1
|
Ahmed MM, Okesanya OJ, Ukoaka BM, Ibrahim AM, Lucero-Prisno DE. Vesicular Stomatitis Virus: Insights into Pathogenesis, Immune Evasion, and Technological Innovations in Oncolytic and Vaccine Development. Viruses 2024; 16:1933. [PMID: 39772239 PMCID: PMC11680291 DOI: 10.3390/v16121933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 11/22/2024] [Accepted: 12/16/2024] [Indexed: 01/11/2025] Open
Abstract
Vesicular stomatitis virus (VSV) represents a significant advancement in therapeutic medicine, offering unique molecular and cellular characteristics that make it exceptionally suitable for medical applications. The bullet-shaped morphology, RNA genome organization, and cytoplasmic replication strategy provide fundamental advantages for both vaccine development and oncolytic applications. VSV's interaction with host cells through the low-density lipoprotein receptor (LDL-R) and its sophisticated transcriptional regulation mechanisms enables precise control over therapeutic applications. The virus demonstrates remarkable versatility through its rapid replication cycle, robust immune response induction, and natural neurotropism. Recent technological innovations in VSV engineering have led to enhanced safety protocols and improved therapeutic modifications, particularly in cancer treatment. Attenuation strategies have successfully addressed safety concerns while maintaining the therapeutic efficacy of the virus. The molecular and cellular interactions of VSV, particularly its immune modulation capabilities and tumor-selective properties, have proven valuable in the development of targeted therapeutic strategies. This review explores these aspects, while highlighting the continuing evolution of VSV-based therapeutic approaches in precision medicine.
Collapse
Affiliation(s)
- Mohamed Mustaf Ahmed
- Faculty of Medicine and Health Sciences, SIMAD University, Mogadishu 252, Somalia
| | - Olalekan John Okesanya
- Department of Medical Laboratory Science, Neuropsychiatric Hospital, Aro, Abeokuta 110101, Nigeria;
| | | | - Adamu Muhammad Ibrahim
- Department of Immunology, School of Medical Laboratory Science, Usmanu Danfodiyo University, Sokoto 840001, Nigeria;
| | - Don Eliseo Lucero-Prisno
- Department of Global Health and Development, London School of Hygiene and Tropical Medicine, London WC1E 7HT, UK;
- Research and Innovation Office, Southern Leyte State University, Leyte 6500, Philippines
- Research and Development Office, Biliran Province State University, Biliran 6549, Philippines
| |
Collapse
|
2
|
Gómez-Márquez J. The Lithbea Domain. Adv Biol (Weinh) 2024; 8:e2300679. [PMID: 38386280 DOI: 10.1002/adbi.202300679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 02/09/2024] [Indexed: 02/23/2024]
Abstract
The tree of life is the evolutionary metaphor for the past and present connections of all cellular organisms. Today, to speak of biodiversity is not only to speak of archaea, bacteria, and eukaryotes, but they should also consider the "new biodiversity" that includes viruses and synthetic organisms, which represent the new forms of life created in laboratories. There is even a third group of artificial entities that, although not living systems, pretend to imitate the living. To embrace and organize all this new biodiversity, I propose the creation of a new domain, with the name Lithbea (from life-on-the-border entites) The criteria for inclusion as members of Lithbea are: i) the acellular nature of the living system, ii) its origin in laboratory manipulation, iii) showing new biological traits, iv) the presence of exogenous genetic elements, v) artificial or inorganic nature. Within Lithbea there are two subdomains: Virworld (from virus world) which includes all viruses, regarded as lifeless living systems, and classified according to the International Committee on Taxonomy of Viruses (ICTV), and ii) Humade (from human-made) which includes all synthetic organisms and artificial entities. The relationships of Lithbea members to the three classical woesian domains and their implications are briefly discussed.
Collapse
Affiliation(s)
- Jaime Gómez-Márquez
- Department of Biochemistry and Molecular Biology, University of Santiago de Compostela, Santiago de Compostela, Galicia, 15782, Spain
| |
Collapse
|
3
|
Mahdizade Ari M, Dadgar L, Elahi Z, Ghanavati R, Taheri B. Genetically Engineered Microorganisms and Their Impact on Human Health. Int J Clin Pract 2024; 2024:6638269. [PMID: 38495751 PMCID: PMC10944348 DOI: 10.1155/2024/6638269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Revised: 11/20/2023] [Accepted: 02/12/2024] [Indexed: 03/19/2024] Open
Abstract
The emergence of antibiotic-resistant strains, the decreased effectiveness of conventional therapies, and the side effects have led researchers to seek a safer, more cost-effective, patient-friendly, and effective method that does not develop antibiotic resistance. With progress in synthetic biology and genetic engineering, genetically engineered microorganisms effective in treatment, prophylaxis, drug delivery, and diagnosis have been developed. The present study reviews the types of genetically engineered bacteria and phages, their impacts on diseases, cancer, and metabolic and inflammatory disorders, the biosynthesis of these modified strains, the route of administration, and their effects on the environment. We conclude that genetically engineered microorganisms can be considered promising candidates for adjunctive treatment of diseases and cancers.
Collapse
Affiliation(s)
- Marzie Mahdizade Ari
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Microbial Biotechnology Research Centre, Iran University of Medical Sciences, Tehran, Iran
| | - Leila Dadgar
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Microbial Biotechnology Research Centre, Iran University of Medical Sciences, Tehran, Iran
| | - Zahra Elahi
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Microbial Biotechnology Research Centre, Iran University of Medical Sciences, Tehran, Iran
| | | | - Behrouz Taheri
- Department of Biotechnology, School of Medicine, Ahvaz Jundishapour University of medical Sciences, Ahvaz, Iran
| |
Collapse
|
4
|
Mateu MG, Valbuena A. Engineering and Bio/Nanotechnological Applications of Virus Particles. Subcell Biochem 2024; 105:823-878. [PMID: 39738964 DOI: 10.1007/978-3-031-65187-8_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
Abstract
Virus particles (VPs) are naturally evolved nanomachines. Their outstanding molecular structures, physical and chemical properties, and biological activities make them potentially useful for many biomedical or technological applications. Natural VPs such as virions or capsids must, however, be modified by genetic and/or chemical engineering in order to become adequate for many specific uses. We present first a general overview of the methods used for obtaining virions and viral capsids, and of genetic and chemical engineering approaches to suitably modify VPs. In the second part of the chapter, we present an updated overview on current or developing applications of engineered VPs as tools, materials, reagents, or nanodevices in biomedicine, biotechnology, or nanotechnology.
Collapse
Affiliation(s)
- Mauricio G Mateu
- Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), and Department of Molecular Biology, Universidad Autónoma de Madrid, Madrid, Spain.
| | - Alejandro Valbuena
- Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), and Department of Molecular Biology, Universidad Autónoma de Madrid, Madrid, Spain
| |
Collapse
|
5
|
Bárcena J, Zamora-Ceballos M, Blanco E. Design of Novel Vaccines Based on Virus-Like Particles. Subcell Biochem 2024; 105:785-821. [PMID: 39738963 DOI: 10.1007/978-3-031-65187-8_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
Abstract
Virus-like particles (VLPs) are formed by viral proteins that, when overexpressed, spontaneously self-assemble into particles that structurally are similar to infectious virus or subviral particles (e.g. the viral capsid). VLPs are appealing as vaccine candidates because their inherent properties (i.e. virus-sized, multimeric antigens, highly organised and repetitive structure, not infectious) are suitable for the induction of safe and efficient humoral and cellular immune responses. VLP-based vaccines have already been licensed for human and veterinary use, and many more vaccine candidates are currently in late stages of evaluation. Moreover, the development of VLPs as platforms for foreign antigen display has further broadened their potential applicability both as prophylactic and therapeutic vaccines. This chapter provides an overview on the design and use of VLPs for the development of new-generation vaccines.
Collapse
Affiliation(s)
- Juan Bárcena
- Centro de Investigación en Sanidad Animal (CISA-INIA/CSIC), Madrid, Spain.
| | | | - Esther Blanco
- Centro de Investigación en Sanidad Animal (CISA-INIA/CSIC), Madrid, Spain
| |
Collapse
|
6
|
Dogbey DM, Torres VES, Fajemisin E, Mpondo L, Ngwenya T, Akinrinmade OA, Perriman AW, Barth S. Technological advances in the use of viral and non-viral vectors for delivering genetic and non-genetic cargos for cancer therapy. Drug Deliv Transl Res 2023; 13:2719-2738. [PMID: 37301780 PMCID: PMC10257536 DOI: 10.1007/s13346-023-01362-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/29/2023] [Indexed: 06/12/2023]
Abstract
The burden of cancer is increasing globally. Several challenges facing its mainstream treatment approaches have formed the basis for the development of targeted delivery systems to carry and distribute anti-cancer payloads to their defined targets. This site-specific delivery of drug molecules and gene payloads to selectively target druggable biomarkers aimed at inducing cell death while sparing normal cells is the principal goal for cancer therapy. An important advantage of a delivery vector either viral or non-viral is the cumulative ability to penetrate the haphazardly arranged and immunosuppressive tumour microenvironment of solid tumours and or withstand antibody-mediated immune response. Biotechnological approaches incorporating rational protein engineering for the development of targeted delivery systems which may serve as vehicles for packaging and distribution of anti-cancer agents to selectively target and kill cancer cells are highly desired. Over the years, these chemically and genetically modified delivery systems have aimed at distribution and selective accumulation of drug molecules at receptor sites resulting in constant maintenance of high drug bioavailability for effective anti-tumour activity. In this review, we highlighted the state-of-the art viral and non-viral drug and gene delivery systems and those under developments focusing on cancer therapy.
Collapse
Affiliation(s)
- Dennis Makafui Dogbey
- South African Research Chair in Cancer Biotechnology, Division of Chemical and Systems Biology, Department of Integrative Biomedical Sciences, University of Cape Town, Cape Town, South Africa
| | | | - Emmanuel Fajemisin
- South African Research Chair in Cancer Biotechnology, Division of Chemical and Systems Biology, Department of Integrative Biomedical Sciences, University of Cape Town, Cape Town, South Africa
| | - Liyabona Mpondo
- South African Research Chair in Cancer Biotechnology, Division of Chemical and Systems Biology, Department of Integrative Biomedical Sciences, University of Cape Town, Cape Town, South Africa
| | - Takunda Ngwenya
- South African Research Chair in Cancer Biotechnology, Division of Chemical and Systems Biology, Department of Integrative Biomedical Sciences, University of Cape Town, Cape Town, South Africa
| | - Olusiji Alex Akinrinmade
- South African Research Chair in Cancer Biotechnology, Division of Chemical and Systems Biology, Department of Integrative Biomedical Sciences, University of Cape Town, Cape Town, South Africa
| | - Adam W Perriman
- School of Cellular and Molecular Medicine, University of Bristol, BS8 1TD, Bristol, UK
| | - Stefan Barth
- South African Research Chair in Cancer Biotechnology, Division of Chemical and Systems Biology, Department of Integrative Biomedical Sciences, University of Cape Town, Cape Town, South Africa.
- Medical Biotechnology and Immunotherapy Research Unit, Institute of Infectious Diseases and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa.
| |
Collapse
|
7
|
Tan JS, Jaffar Ali MNB, Gan BK, Tan WS. Next-generation viral nanoparticles for targeted delivery of therapeutics: Fundamentals, methods, biomedical applications, and challenges. Expert Opin Drug Deliv 2023; 20:955-978. [PMID: 37339432 DOI: 10.1080/17425247.2023.2228202] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 06/19/2023] [Indexed: 06/22/2023]
Abstract
INTRODUCTION Viral nanoparticles (VNPs) are virus-based nanocarriers that have been studied extensively and intensively for biomedical applications. However, their clinical translation is relatively low compared to the predominating lipid-based nanoparticles. Therefore, this article describes the fundamentals, challenges, and solutions of the VNP-based platform, which will leverage the development of next-generation VNPs. AREAS COVERED Different types of VNPs and their biomedical applications are reviewed comprehensively. Strategies and approaches for cargo loading and targeted delivery of VNPs are examined thoroughly. The latest developments in controlled release of cargoes from VNPs and their mechanisms are highlighted too. The challenges faced by VNPs in biomedical applications are identified, and solutions are provided to overcome them. EXPERT OPINION In the development of next-generation VNPs for gene therapy, bioimaging and therapeutic deliveries, focus must be given to reduce their immunogenicity, and increase their stability in the circulatory system. Modular virus-like particles (VLPs) which are produced separately from their cargoes or ligands before all the components are coupled can speed up clinical trials and commercialization. In addition, removal of contaminants from VNPs, cargo delivery across the blood brain barrier (BBB), and targeting of VNPs to organelles intracellularly are challenges that will preoccupy researchers in this decade.
Collapse
Affiliation(s)
- Jia Sen Tan
- Department of Microbiology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Muhamad Norizwan Bin Jaffar Ali
- Department of Microbiology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Bee Koon Gan
- Department of Biological Science, Faculty of Science, National University of Singapore, Singapore
| | - Wen Siang Tan
- Department of Microbiology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| |
Collapse
|
8
|
Marchetti L, Simon-Gracia L, Lico C, Mancuso M, Baschieri S, Santi L, Teesalu T. Targeting of Tomato Bushy Stunt Virus with a Genetically Fused C-End Rule Peptide. NANOMATERIALS (BASEL, SWITZERLAND) 2023; 13:1428. [PMID: 37111013 PMCID: PMC10143547 DOI: 10.3390/nano13081428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 04/18/2023] [Accepted: 04/19/2023] [Indexed: 06/19/2023]
Abstract
Homing peptides are widely used to improve the delivery of drugs, imaging agents, and nanoparticles (NPs) to their target sites. Plant virus-based particles represent an emerging class of structurally diverse nanocarriers that are biocompatible, biodegradable, safe, and cost-effective. Similar to synthetic NPs, these particles can be loaded with imaging agents and/or drugs and functionalized with affinity ligands for targeted delivery. Here we report the development of a peptide-guided Tomato Bushy Stunt Virus (TBSV)-based nanocarrier platform for affinity targeting with the C-terminal C-end rule (CendR) peptide, RPARPAR (RPAR). Flow cytometry and confocal microscopy demonstrated that the TBSV-RPAR NPs bind specifically to and internalize in cells positive for the peptide receptor neuropilin-1 (NRP-1). TBSV-RPAR particles loaded with a widely used anticancer anthracycline, doxorubicin, showed selective cytotoxicity on NRP-1-expressing cells. Following systemic administration in mice, RPAR functionalization conferred TBSV particles the ability to accumulate in the lung tissue. Collectively, these studies show the feasibility of the CendR-targeted TBSV platform for the precision delivery of payloads.
Collapse
Affiliation(s)
- Luca Marchetti
- Laboratory of Biomedical Technologies, Italian National Agency for New Technologies, Energy and Sustainable Economic Development, ENEA, Casaccia Research Center, Via Anguillarese 301, 00123 Rome, Italy
- Department of Agriculture and Forest Sciences (DAFNE), University of Tuscia, Via S. Camillo De Lellis, 01100 Viterbo, Italy
| | - Lorena Simon-Gracia
- Laboratory of Precision and Nanomedicine, Institute of Biomedicine and Translational Medicine, University of Tartu, Ravila 14b, 50090 Tartu, Estonia
| | - Chiara Lico
- Laboratory of Biotechnologies, Italian National Agency for New Technologies, Energy and Sustainable Economic Development, ENEA, Casaccia Research Center, Via Anguillarese 301, 00123 Rome, Italy
| | - Mariateresa Mancuso
- Laboratory of Biomedical Technologies, Italian National Agency for New Technologies, Energy and Sustainable Economic Development, ENEA, Casaccia Research Center, Via Anguillarese 301, 00123 Rome, Italy
| | - Selene Baschieri
- Laboratory of Biotechnologies, Italian National Agency for New Technologies, Energy and Sustainable Economic Development, ENEA, Casaccia Research Center, Via Anguillarese 301, 00123 Rome, Italy
| | - Luca Santi
- Department of Agriculture and Forest Sciences (DAFNE), University of Tuscia, Via S. Camillo De Lellis, 01100 Viterbo, Italy
| | - Tambet Teesalu
- Laboratory of Precision and Nanomedicine, Institute of Biomedicine and Translational Medicine, University of Tartu, Ravila 14b, 50090 Tartu, Estonia
- Materials Research Laboratory, University of California, Santa Barbara, CA 93106, USA
| |
Collapse
|
9
|
Zheng P, Yang Y, Fu Y, He J, Hu Y, Zheng X, Duan B, Wang M, Liu Q, Li W, Li D, Yang Y, Yang Z, Yang X, Huang W, Ma Y. Engineered Norovirus-Derived Nanoparticles as a Plug-and-Play Cancer Vaccine Platform. ACS NANO 2023; 17:3412-3429. [PMID: 36779845 DOI: 10.1021/acsnano.2c08840] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
In recent years, virus-derived self-assembled protein nanoparticles (NPs) have emerged as attractive antigen delivery platforms for developing both preventive and therapeutic vaccines. In this study, we exploited the genetically engineered Norovirus S domain (Nov-S) with SpyCatcher003 fused to the C-terminus to develop a robust, modular, and versatile NP-based carrier platform (Nov-S-Catcher003). The NPs can be conveniently armed in a plug-and-play pattern with SpyTag003-linked antigens. Nov-S-Catcher003 was efficiently expressed in Escherichia coli and self-assembled into highly uniform NPs with a purified protein yield of 97.8 mg/L. The NPs presented high stability at different maintained temperatures and after undergoing differing numbers of freeze-thaw cycles. Tumor vaccine candidates were easily obtained by modifying Nov-S-Catcher003 NPs with SpyTag003-linked tumor antigens. Nov-S-Catcher003-antigen NPs significantly promoted the maturation of bone marrow-derived dendritic cells in vitro and were capable of efficiently migrating to lymph nodes in vivo. In TC-1 and B16F10 tumor-bearing mice, the subcutaneous immunization of NPs elicited robust tumor-specific T-cell immunity, reshaped the tumor microenvironment, and inhibited tumor growth. In the TC-1 model, the NPs even completely abolished established tumors. In conclusion, the Nov-S-Catcher003 system is a promising delivery platform for facilitating the development of NP-based cancer vaccines.
Collapse
Affiliation(s)
- Peng Zheng
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650118, China
| | - Ying Yang
- Cell Biology & Molecular Biology Laboratory of Experimental Teaching Center, Faculty of Basic Medical Science, Kunming Medical University, Kunming 650500, China
| | - Yuting Fu
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650118, China
| | - Jinrong He
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650118, China
| | - Yongmao Hu
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650118, China
- School of Life Sciences, Yunnan University, Kunming 650091, China
| | - Xiao Zheng
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650118, China
- School of Life Sciences, Yunnan University, Kunming 650091, China
| | - Biao Duan
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650118, China
- Kunming Medical University, Kunming 650500, China
| | - Mengzhen Wang
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650118, China
| | - Qingwen Liu
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650118, China
- Kunming Medical University, Kunming 650500, China
| | - Weiran Li
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650118, China
| | - Duo Li
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650118, China
- Department of Acute Infectious Diseases Control and Prevention, Yunnan Provincial Centers for Disease Control and Prevention, Kunming 650034, China
| | - Ying Yang
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650118, China
| | - Zhongqian Yang
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650118, China
| | - Xu Yang
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650118, China
| | - Weiwei Huang
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650118, China
| | - Yanbing Ma
- Laboratory of Molecular Immunology, Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650118, China
| |
Collapse
|
10
|
Holz E, Darwish M, Tesar DB, Shatz-Binder W. A Review of Protein- and Peptide-Based Chemical Conjugates: Past, Present, and Future. Pharmaceutics 2023; 15:600. [PMID: 36839922 PMCID: PMC9959917 DOI: 10.3390/pharmaceutics15020600] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 02/04/2023] [Accepted: 02/07/2023] [Indexed: 02/12/2023] Open
Abstract
Over the past few decades, the complexity of molecular entities being advanced for therapeutic purposes has continued to evolve. A main propellent fueling innovation is the perpetual mandate within the pharmaceutical industry to meet the needs of novel disease areas and/or delivery challenges. As new mechanisms of action are uncovered, and as our understanding of existing mechanisms grows, the properties that are required and/or leveraged to enable therapeutic development continue to expand. One rapidly evolving area of interest is that of chemically enhanced peptide and protein therapeutics. While a variety of conjugate molecules such as antibody-drug conjugates, peptide/protein-PEG conjugates, and protein conjugate vaccines are already well established, others, such as antibody-oligonucleotide conjugates and peptide/protein conjugates using non-PEG polymers, are newer to clinical development. This review will evaluate the current development landscape of protein-based chemical conjugates with special attention to considerations such as modulation of pharmacokinetics, safety/tolerability, and entry into difficult to access targets, as well as bioavailability. Furthermore, for the purpose of this review, the types of molecules discussed are divided into two categories: (1) therapeutics that are enhanced by protein or peptide bioconjugation, and (2) protein and peptide therapeutics that require chemical modifications. Overall, the breadth of novel peptide- or protein-based therapeutics moving through the pipeline each year supports a path forward for the pursuit of even more complex therapeutic strategies.
Collapse
Affiliation(s)
- Emily Holz
- Department of Pharmaceutical Development, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Martine Darwish
- Department of Protein Chemistry, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Devin B. Tesar
- Department of Pharmaceutical Development, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Whitney Shatz-Binder
- Department of Pharmaceutical Development, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
- Department of Protein Chemistry, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| |
Collapse
|
11
|
Kim KR, Lee AS, Kim SM, Heo HR, Kim CS. Virus-like nanoparticles as a theranostic platform for cancer. Front Bioeng Biotechnol 2023; 10:1106767. [PMID: 36714624 PMCID: PMC9878189 DOI: 10.3389/fbioe.2022.1106767] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Accepted: 12/31/2022] [Indexed: 01/15/2023] Open
Abstract
Virus-like nanoparticles (VLPs) are natural polymer-based nanomaterials that mimic viral structures through the hierarchical assembly of viral coat proteins, while lacking viral genomes. VLPs have received enormous attention in a wide range of nanotechnology-based medical diagnostics and therapies, including cancer therapy, imaging, and theranostics. VLPs are biocompatible and biodegradable and have a uniform structure and controllable assembly. They can encapsulate a wide range of therapeutic and diagnostic agents, and can be genetically or chemically modified. These properties have led to sophisticated multifunctional theranostic platforms. This article reviews the current progress in developing and applying engineered VLPs for molecular imaging, drug delivery, and multifunctional theranostics in cancer research.
Collapse
Affiliation(s)
- Kyeong Rok Kim
- Graduate School of Biochemistry, Yeungnam University, Gyeongsan, South Korea
| | - Ae Sol Lee
- Graduate School of Biochemistry, Yeungnam University, Gyeongsan, South Korea
| | - Su Min Kim
- Graduate School of Biochemistry, Yeungnam University, Gyeongsan, South Korea
| | - Hye Ryoung Heo
- Senotherapy-Based Metabolic Disease Control Research Center, Yeungnam University, Gyeongsan, South Korea,*Correspondence: Chang Sup Kim, ; Hye Ryoung Heo,
| | - Chang Sup Kim
- Graduate School of Biochemistry, Yeungnam University, Gyeongsan, South Korea,School of Chemistry and Biochemistry, Yeungnam University, Gyeongsan, South Korea,*Correspondence: Chang Sup Kim, ; Hye Ryoung Heo,
| |
Collapse
|
12
|
Boonyakida J, Khoris IM, Nasrin F, Park EY. Improvement of Modular Protein Display Efficiency in SpyTag-Implemented Norovirus-like Particles. Biomacromolecules 2023; 24:308-318. [PMID: 36475654 DOI: 10.1021/acs.biomac.2c01150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Genetic fusion and chemical conjugation are the most common approaches for displaying a foreign protein on the surface of virus-like particles (VLPs); however, these methods may negatively affect the formation and stability of VLPs. Here, we aimed to develop a modular display platform for protein decoration on norovirus-like particles (NoV-LPs) by combining the NoV-LP scaffold with the SpyTag/SpyCatcher bioconjugation system, as the NoV-LP is an attractive protein nanoparticle to carry foreign proteins for various applications. The SpyTagged-NoV-LPs were prepared by introducing SpyTag peptide into the C-terminus of the norovirus VP1 protein. To increase surface exposure of the SpyTag peptide on the NoV-LPs, two or three repeated extension linkers (EAAAK) were inserted between the SpyTag peptide and VP1 protein. Fluorescence proteins, EGFP and mCherry, were fused to SpyCatcher and employed as SpyTag conjugation partners. These VP1-SpyTag variants and SpyCatcher-fused EGFP and mCherry were separately expressed in silkworm fat bodies and purified. This study reveals that adding an extension linker did not disrupt the VLP formation; instead, it increased the particle size by 4-6 nm. The conjugation efficiency of the VP1-SpyTag variants with the extended linker improved from ∼15-35 to ∼50-63% based on the densitometric analysis, while it was up to 77% based on an optical quantification of EGFP and mCherry. Results indicate that the linker causes the SpyTag peptides to be positioned further away from the C-termini of VP1 and potentially increases the exposure of the SpyTag to the outer surface of the NoV-LPs, allowing more SpyTag/SpyCatcher complex formation on the VLP surface. Our study provides a strategy for enhancing the conjugation efficiency of NoV-LP and demonstrates the platform's utility for developing vaccines or functional nanoparticles.
Collapse
Affiliation(s)
- Jirayu Boonyakida
- Research Institute of Green Science and Technology, Shizuoka University, 836 Ohya, Suruga ward, Shizuoka422-8529, Japan
| | - Indra Memdi Khoris
- Research Institute of Green Science and Technology, Shizuoka University, 836 Ohya, Suruga ward, Shizuoka422-8529, Japan
| | - Fahmida Nasrin
- Research Institute of Green Science and Technology, Shizuoka University, 836 Ohya, Suruga ward, Shizuoka422-8529, Japan
| | - Enoch Y Park
- Research Institute of Green Science and Technology, Shizuoka University, 836 Ohya, Suruga ward, Shizuoka422-8529, Japan
| |
Collapse
|
13
|
Chen J, Cong X. Surface-engineered nanoparticles in cancer immune response and immunotherapy: Current status and future prospects. Biomed Pharmacother 2023; 157:113998. [PMID: 36399829 DOI: 10.1016/j.biopha.2022.113998] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 11/05/2022] [Accepted: 11/09/2022] [Indexed: 11/17/2022] Open
Abstract
Cancer immunotherapy is a therapeutic strategy to inhibit tumor growth and metastasis by intervening in the immune response process. Strategies applied to cancer immunotherapy mainly include blocking immune checkpoints, adoptive transfer of engineered immune cells, cytokine therapy, cancer vaccines, and oncolytic virus infection. However, many factors, such as off-target side effects, immunosuppressive cell infiltration and/or upregulation of immune checkpoint expression, cancer cell heterogeneity, and lack of antigen presentation, affect the therapeutic effect of immunotherapy on cancer. To improve the efficacy of targeted immunotherapy and reduce off-target effects, over the past two decades, nanoparticle delivery platforms have been increasingly used in tumor immunotherapy. However, nanoparticles are still subject to biological barriers and biodistribution challenges, which limit their overall clinical potential. This has prompted a series of engineered nanoparticles to overcome specific obstacles and transfer the accumulation of payloads to tumor-infiltrating immune cells. In recent years, new techniques and chemical methods have been employed to modify or functionalize the surfaces of nanoparticles. This review discusses the recent progress of surface-engineered nanoparticles in inducing tumor immune responses and immunotherapy, as well as future directions for the development of next-generation nanomedicines.
Collapse
Affiliation(s)
- Jun Chen
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang 110000, Liaoning Province, China
| | - Xiufeng Cong
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang 110000, Liaoning Province, China.
| |
Collapse
|
14
|
Ma XY, Hill BD, Hoang T, Wen F. Virus-inspired strategies for cancer therapy. Semin Cancer Biol 2022; 86:1143-1157. [PMID: 34182141 PMCID: PMC8710185 DOI: 10.1016/j.semcancer.2021.06.021] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 06/17/2021] [Accepted: 06/23/2021] [Indexed: 01/27/2023]
Abstract
The intentional use of viruses for cancer therapy dates back over a century. As viruses are inherently immunogenic and naturally optimized delivery vehicles, repurposing viruses for drug delivery, tumor antigen presentation, or selective replication in cancer cells represents a simple and elegant approach to cancer treatment. While early virotherapy was fraught with harsh side effects and low response rates, virus-based therapies have recently seen a resurgence due to newfound abilities to engineer and tune oncolytic viruses, virus-like particles, and virus-mimicking nanoparticles for improved safety and efficacy. However, despite their great potential, very few virus-based therapies have made it through clinical trials. In this review, we present an overview of virus-inspired approaches for cancer therapy, discuss engineering strategies to enhance their mechanisms of action, and highlight their application for overcoming the challenges of traditional cancer therapies.
Collapse
Affiliation(s)
- Xiao Yin Ma
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, United States
| | - Brett D Hill
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, United States
| | - Trang Hoang
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, United States
| | - Fei Wen
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, United States.
| |
Collapse
|
15
|
González-Gamboa I, Caparco AA, McCaskill JM, Steinmetz NF. Bioconjugation Strategies for Tobacco Mild Green Mosaic Virus. Chembiochem 2022; 23:e202200323. [PMID: 35835718 PMCID: PMC9624232 DOI: 10.1002/cbic.202200323] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 07/03/2022] [Indexed: 11/06/2022]
Abstract
Tobacco mild green mosaic virus (TMGMV) is a plant virus closely related to Tobacco mosaic virus (TMV), sharing many of its structural and chemical features. These rod-shaped viruses, comprised of 2130 identical coat protein subunits, have been utilized as nanotechnological platforms for a myriad of applications, ranging from drug delivery to precision agriculture. This versatility for functionalization is due to their chemically active external and internal surfaces. While both viruses are similar, they do exhibit some key differences in their surface chemistry, suggesting the reactive residue distribution on TMGMV should not overlap with TMV. In this work, we focused on the establishment and refinement of chemical bioconjugation strategies to load molecules into or onto TMGMV for targeted delivery. A combination of NHS, EDC, and diazo coupling reactions in combination with click chemistry were used to modify the N-terminus, glutamic/aspartic acid residues, and tyrosines in TMGMV. We report loading with over 600 moieties per TMGMV via diazo-coupling, which is a >3-fold increase compared to previous studies. We also report that cargo can be loaded to the solvent-exposed N-terminus and carboxylates on the exterior/interior surfaces. Mass spectrometry revealed the most reactive sites to be Y12 and Y72, both tyrosine side chains are located on the exterior surface. For the carboxylates, interior E106 (66.53 %) was the most reactive for EDC-propargylamine coupled reactions, with the exterior E145 accounting for >15 % reactivity, overturning previous assumptions that only interior glutamic acid residues are accessible. A deeper understanding of the chemical properties of TMGMV further enables its functionalization and use as a multifunctional nanocarrier platform for applications in medicine and precision farming.
Collapse
Affiliation(s)
- Ivonne González-Gamboa
- Department of NanoEngineering, University of California, San Diego, 9500 Gilman Dr., La Jolla, CA 92093, USA
| | - Adam A Caparco
- Department of NanoEngineering, University of California, San Diego, 9500 Gilman Dr., La Jolla, CA 92093, USA
| | - Justin M McCaskill
- Department of NanoEngineering, University of California, San Diego, 9500 Gilman Dr., La Jolla, CA 92093, USA
| | - Nicole F Steinmetz
- Department of NanoEngineering, University of California, San Diego, 9500 Gilman Dr., La Jolla, CA 92093, USA
- Department of Bioengineering, University of California, San Diego, 9500 Gilman Dr., La Jolla, CA 92093, USA
- Department of Radiology, University of California, San Diego, 9500 Gilman Dr., La Jolla, CA 92093, USA
- Center for Nano-ImmunoEngineering, University of California, San Diego, 9500 Gilman Dr., La Jolla, CA 92093, USA
- Institute for Materials Discovery and Design, University of California, San Diego, 9500 Gilman Dr., La Jolla, CA 92093, USA
- Moores Cancer Center, University of California, San Diego, 9500 Gilman Dr., La Jolla, CA 92093, USA
| |
Collapse
|
16
|
Mejía-Méndez JL, Vazquez-Duhalt R, Hernández LR, Sánchez-Arreola E, Bach H. Virus-like Particles: Fundamentals and Biomedical Applications. Int J Mol Sci 2022; 23:8579. [PMID: 35955711 PMCID: PMC9369363 DOI: 10.3390/ijms23158579] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/25/2022] [Accepted: 07/29/2022] [Indexed: 02/04/2023] Open
Abstract
Nanotechnology is a fast-evolving field focused on fabricating nanoscale objects for industrial, cosmetic, and therapeutic applications. Virus-like particles (VLPs) are self-assembled nanoparticles whose intrinsic properties, such as heterogeneity, and highly ordered structural organization are exploited to prepare vaccines; imaging agents; construct nanobioreactors; cancer treatment approaches; or deliver drugs, genes, and enzymes. However, depending upon the intrinsic features of the native virus from which they are produced, the therapeutic performance of VLPs can vary. This review compiles the recent scientific literature about the fundamentals of VLPs with biomedical applications. We consulted different databases to present a general scenario about viruses and how VLPs are produced in eukaryotic and prokaryotic cell lines to entrap therapeutic cargo. Moreover, the structural classification, morphology, and methods to functionalize the surface of VLPs are discussed. Finally, different characterization techniques required to examine the size, charge, aggregation, and composition of VLPs are described.
Collapse
Affiliation(s)
- Jorge L. Mejía-Méndez
- Departamento de Ciencias Químico Biológicas, Universidad de las Américas Puebla, Santa Catarina Mártir s/n, Cholula 72810, Puebla, Mexico; (J.L.M.-M.); (L.R.H.); (E.S.-A.)
| | - Rafael Vazquez-Duhalt
- Centro de Nanociencias y Nanotecnología UNAM, Km 107 Carretera Tijuana-Ensenada, Ensenada 22860, Baja California, Mexico;
| | - Luis R. Hernández
- Departamento de Ciencias Químico Biológicas, Universidad de las Américas Puebla, Santa Catarina Mártir s/n, Cholula 72810, Puebla, Mexico; (J.L.M.-M.); (L.R.H.); (E.S.-A.)
| | - Eugenio Sánchez-Arreola
- Departamento de Ciencias Químico Biológicas, Universidad de las Américas Puebla, Santa Catarina Mártir s/n, Cholula 72810, Puebla, Mexico; (J.L.M.-M.); (L.R.H.); (E.S.-A.)
| | - Horacio Bach
- Department of Medicine, Division of Infectious Diseases, University of British Columbia, Vancouver, BC V6H 3Z6, Canada
| |
Collapse
|
17
|
Virus-like particles displaying conserved toxin epitopes stimulate polyspecific, murine antibody responses capable of snake venom recognition. Sci Rep 2022; 12:11328. [PMID: 35790745 PMCID: PMC9256628 DOI: 10.1038/s41598-022-13376-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 05/24/2022] [Indexed: 11/14/2022] Open
Abstract
Antivenom is currently the first-choice treatment for snakebite envenoming. However, only a low proportion of antivenom immunoglobulins are specific to venom toxins, resulting in poor dose efficacy and potency. We sought to investigate whether linear venom epitopes displayed on virus like particles can stimulate an antibody response capable of recognising venom toxins from diverse medically important species. Bioinformatically-designed epitopes, corresponding to predicted conserved regions of group I phospholipase A2 and three finger toxins, were engineered for display on the surface of hepatitis B core antigen virus like particles and used to immunise female CD1 mice over a 14 weeks. Antibody responses to all venom epitope virus like particles were detectable by ELISA by the end of the immunisation period, although total antibody and epitope specific antibody titres were variable against the different epitope immunogens. Immunoblots using pooled sera demonstrated recognition of various venom components in a diverse panel of six elapid venoms, representing three continents and four genera. Insufficient antibody yields precluded a thorough assessment of the neutralising ability of the generated antibodies, however we were able to test polyclonal anti-PLA2 IgG from three animals against the PLA2 activity of Naja nigricollis venom, all of which showed no neutralising ability. This study demonstrates proof-of-principle that virus like particles engineered to display conserved toxin linear epitopes can elicit specific antibody responses in mice which are able to recognise a geographically broad range of elapid venoms.
Collapse
|
18
|
Ren F, Yan J, Kontogiannatos D, Wang X, Li J, Swevers L, Sun J. Characterization of virus-like particles assembled by co-expression of BmCPV capsid shell protein and large protrusion protein. Int J Biol Macromol 2022; 209:1656-1664. [PMID: 35460752 DOI: 10.1016/j.ijbiomac.2022.04.097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 04/09/2022] [Accepted: 04/13/2022] [Indexed: 11/18/2022]
Abstract
Bombyx mori cytoplasmic polyhedrosis virus (BmCPV) is a typical single-layer capsid dsRNA virus belonging to the Reoviridae family of the Cypovirus genus. Previous studies have shown that the BmCPV major capsid shell protein (CSP) has the ability to self-assemble into virus-like particles (VLPs), and cryo-electron microscopy of the BmCPV virions has revealed a tight mutual binding region between CSP and another capsid protein known as the Large Protrusion Protein (LPP), which further stabilizes the capsid shell. In this study, the multi-gene baculovirus expression system, Ac-MultiBac, was used to produce both solely CSP-based and CSP-LPP co-assembled VLPs. Transmission electron microscopy (TEM) results showed that addition of LPP did not affect the assembly of VLPs resulting in almost identical structure in both cases. However, ex vivo administration of VLPs to silkworm midgut tissue showed that CSP-based VLPs did not induce a significant transcriptional response in the innate immunity and RNAi gene cascades, compared to the co-assembled CSP-LPP based VLPs and the natural BmCPV virions isolated from polyhedra. The experimental results indicate that CSP and LPP attach tightly ("Plug and Display" model with CSP acting as "catcher" and LPP as "tag") to form VLPs that have a structure similar to that of the native CPV virions. Moreover, our results showed that the formation of VLPs with the two BmCPV capsid proteins is feasible, which can form the basis for the production of BmCPV-based VLPs as a new type of biological material to display exogenous proteins.
Collapse
Affiliation(s)
- Feifei Ren
- Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Jiming Yan
- Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Dimitrios Kontogiannatos
- Insect Molecular Genetics and Biotechnology, Institute of Biosciences and Applications, National Centre for Scientific Research Demokritos, Aghia Paraskevi, Athens 15341, Greece
| | - Xiong Wang
- Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Jingyang Li
- Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Luc Swevers
- Insect Molecular Genetics and Biotechnology, Institute of Biosciences and Applications, National Centre for Scientific Research Demokritos, Aghia Paraskevi, Athens 15341, Greece.
| | - Jingchen Sun
- Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, China.
| |
Collapse
|
19
|
Jansen van Vuren P, Singanallur NB, Keck H, Eschbaumer M, Vosloo W. Chemical inactivation of foot-and-mouth disease virus in bovine tongue epithelium for safe transport and downstream processing. J Virol Methods 2022; 305:114539. [PMID: 35523370 DOI: 10.1016/j.jviromet.2022.114539] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 04/18/2022] [Accepted: 04/30/2022] [Indexed: 11/19/2022]
Abstract
Epithelial tissue or vesicular fluid from an unruptured or recently ruptured vesicle is the sample of choice for confirmatory laboratory diagnosis of foot-and-mouth disease (FMD). However, in 'FMD-free' countries the transport and downstream processing of such samples from potentially infected animals present a biosafety risk, particularly during heightened surveillance, potentially involving decentralised testing in laboratories without adequate biocontainment facilities. In such circumstances, rapid inactivation of virus, if present, prior to transport becomes a necessity, while still maintaining the integrity of diagnostic analytes. Tongue epithelium collected from cattle infected with FMD virus (FMDV) of serotype O (O/ALG/3/2014 - Lineage O/ME-SA/Ind-2001d) or A (A/IRN/22/2015 - Lineage A/ASIA/G-VII) was incubated in the PAXGene Tissue System Fixative (pH 4) and Stabiliser (pH 6.5) components respectively, in McIlvaine's citrate-phosphate buffer (pH 2.6) or in phosphate-buffered saline (PBS, pH 7.4) at room temperature for 2, 6, 24 or 48h. Following incubation, tissues were homogenised and tested by virus isolation and titration using LFBKαVβ6 cells. The integrity of FMD viral RNA was assessed by RT-qPCR (3Dpol coding region), Sanger sequencing of the VP1 region and transfection of LFBKαVβ6 cells to recover infectious virus. Viable virus could be recovered from samples incubated in PBS for at least 48hours. The PAXgene Tissue System Stabiliser component yielded variable results dependent on virus serotype, requiring at least 6hours of incubation to inactivate A/IRN/22/2015 in most samples, whereas the Fixative component required up to 2hours in some samples. McIlvaine's citrate-phosphate buffer rapidly inactivated both viruses within 2hours of incubation. There was no demonstrable degradation of FMD viral RNA resulting from incubation in any of the buffers for up to 48hours, as assessed by RT-qPCR, and 24hours by sequencing and transfection to recover infectious virus. McIlvaine's citrate-phosphate buffer (pH 2.6) is easy to prepare, inexpensive and inactivates serotype A and O FMDV in epithelial tissue within 2hours, while maintaining RNA integrity for downstream diagnostic processes and virus characterisation.
Collapse
Affiliation(s)
- Petrus Jansen van Vuren
- Australian Centre for Disease Preparedness, CSIRO Health and Biosecurity, 5 Portarlington rd, Geelong, VIC, Australia
| | | | - Hanna Keck
- National Reference Laboratory for FMD, Institute of Diagnostic Virology, Friedrich-Loeffler-Institut, Suedufer 10, 17493 Greifswald-Insel Riems, Germany
| | - Michael Eschbaumer
- National Reference Laboratory for FMD, Institute of Diagnostic Virology, Friedrich-Loeffler-Institut, Suedufer 10, 17493 Greifswald-Insel Riems, Germany
| | - Wilna Vosloo
- Australian Centre for Disease Preparedness, CSIRO Health and Biosecurity, 5 Portarlington rd, Geelong, VIC, Australia.
| |
Collapse
|
20
|
Twigg C, Wenk J. Review and Meta‐Analysis: SARS‐CoV‐2 and Enveloped Virus Detection in Feces and Wastewater. CHEMBIOENG REVIEWS 2022. [PMCID: PMC9083821 DOI: 10.1002/cben.202100039] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Detection and quantification of viruses supplies key information on their spread and allows risk assessment for public health. In wastewater, existing detection methods have been focusing on non‐enveloped enteric viruses due to enveloped virus transmission, such as coronaviruses, by the fecal‐oral route being less likely. Since the beginning of the SARS‐CoV‐2 pandemic, interest and importance of enveloped virus detection in wastewater has increased. Here, quantitative studies on SARS‐CoV‐2 occurrence in feces and raw wastewater and other enveloped viruses via quantitative real‐time reverse transcription polymerase chain reaction (RT‐qPCR) during the early stage of the pandemic until April 2021 are reviewed, including statistical evaluation of the positive detection rate and efficiency throughout the detection process involving concentration, extraction, and amplification stages. Optimized and aligned sampling protocols and concentration methods for enveloped viruses, along with SARS‐CoV‐2 surrogates, in wastewater environments may improve low and variable recovery rates providing increased detection efficiency and comparable data on viral load measured across different studies.
Collapse
Affiliation(s)
- Charlotte Twigg
- University of Bath Department of Chemical Engineering and Water Innovation and Research Centre (WIRC@Bath) Claverton Down BA2 7AY Bath Somerset United Kingdom
| | - Jannis Wenk
- University of Bath Department of Chemical Engineering and Water Innovation and Research Centre (WIRC@Bath) Claverton Down BA2 7AY Bath Somerset United Kingdom
| |
Collapse
|
21
|
Tornesello AL, Tagliamonte M, Buonaguro FM, Tornesello ML, Buonaguro L. Virus-like Particles as Preventive and Therapeutic Cancer Vaccines. Vaccines (Basel) 2022; 10:227. [PMID: 35214685 PMCID: PMC8879290 DOI: 10.3390/vaccines10020227] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/27/2022] [Accepted: 01/28/2022] [Indexed: 12/04/2022] Open
Abstract
Virus-like particles (VLPs) are self-assembled viral protein complexes that mimic the native virus structure without being infectious. VLPs, similarly to wild type viruses, are able to efficiently target and activate dendritic cells (DCs) triggering the B and T cell immunities. Therefore, VLPs hold great promise for the development of effective and affordable vaccines in infectious diseases and cancers. Vaccine formulations based on VLPs, compared to other nanoparticles, have the advantage of incorporating multiple antigens derived from different proteins. Moreover, such antigens can be functionalized by chemical modifications without affecting the structural conformation or the antigenicity. This review summarizes the current status of preventive and therapeutic VLP-based vaccines developed against human oncoviruses as well as cancers.
Collapse
Affiliation(s)
- Anna Lucia Tornesello
- Molecular Biology and Viral Oncology Unit, Istituto Nazionale Tumori IRCCS “Fondazione G. Pascale”, via Mariano Semmola, 80131 Napoli, Italy; (F.M.B.); (M.L.T.)
| | - Maria Tagliamonte
- Innovative Immunological Models, Istituto Nazionale Tumori IRCCS “Fondazione G. Pascale”, via Mariano Semmola, 80131 Napoli, Italy; (M.T.); (L.B.)
| | - Franco M. Buonaguro
- Molecular Biology and Viral Oncology Unit, Istituto Nazionale Tumori IRCCS “Fondazione G. Pascale”, via Mariano Semmola, 80131 Napoli, Italy; (F.M.B.); (M.L.T.)
| | - Maria Lina Tornesello
- Molecular Biology and Viral Oncology Unit, Istituto Nazionale Tumori IRCCS “Fondazione G. Pascale”, via Mariano Semmola, 80131 Napoli, Italy; (F.M.B.); (M.L.T.)
| | - Luigi Buonaguro
- Innovative Immunological Models, Istituto Nazionale Tumori IRCCS “Fondazione G. Pascale”, via Mariano Semmola, 80131 Napoli, Italy; (M.T.); (L.B.)
| |
Collapse
|
22
|
Incidence of Phage Capsid Organization on the Resistance to High Energy Proton Beams. APPLIED SCIENCES-BASEL 2022. [DOI: 10.3390/app12030988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
The helical geometry of virus capsid allows simple self-assembly of identical protein subunits with a low request of free energy and a similar spiral path to virus nucleic acid. Consequently, small variations in protein subunits can affect the stability of the entire phage particle. Previously, we observed that rearrangement in the capsid structure of M13 engineered phages affected the resistance to UV-C exposure, while that to H2O2 was mainly ascribable to the amino acids’ sequence of the foreign peptide. Based on these findings, in this work, the resistance to accelerated proton beam exposure (5.0 MeV energy) of the same phage clones was determined at different absorbed doses and dose rates. Then, the number of viral particles able to infect and replicate in the natural host, Escherichia coli F+, was evaluated. By comparing the results with the M13 wild-type vector (pC89), we observed that 12III1 phage clones, with the foreign peptide containing amino acids favorable to carbonylation, exhibited the highest reduction in phage titer associated with a radiation damage (RD) of 35 × 10−3/Gy at 50 dose Gy. On the other hand, P9b phage clones, containing amino acids unfavorable to carbonylation, showed the lowest reduction with an RD of 4.83 × 10−3/Gy at 500 dose Gy. These findings could improve the understanding of the molecular mechanisms underlying the radiation resistance of viruses
Collapse
|
23
|
Li X, Pan C, Sun P, Peng Z, Feng E, Wu J, Wang H, Zhu L. Orthogonal modular biosynthesis of nanoscale conjugate vaccines for vaccination against infection. NANO RESEARCH 2022; 15:1645-1653. [PMID: 34405037 PMCID: PMC8359766 DOI: 10.1007/s12274-021-3713-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 06/17/2021] [Accepted: 06/24/2021] [Indexed: 05/21/2023]
Abstract
UNLABELLED Conjugate vaccines represent one of the most effective means for controlling the occurrence of bacterial diseases. Although nanotechnology has been greatly applied in the field of vaccines, it is seldom used for conjugate vaccine research because it is very difficult to connect polysaccharides and nanocarriers. In this work, an orthogonal and modular biosynthesis method was used to produce nanoconjugate vaccines using the SpyTag/SpyCatcher system. When SpyTag/SpyCatcher system is combined with protein glycosylation technology, bacterial O-polysaccharide obtained from Shigela flexneri 2a can be conjugated onto the surfaces of different virus-like particles (VLPs) in a biocompatible and controlled manner. After confirming the excellent lymph node targeting and humoral immune activation abilities, these nanoconjugate vaccines further induced efficient prophylactic effects against infection in a mouse model. These results demonstrated that natural polysaccharide antigens can be easily connected to VLPs to prepare highly efficient nanoconjugate vaccines. To the best of the researchers' knowledge, this is the first time VLP-based nanoconjugate vaccines are produced efficiently, and this strategy could be applied to develop various pathogenic nanoconjugate vaccines. ELECTRONIC SUPPLEMENTARY MATERIAL Supplementary material (Figs. S1-S9) is available in the online version of this article at 10.1007/s12274-021-3713-4.
Collapse
Affiliation(s)
- Xin Li
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing, 100071 China
| | - Chao Pan
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing, 100071 China
| | - Peng Sun
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing, 100071 China
| | - Zhehui Peng
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing, 100071 China
| | - Erling Feng
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing, 100071 China
| | - Jun Wu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing, 100071 China
| | - Hengliang Wang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing, 100071 China
| | - Li Zhu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing, 100071 China
| |
Collapse
|
24
|
Higgs PL, Appleton JL, Turnbull WB, Fulton DA. Exploiting the Structural Metamorphosis of Polymers to 'Wrap' Micron-Sized Spherical Objects. Chemistry 2021; 27:17647-17654. [PMID: 34665484 DOI: 10.1002/chem.202103216] [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: 09/04/2021] [Indexed: 11/07/2022]
Abstract
There is growing interest in developing methods to 'wrap' nano- and micron-sized biological objects within films that may offer protection, enhance their stability or improve performance. We describe the successful 'wrapping' of lectin-decorated microspheres, which serve as appealing model micron-sized objects, within cross-linked polymer film. This approach utilizes polymer chains able to undergo a structural metamorphosis, from being intramolecularly cross-linked to intermolecularly cross-linked, a process that is triggered by polymer concentration upon the particle surface. Experiments demonstrate that both complementary molecular recognition and the dynamic covalent nature of the crosslinker are required for successful 'wrapping' to occur. This work is significant as it suggests that nano- and micron-sized biological objects such as virus-like particles, bacteria or mammalian cells-all of which may benefit from additional environmental protection or stabilization in emerging applications-may also be 'wrapped' by this approach.
Collapse
Affiliation(s)
- Patrick L Higgs
- Chemistry-School of Natural and Environmental Sciences, Bedson Building, Newcastle University, Newcastle upon Tyne, NE1 7RU, UK
| | - Jordan L Appleton
- Chemistry-School of Natural and Environmental Sciences, Bedson Building, Newcastle University, Newcastle upon Tyne, NE1 7RU, UK
| | - W Bruce Turnbull
- School of Chemistry and Astbury Centre for Structural MolecularBiology, University of Leeds, Leeds, LS2 9JT, UK
| | - David A Fulton
- Chemistry-School of Natural and Environmental Sciences, Bedson Building, Newcastle University, Newcastle upon Tyne, NE1 7RU, UK
| |
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
|
Chimeric virus-like particles (VLPs) designed from shrimp nodavirus (MrNV) capsid protein specifically target EGFR-positive human colorectal cancer cells. Sci Rep 2021; 11:16579. [PMID: 34400669 PMCID: PMC8367941 DOI: 10.1038/s41598-021-95891-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 07/30/2021] [Indexed: 12/24/2022] Open
Abstract
Recombinant MrNV capsid protein has been shown to effectively deliver plasmid DNA and dsRNA into Sf9 insect cells and shrimp tissues. To extend its application to cancer cell-targeting drug delivery, we created three different types of chimeric MrNV virus-like particles (VLPs) (R-MrNV, I-MrNV, and E-MrNV) that have specificity toward the epidermal growth factor receptor (EGFR), a cancer cell biomarker, by incorporating the EGFR-specific GE11 peptide at 3 different locations within the host cell recognition site of the capsid. All three chimeric MrNV-VLPs preserved the ability to form a mulberry-like VLP structure and to encapsulate EGFP DNA plasmid with an efficiency comparable to that previously reported for normal MrNV (N-MrNV). Compared to N-MrNV, the chimeric R-MrNV and E-MrNV carrying the exposed GE-11 peptide showed a significantly enhanced binding and internalization abilities that were specific towards EGFR expression in colorectal cancer cells (SW480). Specific targeting of chimeric MrNV to EGFR was proven by both EGFR silencing with siRNA vector and a competition with excess GE-11 peptide as well as the use of EGFR-negative colorectal cells (SW620) and breast cancer cells (MCF7). We demonstrated here that both chimeric R-MrNV and E-MrNV could be used to encapsulate cargo such as exogenous DNA and deliver it specifically to EGFR-positive cells. Our study presents the potential use of surface-modified VLPs of shrimp virus origin as nanocontainers for targeted cancer drug delivery.
Collapse
|
27
|
Rangel G, Bárcena J, Moreno N, Mata CP, Castón JR, Alejo A, Blanco E. Chimeric RHDV Virus-Like Particles Displaying Foot-and-Mouth Disease Virus Epitopes Elicit Neutralizing Antibodies and Confer Partial Protection in Pigs. Vaccines (Basel) 2021; 9:vaccines9050470. [PMID: 34066934 PMCID: PMC8148555 DOI: 10.3390/vaccines9050470] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 04/29/2021] [Accepted: 05/03/2021] [Indexed: 12/29/2022] Open
Abstract
Currently there is a clear trend towards the establishment of virus-like particles (VLPs) as a powerful tool for vaccine development. VLPs are tunable nanoparticles that can be engineered to be used as platforms for multimeric display of foreign antigens. We have previously reported that VLPs derived from rabbit hemorrhagic disease virus (RHDV) constitute an excellent vaccine vector, capable of inducing specific protective immune responses against inserted heterologous T-cytotoxic and B-cell epitopes. Here, we evaluate the ability of chimeric RHDV VLPs to elicit immune response and protection against Foot-and-Mouth disease virus (FMDV), one of the most devastating livestock diseases. For this purpose, we generated a set of chimeric VLPs containing two FMDV-derived epitopes: a neutralizing B-cell epitope (VP1 (140-158)) and a T-cell epitope [3A (21-35)]. The epitopes were inserted joined or individually at two different locations within the RHDV capsid protein. The immunogenicity and protection potential of the chimeric VLPs were analyzed in the mouse and pig models. Herein we show that the RHDV engineered VLPs displaying FMDV-derived epitopes elicit a robust neutralizing immune response in mice and pigs, affording partial clinical protection against an FMDV challenge in pigs.
Collapse
Affiliation(s)
- Giselle Rangel
- Centro de Investigación en Sanidad Animal (CISA, CSIC-INIA), Valdeolmos, 28130 Madrid, Spain; (G.R.); (J.B.); (N.M.); (A.A.)
| | - Juan Bárcena
- Centro de Investigación en Sanidad Animal (CISA, CSIC-INIA), Valdeolmos, 28130 Madrid, Spain; (G.R.); (J.B.); (N.M.); (A.A.)
| | - Noelia Moreno
- Centro de Investigación en Sanidad Animal (CISA, CSIC-INIA), Valdeolmos, 28130 Madrid, Spain; (G.R.); (J.B.); (N.M.); (A.A.)
| | - Carlos P. Mata
- Department of Structure of Macromolecules, Centro Nacional de Biotecnología/CSIC, Cantoblanco, 28049 Madrid, Spain; (C.P.M.); (J.R.C.)
| | - José R. Castón
- Department of Structure of Macromolecules, Centro Nacional de Biotecnología/CSIC, Cantoblanco, 28049 Madrid, Spain; (C.P.M.); (J.R.C.)
| | - Alí Alejo
- Centro de Investigación en Sanidad Animal (CISA, CSIC-INIA), Valdeolmos, 28130 Madrid, Spain; (G.R.); (J.B.); (N.M.); (A.A.)
| | - Esther Blanco
- Centro de Investigación en Sanidad Animal (CISA, CSIC-INIA), Valdeolmos, 28130 Madrid, Spain; (G.R.); (J.B.); (N.M.); (A.A.)
- Correspondence: ; Tel.: +34-916-202-300
| |
Collapse
|
28
|
Machhi J, Shahjin F, Das S, Patel M, Abdelmoaty MM, Cohen JD, Singh PA, Baldi A, Bajwa N, Kumar R, Vora LK, Patel TA, Oleynikov MD, Soni D, Yeapuri P, Mukadam I, Chakraborty R, Saksena CG, Herskovitz J, Hasan M, Oupicky D, Das S, Donnelly RF, Hettie KS, Chang L, Gendelman HE, Kevadiya BD. Nanocarrier vaccines for SARS-CoV-2. Adv Drug Deliv Rev 2021; 171:215-239. [PMID: 33428995 PMCID: PMC7794055 DOI: 10.1016/j.addr.2021.01.002] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Revised: 12/18/2020] [Accepted: 01/01/2021] [Indexed: 02/07/2023]
Abstract
The SARS-CoV-2 global pandemic has seen rapid spread, disease morbidities and death associated with substantive social, economic and societal impacts. Treatments rely on re-purposed antivirals and immune modulatory agents focusing on attenuating the acute respiratory distress syndrome. No curative therapies exist. Vaccines remain the best hope for disease control and the principal global effort to end the pandemic. Herein, we summarize those developments with a focus on the role played by nanocarrier delivery.
Collapse
Affiliation(s)
- Jatin Machhi
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, NE 68198, USA
| | - Farah Shahjin
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, NE 68198, USA
| | - Srijanee Das
- Department of Pathology and Microbiology, College of Medicine, University of Nebraska Medical Center, NE 68198, USA
| | - Milankumar Patel
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, NE 68198, USA
| | - Mai Mohamed Abdelmoaty
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, NE 68198, USA; Therapeutic Chemistry Department, Pharmaceutical and Drug Industries Research Division, National Research Centre, Giza, Egypt
| | - Jacob D Cohen
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, NE 68198, USA
| | - Preet Amol Singh
- Department of Pharmaceutical Sciences & Technology, Maharaja Ranjit Singh Punjab Technical University, Bathinda, Punjab, India
| | - Ashish Baldi
- Department of Pharmaceutical Sciences & Technology, Maharaja Ranjit Singh Punjab Technical University, Bathinda, Punjab, India
| | - Neha Bajwa
- Department of Pharmaceutical Sciences & Technology, Maharaja Ranjit Singh Punjab Technical University, Bathinda, Punjab, India
| | - Raj Kumar
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Lalit K Vora
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, United Kingdom
| | - Tapan A Patel
- Department of Biological Sciences, P. D. Patel Institute of Applied Sciences (PDPIAS), Charotar University of Science and Technology (CHARUSAT), Changa, Anand 388421, Gujarat, India
| | - Maxim D Oleynikov
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, NE 68198, USA
| | - Dhruvkumar Soni
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, NE 68198, USA
| | - Pravin Yeapuri
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, NE 68198, USA
| | - Insiya Mukadam
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, NE 68198, USA
| | - Rajashree Chakraborty
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, NE 68198, USA
| | - Caroline G Saksena
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, NE 68198, USA
| | - Jonathan Herskovitz
- Department of Pathology and Microbiology, College of Medicine, University of Nebraska Medical Center, NE 68198, USA
| | - Mahmudul Hasan
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, NE 68198, USA
| | - David Oupicky
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Suvarthi Das
- Department of Medicine, Stanford Medical School, Stanford University, Palo Alto, CA 94304, USA
| | - Ryan F Donnelly
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, United Kingdom
| | - Kenneth S Hettie
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, Department of Otolaryngology - Head & Neck Surgery, Stanford University, Palo Alto, CA 94304, USA
| | - Linda Chang
- Departments of Diagnostic Radiology & Nuclear Medicine, and Neurology, University of Maryland, School of Medicine, Baltimore, MD 21201, USA
| | - Howard E Gendelman
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, NE 68198, USA; Department of Pathology and Microbiology, College of Medicine, University of Nebraska Medical Center, NE 68198, USA; Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, NE 68198, USA.
| | - Bhavesh D Kevadiya
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, NE 68198, USA
| |
Collapse
|
29
|
Plano LMD, Franco D, Rizzo MG, Zammuto V, Gugliandolo C, Silipigni L, Torrisi L, Guglielmino SPP. Role of Phage Capsid in the Resistance to UV-C Radiations. Int J Mol Sci 2021; 22:3408. [PMID: 33810266 PMCID: PMC8037334 DOI: 10.3390/ijms22073408] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 03/22/2021] [Accepted: 03/23/2021] [Indexed: 11/24/2022] Open
Abstract
The conformational variation of the viral capsid structure plays an essential role both for the environmental resistance and acid nuclear release during cellular infection. The aim of this study was to evaluate how capsid rearrangement in engineered phages of M13 protects viral DNA and peptide bonds from damage induced by UV-C radiation. From in silico 3D modelling analysis, two M13 engineered phage clones, namely P9b and 12III1, were chosen for (i) chemical features of amino acids sequences, (ii) rearrangements in the secondary structure of their pVIII proteins and (iii) in turn the interactions involved in phage capsid. Then, their resistance to UV-C radiation and hydrogen peroxide (H2O2) was compared to M13 wild-type vector (pC89) without peptide insert. Results showed that both the phage clones acquired an advantage against direct radiation damage, due to a reorganization of interactions in the capsid for an increase of H-bond and steric interactions. However, only P9b had an increase in resistance against H2O2. These results could help to understand the molecular mechanisms involved in the stability of new virus variants, also providing quick and necessary information to develop effective protocols in the virus inactivation for human activities, such as safety foods and animal-derived materials.
Collapse
Affiliation(s)
- Laura Maria De Plano
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy; (L.M.D.P.); (M.G.R.); (V.Z.); (C.G.); (S.P.P.G.)
| | - Domenico Franco
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy; (L.M.D.P.); (M.G.R.); (V.Z.); (C.G.); (S.P.P.G.)
| | - Maria Giovanna Rizzo
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy; (L.M.D.P.); (M.G.R.); (V.Z.); (C.G.); (S.P.P.G.)
| | - Vincenzo Zammuto
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy; (L.M.D.P.); (M.G.R.); (V.Z.); (C.G.); (S.P.P.G.)
| | - Concetta Gugliandolo
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy; (L.M.D.P.); (M.G.R.); (V.Z.); (C.G.); (S.P.P.G.)
| | - Letteria Silipigni
- Department of Mathematical and Computational Sciences, Physical Sciences and Earth Sciences, University of Messina, 98166 Messina, Italy; (L.S.); (L.T.)
| | - Lorenzo Torrisi
- Department of Mathematical and Computational Sciences, Physical Sciences and Earth Sciences, University of Messina, 98166 Messina, Italy; (L.S.); (L.T.)
| | - Salvatore P. P. Guglielmino
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy; (L.M.D.P.); (M.G.R.); (V.Z.); (C.G.); (S.P.P.G.)
| |
Collapse
|
30
|
Tan FH, Kong JC, Ng JF, Alitheen NB, Wong CL, Yong CY, Lee KW. Recombinant turnip yellow mosaic virus coat protein as a potential nanocarrier. J Appl Microbiol 2021; 131:2072-2080. [PMID: 33629458 DOI: 10.1111/jam.15048] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 02/17/2021] [Accepted: 02/19/2021] [Indexed: 01/13/2023]
Abstract
AIMS To display a short peptide (GSRSHHHHHH) at the C-terminal end of turnip yellow mosaic virus coat protein (TYMVc) and to study its assembly into virus-like particles (TYMVcHis6 VLPs). METHODS AND RESULTS In this study, recombinant TYMVcHis6 expressed in Escherichia coli self-assembled into VLPs of approximately 30-32 nm. SDS-PAGE and Western blot analysis of protein fractions from the immobilized metal affinity chromatography (IMAC) showed that TYMVcHis6 VLPs interacted strongly with nickel ligands in IMAC column, suggesting that the fusion peptide is protruding out from the surface of VLPs. These VLPs are highly stable over a wide pH range from 3·0 to 11·0 at different temperatures. At pH 11·0, specifically, the VLPs remained intact up to 75°C. Additionally, the disassembly and reassembly of TYMVcHis6 VLPs were studied in vitro. Dynamic light scattering and transmission electron microscopy analysis revealed that TYMVcHis6 VLPs were dissociated by 7 mol l-1 urea and 2 mol l-1 guanidine hydrochloride (GdnHCl) without impairing their reassembly property. CONCLUSIONS A 10-residue peptide was successfully displayed on the surface of TYMVcHis6 VLPs. This chimera demonstrated high stability under extreme thermal conditions with varying pH and was able to dissociate and reassociate into VLPs by chemical denaturants. SIGNIFICANCE AND IMPACT OF THE STUDY This is the first C-terminally modified TYMVc produced in E. coli. The C-terminal tail which is exposed on the surface can be exploited as a useful site to display multiple copies of functional ligands. The ability of the chimeric VLPs to self-assemble after undergo chemical denaturation indicates its potential role to serve as a nanocarrier for use in targeted drug delivery.
Collapse
Affiliation(s)
- F H Tan
- School of Biosciences, Faculty of Health and Medical Sciences, Taylor's University, Subang Jaya, Selangor, Malaysia
| | - J C Kong
- School of Biosciences, Faculty of Health and Medical Sciences, Taylor's University, Subang Jaya, Selangor, Malaysia
| | - J F Ng
- School of Biosciences, Faculty of Health and Medical Sciences, Taylor's University, Subang Jaya, Selangor, Malaysia
| | - N B Alitheen
- Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - C L Wong
- Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - C Y Yong
- Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - K W Lee
- School of Biosciences, Faculty of Health and Medical Sciences, Taylor's University, Subang Jaya, Selangor, Malaysia
| |
Collapse
|
31
|
Wu Y, Li J, Shin HJ. Self-assembled Viral Nanoparticles as Targeted Anticancer Vehicles. BIOTECHNOL BIOPROC E 2021; 26:25-38. [PMID: 33584104 PMCID: PMC7872722 DOI: 10.1007/s12257-020-0383-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 01/04/2021] [Accepted: 01/06/2021] [Indexed: 12/31/2022]
Abstract
Viral nanoparticles (VNPs) comprise a variety of mammalian viruses, plant viruses, and bacteriophages, that have been adopted as building blocks and supra-molecular templates in nanotechnology. VNPs demonstrate the dynamic, monodisperse, polyvalent, and symmetrical architectures which represent examples of such biological templates. These programmable scaffolds have been exploited for genetic and chemical manipulation for displaying of targeted moieties together with encapsulation of various payloads for diagnosis or therapeutic intervention. The drug delivery system based on VNPs offer diverse advantages over synthetic nanoparticles, including biocompatibility, biodegradability, water solubility, and high uptake capability. Here we summarize the recent progress of VNPs especially as targeted anticancer vehicles from the encapsulation and surface modification mechanisms, involved viruses and VNPs, to their application potentials.
Collapse
Affiliation(s)
- Yuanzheng Wu
- Ecology Institute, Qilu University of Technology (Shandong Academy of Sciences), Shandong Provincial Key Laboratory of Applied Microbiology, Jinan, 250103 China
| | - Jishun Li
- Ecology Institute, Qilu University of Technology (Shandong Academy of Sciences), Shandong Provincial Key Laboratory of Applied Microbiology, Jinan, 250103 China
| | - Hyun-Jae Shin
- Department of Biochemical and Polymer Engineering, Chosun University, Gwangju, 61452 Korea
| |
Collapse
|
32
|
Chan SK, Du P, Ignacio C, Mehta S, Newton IG, Steinmetz NF. Biomimetic Virus-Like Particles as Severe Acute Respiratory Syndrome Coronavirus 2 Diagnostic Tools. ACS NANO 2021; 15:1259-1272. [PMID: 33237727 PMCID: PMC7724985 DOI: 10.1021/acsnano.0c08430] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 11/19/2020] [Indexed: 05/07/2023]
Abstract
Coronavirus disease 2019 (COVID-19) is a highly transmissible disease that has affected more than 90% of the countries worldwide. At least 17 million individuals have been infected, and some countries are still battling first or second waves of the pandemic. Nucleic acid tests, especially reverse transcription polymerase chain reaction (RT-PCR), have become the workhorse for early detection of COVID-19 infection. Positive controls for the molecular assays have been developed to validate each test and to provide high accuracy. However, most available positive controls require cold-chain distribution and cannot serve as full-process control. To overcome these shortcomings, we report the production of biomimetic virus-like particles (VLPs) as SARS-CoV-2 positive controls. A SARS-CoV-2 detection module for RT-PCR was encapsidated into VLPs from a bacteriophage and a plant virus. The chimeric VLPs were obtained either by in vivo reconstitution and coexpression of the target detection module and coat proteins or by in vitro assembly of purified detection module RNA sequences and coat proteins. These VLP-based positive controls mimic SARS-CoV-2 packaged ribonucleic acid (RNA) while being noninfectious. Most importantly, we demonstrated that the positive controls are scalable, stable, and can serve broadly as controls, from RNA extraction to PCR in clinical settings.
Collapse
Affiliation(s)
- Soo Khim Chan
- Department of NanoEngineering,
Department of Medicine,
Department of Radiology,
Department of Bioengineering,
Center for Nano-ImmunoEngineering,
Moores Cancer Center, Institute for
Materials Discovery and Design, and Veterans
Administration San Diego Healthcare System, University of
California San Diego, 9500 Gilman Drive, La Jolla,
California 92039, United States
| | - Pinyi Du
- Department of NanoEngineering,
Department of Medicine,
Department of Radiology,
Department of Bioengineering,
Center for Nano-ImmunoEngineering,
Moores Cancer Center, Institute for
Materials Discovery and Design, and Veterans
Administration San Diego Healthcare System, University of
California San Diego, 9500 Gilman Drive, La Jolla,
California 92039, United States
| | - Caroline Ignacio
- Department of NanoEngineering,
Department of Medicine,
Department of Radiology,
Department of Bioengineering,
Center for Nano-ImmunoEngineering,
Moores Cancer Center, Institute for
Materials Discovery and Design, and Veterans
Administration San Diego Healthcare System, University of
California San Diego, 9500 Gilman Drive, La Jolla,
California 92039, United States
| | - Sanjay Mehta
- Department of NanoEngineering,
Department of Medicine,
Department of Radiology,
Department of Bioengineering,
Center for Nano-ImmunoEngineering,
Moores Cancer Center, Institute for
Materials Discovery and Design, and Veterans
Administration San Diego Healthcare System, University of
California San Diego, 9500 Gilman Drive, La Jolla,
California 92039, United States
| | - Isabel G. Newton
- Department of NanoEngineering,
Department of Medicine,
Department of Radiology,
Department of Bioengineering,
Center for Nano-ImmunoEngineering,
Moores Cancer Center, Institute for
Materials Discovery and Design, and Veterans
Administration San Diego Healthcare System, University of
California San Diego, 9500 Gilman Drive, La Jolla,
California 92039, United States
| | - Nicole F. Steinmetz
- Department of NanoEngineering,
Department of Medicine,
Department of Radiology,
Department of Bioengineering,
Center for Nano-ImmunoEngineering,
Moores Cancer Center, Institute for
Materials Discovery and Design, and Veterans
Administration San Diego Healthcare System, University of
California San Diego, 9500 Gilman Drive, La Jolla,
California 92039, United States
| |
Collapse
|
33
|
Modular vaccine platform based on the norovirus-like particle. J Nanobiotechnology 2021; 19:25. [PMID: 33468139 PMCID: PMC7815183 DOI: 10.1186/s12951-021-00772-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 01/08/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Virus-like particle (VLP) vaccines have recently emerged as a safe and effective alternative to conventional vaccine technologies. The strong immunogenic effects of VLPs can be harnessed for making vaccines against any pathogen by decorating VLPs with antigens from the pathogen. Producing the antigenic pathogen fragments and the VLP platform separately makes vaccine development rapid and convenient. Here we decorated the norovirus-like particle with two conserved influenza antigens and tested for the immunogenicity of the vaccine candidates in BALB/c mice. RESULTS SpyTagged noro-VLP was expressed with high efficiency in insect cells and purified using industrially scalable methods. Like the native noro-VLP, SpyTagged noro-VLP is stable for months when refrigerated in a physiological buffer. The conserved influenza antigens were produced separately as SpyCatcher fusions in E. coli before covalent conjugation on the surface of noro-VLP. The noro-VLP had a high adjuvant effect, inducing high titers of antibody production against the antigens presented on its surface. CONCLUSIONS The modular noro-VLP vaccine platform presented here offers a rapid, convenient and safe method to present various soluble protein antigens to the immune system for vaccination and antibody production purposes.
Collapse
|
34
|
Ren F, Swevers L, Lu Q, Zhao Y, Yan J, Li H, Sun J. Effect of mutations in capsid shell protein on the assembly of BmCPV virus-like particles. J Gen Virol 2020; 102. [PMID: 33331809 DOI: 10.1099/jgv.0.001542] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Bombyx mori cytoplasmic polyhedrosis virus (BmCPV) is a typical single-layer capsid dsRNA virus belonging to the genus Cypovirus in the family Reoviridae. The results of cryo-electron microscopy showed that the BmCPV capsid consists of 60 asymmetric units, and each asymmetric unit contains one turret protein (TP), two large protrusion proteins (LPP) and two capsid shell proteins (CSP). CSP has the ability to self-assemble into virus-like particles (VLPs), and the small protrusion domain (SPD) in CSP may play an essential role in the assembly of viral capsids. In this study, three critical amino acid sites, D828, S829 and V945, in the SPD were efficiently mutated (point mutation) based on the principle of PCR circular mutagenesis. Moreover, a multi-gene expression system, Ac-MultiBac baculovirus, was used to produce eight different recombinant VLPs in vitro. Transmission electron microscopy showed that the single site and double site mutations had little effect on the efficiency and morphology of the assembly of VLPs. Still, the simultaneous mutation of the three sites had a significant impact. The experimental results demonstrate that the SPD of CSP plays an essential role in assembly of the viral capsid, which lays the foundation for further analysis of the molecular and structural mechanism of BmCPV capsid assembly.
Collapse
Affiliation(s)
- Feifei Ren
- Insect Molecular Genetics and Biotechnology, Institute of Biosciences and Applications, National Centre for Scientific Research Demokritos, Aghia Paraskevi, Athens, Greece.,Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, PR China
| | - Luc Swevers
- Insect Molecular Genetics and Biotechnology, Institute of Biosciences and Applications, National Centre for Scientific Research Demokritos, Aghia Paraskevi, Athens, Greece
| | - Qiuyuan Lu
- Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, PR China
| | - Yongchao Zhao
- Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, PR China
| | - Jiming Yan
- Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, PR China
| | - Haiyun Li
- Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, PR China
| | - Jingchen Sun
- Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou 510642, PR China
| |
Collapse
|
35
|
Aljabali AA, Obeid MA. Inorganic-organic Nanomaterials for Therapeutics and Molecular Imaging Applications. ACTA ACUST UNITED AC 2020. [DOI: 10.2174/2210681209666190807145229] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Background::
Surface modification of nanoparticles with targeting moieties can be
achieved through bioconjugation chemistries to impart new Functionalities. Various polymeric
nanoparticles have been used for the formulation of nanoparticles such as naturally-occurring
protein cages, virus-like particles, polymeric saccharides, and liposomes. These polymers have
been proven to be biocompatible, side effects free and degradable with no toxicity.
Objectives::
This paper reviews available literature on the nanoparticles pharmaceutical and medical
applications. The review highlights and updates the customized solutions for selective drug
delivery systems that allow high-affinity binding between nanoparticles and the target receptors.
Methods::
Bibliographic databases and web-search engines were used to retrieve studies that assessed
the usability of nanoparticles in the pharmaceutical and medical fields. Data were extracted
on each system in vivo and in vitro applications, its advantages and disadvantages, and its ability to
be chemically and genetically modified to impart new functionalities. Finally, a comparison
between naturally occurring and their synthetic counterparts was carried out.
Results::
The results showed that nanoparticles-based systems could have promising applications in
diagnostics, cell labeling, contrast agents (Magnetic Resonance Imaging and Computed Tomography),
antimicrobial agents, and as drug delivery systems. However, precautions should be taken
to avoid or minimize toxic effect or incompatibility of nanoparticles-based systems with the biological
systems in case of pharmaceutical or medical applications.
Conclusion::
This review presented a summary of recent developments in the field of pharmaceutical
nanotechnology and highlighted the challenges and the merits that some of the nanoparticles-
based systems both in vivo and in vitro systems.
Collapse
Affiliation(s)
- Alaa A.A. Aljabali
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Yarmouk University, P.O. BOX 566, Irbid 21163, Jordan
| | - Mohammad A. Obeid
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Yarmouk University, P.O. BOX 566, Irbid 21163, Jordan
| |
Collapse
|
36
|
Gan BK, Rullah K, Yong CY, Ho KL, Omar AR, Alitheen NB, Tan WS. Targeted delivery of 5-fluorouracil-1-acetic acid (5-FA) to cancer cells overexpressing epithelial growth factor receptor (EGFR) using virus-like nanoparticles. Sci Rep 2020; 10:16867. [PMID: 33033330 PMCID: PMC7545207 DOI: 10.1038/s41598-020-73967-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 09/23/2020] [Indexed: 11/08/2022] Open
Abstract
Chemotherapy is widely used in cancer treatments. However, non-specific distribution of chemotherapeutic agents to healthy tissues and normal cells in the human body always leads to adverse side effects and disappointing therapeutic outcomes. Therefore, the main aim of this study was to develop a targeted drug delivery system based on the hepatitis B virus-like nanoparticle (VLNP) for specific delivery of 5-fluorouracil-1-acetic acid (5-FA) to cancer cells expressing epithelial growth factor receptor (EGFR). 5-FA was synthesized from 5-fluorouracil (5-FU), and it was found to be less toxic than the latter in cancer cells expressing different levels of EGFR. The cytotoxicity of 5-FA increased significantly after being conjugated on the VLNP. A cell penetrating peptide (CPP) of EGFR was displayed on the VLNP via the nanoglue concept, for targeted delivery of 5-FA to A431, HT29 and HeLa cells. The results showed that the VLNP displaying the CPP and harboring 5-FA internalized the cancer cells and killed them in an EGFR-dependent manner. This study demonstrated that the VLNP can be used to deliver chemically modified 5-FU derivatives to cancer cells overexpressing EGFR, expanding the applications of the VLNP in targeted delivery of chemotherapeutic agents to cancer cells overexpressing this transmembrane receptor.
Collapse
Affiliation(s)
- Bee Koon Gan
- Institute of Bioscience, Universiti Putra Malaysia, 43400, UPM Serdang, Selangor, Malaysia
| | - Kamal Rullah
- Department of Pharmaceutical Chemistry, Kulliyyah of Pharmacy (KOP), International Islamic University Malaysia (IIUM), 25200, Kuantan, Pahang, Malaysia
| | - Chean Yeah Yong
- Institute of Bioscience, Universiti Putra Malaysia, 43400, UPM Serdang, Selangor, Malaysia
| | - Kok Lian Ho
- Department of Pathology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400, UPM Serdang, Selangor, Malaysia
| | - Abdul Rahman Omar
- Institute of Bioscience, Universiti Putra Malaysia, 43400, UPM Serdang, Selangor, Malaysia
- Department of Veterinary Pathology and Microbiology, Faculty of Veterinary Medicine, Universiti Putra Malaysia, 43400, UPM Serdang, Selangor, Malaysia
| | - Noorjahan Banu Alitheen
- Institute of Bioscience, Universiti Putra Malaysia, 43400, UPM Serdang, Selangor, Malaysia
- Department of Cell and Molecular Biology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400, UPM Serdang, Selangor, Malaysia
| | - Wen Siang Tan
- Institute of Bioscience, Universiti Putra Malaysia, 43400, UPM Serdang, Selangor, Malaysia.
- Department of Microbiology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400, UPM Serdang, Selangor, Malaysia.
| |
Collapse
|
37
|
Zhou K, Zhou Y, Yang H, Jin H, Ke Y, Wang Q. Interfacially Bridging Covalent Network Yields Hyperstable and Ultralong Virus-Based Fibers for Engineering Functional Materials. Angew Chem Int Ed Engl 2020; 59:18249-18255. [PMID: 32643299 DOI: 10.1002/anie.202008670] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Indexed: 11/07/2022]
Abstract
We present a strategy of interfacially bridging covalent network within tobacco mosaic virus (TMV) virus-like particles (VLPs). We arranged T103C cysteine to laterally conjugate adjacent subunits. In the axis direction, we set A74C mutation and systematically investigated candidate from E50C to P54C as the other thiol function site, for forming longitudinal disulfide bond chains. Significantly, the T103C-TMV-E50C-A74C shows the highest robustness in assembly capability and structural stability with the largest length, for TMV VLP to date. The fibers with lengths from several to a dozen of micrometers even survive under pH 13. The robust nature of this TMV VLP allows for reducer-free synthesis of excellent electrocatalysts for application in harshly alkaline hydrogen evolution.
Collapse
Affiliation(s)
- Kun Zhou
- Institute of New Materials and Industrial Technologies, Wenzhou University, Wenzhou, 325035, China.,CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine and i-Lab, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
| | - Yihao Zhou
- CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine and i-Lab, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China.,School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei, 230026, China
| | - Hongchao Yang
- CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine and i-Lab, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
| | - Huile Jin
- Institute of New Materials and Industrial Technologies, Wenzhou University, Wenzhou, 325035, China
| | - Yonggang Ke
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30322, USA
| | - Qiangbin Wang
- CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine and i-Lab, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China.,School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei, 230026, China
| |
Collapse
|
38
|
Kiss B, Mudra D, Török G, Mártonfalvi Z, Csík G, Herényi L, Kellermayer M. Single-particle virology. Biophys Rev 2020; 12:1141-1154. [PMID: 32880826 PMCID: PMC7471434 DOI: 10.1007/s12551-020-00747-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 08/18/2020] [Indexed: 01/02/2023] Open
Abstract
The development of advanced experimental methodologies, such as optical tweezers, scanning-probe and super-resolved optical microscopies, has led to the evolution of single-molecule biophysics, a field of science that allows direct access to the mechanistic detail of biomolecular structure and function. The extension of single-molecule methods to the investigation of particles such as viruses permits unprecedented insights into the behavior of supramolecular assemblies. Here we address the scope of viral exploration at the level of individual particles. In an era of increased awareness towards virology, single-particle approaches are expected to facilitate the in-depth understanding, and hence combating, of viral diseases.
Collapse
Affiliation(s)
- Bálint Kiss
- Department of Biophysics and Radiation Biology, Semmelweis University, Budapest, Hungary
| | - Dorottya Mudra
- Department of Biophysics and Radiation Biology, Semmelweis University, Budapest, Hungary
| | - György Török
- Department of Biophysics and Radiation Biology, Semmelweis University, Budapest, Hungary
| | - Zsolt Mártonfalvi
- Department of Biophysics and Radiation Biology, Semmelweis University, Budapest, Hungary
| | - Gabriella Csík
- Department of Biophysics and Radiation Biology, Semmelweis University, Budapest, Hungary
| | - Levente Herényi
- Department of Biophysics and Radiation Biology, Semmelweis University, Budapest, Hungary
| | - Miklós Kellermayer
- Department of Biophysics and Radiation Biology, Semmelweis University, Budapest, Hungary.
| |
Collapse
|
39
|
Zhou K, Zhou Y, Yang H, Jin H, Ke Y, Wang Q. Interfacially Bridging Covalent Network Yields Hyperstable and Ultralong Virus‐Based Fibers for Engineering Functional Materials. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.202008670] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Kun Zhou
- Institute of New Materials and Industrial Technologies Wenzhou University Wenzhou 325035 China
- CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine and i-Lab Suzhou Institute of Nano-Tech and Nano-Bionics Chinese Academy of Sciences Suzhou 215123 China
| | - Yihao Zhou
- CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine and i-Lab Suzhou Institute of Nano-Tech and Nano-Bionics Chinese Academy of Sciences Suzhou 215123 China
- School of Nano-Tech and Nano-Bionics University of Science and Technology of China Hefei 230026 China
| | - Hongchao Yang
- CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine and i-Lab Suzhou Institute of Nano-Tech and Nano-Bionics Chinese Academy of Sciences Suzhou 215123 China
| | - Huile Jin
- Institute of New Materials and Industrial Technologies Wenzhou University Wenzhou 325035 China
| | - Yonggang Ke
- Wallace H. Coulter Department of Biomedical Engineering Georgia Institute of Technology and Emory University Atlanta GA 30322 USA
| | - Qiangbin Wang
- CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine and i-Lab Suzhou Institute of Nano-Tech and Nano-Bionics Chinese Academy of Sciences Suzhou 215123 China
- School of Nano-Tech and Nano-Bionics University of Science and Technology of China Hefei 230026 China
| |
Collapse
|
40
|
Zackova Suchanova J, Hejtmankova A, Neburkova J, Cigler P, Forstova J, Spanielova H. The Protein Corona Does Not Influence Receptor-Mediated Targeting of Virus-like Particles. Bioconjug Chem 2020; 31:1575-1585. [DOI: 10.1021/acs.bioconjchem.0c00240] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Jirina Zackova Suchanova
- Department of Genetics and Microbiology, Faculty of Science, Charles University, Viničná 5, 128 44 Prague 2, Czech Republic
| | - Alzbeta Hejtmankova
- Department of Genetics and Microbiology, Faculty of Science, Charles University, Viničná 5, 128 44 Prague 2, Czech Republic
| | - Jitka Neburkova
- Institute of Organic Chemistry and Biochemistry of the CAS, Flemingovo nám. 2, 166 10 Prague 6, Czech Republic
| | - Petr Cigler
- Institute of Organic Chemistry and Biochemistry of the CAS, Flemingovo nám. 2, 166 10 Prague 6, Czech Republic
| | - Jitka Forstova
- Department of Genetics and Microbiology, Faculty of Science, Charles University, Viničná 5, 128 44 Prague 2, Czech Republic
| | - Hana Spanielova
- Department of Genetics and Microbiology, Faculty of Science, Charles University, Viničná 5, 128 44 Prague 2, Czech Republic
- Institute of Organic Chemistry and Biochemistry of the CAS, Flemingovo nám. 2, 166 10 Prague 6, Czech Republic
| |
Collapse
|
41
|
Algorithm-Based Liquid Formulation Development Including a DoE Concept Predicts Long-Term Viral Vector Stability. J Pharm Sci 2020; 109:818-829. [DOI: 10.1016/j.xphs.2019.10.063] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 10/29/2019] [Accepted: 10/30/2019] [Indexed: 11/20/2022]
|
42
|
Horsington J, Eschbaumer M, Singanallur NB, Vosloo W. Inactivation of foot-and-mouth disease virus in epithelium samples for safe transport and processing in low-containment laboratories. J Virol Methods 2019; 276:113770. [PMID: 31705919 DOI: 10.1016/j.jviromet.2019.113770] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 11/04/2019] [Accepted: 11/05/2019] [Indexed: 10/25/2022]
Abstract
During a foot-and-mouth disease (FMD) outbreak, transport and testing of potentially infectious samples, including epithelium from suspect lesions, presents a biosafety risk, particularly in FMD-free countries. Therefore, treatment to inactivate virus prior to transport is important. Tongue epithelium from cattle infected with FMD virus (FMDV) serotype O (O ALG/3/2014 - Lineage O/ME-SA/Ind-2001d) or A (A IRN/22/2015 - Lineage A/ASIA/G-VII) was incubated in RNAlater, RNA Shield or phosphate-buffered saline (pH 7.4) at room temperature for 2, 6, 24 or 48 h. After incubation, tissues were homogenised and tested by virus titration. Viral RNA in the homogenate was quantified by RT-qPCR, used for sequencing, and transfected into LFBKαVβ6 cells to recover infectious virus. RNAlater reduced A IRN/22/2015 titres by 4 log10 after 24 h, and completely after 48 h incubation. While O ALG/3/2014 was detected by VI after 2, 6 and 24 h, titration yielded no infectious virus, likely as a result of freeze-thawing. RNA Shield was cytotoxic at high concentrations but was effective at inactivating both strains after 24 h. Regardless of reagent or inactivation period, RT-qPCR, VP1 sequencing, and transfection of RNA to recover infectious virus were possible. RNA Shield appears a better choice for FMDV inactivation in tissues, however 24 h incubation is recommended.
Collapse
Affiliation(s)
- Jacquelyn Horsington
- Australian Animal Health Laboratory, CSIRO Health and Biosecurity, 5 Portarlington Rd, Geelong, VIC, Australia
| | - Michael Eschbaumer
- Institute of Diagnostic Virology, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Suedufer 10, 17493, Greifswald, Germany
| | | | - Wilna Vosloo
- Australian Animal Health Laboratory, CSIRO Health and Biosecurity, 5 Portarlington Rd, Geelong, VIC, Australia.
| |
Collapse
|
43
|
Virus capsid assembly across different length scales inspire the development of virus-based biomaterials. Curr Opin Virol 2019; 36:38-46. [PMID: 31071601 DOI: 10.1016/j.coviro.2019.02.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Revised: 02/12/2019] [Accepted: 02/25/2019] [Indexed: 01/26/2023]
Abstract
In biology, there are an abundant number of self-assembled structures organized according to hierarchical levels of complexity. In some examples, the assemblies formed at each level exhibit unique properties and behaviors not present in individual components. Viruses are an example of such where first individual subunits come together to form a capsid structure, some utilizing a scaffolding protein to template or catalyze the capsid formation. Increasing the level of complexity, the viral capsids can then be used as building blocks of higher-level assemblies. This has inspired scientists to design and construct virus capsid-based functional nano-materials. This review provides some insight into the assembly of virus capsids across several length scales, and certain properties that arise at different levels, providing examples found in naturally occurring systems and those that are synthetically designed.
Collapse
|
44
|
Chen MY, Butler SS, Chen W, Suh J. Physical, chemical, and synthetic virology: Reprogramming viruses as controllable nanodevices. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2019; 11:e1545. [PMID: 30411529 PMCID: PMC6461522 DOI: 10.1002/wnan.1545] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 08/03/2018] [Accepted: 10/04/2018] [Indexed: 01/24/2023]
Abstract
The fields of physical, chemical, and synthetic virology work in partnership to reprogram viruses as controllable nanodevices. Physical virology provides the fundamental biophysical understanding of how virus capsids assemble, disassemble, display metastability, and assume various configurations. Chemical virology considers the virus capsid as a chemically addressable structure, providing chemical pathways to modify the capsid exterior, interior, and subunit interfaces. Synthetic virology takes an engineering approach, modifying the virus capsid through rational, combinatorial, and bioinformatics-driven design strategies. Advances in these three subfields of virology aim to develop virus-based materials and tools that can be applied to solve critical problems in biomedicine and biotechnology, including applications in gene therapy and drug delivery, diagnostics, and immunotherapy. Examples discussed include mammalian viruses, such as adeno-associated virus (AAV), plant viruses, such as cowpea mosaic virus (CPMV), and bacterial viruses, such as Qβ bacteriophage. Importantly, research efforts in physical, chemical, and synthetic virology have further unraveled the design principles foundational to the form and function of viruses. This article is categorized under: Diagnostic Tools > Diagnostic Nanodevices Biology-Inspired Nanomaterials > Protein and Virus-Based Structures.
Collapse
Affiliation(s)
| | - Susan S Butler
- Department of Bioengineering, Rice University, Houston, Texas
| | - Weitong Chen
- Department of Chemical and Biomolecular Engineering, Rice University, Houston, Texas
| | - Junghae Suh
- Department of Bioengineering, Rice University, Houston, Texas
- Systems, Synthetic, and Physical Biology Program, Rice University, Houston, Texas
| |
Collapse
|
45
|
Charlton Hume HK, Vidigal J, Carrondo MJT, Middelberg APJ, Roldão A, Lua LHL. Synthetic biology for bioengineering virus-like particle vaccines. Biotechnol Bioeng 2019; 116:919-935. [PMID: 30597533 PMCID: PMC7161758 DOI: 10.1002/bit.26890] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 11/08/2018] [Accepted: 11/29/2018] [Indexed: 12/13/2022]
Abstract
Vaccination is the most effective method of disease prevention and control. Many viruses and bacteria that once caused catastrophic pandemics (e.g., smallpox, poliomyelitis, measles, and diphtheria) are either eradicated or effectively controlled through routine vaccination programs. Nonetheless, vaccine manufacturing remains incredibly challenging. Viruses exhibiting high antigenic diversity and high mutation rates cannot be fairly contested using traditional vaccine production methods and complexities surrounding the manufacturing processes, which impose significant limitations. Virus-like particles (VLPs) are recombinantly produced viral structures that exhibit immunoprotective traits of native viruses but are noninfectious. Several VLPs that compositionally match a given natural virus have been developed and licensed as vaccines. Expansively, a plethora of studies now confirms that VLPs can be designed to safely present heterologous antigens from a variety of pathogens unrelated to the chosen carrier VLPs. Owing to this design versatility, VLPs offer technological opportunities to modernize vaccine supply and disease response through rational bioengineering. These opportunities are greatly enhanced with the application of synthetic biology, the redesign and construction of novel biological entities. This review outlines how synthetic biology is currently applied to engineer VLP functions and manufacturing process. Current and developing technologies for the identification of novel target-specific antigens and their usefulness for rational engineering of VLP functions (e.g., presentation of structurally diverse antigens, enhanced antigen immunogenicity, and improved vaccine stability) are described. When applied to manufacturing processes, synthetic biology approaches can also overcome specific challenges in VLP vaccine production. Finally, we address several challenges and benefits associated with the translation of VLP vaccine development into the industry.
Collapse
Affiliation(s)
- Hayley K. Charlton Hume
- The University of Queensland, Australian Institute of Bioengineering and NanotechnologySt LuciaQueenslandAustralia
| | - João Vidigal
- Health & Pharma Division, Animal Cell Technology Unit, Instituto de Biologia Experimental e Tecnológica (iBET)OeirasPortugal
- Health & Pharma Division, Animal Cell Technology Unit, Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da RepúblicaOeirasPortugal
| | - Manuel J. T. Carrondo
- Health & Pharma Division, Animal Cell Technology Unit, Instituto de Biologia Experimental e Tecnológica (iBET)OeirasPortugal
| | - Anton P. J. Middelberg
- Faculty of Engineering, Computer and Mathematical Sciences, The University of AdelaideAdelaideSouth AustraliaAustralia
| | - António Roldão
- Health & Pharma Division, Animal Cell Technology Unit, Instituto de Biologia Experimental e Tecnológica (iBET)OeirasPortugal
- Health & Pharma Division, Animal Cell Technology Unit, Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da RepúblicaOeirasPortugal
| | | |
Collapse
|
46
|
Vishnu Vardhan GP, Hema M, Sushmitha C, Savithri HS, Natraj U, Murthy MRN. Development of sesbania mosaic virus nanoparticles for imaging. Arch Virol 2018; 164:497-507. [PMID: 30430265 DOI: 10.1007/s00705-018-4097-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Accepted: 10/22/2018] [Indexed: 11/30/2022]
Abstract
The capsids of viruses have a high degree of symmetry. Therefore, virus nanoparticles (VNPs) can be programmed to display many imaging agents precisely. Plant VNPs are biocompatible, biodegradable and non-infectious to mammals. We have carried out bioconjugation of sesbania mosaic virus (SeMV), a well characterized plant virus, with fluorophores using reactive lysine-N-hydroxysuccinimide ester and cysteine-maleimide chemistries. Monitoring of cellular internalization of labelled SeMV nanoparticles (NPs) by confocal microscopy and flow cytometry showed that the particles have a natural preference for entry into MDA-MB-231 (breast cancer) cells, although they could also enter various other cell lines. The fluorescence of SeMV NPs labelled via the cysteines with Cy5.5 dye was found to be more stable and was detectable with greater sensitivity than that of particles labelled via the lysines with Alexa Fluor. Live-cell imaging using SeMV internally labelled with Cy5.5 showed that it could bind to MDA-MB-231 cells in less than 5 minutes and enter the cells within 15 minutes. The particles undergo endolysosomal degradation by 6 h as evidenced by their co-localization with LAMP-1. Far-western blot analysis with a HeLa cell membrane protein fraction showed that SeMV interacts with 54-, 35- and 33-kDa proteins, which were identified by mass spectrometry as vimentin, voltage-dependent anion-selective channel protein (VDAC1), and annexin A2 isoform 2 (ANXA2), respectively, suggesting that the particles may bind and enter the cell through these proteins. The results presented here demonstrate that the SeMV NPs provide a new platform technology that could be used to develop in vivo imaging and targeted drug delivery agents for cancer diagnosis and therapy.
Collapse
Affiliation(s)
- G P Vishnu Vardhan
- Department of Biochemistry, Indian Institute of Science, Bengaluru, India
| | - M Hema
- Department of Virology, Sri Venkateswara University, Tirupati, India.
| | - C Sushmitha
- Department of Biochemistry, Indian Institute of Science, Bengaluru, India
| | - H S Savithri
- Department of Biochemistry, Indian Institute of Science, Bengaluru, India.
| | - Usha Natraj
- Department of Biochemistry, Indian Institute of Science, Bengaluru, India
| | - M R N Murthy
- Molecular Biophysics Unit, Indian Institute of Science, Bengaluru, India
| |
Collapse
|
47
|
Wang X, Xiao Y, Hao H, Zhang Y, Xu X, Tang R. Therapeutic Potential of Biomineralization‐Based Engineering. ADVANCED THERAPEUTICS 2018. [DOI: 10.1002/adtp.201800079] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Xiaoyu Wang
- Qiushi Academy for Advanced StudiesZhejiang University No. 38 Zheda Road Hangzhou Zhejiang 310027 China
| | - Yun Xiao
- Center for Biomaterials and Biopathways, Department of ChemistryZhejiang University No. 38 Zheda Road Hangzhou Zhejiang 310027 China
| | - Haibin Hao
- Center for Biomaterials and Biopathways, Department of ChemistryZhejiang University No. 38 Zheda Road Hangzhou Zhejiang 310027 China
| | - Ying Zhang
- Center for Biomaterials and Biopathways, Department of ChemistryZhejiang University No. 38 Zheda Road Hangzhou Zhejiang 310027 China
| | - Xurong Xu
- Qiushi Academy for Advanced StudiesZhejiang University No. 38 Zheda Road Hangzhou Zhejiang 310027 China
| | - Ruikang Tang
- Qiushi Academy for Advanced StudiesZhejiang University No. 38 Zheda Road Hangzhou Zhejiang 310027 China
- Center for Biomaterials and Biopathways, Department of ChemistryZhejiang University No. 38 Zheda Road Hangzhou Zhejiang 310027 China
| |
Collapse
|
48
|
Shan W, Zhang D, Wu Y, Lv X, Hu B, Zhou X, Ye S, Bi S, Ren L, Zhang X. Modularized peptides modified HBc virus-like particles for encapsulation and tumor-targeted delivery of doxorubicin. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2018; 14:725-734. [DOI: 10.1016/j.nano.2017.12.002] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2017] [Revised: 11/12/2017] [Accepted: 12/03/2017] [Indexed: 01/08/2023]
|
49
|
Riekel C, Burghammer M, Snigirev I, Rosenthal M. Microstructural metrology of tobacco mosaic virus nanorods during radial compression and heating. SOFT MATTER 2018; 14:194-204. [PMID: 29138785 DOI: 10.1039/c7sm01332a] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
We determined stress-induced deformations and the thermal stability of nanorod-shaped tobacco mosaic virus (TMV) capsids in coffee-ring structures by X-ray nanodiffraction. The hexagonal capsids lattice transforms under compression in the outer boundary zone of the coffee-ring into a tetragonal lattice. The helical pitch of the nanorods increases by about 2.5% across the outer boundary zone while the lateral distance between nanorods decreases continuously across the whole coffee-ring structure by about 2% due to compressive forces. The diffraction patterns show a mixture of helical scattering and Bragg peaks attributed to a lattice of nanorods interlocked by their helical grooves. Thermo-nanodiffraction reveals water loss up to about 100 °C resulting in a reduction of the helical pitch by about 6% with respect to its maximum value and a reduction of the nanorods separation by about 0.5 nm. Up to about 200 °C the pitch is increasing again by about 2%. Secondary crystallization in the bulk reaches a maximum at 150-160 °C. At higher temperatures the crystallinity is continuously decreasing up to about 220 °C. Above about 200 °C and depending on the heating history, the nanorods start disintegrating into small, randomly oriented aggregates.
Collapse
Affiliation(s)
- C Riekel
- The European Synchrotron, ESRF, CS40220, F-38043 Grenoble Cedex 9, France.
| | | | | | | |
Collapse
|
50
|
Carrillo PJP, Medrano M, Valbuena A, Rodríguez-Huete A, Castellanos M, Pérez R, Mateu MG. Amino Acid Side Chains Buried along Intersubunit Interfaces in a Viral Capsid Preserve Low Mechanical Stiffness Associated with Virus Infectivity. ACS NANO 2017; 11:2194-2208. [PMID: 28117975 DOI: 10.1021/acsnano.6b08549] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Single-molecule experimental techniques and theoretical approaches reveal that important aspects of virus biology can be understood in biomechanical terms at the nanoscale. A detailed knowledge of the relationship in virus capsids between small structural changes caused by single-point mutations and changes in mechanical properties may provide further physics-based insights into virus function; it may also facilitate the engineering of viral nanoparticles with improved mechanical behavior. Here, we used the minute virus of mice to undertake a systematic experimental study on the contribution to capsid stiffness of amino acid side chains at interprotein interfaces and the specific noncovalent interactions they establish. Selected side chains were individually truncated by introducing point mutations to alanine, and the effects on local and global capsid stiffness were determined using atomic force microscopy. The results revealed that, in the natural virus capsid, multiple, mostly hydrophobic, side chains buried along the interfaces between subunits preserve a comparatively low stiffness of most (S2 and S3) regions. Virtually no point mutation tested substantially reduced stiffness, whereas most mutations increased stiffness of the S2/S3 regions. This stiffening was invariably associated with reduced virus yields during cell infection. The experimental evidence suggests that a comparatively low stiffness at S3/S2 capsid regions may have been biologically selected because it facilitates capsid assembly, increasing infectious virus yields. This study demonstrated also that knowledge of individual amino acid side chains and biological pressures that determine the physical behavior of a protein nanoparticle may be used for engineering its mechanical properties.
Collapse
Affiliation(s)
- Pablo José P Carrillo
- Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), Universidad Autónoma de Madrid , c/Nicolás Cabrera 1, Cantoblanco, 28049 Madrid, Spain
| | - María Medrano
- Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), Universidad Autónoma de Madrid , c/Nicolás Cabrera 1, Cantoblanco, 28049 Madrid, Spain
| | - Alejandro Valbuena
- Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), Universidad Autónoma de Madrid , c/Nicolás Cabrera 1, Cantoblanco, 28049 Madrid, Spain
| | - Alicia Rodríguez-Huete
- Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), Universidad Autónoma de Madrid , c/Nicolás Cabrera 1, Cantoblanco, 28049 Madrid, Spain
| | - Milagros Castellanos
- Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), Universidad Autónoma de Madrid , c/Nicolás Cabrera 1, Cantoblanco, 28049 Madrid, Spain
| | - Rebeca Pérez
- Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), Universidad Autónoma de Madrid , c/Nicolás Cabrera 1, Cantoblanco, 28049 Madrid, Spain
| | - Mauricio G Mateu
- Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), Universidad Autónoma de Madrid , c/Nicolás Cabrera 1, Cantoblanco, 28049 Madrid, Spain
| |
Collapse
|