunity to Tick-Borne Encephalitis Virus NS3 Protein Induced with a Recombinant Modified Vaccinia Virus Ankara Fails to Afford Mice Protection against TBEV Infection. Vaccines (Basel) 2024; 12:105. [PMID: 38276677 PMCID: PMC10819467 DOI: 10.3390/vaccines12010105] [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: 11/29/2023] [Revised: 01/16/2024] [Accepted: 01/17/2024] [Indexed: 01/27/2024] Open
Abstract
Tick-borne encephalitis (TBE) is a serious neurological disease caused by TBE virus (TBEV). Because antiviral treatment options are not available, vaccination is the key prophylactic measure against TBEV infections. Despite the availability of effective vaccines, cases of vaccination breakthrough infections have been reported. The multienzymatic non-structural protein 3 (NS3) of orthoflaviviruses plays an important role in polyprotein processing and virus replication. In the present study, we evaluated NS3 of TBEV as a potential vaccine target for the induction of protective immunity. To this end, a recombinant modified vaccinia virus Ankara that drives the expression of the TBEV NS3 gene (MVA-NS3) was constructed. MVA-NS3 was used to immunize C57BL/6 mice. It induced NS3-specific immune responses, in particular T cell responses, especially against the helicase domain of NS3. However, MVA-NS3-immunized mice were not protected from subsequent challenge infection with a lethal dose of the TBEV strain Neudoerfl, indicating that in contrast to immunity to prME and NS1, NS3-specific immunity is not an independent correlate of protection against TBEV in this mouse model.
Collapse
Affiliation(s)
- Mareike Kubinski
- Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine Hannover, Foundation, Bünteweg 17, 30559 Hannover, Germany; (M.K.); (J.B.); (T.G.); (A.A.); (A.D.M.E.O.); (C.K.P.)
| | - Jana Beicht
- Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine Hannover, Foundation, Bünteweg 17, 30559 Hannover, Germany; (M.K.); (J.B.); (T.G.); (A.A.); (A.D.M.E.O.); (C.K.P.)
| | - Thomas Gerlach
- Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine Hannover, Foundation, Bünteweg 17, 30559 Hannover, Germany; (M.K.); (J.B.); (T.G.); (A.A.); (A.D.M.E.O.); (C.K.P.)
| | - Amare Aregay
- Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine Hannover, Foundation, Bünteweg 17, 30559 Hannover, Germany; (M.K.); (J.B.); (T.G.); (A.A.); (A.D.M.E.O.); (C.K.P.)
| | - Albert D. M. E. Osterhaus
- Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine Hannover, Foundation, Bünteweg 17, 30559 Hannover, Germany; (M.K.); (J.B.); (T.G.); (A.A.); (A.D.M.E.O.); (C.K.P.)
| | - Alina Tscherne
- Division of Virology, Institute for Infectious Diseases and Zoonoses, Ludwig Maximilian University Munich, Sonnenstraße 24, 85764 Oberschleißheim, Germany; (A.T.)
- German Center for Infection Research (DZIF), Partner Site Munich, 80802 Munich, Germany
| | - Gerd Sutter
- Division of Virology, Institute for Infectious Diseases and Zoonoses, Ludwig Maximilian University Munich, Sonnenstraße 24, 85764 Oberschleißheim, Germany; (A.T.)
- German Center for Infection Research (DZIF), Partner Site Munich, 80802 Munich, Germany
| | - Chittappen Kandiyil Prajeeth
- Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine Hannover, Foundation, Bünteweg 17, 30559 Hannover, Germany; (M.K.); (J.B.); (T.G.); (A.A.); (A.D.M.E.O.); (C.K.P.)
| | - Guus F. Rimmelzwaan
- Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine Hannover, Foundation, Bünteweg 17, 30559 Hannover, Germany; (M.K.); (J.B.); (T.G.); (A.A.); (A.D.M.E.O.); (C.K.P.)
| |
Collapse
|
9
|
Mysorekar I, Michita R, Tran L, Bark S, Kumar D, Toner S, Jose J, Narayanan A. Zika Virus NS1 Drives Tunneling Nanotube Formation for Mitochondrial Transfer, Enhanced Survival, Interferon Evasion, and Stealth Transmission in Trophoblasts. RESEARCH SQUARE 2023:rs.3.rs-3674059. [PMID: 38106210 PMCID: PMC10723532 DOI: 10.21203/rs.3.rs-3674059/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
Zika virus (ZIKV) infection continues to pose a significant public health concern due to limited available preventive measures and treatments. ZIKV is unique among flaviviruses in its vertical transmission capacity (i.e., transmission from mother to fetus) yet the underlying mechanisms remain incompletely understood. Here, we show that both African and Asian lineages of ZIKV induce tunneling nanotubes (TNTs) in placental trophoblasts and multiple other mammalian cell types. Amongst investigated flaviviruses, only ZIKV strains trigger TNTs. We show that ZIKV-induced TNTs facilitate transfer of viral particles, proteins, and RNA to neighboring uninfected cells. ZIKV TNT formation is driven exclusively via its non-structural protein 1 (NS1); specifically, the N-terminal region (50 aa) of membrane-bound NS1 is necessary and sufficient for triggering TNT formation in host cells. Using affinity purification-mass spectrometry of cells infected with wild-type NS1 or non-TNT forming NS1 (pNS1ΔTNT) proteins, we found mitochondrial proteins are dominant NS1-interacting partners, consistent with the elevated mitochondrial mass we observed in infected trophoblasts. We demonstrate that mitochondria are siphoned via TNTs from healthy to ZIKV-infected cells, both homotypically and heterotypically, and inhibition of mitochondrial respiration reduced viral replication in trophoblast cells. Finally, ZIKV strains lacking TNT capabilities due to mutant NS1 elicited a robust antiviral IFN-λ 1/2/3 response, indicating ZIKV's TNT-mediated trafficking also allows ZIKV cell-cell transmission that is camouflaged from host defenses. Together, our findings identify a new stealth mechanism that ZIKV employs for intercellular spread among placental trophoblasts, evasion of antiviral interferon response, and the hijacking of mitochondria to augment its propagation and survival. Discerning the mechanisms of ZIKV intercellular strategies offers a basis for novel therapeutic developments targeting these interactions to limit its dissemination.
Collapse
|
10
|
Kuhn RJ, Barrett ADT, Desilva AM, Harris E, Kramer LD, Montgomery RR, Pierson TC, Sette A, Diamond MS. A Prototype-Pathogen Approach for the Development of Flavivirus Countermeasures. J Infect Dis 2023; 228:S398-S413. [PMID: 37849402 PMCID: PMC10582523 DOI: 10.1093/infdis/jiad193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 05/28/2023] [Indexed: 10/19/2023] Open
Abstract
Flaviviruses are a genus within the Flaviviridae family of positive-strand RNA viruses and are transmitted principally through mosquito and tick vectors. These viruses are responsible for hundreds of millions of human infections worldwide per year that result in a range of illnesses from self-limiting febrile syndromes to severe neurotropic and viscerotropic diseases and, in some cases, death. A vaccine against the prototype flavivirus, yellow fever virus, has been deployed for 85 years and is highly effective. While vaccines against some medically important flaviviruses are available, others have proven challenging to develop. The emergence and spread of flaviviruses, including dengue virus and Zika virus, demonstrate their pandemic potential. This review highlights the gaps in knowledge that need to be addressed to allow for the rapid development of vaccines against emerging flaviviruses in the future.
Collapse
Affiliation(s)
- Richard J Kuhn
- Department of Biological Sciences, Purdue University, West Lafayette, Indiana, USA
- Purdue Institute of Inflammation, Immunology, and Infectious Disease, Purdue University, West Lafayette, Indiana, USA
| | - Alan D T Barrett
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, USA
- Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, Texas, USA
| | - Aravinda M Desilva
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Eva Harris
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California Berkeley, Berkeley, California, USA
| | - Laura D Kramer
- School of Public Health, State University of New York at Albany, Albany, New York, USA
| | - Ruth R Montgomery
- Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Theodore C Pierson
- Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Alessandro Sette
- Division of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, California, USA
- Department of Medicine, University of California in San Diego, San Diego, California, USA
| | - Michael S Diamond
- Department of Medicine, Washington University School of Medicine, St Louis, Missouri, USA
- Department of Molecular Microbiology and Pathology and Immunology, Washington University School of Medicine, St Louis, Missouri, USA
| |
Collapse
|
11
|
Tang J, Fu M, Xu C, Xue B, Zhou A, Chen S, Zhao H, Zhou Y, Chen J, Yang Q, Chen X. Development of a novel virus-like particle-based vaccine for preventing tick-borne encephalitis virus infection. Virol Sin 2023; 38:767-777. [PMID: 37328107 PMCID: PMC10590693 DOI: 10.1016/j.virs.2023.06.003] [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: 03/12/2023] [Accepted: 06/12/2023] [Indexed: 06/18/2023] Open
Abstract
Tick-borne encephalitis virus (TBEV) is an important tick-borne pathogen that poses as a serious public health concern. The coverage and immunogenicity of the currently available vaccines against TBEV are relatively low; therefore, it is crucial to develop novel and effective vaccines against TBEV. The present study describes a novel strategy for the assembly of virus-like particles (VLPs) by co-expressing the structural (core/prM/E) and non-structural (NS2B/NS3Pro) proteins of TBEV. The efficacy of the VLPs was subsequently evaluated in C57BL/6 mice, and the resultant IgG serum could neutralize both Far-Eastern and European subtypes of TBEV. These findings indicated that the VLP-based vaccine elicited the production of cross-subtype reactive antibodies. The VLPs provided protection to mice lacking the type I interferon receptor (IFNAR-/-) against lethal TBEV challenge, with undetectable viral load in brain and intestinal tissues. Furthermore, the group that received the VLP vaccine did not exhibit significant pathological changes and the inflammatory factors were significantly suppressed compared to the control group. Immunization with the VLP vaccine induced the production of multiple-cytokine-producing antiviral CD4+ T cells in vivo, including TNF-α+, IL-2+, and IFN-γ+ T cells. Altogether, the findings suggest that noninfectious VLPs can serve as a potentially safe and effective vaccine candidate against diverse subtypes of TBEV.
Collapse
Affiliation(s)
- Jielin Tang
- Guangzhou National Laboratory, Guangzhou, 510005, China; State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Muqing Fu
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Chonghui Xu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Bao Xue
- Guangzhou National Laboratory, Guangzhou, 510005, China; State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Anqi Zhou
- GMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Sijie Chen
- GMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - He Zhao
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Yuan Zhou
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Jizheng Chen
- Guangzhou National Laboratory, Guangzhou, 510005, China; State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China; State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, 511436, China
| | - Qi Yang
- Guangzhou National Laboratory, Guangzhou, 510005, China; State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China.
| | - Xinwen Chen
- Guangzhou National Laboratory, Guangzhou, 510005, China; State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China; State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, 511436, China.
| |
Collapse
|
12
|
Gupta Y, Baranwal M, Chudasama B. Immunoinformatics-Based Identification of the Conserved Immunogenic Peptides Targeting of Zika Virus Precursor Membrane Protein. Viral Immunol 2023; 36:503-519. [PMID: 37486711 DOI: 10.1089/vim.2023.0015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/25/2023] Open
Abstract
Zika virus infections lead to neurological complications such as congenital Zika syndrome and Guillain-Barré syndrome. Rising Zika infections in newborns and adults have triggered the need for vaccine development. In the current study, the precursor membrane (prM) protein of the Zika virus is explored for its functional importance and design of epitopes enriched conserved peptides with the usage of different immunoinformatics approach. Phylogenetic and mutational analyses inferred that the prM protein is highly conserved. Three conserved peptides containing multiple T and B cell epitopes were designed by employing different epitope prediction algorithms. IEDB population coverage analysis of selected peptides in six different continents has shown the population coverage of 60-99.8% (class I HLA) and 80-100% (class II HLA). Molecular docking of selected peptides/epitopes was carried out with each of class I and II HLA alleles using HADDOCK. A majority of peptide-HLA complex (pHLA) have HADDOCK scores found to be comparable and more than native-HLA complex representing the good binding interaction of peptides to HLA. Molecular dynamics simulation with best docked pHLA complexes revealed that pHLA complexes are stable with RMSD <5.5Å. Current work highlights the importance of prM as a strong antigenic protein and selected peptides have the potential to elicit humoral and cell-mediated immune responses.
Collapse
Affiliation(s)
- Yogita Gupta
- Department of Biotechnology, Thapar Institute of Engineering and Technology, Patiala, India
| | - Manoj Baranwal
- Department of Biotechnology, Thapar Institute of Engineering and Technology, Patiala, India
| | - Bhupendra Chudasama
- School of Physics & Materials Science, Thapar Institute of Engineering and Technology, Patiala, India
| |
Collapse
|
13
|
Wang S, Liang B, Wang W, Li L, Feng N, Zhao Y, Wang T, Yan F, Yang S, Xia X. Viral vectored vaccines: design, development, preventive and therapeutic applications in human diseases. Signal Transduct Target Ther 2023; 8:149. [PMID: 37029123 PMCID: PMC10081433 DOI: 10.1038/s41392-023-01408-5] [Citation(s) in RCA: 81] [Impact Index Per Article: 40.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 03/06/2023] [Accepted: 03/14/2023] [Indexed: 04/09/2023] Open
Abstract
Human diseases, particularly infectious diseases and cancers, pose unprecedented challenges to public health security and the global economy. The development and distribution of novel prophylactic and therapeutic vaccines are the prioritized countermeasures of human disease. Among all vaccine platforms, viral vector vaccines offer distinguished advantages and represent prominent choices for pathogens that have hampered control efforts based on conventional vaccine approaches. Currently, viral vector vaccines remain one of the best strategies for induction of robust humoral and cellular immunity against human diseases. Numerous viruses of different families and origins, including vesicular stomatitis virus, rabies virus, parainfluenza virus, measles virus, Newcastle disease virus, influenza virus, adenovirus and poxvirus, are deemed to be prominent viral vectors that differ in structural characteristics, design strategy, antigen presentation capability, immunogenicity and protective efficacy. This review summarized the overall profile of the design strategies, progress in advance and steps taken to address barriers to the deployment of these viral vector vaccines, simultaneously highlighting their potential for mucosal delivery, therapeutic application in cancer as well as other key aspects concerning the rational application of these viral vector vaccines. Appropriate and accurate technological advances in viral vector vaccines would consolidate their position as a leading approach to accelerate breakthroughs in novel vaccines and facilitate a rapid response to public health emergencies.
Collapse
Affiliation(s)
- Shen Wang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Bo Liang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Weiqi Wang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
- College of Veterinary Medicine, Jilin University, Changchun, China
| | - Ling Li
- China National Research Center for Exotic Animal Diseases, China Animal Health and Epidemiology Center, Qingdao, China
| | - Na Feng
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Yongkun Zhao
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Tiecheng Wang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Feihu Yan
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China.
| | - Songtao Yang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China.
| | - Xianzhu Xia
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China.
| |
Collapse
|
14
|
Wu B, Qi Z, Qian X. Recent Advancements in Mosquito-Borne Flavivirus Vaccine Development. Viruses 2023; 15:813. [PMID: 37112794 PMCID: PMC10143207 DOI: 10.3390/v15040813] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 03/21/2023] [Accepted: 03/21/2023] [Indexed: 04/29/2023] Open
Abstract
Lately, the global incidence of flavivirus infection has been increasing dramatically and presents formidable challenges for public health systems around the world. Most clinically significant flaviviruses are mosquito-borne, such as the four serotypes of dengue virus, Zika virus, West Nile virus, Japanese encephalitis virus and yellow fever virus. Until now, no effective antiflaviviral drugs are available to fight flaviviral infection; thus, a highly immunogenic vaccine would be the most effective weapon to control the diseases. In recent years, flavivirus vaccine research has made major breakthroughs with several vaccine candidates showing encouraging results in preclinical and clinical trials. This review summarizes the current advancement, safety, efficacy, advantages and disadvantages of vaccines against mosquito-borne flaviviruses posing significant threats to human health.
Collapse
Affiliation(s)
| | - Zhongtian Qi
- Department of Microbiology, Faculty of Naval Medicine, Naval Medical University, Shanghai 200433, China;
| | - Xijing Qian
- Department of Microbiology, Faculty of Naval Medicine, Naval Medical University, Shanghai 200433, China;
| |
Collapse
|
15
|
Poveda Cuevas SA, Barroso da Silva FL, Etchebest C. NS1 from Two Zika Virus Strains Differently Interact with a Membrane: Insights to Understand Their Differential Virulence. J Chem Inf Model 2023; 63:1386-1400. [PMID: 36780300 DOI: 10.1021/acs.jcim.2c01461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/14/2023]
Abstract
Zika virus (ZIKV) from Uganda (UG) expresses a phenotype related to fetal loss, whereas the variant from Brazil (BR) induces microcephaly in neonates. The differential virulence has a direct relation to biomolecular mechanisms that make one strain more aggressive than the other. The nonstructural protein 1 (NS1) is a key viral toxin to comprehend these viral discrepancies because of its versatility in many processes of the virus life cycle. Here, we aim to examine through coarse-grained models and molecular dynamics simulations the protein-membrane interactions for both NS1ZIKV-UG and NS1ZIKV-BR dimers. A first evaluation allowed us to establish that the NS1 proteins, in the membrane presence, explore new conformational spaces when compared to systems simulated without a lipid bilayer. These events derive from both differential coupling patterns and discrepant binding affinities to the membrane. The N-terminal domain, intertwined loop, and greasy finger proposed previously as binding membrane regions were also computationally confirmed by us. The anchoring sites have aromatic and ionizable residues that manage the assembly of NS1 toward the membrane, especially for the Ugandan variant. Furthermore, in the presence of the membrane, the difference in the dynamic cross-correlation of residues between the two strains is particularly pronounced in the putative epitope regions. This suggests that the protein-membrane interaction induces changes in the distal part related to putative epitopes. Taken together, these results open up new strategies for the treatment of flaviviruses that would specifically target these dynamic differences.
Collapse
Affiliation(s)
- Sergio Alejandro Poveda Cuevas
- Programa Interunidades em Bioinformática, Universidade de São Paulo, Rua do Matão, 1010, São Paulo, São Paulo BR-05508-090, Brazil.,Departamento de Ciências Biomoleculares, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Av. do Café, s/no-Campus da USP, Ribeirão Preto, São Paulo BR-14040-903, Brazil.,Goethe University Frankfurt, Institute of Biochemistry II, Theodor-Stern-Kai 7, Frankfurt am Main, Hesse DE-60590, Germany.,Faculdade de Ciências Farmacêuticas de Ribeirão Preto, University of São Paulo and Université de Paris International Laboratory in Structural Bioinformatics, Av. do Café, s/no-Campus da USP, Bloco B, Ribeirão Preto, São Paulo BR-14040-903, Brazil
| | - Fernando L Barroso da Silva
- Programa Interunidades em Bioinformática, Universidade de São Paulo, Rua do Matão, 1010, São Paulo, São Paulo BR-05508-090, Brazil.,Departamento de Ciências Biomoleculares, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Av. do Café, s/no-Campus da USP, Ribeirão Preto, São Paulo BR-14040-903, Brazil.,Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, North Carolina 27695, United States.,Faculdade de Ciências Farmacêuticas de Ribeirão Preto, University of São Paulo and Université de Paris International Laboratory in Structural Bioinformatics, Av. do Café, s/no-Campus da USP, Bloco B, Ribeirão Preto, São Paulo BR-14040-903, Brazil
| | - Catherine Etchebest
- Université Paris Cité and Université des Antilles, INSERM, Biologie Intégrée du Globule Rouge, F-75015 Paris, France.,Faculdade de Ciências Farmacêuticas de Ribeirão Preto, University of São Paulo and Université de Paris International Laboratory in Structural Bioinformatics, Av. do Café, s/no-Campus da USP, Bloco B, Ribeirão Preto, São Paulo BR-14040-903, Brazil
| |
Collapse
|
16
|
Dahiya N, Yadav M, Singh H, Jakhar R, Sehrawat N. ZIKV: Epidemiology, infection mechanism and current therapeutics. FRONTIERS IN TROPICAL DISEASES 2023. [DOI: 10.3389/fitd.2022.1059283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
The Zika virus (ZIKV) is a vector-borne flavivirus that has been detected in 87 countries worldwide. Outbreaks of ZIKV infection have been reported from various places around the world and the disease has been declared a public health emergency of international concern. ZIKV has two modes of transmission: vector and non-vector. The ability of ZIKV to vertically transmit in its competent vectors, such as Aedes aegypti and Aedes albopictus, helps it to cope with adverse conditions, and this could be the reason for the major outbreaks that occur from time to time. ZIKV outbreaks are a global threat and, therefore, there is a need for safe and effective drugs and vaccines to fight the virus. In more than 80% of cases, ZIKV infection is asymptomatic and leads to complications, such as microcephaly in newborns and Guillain–Barré syndrome (GBS) in adults. Drugs such as sofosbuvir, chloroquine, and suramin have been found to be effective against ZIKV infections, but further evaluation of their safety in pregnant women is needed. Although temoporfin can be given to pregnant women, it needs to be tested further for side effects. Many vaccine types based on protein, vector, DNA, and mRNA have been formulated. Some vaccines, such as mRNA-1325 and VRC-ZKADNA090-00-VP, have reached Phase II clinical trials. Some new techniques should be used for formulating and testing the efficacy of vaccines. Although there have been no recent outbreaks of ZIKV infection, several studies have shown continuous circulation of ZIKV in mosquito vectors, and there is a risk of re-emergence of ZIKV in the near future. Therefore, vaccines and drugs for ZIKV should be tested further, and safe and effective therapeutic techniques should be licensed for use during outbreaks.
Collapse
|
17
|
Salampe M, Mamada SS, Evary YM, Mitra S, Bin Emran T, Harapan H, Nainu F, Simal-Gandara J. Promising Marine Natural Products for Tackling Viral Outbreaks: A Focus on Possible Targets and Structure-activity Relationship. Curr Top Med Chem 2023; 23:1352-1379. [PMID: 36045529 DOI: 10.2174/1568026622666220831114838] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 05/11/2022] [Accepted: 05/26/2022] [Indexed: 11/22/2022]
Abstract
Recently, people worldwide have experienced several outbreaks caused by viruses that have attracted much interest globally, such as HIV, Zika, Ebola, and the one being faced, SARSCoV- 2 viruses. Unfortunately, the availability of drugs giving satisfying outcomes in curing those diseases is limited. Therefore, it is necessary to dig deeper to provide compounds that can tackle the causative viruses. Meanwhile, the efforts to explore marine natural products have been gaining great interest as the products have consistently shown several promising biological activities, including antiviral activity. This review summarizes some products extracted from marine organisms, such as seaweeds, seagrasses, sponges, and marine bacteria, reported in recent years to have potential antiviral activities tested through several methods. The mechanisms by which those compounds exert their antiviral effects are also described here, with several main mechanisms closely associated with the ability of the products to block the entry of the viruses into the host cells, inhibiting replication or transcription of the viral genetic material, and disturbing the assembly of viral components. In addition, the structure-activity relationship of the compounds is also highlighted by focusing on six groups of marine compounds, namely sulfated polysaccharides, phlorotannins, terpenoids, lectins, alkaloids, and flavonoids. In conclusion, due to their uniqueness compared to substances extracted from terrestrial sources, marine organisms provide abundant products having promising activities as antiviral agents that can be explored to tackle virus-caused outbreaks.
Collapse
Affiliation(s)
| | - Sukamto Salang Mamada
- Department of Pharmacy, Faculty of Pharmacy, Hasanuddin University, Makassar, 90245, Indonesia
| | - Yayu Mulsiani Evary
- Department of Pharmaceutical Science and Technology, Faculty of Pharmacy, Hasanuddin University, Makassar, 90245, Indonesia
| | - Saikat Mitra
- Department of Pharmacy, Faculty of Pharmacy, University of Dhaka, Dhaka, 1000, Bangladesh
| | - Talha Bin Emran
- Department of Pharmacy, BGC Trust University Bangladesh, Chittagong, 4381, Bangladesh
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka ,1207, Bangladesh
| | - Harapan Harapan
- Department of Microbiology, School of Medicine, Universitas Syiah Kuala, Banda Aceh, 23111, Indonesia
| | - Firzan Nainu
- Department of Pharmacy, Faculty of Pharmacy, Hasanuddin University, Makassar, 90245, Indonesia
| | - Jesus Simal-Gandara
- Nutrition and Bromatology Group, Department of Analytical and Food Chemistry, Faculty of Food Science and Technology, University of Vigo, Ourense Campus, E32004 Ourense, Spain
| |
Collapse
|
18
|
Evaluation of Zika virus DNA vaccines based on NS1 and domain III of E. Int Immunopharmacol 2022; 113:109308. [DOI: 10.1016/j.intimp.2022.109308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 09/30/2022] [Accepted: 09/30/2022] [Indexed: 11/05/2022]
|
19
|
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
|
20
|
Lu M, Chamblee M, Zhang Y, Ye C, Dravid P, Park JG, Mahesh KC, Trivedi S, Murthy S, Sharma H, Cassady C, Chaiwatpongsakorn S, Liang X, Yount JS, Boyaka PN, Peeples ME, Martinez-Sobrido L, Kapoor A, Li J. SARS-CoV-2 prefusion spike protein stabilized by six rather than two prolines is more potent for inducing antibodies that neutralize viral variants of concern. Proc Natl Acad Sci U S A 2022; 119:e2110105119. [PMID: 35994646 PMCID: PMC9436349 DOI: 10.1073/pnas.2110105119] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 07/11/2022] [Indexed: 11/18/2022] Open
Abstract
The spike (S) protein of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the main target for neutralizing antibodies (NAbs). The S protein trimer is anchored in the virion membrane in its prefusion (preS) but metastable form. The preS protein has been stabilized by introducing two or six proline substitutions, to generate stabilized, soluble 2P or HexaPro (6P) preS proteins. Currently, it is not known which form is the most immunogenic. Here, we generated recombinant vesicular stomatitis virus (rVSV) expressing preS-2P, preS-HexaPro, and native full-length S, and compared their immunogenicity in mice and hamsters. The rVSV-preS-HexaPro produced and secreted significantly more preS protein compared to rVSV-preS-2P. Importantly, rVSV-preS-HexaPro triggered significantly more preS-specific serum IgG antibody than rVSV-preS-2P in both mice and hamsters. Antibodies induced by preS-HexaPro neutralized the B.1.1.7, B.1.351, P.1, B.1.427, and B.1.617.2 variants approximately two to four times better than those induced by preS-2P. Furthermore, preS-HexaPro induced a more robust Th1-biased cellular immune response than preS-2P. A single dose (104 pfu) immunization with rVSV-preS-HexaPro and rVSV-preS-2P provided complete protection against challenge with mouse-adapted SARS-CoV-2 and B.1.617.2 variant, whereas rVSV-S only conferred partial protection. When the immunization dose was lowered to 103 pfu, rVSV-preS-HexaPro induced two- to sixfold higher antibody responses than rVSV-preS-2P in hamsters. In addition, rVSV-preS-HexaPro conferred 70% protection against lung infection whereas only 30% protection was observed in the rVSV-preS-2P. Collectively, our data demonstrate that both preS-2P and preS-HexaPro are highly efficacious but preS-HexaPro is more immunogenic and protective, highlighting the advantages of using preS-HexaPro in the next generation of SARS-CoV-2 vaccines.
Collapse
Affiliation(s)
- Mijia Lu
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, 43210
| | - Michelle Chamblee
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, 43210
| | - Yuexiu Zhang
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, 43210
| | - Chengjin Ye
- Texas Biomedical Research Institute, San Antonio, TX, 78227
| | - Piyush Dravid
- Center for Vaccines and Immunity, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH, 43205
| | - Jun-Gyu Park
- Texas Biomedical Research Institute, San Antonio, TX, 78227
| | - KC Mahesh
- Center for Vaccines and Immunity, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH, 43205
| | - Sheetal Trivedi
- Center for Vaccines and Immunity, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH, 43205
| | - Satyapramod Murthy
- Center for Vaccines and Immunity, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH, 43205
| | - Himanshu Sharma
- Center for Vaccines and Immunity, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH, 43205
| | - Cole Cassady
- Center for Vaccines and Immunity, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH, 43205
| | - Supranee Chaiwatpongsakorn
- Center for Vaccines and Immunity, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH, 43205
| | - Xueya Liang
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, 43210
| | - Jacob S. Yount
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH, 43210
- Infectious Disease Institute, The Ohio State University, Columbus, OH, 43210
| | - Prosper N. Boyaka
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, 43210
- Infectious Disease Institute, The Ohio State University, Columbus, OH, 43210
| | - Mark E. Peeples
- Center for Vaccines and Immunity, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH, 43205
- Infectious Disease Institute, The Ohio State University, Columbus, OH, 43210
- Department of Pediatrics, College of Medicine, The Ohio State University, Columbus, OH, 43210
| | | | - Amit Kapoor
- Center for Vaccines and Immunity, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH, 43205
- Infectious Disease Institute, The Ohio State University, Columbus, OH, 43210
- Department of Pediatrics, College of Medicine, The Ohio State University, Columbus, OH, 43210
| | - Jianrong Li
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, 43210
- Infectious Disease Institute, The Ohio State University, Columbus, OH, 43210
| |
Collapse
|
21
|
Poveda-Cuevas SA, Etchebest C, da Silva FLB. Self-association features of NS1 proteins from different flaviviruses. Virus Res 2022; 318:198838. [PMID: 35662566 DOI: 10.1016/j.virusres.2022.198838] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 05/26/2022] [Accepted: 05/27/2022] [Indexed: 12/22/2022]
Abstract
Flaviviruses comprise a large group of arboviral species that are distributed in several countries of the tropics, neotropics, and some temperate zones. Since they can produce neurological pathologies or vascular damage, there has been intense research seeking better diagnosis and treatments for their infections in the last decades. The flavivirus NS1 protein is a relevant clinical target because it is involved in viral replication, immune evasion, and virulence. Being a key factor in endothelial and tissue-specific modulation, NS1 has been largely studied to understand the molecular mechanisms exploited by the virus to reprogram host cells. A central part of the viral maturation processes is the NS1 oligomerization because many stages rely on these protein-protein assemblies. In the present study, the self-associations of NS1 proteins from Zika, Dengue, and West Nile viruses are examined through constant-pH coarse-grained biophysical simulations. Free energies of interactions were estimated for different oligomeric states and pH conditions. Our results show that these proteins can form both dimers and tetramers under conditions near physiological pH even without the presence of lipids. Moreover, pH plays an important role mainly controlling the regimes where van der Waals interactions govern their association. Finally, despite the similarity at the sequence level, we found that each flavivirus has a well-characteristic protein-protein interaction profile. These specific features can provide new hints for the development of binders both for better diagnostic tools and the formulation of new therapeutic drugs.
Collapse
Affiliation(s)
- Sergio A Poveda-Cuevas
- Universidade de São Paulo, Programa Interunidades em Bioinformática, Rua do Matão, 1010, BR-05508-090 São Paulo, São Paulo, Brazil; Universidade de São Paulo, Departamento de Ciências Biomoleculares, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Av. do Café, s/no-Campus da USP, BR-14040-903 Ribeirão Preto, São Paulo, Brazil; University of São Paulo and Université de Paris International Laboratory in Structural Bioinformatics, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Av. do Café, s/no-Campus da USP, Bloco B, BR-14040-903 Ribeirão Preto, São Paulo, Brazil.; Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, NC 27695, United States
| | - Catherine Etchebest
- Université Paris Cité, Biologie Intégrée du Globule Rouge, Equipe 2, INSERM, F-75015 Paris, France; University of São Paulo and Université de Paris International Laboratory in Structural Bioinformatics, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Av. do Café, s/no-Campus da USP, Bloco B, BR-14040-903 Ribeirão Preto, São Paulo, Brazil
| | - Fernando L Barroso da Silva
- Universidade de São Paulo, Programa Interunidades em Bioinformática, Rua do Matão, 1010, BR-05508-090 São Paulo, São Paulo, Brazil; Universidade de São Paulo, Departamento de Ciências Biomoleculares, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Av. do Café, s/no-Campus da USP, BR-14040-903 Ribeirão Preto, São Paulo, Brazil; University of São Paulo and Université de Paris International Laboratory in Structural Bioinformatics, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Av. do Café, s/no-Campus da USP, Bloco B, BR-14040-903 Ribeirão Preto, São Paulo, Brazil..
| |
Collapse
|
22
|
Rong H, Qi M, Pan J, Sun Y, Gao J, Zhang X, Li W, Zhang B, Zhang XE, Cui Z. Self-Assembling Nanovaccine Confers Complete Protection Against Zika Virus Without Causing Antibody-Dependent Enhancement. Front Immunol 2022; 13:905431. [PMID: 35615356 PMCID: PMC9124840 DOI: 10.3389/fimmu.2022.905431] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Accepted: 04/14/2022] [Indexed: 01/20/2023] Open
Abstract
The Zika virus (ZIKV) epidemic poses a substantial threat to the public, and the development of safe and effective vaccines is a demanding challenge. In this study, we constructed a kind of self-assembling nanovaccine which confers complete protection against ZIKV infection. The ZIKV envelop protein domain III (zEDIII) was presented on recombinant human heavy chain ferritin (rHF) to form the zEDIII-rHF nanoparticle. Immunization of mice with zEDIII-rHF nanoparticle in the absence of an adjuvant induced robust humoral and cellular immune responses. zEDIII-rHF vaccination conferred complete protection against lethal infection with ZIKV and eliminated pathological symptoms in the brain. Importantly, the zEDIII-rHF nanovaccine induced immune response did not cross-react with dengue virus-2, overcoming the antibody-dependent enhancement (ADE) problem that is a safety concern for ZIKV vaccine development. Our constructed zEDIII-rHF nanovaccine, with superior protective performance and avoidance of ADE, provides an effective and safe vaccine candidate against ZIKV.
Collapse
Affiliation(s)
- Heng Rong
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Mi Qi
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Jingdi Pan
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yuhan Sun
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Jiawang Gao
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xiaowei Zhang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China
| | - Wei Li
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China
| | - Bo Zhang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China
| | - Xian-En Zhang
- Faculty of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Zongqiang Cui
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, China
- University of Chinese Academy of Sciences, Beijing, China
- *Correspondence: Zongqiang Cui,
| |
Collapse
|
23
|
Measles-based Zika vaccine induces long-term immunity and requires NS1 antibodies to protect the female reproductive tract. NPJ Vaccines 2022; 7:43. [PMID: 35440656 PMCID: PMC9018676 DOI: 10.1038/s41541-022-00464-2] [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: 11/10/2021] [Accepted: 02/21/2022] [Indexed: 01/09/2023] Open
Abstract
Zika virus (ZIKV) can cause devastating effects in the unborn fetus of pregnant women. To develop a candidate vaccine that can protect human fetuses, we generated a panel of live measles vaccine (MV) vectors expressing ZIKV-E and -NS1. Our MV-based ZIKV-E vaccine, MV-E2, protected mice from the non-lethal Zika Asian strain (PRVABC59) and the lethal African strain (MR766) challenge. Despite 100% survival of the MV-E2 mice, however, complete viral clearance was not achieved in the brain and reproductive tract of the lethally challenged mice. We then tested MV-based vaccines that expressed E and NS1 together or separately in two different vaccines. We observed complete clearance of ZIKV from the female reproductive tract and complete fetal protection in the lethal African challenge model in animals that received the dual antigen vaccines. Additionally, MV-E2 and MV-NS1, when administered together, induced durable plasma cell responses. Our findings suggest that NS1 antibodies are required to enhance the protection of ZIKV-E antibodies in the female reproductive tract.
Collapse
|
24
|
Nazerai L, Buus S, Stryhn A, Thomsen AR, Christensen JP. Efficient Control of Zika Virus Infection Induced by a Non-Replicating Adenovector Encoding Zika Virus NS1/NS2 Antigens Fused to the MHC Class II-Associated Invariant Chain. Viruses 2021; 13:2215. [PMID: 34835021 PMCID: PMC8625593 DOI: 10.3390/v13112215] [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: 09/22/2021] [Revised: 11/01/2021] [Accepted: 11/01/2021] [Indexed: 11/16/2022] Open
Abstract
It is generally believed that a successful Zika virus (ZIKV) vaccine should induce neutralizing antibodies against the ZIKV envelope (E) protein to efficiently halt viral infection. However, E-specific neutralizing antibodies have been implicated in a phenomenon called antibody-dependent enhancement, which represents an ongoing concern in the flavivirus-vaccinology field. In this report, we investigated the vaccination potential of replication-deficient adenoviral vectors encoding the ZIKV non-structural proteins 1 and 2 (NS1/NS2) and employed the strategy of linking the antigens to the MHC-II associated invariant chain (li) to improve immunogenicity and by inference, the level of protection. We demonstrated that li-linkage enhanced the production of anti-NS1 antibodies and induced an accelerated and prolonged polyfunctional CD8 T cell response in mice, which ultimately resulted in a high degree of protection against ZIKV infection of the CNS.
Collapse
Affiliation(s)
| | | | | | | | - Jan Pravsgaard Christensen
- Department of Immunology and Microbiology, University of Copenhagen, 2200 Copenhagen, Denmark; (L.N.); (S.B.); (A.S.); (A.R.T.)
| |
Collapse
|
25
|
Lu M, Zhang Y, Dravid P, Li A, Zeng C, KC M, Trivedi S, Sharma H, Chaiwatpongsakorn S, Zani A, Kenney A, Cai C, Ye C, Liang X, Qiu J, Martinez-Sobrido L, Yount JS, Boyaka PN, Liu SL, Peeples ME, Kapoor A, Li J. A Methyltransferase-Defective Vesicular Stomatitis Virus-Based SARS-CoV-2 Vaccine Candidate Provides Complete Protection against SARS-CoV-2 Infection in Hamsters. J Virol 2021; 95:e0059221. [PMID: 34379509 PMCID: PMC8475528 DOI: 10.1128/jvi.00592-21] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 08/02/2021] [Indexed: 01/11/2023] Open
Abstract
The current pandemic of coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has led to dramatic economic and health burdens. Although the worldwide SARS-CoV-2 vaccination campaign has begun, exploration of other vaccine candidates is needed due to uncertainties with the current approved vaccines, such as durability of protection, cross-protection against variant strains, and costs of long-term production and storage. In this study, we developed a methyltransferase-defective recombinant vesicular stomatitis virus (mtdVSV)-based SARS-CoV-2 vaccine candidate. We generated mtdVSVs expressing SARS-CoV-2 full-length spike (S) protein, S1, or its receptor-binding domain (RBD). All of these recombinant viruses grew to high titers in mammalian cells despite high attenuation in cell culture. The SARS-CoV-2 S protein and its truncations were highly expressed by the mtdVSV vector. These mtdVSV-based vaccine candidates were completely attenuated in both immunocompetent and immunocompromised mice. Among these constructs, mtdVSV-S induced high levels of SARS-CoV-2-specific neutralizing antibodies (NAbs) and Th1-biased T-cell immune responses in mice. In Syrian golden hamsters, the serum levels of SARS-CoV-2-specific NAbs triggered by mtdVSV-S were higher than the levels of NAbs in convalescent plasma from recovered COVID-19 patients. In addition, hamsters immunized with mtdVSV-S were completely protected against SARS-CoV-2 replication in lung and nasal turbinate tissues, cytokine storm, and lung pathology. Collectively, our data demonstrate that mtdVSV expressing SARS-CoV-2 S protein is a safe and highly efficacious vaccine candidate against SARS-CoV-2 infection. IMPORTANCE Viral mRNA cap methyltransferase (MTase) is essential for mRNA stability, protein translation, and innate immune evasion. Thus, viral mRNA cap MTase activity is an excellent target for development of live attenuated or live vectored vaccine candidates. Here, we developed a panel of MTase-defective recombinant vesicular stomatitis virus (mtdVSV)-based SARS-CoV-2 vaccine candidates expressing full-length S, S1, or several versions of the RBD. These mtdVSV-based vaccine candidates grew to high titers in cell culture and were completely attenuated in both immunocompetent and immunocompromised mice. Among these vaccine candidates, mtdVSV-S induces high levels of SARS-CoV-2-specific neutralizing antibodies (Nabs) and Th1-biased immune responses in mice. Syrian golden hamsters immunized with mtdVSV-S triggered SARS-CoV-2-specific NAbs at higher levels than those in convalescent plasma from recovered COVID-19 patients. Furthermore, hamsters immunized with mtdVSV-S were completely protected against SARS-CoV-2 challenge. Thus, mtdVSV is a safe and highly effective vector to deliver SARS-CoV-2 vaccine.
Collapse
Affiliation(s)
- Mijia Lu
- Department of Veterinary Biosciences, The Ohio State University, Columbus, Ohio, USA
| | - Yuexiu Zhang
- Department of Veterinary Biosciences, The Ohio State University, Columbus, Ohio, USA
| | - Piyush Dravid
- Center for Vaccines and Immunity, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, Ohio, USA
| | - Anzhong Li
- Department of Veterinary Biosciences, The Ohio State University, Columbus, Ohio, USA
| | - Cong Zeng
- Department of Veterinary Biosciences, The Ohio State University, Columbus, Ohio, USA
| | - Mahesh KC
- Center for Vaccines and Immunity, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, Ohio, USA
| | - Sheetal Trivedi
- Center for Vaccines and Immunity, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, Ohio, USA
| | - Himanshu Sharma
- Center for Vaccines and Immunity, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, Ohio, USA
| | - Supranee Chaiwatpongsakorn
- Center for Vaccines and Immunity, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, Ohio, USA
| | - Ashley Zani
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Adam Kenney
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Chuanxi Cai
- Department of Surgery, College of Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Chengjin Ye
- Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Xueya Liang
- Department of Veterinary Biosciences, The Ohio State University, Columbus, Ohio, USA
| | - Jianming Qiu
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | | | - Jacob S. Yount
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Prosper N. Boyaka
- Department of Veterinary Biosciences, The Ohio State University, Columbus, Ohio, USA
- Infectious Disease Institute, The Ohio State University, Columbus, Ohio, USA
| | - Shan-Lu Liu
- Department of Veterinary Biosciences, The Ohio State University, Columbus, Ohio, USA
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, Ohio, USA
- Center for Retrovirus Research, The Ohio State University, Columbus, Ohio, USA
- Infectious Disease Institute, The Ohio State University, Columbus, Ohio, USA
| | - Mark E. Peeples
- Center for Vaccines and Immunity, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, Ohio, USA
- Department of Pediatrics, College of Medicine, The Ohio State University, Columbus, Ohio, USA
- Infectious Disease Institute, The Ohio State University, Columbus, Ohio, USA
| | - Amit Kapoor
- Center for Vaccines and Immunity, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, Ohio, USA
- Department of Pediatrics, College of Medicine, The Ohio State University, Columbus, Ohio, USA
- Infectious Disease Institute, The Ohio State University, Columbus, Ohio, USA
| | - Jianrong Li
- Department of Veterinary Biosciences, The Ohio State University, Columbus, Ohio, USA
- Infectious Disease Institute, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
26
|
Current Progress in the Development of Zika Virus Vaccines. Vaccines (Basel) 2021; 9:vaccines9091004. [PMID: 34579241 PMCID: PMC8472938 DOI: 10.3390/vaccines9091004] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Revised: 09/04/2021] [Accepted: 09/07/2021] [Indexed: 11/17/2022] Open
Abstract
Zika virus (ZIKV) is an arbovirus first discovered in the Americas. ZIKV infection is insidious based on its mild clinical symptoms observed after infection. In Brazil, after 2015, ZIKV infection broke out on a large scale, and many infected pregnant women gave birth to babies with microcephaly. The teratogenic effects of the virus on the fetus and its effects on nerves and the immune system have attracted great attention. Currently, no specific prophylactics or therapeutics are clinically available to treat ZIKV infection. Development of a safe and effective vaccine is essential to prevent the rise of any potential pandemic. In this review, we summarize the latest research on Zika vaccine development based on different strategies, including DNA vaccines, subunit vaccines, live-attenuated vaccines, virus-vector-based vaccines, inactivated vaccines, virus-like particles (VLPs), mRNA-based vaccines, and others. We anticipate that this review will facilitate further progress toward the development of effective and safe vaccines against ZIKV infection.
Collapse
|
27
|
Liu G, Cao W, Salawudeen A, Zhu W, Emeterio K, Safronetz D, Banadyga L. Vesicular Stomatitis Virus: From Agricultural Pathogen to Vaccine Vector. Pathogens 2021; 10:1092. [PMID: 34578125 PMCID: PMC8470541 DOI: 10.3390/pathogens10091092] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 08/23/2021] [Accepted: 08/25/2021] [Indexed: 11/16/2022] Open
Abstract
Vesicular stomatitis virus (VSV), which belongs to the Vesiculovirus genus of the family Rhabdoviridae, is a well studied livestock pathogen and prototypic non-segmented, negative-sense RNA virus. Although VSV is responsible for causing economically significant outbreaks of vesicular stomatitis in cattle, horses, and swine, the virus also represents a valuable research tool for molecular biologists and virologists. Indeed, the establishment of a reverse genetics system for the recovery of infectious VSV from cDNA transformed the utility of this virus and paved the way for its use as a vaccine vector. A highly effective VSV-based vaccine against Ebola virus recently received clinical approval, and many other VSV-based vaccines have been developed, particularly for high-consequence viruses. This review seeks to provide a holistic but concise overview of VSV, covering the virus's ascension from perennial agricultural scourge to promising medical countermeasure, with a particular focus on vaccines.
Collapse
Affiliation(s)
- Guodong Liu
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB R3E 3R2, Canada
| | - Wenguang Cao
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB R3E 3R2, Canada
| | - Abdjeleel Salawudeen
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Wenjun Zhu
- Canadian Food Inspection Agency, National Centre for Foreign Animal Disease, Winnipeg, MB R3E 3M4, Canada
| | - Karla Emeterio
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB R3E 3R2, Canada
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - David Safronetz
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB R3E 3R2, Canada
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Logan Banadyga
- Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB R3E 3R2, Canada
| |
Collapse
|
28
|
Carpio KL, Barrett ADT. Flavivirus NS1 and Its Potential in Vaccine Development. Vaccines (Basel) 2021; 9:622. [PMID: 34207516 PMCID: PMC8229460 DOI: 10.3390/vaccines9060622] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 06/03/2021] [Accepted: 06/04/2021] [Indexed: 12/19/2022] Open
Abstract
The Flavivirus genus contains many important human pathogens, including dengue, Japanese encephalitis (JE), tick-borne encephalitis (TBE), West Nile (WN), yellow fever (YF) and Zika (ZIK) viruses. While there are effective vaccines for a few flavivirus diseases (JE, TBE and YF), the majority do not have vaccines, including WN and ZIK. The flavivirus nonstructural 1 (NS1) protein has an unusual structure-function because it is glycosylated and forms different structures to facilitate different roles intracellularly and extracellularly, including roles in the replication complex, assisting in virus assembly, and complement antagonism. It also plays a role in protective immunity through antibody-mediated cellular cytotoxicity, and anti-NS1 antibodies elicit passive protection in animal models against a virus challenge. Historically, NS1 has been used as a diagnostic marker for the flavivirus infection due to its complement fixing properties and specificity. Its role in disease pathogenesis, and the strong humoral immune response resulting from infection, makes NS1 an excellent target for inclusion in candidate flavivirus vaccines.
Collapse
Affiliation(s)
- Kassandra L. Carpio
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555, USA;
| | - Alan D. T. Barrett
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA
- Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, TX 77555, USA
| |
Collapse
|
29
|
Jin H, Jiao C, Cao Z, Huang P, Chi H, Bai Y, Liu D, Wang J, Feng N, Li N, Zhao Y, Wang T, Gao Y, Yang S, Xia X, Wang H. An inactivated recombinant rabies virus displaying the Zika virus prM-E induces protective immunity against both pathogens. PLoS Negl Trop Dis 2021; 15:e0009484. [PMID: 34086672 PMCID: PMC8208564 DOI: 10.1371/journal.pntd.0009484] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 06/16/2021] [Accepted: 05/17/2021] [Indexed: 12/15/2022] Open
Abstract
The global spread of Zika virus (ZIKV), which caused a pandemic associated with Congenital Zika Syndrome and neuropathology in newborns and adults, prompted the pursuit of a safe and effective vaccine. Here, three kinds of recombinant rabies virus (RABV) encoding the prM-E protein of ZIKV were constructed: ZI-D (prM-E), ZI-E (transmembrane domain (TM) of prM-E replaced with RABV G) and ZI-F (signal peptide and TM domain of prM-E replaced with the region of RABV G). When the TM of prM-E was replaced with the region of RABV G (termed ZI-E), it promoted ZIKV E protein localization on the cell membrane and assembly on recombinant viruses. In addition, the change in the signal peptide with RABV G (termed ZI-F) was not conducive to foreign protein expression. The immunogenicity of recombinant viruses mixed with a complex adjuvant of ISA 201 VG and poly(I:C) was tested in BALB/c mice. After immunization with ZI-E, the anti-ZIKV IgG antibody lasted for at least 10 weeks. The titers of neutralizing antibodies (NAbs) against ZIKV and RABV at week 6 were all greater than the protective titers. Moreover, ZI-E stimulated the proliferation of splenic lymphocytes and promoted the secretion of cytokines. It also promoted the production of central memory T cells (TCMs) among CD4+/CD8+ T cells and stimulated B cell activation and maturation. These results indicate that ZI-E could induce ZIKV-specific humoral and cellular immune responses, which have the potential to be developed into a promising vaccine for protection against both ZIKV and RABV infections.
Collapse
Affiliation(s)
- Hongli Jin
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, China
| | - Cuicui Jiao
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Zengguo Cao
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, China
| | - Pei Huang
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, China
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China
| | - Hang Chi
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, China
| | - Yujie Bai
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Di Liu
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China
| | - Jianzhong Wang
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China
| | - Na Feng
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, China
| | - Nan Li
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, China
| | - Yongkun Zhao
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, China
| | - Tiecheng Wang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, China
| | - Yuwei Gao
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, China
| | - Songtao Yang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, China
| | - Xianzhu Xia
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, China
- * E-mail: (XX); (HW)
| | - Hualei Wang
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, China
- * E-mail: (XX); (HW)
| |
Collapse
|
30
|
Dey D, Poudyal S, Rehman A, Hasan SS. Structural and biochemical insights into flavivirus proteins. Virus Res 2021; 296:198343. [PMID: 33607183 DOI: 10.1016/j.virusres.2021.198343] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 02/10/2021] [Accepted: 02/11/2021] [Indexed: 01/01/2023]
Abstract
Flaviviruses are the fastest spreading arthropod-borne viruses that cause severe symptoms such as hepatitis, hemorrhagic fever, encephalitis, and congenital deformities. Nearly 40 % of the entire human population is at risk of flavivirus epidemics. Yet, effective vaccination is restricted only to a few flaviviruses such as yellow fever and Japanese encephalitis viruses, and most recently for select cases of dengue virus infections. Despite the global spread of dengue virus, and emergence of new threats such as Zika virus and a new genotype of Japanese encephalitis virus, insights into flavivirus targets for potentially broad-spectrum vaccination are limited. In this review article, we highlight biochemical and structural differences in flavivirus proteins critical for virus assembly and host interactions. A comparative sequence analysis of pH-responsive properties of viral structural proteins identifies trends in conservation of complementary acidic-basic character between interacting viral structural proteins. This is highly relevant to the understanding of pH-sensitive differences in virus assembly in organelles such as neutral ER and acidic Golgi. Surface residues in viral interfaces identified by structural approaches are shown to demonstrate partial conservation, further reinforcing virus-specificity in assembly and interactions with host proteins. A comparative analysis of epitope conservation in emerging flaviviruses identifies therapeutic antibody candidates that have potential as broad spectrum anti-virals, thus providing a path towards development of vaccines.
Collapse
Affiliation(s)
- Debajit Dey
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, 108 N. Greene Street, Baltimore MD 21201, USA
| | - Shishir Poudyal
- Department of Biological Sciences, Purdue University, 915 W. State Street, West Lafayette IN 47907, USA
| | - Asma Rehman
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, 108 N. Greene Street, Baltimore MD 21201, USA
| | - S Saif Hasan
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, 108 N. Greene Street, Baltimore MD 21201, USA; University of Maryland Marlene and Stewart Greenebaum Cancer Center, University of Maryland Medical Center, 22. S. Greene St. Baltimore MD 21201, USA; Center for Biomolecular Therapeutics, University of Maryland School of Medicine, 9600 Gudelsky Drive, Rockville MD 20850, USA.
| |
Collapse
|
31
|
Yu L, Liu X, Ye X, Su W, Zhang X, Deng W, Luo J, Xiang M, Guo W, Zhang S, Xu W, Yan Q, Wang Q, Cui Y, Wu C, Guo W, Niu X, Zhang F, Lei C, Qu L, Chen L, Feng L. Monoclonal Antibodies against Zika Virus NS1 Protein Confer Protection via Fc γ Receptor-Dependent and -Independent Pathways. mBio 2021; 12:e03179-20. [PMID: 33563822 PMCID: PMC7885117 DOI: 10.1128/mbio.03179-20] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 12/14/2020] [Indexed: 12/22/2022] Open
Abstract
Zika virus (ZIKV) infection during pregnancy causes congenital defects such as fetal microcephaly. Monoclonal antibodies (MAbs) against the nonstructural protein 1 (NS1) have the potential to suppress ZIKV pathogenicity without enhancement of disease, but the pathways through which they confer protection remain obscure. Here, we report two types of NS1-targeted human MAbs that inhibit ZIKV infection through distinct mechanisms. MAbs 3G2 and 4B8 show a better efficacy than MAb 4F10 in suppressing ZIKV infection in C57BL/6 neonatal mice. Unlike MAb 4F10 that mainly triggers antibody-dependent cell-mediated cytotoxicity (ADCC), MAbs 3G2 and 4B8 not only trigger ADCC but inhibit ZIKV infection without Fcγ receptor-bearing effector cells, possibly at postentry stages. Destroying the Fc-mediated effector function of MAbs 3G2 and 4B8 reduces but does not abolish their protective effects, whereas destroying the effector function of MAb 4F10 eliminates the protective effects, suggesting that MAbs 3G2 and 4B8 engage both Fcγ receptor-dependent and -independent pathways. Further analysis reveals that MAbs 3G2 and 4B8 target the N-terminal region of NS1 protein, whereas MAb 4F10 targets the C-terminal region, implying that the protective efficacy of an NS1-targeted MAb may be associated with its epitope recognition. Our results illustrate that NS1-targeted MAbs have multifaceted protective effects and provide insights for the development of NS1-based vaccines and therapeutics.IMPORTANCE Zika virus (ZIKV) is a mosquito-borne flavivirus that has been linked to congenital microcephaly during recent epidemics. No licensed antiviral drug or vaccine is available. Monoclonal antibodies (MAbs) against the nonstructural protein 1 (NS1) inhibit ZIKV pathogenicity but do not enhance the disease as envelope protein-targeted MAbs do. However, the protection mechanisms are not fully understood. Here, we show that in the presence or absence of Fcγ receptor-bearing effector cells, NS1-targeted human MAbs 3G2 and 4B8 inhibit ZIKV infection. Compared to MAb 4F10 that has no inhibitory effects without effector cells, 3G2 and 4B8 confer better protection in ZIKV-infected neonatal mice. Destroying the Fc-mediated effector function reduces but does not abolish the protection of 3G2 and 4B8, suggesting that they engage both Fcγ receptor-dependent and -independent pathways. The protective efficacy of NS1-targeted MAbs may be associated with their epitope recognition. Our findings will help to develop NS1-based vaccines and therapeutics.
Collapse
Affiliation(s)
- Lei Yu
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Xinglong Liu
- State Key Laboratories of Respiratory Diseases, Guangdong Provincial Key Laboratory of Computational Biomedicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Xianmiao Ye
- State Key Laboratories of Respiratory Diseases, Guangdong Provincial Key Laboratory of Computational Biomedicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Wan Su
- School of Biomedical Sciences, Huaqiao University, Quanzhou, China
| | - Xiaoyan Zhang
- State Key Laboratories of Respiratory Diseases, Guangdong Provincial Key Laboratory of Computational Biomedicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Weiqi Deng
- State Key Laboratories of Respiratory Diseases, Guangdong Provincial Key Laboratory of Computational Biomedicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Jia Luo
- State Key Laboratories of Respiratory Diseases, Guangdong Provincial Key Laboratory of Computational Biomedicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Mengrong Xiang
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Wenjing Guo
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Shengnan Zhang
- State Key Laboratories of Respiratory Diseases, Guangdong Provincial Key Laboratory of Computational Biomedicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Wei Xu
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Qihong Yan
- State Key Laboratories of Respiratory Diseases, Guangdong Provincial Key Laboratory of Computational Biomedicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Qian Wang
- State Key Laboratories of Respiratory Diseases, Guangdong Provincial Key Laboratory of Computational Biomedicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yilan Cui
- State Key Laboratories of Respiratory Diseases, Guangdong Provincial Key Laboratory of Computational Biomedicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Caixia Wu
- State Key Laboratories of Respiratory Diseases, Guangdong Provincial Key Laboratory of Computational Biomedicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Wenjing Guo
- State Key Laboratories of Respiratory Diseases, Guangdong Provincial Key Laboratory of Computational Biomedicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Xuefeng Niu
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Fuchun Zhang
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Chunliang Lei
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Linbing Qu
- State Key Laboratories of Respiratory Diseases, Guangdong Provincial Key Laboratory of Computational Biomedicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Ling Chen
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, China
- State Key Laboratories of Respiratory Diseases, Guangdong Provincial Key Laboratory of Computational Biomedicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- School of Biomedical Sciences, Huaqiao University, Quanzhou, China
- Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Liqiang Feng
- State Key Laboratories of Respiratory Diseases, Guangdong Provincial Key Laboratory of Computational Biomedicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China
- University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
32
|
Zhang X, Li G, Chen G, Zhu N, Wu D, Wu Y, James TD. Recent progresses and remaining challenges for the detection of Zika virus. Med Res Rev 2021; 41:2039-2108. [PMID: 33559917 DOI: 10.1002/med.21786] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 12/23/2020] [Accepted: 01/05/2021] [Indexed: 12/26/2022]
Abstract
Zika virus (ZIKV) has emerged as a particularly notorious mosquito-borne flavivirus, which can lead to a devastating congenital syndrome in the fetuses of pregnant mothers (e.g., microcephaly, spasticity, craniofacial disproportion, miscarriage, and ocular abnormalities) and cause the autoimmune disorder Guillain-Barre' syndrome of adults. Due to its severity and rapid dispersal over several continents, ZIKV has been acknowledged to be a global health concern by the World Health Organization. Unfortunately, the ZIKV has recently resurged in India with the potential for devastating effects. Researchers from all around the world have worked tirelessly to develop effective detection strategies and vaccines for the prevention and control of ZIKV infection. In this review, we comprehensively summarize the most recent research into ZIKV, including the structural biology and evolution, historical overview, pathogenesis, symptoms, and transmission. We then focus on the detection strategies for ZIKV, including viral isolation, serological assays, molecular assays, sensing methods, reverse transcription loop mediated isothermal amplification, transcription-mediated amplification technology, reverse transcription strand invasion based amplification, bioplasmonic paper-based device, and reverse transcription isothermal recombinase polymerase amplification. To conclude, we examine the limitations of currently available strategies for the detection of ZIKV, and outline future opportunities and research challenges.
Collapse
Affiliation(s)
- Xianlong Zhang
- School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi'an, China
| | - Guoliang Li
- School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi'an, China
| | - Guang Chen
- School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi'an, China
| | - Niu Zhu
- Department of Public Health, Xi'an Medical University, Xi'an, China
| | - Di Wu
- Institute for Global Food Security, Queen's University Belfast, Belfast, UK
| | - Yongning Wu
- NHC Key Laboratory of Food Safety Risk Assessment, Food Safety Research Unit (2019RU014) of Chinese Academy of Medical Science, China National Center for Food Safety Risk Assessment, Beijing, China
| | - Tony D James
- Department of Chemistry, University of Bath, Bath, UK.,School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang, China
| |
Collapse
|
33
|
Pereira LR, Alves RPDS, Sales NS, Andreata-Santos R, Venceslau-Carvalho AA, Pereira SS, Castro-Amarante MF, Rodrigues-Jesus MJ, Favaro MTDP, Chura-Chambi RM, Morganti L, Ferreira LCDS. Enhanced Immune Responses and Protective Immunity to Zika Virus Induced by a DNA Vaccine Encoding a Chimeric NS1 Fused With Type 1 Herpes Virus gD Protein. FRONTIERS IN MEDICAL TECHNOLOGY 2020; 2:604160. [PMID: 35047887 PMCID: PMC8757838 DOI: 10.3389/fmedt.2020.604160] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 11/11/2020] [Indexed: 12/30/2022] Open
Abstract
Zika virus (ZIKV) is a globally-distributed flavivirus transmitted to humans by Aedes mosquitoes, usually causing mild symptoms that may evolve to severe conditions, including neurological alterations, such as neonatal microcephaly and Guillain-Barré syndrome. Due to the absence of specific and effective preventive methods, we designed a new subunit vaccine based on a DNA vector (pgDNS1-ZIKV) encoding the non-structural protein 1 (NS1) genetically fused to the Herpes Simplex Virus (HSV) glycoprotein D (gD) protein. Recombinant plasmids were replicated in Escherichia coli and the expression of the target protein was confirmed in transfected HEK293 cells. C57BL/6 and AB6 (IFNAR1-/-) mice were i.m. immunized by electroporation in order to evaluate pgDNS1-ZIKV immunogenicity. After two doses, high NS1-specific IgG antibody titers were measured in serum samples collected from pgDNS1-ZIKV-immunized mice. The NS1-specific antibodies were capable to bind the native protein expressed in infected mammalian cells. Immunization with pgDNS1-ZIKV increased both humoral and cellular immune responses regarding mice immunized with a ZIKV NS1 encoding vaccine. Immunization with pgDNS1-ZIKV reduced viremia and morbidity scores leading to enhanced survival of immunodeficient AB6 mice challenged with a lethal virus load. These results give support to the use of ZIKV NS1 as a target antigen and further demonstrate the relevant adjuvant effects of HSV-1 gD.
Collapse
Affiliation(s)
- Lennon Ramos Pereira
- Laboratory of Vaccine Development, Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Rúbens Prince dos Santos Alves
- Laboratory of Vaccine Development, Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Natiely Silva Sales
- Laboratory of Vaccine Development, Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Robert Andreata-Santos
- Laboratory of Vaccine Development, Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Aléxia Adrianne Venceslau-Carvalho
- Laboratory of Vaccine Development, Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Samuel Santos Pereira
- Laboratory of Vaccine Development, Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Maria Fernanda Castro-Amarante
- Laboratory of Vaccine Development, Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Mônica Josiane Rodrigues-Jesus
- Laboratory of Vaccine Development, Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Marianna Teixeira de Pinho Favaro
- Laboratory of Vaccine Development, Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | | | - Ligia Morganti
- Biotechnology Center, Institute of Energy and Nuclear Research (IPEN), São Paulo, Brazil
| | - Luís Carlos de Souza Ferreira
- Laboratory of Vaccine Development, Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil,*Correspondence: Luís Carlos de Souza Ferreira
| |
Collapse
|
34
|
Zhan Y, Pang Z, Du Y, Wang W, Yang Y, Wang W, Gao GF, Huang B, Deng Y, Tan W. NS1-based DNA vaccination confers mouse protective immunity against ZIKV challenge. INFECTION GENETICS AND EVOLUTION 2020; 85:104521. [DOI: 10.1016/j.meegid.2020.104521] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Revised: 08/24/2020] [Accepted: 08/27/2020] [Indexed: 02/06/2023]
|
35
|
Wessel AW, Kose N, Bombardi RG, Roy V, Chantima W, Mongkolsapaya J, Edeling MA, Nelson CA, Bosch I, Alter G, Screaton GR, Fremont DH, Crowe JE, Diamond MS. Antibodies targeting epitopes on the cell-surface form of NS1 protect against Zika virus infection during pregnancy. Nat Commun 2020; 11:5278. [PMID: 33077712 PMCID: PMC7572419 DOI: 10.1038/s41467-020-19096-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 09/29/2020] [Indexed: 12/18/2022] Open
Abstract
There are no licensed therapeutics or vaccines available against Zika virus (ZIKV) to counteract its potential for congenital disease. Antibody-based countermeasures targeting the ZIKV envelope protein have been hampered by concerns for cross-reactive responses that induce antibody-dependent enhancement (ADE) of heterologous flavivirus infection. Nonstructural protein 1 (NS1) is a membrane-associated and secreted glycoprotein that functions in flavivirus replication and immune evasion but is absent from the virion. Although some studies suggest that antibodies against ZIKV NS1 are protective, their activity during congenital infection is unknown. Here we develop mouse and human anti-NS1 monoclonal antibodies that protect against ZIKV in both non-pregnant and pregnant mice. Avidity of antibody binding to cell-surface NS1 along with Fc effector functions engagement correlate with protection in vivo. Protective mAbs map to exposed epitopes in the wing domain and loop face of the β-platform. Anti-NS1 antibodies provide an alternative strategy for protection against congenital ZIKV infection without causing ADE.
Collapse
Affiliation(s)
- Alex W Wessel
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Nurgun Kose
- Departments of Pediatrics, Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Robin G Bombardi
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Vicky Roy
- Ragon Institute of MGH, MIT, and Harvard University, Cambridge, MA, 02139, USA
| | - Warangkana Chantima
- Nuffield Department of Medicine, Wellcome Centre for Human Genetics, University of Oxford, Oxford, OX3 7BN, UK
| | - Juthathip Mongkolsapaya
- Nuffield Department of Medicine, Wellcome Centre for Human Genetics, University of Oxford, Oxford, OX3 7BN, UK
- Dengue Hemorrhagic Fever Unit, Faculty of Medicine, Office for Research and Development, Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Melissa A Edeling
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Christopher A Nelson
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Irene Bosch
- E25Bio, Inc., The Engine of MIT, Cambridge, MA, 02139, USA
| | - Galit Alter
- Ragon Institute of MGH, MIT, and Harvard University, Cambridge, MA, 02139, USA
| | - Gavin R Screaton
- Nuffield Department of Medicine, Wellcome Centre for Human Genetics, University of Oxford, Oxford, OX3 7BN, UK
| | - David H Fremont
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO, 63110, USA
- The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - James E Crowe
- Departments of Pediatrics, Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Michael S Diamond
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, 63110, USA.
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, 63110, USA.
- The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, MO, 63110, USA.
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA.
| |
Collapse
|
36
|
Zhang N, Li C, Jiang S, Du L. Recent Advances in the Development of Virus-Like Particle-Based Flavivirus Vaccines. Vaccines (Basel) 2020; 8:vaccines8030481. [PMID: 32867194 PMCID: PMC7565697 DOI: 10.3390/vaccines8030481] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Revised: 08/21/2020] [Accepted: 08/24/2020] [Indexed: 01/07/2023] Open
Abstract
Flaviviruses include several medically important viruses, such as Zika virus (ZIKV), Dengue virus (DENV), West Nile virus (WNV) and Japanese encephalitis virus (JEV). They have expanded in geographic distribution and refocused international attention in recent years. Vaccination is one of the most effective public health strategies for combating flavivirus infections. In this review, we summarized virus-like particle (VLP)-based vaccines against the above four mentioned flaviviruses. Potential strategies to improve the efficacy of VLP-based flavivirus vaccines were also illustrated. The applications of flavivirus VLPs as tools for viral detection and antiviral drug screening were finally proposed.
Collapse
Affiliation(s)
- Naru Zhang
- Department of Clinical Medicine, School of Medicine, Zhejiang University City College, Hangzhou 310015, China; (N.Z.); (C.L.)
| | - Chaoqun Li
- Department of Clinical Medicine, School of Medicine, Zhejiang University City College, Hangzhou 310015, China; (N.Z.); (C.L.)
| | - Shibo Jiang
- School of Basic Medical Sciences, Fudan University, Shanghai 200433, China
- Lindsley F. Kimball Research Institute, New York Blood Center, New York, NY 10065, USA
- Correspondence: (S.J.); (L.D.)
| | - Lanying Du
- Lindsley F. Kimball Research Institute, New York Blood Center, New York, NY 10065, USA
- Correspondence: (S.J.); (L.D.)
| |
Collapse
|
37
|
Vesicular Stomatitis Virus and DNA Vaccines Expressing Zika Virus Nonstructural Protein 1 Induce Substantial but Not Sterilizing Protection against Zika Virus Infection. J Virol 2020; 94:JVI.00048-20. [PMID: 32554698 DOI: 10.1128/jvi.00048-20] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Accepted: 06/10/2020] [Indexed: 11/20/2022] Open
Abstract
The nonstructural protein 1 (NS1) of several flaviviruses, including West Nile, dengue, and yellow fever viruses, is capable of inducing variable degrees of protection against flavivirus infection in animal models. However, the immunogenicity of NS1 protein of Zika virus (ZIKV) is less understood. Here, we determined the efficacy of ZIKV NS1-based vaccine candidates using two delivery platforms, methyltransferase-defective recombinant vesicular stomatitis virus (mtdVSV) and a DNA vaccine. We first show that expression of ZIKV NS1 could be significantly enhanced by optimizing the signal peptide. A single dose of mtdVSV-NS1-based vaccine or two doses of DNA vaccine induced high levels of NS1-specfic antibody and T cell immune responses but provided only partial protection against ZIKV viremia in BALB/c mice. In Ifnar1-/- mice, neither NS1-based vaccine provided protection against a lethal high dose (105 PFU) ZIKV challenge, but mtdVSV-NS1-based vaccine prevented deaths from a low dose (103 PFU) challenge, though they experienced viremia and body weight loss. We conclude that ZIKV NS1 alone conferred substantial, but not complete, protection against ZIKV infection. Nevertheless, these results highlight the value of ZIKV NS1 for vaccine development.IMPORTANCE Most Zika virus (ZIKV) vaccine research has focused on the E or prM-E proteins and the induction of high levels of neutralizing antibodies. However, these ZIKV neutralizing antibodies cross-react with other flaviviruses, which may aggravate the disease via an antibody-dependent enhancement (ADE) mechanism. ZIKV NS1 protein may be an alternative antigen for vaccine development, since antibodies to NS1 do not bind to the virion, thereby eliminating the risk of ADE. Here, we show that recombinant VSV and DNA vaccines expressing NS1, alone, confer partial protection against ZIKV infection in both immunocompetent and immunodeficient mice, highlighting the value of NS1 as a potential vaccine candidate.
Collapse
|
38
|
Kubinski M, Beicht J, Gerlach T, Volz A, Sutter G, Rimmelzwaan GF. Tick-Borne Encephalitis Virus: A Quest for Better Vaccines against a Virus on the Rise. Vaccines (Basel) 2020; 8:E451. [PMID: 32806696 PMCID: PMC7564546 DOI: 10.3390/vaccines8030451] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 08/06/2020] [Accepted: 08/10/2020] [Indexed: 12/15/2022] Open
Abstract
Tick-borne encephalitis virus (TBEV), a member of the family Flaviviridae, is one of the most important tick-transmitted viruses in Europe and Asia. Being a neurotropic virus, TBEV causes infection of the central nervous system, leading to various (permanent) neurological disorders summarized as tick-borne encephalitis (TBE). The incidence of TBE cases has increased due to the expansion of TBEV and its vectors. Since antiviral treatment is lacking, vaccination against TBEV is the most important protective measure. However, vaccination coverage is relatively low and immunogenicity of the currently available vaccines is limited, which may account for the vaccine failures that are observed. Understanding the TBEV-specific correlates of protection is of pivotal importance for developing novel and improved TBEV vaccines. For affording robust protection against infection and development of TBE, vaccines should induce both humoral and cellular immunity. In this review, the adaptive immunity induced upon TBEV infection and vaccination as well as novel approaches to produce improved TBEV vaccines are discussed.
Collapse
Affiliation(s)
- Mareike Kubinski
- Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine Hannover, Foundation (TiHo), Buenteweg 17, 30559 Hannover, Germany; (M.K.); (J.B.); (T.G.)
| | - Jana Beicht
- Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine Hannover, Foundation (TiHo), Buenteweg 17, 30559 Hannover, Germany; (M.K.); (J.B.); (T.G.)
| | - Thomas Gerlach
- Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine Hannover, Foundation (TiHo), Buenteweg 17, 30559 Hannover, Germany; (M.K.); (J.B.); (T.G.)
| | - Asisa Volz
- Institute of Virology, University of Veterinary Medicine Hannover, Foundation (TiHo), Buenteweg 17, 30559 Hannover, Germany;
| | - Gerd Sutter
- Institute for Infectious Diseases and Zoonoses, Ludwig-Maximilians-University (LMU) Munich, Veterinaerstr. 13, 80539 Munich, Germany;
| | - Guus F. Rimmelzwaan
- Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine Hannover, Foundation (TiHo), Buenteweg 17, 30559 Hannover, Germany; (M.K.); (J.B.); (T.G.)
| |
Collapse
|
39
|
Ezzemani W, Windisch MP, Kettani A, Altawalah H, Nourlil J, Benjelloun S, Ezzikouri S. Immuno-informatics-based identification of novel potential B cell and T cell epitopes to fight Zika virus infections. Infect Disord Drug Targets 2020; 21:572-581. [PMID: 32778040 DOI: 10.2174/1871526520666200810153657] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Revised: 05/23/2020] [Accepted: 06/23/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Globally, the recent outbreak of Zika virus (ZIKV) in Brazil, Asia Pacific, and other countries highlighted the unmet medical needs. Currently, there are neither effective vaccines nor therapeutics available to prevent or treat ZIKV infection. OBJECTIVE In this study, we aimed to design an epitope-based vaccine for ZIKV using an in silico approach to predict and analyze B- and T-cell epitopes. METHODS The prediction of the most antigenic epitopes has targeted the capsid and the envelope proteins as well as nonstructural proteins NS5 and NS3 using immune-informatics tools PROTPARAM, CFSSP, PSIPRED, and Vaxijen v2.0. B and T-cell epitopes were predicted using ABCpred, IEDB, TepiTool, and their toxicity were evaluated using ToxinPred. The 3-dimensional epitope structures were generated by PEP-FOLD. Energy minimization was performed using Swiss-Pdb Viewer, and molecular docking was conducted using PatchDock and FireDock server. RESULTS As a result, we predicted 307 epitopes of MHCI (major histocompatibility complex class I) and 102 epitopes of MHCII (major histocompatibility complex class II). Based on immunogenicity and antigenicity scores, we identified the four most antigenic MHC I epitopes: MVLAILAFLR (HLA-A*68 :01), ETLHGTVTV (HLA-A*68 :02), DENHPYRTW (HLA-B*44 :02),QEGVFHTMW (HLA-B*44 :03) and TASGRVIEEW (HLA-B*58:01), and MHC II epitopes: IIKKFKKDLAAMLRI (HLA-DRB3*02 :02), ENSKMMLELDPPFGD (HLA-DRB3*01:01), HAETWFFDENHPYRT (HLA-DRB3*01:01), TDGVYRVMTRRLLGS (HLA-DRB1*11 :01), and DGCWYGMEIRPRKEP (HLA-DRB5*01:01). CONCLUSION This study provides novel potential B cell and T cell epitopes to fight Zika virus infections and may prompt further development of vaccines against ZIKV and other emerging infectious diseases. However, further investigations for protective immune response by in vitro and in vivo studies to ratify the immunogenicity, safety of the predicted structure, and ultimately the vaccine properties to prevent ZIKV infections are warranted.
Collapse
Affiliation(s)
- Wahiba Ezzemani
- Virology Unit, Viral Hepatitis Laboratory, Institut Pasteur du Maroc, Casablanca. Morocco
| | - Marc P Windisch
- Applied Molecular Virology Laboratory, Discovery Biology Department, Institut Pasteur Korea, Seongnamsi, Gyeonggi-do. South Korea
| | - Anass Kettani
- Laboratoire de Biologie et Santé (URAC34), Département de Biologie, Faculté des Sciences Ben Msik, Hassan II University Of Casablanca. Morocco
| | - Haya Altawalah
- Department of Microbiology, Faculty of Medicine, Kuwait University. Kuwait
| | - Jalal Nourlil
- Medical Virology and BSL3 Laboratory, Institut Pasteur du Maroc, Casablanca. Morocco
| | - Soumaya Benjelloun
- Virology Unit, Viral Hepatitis Laboratory, Institut Pasteur du Maroc, Casablanca. Morocco
| | - Sayeh Ezzikouri
- Laboratoire de Biologie et Santé (URAC34), Département de Biologie, Faculté des Sciences Ben Msik, Hassan II University Of Casablanca. Morocco
| |
Collapse
|
40
|
In HJ, Lee YH, Jang S, Lim HJ, Kim MY, Kim JA, Yoo JS, Chung GT, Kim YJ. Enhanced effect of modified Zika virus E antigen on the immunogenicity of DNA vaccine. Virology 2020; 549:25-31. [PMID: 32818729 DOI: 10.1016/j.virol.2020.07.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 07/10/2020] [Accepted: 07/21/2020] [Indexed: 01/21/2023]
Abstract
It has been reported worldwide that the Zika virus (ZIKV) could be transmitted through placentas and sexual contact. ZIKV can also cause Guillain-Barre syndrome, microcephaly and neurological abnormalities. However, there are no approved vaccines available. We constructed six DNA vaccine candidates and tested the immunogenicity. Tandem repeated envelope domain Ⅲ (ED Ⅲ × 3) induced highly total IgG and neutralization antibody, as well as CD8+ T cell responses. Also, stem region-removed envelope (E ΔSTEM) elicited a robust production of IFN-γ in mice. To examine in vivo protection, we used mice treated with an IFNAR1 blocking antibody before and after the challenge. Vaccination with the two candidates led to a decline in the level of viral RNAs in organs. Moreover, the sera from the vaccinated mice did not enhance the infection of Dengue virus in K562 cells. These findings suggest the potential for the development of a novel ZIKV DNA vaccine.
Collapse
Affiliation(s)
- Hyun Ju In
- Division of Vaccine Research, Korea National Institute of Health, Korea Centers for Disease Control and Prevention, CheongJu, Chungbuk, 28160, Republic of Korea
| | - Yun Ha Lee
- Division of Vaccine Research, Korea National Institute of Health, Korea Centers for Disease Control and Prevention, CheongJu, Chungbuk, 28160, Republic of Korea
| | - Sundong Jang
- College of Pharmacy, Chungbuk National University, CheongJu, Chungbuk, 28160, Republic of Korea
| | - Hee Ji Lim
- Division of Vaccine Research, Korea National Institute of Health, Korea Centers for Disease Control and Prevention, CheongJu, Chungbuk, 28160, Republic of Korea
| | - Mi Young Kim
- Division of Vaccine Research, Korea National Institute of Health, Korea Centers for Disease Control and Prevention, CheongJu, Chungbuk, 28160, Republic of Korea
| | - Joo Ae Kim
- Division of Vaccine Research, Korea National Institute of Health, Korea Centers for Disease Control and Prevention, CheongJu, Chungbuk, 28160, Republic of Korea
| | - Jung-Sik Yoo
- Division of Vaccine Research, Korea National Institute of Health, Korea Centers for Disease Control and Prevention, CheongJu, Chungbuk, 28160, Republic of Korea
| | - Gyung Tae Chung
- Division of Vaccine Research, Korea National Institute of Health, Korea Centers for Disease Control and Prevention, CheongJu, Chungbuk, 28160, Republic of Korea
| | - You-Jin Kim
- Division of Vaccine Research, Korea National Institute of Health, Korea Centers for Disease Control and Prevention, CheongJu, Chungbuk, 28160, Republic of Korea.
| |
Collapse
|
41
|
Zhang M, Sun J, Li M, Jin X. Modified mRNA-LNP Vaccines Confer Protection against Experimental DENV-2 Infection in Mice. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2020; 18:702-712. [PMID: 32913878 PMCID: PMC7452130 DOI: 10.1016/j.omtm.2020.07.013] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 07/16/2020] [Indexed: 12/12/2022]
Abstract
Dengue virus (DENV) infection is a major global public health concern, and there is no effective vaccine for it. In this study, we describe the design and characterization of three nucleotide-modified mRNA vaccines (prME-mRNA, E80-mRNA, and NS1-mRNA) for DENV-2. Our results showed that vaccination with E80-mRNA alone or a combination of E80-mRNA and NS1-mRNA can induce high levels of neutralizing antibodies and antigen-specific T cell responses; furthermore, these vaccines confer complete protection against DENV-2 challenge in immunocompetent mice. These data provide foundations for further development of a tetravalent DENV vaccine based on nucleotide-modified mRNA.
Collapse
Affiliation(s)
- Mengling Zhang
- 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.,University of Chinese Academy of Sciences, Beijing, China.,Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - 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
| | - Min Li
- 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
| | - Xia Jin
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| |
Collapse
|
42
|
TRiC/CCT Complex, a Binding Partner of NS1 Protein, Supports the Replication of Zika Virus in Both Mammalians and Mosquitoes. Viruses 2020; 12:v12050519. [PMID: 32397176 PMCID: PMC7290343 DOI: 10.3390/v12050519] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 05/02/2020] [Accepted: 05/06/2020] [Indexed: 11/23/2022] Open
Abstract
Mosquito-borne Zika virus (ZIKV) can cause congenital microcephaly and Guillain–Barré syndrome, among other symptoms. Specific treatments and vaccines for ZIKV are not currently available. To further understand the host factors that support ZIKV replication, we used mass spectrometry to characterize mammalian proteins that associate with the ZIKV NS1 protein and identified the TRiC/CCT complex as an interacting partner. Furthermore, the suppression of CCT2, one of the critical components of the TRiC/CCT complex, inhibited ZIKV replication in both mammalian cells and mosquitoes. These results highlight an important role for the TRiC/CCT complex in ZIKV infection, suggesting that the TRiC/CCT complex may be a promising therapeutic target.
Collapse
|
43
|
Subramaniam KS, Lant S, Goodwin L, Grifoni A, Weiskopf D, Turtle L. Two Is Better Than One: Evidence for T-Cell Cross-Protection Between Dengue and Zika and Implications on Vaccine Design. Front Immunol 2020; 11:517. [PMID: 32269575 PMCID: PMC7109261 DOI: 10.3389/fimmu.2020.00517] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 03/06/2020] [Indexed: 12/13/2022] Open
Abstract
Dengue virus (DENV, family Flaviviridae, genus Flavivirus) exists as four distinct serotypes. Generally, immunity after infection with one serotype is protective and lifelong, though exceptions have been described. However, secondary infection with a different serotype can result in more severe disease for a minority of patients. Host responses to the first DENV infection involve the development of both cross-reactive antibody and T cell responses, which, depending upon their precise balance, may mediate protection or enhance disease upon secondary infection with a different serotype. Abundant evidence now exists that responses elicited by DENV infection can cross-react with other members of the genus Flavivirus, particularly Zika virus (ZIKV). Cohort studies have shown that prior DENV immunity is associated with protection against Zika. Cross-reactive antibody responses may enhance infection with flaviviruses, which likely accounts for the cases of severe disease seen during secondary DENV infections. Data for T cell responses are contradictory, and even though cross-reactive T cell responses exist, their clinical significance is uncertain. Recent mouse experiments, however, show that cross-reactive T cells are capable of mediating protection against ZIKV. In this review, we summarize and discuss the evidence that T cell responses may, at least in part, explain the cross-protection seen against ZIKV from DENV infection, and that T cell antigens should therefore be included in putative Zika vaccines.
Collapse
Affiliation(s)
- Krishanthi S. Subramaniam
- NIHR Health Protection Research Unit in Emerging and Zoonotic Infections, Centre for Global Vaccine Research, Institute of Infection and Global Health, University of Liverpool, Liverpool, United Kingdom
| | - Suzannah Lant
- NIHR Health Protection Research Unit in Emerging and Zoonotic Infections, Centre for Global Vaccine Research, Institute of Infection and Global Health, University of Liverpool, Liverpool, United Kingdom
| | - Lynsey Goodwin
- NIHR Health Protection Research Unit in Emerging and Zoonotic Infections, Centre for Global Vaccine Research, Institute of Infection and Global Health, University of Liverpool, Liverpool, United Kingdom
| | - Alba Grifoni
- Division of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA, United States
| | - Daniela Weiskopf
- Division of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA, United States
| | - Lance Turtle
- NIHR Health Protection Research Unit in Emerging and Zoonotic Infections, Centre for Global Vaccine Research, Institute of Infection and Global Health, University of Liverpool, Liverpool, United Kingdom
- Tropical and Infectious Disease Unit, Liverpool University Hospitals, Liverpool, United Kingdom
| |
Collapse
|
44
|
Collins ND, Widen SG, Li L, Swetnam DM, Shi PY, Tesh RB, Sarathy VV. Inter- and intra-lineage genetic diversity of wild-type Zika viruses reveals both common and distinctive nucleotide variants and clusters of genomic diversity. Emerg Microbes Infect 2019; 8:1126-1138. [PMID: 31355708 PMCID: PMC6711133 DOI: 10.1080/22221751.2019.1645572] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Zika virus (ZIKV) strains belong to the East African, West African, and Asian/American phylogenetic lineages. RNA viruses, like ZIKV, exist as populations of genetically-related sequences whose heterogeneity may impact viral fitness, evolution, and virulence. Genetic diversity of representative ZIKVs from each lineage was examined using next generation sequencing (NGS) paired with downstream entropy and single nucleotide variant (SNV) analysis. Comparisons showed that inter-lineage diversity was statistically supported, while intra-lineage diversity. Intra-lineage diversity was significant for East but not West Africa strains. Furthermore, intra-lineage diversity for the Asian/American lineage was not supported for human serum isolates; however, a placenta isolate differed significantly. Relative in the pre-membrane/membrane (prM/M) gene of several ZIKV strains. Additionally, the East African lineage contained a greater number of synonymous SNVs, while a greater number of non-synonymous SNVs were identified for American strains. Further, inter-lineage SNVs were dispersed throughout the genome, whereas intra-lineage non-synonymous SNVs for Asian/American strains clustered within prM/M and NS1 gene. This comprehensive analysis of ZIKV genetic diversity provides a repository of SNV positions across lineages. We posit that increased non-synonymous SNV populations and increased relative genetic diversity of the prM/M and NS1 proteins provides more evidence for their role in ZIKV virulence and fitness.
Collapse
Affiliation(s)
- Natalie D Collins
- a Department of Microbiology and Immunology, University of Texas Medical Branch , Galveston , USA
| | - Steven G Widen
- b Department of Biochemistry and Molecular Biology, University of Texas Medical Branch , Galveston , USA
| | - Li Li
- c Department of Pathology, University of Texas Medical Branch , Galveston , USA
| | - Daniele M Swetnam
- d Department of Pathology, Microbiology and Immunology, School of Veterinary Medicine at University of California , Davis , USA
| | - Pei-Yong Shi
- b Department of Biochemistry and Molecular Biology, University of Texas Medical Branch , Galveston , USA
| | - Robert B Tesh
- c Department of Pathology, University of Texas Medical Branch , Galveston , USA
| | - Vanessa V Sarathy
- c Department of Pathology, University of Texas Medical Branch , Galveston , USA.,e Sealy Institute for Vaccine Sciences, Institute for Human Infections and Immunity, University of Texas Medical Branch , Galveston , USA
| |
Collapse
|
45
|
Grubor-Bauk B, Wijesundara DK, Masavuli M, Abbink P, Peterson RL, Prow NA, Larocca RA, Mekonnen ZA, Shrestha A, Eyre NS, Beard MR, Gummow J, Carr J, Robertson SA, Hayball JD, Barouch DH, Gowans EJ. NS1 DNA vaccination protects against Zika infection through T cell-mediated immunity in immunocompetent mice. SCIENCE ADVANCES 2019; 5:eaax2388. [PMID: 31844662 PMCID: PMC6905874 DOI: 10.1126/sciadv.aax2388] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 10/08/2019] [Indexed: 05/08/2023]
Abstract
The causal association of Zika virus (ZIKV) with microcephaly, congenital malformations in infants, and Guillain-Barré syndrome in adults highlights the need for effective vaccines. Thus far, efforts to develop ZIKV vaccines have focused on the viral envelope. ZIKV NS1 as a vaccine immunogen has not been fully explored, although it can circumvent the risk of antibody-dependent enhancement of ZIKV infection, associated with envelope antibodies. Here, we describe a novel DNA vaccine encoding a secreted ZIKV NS1, that confers rapid protection from systemic ZIKV infection in immunocompetent mice. We identify novel NS1 T cell epitopes in vivo and show that functional NS1-specific T cell responses are critical for protection against ZIKV infection. We demonstrate that vaccine-induced anti-NS1 antibodies fail to confer protection in the absence of a functional T cell response. This highlights the importance of using NS1 as a target for T cell-based ZIKV vaccines.
Collapse
Affiliation(s)
- B. Grubor-Bauk
- Discipline of Surgery, University of Adelaide and Basil Hetzel Institute for Translational Health Research, Adelaide, SA 5005, Australia
- Corresponding author.
| | - D. K. Wijesundara
- Discipline of Surgery, University of Adelaide and Basil Hetzel Institute for Translational Health Research, Adelaide, SA 5005, Australia
| | - M. Masavuli
- Discipline of Surgery, University of Adelaide and Basil Hetzel Institute for Translational Health Research, Adelaide, SA 5005, Australia
| | - P. Abbink
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - R. L. Peterson
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - N. A. Prow
- Experimental Therapeutics Laboratory, Cancer Research Institute, School of Pharmacy and Medical Science, University of South Australia, Adelaide, SA 5000, Australia
- QIMR Berghofer Medical Research Institute, Brisbane, QLD 4029, Australia
- Australian Infectious Diseases Research Centre, Brisbane, QLD 4072, Australia
| | - R. A. Larocca
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Z. A. Mekonnen
- Discipline of Surgery, University of Adelaide and Basil Hetzel Institute for Translational Health Research, Adelaide, SA 5005, Australia
| | - A. Shrestha
- Discipline of Surgery, University of Adelaide and Basil Hetzel Institute for Translational Health Research, Adelaide, SA 5005, Australia
| | - N. S. Eyre
- Research Centre for Infectious Diseases, School of Biological Sciences, University of Adelaide, Adelaide, SA 5005, Australia
| | - M. R. Beard
- Research Centre for Infectious Diseases, School of Biological Sciences, University of Adelaide, Adelaide, SA 5005, Australia
| | - J. Gummow
- Gene Silencing and Expression Core Facility, Adelaide Health and Medical Sciences, Robinson Research Institute, University of Adelaide, Adelaide, SA 5005, Australia
| | - J. Carr
- Microbiology and Infectious Diseases, College of Medicine and Public Health, Flinders University, Adelaide, SA 5042, Australia
| | - S. A. Robertson
- Robinson Research Institute, School of Medicine, University of Adelaide, Adelaide, SA 5005, Australia
| | - J. D. Hayball
- Experimental Therapeutics Laboratory, Cancer Research Institute, School of Pharmacy and Medical Science, University of South Australia, Adelaide, SA 5000, Australia
- Robinson Research Institute, School of Medicine, University of Adelaide, Adelaide, SA 5005, Australia
| | - D. H. Barouch
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA
| | - E. J. Gowans
- Discipline of Surgery, University of Adelaide and Basil Hetzel Institute for Translational Health Research, Adelaide, SA 5005, Australia
| |
Collapse
|
46
|
Annamalai AS, Pattnaik A, Sahoo BR, Guinn ZP, Bullard BL, Weaver EA, Steffen D, Natarajan SK, Petro TM, Pattnaik AK. An Attenuated Zika Virus Encoding Non-Glycosylated Envelope (E) and Non-Structural Protein 1 (NS1) Confers Complete Protection against Lethal Challenge in a Mouse Model. Vaccines (Basel) 2019; 7:vaccines7030112. [PMID: 31547297 PMCID: PMC6789518 DOI: 10.3390/vaccines7030112] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 08/30/2019] [Accepted: 09/07/2019] [Indexed: 12/15/2022] Open
Abstract
Zika virus (ZIKV), a mosquito-transmitted flavivirus, emerged in the last decade causing serious human diseases, including congenital microcephaly in newborns and Guillain-Barré syndrome in adults. Although many vaccine platforms are at various stages of development, no licensed vaccines are currently available. Previously, we described a mutant MR766 ZIKV (m2MR) bearing an E protein mutation (N154A) that prevented its glycosylation, resulting in attenuation and defective neuroinvasion. To further attenuate m2MR for its potential use as a live viral vaccine, we incorporated additional mutations into m2MR by substituting the asparagine residues in the glycosylation sites (N130 and N207) of NS1 with alanine residues. Examination of pathogenic properties revealed that the virus (m5MR) carrying mutations in E (N154A) and NS1 (N130A and N207A) was fully attenuated with no disease signs in infected mice, inducing high levels of humoral and cell-mediated immune responses, and protecting mice from subsequent lethal virus challenge. Furthermore, passive transfer of sera from m5MR-infected mice into naïve animals resulted in complete protection from lethal challenge. The immune sera from m5MR-infected animals neutralized both African and Asian lineage viruses equally well, suggesting that m5MR virus could be developed as a potentially broad live virus vaccine candidate.
Collapse
Affiliation(s)
- Arun S Annamalai
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA.
- Nebraska Center for Virology, University of Nebraska-Lincoln, Lincoln, NE 68583, USA.
| | - Aryamav Pattnaik
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA.
- Nebraska Center for Virology, University of Nebraska-Lincoln, Lincoln, NE 68583, USA.
| | - Bikash R Sahoo
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA.
- Nebraska Center for Virology, University of Nebraska-Lincoln, Lincoln, NE 68583, USA.
| | - Zack P Guinn
- Department of Oral Biology, University of Nebraska Medical Center, Lincoln, NE 68583, USA.
| | - Brianna L Bullard
- Nebraska Center for Virology, University of Nebraska-Lincoln, Lincoln, NE 68583, USA.
- School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA.
| | - Eric A Weaver
- Nebraska Center for Virology, University of Nebraska-Lincoln, Lincoln, NE 68583, USA.
- School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA.
| | - David Steffen
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA.
| | - Sathish Kumar Natarajan
- Nebraska Center for Virology, University of Nebraska-Lincoln, Lincoln, NE 68583, USA.
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA.
| | - Thomas M Petro
- Nebraska Center for Virology, University of Nebraska-Lincoln, Lincoln, NE 68583, USA.
- Department of Oral Biology, University of Nebraska Medical Center, Lincoln, NE 68583, USA.
| | - Asit K Pattnaik
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA.
- Nebraska Center for Virology, University of Nebraska-Lincoln, Lincoln, NE 68583, USA.
| |
Collapse
|
47
|
Fathi A, Dahlke C, Addo MM. Recombinant vesicular stomatitis virus vector vaccines for WHO blueprint priority pathogens. Hum Vaccin Immunother 2019; 15:2269-2285. [PMID: 31368826 PMCID: PMC6816421 DOI: 10.1080/21645515.2019.1649532] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
The devastating Ebola virus (EBOV) outbreak in West Africa in 2013-2016 has flagged the need for the timely development of vaccines for high-threat pathogens. To be better prepared for new epidemics, the WHO has compiled a list of priority pathogens that are likely to cause future outbreaks and for which R&D efforts are, therefore, paramount (R&D Blueprint: https://www.who.int/blueprint/priority-diseases/en/ ). To this end, the detailed characterization of vaccine platforms is needed. The vesicular stomatitis virus (VSV) has been established as a robust vaccine vector backbone for infectious diseases for well over a decade. The recent clinical trials testing the vaccine candidate VSV-EBOV against EBOV disease now have added a substantial amount of clinical data and suggest VSV to be an ideal vaccine vector candidate for outbreak pathogens. In this review, we discuss insights gained from the clinical VSV-EBOV vaccine trials as well as from animal studies investigating vaccine candidates for Blueprint pathogens.
Collapse
Affiliation(s)
- Anahita Fathi
- Department of Medicine, Division of Infectious Diseases, University Medical-Center Hamburg-Eppendorf , Hamburg , Germany.,Department for Clinical Immunology of Infectious Diseases, Bernhard Nocht Institute for Tropical Medicine , Hamburg , Germany.,German Center for Infection Research, Hamburg-Lübeck-Borstel-Riems , Germany
| | - Christine Dahlke
- Department of Medicine, Division of Infectious Diseases, University Medical-Center Hamburg-Eppendorf , Hamburg , Germany.,Department for Clinical Immunology of Infectious Diseases, Bernhard Nocht Institute for Tropical Medicine , Hamburg , Germany.,German Center for Infection Research, Hamburg-Lübeck-Borstel-Riems , Germany
| | - Marylyn M Addo
- Department of Medicine, Division of Infectious Diseases, University Medical-Center Hamburg-Eppendorf , Hamburg , Germany.,Department for Clinical Immunology of Infectious Diseases, Bernhard Nocht Institute for Tropical Medicine , Hamburg , Germany.,German Center for Infection Research, Hamburg-Lübeck-Borstel-Riems , Germany
| |
Collapse
|
48
|
Muruato AE, Shan C, Fontes-Garfias CR, Liu Y, Cao Z, Gao Q, Weaver SC, Shi PY. Genetic stability of live-attenuated Zika vaccine candidates. Antiviral Res 2019; 171:104596. [PMID: 31493417 DOI: 10.1016/j.antiviral.2019.104596] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 08/30/2019] [Accepted: 09/02/2019] [Indexed: 12/28/2022]
Abstract
Zika virus (ZIKV) has drawn global attention as the etiologic agent of Zika Congenital Syndrome in babies born to infected pregnant women. To prevent future ZIKV outbreaks and protect persons at risk for severe disease, we developed two live-attenuated vaccine (LAV) candidates containing 10- or 20-nucleotide deletions in the 3'UTR of the viral genome (Δ10 and Δ20). After a single-dose immunization, both Δ10 and Δ20 LAVs protected mice and non-human primates against ZIKV infection. Here, we characterized the stability, safety, and efficacy of the LAVs after continuously culturing them on manufacture Vero cells for ten rounds. Whole genome sequencing showed that passage 10 (P10) LAVs retained the engineered Δ10 and Δ20 deletions; one to four additional mutations emerged at different regions of the genome. In A129 mice, the P10 LAVs exhibited viremia higher than the un-passaged LAVs, but lower than wild-type ZIKV; unlike wild-type ZIKV-infected mice, none of the P10 LAV-infected mice developed disease or death, demonstrating that the P10 LAVs remained attenuated. Mice immunized with a single dose of the P10 LAVs developed robust neutralizing antibody titers (1/1,000 to 1/10,000) and were protected against epidemic ZIKV challenge. The P10 LAVs did not exhibit increased neurovirulence. Intracranial inoculation of one-day-old CD1 pups with 103 focus-forming units of the P10 Δ10 and Δ20 LAVs resulted in 100% and ≥80% survival, respectively. Furthermore, the P10 LAVs remained incompetent in infecting Aedes aegypti mosquitoes after intrathoracic microinjection. Our results support the phenotypic stability and further development of these promising LAVs for ZIKV.
Collapse
Affiliation(s)
- Antonio E Muruato
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX, USA; Institute for Human Infections & Immunity, University of Texas Medical Branch, Galveston, TX, USA; Institute for Translational Science, University of Texas Medical Branch, Galveston, TX, USA
| | - Chao Shan
- Department of Biochemistry & Molecular Biology, University of Texas Medical Branch, Galveston, TX, USA
| | - Camila R Fontes-Garfias
- Department of Biochemistry & Molecular Biology, University of Texas Medical Branch, Galveston, TX, USA
| | - Yang Liu
- Department of Biochemistry & Molecular Biology, University of Texas Medical Branch, Galveston, TX, USA
| | - Zengguo Cao
- Department of Biochemistry & Molecular Biology, University of Texas Medical Branch, Galveston, TX, USA
| | - Qiang Gao
- Sinovac Bioteck Co., Ltd., Beijing, China
| | - Scott C Weaver
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX, USA; Institute for Human Infections & Immunity, University of Texas Medical Branch, Galveston, TX, USA; Institute for Translational Science, University of Texas Medical Branch, Galveston, TX, USA; Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, TX, USA; Sealy Center for Structural Biology & Molecular Biophysics, University of Texas Medical Branch, Galveston, TX, USA
| | - Pei-Yong Shi
- Institute for Human Infections & Immunity, University of Texas Medical Branch, Galveston, TX, USA; Institute for Translational Science, University of Texas Medical Branch, Galveston, TX, USA; Department of Biochemistry & Molecular Biology, University of Texas Medical Branch, Galveston, TX, USA; Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, TX, USA; Sealy Center for Structural Biology & Molecular Biophysics, University of Texas Medical Branch, Galveston, TX, USA.
| |
Collapse
|
49
|
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
|
50
|
Hu Y, Sun L. Systematic Analysis of Structure Similarity between Zika Virus and Other Flaviviruses. ACS Infect Dis 2019; 5:1070-1080. [PMID: 31038920 DOI: 10.1021/acsinfecdis.9b00047] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Zika virus (ZIKV) infection has caused global concern because of its association with fetal microcephaly and serious neurological complications in adults since 2016. Currently, no specific anti-ZIKV therapy is available to control ZIKV infection. During the last couple of years, the intensive investigation of ZIKV structure has provided significant information for structure-based vaccine and drug design. In this review, we summarized the research progress on the structures of ZIKV and its component proteins. We analyzed the structure identity and the differences between ZIKV and other flaviviruses. This information is crucial to guiding structure-based anti-ZIKV inhibitors and vaccine discovery.
Collapse
Affiliation(s)
- Yuxia Hu
- The Fifth People’s Hospital of Shanghai and Institutes of Biomedical Sciences, Fudan University, 131 Dongan Road, Shanghai 20032, China
| | - Lei Sun
- The Fifth People’s Hospital of Shanghai and Institutes of Biomedical Sciences, Fudan University, 131 Dongan Road, Shanghai 20032, China
| |
Collapse
|