1
|
Gloanec N, Guyard-Nicodème M, Chemaly M, Dory D. Reverse vaccinology: A strategy also used for identifying potential vaccine antigens in poultry. Vaccine 2025; 48:126756. [PMID: 39855107 DOI: 10.1016/j.vaccine.2025.126756] [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: 11/07/2024] [Revised: 01/13/2025] [Accepted: 01/15/2025] [Indexed: 01/27/2025]
Abstract
Vaccination of livestock plays a major role in improving animal health, welfare and productivity, but also in public health by preventing zoonotic diseases. Advances in bioinformatics and whole-genome sequencing techniques since the 2000s have led to the development of genome-based vaccinology, called reverse vaccinology. Reverse vaccinology is a rapid and competitive strategy that uses pathogen genome sequences to screen for and identify potential vaccine antigens and, unlike conventional methods, does not require culturing the pathogenic microorganism, at least initially. Based on in silico approaches and dedicated software, reverse vaccinology has led to the identification of a wide range of proteins as putative vaccine candidates against human pathogens and has been applied more recently to several animal diseases. After a brief overview of the principle of the approach and its applications in human medicine, this review focuses on the use of reverse vaccinology for the development of vaccines specifically for poultry, a representative example of livestock vaccination, and discusses the important points to consider when using this method.
Collapse
Affiliation(s)
- Noémie Gloanec
- GVB-Viral Genetics and Biosafety Unit, French Agency for Food, Environmental and Occupational Health & Safety (ANSES), France; HQPAP-Unit of Hygiene and Quality of Poultry and Pork Products, French Agency for Food, Environmental and Occupational Health & Safety (ANSES), France; UFR of Life Sciences Environment, University of Rennes 1, 35065 Rennes, France
| | - Muriel Guyard-Nicodème
- HQPAP-Unit of Hygiene and Quality of Poultry and Pork Products, French Agency for Food, Environmental and Occupational Health & Safety (ANSES), France.
| | - Marianne Chemaly
- HQPAP-Unit of Hygiene and Quality of Poultry and Pork Products, French Agency for Food, Environmental and Occupational Health & Safety (ANSES), France.
| | - Daniel Dory
- GVB-Viral Genetics and Biosafety Unit, French Agency for Food, Environmental and Occupational Health & Safety (ANSES), France; VIPAC-Avian and Rabbit Virology, Immunology and Parasitology Unit, French Agency for Food, Environmental and Occupational Health & Safety (ANSES), France.
| |
Collapse
|
2
|
Olaya-Bravo K, Martínez-Flores D, Rodríguez-Hernández AP, Tobías-Juárez I, Castro-Rodríguez JA, Sampieri A, Vaca L. Resolving viral structural complexity by super-resolution microscopy. Arch Virol 2024; 170:5. [PMID: 39652240 DOI: 10.1007/s00705-024-06192-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 10/08/2024] [Indexed: 12/17/2024]
Abstract
In this review, we discuss different super-resolution microscopy (SRM) techniques employed to study viral structures and virus composition with nanometric resolution. We describe the basic principles of the different microscopy methods utilized to break the light diffraction limit, enabling the study of protein composition in viral structures. Finally, we demonstrate for the first time the differential spatial distribution of two structural proteins in an individual baculovirus using single-molecule super-resolution microscopy. We discuss the future of these powerful methods for virology, medicine, and biotechnology applications.
Collapse
Affiliation(s)
- Kevin Olaya-Bravo
- Departamento de BIologia Celular y del Desarrollo. Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico
| | - Daniel Martínez-Flores
- Departamento de BIologia Celular y del Desarrollo. Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico
| | - Aaron Pavel Rodríguez-Hernández
- Departamento de BIologia Celular y del Desarrollo. Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico
| | - Ileana Tobías-Juárez
- Departamento de BIologia Celular y del Desarrollo. Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico
| | - Jorge A Castro-Rodríguez
- Departamento de BIologia Celular y del Desarrollo. Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico
| | - Alicia Sampieri
- Departamento de BIologia Celular y del Desarrollo. Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico
| | - Luis Vaca
- Departamento de BIologia Celular y del Desarrollo. Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico.
| |
Collapse
|
3
|
Khudainazarova NS, Granovskiy DL, Kondakova OA, Ryabchevskaya EM, Kovalenko AO, Evtushenko EA, Arkhipenko MV, Nikitin NA, Karpova OV. Prokaryote- and Eukaryote-Based Expression Systems: Advances in Post-Pandemic Viral Antigen Production for Vaccines. Int J Mol Sci 2024; 25:11979. [PMID: 39596049 PMCID: PMC11594041 DOI: 10.3390/ijms252211979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 11/01/2024] [Accepted: 11/02/2024] [Indexed: 11/28/2024] Open
Abstract
This review addresses the ongoing global challenge posed by emerging and evolving viral diseases, underscoring the need for innovative vaccine development strategies. It focuses on the modern approaches to creating vaccines based on recombinant proteins produced in different expression systems, including bacteria, yeast, plants, insects, and mammals. This review analyses the advantages, limitations, and applications of these expression systems for producing vaccine antigens, as well as strategies for designing safer, more effective, and potentially 'universal' antigens. The review discusses the development of vaccines for a range of viral diseases, excluding SARS-CoV-2, which has already been extensively studied. The authors present these findings with the aim of contributing to ongoing research and advancing the development of antiviral vaccines.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Nikolai A. Nikitin
- Department of Virology, Faculty of Biology, Lomonosov Moscow State University, 119991 Moscow, Russia; (N.S.K.); (D.L.G.); (O.A.K.); (E.M.R.); (A.O.K.); (E.A.E.); (M.V.A.); (O.V.K.)
| | | |
Collapse
|
4
|
Cheng X, Zhao W, Liang G, Lu H, Fu Y, Li Y, Cui F. Construction of cytomegalovirus promoter-driven gene expression system in Laodelphax striatellus. INSECT SCIENCE 2024; 31:720-732. [PMID: 38339806 DOI: 10.1111/1744-7917.13333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 12/18/2023] [Accepted: 01/08/2024] [Indexed: 02/12/2024]
Abstract
The small brown planthopper (SBPH, Laodelphax striatellus) is a significant rice pest, responsible for transmitting rice stripe virus (RSV) in a persistent and propagative manner. RSV is one of the most detrimental rice viruses, causing rice stripe disease, which results in considerable loss of rice grain yield. While RNA interference and gene knockout techniques have enabled gene downregulation in SBPH, no system currently exists for the overexpression of endogenous or exogenous genes. Consequently, the development of a protein expression system for SBPH is imperative to serve as a technical foundation for pest control and gene function investigations. This study aimed to construct an expression vector using the promoter of the constitutive-expressed tubulin gene of SBPH, and promoter of human cytomegalovirus (CMV). Fluorescence experiments demonstrated that both tubulin and CMV promoter could drive green fluorescent protein (GFP) expression in SBPH, and could also facilitate the expression of a nucleocapsid protein (NP) -GFP fusion protein containing viral NP with comparable efficiency. Through expression vector optimization, we have identified that the 3 tandem CMV promoters display a significantly higher promoter activity compared with both the 2 tandem CMV promoters and the single CMV promoter. In addition, the incorporation of Star polycation nanoparticles significantly enhanced the expression efficiency in SBPH. These results provide a promising technical platform for investigating gene functions in SBPH.
Collapse
Affiliation(s)
- Xiaohui Cheng
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Wan Zhao
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Guohua Liang
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- School of Life Sciences, Hebei University, Baoding, Hebei, China
| | - Hong Lu
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yumei Fu
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yiming Li
- School of Life Sciences, Hebei University, Baoding, Hebei, China
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Institute of Wetland Ecology & Clone Ecology/Zhejiang Provincial Key Laboratory of Plant Evolutionary Ecology and Conservation, Taizhou University, Taizhou, Zhejiang, China
| | - Feng Cui
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
5
|
Fan G, Li Y, Ye Q, Niu Q, Zhao X, Chen L, Gu Q, Zhang Y, Wei X, Wu S, Wu Q, Wu Y. Animal-derived free hydrolysate in animal cell culture: Current research and application advances. J Tissue Eng 2024; 15:20417314241300388. [PMID: 39649943 PMCID: PMC11624555 DOI: 10.1177/20417314241300388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 11/01/2024] [Indexed: 12/11/2024] Open
Abstract
Fetal bovine serum (FBS) plays a crucial role in the composition of animal cell culture medium. However, conventional serum-based medium face numerous challenges. The use of animal-derived free hydrolysate (ADFH) has garnered significant attention in research and applications as a viable alternative to FBS-containing medium in animal cell culture. This article provides a comprehensive overview of the effects, mechanisms of action, and applications of ADFH in animal cell culture. ADFH serves as an effective substitute for FBS-containing medium, enhancing various cellular processes, including cell proliferation, viability, protein synthesis, production, survival, and stability. Several mechanisms of action for ADFH have been elucidated through scientific investigations, such as nutrient provision, activation of signaling pathways, regulation of protein synthesis and folding, protection against oxidative damage and apoptosis, as well as cell cycle regulation. Researches and applications of ADFH represent a promising approach to overcoming the limitations of FBS-containing medium and advancing the field of animal cell culture. This review provides a theoretical foundation for promoting the development of sustainable and alternative hydrolysates, as well as the continued progress of animal cell culture.
Collapse
Affiliation(s)
- Guanghan Fan
- National Health Commission Science and Technology Innovation Platform for Nutrition and Safety of Microbial Food, Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
- Department of Food Science and Engineering, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China
| | - Ying Li
- National Health Commission Science and Technology Innovation Platform for Nutrition and Safety of Microbial Food, Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Qinghua Ye
- National Health Commission Science and Technology Innovation Platform for Nutrition and Safety of Microbial Food, Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Qinya Niu
- National Health Commission Science and Technology Innovation Platform for Nutrition and Safety of Microbial Food, Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
- Department of Food Science and Engineering, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China
| | - Xinyu Zhao
- Guangdong Kehuan Biological Science and Technology Co. Ltd., Guangzhou, Guangdong, China
| | - Ling Chen
- National Health Commission Science and Technology Innovation Platform for Nutrition and Safety of Microbial Food, Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Qihui Gu
- National Health Commission Science and Technology Innovation Platform for Nutrition and Safety of Microbial Food, Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Youxiong Zhang
- National Health Commission Science and Technology Innovation Platform for Nutrition and Safety of Microbial Food, Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Xianhu Wei
- National Health Commission Science and Technology Innovation Platform for Nutrition and Safety of Microbial Food, Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Shi Wu
- National Health Commission Science and Technology Innovation Platform for Nutrition and Safety of Microbial Food, Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Qingping Wu
- National Health Commission Science and Technology Innovation Platform for Nutrition and Safety of Microbial Food, Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
- Department of Food Science and Engineering, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China
| | - Yuwei Wu
- National Health Commission Science and Technology Innovation Platform for Nutrition and Safety of Microbial Food, Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| |
Collapse
|
6
|
Chauhan S, Khasa YP. Challenges and Opportunities in the Process Development of Chimeric Vaccines. Vaccines (Basel) 2023; 11:1828. [PMID: 38140232 PMCID: PMC10747103 DOI: 10.3390/vaccines11121828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 07/22/2023] [Accepted: 08/04/2023] [Indexed: 12/24/2023] Open
Abstract
Vaccines are integral to human life to protect them from life-threatening diseases. However, conventional vaccines often suffer limitations like inefficiency, safety concerns, unavailability for non-culturable microbes, and genetic variability among pathogens. Chimeric vaccines combine multiple antigen-encoding genes of similar or different microbial strains to protect against hyper-evolving drug-resistant pathogens. The outbreaks of dreadful diseases have led researchers to develop economical chimeric vaccines that can cater to a large population in a shorter time. The process development begins with computationally aided omics-based approaches to design chimeric vaccines. Furthermore, developing these vaccines requires optimizing upstream and downstream processes for mass production at an industrial scale. Owing to the complex structures and complicated bioprocessing of evolving pathogens, various high-throughput process technologies have come up with added advantages. Recent advancements in high-throughput tools, process analytical technology (PAT), quality-by-design (QbD), design of experiments (DoE), modeling and simulations, single-use technology, and integrated continuous bioprocessing have made scalable production more convenient and economical. The paradigm shift to innovative strategies requires significant attention to deal with major health threats at the global scale. This review outlines the challenges and emerging avenues in the bioprocess development of chimeric vaccines.
Collapse
Affiliation(s)
| | - Yogender Pal Khasa
- Department of Microbiology, University of Delhi South Campus, New Delhi 110021, India;
| |
Collapse
|
7
|
Zhang R, Duan X, Liu Y, Xu J, Al-bashari AAG, Ye P, Ye Q, He Y. The Application of Mesenchymal Stem Cells in Future Vaccine Synthesis. Vaccines (Basel) 2023; 11:1631. [PMID: 38005963 PMCID: PMC10675160 DOI: 10.3390/vaccines11111631] [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: 09/17/2023] [Revised: 10/09/2023] [Accepted: 10/20/2023] [Indexed: 11/26/2023] Open
Abstract
Vaccines have significant potential in treating and/or preventing diseases, yet there remain challenges in developing effective vaccines against some diseases, such as AIDS and certain tumors. Mesenchymal stem cells (MSCs), a subset of cells with low immunogenicity, high proliferation potential, and an abundant source of extracellular vesicles (EVs), represent one of the novel and promising vaccine platforms. This review describes the unique features and potential mechanisms of MSCs as a novel vaccine platform. We also cover aspects such as the safety and stability of MSCs that warrant future in-depth studies.
Collapse
Affiliation(s)
- Rui Zhang
- Center of Regenerative Medicine & Department of Stomatology, Renmin Hospital of Wuhan University, Wuhan 430060, China; (R.Z.); (X.D.); (Y.L.); (A.A.G.A.-b.)
| | - Xingxiang Duan
- Center of Regenerative Medicine & Department of Stomatology, Renmin Hospital of Wuhan University, Wuhan 430060, China; (R.Z.); (X.D.); (Y.L.); (A.A.G.A.-b.)
| | - Ye Liu
- Center of Regenerative Medicine & Department of Stomatology, Renmin Hospital of Wuhan University, Wuhan 430060, China; (R.Z.); (X.D.); (Y.L.); (A.A.G.A.-b.)
| | - Jia Xu
- Australian Rivers Institute and School of Environment and Science, Griffith University, Nathan, QLD 4111, Australia;
| | - Abdullkhaleg Ali Ghaleb Al-bashari
- Center of Regenerative Medicine & Department of Stomatology, Renmin Hospital of Wuhan University, Wuhan 430060, China; (R.Z.); (X.D.); (Y.L.); (A.A.G.A.-b.)
| | - Peng Ye
- Department of Pharmacy, Renmin Hospital of Wuhan University, Wuhan 430060, China;
| | - Qingsong Ye
- Center of Regenerative Medicine & Department of Stomatology, Renmin Hospital of Wuhan University, Wuhan 430060, China; (R.Z.); (X.D.); (Y.L.); (A.A.G.A.-b.)
| | - Yan He
- Institute of Regenerative and Translational Medicine, Department of Stomatology, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
8
|
Ma C, Li X, Ding W, Zhang X, Chen H, Feng Y. Effects of hTERT transfection on the telomere and telomerase of Periplaneta americana cells in vitro. AMB Express 2023; 13:118. [PMID: 37864620 PMCID: PMC10590340 DOI: 10.1186/s13568-023-01624-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Accepted: 10/03/2023] [Indexed: 10/23/2023] Open
Abstract
Telomere and telomerase are crucial factors in cell division and chromosome stability. Telomerase activity in most cells depends on the transcription control by the telomerase reverse transcriptase (TERT). The introduction of an exogenous human TERT (hTERT) in cultured cells could enhance telomerase activity and elongate the lifespan of various cells. Telomere elongation mechanisms vary between insects and are complex and unusual. Whether the use of exogenous hTERT can immortalize primary insect cells remains to be investigated. In this study, we used a recombinant virus expressing hTERT to infect primary cultured cells of Periplaneta americana and evaluated its effects on insect cell immortalization. We found that hTERT was successfully expressed and promoted the growth of P. americana cells, shortening their doubling time. This was due to the ability of hTERT to increase the activity of telomerase in P. americana cells, thus prolonging the telomeres. Our study lays the foundation for understanding the mechanisms of telomere elongation in P. americana, and suggests that the introduction of hTERT into insect cells could be an efficient way to establish certain insect cell lines.
Collapse
Affiliation(s)
- Chenjing Ma
- Key Laboratory of Breeding and Utilization of Resource Insects of National Forestry and Grassland Administration, Institute of Highland Forest Science, Chinese Academy of Forestry, Kunming, Yunnan Province, 650224, China
- Nanjing Forestry University, Nanjing, Jiangsu Province, 210037, China
| | - Xian Li
- Key Laboratory of Breeding and Utilization of Resource Insects of National Forestry and Grassland Administration, Institute of Highland Forest Science, Chinese Academy of Forestry, Kunming, Yunnan Province, 650224, China
| | - Weifeng Ding
- Key Laboratory of Breeding and Utilization of Resource Insects of National Forestry and Grassland Administration, Institute of Highland Forest Science, Chinese Academy of Forestry, Kunming, Yunnan Province, 650224, China
| | - Xin Zhang
- Key Laboratory of Breeding and Utilization of Resource Insects of National Forestry and Grassland Administration, Institute of Highland Forest Science, Chinese Academy of Forestry, Kunming, Yunnan Province, 650224, China.
| | - Hang Chen
- Key Laboratory of Breeding and Utilization of Resource Insects of National Forestry and Grassland Administration, Institute of Highland Forest Science, Chinese Academy of Forestry, Kunming, Yunnan Province, 650224, China
| | - Ying Feng
- Key Laboratory of Breeding and Utilization of Resource Insects of National Forestry and Grassland Administration, Institute of Highland Forest Science, Chinese Academy of Forestry, Kunming, Yunnan Province, 650224, China
| |
Collapse
|
9
|
Ahata B, Akçapınar GB. CCHFV vaccine development, current challenges, limitations, and future directions. Front Immunol 2023; 14:1238882. [PMID: 37753088 PMCID: PMC10518622 DOI: 10.3389/fimmu.2023.1238882] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 08/22/2023] [Indexed: 09/28/2023] Open
Abstract
Crimean-Congo hemorrhagic fever (CCHF) is the most prevalent tick-borne viral disease affecting humans. The disease is life-threatening in many regions of the developing world, including Africa, Asia, the Middle East, and Southern Europe. In line with the rapidly increasing disease prevalence, various vaccine strategies are under development. Despite a large number of potential vaccine candidates, there are no approved vaccines as of yet. This paper presents a detailed comparative analysis of current efforts to develop vaccines against CCHFV, limitations associated with current efforts, and future research directions.
Collapse
Affiliation(s)
- Büşra Ahata
- Department of Medical Biotechnology, Institute of Health Sciences, Acıbadem Mehmet Ali Aydınlar University, Istanbul, Türkiye
- Health Institutes of Turkey, Istanbul, Türkiye
| | - Günseli Bayram Akçapınar
- Department of Medical Biotechnology, Institute of Health Sciences, Acıbadem Mehmet Ali Aydınlar University, Istanbul, Türkiye
| |
Collapse
|
10
|
Alenton RRR, Mai HN, Dhar AK. Engineering a replication-incompetent viral vector for the delivery of therapeutic RNA in crustaceans. PNAS NEXUS 2023; 2:pgad278. [PMID: 37693213 PMCID: PMC10485883 DOI: 10.1093/pnasnexus/pgad278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 07/05/2023] [Accepted: 08/15/2023] [Indexed: 09/12/2023]
Abstract
Viral disease pandemics are a major cause of economic losses in crustacean farming worldwide. While RNA interference (RNAi)-based therapeutics have shown promise at a laboratory scale, without an effective oral delivery platform, RNA-based therapy will not reach its potential against controlling viral diseases in crustaceans. Using a reverse-engineered shrimp RNA virus, Macrobrachium rosenbergii nodavirus (MrNV), we have developed a shrimp viral vector for delivering an engineered RNA cargo. By replacing the RNA-dependent RNA polymerase (RdRp) protein-coding region of MrNV with a cargo RNA encoding green fluorescent protein (GFP) as a proof-of-concept, we generated a replication-incompetent mutant MrNV(ΔRdRp) carrying the GFP RNA cargo resulting in MrNV(ΔRdRp)-GFP. Upon incorporating MrNV(ΔRdRp)-GFP in the diet of the marine Pacific white shrimp (Penaeus vannamei), MrNV(ΔRdRp) particles were visualized in hemocytes demonstrating successful vector internalization. Fluorescence imaging of hemocytes showed the expression of GFP protein and the MrNV capsid RNA (RNA2) as well as the incorporated GFP RNA cargo. Detection of cargo RNA in hepatopancreas and pleopods indicated the systemic spread of the viral vector. The quantitative load of both the MrNV RNA2 and GFP RNA progressively diminished within 8 days postadministration of the viral vector, which indicated a lack of MrNV(ΔRdRp)-GFP replication in shrimp. In addition, no pathological hallmarks of the wild-type MrNV infection were detected using histopathology in the target tissue of treated shrimp. The data unequivocally demonstrated the successful engineering of a replication-incompetent viral vector for RNA delivery, paving the way for the oral delivery of antiviral therapeutics in farmed crustaceans.
Collapse
Affiliation(s)
- Rod Russel R Alenton
- Aquaculture Pathology Laboratory, School of Animal and Comparative Biomedical Sciences, The University of Arizona, Tucson, AZ 85721, USA
| | - Hung N Mai
- Aquaculture Pathology Laboratory, School of Animal and Comparative Biomedical Sciences, The University of Arizona, Tucson, AZ 85721, USA
| | - Arun K Dhar
- Aquaculture Pathology Laboratory, School of Animal and Comparative Biomedical Sciences, The University of Arizona, Tucson, AZ 85721, USA
| |
Collapse
|
11
|
Honda-Okubo Y, Sakala IG, André G, Tarbet EB, Hurst BL, Petrovsky N. An Advax-CpG55.2 adjuvanted recombinant hemagglutinin vaccine provides immunity against H7N9 influenza in adult and neonatal mice. Vaccine 2023; 41:5592-5602. [PMID: 37532610 DOI: 10.1016/j.vaccine.2023.07.061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 07/25/2023] [Accepted: 07/26/2023] [Indexed: 08/04/2023]
Abstract
There is a major unmet need for strategies to improve the immunogenicity and effectiveness of pandemic influenza vaccines, particularly in poor responder populations such as neonates. Recombinant protein approaches to pandemic influenza offer advantages over more traditional inactivated virus approaches, as they are free of problems such as egg adaptation or need for high level biosecurity containment for manufacture. However, a weakness of recombinant proteins is their low immunogenicity. We asked whether the use of an inulin polysaccharide adjuvant (Advax) alone or combined with a TLR9 agonist (CpG55.2) would enhance the immunogenicity and protection of a recombinant hemagglutinin vaccine against H7N9 influenza (rH7HA), including in neonatal mice. Advax adjuvant induced predominantly IgG1 responses against H7HA, whereas Advax-CpG55.2 adjuvant also induced IgG2a, IgG2b and IgG3 responses, consistent with the TLR9 agonist component inducing a Th1 bias. Advax-CpG55.2 adjuvanted rH7HA induced high serum neutralizing antibody titers in adult mice. In newborns it similarly overcame immune hypo-responsiveness and enhanced serum anti-rH7HA IgG levels in 7-day-old BALB/C and C57BL/6 mice. Immunized adult mice were protected against a lethal H7N9 virus challenge. When formulated with Advax-CpG55.2 adjuvant, greater protection was seen with rH7HA than with inactivated H7 whole virus antigen. Advax-CpG55.2 adjuvanted rH7HA represents a promising influenza vaccine platform for further development.
Collapse
Affiliation(s)
- Yoshikazu Honda-Okubo
- Vaxine Pty Ltd, Bedford Park, Adelaide, SA 5042, Australia; Flinders University, Bedford Park, Adelaide, SA 5042, Australia
| | - Isaac G Sakala
- Vaxine Pty Ltd, Bedford Park, Adelaide, SA 5042, Australia; Flinders University, Bedford Park, Adelaide, SA 5042, Australia
| | | | - E Bart Tarbet
- Institute for Antiviral Research, Department of Animal, Dairy, and Veterinary Sciences, 5600 Old Main Hill, Utah State University, Logan, UT 84322, USA
| | - Brett L Hurst
- Institute for Antiviral Research, Department of Animal, Dairy, and Veterinary Sciences, 5600 Old Main Hill, Utah State University, Logan, UT 84322, USA
| | | |
Collapse
|
12
|
Soto ER, Specht CA, Lee CK, Levitz SM, Ostroff GR. One Step Purification-Vaccine Delivery System. Pharmaceutics 2023; 15:pharmaceutics15051390. [PMID: 37242632 DOI: 10.3390/pharmaceutics15051390] [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: 03/28/2023] [Revised: 04/24/2023] [Accepted: 04/27/2023] [Indexed: 05/28/2023] Open
Abstract
Glucan particles (GPs) are hollow, porous 3-5 µm microspheres derived from the cell walls of Baker's yeast (Saccharomyces cerevisiae). Their 1,3-β-glucan outer shell allows for receptor-mediated uptake by macrophages and other phagocytic innate immune cells expressing β-glucan receptors. GPs have been used for the targeted delivery of a wide range of payloads, including vaccines and nanoparticles, encapsulated inside the hollow cavity of GPs. In this paper, we describe the methods to prepare GP-encapsulated nickel nanoparticles (GP-Ni) for the binding of histidine (His)-tagged proteins. His-tagged Cda2 cryptococcal antigens were used as payloads to demonstrate the efficacy of this new GP vaccine encapsulation approach. The GP-Ni-Cda2 vaccine was shown to be comparable to our previous approach utilizing mouse serum albumin (MSA) and yeast RNA trapping of Cda2 in GPs in a mouse infection model. This novel GP-Ni approach allows for the one-step binding of His-tagged vaccine antigens and encapsulation in an effective delivery vehicle to target vaccines to antigen-presenting cells (APCs), antigen discovery, and vaccine development.
Collapse
Affiliation(s)
- Ernesto R Soto
- Program in Molecular Medicine, UMass Chan Medical School, Worcester, MA 01605, USA
| | - Charles A Specht
- Department of Medicine, UMass Chan Medical School, Worcester, MA 01605, USA
| | - Chrono K Lee
- Department of Medicine, UMass Chan Medical School, Worcester, MA 01605, USA
| | - Stuart M Levitz
- Department of Medicine, UMass Chan Medical School, Worcester, MA 01605, USA
| | - Gary R Ostroff
- Program in Molecular Medicine, UMass Chan Medical School, Worcester, MA 01605, USA
| |
Collapse
|
13
|
Chen C, Wang Z, Sun Z, Li W, Dimitrov DS. Development of an efficient method for selection of stable cell pools for protein expression and surface display with Expi293F cells. Cell Biochem Funct 2023; 41:355-364. [PMID: 36864545 DOI: 10.1002/cbf.3787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 02/18/2023] [Indexed: 03/04/2023]
Abstract
Compare with transient expression, stable cell lines generally have higher productivity and better quality for protein expression. However, selection of stable cell line is time-consuming and laborious. Here we describe an optimized selection method to achieve high-efficient stable cell pools with Expi293F suspension cells. This method only takes 2-3 weeks to generate stable cell pools with 2- to 10-fold higher productivity than transient gene expression (TGE). In fed-batch culture with Yeastolate, >1 g/L yield was achieved with our KTN0239-IgG stable cell pool in shaker flasks. This method can be also applied to efficiently display proteins on the cell surface.
Collapse
Affiliation(s)
- Chuan Chen
- Division of Infectious Diseases, Department of Medicine, Center for Antibody Therapeutics, University of Pittsburgh Medical School, Pittsburgh, Pennsylvania, USA
| | - Zening Wang
- Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Zehua Sun
- Division of Infectious Diseases, Department of Medicine, Center for Antibody Therapeutics, University of Pittsburgh Medical School, Pittsburgh, Pennsylvania, USA.,Abound Bio, Pittsburgh, Pennsylvania, USA
| | - Wei Li
- Division of Infectious Diseases, Department of Medicine, Center for Antibody Therapeutics, University of Pittsburgh Medical School, Pittsburgh, Pennsylvania, USA
| | - Dimiter S Dimitrov
- Division of Infectious Diseases, Department of Medicine, Center for Antibody Therapeutics, University of Pittsburgh Medical School, Pittsburgh, Pennsylvania, USA.,Abound Bio, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
14
|
Sullivan HM, Krupinska E, Rasmussen AA, Orozco Rodriguez JM, Knecht W. Recombinant Protein Production Using the Baculovirus Expression Vector System (BEVS). Methods Mol Biol 2023; 2652:55-77. [PMID: 37093470 DOI: 10.1007/978-1-0716-3147-8_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2023]
Abstract
The baculovirus expression vector system (BEVS) is one of the most popular eukaryotic systems for recombinant protein production. The focus of our protein production platform is the provision of recombinant proteins for research use, where generally only small quantities are required, in the range of tens of micrograms to a few hundred milligrams. Here, we present methods that reflect our standard operating procedures and setup to be able to frequently, and often repeatedly, produce many different types of proteins.
Collapse
Affiliation(s)
- Heather McDuffie Sullivan
- Lund Protein Production Platform (LP3) & Protein Production Sweden (PPS), Department of Biology, Lund University, Lund, Sweden
| | - Ewa Krupinska
- Lund Protein Production Platform (LP3) & Protein Production Sweden (PPS), Department of Biology, Lund University, Lund, Sweden
| | - Anna Andersson Rasmussen
- Lund Protein Production Platform (LP3) & Protein Production Sweden (PPS), Department of Biology, Lund University, Lund, Sweden
| | - Juan Manuel Orozco Rodriguez
- Lund Protein Production Platform (LP3) & Protein Production Sweden (PPS), Department of Biology, Lund University, Lund, Sweden
| | - Wolfgang Knecht
- Lund Protein Production Platform (LP3) & Protein Production Sweden (PPS), Department of Biology, Lund University, Lund, Sweden.
| |
Collapse
|
15
|
Pidre ML, Arrías PN, Amorós Morales LC, Romanowski V. The Magic Staff: A Comprehensive Overview of Baculovirus-Based Technologies Applied to Human and Animal Health. Viruses 2022; 15:80. [PMID: 36680120 PMCID: PMC9863858 DOI: 10.3390/v15010080] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/19/2022] [Accepted: 12/23/2022] [Indexed: 12/29/2022] Open
Abstract
Baculoviruses are enveloped, insect-specific viruses with large double-stranded DNA genomes. Among all the baculovirus species, Autographa californica multiple nucleopolyhedrovirus (AcMNPV) is the most studied. Due to its characteristics regarding biosafety, narrow host range and the availability of different platforms for modifying its genome, AcMNPV has become a powerful biotechnological tool. In this review, we will address the most widespread technological applications of baculoviruses. We will begin by summarizing their natural cycle both in larvae and in cell culture and how it can be exploited. Secondly, we will explore the different baculovirus-based protein expression systems (BEVS) and their multiple applications in the pharmaceutical and biotechnological industry. We will focus particularly on the production of vaccines, many of which are either currently commercialized or in advanced stages of development (e.g., Novavax, COVID-19 vaccine). In addition, recombinant baculoviruses can be used as efficient gene transduction and protein expression vectors in vertebrate cells (e.g., BacMam). Finally, we will extensively describe various gene therapy strategies based on baculoviruses applied to the treatment of different diseases. The main objective of this work is to provide an extensive up-to-date summary of the different biotechnological applications of baculoviruses, emphasizing the genetic modification strategies used in each field.
Collapse
Affiliation(s)
| | | | | | - Víctor Romanowski
- Instituto de Biotecnología y Biología Molecular (IBBM), Universidad Nacional de La Plata (UNLP) and Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), La Plata 1900, Argentina
| |
Collapse
|
16
|
Liu C, Wang M, Zhang H, Li C, Zhang T, Liu H, Zhu S, Chen J. Tumor microenvironment and immunotherapy of oral cancer. Eur J Med Res 2022; 27:198. [PMID: 36209263 PMCID: PMC9547678 DOI: 10.1186/s40001-022-00835-4] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 09/22/2022] [Indexed: 11/10/2022] Open
Abstract
Oral cancer is one of the most common malignant tumors of the head and neck, not only affects the appearance, but also affects eating and even endangers life. The clinical treatments of oral cancer mainly include surgery, radiotherapy, and chemotherapy. However, unsatisfactory therapeutic effect and toxic side effects are still the main problems in clinical treatment. Tumor microenvironment (TME) is not only closely related to the occurrence, growth, and metastasis of tumor but also works in the diagnosis, prevention, and treatment of tumor and prognosis. Future studies should continue to investigate the relationship of TME and oral cancer therapy. This purpose of this review was to analyze the characteristics of oral cancer microenvironment, summarize the traditional oral cancer therapy and immunotherapy strategies, and finally prospect the development prospects of oral cancer immunotherapy. Immunotherapy targeting tumor microenvironment is expected to provide a new strategy for clinical treatment of oral cancer.
Collapse
Affiliation(s)
- Chang Liu
- Hospital of Stomatology, Jilin University, Changchun, 130021, People's Republic of China
| | - Min Wang
- Hospital of Stomatology, Jilin University, Changchun, 130021, People's Republic of China
| | - Haiyang Zhang
- Hospital of Stomatology, Jilin University, Changchun, 130021, People's Republic of China
| | - Chunyan Li
- Hospital of Stomatology, Jilin University, Changchun, 130021, People's Republic of China
| | - Tianshou Zhang
- Hospital of Stomatology, Jilin University, Changchun, 130021, People's Republic of China
| | - Hong Liu
- Hospital of Stomatology, Jilin University, Changchun, 130021, People's Republic of China
| | - Song Zhu
- Hospital of Stomatology, Jilin University, Changchun, 130021, People's Republic of China.
| | - Jie Chen
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, People's Republic of China.
| |
Collapse
|
17
|
Wanarska M, Krajewska-Przybyszewska E, Wicka-Grochocka M, Cieśliński H, Pawlak-Szukalska A, Białkowska AM, Turkiewicz M, Florczak T, Gromek E, Krysiak J, Filipowicz N. A New Expression System Based on Psychrotolerant Debaryomyces macquariensis Yeast and Its Application to the Production of Cold-Active β-d-Galactosidase from Paracoccus sp. 32d. Int J Mol Sci 2022; 23:ijms231911691. [PMID: 36232994 PMCID: PMC9569826 DOI: 10.3390/ijms231911691] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 09/24/2022] [Accepted: 09/26/2022] [Indexed: 12/03/2022] Open
Abstract
Yeasts provide attractive host/vector systems for heterologous gene expression. The currently used yeast-based expression platforms include mesophilic and thermotolerant species. A eukaryotic expression system working at low temperatures could be particularly useful for the production of thermolabile proteins and proteins that tend to form insoluble aggregates. For this purpose, an expression system based on an Antarctic psychrotolerant yeast Debaryomyces macquariensis strain D50 that is capable of growing at temperatures ranging from 0 to 30 °C has been developed. The optimal physical culture conditions for D. macquariensis D50 in a fermenter are as follows: temperature 20 °C, pH 5.5, aeration rate of 1.5 vvm, and a stirring speed of 300 rpm. Four integrative plasmid vectors equipped with an expression cassette containing the constitutive GAP promoter and CYC1 transcriptional terminator from D. macquariensis D50 were constructed and used to clone and express a gene-encoding cold-active β-d-galactosidase of Paracoccus sp. 32d. The yield was 1150 U/L of recombinant yeast culture. Recombinant D. macquariensis D50 strains were mitotically stable under both selective and non-selective conditions. The D. macquariensis D50 host/vector system has been successfully utilized for the synthesis of heterologous thermolabile protein, and it can be an alternative to other microbial expression systems.
Collapse
Affiliation(s)
- Marta Wanarska
- Department of Molecular Biotechnology and Microbiology, Faculty of Chemistry, Gdansk University of Technology, Narutowicza 11/12, 80-233 Gdansk, Poland
- Correspondence:
| | - Ewelina Krajewska-Przybyszewska
- Department of Molecular Biotechnology and Microbiology, Faculty of Chemistry, Gdansk University of Technology, Narutowicza 11/12, 80-233 Gdansk, Poland
| | - Monika Wicka-Grochocka
- Department of Molecular Biotechnology and Microbiology, Faculty of Chemistry, Gdansk University of Technology, Narutowicza 11/12, 80-233 Gdansk, Poland
| | - Hubert Cieśliński
- Department of Molecular Biotechnology and Microbiology, Faculty of Chemistry, Gdansk University of Technology, Narutowicza 11/12, 80-233 Gdansk, Poland
| | - Anna Pawlak-Szukalska
- Department of Molecular Biotechnology and Microbiology, Faculty of Chemistry, Gdansk University of Technology, Narutowicza 11/12, 80-233 Gdansk, Poland
| | - Aneta M. Białkowska
- Institute of Molecular and Industrial Biotechnology, Faculty of Biotechnology and Food Sciences, Lodz University of Technology, Stefanowskiego 2/22, 90-573 Lodz, Poland
| | - Marianna Turkiewicz
- Institute of Molecular and Industrial Biotechnology, Faculty of Biotechnology and Food Sciences, Lodz University of Technology, Stefanowskiego 2/22, 90-573 Lodz, Poland
| | - Tomasz Florczak
- Institute of Molecular and Industrial Biotechnology, Faculty of Biotechnology and Food Sciences, Lodz University of Technology, Stefanowskiego 2/22, 90-573 Lodz, Poland
| | - Ewa Gromek
- Institute of Molecular and Industrial Biotechnology, Faculty of Biotechnology and Food Sciences, Lodz University of Technology, Stefanowskiego 2/22, 90-573 Lodz, Poland
| | - Joanna Krysiak
- Institute of Molecular and Industrial Biotechnology, Faculty of Biotechnology and Food Sciences, Lodz University of Technology, Stefanowskiego 2/22, 90-573 Lodz, Poland
| | - Natalia Filipowicz
- Department of Molecular Biotechnology and Microbiology, Faculty of Chemistry, Gdansk University of Technology, Narutowicza 11/12, 80-233 Gdansk, Poland
| |
Collapse
|
18
|
Identification and characterization of coiled-coil motifs across Autographa californica multiple nucleopolyhedrovirus genome. Heliyon 2022; 8:e10588. [PMID: 36132175 PMCID: PMC9483598 DOI: 10.1016/j.heliyon.2022.e10588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 05/15/2022] [Accepted: 09/05/2022] [Indexed: 12/02/2022] Open
Abstract
Coiled coils (CCs) are protein structural motifs universally found in proteins and mediate a plethora of biological interactions, and thus their reliable annotation is crucial for studies of protein structure and function. Autographa californica multiple nucleopolyhedrovirus (AcMNPV) is a large double-stranded DNA (dsDNA) virus and encodes 154 proteins. In this study, genome-wide scans of previously uncharacterized CC motifs throughout AcMNPV was conducted using CC prediction software. In total, 24 CC motifs in 19 CC proteins with high confidence were identified. The characteristic of viral CC motifs were analyzed. The CC proteins could be divided into 12 viral structural proteins and 7 non-structural proteins, including viral membrane fusion proteins, enzymes, and transcription factors. Moreover, CC motifs are conserved in the baculoviral orthologs of 14 of the 19 proteins. It is noted that five CC proteins, including Ac51, Ac66, Exon0, Ac13, and GP16, were previously identified to function in the nuclear egress of nucleocapsids, and Ac66 contains multiple CC motifs, the longest of which comprises 252 amino acids, suggesting a role of CC motifs in this process. Taken together, the CC motifs identified in this study are valuable resource for studying protein function and protein interaction networks during virus replication.
Collapse
|
19
|
Choque-Guevara R, Poma-Acevedo A, Montesinos-Millán R, Rios-Matos D, Gutiérrez-Manchay K, Montalvan-Avalos A, Quiñones-Garcia S, Cauti-Mendoza MDG, Agurto-Arteaga A, Ramirez-Ortiz I, Criollo-Orozco M, Huaccachi-Gonzales E, Romero YK, Perez-Martinez N, Isasi-Rivas G, Sernaque-Aguilar Y, Villanueva-Pérez D, Ygnacio F, Vallejos-Sánchez K, Fernández-Sánchez M, Guevara-Sarmiento LA, Fernández-Díaz M, Zimic M. Squalene in oil-based adjuvant improves the immunogenicity of SARS-CoV-2 RBD and confirms safety in animal models. PLoS One 2022; 17:e0269823. [PMID: 35998134 PMCID: PMC9397949 DOI: 10.1371/journal.pone.0269823] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 05/30/2022] [Indexed: 01/09/2023] Open
Abstract
COVID-19 pandemic has accelerated the development of vaccines against its etiologic agent, SARS-CoV-2. However, the emergence of new variants of the virus lead to the generation of new alternatives to improve the current sub-unit vaccines in development. In the present report, the immunogenicity of the Spike RBD of SARS-CoV-2 formulated with an oil-in-water emulsion and a water-in-oil emulsion with squalene was evaluated in mice and hamsters. The RBD protein was expressed in insect cells and purified by chromatography until >95% purity. The protein was shown to have the appropriate folding as determined by ELISA and flow cytometry binding assays to its receptor, as well as by its detection by hamster immune anti-S1 sera under non-reducing conditions. In immunization assays, although the cellular immune response elicited by both adjuvants were similar, the formulation based in water-in-oil emulsion and squalene generated an earlier humoral response as determined by ELISA. Similarly, this formulation was able to stimulate neutralizing antibodies in hamsters. The vaccine candidate was shown to be safe, as demonstrated by the histopathological analysis in lungs, liver and kidney. These results have shown the potential of this formulation vaccine to be evaluated in a challenge against SARS-CoV-2 and determine its ability to confer protection.
Collapse
Affiliation(s)
| | | | | | - Dora Rios-Matos
- Laboratorios de investigación y desarrollo, FARVET SAC, Chincha, Ica, Perú
| | | | | | - Stefany Quiñones-Garcia
- Laboratorios de investigación y desarrollo, FARVET SAC, Chincha, Ica, Perú
- Laboratorio de Bioinformática, Biología Molecular y Desarrollos Tecnológicos, Laboratorios de Investigación y Desarrollo, Facultad de Ciencias y Filosofía, Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Maria de Grecia Cauti-Mendoza
- Laboratorios de investigación y desarrollo, FARVET SAC, Chincha, Ica, Perú
- Laboratorio de Bioinformática, Biología Molecular y Desarrollos Tecnológicos, Laboratorios de Investigación y Desarrollo, Facultad de Ciencias y Filosofía, Universidad Peruana Cayetano Heredia, Lima, Peru
| | | | | | | | | | - Yomara K. Romero
- Laboratorio de Bioinformática, Biología Molecular y Desarrollos Tecnológicos, Laboratorios de Investigación y Desarrollo, Facultad de Ciencias y Filosofía, Universidad Peruana Cayetano Heredia, Lima, Peru
| | | | - Gisela Isasi-Rivas
- Laboratorios de investigación y desarrollo, FARVET SAC, Chincha, Ica, Perú
| | | | | | - Freddy Ygnacio
- Laboratorios de investigación y desarrollo, FARVET SAC, Chincha, Ica, Perú
| | - Katherine Vallejos-Sánchez
- Laboratorio de Bioinformática, Biología Molecular y Desarrollos Tecnológicos, Laboratorios de Investigación y Desarrollo, Facultad de Ciencias y Filosofía, Universidad Peruana Cayetano Heredia, Lima, Peru
| | | | | | | | - Mirko Zimic
- Laboratorios de investigación y desarrollo, FARVET SAC, Chincha, Ica, Perú
- Laboratorio de Bioinformática, Biología Molecular y Desarrollos Tecnológicos, Laboratorios de Investigación y Desarrollo, Facultad de Ciencias y Filosofía, Universidad Peruana Cayetano Heredia, Lima, Peru
- * E-mail:
| | | |
Collapse
|
20
|
Quinlan EJ, Chubet R, Leonardi P. A novel SARS-CoV-2 subunit vaccine engineered on an immune-activating platform technology. Hum Vaccin Immunother 2022; 18:2062971. [PMID: 35801956 DOI: 10.1080/21645515.2022.2062971] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
While there are several SARS-CoV-2 vaccines currently available, additional options must be provided that are safe, effective, and affordable for the entire global population. We have developed a novel immune activating platform technology that will fill this need. This recombinant platform protein is produced in insect cells using baculoviral expression technology similar to what is currently used for several other approved vaccines as well as employed by myriad GMP facilities globally. Thus, infrastructure exists for rapid scale up following initial optimizations. Here we report initial results for a SARS-CoV-2 vaccine (OMN008) based on our platform technology. Unadjuvanted OMN008 vaccination resulted in robust antigenicity and neutralization. Additionally, OMN008 vaccination induced a specific CD8 T-cell response. All of these results taken together indicate OMN008 may be an excellent candidate to fill gaps left by the currently available vaccines. Further testing is necessary to fully optimize production; however, overall cost of production should remain low given the simple formulation of this recombinant platform.
Collapse
Affiliation(s)
| | - Richard Chubet
- Research and Development, OmniCyte LLC, Guilford, CT, USA
| | - Peter Leonardi
- Research and Development, OmniCyte LLC, Guilford, CT, USA
| |
Collapse
|
21
|
The effect of different insect cell culture media on the efficiency of protein production by Spodoptera frugiperda cells. ELECTRON J BIOTECHN 2022. [DOI: 10.1016/j.ejbt.2022.01.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
|
22
|
Koda S, Zhu XQ, Zheng KY, Yan C. Molecular Mechanisms of Clonorchis sinensis-Host Interactions and Implications for Vaccine Development. Front Cell Dev Biol 2022; 9:781768. [PMID: 35118069 PMCID: PMC8804234 DOI: 10.3389/fcell.2021.781768] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 12/14/2021] [Indexed: 12/12/2022] Open
Abstract
Infections caused by Clonorchis sinensis remain a significant public health challenge for both humans and animals, causing pyogenic cholangitis, cholelithiasis, cholecystitis, biliary fibrosis, and even cholangiocarcinoma. However, the strategies used by the parasite and the immunological mechanisms used by the host have not yet been fully understood. With the advances in technologies and the accumulated knowledge of host-parasite interactions, many vaccine candidates against liver flukes have been investigated using different strategies. In this review, we explore and analyze in-depth the immunological mechanisms involved in the pathogenicity of C. sinensis. We highlight the different mechanisms by which the parasite interacts with its host to induce immune responses. All together, these data will allow us to have a better understanding of molecular mechansism of host-parasite interactions, which may shed lights on the development of an effective vaccine against C. sinensis.
Collapse
Affiliation(s)
- Stephane Koda
- Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Laboratory of Infection and Immunity, Department of Pathogenic Biology and Immunology, National Experimental Demonstration Center for Basic Medicine Education, Xuzhou Medical University, Xuzhou, China
| | - Xing-Quan Zhu
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, China
| | - Kui-Yang Zheng
- Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Laboratory of Infection and Immunity, Department of Pathogenic Biology and Immunology, National Experimental Demonstration Center for Basic Medicine Education, Xuzhou Medical University, Xuzhou, China
- *Correspondence: Kui-Yang Zheng, ; Chao Yan,
| | - Chao Yan
- Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Laboratory of Infection and Immunity, Department of Pathogenic Biology and Immunology, National Experimental Demonstration Center for Basic Medicine Education, Xuzhou Medical University, Xuzhou, China
- *Correspondence: Kui-Yang Zheng, ; Chao Yan,
| |
Collapse
|
23
|
Cox MMJ. Innovations in the Insect Cell Expression System for Industrial Recombinant Vaccine Antigen Production. Vaccines (Basel) 2021; 9:vaccines9121504. [PMID: 34960250 PMCID: PMC8707663 DOI: 10.3390/vaccines9121504] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 12/16/2021] [Accepted: 12/16/2021] [Indexed: 12/22/2022] Open
Abstract
The insect cell expression system has previously been proposed as the preferred biosecurity strategy for production of any vaccine, particularly for future influenza pandemic vaccines. The development and regulatory risk for new vaccine candidates is shortened as the platform is already in use for the manufacturing of the FDA-licensed seasonal recombinant influenza vaccine Flublok®. Large-scale production capacity is in place and could be used to produce other antigens as well. However, as demonstrated by the 2019 SARS-CoV-2 pandemic the insect cell expression system has limitations that need to be addressed to ensure that recombinant antigens will indeed play a role in combating future pandemics. The greatest challenge may be the ability to produce an adequate quantity of purified antigen in an accelerated manner. This review summarizes recent innovations in technology areas important for enhancing recombinant-protein production levels and shortening development timelines. Opportunities for increasing product concentrations through vector development, cell line engineering, or bioprocessing and for shortening timelines through standardization of manufacturing processes will be presented.
Collapse
|
24
|
Salem R, El-Kholy AA, Waly FR, Ayman D, Sakr A, Hussein M. Generation and utility of a single-chain fragment variable monoclonal antibody platform against a baculovirus expressed recombinant receptor binding domain of SARS-CoV-2 spike protein. Mol Immunol 2021; 141:287-296. [PMID: 34915268 PMCID: PMC8660258 DOI: 10.1016/j.molimm.2021.12.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 12/01/2021] [Accepted: 12/05/2021] [Indexed: 02/08/2023]
Abstract
As the second wave of COVID-19 launched, various variants of Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) have emerged with a dramatic global spread amongst millions of people causing unprecedented case fatalities and economic shut-downs. That initiated a necessity for developing specific diagnostics and therapeutics along with vaccines to control such a pandemic. This endeavor describes generation of murine derived recombinant single-chain fragment variable (scFv) as a monoclonal antibody (MAb) platform targeting the receptor binding domain (RBD) of Spike protein of SARS-CoV-2. A specific synthesized RBD coding sequence was cloned and expressed in Baculovirus expression system. The recombinant RBD (rRBD) was ascertained to be at the proper encoding size of ∼ 600bp and expressed protein of the molecular weight of ∼ 21KDa. Purified rRBD was proved genuinely antigenic and immunogenic, exhibiting specific reactivity to anti-SARS-CoV-2 antibody in an indirect enzyme-linked immunosorbent assay (ELISA), and inducing strong seroconversion in immunized mice. The scFv phage display library against rRBD was successfully constructed, revealing ∼ 90 % recombination frequency, and great enriching factor reaching 88 % and 25 % in polyclonal Ab-based and MAb-based ELISAs, respectively. Typically, three unique scFvs were generated, selected, purified and molecularly identified. That was manifested by their: accurate structure, close relation to the mouse immunoglobulin (Ig) superfamily, right anchored six complementarily-determining regions (CDRs) as three within variable heavy (vH) and variable light (vL) regions each, and proper configuration of the three-dimensional (3D) structure. Besides, their expression downstream in a non-suppressive amber codon of E. coli strain SS32 created a distinct protein band at an apparent molecular weight of ∼ 27KDa. Moreover, the purified scFvs showed authentic immunoreactivity and specificity to both rRBD and SARS-CoV-2 in western blot and ELISA. Accordingly, these developed scFvs platform might be a functional candidate for research, inexpensive diagnostics and therapeutics, mitigating spread of COVID-19.
Collapse
Affiliation(s)
- Reda Salem
- Agricultural Genetic Engineering Research Institute (AGERI), ARC, 12619, Giza, Egypt.
| | - Alaa A El-Kholy
- Veterinary Sera and Vaccines Research Institute (VSVRI), ARC, Abbassia, P.O. Box #131, 11381, Cairo, Egypt
| | - Fatma R Waly
- Agricultural Genetic Engineering Research Institute (AGERI), ARC, 12619, Giza, Egypt
| | - Dalia Ayman
- Agricultural Genetic Engineering Research Institute (AGERI), ARC, 12619, Giza, Egypt
| | - Aya Sakr
- Agricultural Genetic Engineering Research Institute (AGERI), ARC, 12619, Giza, Egypt
| | - Mai Hussein
- Agricultural Genetic Engineering Research Institute (AGERI), ARC, 12619, Giza, Egypt
| |
Collapse
|
25
|
Targovnik AM, Simonin JA, Mc Callum GJ, Smith I, Cuccovia Warlet FU, Nugnes MV, Miranda MV, Belaich MN. Solutions against emerging infectious and noninfectious human diseases through the application of baculovirus technologies. Appl Microbiol Biotechnol 2021; 105:8195-8226. [PMID: 34618205 PMCID: PMC8495437 DOI: 10.1007/s00253-021-11615-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 09/20/2021] [Accepted: 09/21/2021] [Indexed: 12/23/2022]
Abstract
Abstract
Baculoviruses are insect pathogens widely used as biotechnological tools in different fields of life sciences and technologies. The particular biology of these entities (biosafety viruses 1; large circular double-stranded DNA genomes, infective per se; generally of narrow host range on insect larvae; many of the latter being pests in agriculture) and the availability of molecular-biology procedures (e.g., genetic engineering to edit their genomes) and cellular resources (availability of cell lines that grow under in vitro culture conditions) have enabled the application of baculoviruses as active ingredients in pest control, as systems for the expression of recombinant proteins (Baculovirus Expression Vector Systems—BEVS) and as viral vectors for gene delivery in mammals or to display antigenic proteins (Baculoviruses applied on mammals—BacMam). Accordingly, BEVS and BacMam technologies have been introduced in academia because of their availability as commercial systems and ease of use and have also reached the human pharmaceutical industry, as incomparable tools in the development of biological products such as diagnostic kits, vaccines, protein therapies, and—though still in the conceptual stage involving animal models—gene therapies. Among all the baculovirus species, the Autographa californica multiple nucleopolyhedrovirus has been the most highly exploited in the above utilities for the human-biotechnology field. This review highlights the main achievements (in their different stages of development) of the use of BEVS and BacMam technologies for the generation of products for infectious and noninfectious human diseases. Key points • Baculoviruses can assist as biotechnological tools in human health problems. • Vaccines and diagnosis reagents produced in the baculovirus platform are described. • The use of recombinant baculovirus for gene therapy–based treatment is reviewed.
Collapse
Affiliation(s)
- Alexandra Marisa Targovnik
- Cátedra de Biotecnología, Departamento de Microbiología, Inmunología, Biotecnología y Genética, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Junín 956, Buenos Aires, 1113, Argentina.
- Instituto de Nanobiotecnología (NANOBIOTEC), Facultad de Farmacia y Bioquímica, CONICET -Universidad de Buenos Aires, Junín 956, Sexto Piso, C1113AAD, 1113, Buenos Aires, Argentina.
| | - Jorge Alejandro Simonin
- Laboratorio de Ingeniería Genética y Biología Celular y Molecular, Área Virosis de Insectos, Instituto de Microbiología Básica y Aplicada, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes, Buenos Aires, Argentina
| | - Gregorio Juan Mc Callum
- Cátedra de Biotecnología, Departamento de Microbiología, Inmunología, Biotecnología y Genética, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Junín 956, Buenos Aires, 1113, Argentina
- Instituto de Nanobiotecnología (NANOBIOTEC), Facultad de Farmacia y Bioquímica, CONICET -Universidad de Buenos Aires, Junín 956, Sexto Piso, C1113AAD, 1113, Buenos Aires, Argentina
| | - Ignacio Smith
- Cátedra de Biotecnología, Departamento de Microbiología, Inmunología, Biotecnología y Genética, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Junín 956, Buenos Aires, 1113, Argentina
- Instituto de Nanobiotecnología (NANOBIOTEC), Facultad de Farmacia y Bioquímica, CONICET -Universidad de Buenos Aires, Junín 956, Sexto Piso, C1113AAD, 1113, Buenos Aires, Argentina
| | - Franco Uriel Cuccovia Warlet
- Laboratorio de Ingeniería Genética y Biología Celular y Molecular, Área Virosis de Insectos, Instituto de Microbiología Básica y Aplicada, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes, Buenos Aires, Argentina
| | - María Victoria Nugnes
- Laboratorio de Ingeniería Genética y Biología Celular y Molecular, Área Virosis de Insectos, Instituto de Microbiología Básica y Aplicada, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes, Buenos Aires, Argentina
| | - María Victoria Miranda
- Cátedra de Biotecnología, Departamento de Microbiología, Inmunología, Biotecnología y Genética, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Junín 956, Buenos Aires, 1113, Argentina
- Instituto de Nanobiotecnología (NANOBIOTEC), Facultad de Farmacia y Bioquímica, CONICET -Universidad de Buenos Aires, Junín 956, Sexto Piso, C1113AAD, 1113, Buenos Aires, Argentina
| | - Mariano Nicolás Belaich
- Laboratorio de Ingeniería Genética y Biología Celular y Molecular, Área Virosis de Insectos, Instituto de Microbiología Básica y Aplicada, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes, Buenos Aires, Argentina
| |
Collapse
|
26
|
Porcine Circovirus (PCV) Genotype 2d-Based Virus-like Particles (VLPs) Induced Broad Cross-Neutralizing Antibodies against Diverse Genotypes and Provided Protection in Dual-Challenge Infection of a PCV2d Virus and a Type 1 Porcine Reproductive and Respiratory Syndrome Virus (PRRSV). Pathogens 2021; 10:pathogens10091145. [PMID: 34578177 PMCID: PMC8464671 DOI: 10.3390/pathogens10091145] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 08/24/2021] [Accepted: 09/01/2021] [Indexed: 11/29/2022] Open
Abstract
As PCV2d infection has been continuously reported in swine farms in which pigs were vaccinated with PCV2a- or 2d-based vaccines, we attempted to develop a novel vaccine using a PCV2d-based capsid to enhance its protective efficacy. In this study, recombinant virus-like particles (VLPs) of rPCV2a, rPCV2b and rPCV2d were synthesized from the capsid proteins of PCV2a, PCV2b and PCV2d field isolates, respectively. A cross-neutralization assay between the VLPs induced antisera and the field isolates demonstrated the broad cross-neutralizing activities of the rPCV2d-induced antisera. Then, the protective efficacy of rPCV2d as a vaccine candidate was investigated in commercial pigs by rPCV2d vaccination and a single- or dual-challenge infection using a PCV2d strain and a type 1 PRRSV strain. High levels of anti-PCV2d IgG and neutralizing antibodies were induced 3 weeks after vaccination. After the challenge infection, the average ADWG values of the vaccinated group were higher than those of the unvaccinated group. None or a significantly low amount of (p < 0.05) reduced PCV2 genomic DNA was found in the blood, saliva and tissues of the vaccinated pigs, when compared to the unvaccinated group. Moreover, macroscopic and microscopic lesions in the tissues were significantly (p < 0.05) reduced in the vaccinated groups. This study therefore suggests that rPCV2d may be highly useful for the control of diverse field genotypes.
Collapse
|
27
|
Verdecia M, Kokai-Kun JF, Kibbey M, Acharya S, Venema J, Atouf F. COVID-19 vaccine platforms: Delivering on a promise? Hum Vaccin Immunother 2021; 17:2873-2893. [PMID: 34033528 PMCID: PMC8381795 DOI: 10.1080/21645515.2021.1911204] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 03/24/2021] [Indexed: 12/13/2022] Open
Abstract
The emergence of the novel SARS-CoV-2 and COVID-19 has brought into sharp focus the need for a vaccine to prevent this disease. Vaccines have saved millions of lives since their introduction to the public over 200 years ago. The potential for vaccination reached new heights in the mid-20th century with the development of technologies that expanded the ability to create novel vaccines. Since then, there has been continued technological advancement in vaccine development. The resulting platforms provide the promise for solutions for many infectious diseases, including those that have been with us for decades as well as those just now emerging. Each vaccine platform represents a different technology with a unique set of advantages and challenges, especially when considering manufacturing. Therefore, it is essential to understand each platform as a separate product and process with its specific quality considerations. This review outlines the relevant platforms for developing a vaccine for SARS-CoV-2 and discusses the advantages and disadvantages of each.
Collapse
Affiliation(s)
- Mark Verdecia
- United States Pharmacopeial Convention, Rockville, MD, USA
| | | | - Maura Kibbey
- United States Pharmacopeial Convention, Rockville, MD, USA
| | - Sarita Acharya
- United States Pharmacopeial Convention, Rockville, MD, USA
| | - Jaap Venema
- United States Pharmacopeial Convention, Rockville, MD, USA
| | - Fouad Atouf
- United States Pharmacopeial Convention, Rockville, MD, USA
| |
Collapse
|
28
|
Cid R, Bolívar J. Platforms for Production of Protein-Based Vaccines: From Classical to Next-Generation Strategies. Biomolecules 2021; 11:1072. [PMID: 34439738 PMCID: PMC8394948 DOI: 10.3390/biom11081072] [Citation(s) in RCA: 86] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 07/16/2021] [Accepted: 07/17/2021] [Indexed: 12/12/2022] Open
Abstract
To date, vaccination has become one of the most effective strategies to control and reduce infectious diseases, preventing millions of deaths worldwide. The earliest vaccines were developed as live-attenuated or inactivated pathogens, and, although they still represent the most extended human vaccine types, they also face some issues, such as the potential to revert to a pathogenic form of live-attenuated formulations or the weaker immune response associated with inactivated vaccines. Advances in genetic engineering have enabled improvements in vaccine design and strategies, such as recombinant subunit vaccines, have emerged, expanding the number of diseases that can be prevented. Moreover, antigen display systems such as VLPs or those designed by nanotechnology have improved the efficacy of subunit vaccines. Platforms for the production of recombinant vaccines have also evolved from the first hosts, Escherichia coli and Saccharomyces cerevisiae, to insect or mammalian cells. Traditional bacterial and yeast systems have been improved by engineering and new systems based on plants or insect larvae have emerged as alternative, low-cost platforms. Vaccine development is still time-consuming and costly, and alternative systems that can offer cost-effective and faster processes are demanding to address infectious diseases that still do not have a treatment and to face possible future pandemics.
Collapse
Affiliation(s)
- Raquel Cid
- ADL Bionatur Solutions S.A., Av. del Desarrollo Tecnológico 11, 11591 Jerez de la Frontera, Spain
| | - Jorge Bolívar
- Department of Biomedicine, Biotechnology and Public Health-Biochemistry and Molecular Biology, Campus Universitario de Puerto Real, University of Cadiz, 11510 Puerto Real, Spain
| |
Collapse
|
29
|
Chen J, Wang J, Zhang J, Ly H. Advances in Development and Application of Influenza Vaccines. Front Immunol 2021; 12:711997. [PMID: 34326849 PMCID: PMC8313855 DOI: 10.3389/fimmu.2021.711997] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Accepted: 06/24/2021] [Indexed: 12/24/2022] Open
Abstract
Influenza A virus is one of the most important zoonotic pathogens that can cause severe symptoms and has the potential to cause high number of deaths and great economic loss. Vaccination is still the best option to prevent influenza virus infection. Different types of influenza vaccines, including live attenuated virus vaccines, inactivated whole virus vaccines, virosome vaccines, split-virion vaccines and subunit vaccines have been developed. However, they have several limitations, such as the relatively high manufacturing cost and long production time, moderate efficacy of some of the vaccines in certain populations, and lack of cross-reactivity. These are some of the problems that need to be solved. Here, we summarized recent advances in the development and application of different types of influenza vaccines, including the recent development of viral vectored influenza vaccines. We also described the construction of other vaccines that are based on recombinant influenza viruses as viral vectors. Information provided in this review article might lead to the development of safe and highly effective novel influenza vaccines.
Collapse
Affiliation(s)
- Jidang Chen
- School of Life Science and Engineering, Foshan University, Foshan, China
| | - Jiehuang Wang
- School of Life Science and Engineering, Foshan University, Foshan, China
| | - Jipei Zhang
- School of Life Science and Engineering, Foshan University, Foshan, China
| | - Hinh Ly
- Department of Veterinary & Biomedical Sciences, University of Minnesota, Twin Cities, MN, United States
| |
Collapse
|
30
|
Gałęcki R, Zielonka Ł, Zasȩpa M, Gołȩbiowska J, Bakuła T. Potential Utilization of Edible Insects as an Alternative Source of Protein in Animal Diets in Poland. FRONTIERS IN SUSTAINABLE FOOD SYSTEMS 2021. [DOI: 10.3389/fsufs.2021.675796] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The selection of high-protein raw materials that could be utilized in the production of animal feed is limited in Poland. The aim of this study was to analyze the needs and possibilities associated with the utilization of insect protein in animal nutrition in Poland. In the future, insects could become an effective solution to meeting the nutritional requirements of a growing population. Insect protein is already used in the production of fish feed in the European Union (EU). Legislative decisions on the introduction of this alternative feed source have to be based on the results of studies investigating the safety of insect protein for farmed animals. Diets containing insect protein and their influence on animals need to be thoroughly analyzed. In the future, insect farming could become a novel branch of agriculture, and it could create new opportunities for Polish farmers who were affected by the African swine fever (ASF) virus. Insect farms could create new jobs, promote innovative business development, and increase food and feed production. Entomophagy is a new and controversial concept for Polish consumers, but in the future, it could offer a viable solution to feeding the world's growing population.
Collapse
|
31
|
Dias MM, Vidigal J, Sequeira DP, Alves PM, Teixeira AP, Roldão A. Insect High FiveTM cell line development using site-specific flipase recombination technology. G3-GENES GENOMES GENETICS 2021; 11:6274903. [PMID: 33982066 PMCID: PMC8763235 DOI: 10.1093/g3journal/jkab166] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Accepted: 05/24/2021] [Indexed: 11/14/2022]
Abstract
Insect Trichoplusia ni High FiveTM (Hi5) cells have been widely explored for production of heterologous proteins, traditionally mostly using the lytic baculovirus expression vector system (BEVS), and more recently using virus-free transient gene expression systems. Stable expression in such host cells would circumvent the drawbacks associated with both systems when it comes to scale-up and implementation of more efficient high-cell density process modes for the manufacturing of biologics. In this work, we combined Flipase (Flp) recombinase-mediated cassette exchange (RMCE) with fluorescence-activated cell sorting (FACS) for generating a stable master clonal Hi5 cell line with the flexibility to express single or multiple proteins of interest from a tagged genomic locus. The 3-step protocol herein implemented consisted of (i) introducing the RMCE docking cassette into the cell genome by random integration followed by selection in Hygromycin B and FACS (Hi5-tagging population), (ii) eliminating cells tagged in loci with low recombination efficiency by transfecting the tagging population with an eGFP-containing target cassette followed by selection in G418 and FACS (Hi5-RMCE population), and (iii) isolation of pure eGFP-expressing cells by FACS and expansion to suspension cultures (Hi5-RMCE master clone). Exchangeability of the locus in the master clone was demonstrated in small-scale suspension cultures by replacing the target cassette by one containing a single protein (i.e. iCherry, as an intracellular protein model) or two proteins (i.e. influenza HA and M1 for virus-like particles production, as an extracellular protein model). Overall, the stable insect Hi5 cell platform herein assembled has the potential to assist and accelerate biologics development.
Collapse
Affiliation(s)
- Mafalda M Dias
- IBET, Instituto de Biologia Experimental e Tecnológica, 2780-901 Oeiras, Portugal.,Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, 2780-901 Oeiras, Portugal
| | - João Vidigal
- IBET, Instituto de Biologia Experimental e Tecnológica, 2780-901 Oeiras, Portugal.,Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, 2780-901 Oeiras, Portugal
| | - Daniela P Sequeira
- IBET, Instituto de Biologia Experimental e Tecnológica, 2780-901 Oeiras, Portugal.,Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, 2780-901 Oeiras, Portugal.,Department of Molecular Life Sciences, University of Zurich, 8057 Zurich, Switzerland
| | - Paula M Alves
- IBET, Instituto de Biologia Experimental e Tecnológica, 2780-901 Oeiras, Portugal.,Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, 2780-901 Oeiras, Portugal
| | - Ana P Teixeira
- IBET, Instituto de Biologia Experimental e Tecnológica, 2780-901 Oeiras, Portugal.,Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, 2780-901 Oeiras, Portugal.,ETH Zurich, Department of Biosystems Science and Engineering, Mattenstrasse 26, 4058 - Basel, Switzerland
| | - António Roldão
- IBET, Instituto de Biologia Experimental e Tecnológica, 2780-901 Oeiras, Portugal.,Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, 2780-901 Oeiras, Portugal
| |
Collapse
|
32
|
Attia YA, El-Saadony MT, Swelum AA, Qattan SYA, Al-Qurashi AD, Asiry KA, Shafi ME, Elbestawy AR, Gado AR, Khafaga AF, Hussein EOS, Ba-Awadh H, Tiwari R, Dhama K, Alhussaini B, Alyileili SR, El-Tarabily KA, Abd El-Hack ME. COVID-19: pathogenesis, advances in treatment and vaccine development and environmental impact-an updated review. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2021; 28:22241-22264. [PMID: 33733422 PMCID: PMC7969349 DOI: 10.1007/s11356-021-13018-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/26/2020] [Accepted: 02/15/2021] [Indexed: 05/08/2023]
Abstract
Diseases negatively impact the environment, causing many health risks and the spread of pollution and hazards. A novel coronavirus, severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2) has led to a recent respiratory syndrome epidemic in humans. In December 2019, the sudden emergence of this new coronavirus and the subsequent severe disease it causes created a serious global health threat and hazards. This is in contrast to the two aforementioned coronaviruses, SARS-CoV-2 (in 2002) and middle east respiratory syndrome coronavirus MERS-CoV (in 2012), which were much more easily contained. The World Health Organization (WHO) dubbed this contagious respiratory disease an "epidemic outbreak" in March 2020. More than 80 companies and research institutions worldwide are working together, in cooperation with many governmental agencies, to develop an effective vaccine. To date, six authorized vaccines have been registered. Up till now, no approved drugs and drug scientists are racing from development to clinical trials to find new drugs for COVID-19. Wild animals, such as snakes, bats, and pangolins are the main sources of coronaviruses, as determined by the sequence homology between MERS-CoV and viruses in these animals. Human infection is caused by inhalation of respiratory droplets. To date, the only available treatment protocol for COVID-19 is based on the prevalent clinical signs. This review aims to summarize the current information regarding the origin, evolution, genomic organization, epidemiology, and molecular and cellular characteristics of SARS-CoV-2 as well as the diagnostic and treatment approaches for COVID-19 and its impact on global health, environment, and economy.
Collapse
Affiliation(s)
- Youssef A Attia
- Agriculture Department, Faculty of Environmental Sciences, King Abdulaziz University, P.O. Box 80208, Jeddah, 21589, Saudi Arabia.
- The Strategic Center to Kingdom Vision Realization, King Abdulaziz University, Jeddah, Saudi Arabia.
- Animal and Poultry Production Department, Faculty of Agriculture, Damanhour University, Damanhour, Egypt.
| | - Mohamed T El-Saadony
- Department of Agricultural Microbiology, Faculty of Agriculture, Zagazig University, Zagazig, 44511, Egypt
| | - Ayman A Swelum
- Department of Animal Production, College of Food and Agriculture Sciences, King Saud University, Riyadh, 11451, Saudi Arabia.
- Department of Theriogenology, Faculty of Veterinary Medicine, Zagazig University, Sharkia, Zagazig, 44519, Egypt.
| | - Shaza Y A Qattan
- Department of Biological Sciences, Microbiology, King Abdulaziz University, P.O. Box 80203, Jeddah, 21589, Saudi Arabia
| | - Adel D Al-Qurashi
- Agriculture Department, Faculty of Environmental Sciences, King Abdulaziz University, P.O. Box 80208, Jeddah, 21589, Saudi Arabia
| | - Khalid A Asiry
- Agriculture Department, Faculty of Environmental Sciences, King Abdulaziz University, P.O. Box 80208, Jeddah, 21589, Saudi Arabia
| | - Manal E Shafi
- Department of Biological Sciences, Zoology, King Abdulaziz University, P.O. Box 80203, Jeddah, 21589, Saudi Arabia
| | - Ahmed R Elbestawy
- Poultry and Fish Diseases Department, Faculty of Veterinary Medicine, Damanhour University, Damanhur, 22511, Egypt
| | - Ahmed R Gado
- Poultry and Fish Diseases Department, Faculty of Veterinary Medicine, Damanhour University, Damanhur, 22511, Egypt
| | - Asmaa F Khafaga
- Department of Pathology, Faculty of Veterinary Medicine, Alexandria University, Edfina, Alexandria, 22758, Egypt
| | - Elsayed O S Hussein
- Department of Animal Production, College of Food and Agriculture Sciences, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Hani Ba-Awadh
- Department of Animal Production, College of Food and Agriculture Sciences, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Ruchi Tiwari
- Department of Veterinary Microbiology and Immunology, College of Veterinary Sciences, Uttar Pradesh Pandit Deen Dayal Upadhyaya Pashu Chikitsa Vigyan Vishwavidyalaya Evam Go Anusandhan Sansthan (DUVASU), Mathura, 281001, India
| | - Kuldeep Dhama
- Division of Pathology, Indian Veterinary Research Institute (IVRI), Izatnagar-243, Bareilly, Uttar Pradesh, 122, India
| | - Bakr Alhussaini
- Department of Pediatric, Faculty of Medicine, King Abdualziz University, Jeddah, Saudi Arabia
| | - Salem R Alyileili
- Department of Integrative Agriculture, College of Food and Agriculture, United Arab Emirates University, 15551, Al-Ain, United Arab Emirates
| | - Khaled A El-Tarabily
- Department of Biology, College of Science, United Arab Emirates University, 15551, Al-Ain, United Arab Emirates.
- Harry Butler Institute, Murdoch University, Murdoch, Western Australia, 6150, Australia.
| | - Mohamed E Abd El-Hack
- Department of Poultry, Faculty of Agriculture, Zagazig University, Zagazig, 44511, Egypt
| |
Collapse
|
33
|
Aida V, Pliasas VC, Neasham PJ, North JF, McWhorter KL, Glover SR, Kyriakis CS. Novel Vaccine Technologies in Veterinary Medicine: A Herald to Human Medicine Vaccines. Front Vet Sci 2021; 8:654289. [PMID: 33937377 PMCID: PMC8083957 DOI: 10.3389/fvets.2021.654289] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 03/17/2021] [Indexed: 01/10/2023] Open
Abstract
The success of inactivated and live-attenuated vaccines has enhanced livestock productivity, promoted food security, and attenuated the morbidity and mortality of several human, animal, and zoonotic diseases. However, these traditional vaccine technologies are not without fault. The efficacy of inactivated vaccines can be suboptimal with particular pathogens and safety concerns arise with live-attenuated vaccines. Additionally, the rate of emerging infectious diseases continues to increase and with that the need to quickly deploy new vaccines. Unfortunately, first generation vaccines are not conducive to such urgencies. Within the last three decades, veterinary medicine has spearheaded the advancement in novel vaccine development to circumvent several of the flaws associated with classical vaccines. These third generation vaccines, including DNA, RNA and recombinant viral-vector vaccines, induce both humoral and cellular immune response, are economically manufactured, safe to use, and can be utilized to differentiate infected from vaccinated animals. The present article offers a review of commercially available novel vaccine technologies currently utilized in companion animal, food animal, and wildlife disease control.
Collapse
Affiliation(s)
- Virginia Aida
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL, United States
- Emory-University of Georgia (UGA) Center of Excellence for Influenza Research and Surveillance (CEIRS), Auburn, AL, United States
| | - Vasilis C. Pliasas
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL, United States
- Emory-University of Georgia (UGA) Center of Excellence for Influenza Research and Surveillance (CEIRS), Auburn, AL, United States
| | - Peter J. Neasham
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL, United States
- Emory-University of Georgia (UGA) Center of Excellence for Influenza Research and Surveillance (CEIRS), Auburn, AL, United States
| | - J. Fletcher North
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL, United States
- Emory-University of Georgia (UGA) Center of Excellence for Influenza Research and Surveillance (CEIRS), Auburn, AL, United States
| | - Kirklin L. McWhorter
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL, United States
- Department of Chemistry, Emory University, Atlanta, GA, United States
| | - Sheniqua R. Glover
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL, United States
- Emory-University of Georgia (UGA) Center of Excellence for Influenza Research and Surveillance (CEIRS), Auburn, AL, United States
| | - Constantinos S. Kyriakis
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL, United States
- Emory-University of Georgia (UGA) Center of Excellence for Influenza Research and Surveillance (CEIRS), Auburn, AL, United States
- Center for Vaccines and Immunology, University of Georgia, Athens, GA, United States
| |
Collapse
|
34
|
Perera PY, Perera LP. Development of leading first-generation vaccines against SARS-CoV-2. Microbes Infect 2021; 23:104841. [PMID: 34022375 PMCID: PMC8133830 DOI: 10.1016/j.micinf.2021.104841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 05/03/2021] [Accepted: 05/10/2021] [Indexed: 01/29/2023]
Abstract
SARS-CoV-2 has infected more than 167 million individuals globally. Highly effective and safe vaccines are required to accelerate the development of herd immunity to end the pandemic. This review focuses on vaccines that are being developed at unprecedented speed globally and are completing late phase clinical trials to meet this urgent need.
Collapse
Affiliation(s)
- Pin-Yu Perera
- Pathology and Laboratory Medicine, Veterans Affairs Medical Center, Washington, DC, USA.
| | - Liyanage P Perera
- Lymphoid Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
35
|
Tokumoto S, Miyata Y, Usui K, Deviatiiarov R, Ohkawa T, Kondratieva S, Shagimardanova E, Gusev O, Cornette R, Itoh M, Hayashizaki Y, Kikawada T. Development of a Tet-On Inducible Expression System for the Anhydrobiotic Cell Line, Pv11. INSECTS 2020; 11:E781. [PMID: 33187095 PMCID: PMC7696976 DOI: 10.3390/insects11110781] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 11/02/2020] [Accepted: 11/09/2020] [Indexed: 12/28/2022]
Abstract
The Pv11 cell line established from an African chironomid, Polypedilum vanderplanki, is the only cell line tolerant to complete desiccation. In Pv11 cells, a constitutive expression system for Pv11 cells was previously exploited and several reporter genes were successfully expressed. Here we report the identification of an effective minimal promoter for Pv11 cells and its application to the Tet-On inducible expression system. First, using a luciferase reporter assay, we showed that a 202 bp deletion fragment derived from the constitutively active 121-promoter functions in Pv11 cells as an appropriate minimal promoter with the Tet-On inducible expression system. The AcGFP1 (Aequorea coerulescens green fluorescent protein) was also successfully expressed in Pv11 cells using the inducible system. In addition to these reporter genes, the avian myeloblastosis virus reverse transcriptase α subunit (AMV RTα), which is one of the most widely commercially available RNA-dependent DNA polymerases, was successfully expressed through the inducible expression system and its catalytic activity was verified. These results demonstrate the establishment of an inducible expression system in cells that can be preserved in the dry state and highlight a possible application to the production of large and complex proteins.
Collapse
Affiliation(s)
- Shoko Tokumoto
- Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa 277-8562, Japan; (S.T.); (R.C.)
| | - Yugo Miyata
- Institute of Agrobiological Sciences, National Institute of Agriculture and Food Research Organization (NARO), Tsukuba 305-0851, Japan;
| | - Kengo Usui
- RIKEN Center for Integrative Medical Sciences (IMS), Yokohama 230-0045, Japan; (K.U.); (T.O.)
| | - Ruslan Deviatiiarov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Tatarstan 420012, Russia; (R.D.); (S.K.); (E.S.); (O.G.)
| | - Takahiro Ohkawa
- RIKEN Center for Integrative Medical Sciences (IMS), Yokohama 230-0045, Japan; (K.U.); (T.O.)
| | - Sabina Kondratieva
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Tatarstan 420012, Russia; (R.D.); (S.K.); (E.S.); (O.G.)
| | - Elena Shagimardanova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Tatarstan 420012, Russia; (R.D.); (S.K.); (E.S.); (O.G.)
| | - Oleg Gusev
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Tatarstan 420012, Russia; (R.D.); (S.K.); (E.S.); (O.G.)
- RIKEN Preventive Medicine and Diagnosis Innovation Program, Wako 351-0198, Japan; (M.I.); (Y.H.)
| | - Richard Cornette
- Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa 277-8562, Japan; (S.T.); (R.C.)
| | - Masayoshi Itoh
- RIKEN Preventive Medicine and Diagnosis Innovation Program, Wako 351-0198, Japan; (M.I.); (Y.H.)
| | - Yoshihide Hayashizaki
- RIKEN Preventive Medicine and Diagnosis Innovation Program, Wako 351-0198, Japan; (M.I.); (Y.H.)
| | - Takahiro Kikawada
- Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa 277-8562, Japan; (S.T.); (R.C.)
- Institute of Agrobiological Sciences, National Institute of Agriculture and Food Research Organization (NARO), Tsukuba 305-0851, Japan;
| |
Collapse
|
36
|
Roos N, Breiner B, Preuss L, Lilie H, Hipp K, Herrmann H, Horn T, Biener R, Iftner T, Simon C. Optimized production strategy of the major capsid protein HPV 16L1 non-assembly variant in E. coli. Protein Expr Purif 2020; 175:105690. [DOI: 10.1016/j.pep.2020.105690] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 06/19/2020] [Accepted: 06/21/2020] [Indexed: 12/31/2022]
|
37
|
Ecker JW, Kirchenbaum GA, Pierce SR, Skarlupka AL, Abreu RB, Cooper RE, Taylor-Mulneix D, Ross TM, Sautto GA. High-Yield Expression and Purification of Recombinant Influenza Virus Proteins from Stably-Transfected Mammalian Cell Lines. Vaccines (Basel) 2020; 8:vaccines8030462. [PMID: 32825605 PMCID: PMC7565037 DOI: 10.3390/vaccines8030462] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 08/15/2020] [Accepted: 08/17/2020] [Indexed: 12/21/2022] Open
Abstract
Influenza viruses infect millions of people each year, resulting in significant morbidity and mortality in the human population. Therefore, generation of a universal influenza virus vaccine is an urgent need and would greatly benefit public health. Recombinant protein technology is an established vaccine platform and has resulted in several commercially available vaccines. Herein, we describe the approach for developing stable transfected human cell lines for the expression of recombinant influenza virus hemagglutinin (HA) and recombinant influenza virus neuraminidase (NA) proteins for the purpose of in vitro and in vivo vaccine development. HA and NA are the main surface glycoproteins on influenza virions and the major antibody targets. The benefits for using recombinant proteins for in vitro and in vivo assays include the ease of use, high level of purity and the ability to scale-up production. This work provides guidelines on how to produce and purify recombinant proteins produced in mammalian cell lines through either transient transfection or generation of stable cell lines from plasmid creation through the isolation step via Immobilized Metal Affinity Chromatography (IMAC). Collectively, the establishment of this pipeline has facilitated large-scale production of recombinant HA and NA proteins to high purity and with consistent yields, including glycosylation patterns that are very similar to proteins produced in a human host.
Collapse
Affiliation(s)
- Jeffrey W. Ecker
- Center for Vaccines and Immunology, University of Georgia, Athens, GA 30602, USA; (J.W.E.); (G.A.K.); (S.R.P.); (A.L.S.); (R.B.A.); (R.E.C.); (D.T.-M.); (T.M.R.)
| | - Greg A. Kirchenbaum
- Center for Vaccines and Immunology, University of Georgia, Athens, GA 30602, USA; (J.W.E.); (G.A.K.); (S.R.P.); (A.L.S.); (R.B.A.); (R.E.C.); (D.T.-M.); (T.M.R.)
| | - Spencer R. Pierce
- Center for Vaccines and Immunology, University of Georgia, Athens, GA 30602, USA; (J.W.E.); (G.A.K.); (S.R.P.); (A.L.S.); (R.B.A.); (R.E.C.); (D.T.-M.); (T.M.R.)
| | - Amanda L. Skarlupka
- Center for Vaccines and Immunology, University of Georgia, Athens, GA 30602, USA; (J.W.E.); (G.A.K.); (S.R.P.); (A.L.S.); (R.B.A.); (R.E.C.); (D.T.-M.); (T.M.R.)
| | - Rodrigo B. Abreu
- Center for Vaccines and Immunology, University of Georgia, Athens, GA 30602, USA; (J.W.E.); (G.A.K.); (S.R.P.); (A.L.S.); (R.B.A.); (R.E.C.); (D.T.-M.); (T.M.R.)
| | - R. Ethan Cooper
- Center for Vaccines and Immunology, University of Georgia, Athens, GA 30602, USA; (J.W.E.); (G.A.K.); (S.R.P.); (A.L.S.); (R.B.A.); (R.E.C.); (D.T.-M.); (T.M.R.)
| | - Dawn Taylor-Mulneix
- Center for Vaccines and Immunology, University of Georgia, Athens, GA 30602, USA; (J.W.E.); (G.A.K.); (S.R.P.); (A.L.S.); (R.B.A.); (R.E.C.); (D.T.-M.); (T.M.R.)
| | - Ted M. Ross
- Center for Vaccines and Immunology, University of Georgia, Athens, GA 30602, USA; (J.W.E.); (G.A.K.); (S.R.P.); (A.L.S.); (R.B.A.); (R.E.C.); (D.T.-M.); (T.M.R.)
- Department of Infectious Diseases, University of Georgia, Athens, GA 30602, USA
| | - Giuseppe A. Sautto
- Center for Vaccines and Immunology, University of Georgia, Athens, GA 30602, USA; (J.W.E.); (G.A.K.); (S.R.P.); (A.L.S.); (R.B.A.); (R.E.C.); (D.T.-M.); (T.M.R.)
- Correspondence: ; Tel.: +1-706-542-6711
| |
Collapse
|
38
|
Matsuda T, Tanijima T, Hirose A, Masumi-Koizumi K, Katsuda T, Yamaji H. Production of influenza virus-like particles using recombinant insect cells. Biochem Eng J 2020; 163:107757. [PMID: 32834743 PMCID: PMC7427601 DOI: 10.1016/j.bej.2020.107757] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 07/22/2020] [Accepted: 08/13/2020] [Indexed: 01/23/2023]
Abstract
Influenza A virus-like particles (VLPs) were produced using recombinant insect cells. VLPs were produced using insect cells as host cells without using a baculovirus. A secretory form of VLPs consists of hemagglutinin and matrix protein 1. The VLP productivity is comparable to that of the baculovirus–insect cell system.
Virus-like particles (VLPs) are hollow nanoparticles composed of recombinant viral surface proteins without a virus genome. In the present study, we investigated the production of influenza VLPs using recombinant insect cells. DNA fragments encoding influenza A virus hemagglutinin (HA) and matrix protein 1 (M1) were cloned with the Drosophila BiP signal sequence in plasmid vectors containing a blasticidin and a neomycin resistance gene, respectively. After Trichoplusia ni BTI-TN-5B1-4 (High Five) cells were co-transfected with a pair of constructed plasmid vectors, stably transformed cells were established via incubation with blasticidin and G418. Western blot analyses showed that recombinant High Five cells secreted HA and M1 proteins into the culture supernatant. Immunoprecipitation of the culture supernatant with an anti-HA antibody and transmission electron microscopy suggested that secreted HA and M1 proteins were in a particulate structure with a morphology similar to that of an influenza virus. Hemagglutination assay indicated that expressed HA molecules retained hemagglutination activity. In a shake-flask culture, recombinant cells achieved a high HA yield (≈ 10 μg/ml) comparable to the yields obtained using the baculovirus–insect cell system. Recombinant insect cells may serve as excellent platforms for the efficient production of influenza VLPs for use as safe and effective vaccines and diagnostic antigens.
Collapse
Affiliation(s)
- Takuya Matsuda
- Department of Chemical Science and Engineering, Graduate School of Engineering, Kobe University, 1-1 Rokkodai, Nada, Kobe 657-8501, Japan
| | - Toshikazu Tanijima
- Department of Chemical Science and Engineering, Graduate School of Engineering, Kobe University, 1-1 Rokkodai, Nada, Kobe 657-8501, Japan
| | - Akito Hirose
- Department of Chemical Science and Engineering, Graduate School of Engineering, Kobe University, 1-1 Rokkodai, Nada, Kobe 657-8501, Japan
| | - Kyoko Masumi-Koizumi
- Department of Chemical Science and Engineering, Graduate School of Engineering, Kobe University, 1-1 Rokkodai, Nada, Kobe 657-8501, Japan
| | - Tomohisa Katsuda
- Department of Chemical Science and Engineering, Graduate School of Engineering, Kobe University, 1-1 Rokkodai, Nada, Kobe 657-8501, Japan
| | - Hideki Yamaji
- Department of Chemical Science and Engineering, Graduate School of Engineering, Kobe University, 1-1 Rokkodai, Nada, Kobe 657-8501, Japan
| |
Collapse
|
39
|
Holographic Imaging of Insect Cell Cultures: Online Non-Invasive Monitoring of Adeno-Associated Virus Production and Cell Concentration. Processes (Basel) 2020. [DOI: 10.3390/pr8040487] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
The insect cell-baculovirus vector system has become one of the favorite platforms for the expression of viral vectors for vaccination and gene therapy purposes. As it is a lytic system, it is essential to balance maximum recombinant product expression with harvest time, minimizing product exposure to detrimental proteases. With this purpose, new bioprocess monitoring solutions are needed to accurately estimate culture progression. Herein, we used online digital holographic microscopy (DHM) to monitor bioreactor cultures of Sf9 insect cells. Batches of baculovirus-infected Sf9 cells producing recombinant adeno-associated virus (AAV) and non-infected cells were used to evaluate DHM prediction capabilities for viable cell concentration, culture viability and AAV titer. Over 30 cell-related optical attributes were quantified using DHM, followed by a forward stepwise regression to select the most significant (p < 0.05) parameters for each variable. We then applied multiple linear regression to obtain models which were able to predict culture variables with root mean squared errors (RMSE) of 7 × 105 cells/mL, 3% for cell viability and 2 × 103 AAV/cell for 3-fold cross-validation. Overall, this work shows that DHM can be implemented for online monitoring of Sf9 concentration and viability, also permitting to monitor product titer, namely AAV, or culture progression in lytic systems, making it a valuable tool to support the time of harvest decision and for the establishment of controlled feeding strategies.
Collapse
|
40
|
A Heterologous Viral Protein Scaffold for Chimeric Antigen Design: An Example PCV2 Virus Vaccine Candidate. Viruses 2020; 12:v12040385. [PMID: 32244384 PMCID: PMC7232224 DOI: 10.3390/v12040385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 03/09/2020] [Accepted: 03/15/2020] [Indexed: 12/05/2022] Open
Abstract
Recombinant vaccines have low-cost manufacturing, regulatory requirements, and reduced side effects compared to attenuated or inactivated vaccines. In the porcine industry, post-weaning multisystemic disease syndrome generates economic losses, characterized by progressive weight loss and weakness in piglets, and it is caused by porcine circovirus type 2 (PCV2). We designed a chimeric antigen (Qm1) to assemble the main exposed epitopes of the Cap-PCV2 protein on the capsid protein of the tobacco necrosis virus (TNV). This design was based on the Cap-N-terminal of an isolated PCV2 virus obtained in Chile. The virus was characterized, and the sequence was clustered within the PCV2 genotype b clade. This chimeric protein was expressed as inclusion bodies in both monomeric and multimeric forms, suggesting a high-molecular-weight aggregate formation. Pigs immunized with Qm1 elicited a strong and specific antibody response, which reduced the viral loads after the PCV2 challenge. In conclusion, the implemented design allowed for the generation of an effective vaccine candidate. Our proposal could be used to express the domains or fragments of antigenic proteins, whose structural complexity does not allow for low-cost production in Escherichia coli. Hence, other antigen domains could be integrated into the TNV backbone for suitable antigenicity and immunogenicity. This work represents new biotechnological strategies, with a reduction in the costs associated with vaccine development.
Collapse
|
41
|
Montero DA, Del Canto F, Salazar JC, Céspedes S, Cádiz L, Arenas-Salinas M, Reyes J, Oñate Á, Vidal RM. Immunization of mice with chimeric antigens displaying selected epitopes confers protection against intestinal colonization and renal damage caused by Shiga toxin-producing Escherichia coli. NPJ Vaccines 2020; 5:20. [PMID: 32194997 PMCID: PMC7067774 DOI: 10.1038/s41541-020-0168-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 02/14/2020] [Indexed: 12/14/2022] Open
Abstract
Shiga toxin-producing Escherichia coli (STEC) cause diarrhea and dysentery, which may progress to hemolytic uremic syndrome (HUS). Vaccination has been proposed as a preventive approach against STEC infection; however, there is no vaccine for humans and those used in animals reduce but do not eliminate the intestinal colonization of STEC. The OmpT, Cah and Hes proteins are widely distributed among clinical STEC strains and are recognized by serum IgG and IgA in patients with HUS. Here, we develop a vaccine formulation based on two chimeric antigens containing epitopes of OmpT, Cah and Hes proteins against STEC strains. Intramuscular and intranasal immunization of mice with these chimeric antigens elicited systemic and local long-lasting humoral responses. However, the class of antibodies generated was dependent on the adjuvant and the route of administration. Moreover, while intramuscular immunization with the combination of the chimeric antigens conferred protection against colonization by STEC O157:H7, the intranasal conferred protection against renal damage caused by STEC O91:H21. This preclinical study supports the potential use of this formulation based on recombinant chimeric proteins as a preventive strategy against STEC infections.
Collapse
Affiliation(s)
- David A Montero
- 1Programa de Microbiología y Micología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile.,2Programa Disciplinario de Inmunología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Felipe Del Canto
- 1Programa de Microbiología y Micología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Juan C Salazar
- 1Programa de Microbiología y Micología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Sandra Céspedes
- 1Programa de Microbiología y Micología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Leandro Cádiz
- 1Programa de Microbiología y Micología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Mauricio Arenas-Salinas
- 3Centro de Bioinformática y Simulación Molecular, Facultad de Ingeniería, Universidad de Talca, Talca, Chile
| | - José Reyes
- 4Departamento de Microbiología, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Ángel Oñate
- 4Departamento de Microbiología, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Roberto M Vidal
- 1Programa de Microbiología y Micología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile.,5Instituto Milenio de Inmunología e Inmunoterapia, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| |
Collapse
|
42
|
Lye PY, Noor SM, Shohaimi SA, Junoh NF, Tan SC, Iwamoto S, Kotani E, Norazmi MN, Nagamine T, Mori H, Liew MW. Process development for quantitation and vaccine efficacy assessment of recombinant hemagglutinin-neuraminidase. Process Biochem 2020. [DOI: 10.1016/j.procbio.2019.11.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
43
|
Abstract
Baculoviruses are arthropod-specific, enveloped viruses with circular, supercoiled double-stranded deoxyribonucleic acid genomes. While many viruses are studied to seek solutions for their adverse impact on human, veterinary, and plant health, the study of baculoviruses was stimulated initially by their potential utility to control insect pests. Later, the utility of baculovirus as gene expression vectors was evidenced leading to numerous applications. Several strategies are employed to obtain recombinant viruses that express large quantities of heterologous proteins. A major step forward was the development of bacmid technology (the construction of bacterial artificial chromosomes containing the genome of the baculovirus) which allows the manipulation of the baculovirus genome in bacteria. With this technology, foreign genes can be introduced into the bacmid by homologous and site-directed recombination or by transposition. Baculoviruses have been used to explore fundamental questions in molecular biology such as the nature of programmed cell-death. Moreover, the ability of baculoviruses to transduce mammalian cells led to the consideration of their use as gene-therapy and vaccine vectors. Strategies for genetic engineering of baculoviruses have been developed to meet the requirements of new application areas. Display of foreign proteins on the surface of virions or in nucleocapsid structures, the assembly of expressed proteins to form virus-like particles or protein complexes have been explored and validated as vaccines. The aim of this chapter is to update the areas of application of the baculoviruses in protein expression, alternative vaccine designs and gene therapy of infectious diseases and genetic disorders. Finally, we review the baculovirus-derived products on the market and in the pipeline for biomedical and veterinary use.
Collapse
|
44
|
Dai X, Jian C, Na L, Wang X, Dai Y, Li D. Production and characterization of Hantaan virus-like particles from baculovirus expression system. Biochem Eng J 2019. [DOI: 10.1016/j.bej.2019.107373] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
45
|
The Pfs230 N-terminal fragment, Pfs230D1+: expression and characterization of a potential malaria transmission-blocking vaccine candidate. Malar J 2019; 18:356. [PMID: 31703583 PMCID: PMC6839146 DOI: 10.1186/s12936-019-2989-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Accepted: 10/24/2019] [Indexed: 12/31/2022] Open
Abstract
Background Control and elimination of malaria can be accelerated by transmission-blocking interventions such as vaccines. A surface antigen of Plasmodium falciparum gametocytes, Pfs230, is a leading vaccine target antigen, and has recently progressed to experimental clinical trials. To support vaccine product development, an N-terminal Pfs230 antigen was designed to increase yield, as well as to improve antigen quality, integrity, and homogeneity. Methods A scalable baculovirus expression system was used to express the Pfs230D1+ construct (aa 552–731), which was subsequently purified and analysed. Pfs230D1+ was designed to avoid glycosylation and protease digestion, thereby potentially increasing homogeneity and stability. The resulting Pfs230D1+ protein was compared to a previous iteration of the Pfs230 N-terminal domain, Pfs230C1 (aa 443–731), through physiochemical characterization and in vivo analysis. The induction of functional antibody responses was confirmed via the standard membrane feeding assay (SMFA). Results Pfs230D1+ was produced and purified to an overall yield of 23 mg/L culture supernatant, a twofold yield increase over Pfs230C1. The Pfs230D1+ protein migrated as a single band via SDS-PAGE and was detected by anti-Pfs230C1 monoclonal antibodies. Evaluation by SDS-PAGE, chromatography (size-exclusion and reversed phase) and capillary isoelectric focusing demonstrated the molecule had improved homogeneity in terms of size, conformation, and charge. Intact mass spectrometry confirmed its molecular weight and that it was free of glycosylation, a key difference to the prior Pfs230C1 protein. The correct formation of the two intramolecular disulfide bonds was initially inferred by binding of a conformation specific monoclonal antibody and directly confirmed by LC/MS and peptide mapping. When injected into mice the Pfs230D1+ protein elicited antibodies that demonstrated transmission-reducing activity, via SMFA, comparable to Pfs230C1. Conclusion By elimination of an O-glycosylation site, a potential N-glycosylation site, and two proteolytic cleavage sites, an improved N-terminal Pfs230 fragment was produced, termed D1+, which is non-glycosylated, homogeneous, and biologically active. An intact protein at higher yield than that previously observed for the Pfs230C1 fragment was achieved. The results indicate that Pfs230D1+ protein produced in the baculovirus expression system is an attractive antigen for transmission-blocking vaccine development.
Collapse
|
46
|
Yang Z, Ma Y, Zhao H, Yuan Y, Kim BYS. Nanotechnology platforms for cancer immunotherapy. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2019; 12:e1590. [PMID: 31696664 DOI: 10.1002/wnan.1590] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 08/22/2019] [Accepted: 08/23/2019] [Indexed: 12/18/2022]
Abstract
Various cancer therapies have advanced remarkably over the past decade. Unlike the direct therapeutic targeting of tumor cells, cancer immunotherapy is a new strategy that boosts the host's immune system to detect specific cancer cells for efficient elimination. Nanoparticles incorporating immunomodulatory agents can activate immune cells and modulate the tumor microenvironment to enhance antitumor immunity. Such nanoparticle-based cancer immunotherapies have received considerable attention and have been extensively studied in recent years. This review thus focuses on nanoparticle-based platforms (especially naturally derived nanoparticles and synthetic nanoparticles) utilized in recent advances; summarizes delivery systems that incorporate various immune-modulating agents, including peptides and nucleic acids, immune checkpoint inhibitors, and other small immunostimulating agents; and introduces combinational cancer immunotherapy with nanoparticles, especially nanoparticle-based photo-immunotherapy and nanoparticle-based chemo-immunotherapy. Undoubtedly, the recent studies introduced in this review prove that nanoparticle-incorporated cancer immunotherapy is a highly promising treatment modality for patients with cancer. Nonetheless further research is needed to solve safety concerns and improve efficacy of nanoplatform-based cancer immunotherapy for future clinical application. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease.
Collapse
Affiliation(s)
- Zhaogang Yang
- Department of Radiation Oncology, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Yifan Ma
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio
| | - Hai Zhao
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Yuan Yuan
- Engineering Research Center for Biomaterials of Ministry of Education, East China University of Science and Technology, Shanghai, China
| | - Betty Y S Kim
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
47
|
Pais DAM, Portela RMC, Carrondo MJT, Isidro IA, Alves PM. Enabling PAT in insect cell bioprocesses:
In situ
monitoring of recombinant adeno‐associated virus production by fluorescence spectroscopy. Biotechnol Bioeng 2019; 116:2803-2814. [DOI: 10.1002/bit.27117] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 04/22/2019] [Accepted: 07/09/2019] [Indexed: 12/25/2022]
Affiliation(s)
- Daniel A. M. Pais
- iBET, Instituto de Biologia Experimental e Tecnológica Oeiras Portugal
- Instituto de Tecnologia Química e Biológica António XavierUniversidade Nova de Lisboa Oeiras Portugal
| | - Rui M. C. Portela
- iBET, Instituto de Biologia Experimental e Tecnológica Oeiras Portugal
| | | | - Inês A. Isidro
- iBET, Instituto de Biologia Experimental e Tecnológica Oeiras Portugal
- Instituto de Tecnologia Química e Biológica António XavierUniversidade Nova de Lisboa Oeiras Portugal
| | - Paula M. Alves
- iBET, Instituto de Biologia Experimental e Tecnológica Oeiras Portugal
- Instituto de Tecnologia Química e Biológica António XavierUniversidade Nova de Lisboa Oeiras Portugal
| |
Collapse
|
48
|
Amann T, Schmieder V, Faustrup Kildegaard H, Borth N, Andersen MR. Genetic engineering approaches to improve posttranslational modification of biopharmaceuticals in different production platforms. Biotechnol Bioeng 2019; 116:2778-2796. [PMID: 31237682 DOI: 10.1002/bit.27101] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 03/27/2019] [Accepted: 06/18/2019] [Indexed: 12/18/2022]
Abstract
The number of approved biopharmaceuticals, where product quality attributes remain of major importance, is increasing steadily. Within the available variety of expression hosts, the production of biopharmaceuticals faces diverse limitations with respect to posttranslational modifications (PTM), while different biopharmaceuticals demand different forms and specifications of PTMs for proper functionality. With the growing toolbox of genetic engineering technologies, it is now possible to address general as well as host- or biopharmaceutical-specific product quality obstacles. In this review, we present diverse expression systems derived from mammalians, bacteria, yeast, plants, and insects as well as available genetic engineering tools. We focus on genes for knockout/knockdown and overexpression for meaningful approaches to improve biopharmaceutical PTMs and discuss their applicability as well as future trends in the field.
Collapse
Affiliation(s)
- Thomas Amann
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Valerie Schmieder
- acib GmbH-Austrian Centre of Industrial Biotechnology, Graz, Austria.,Department of Biotechnology, BOKU University of Natural Resources and Life Sciences, Vienna, Austria
| | - Helene Faustrup Kildegaard
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Nicole Borth
- Department of Biotechnology, BOKU University of Natural Resources and Life Sciences, Vienna, Austria
| | - Mikael Rørdam Andersen
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kongens Lyngby, Denmark
| |
Collapse
|
49
|
Deepak PR, Saravanan P, Biswal JK, Basagoudanavar SH, Dechamma HJ, Umapathi V, Sreenivasa BP, Tamilselvan RP, Krishnaswamy N, Zaffer I, Sanyal A. Generation of acid resistant virus like particles of vaccine strains of foot-and-mouth disease virus (FMDV). Biologicals 2019; 60:28-35. [PMID: 31221554 DOI: 10.1016/j.biologicals.2019.06.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Revised: 05/17/2019] [Accepted: 06/10/2019] [Indexed: 10/26/2022] Open
Abstract
Foot-and-mouth disease (FMD) is a contagious viral disease affecting cloven hoofed livestock. Insect cell expressed virus like particles (VLPs) are potential alternative to overcome the limitations of inactivated vaccine. However, at pH < 6.5, virus particles disassociate into pentameric structure resulting in loss of antigenicity. Accordingly, we generated seven mutant VLPs containing mutations in the structural genes of FMDV vaccine strains (N17D and/or H145Y for serotypes O/IND/R2/75 and Asia1/IND/63/72; and H142D for serotype A/IND/40/00) by PCR based site directed mutagenesis. Acid resistant VLPs produced by baculovirus expression system were tested for acid stability at pH 7.5, 6.5, 6.0 and 5.5 followed by reactivity in sandwich-ELISA (s-ELISA), which revealed mutant-1 (N17D) of serotype O and Asia1 retained the antigenicity in s-ELISA even at pH 5.5 as compared to other VLPs and wild-types. Further, the 75S empty capsids obtained in sucrose density gradient, when tested in liquid phase blocking ELISA (LPBE) in comparison to cell culture antigen indicated that the VLPs were stable at acidic pH. Transmission electron microscopy of OM-1 confirmed the intact morphology of the empty VLPs. It is concluded that acid resistant VLPs could be useful for developing new generation vaccine or diagnostic for FMDV.
Collapse
Affiliation(s)
- P R Deepak
- ICAR-Indian Veterinary Research Institute, Hebbal Campus, Bengaluru, Karnataka, 560 024, India
| | - P Saravanan
- ICAR-Indian Veterinary Research Institute, Hebbal Campus, Bengaluru, Karnataka, 560 024, India.
| | - J K Biswal
- ICAR-Directorate on Foot-and-Mouth Disease, FMD Vaccination Monitoring and Sero-surveillance Unit, Indian Veterinary Research Institute, Hebbal Campus, Bengaluru, Karnataka, 560 024, India
| | - S H Basagoudanavar
- ICAR-Indian Veterinary Research Institute, Hebbal Campus, Bengaluru, Karnataka, 560 024, India
| | - H J Dechamma
- ICAR-Indian Veterinary Research Institute, Hebbal Campus, Bengaluru, Karnataka, 560 024, India
| | - V Umapathi
- ICAR-Indian Veterinary Research Institute, Hebbal Campus, Bengaluru, Karnataka, 560 024, India
| | - B P Sreenivasa
- ICAR-Indian Veterinary Research Institute, Hebbal Campus, Bengaluru, Karnataka, 560 024, India
| | - R P Tamilselvan
- ICAR-Indian Veterinary Research Institute, Hebbal Campus, Bengaluru, Karnataka, 560 024, India
| | - N Krishnaswamy
- ICAR-Indian Veterinary Research Institute, Hebbal Campus, Bengaluru, Karnataka, 560 024, India
| | - I Zaffer
- ICAR-Indian Veterinary Research Institute, Hebbal Campus, Bengaluru, Karnataka, 560 024, India
| | - A Sanyal
- ICAR-Indian Veterinary Research Institute, Hebbal Campus, Bengaluru, Karnataka, 560 024, India
| |
Collapse
|
50
|
Mc Callum G, Arregui M, Smith I, Bracco L, Wolman F, Cascone O, Targovnik A, Miranda M. Recombinant protein purification in baculovirus-infected Rachiplusia nu larvae: An approach towards a rational design of downstream processing strategies based on chromatographic behavior of proteins. Protein Expr Purif 2019; 158:44-50. [DOI: 10.1016/j.pep.2019.02.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 02/13/2019] [Accepted: 02/13/2019] [Indexed: 12/11/2022]
|