1
|
Ahmadivand S, Fux R, Palić D. Role of T Follicular Helper Cells in Viral Infections and Vaccine Design. Cells 2025; 14:508. [PMID: 40214462 PMCID: PMC11987902 DOI: 10.3390/cells14070508] [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: 01/25/2025] [Revised: 03/20/2025] [Accepted: 03/26/2025] [Indexed: 04/14/2025] Open
Abstract
T follicular helper (Tfh) cells are a specialized subset of CD4+ T lymphocytes that are essential for the development of long-lasting humoral immunity. Tfh cells facilitate B lymphocyte maturation, promote germinal center formation, and drive high-affinity antibody production. Our current knowledge of Tfh interactions with the humoral immune system effectors suggests that they have a critical role in supporting the immune response against viral infections. This review discusses the mechanisms through which Tfh cells influence anti-viral immunity, highlighting their interactions with B cells and their impact on antibody quality and quantity. We explore the role of Tfh cells in viral infections and examine how vaccine design can be improved to enhance Tfh cell responses. Innovative vaccine platforms, such as mRNA vaccines and self-assembling protein nanoplatforms (SAPNs), are promising strategies to enhance Tfh cell activation. Their integration and synergistic combination could further enhance immunity and Tfh responses (SAPN-RNA vaccines). In summary, we provide a comprehensive overview of the current insights into Tfh cells' role during viral infections, emphasizing their potential as strategic targets for innovative vaccine development.
Collapse
Affiliation(s)
- Sohrab Ahmadivand
- Faculty of Veterinary Medicine, Ludwig-Maximilians-University Munich, 80539 Munich, Germany
| | - Robert Fux
- Institute for Infectious Diseases and Zoonoses, Ludwig-Maximilians-University Munich, 80539 Munich, Germany;
| | - Dušan Palić
- Faculty of Veterinary Medicine, Ludwig-Maximilians-University Munich, 80539 Munich, Germany
| |
Collapse
|
2
|
Maltseva M, Keeshan A, Cooper C, Langlois MA. Immune imprinting: The persisting influence of the first antigenic encounter with rapidly evolving viruses. Hum Vaccin Immunother 2024; 20:2384192. [PMID: 39149872 PMCID: PMC11328881 DOI: 10.1080/21645515.2024.2384192] [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: 05/02/2024] [Revised: 07/08/2024] [Accepted: 07/21/2024] [Indexed: 08/17/2024] Open
Abstract
Immune imprinting is a phenomenon that stems from the fundamentals of immunological memory. Upon recurrent exposures to an evolving pathogen, the immune system must weigh the benefits of rapidly recalling established antibody repertoires with greater affinity to the initial variant or invest additional time and energy in producing de novo responses specific to the emerging variant. In this review, we delve into the mechanistic complexities of immune imprinting and its role in shaping subsequent immune responses, both de novo and recall, against rapidly evolving respiratory viruses such as influenza and coronaviruses. By exploring the duality of immune imprinting, we examine its potential to both enhance or hinder immune protection against disease, while emphasizing the role of host and viral factors. Finally, we explore how different vaccine platforms may affect immune imprinting and comment on vaccine strategies that can favor de novo variant-specific antibody responses.
Collapse
Affiliation(s)
- Mariam Maltseva
- Department of Biochemistry, Microbiology & Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Canada
| | - Alexa Keeshan
- School of Epidemiology and Public Health, University of Ottawa Faculty of Medicine, Ottawa, ON, Canada
- Clinical Epidemiology Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Curtis Cooper
- School of Epidemiology and Public Health, University of Ottawa Faculty of Medicine, Ottawa, ON, Canada
- Clinical Epidemiology Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Department of Medicine, University of Ottawa, Ottawa, ON, Canada
- Division of Infectious Diseases, Ottawa Hospital Research Institute Clinical Epidemiology Program, Ottawa, ON, Canada
| | - Marc-André Langlois
- Department of Biochemistry, Microbiology & Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Canada
- Center for Infection, Immunity, and Inflammation (CI3), University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
3
|
Cao N, Li Y, Zhang H, Liu X, Liu S, Lu M, Hu Z, Tian L, Li X, Qian P. A nanoparticle vaccine based on the VP1 21-26 and VP2 structural proteins of Senecavirus A induces robust protective immune responses. Vet Microbiol 2024; 296:110198. [PMID: 39067145 DOI: 10.1016/j.vetmic.2024.110198] [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: 05/10/2024] [Revised: 07/20/2024] [Accepted: 07/24/2024] [Indexed: 07/30/2024]
Abstract
Senecavirus A (SVA) is a causative agent that can cause vesicular disease in swine, which causes a great threat to the swine husbandry in the world. Therefore, it is necessary to develop a vaccine that can effectively prevent the spread of SVA. In this study, we developed a 24-polymeric nano-scaffold using β-annulus peptide from tomato bushy effect virus (TBSV) by coupling this antigen to SVA B cell epitope VP121-26 and VP2 proteins via linkers, respectively. The SVA-based nanoparticle protein of the VP1(B)-β-VP2 was expressed and purified by low-cost prokaryotic system to prepare a SVA nanoparticle vaccine. The immunological protective effect of SVA nanoparticle vaccine was evaluated in mouse and swine models, respectively. The results suggested that both mice and swine could induce high levels SVA neutralizing antibodies and IgG antibodies after two doses immunization. In addition, the swine challenge protection experiment showed that the protection rate of immune SVA nanoparticle vaccine and SVA inactivated vaccine both were 80 %, while the negative control had no protection effect. It demonstrated that SVA nanoparticle vaccine effectively prevented SVA infection in swine. In summary, the preparation of SVA vaccine by using β-annulus peptide is a promising candidate vaccine for prevent SVA transmission, and provides a new idea for the development of novel SVA vaccines.
Collapse
Affiliation(s)
- Nan Cao
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, Hubei 430070, China; College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei 430070, China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei 430070, China
| | - Yamei Li
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, Hubei 430070, China; College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei 430070, China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei 430070, China
| | - Huawei Zhang
- Wuhan Keqian Biological Co., Ltd, Wuhan, Hubei, China
| | - Xiangzu Liu
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, Hubei 430070, China; College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei 430070, China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei 430070, China
| | - Shudan Liu
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, Hubei 430070, China; College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei 430070, China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei 430070, China
| | - Mingxing Lu
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, Hubei 430070, China; College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei 430070, China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei 430070, China
| | - Zihui Hu
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, Hubei 430070, China; College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei 430070, China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei 430070, China
| | - Linxing Tian
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, Hubei 430070, China; College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei 430070, China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei 430070, China
| | - Xiangmin Li
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, Hubei 430070, China; College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei 430070, China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei 430070, China.
| | - Ping Qian
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, Hubei 430070, China; College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei 430070, China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei 430070, China.
| |
Collapse
|
4
|
Kulma M, Šakanović A, Bedina-Zavec A, Caserman S, Omersa N, Šolinc G, Orehek S, Hafner-Bratkovič I, Kuhar U, Slavec B, Krapež U, Ocepek M, Kobayashi T, Kwiatkowska K, Jerala R, Podobnik M, Anderluh G. Sequestration of membrane cholesterol by cholesterol-binding proteins inhibits SARS-CoV-2 entry into Vero E6 cells. Biochem Biophys Res Commun 2024; 716:149954. [PMID: 38704887 DOI: 10.1016/j.bbrc.2024.149954] [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: 02/13/2024] [Revised: 03/26/2024] [Accepted: 04/15/2024] [Indexed: 05/07/2024]
Abstract
Membrane lipids and proteins form dynamic domains crucial for physiological and pathophysiological processes, including viral infection. Many plasma membrane proteins, residing within membrane domains enriched with cholesterol (CHOL) and sphingomyelin (SM), serve as receptors for attachment and entry of viruses into the host cell. Among these, human coronaviruses, including severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), use proteins associated with membrane domains for initial binding and internalization. We hypothesized that the interaction of lipid-binding proteins with CHOL in plasma membrane could sequestrate lipids and thus affect the efficiency of virus entry into host cells, preventing the initial steps of viral infection. We have prepared CHOL-binding proteins with high affinities for lipids in the plasma membrane of mammalian cells. Binding of the perfringolysin O domain four (D4) and its variant D4E458L to membrane CHOL impaired the internalization of the receptor-binding domain of the SARS-CoV-2 spike protein and the pseudovirus complemented with the SARS-CoV-2 spike protein. SARS-CoV-2 replication in Vero E6 cells was also decreased. Overall, our results demonstrate that the integrity of CHOL-rich membrane domains and the accessibility of CHOL in the membrane play an essential role in SARS-CoV-2 cell entry.
Collapse
Affiliation(s)
- Magdalena Kulma
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Hajdrihova 19, 1000, Ljubljana, Slovenia
| | - Aleksandra Šakanović
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Hajdrihova 19, 1000, Ljubljana, Slovenia
| | - Apolonija Bedina-Zavec
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Hajdrihova 19, 1000, Ljubljana, Slovenia
| | - Simon Caserman
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Hajdrihova 19, 1000, Ljubljana, Slovenia
| | - Neža Omersa
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Hajdrihova 19, 1000, Ljubljana, Slovenia
| | - Gašper Šolinc
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Hajdrihova 19, 1000, Ljubljana, Slovenia
| | - Sara Orehek
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Hajdrihova 19, 1000 Ljubljana, Slovenia
| | - Iva Hafner-Bratkovič
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Hajdrihova 19, 1000 Ljubljana, Slovenia; EN-FIST Centre of Excellence, Trg Osvobodilne Fronte 13, 1000, Ljubljana, Slovenia
| | - Urška Kuhar
- Veterinary Faculty, University of Ljubljana, Gerbičeva 60, 1000, Ljubljana, Slovenia
| | - Brigita Slavec
- Veterinary Faculty, University of Ljubljana, Gerbičeva 60, 1000, Ljubljana, Slovenia
| | - Uroš Krapež
- Veterinary Faculty, University of Ljubljana, Gerbičeva 60, 1000, Ljubljana, Slovenia
| | - Matjaž Ocepek
- Veterinary Faculty, University of Ljubljana, Gerbičeva 60, 1000, Ljubljana, Slovenia
| | - Toshihide Kobayashi
- Lipid Biology Laboratory, RIKEN, 2-1, Hirosawa, Wako-shi, Saitama, 351-0198, Japan; UMR 7021 CNRS, Université de Strasbourg, F-67401, Illkirch, France
| | - Katarzyna Kwiatkowska
- Laboratory of Molecular Membrane Biology, Nencki Institute of Experimental Biology of the Polish Academy of Sciences, 3 Pasteur St., 02-093, Warsaw, Poland
| | - Roman Jerala
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, Hajdrihova 19, 1000 Ljubljana, Slovenia; EN-FIST Centre of Excellence, Trg Osvobodilne Fronte 13, 1000, Ljubljana, Slovenia
| | - Marjetka Podobnik
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Hajdrihova 19, 1000, Ljubljana, Slovenia
| | - Gregor Anderluh
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Hajdrihova 19, 1000, Ljubljana, Slovenia.
| |
Collapse
|
5
|
Sobczak JM, Barkovska I, Balke I, Rothen DA, Mohsen MO, Skrastina D, Ogrina A, Martina B, Jansons J, Bogans J, Vogel M, Bachmann MF, Zeltins A. Identifying Key Drivers of Efficient B Cell Responses: On the Role of T Help, Antigen-Organization, and Toll-like Receptor Stimulation for Generating a Neutralizing Anti-Dengue Virus Response. Vaccines (Basel) 2024; 12:661. [PMID: 38932390 PMCID: PMC11209419 DOI: 10.3390/vaccines12060661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 05/14/2024] [Accepted: 05/23/2024] [Indexed: 06/28/2024] Open
Abstract
T help (Th), stimulation of toll-like receptors (pathogen-associated molecular patterns, PAMPs), and antigen organization and repetitiveness (pathogen-associated structural patterns, PASPs) were shown numerous times to be important in driving B-cell and antibody responses. In this study, we dissected the individual contributions of these parameters using newly developed "Immune-tag" technology. As model antigens, we used eGFP and the third domain of the dengue virus 1 envelope protein (DV1 EDIII), the major target of virus-neutralizing antibodies. The respective proteins were expressed alone or genetically fused to the N-terminal fragment of the cucumber mosaic virus (CMV) capsid protein-nCMV, rendering the antigens oligomeric. In a step-by-step manner, RNA was attached as a PAMP, and/or a universal Th-cell epitope was genetically added for additional Th. Finally, a PASP was added to the constructs by displaying the antigens highly organized and repetitively on the surface of CMV-derived virus-like particles (CuMV VLPs). Sera from immunized mice demonstrated that each component contributed stepwise to the immunogenicity of both proteins. All components combined in the CuMV VLP platform induced by far the highest antibody responses. In addition, the DV1 EDIII induced high levels of DENV-1-neutralizing antibodies only if displayed on VLPs. Thus, combining multiple cues typically associated with viruses results in optimal antibody responses.
Collapse
Affiliation(s)
- Jan M. Sobczak
- Department of Immunology, University Clinic of Rheumatology and Immunology, Inselspital, CH-3010 Bern, Switzerland; (D.A.R.); (M.O.M.); (M.V.); (M.F.B.)
- Department of BioMedical Research, University of Bern, CH-3008 Bern, Switzerland
| | - Irena Barkovska
- Latvian Biomedical Research and Study Centre, LV-1067 Riga, Latvia; (I.B.); (I.B.); (D.S.); (A.O.); (J.J.); (J.B.); (A.Z.)
| | - Ina Balke
- Latvian Biomedical Research and Study Centre, LV-1067 Riga, Latvia; (I.B.); (I.B.); (D.S.); (A.O.); (J.J.); (J.B.); (A.Z.)
| | - Dominik A. Rothen
- Department of Immunology, University Clinic of Rheumatology and Immunology, Inselspital, CH-3010 Bern, Switzerland; (D.A.R.); (M.O.M.); (M.V.); (M.F.B.)
- Department of BioMedical Research, University of Bern, CH-3008 Bern, Switzerland
| | - Mona O. Mohsen
- Department of Immunology, University Clinic of Rheumatology and Immunology, Inselspital, CH-3010 Bern, Switzerland; (D.A.R.); (M.O.M.); (M.V.); (M.F.B.)
- Department of BioMedical Research, University of Bern, CH-3008 Bern, Switzerland
| | - Dace Skrastina
- Latvian Biomedical Research and Study Centre, LV-1067 Riga, Latvia; (I.B.); (I.B.); (D.S.); (A.O.); (J.J.); (J.B.); (A.Z.)
| | - Anete Ogrina
- Latvian Biomedical Research and Study Centre, LV-1067 Riga, Latvia; (I.B.); (I.B.); (D.S.); (A.O.); (J.J.); (J.B.); (A.Z.)
| | - Byron Martina
- Artemis Bioservices, 2629 JD Delft, The Netherlands;
- Protinhi Therapeutics, 6534 AT Nijmegen, The Netherlands
| | - Juris Jansons
- Latvian Biomedical Research and Study Centre, LV-1067 Riga, Latvia; (I.B.); (I.B.); (D.S.); (A.O.); (J.J.); (J.B.); (A.Z.)
| | - Janis Bogans
- Latvian Biomedical Research and Study Centre, LV-1067 Riga, Latvia; (I.B.); (I.B.); (D.S.); (A.O.); (J.J.); (J.B.); (A.Z.)
| | - Monique Vogel
- Department of Immunology, University Clinic of Rheumatology and Immunology, Inselspital, CH-3010 Bern, Switzerland; (D.A.R.); (M.O.M.); (M.V.); (M.F.B.)
- Department of BioMedical Research, University of Bern, CH-3008 Bern, Switzerland
| | - Martin F. Bachmann
- Department of Immunology, University Clinic of Rheumatology and Immunology, Inselspital, CH-3010 Bern, Switzerland; (D.A.R.); (M.O.M.); (M.V.); (M.F.B.)
- Department of BioMedical Research, University of Bern, CH-3008 Bern, Switzerland
- Nuffield Department of Medicine, The Jenner Institute, University of Oxford, Oxford OX3 7BN, UK
| | - Andris Zeltins
- Latvian Biomedical Research and Study Centre, LV-1067 Riga, Latvia; (I.B.); (I.B.); (D.S.); (A.O.); (J.J.); (J.B.); (A.Z.)
| |
Collapse
|
6
|
Pandey KK, Sahoo BR, Pattnaik AK. Protein Nanoparticles as Vaccine Platforms for Human and Zoonotic Viruses. Viruses 2024; 16:936. [PMID: 38932228 PMCID: PMC11209504 DOI: 10.3390/v16060936] [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: 05/07/2024] [Revised: 05/31/2024] [Accepted: 06/07/2024] [Indexed: 06/28/2024] Open
Abstract
Vaccines are one of the most effective medical interventions, playing a pivotal role in treating infectious diseases. Although traditional vaccines comprise killed, inactivated, or live-attenuated pathogens that have resulted in protective immune responses, the negative consequences of their administration have been well appreciated. Modern vaccines have evolved to contain purified antigenic subunits, epitopes, or antigen-encoding mRNAs, rendering them relatively safe. However, reduced humoral and cellular responses pose major challenges to these subunit vaccines. Protein nanoparticle (PNP)-based vaccines have garnered substantial interest in recent years for their ability to present a repetitive array of antigens for improving immunogenicity and enhancing protective responses. Discovery and characterisation of naturally occurring PNPs from various living organisms such as bacteria, archaea, viruses, insects, and eukaryotes, as well as computationally designed structures and approaches to link antigens to the PNPs, have paved the way for unprecedented advances in the field of vaccine technology. In this review, we focus on some of the widely used naturally occurring and optimally designed PNPs for their suitability as promising vaccine platforms for displaying native-like antigens from human viral pathogens for protective immune responses. Such platforms hold great promise in combating emerging and re-emerging infectious viral diseases and enhancing vaccine efficacy and safety.
Collapse
Affiliation(s)
- Kush K. Pandey
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA; (K.K.P.); (B.R.S.)
- Nebraska Center for Virology, University of Nebraska-Lincoln, Lincoln, NE 68583, USA
| | - Bikash R. Sahoo
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA; (K.K.P.); (B.R.S.)
- Nebraska Center for Virology, University of Nebraska-Lincoln, Lincoln, NE 68583, USA
| | - Asit K. Pattnaik
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA; (K.K.P.); (B.R.S.)
- Nebraska Center for Virology, University of Nebraska-Lincoln, Lincoln, NE 68583, USA
| |
Collapse
|
7
|
Govednik T, Lainšček D, Kuhar U, Lachish M, Janežič S, Štrbenc M, Krapež U, Jerala R, Atlas D, Manček-Keber M. TXM peptides inhibit SARS-CoV-2 infection, syncytia formation, and lower inflammatory consequences. Antiviral Res 2024; 222:105806. [PMID: 38211737 DOI: 10.1016/j.antiviral.2024.105806] [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: 07/07/2023] [Revised: 12/23/2023] [Accepted: 01/08/2024] [Indexed: 01/13/2024]
Abstract
After three years of the SARS-CoV-2 pandemic, the search and availability of relatively low-cost benchtop therapeutics for people not at high risk for a severe disease are still ongoing. Although vaccines and new SARS-CoV-2 variants reduce the death toll, the long COVID-19 along with neurologic symptoms can develop and persist even after a mild initial infection. Reinfections, which further increase the risk of sequelae in multiple organ systems as well as the risk of death, continue to require caution. The spike protein of SARS-CoV-2 is an important target for both vaccines and therapeutics. The presence of disulfide bonds in the receptor binding domain (RBD) of the spike protein is essential for its binding to the human ACE2 receptor and cell entry. Here, we demonstrate that thiol-reducing peptides based on the active site of oxidoreductase thioredoxin 1, called thioredoxin mimetic (TXM) peptides, can prevent syncytia formation, SARS-CoV-2 entry into cells, and infection in a mouse model. We also show that TXM peptides inhibit the redox-sensitive HIV pseudotyped viral cell entry. These results support disulfide targeting as a common therapeutic strategy for treating infections caused by viruses using redox-sensitive fusion. Furthermore, TXM peptides exert anti-inflammatory properties by lowering the activation of NF-κB and IRF signaling pathways, mitogen-activated protein kinases (MAPKs) and lipopolysaccharide (LPS)-induced cytokines in mice. The antioxidant and anti-inflammatory effects of the TXM peptides, which also cross the blood-brain barrier, in combination with prevention of viral infections, may provide a beneficial clinical strategy to lower viral infections and mitigate severe consequences of COVID-19.
Collapse
Affiliation(s)
- Tea Govednik
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, 1000, Ljubljana, Slovenia; Graduate School of Biomedicine, University of Ljubljana, 1000, Ljubljana, Slovenia
| | - Duško Lainšček
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, 1000, Ljubljana, Slovenia; Centre of Excellence EN-FIST, 1000, Ljubljana, Slovenia
| | - Urška Kuhar
- Institute for Microbiology and Parasitology, Veterinary Faculty, University of Ljubljana, 1000, Ljubljana, Slovenia
| | - Marva Lachish
- Hebrew University of Jerusalem, Jerusalem, 91904, Israel
| | - Sandra Janežič
- National Laboratory of Health, Environment and Food, 2000, Maribor, Slovenia
| | - Malan Štrbenc
- Institute for Preclinical Sciences, Veterinary Faculty, University of Ljubljana, 1000, Ljubljana, Slovenia
| | - Uroš Krapež
- Institute of Poultry, Birds, Small Mammals and Reptiles, Veterinary Faculty, University of Ljubljana, 1000, Ljubljana, Slovenia
| | - Roman Jerala
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, 1000, Ljubljana, Slovenia; Centre of Excellence EN-FIST, 1000, Ljubljana, Slovenia
| | - Daphne Atlas
- Hebrew University of Jerusalem, Jerusalem, 91904, Israel.
| | - Mateja Manček-Keber
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, 1000, Ljubljana, Slovenia; Centre of Excellence EN-FIST, 1000, Ljubljana, Slovenia.
| |
Collapse
|
8
|
Chen K, Jiang M, Liu J, Huang D, Yang YR. DNA nanostructures as biomolecular scaffolds for antigen display. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2024; 16:e1921. [PMID: 37562787 DOI: 10.1002/wnan.1921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 07/17/2023] [Accepted: 07/18/2023] [Indexed: 08/12/2023]
Abstract
Nanoparticle-based vaccines offer a multivalent approach for antigen display, efficiently activating T and B cells in the lymph nodes. Among various nanoparticle design strategies, DNA nanotechnology offers an innovative alternative platform, featuring high modularity, spatial addressing, nanoscale regulation, high functional group density, and lower self-antigenicity. This review delves into the potential of DNA nanostructures as biomolecular scaffolds for antigen display, addressing: (1) immunological mechanisms behind nanovaccines and commonly used nanoparticles in their design, (2) techniques for characterizing protein NP-antigen complexes, (3) advancements in DNA nanotechnology and DNA-protein assembly approach, (4) strategies for precise antigen presentation on DNA scaffolds, and (5) current applications and future possibilities of DNA scaffolds in antigen display. This analysis aims to highlight the transformative potential of DNA nanoscaffolds in immunology and vaccinology. This article is categorized under: Biology-Inspired Nanomaterials > Nucleic Acid-Based Structures Biology-Inspired Nanomaterials > Protein and Virus-Based Structures.
Collapse
Affiliation(s)
- Kun Chen
- CAS Key Laboratory of Nanosystem and Hierarchical Fabrication, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
- Materials Science and Engineering, Beijing University of Chemical Technology, Beijing, China
| | - Ming Jiang
- CAS Key Laboratory of Nanosystem and Hierarchical Fabrication, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
| | - Jin Liu
- CAS Key Laboratory of Nanosystem and Hierarchical Fabrication, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
- Tangdu Hospital, Air Force Medical University, Xi'an, China
| | - Deli Huang
- Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Yuhe R Yang
- CAS Key Laboratory of Nanosystem and Hierarchical Fabrication, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
9
|
Roier S, Mangala Prasad V, McNeal MM, Lee KK, Petsch B, Rauch S. mRNA-based VP8* nanoparticle vaccines against rotavirus are highly immunogenic in rodents. NPJ Vaccines 2023; 8:190. [PMID: 38129390 PMCID: PMC10739717 DOI: 10.1038/s41541-023-00790-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 12/05/2023] [Indexed: 12/23/2023] Open
Abstract
Despite the availability of live-attenuated oral vaccines, rotavirus remains a major cause of severe childhood diarrhea worldwide. Due to the growing demand for parenteral rotavirus vaccines, we developed mRNA-based vaccine candidates targeting the viral spike protein VP8*. Our monomeric P2 (universal T cell epitope)-VP8* mRNA design is equivalent to a protein vaccine currently in clinical development, while LS (lumazine synthase)-P2-VP8* was designed to form nanoparticles. Cyro-electron microscopy and western blotting-based data presented here suggest that proteins derived from LS-P2-VP8* mRNA are secreted in vitro and self-assemble into 60-mer nanoparticles displaying VP8*. mRNA encoded VP8* was immunogenic in rodents and introduced both humoral and cellular responses. LS-P2-VP8* induced superior humoral responses to P2-VP8* in guinea pigs, both as monovalent and trivalent vaccines, with encouraging responses detected against the most prevalent P genotypes. Overall, our data provide evidence that trivalent LS-P2-VP8* represents a promising mRNA-based next-generation rotavirus vaccine candidate.
Collapse
Affiliation(s)
| | - Vidya Mangala Prasad
- Department of Medicinal Chemistry, University of Washington, Seattle, WA, USA
- Molecular Biophysics Unit, Indian Institute of Science, Bangalore, India
| | - Monica M McNeal
- Department of Pediatrics, University of Cincinnati College of Medicine, Division of Infectious Diseases, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Kelly K Lee
- Department of Medicinal Chemistry, University of Washington, Seattle, WA, USA
| | | | | |
Collapse
|
10
|
Štrbenc M, Kuhar U, Lainšček D, Orehek S, Slavec B, Krapež U, Malovrh T, Majdič G. Rehoming and Other Refinements and Replacement in Procedures Using Golden Hamsters in SARS-CoV-2 Vaccine Research. Animals (Basel) 2023; 13:2616. [PMID: 37627407 PMCID: PMC10451472 DOI: 10.3390/ani13162616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 08/10/2023] [Accepted: 08/11/2023] [Indexed: 08/27/2023] Open
Abstract
Effective vaccines are needed to fight the COVID-19 pandemic. Forty golden hamsters were inoculated with two promising vaccine candidates and eighteen animals were used in pilot trials with viral challenge. ELISA assays were performed to determine endpoint serum titres for specific antibodies and virus neutralisation tests were used to evaluate the efficacy of antibodies. All tests with serum from vaccinated hamsters were negative even after booster vaccinations and changes in vaccination protocol. We concluded that antibodies did not have sufficient neutralising properties. Refinements were observed at all steps, and the in vitro method (virus neutralisation test) presented a replacement measure and ultimately lead to a reduction in the total number of animals used in the project. The institutional animal welfare officer and institutional designated veterinarian approved the reuse or rehoming of the surplus animals. Simple socialization procedures were performed and ultimately 19 animals were rehomed, and feedback was collected. Recently, FELASA published recommendations for rehoming of animals used for scientific and educational purposes, with species-specific guidelines, including mice, rats, and rabbits. Based on our positive experience and feedback from adopters, we concluded that the rehoming of rodents, including hamsters, is not only possible, but highly recommended.
Collapse
Affiliation(s)
- Malan Štrbenc
- Institute for Preclinical Sciences, Veterinary Faculty, University of Ljubljana, 1000 Ljubljana, Slovenia;
| | - Urška Kuhar
- Institute for Microbiology and Parasitology, Veterinary Faculty, University of Ljubljana, 1000 Ljubljana, Slovenia; (U.K.); (T.M.)
| | - Duško Lainšček
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, 1000 Ljubljana, Slovenia; (D.L.); (S.O.)
| | - Sara Orehek
- Department of Synthetic Biology and Immunology, National Institute of Chemistry, 1000 Ljubljana, Slovenia; (D.L.); (S.O.)
| | - Brigita Slavec
- Institute of Poultry, Birds, Small Mammals and Reptiles, Veterinary Faculty, University of Ljubljana, 1000 Ljubljana, Slovenia; (B.S.); (U.K.)
| | - Uroš Krapež
- Institute of Poultry, Birds, Small Mammals and Reptiles, Veterinary Faculty, University of Ljubljana, 1000 Ljubljana, Slovenia; (B.S.); (U.K.)
| | - Tadej Malovrh
- Institute for Microbiology and Parasitology, Veterinary Faculty, University of Ljubljana, 1000 Ljubljana, Slovenia; (U.K.); (T.M.)
| | - Gregor Majdič
- Institute for Preclinical Sciences, Veterinary Faculty, University of Ljubljana, 1000 Ljubljana, Slovenia;
| |
Collapse
|
11
|
Tursi NJ, Xu Z, Kulp DW, Weiner DB. Gene-encoded nanoparticle vaccine platforms for in vivo assembly of multimeric antigen to promote adaptive immunity. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2023; 15:e1880. [PMID: 36807845 PMCID: PMC10665986 DOI: 10.1002/wnan.1880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Revised: 01/14/2023] [Accepted: 01/19/2023] [Indexed: 02/23/2023]
Abstract
Nanoparticle vaccines are a diverse category of vaccines for the prophylaxis or treatment of various diseases. Several strategies have been employed for their optimization, especially to enhance vaccine immunogenicity and generate potent B-cell responses. Two major modalities utilized for particulate antigen vaccines include using nanoscale structures for antigen delivery and nanoparticles that are themselves vaccines due to antigen display or scaffolding-the latter of which we will define as "nanovaccines." Multimeric antigen display has a variety of immunological benefits compared to monomeric vaccines mediated through potentiating antigen-presenting cell presentation and enhancing antigen-specific B-cell responses through B-cell activation. The majority of nanovaccine assembly is done in vitro using cell lines. However, in vivo assembly of scaffolded vaccines potentiated using nucleic acids or viral vectors is a burgeoning modality of nanovaccine delivery. Several advantages to in vivo assembly exist, including lower costs of production, fewer production barriers, as well as more rapid development of novel vaccine candidates for emerging diseases such as SARS-CoV-2. This review will characterize the methods for de novo assembly of nanovaccines in the host using methods of gene delivery including nucleic acid and viral vectored vaccines. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Infectious Disease Biology-Inspired Nanomaterials > Nucleic Acid-Based Structures Biology-Inspired Nanomaterials > Protein and Virus-Based Structures Therapeutic Approaches and Drug Discovery > Emerging Technologies.
Collapse
Affiliation(s)
- Nicholas J. Tursi
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, Pennsylvania, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Ziyang Xu
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, Pennsylvania, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Daniel W. Kulp
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, Pennsylvania, USA
| | - David B. Weiner
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, Pennsylvania, USA
| |
Collapse
|
12
|
Kassardjian A, Sun E, Sookhoo J, Muthuraman K, Boligan KF, Kucharska I, Rujas E, Jetha A, Branch DR, Babiuk S, Barber B, Julien JP. Modular adjuvant-free pan-HLA-DR-immunotargeting subunit vaccine against SARS-CoV-2 elicits broad sarbecovirus-neutralizing antibody responses. Cell Rep 2023; 42:112391. [PMID: 37053069 PMCID: PMC10067452 DOI: 10.1016/j.celrep.2023.112391] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 02/14/2023] [Accepted: 03/29/2023] [Indexed: 04/05/2023] Open
Abstract
Subunit vaccines typically require co-administration with an adjuvant to elicit protective immunity, adding development hurdles that can impede rapid pandemic responses. To circumvent the need for adjuvant in a severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) subunit vaccine, we engineer a thermostable immunotargeting vaccine (ITV) that leverages the pan-HLA-DR monoclonal antibody 44H10 to deliver the viral spike protein receptor-binding domain (RBD) to antigen-presenting cells. X-ray crystallography shows that 44H10 binds to a conserved epitope on HLA-DR, providing the basis for its broad HLA-DR reactivity. Adjuvant-free ITV immunization in rabbits and ferrets induces robust anti-RBD antibody responses that neutralize SARS-CoV-2 variants of concern and protect recipients from SARS-CoV-2 challenge. We demonstrate that the modular nature of the ITV scaffold with respect to helper T cell epitopes and diverse RBD antigens facilitates broad sarbecovirus neutralization. Our findings support anti-HLA-DR immunotargeting as an effective means to induce strong antibody responses to subunit antigens without requiring an adjuvant.
Collapse
Affiliation(s)
- Audrey Kassardjian
- Program in Molecular Medicine, The Hospital for Sick Children Research Institute, Toronto, ON M5G 0A4, Canada; Department of Immunology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Eric Sun
- Program in Molecular Medicine, The Hospital for Sick Children Research Institute, Toronto, ON M5G 0A4, Canada; Department of Immunology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Jamie Sookhoo
- Canadian Food Inspection Agency, National Centre for Foreign Animal Disease, Winnipeg, MB R3E 3M4, Canada; Department of Immunology, University of Manitoba, Winnipeg, MB R3E 0T5, Canada
| | - Krithika Muthuraman
- Program in Molecular Medicine, The Hospital for Sick Children Research Institute, Toronto, ON M5G 0A4, Canada; Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada
| | | | - Iga Kucharska
- Program in Molecular Medicine, The Hospital for Sick Children Research Institute, Toronto, ON M5G 0A4, Canada
| | - Edurne Rujas
- Program in Molecular Medicine, The Hospital for Sick Children Research Institute, Toronto, ON M5G 0A4, Canada; Ikerbasque, Basque Foundation for Science, 48013 Bilbao, Spain; Pharmacokinetic, Nanotechnology and Gene Therapy Group, Faculty of Pharmacy, University of the Basque Country UPV/EHU, 01006 Vitoria, Spain; Bioaraba, Microbiology, Infectious Disease, Antimicrobial Agents, and Gene Therapy, 01006 Vitoria, Spain
| | - Arif Jetha
- Program in Molecular Medicine, The Hospital for Sick Children Research Institute, Toronto, ON M5G 0A4, Canada
| | - Donald R Branch
- Canadian Blood Services, Keenan Research Centre, Toronto, ON M5B 1W8, Canada; University of Toronto, Departments of Medicine and Laboratory Medicine and Pathobiology, Toronto, ON M5S 1A8, Canada
| | - Shawn Babiuk
- Canadian Food Inspection Agency, National Centre for Foreign Animal Disease, Winnipeg, MB R3E 3M4, Canada; Department of Immunology, University of Manitoba, Winnipeg, MB R3E 0T5, Canada
| | - Brian Barber
- Department of Immunology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Jean-Philippe Julien
- Program in Molecular Medicine, The Hospital for Sick Children Research Institute, Toronto, ON M5G 0A4, Canada; Department of Immunology, University of Toronto, Toronto, ON M5S 1A8, Canada; Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada.
| |
Collapse
|
13
|
Tapia D, Reyes-Sandoval A, Sanchez-Villamil JI. Protein-based Nanoparticle Vaccine Approaches Against Infectious Diseases. Arch Med Res 2023; 54:168-175. [PMID: 36894463 DOI: 10.1016/j.arcmed.2023.02.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 01/10/2023] [Accepted: 02/02/2023] [Indexed: 03/09/2023]
Abstract
The field of vaccine development has seen an increase in the number of rationally designed technologies that increase effectiveness against vaccine-resistant pathogens, while not compromising safety. Yet, there is still an urgent need to expand and further understand these platforms against complex pathogens that often evade protective responses. Nanoscale platforms have been at the center of new studies, especially in the wake of the coronavirus disease 2019 (COVID-19), with the aim of deploying safe and effective vaccines in a short time period. The intrinsic properties of protein-based nanoparticles, such as biocompatibility, flexible physicochemical characteristics, and variety have made them an attractive platform against different infectious disease agents. In the past decade, several studies have tested both lumazine synthase-, ferritin-, and albumin-based nanoplatforms against a wide range of complex pathogens in pre-clinical studies. Owed to their success in pre-clinical studies, several studies are undergoing human clinical trials or are near an initial phase. In this review we highlight the different protein-based platforms, mechanisms of synthesis, and effectiveness of these over the past decade. In addition, some challenges, and future directions to increase their effectiveness are also highlighted. Taken together, protein-based nanoscaffolds have proven to be an effective means to design rationally designed vaccines, especially against complex pathogens and emerging infectious diseases.
Collapse
Affiliation(s)
- Daniel Tapia
- The Ragon Institute of Massachusetts General Hospital, The Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA
| | - Arturo Reyes-Sandoval
- Instituto Politécnico Nacional, Escuela Nacional de Ciencias Biológicas, Laboratorio Nacional de Vacunología y Virus Tropicales, Ciudad de México, México
| | - Javier I Sanchez-Villamil
- Instituto Politécnico Nacional, Centro de Investigación en Ciencia Aplicada y Tecnología Avanzada, Unidad Morelos, Atlacholoaya, Morelos, México.
| |
Collapse
|
14
|
Kim SA, Lee Y, Ko Y, Kim S, Kim GB, Lee NK, Ahn W, Kim N, Nam GH, Lee EJ, Kim IS. Protein-based nanocages for vaccine development. J Control Release 2023; 353:767-791. [PMID: 36516900 DOI: 10.1016/j.jconrel.2022.12.022] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 12/02/2022] [Accepted: 12/09/2022] [Indexed: 12/23/2022]
Abstract
Protein nanocages have attracted considerable attention in various fields of nanomedicine due to their intrinsic properties, including biocompatibility, biodegradability, high structural stability, and ease of modification of their surfaces and inner cavities. In vaccine development, these protein nanocages are suited for efficient targeting to and retention in the lymph nodes and can enhance immunogenicity through various mechanisms, including excellent uptake by antigen-presenting cells and crosslinking with multiple B cell receptors. This review highlights the superiority of protein nanocages as antigen delivery carriers based on their physiological and immunological properties such as biodistribution, immunogenicity, stability, and multifunctionality. With a focus on design, we discuss the utilization and efficacy of protein nanocages such as virus-like particles, caged proteins, and artificial caged proteins against cancer and infectious diseases such as coronavirus disease 2019 (COVID-19). In addition, we summarize available knowledge on the protein nanocages that are currently used in clinical trials and provide a general outlook on conventional distribution techniques and hurdles faced, particularly for therapeutic cancer vaccines.
Collapse
Affiliation(s)
- Seong A Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, Republic of Korea; Chemical & Biological Integrative Research Center, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Yeram Lee
- Department of Chemical Engineering, Kyungpook National University, Daegu, Republic of Korea
| | - Yeju Ko
- Department of Chemical Engineering, Kyungpook National University, Daegu, Republic of Korea
| | - Seohyun Kim
- Department of Research and Development, SHIFTBIO INC., Seoul, Republic of Korea
| | - Gi Beom Kim
- Department of Research and Development, SHIFTBIO INC., Seoul, Republic of Korea
| | - Na Kyeong Lee
- Chemical & Biological Integrative Research Center, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Wonkyung Ahn
- Department of Chemical Engineering, Kyungpook National University, Daegu, Republic of Korea
| | - Nayeon Kim
- Department of Chemical Engineering, Kyungpook National University, Daegu, Republic of Korea
| | - Gi-Hoon Nam
- Department of Research and Development, SHIFTBIO INC., Seoul, Republic of Korea; Department of Biochemistry & Molecular Biology, Korea University College of Medicine, Seoul, Republic of Korea
| | - Eun Jung Lee
- Department of Chemical Engineering, Kyungpook National University, Daegu, Republic of Korea.
| | - In-San Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, Republic of Korea; Chemical & Biological Integrative Research Center, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul, Republic of Korea; Department of Chemical Engineering, Kyungpook National University, Daegu, Republic of Korea.
| |
Collapse
|
15
|
Kraft JC, Pham MN, Shehata L, Brinkkemper M, Boyoglu-Barnum S, Sprouse KR, Walls AC, Cheng S, Murphy M, Pettie D, Ahlrichs M, Sydeman C, Johnson M, Blackstone A, Ellis D, Ravichandran R, Fiala B, Wrenn S, Miranda M, Sliepen K, Brouwer PJM, Antanasijevic A, Veesler D, Ward AB, Kanekiyo M, Pepper M, Sanders RW, King NP. Antigen- and scaffold-specific antibody responses to protein nanoparticle immunogens. Cell Rep Med 2022; 3:100780. [PMID: 36206752 PMCID: PMC9589121 DOI: 10.1016/j.xcrm.2022.100780] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 05/27/2022] [Accepted: 09/22/2022] [Indexed: 11/29/2022]
Abstract
Protein nanoparticle scaffolds are increasingly used in next-generation vaccine designs, and several have established records of clinical safety and efficacy. Yet the rules for how immune responses specific to nanoparticle scaffolds affect the immunogenicity of displayed antigens have not been established. Here we define relationships between anti-scaffold and antigen-specific antibody responses elicited by protein nanoparticle immunogens. We report that dampening anti-scaffold responses by physical masking does not enhance antigen-specific antibody responses. In a series of immunogens that all use the same nanoparticle scaffold but display four different antigens, only HIV-1 envelope glycoprotein (Env) is subdominant to the scaffold. However, we also demonstrate that scaffold-specific antibody responses can competitively inhibit antigen-specific responses when the scaffold is provided in excess. Overall, our results suggest that anti-scaffold antibody responses are unlikely to suppress antigen-specific antibody responses for protein nanoparticle immunogens in which the antigen is immunodominant over the scaffold.
Collapse
Affiliation(s)
- John C Kraft
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA; Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
| | - Minh N Pham
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA; Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
| | - Laila Shehata
- Department of Immunology, University of Washington, Seattle, WA 98195, USA
| | - Mitch Brinkkemper
- Department of Medical Microbiology, Amsterdam UMC, University of Amsterdam, Amsterdam Infection and Immunity Institute, 1105 AZ Amsterdam, the Netherlands
| | - Seyhan Boyoglu-Barnum
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Kaitlin R Sprouse
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Alexandra C Walls
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA; Howard Hughes Medical Institute, Seattle, WA 98195, USA
| | - Suna Cheng
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA; Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
| | - Mike Murphy
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA; Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
| | - Deleah Pettie
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA; Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
| | - Maggie Ahlrichs
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA; Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
| | - Claire Sydeman
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA; Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
| | - Max Johnson
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA; Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
| | - Alyssa Blackstone
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA; Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
| | - Daniel Ellis
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA; Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
| | - Rashmi Ravichandran
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA; Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
| | - Brooke Fiala
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA; Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
| | - Samuel Wrenn
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA; Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
| | - Marcos Miranda
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA; Institute for Protein Design, University of Washington, Seattle, WA 98195, USA
| | - Kwinten Sliepen
- Department of Medical Microbiology, Amsterdam UMC, University of Amsterdam, Amsterdam Infection and Immunity Institute, 1105 AZ Amsterdam, the Netherlands
| | - Philip J M Brouwer
- Department of Medical Microbiology, Amsterdam UMC, University of Amsterdam, Amsterdam Infection and Immunity Institute, 1105 AZ Amsterdam, the Netherlands
| | - Aleksandar Antanasijevic
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - David Veesler
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA; Howard Hughes Medical Institute, Seattle, WA 98195, USA
| | - Andrew B Ward
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Masaru Kanekiyo
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Marion Pepper
- Department of Immunology, University of Washington, Seattle, WA 98195, USA
| | - Rogier W Sanders
- Department of Medical Microbiology, Amsterdam UMC, University of Amsterdam, Amsterdam Infection and Immunity Institute, 1105 AZ Amsterdam, the Netherlands; Department of Microbiology and Immunology, Weill Medical College of Cornell University, New York, NY 10021, USA
| | - Neil P King
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA; Institute for Protein Design, University of Washington, Seattle, WA 98195, USA.
| |
Collapse
|
16
|
Kou S, Chen W, Sun C, Sun F. SpyStapler-mediated assembly of nanoparticle vaccines. NANO RESEARCH 2022; 16:2821-2828. [PMID: 36258758 PMCID: PMC9561328 DOI: 10.1007/s12274-022-4951-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 08/22/2022] [Accepted: 08/23/2022] [Indexed: 06/16/2023]
Abstract
UNLABELLED The COVID-19 pandemic caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has wreaked havoc around the globe, with no end in sight. The rapid emergence of viral mutants, marked by rapid transmission and effective immune evasion, has also posed unprecedented challenges for vaccine development, not least in its speed, mass production, and distribution. Here we report a versatile "plug-and-display" strategy for creating protein vaccines, including those against malaria parasites and SARS-CoV-2, through the combined use of the intrinsically disordered protein ligase SpyStapler and computationally designed viral-like particles. The resulting protein nanoparticles harboring multiple antigens induce potent neutralizing antibody responses in mice, substantially stronger than those induced by the corresponding free antigens. This modular vaccine design enabled by SpyStapler furnishes us with a new weapon for combatting infectious diseases. ELECTRONIC SUPPLEMENTARY MATERIAL Supplementary material (further details of the protein sequences, cloning procedures, TEM imaging, ELISA details, and reaction controls) is available in the online version of this article at 10.1007/s12274-022-4951-9.
Collapse
Affiliation(s)
- Songzi Kou
- Biomedical Research Institute, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, 518036 China
- Greater Bay Biomedical Innocenter, Shenzhen Bay Laboratory, Shenzhen, 518132 China
| | - Weitao Chen
- Biomedical Research Institute, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, 518036 China
| | - Chenbo Sun
- Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Fei Sun
- Biomedical Research Institute, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, 518036 China
- Greater Bay Biomedical Innocenter, Shenzhen Bay Laboratory, Shenzhen, 518132 China
- Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| |
Collapse
|
17
|
Bergant V, Yamada S, Grass V, Tsukamoto Y, Lavacca T, Krey K, Mühlhofer MT, Wittmann S, Ensser A, Herrmann A, Vom Hemdt A, Tomita Y, Matsuyama S, Hirokawa T, Huang Y, Piras A, Jakwerth CA, Oelsner M, Thieme S, Graf A, Krebs S, Blum H, Kümmerer BM, Stukalov A, Schmidt-Weber CB, Igarashi M, Gramberg T, Pichlmair A, Kato H. Attenuation of SARS-CoV-2 replication and associated inflammation by concomitant targeting of viral and host cap 2'-O-ribose methyltransferases. EMBO J 2022; 41:e111608. [PMID: 35833542 PMCID: PMC9350232 DOI: 10.15252/embj.2022111608] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Revised: 06/23/2022] [Accepted: 06/27/2022] [Indexed: 12/12/2022] Open
Abstract
The SARS‐CoV‐2 infection cycle is a multistage process that relies on functional interactions between the host and the pathogen. Here, we repurposed antiviral drugs against both viral and host enzymes to pharmaceutically block methylation of the viral RNA 2'‐O‐ribose cap needed for viral immune escape. We find that the host cap 2'‐O‐ribose methyltransferase MTr1 can compensate for loss of viral NSP16 methyltransferase in facilitating virus replication. Concomitant inhibition of MTr1 and NSP16 efficiently suppresses SARS‐CoV‐2 replication. Using in silico target‐based drug screening, we identify a bispecific MTr1/NSP16 inhibitor with anti‐SARS‐CoV‐2 activity in vitro and in vivo but with unfavorable side effects. We further show antiviral activity of inhibitors that target independent stages of the host SAM cycle providing the methyltransferase co‐substrate. In particular, the adenosylhomocysteinase (AHCY) inhibitor DZNep is antiviral in in vitro, in ex vivo, and in a mouse infection model and synergizes with existing COVID‐19 treatments. Moreover, DZNep exhibits a strong immunomodulatory effect curbing infection‐induced hyperinflammation and reduces lung fibrosis markers ex vivo. Thus, multispecific and metabolic MTase inhibitors constitute yet unexplored treatment options against COVID‐19.
Collapse
Affiliation(s)
- Valter Bergant
- Institute of Virology, School of Medicine, Technical University of Munich (TUM), Munich, Germany
| | - Shintaro Yamada
- Institute of Cardiovascular Immunology, University Hospital Bonn (UKB), Bonn, Germany
| | - Vincent Grass
- Institute of Virology, School of Medicine, Technical University of Munich (TUM), Munich, Germany
| | - Yuta Tsukamoto
- Institute of Cardiovascular Immunology, University Hospital Bonn (UKB), Bonn, Germany
| | - Teresa Lavacca
- Institute of Virology, School of Medicine, Technical University of Munich (TUM), Munich, Germany
| | - Karsten Krey
- Institute of Virology, School of Medicine, Technical University of Munich (TUM), Munich, Germany
| | - Maria-Teresa Mühlhofer
- Institute of Virology, School of Medicine, Technical University of Munich (TUM), Munich, Germany
| | - Sabine Wittmann
- Institute of Clinical and Molecular Virology, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Armin Ensser
- Institute of Clinical and Molecular Virology, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Alexandra Herrmann
- Institute of Clinical and Molecular Virology, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Anja Vom Hemdt
- Institute of Virology, Medical Faculty, University of Bonn, Bonn, Germany
| | - Yuriko Tomita
- Department of Virology III, National Institute of Infectious Diseases (NIID), Tokyo, Japan
| | - Shutoku Matsuyama
- Department of Virology III, National Institute of Infectious Diseases (NIID), Tokyo, Japan
| | - Takatsugu Hirokawa
- Transborder Medical Research Center, University of Tsukuba, Tsukuba, Japan.,Division of Biomedical Science, University of Tsukuba, Tsukuba, Japan.,Cellular and Molecular Biotechnology Research Institute, National Institute of Advanced Industrial Science and Technology, Tokyo, Japan
| | - Yiqi Huang
- Institute of Virology, School of Medicine, Technical University of Munich (TUM), Munich, Germany
| | - Antonio Piras
- Institute of Virology, School of Medicine, Technical University of Munich (TUM), Munich, Germany
| | - Constanze A Jakwerth
- Center for Allergy & Environment (ZAUM), Technical University of Munich (TUM) and Helmholtz Center Munich, German Research Center for Environmental Health, Member of the German Center for Lung Research (DZL), CPC-M, Munich, Germany
| | - Madlen Oelsner
- Center for Allergy & Environment (ZAUM), Technical University of Munich (TUM) and Helmholtz Center Munich, German Research Center for Environmental Health, Member of the German Center for Lung Research (DZL), CPC-M, Munich, Germany
| | - Susanne Thieme
- Laboratory for functional genome analysis (LAFUGA), Gene Centre, Ludwig Maximilian University of Munich (LMU), Munich, Germany
| | - Alexander Graf
- Laboratory for functional genome analysis (LAFUGA), Gene Centre, Ludwig Maximilian University of Munich (LMU), Munich, Germany
| | - Stefan Krebs
- Laboratory for functional genome analysis (LAFUGA), Gene Centre, Ludwig Maximilian University of Munich (LMU), Munich, Germany
| | - Helmut Blum
- Laboratory for functional genome analysis (LAFUGA), Gene Centre, Ludwig Maximilian University of Munich (LMU), Munich, Germany
| | - Beate M Kümmerer
- Institute of Virology, Medical Faculty, University of Bonn, Bonn, Germany.,German Centre for Infection Research (DZIF), partner site Bonn-Cologne, Bonn, Germany
| | - Alexey Stukalov
- Institute of Virology, School of Medicine, Technical University of Munich (TUM), Munich, Germany
| | - Carsten B Schmidt-Weber
- Center for Allergy & Environment (ZAUM), Technical University of Munich (TUM) and Helmholtz Center Munich, German Research Center for Environmental Health, Member of the German Center for Lung Research (DZL), CPC-M, Munich, Germany
| | - Manabu Igarashi
- International Collaboration Unit, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Hokkaido, Japan.,Division of Global Epidemiology, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Thomas Gramberg
- Institute of Clinical and Molecular Virology, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Andreas Pichlmair
- Institute of Virology, School of Medicine, Technical University of Munich (TUM), Munich, Germany.,German Center for Infection Research (DZIF), Munich partner site, Germany
| | - Hiroki Kato
- Institute of Cardiovascular Immunology, University Hospital Bonn (UKB), Bonn, Germany
| |
Collapse
|
18
|
Kim SA, Kim S, Kim GB, Goo J, Kim N, Lee Y, Nam GH, Lim S, Kim T, Chang KH, Lee TG, Kim IS, Lee EJ. A Multivalent Vaccine Based on Ferritin Nanocage Elicits Potent Protective Immune Responses against SARS-CoV-2 Mutations. Int J Mol Sci 2022; 23:6123. [PMID: 35682801 PMCID: PMC9181758 DOI: 10.3390/ijms23116123] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 05/27/2022] [Accepted: 05/27/2022] [Indexed: 02/05/2023] Open
Abstract
The SARS-CoV-2 pandemic has created a global public crisis and heavily affected personal lives, healthcare systems, and global economies. Virus variants are continuously emerging, and, thus, the pandemic has been ongoing for over two years. Vaccines were rapidly developed based on the original SARS-CoV-2 (Wuhan-Hu-1) to build immunity against the coronavirus disease. However, they had a very low effect on the virus' variants due to their low cross-reactivity. In this study, a multivalent SARS-CoV-2 vaccine was developed using ferritin nanocages, which display the spike protein from the Wuhan-Hu-1, B.1.351, or B.1.429 SARS-CoV-2 on their surfaces. We show that the mixture of three SARS-CoV-2 spike-protein-displaying nanocages elicits CD4+ and CD8+ T cells and B-cell immunity successfully in vivo. Furthermore, they generate a more consistent antibody response against the B.1.351 and B.1.429 variants than a monovalent vaccine. This leads us to believe that the proposed ferritin-nanocage-based multivalent vaccine platform will provide strong protection against emerging SARS-CoV-2 variants of concern (VOCs).
Collapse
Affiliation(s)
- Seong A. Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Korea;
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul 02456, Korea;
| | - Seohyun Kim
- Department of Research and Development, ShiftBio, Seoul 02751, Korea; (S.K.); (G.B.K.); (G.-H.N.)
| | - Gi Beom Kim
- Department of Research and Development, ShiftBio, Seoul 02751, Korea; (S.K.); (G.B.K.); (G.-H.N.)
| | - Jiyoung Goo
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul 02456, Korea;
- KHU-KIST Department of Converging Science and Technology, Kyunghee University, Seoul 02447, Korea
| | - Nayeon Kim
- Department of Chemical Engineering, Kyungpook National University, Daegu 41566, Korea; (N.K.); (Y.L.)
| | - Yeram Lee
- Department of Chemical Engineering, Kyungpook National University, Daegu 41566, Korea; (N.K.); (Y.L.)
| | - Gi-Hoon Nam
- Department of Research and Development, ShiftBio, Seoul 02751, Korea; (S.K.); (G.B.K.); (G.-H.N.)
| | - Seungho Lim
- R&D Department Drug Development Division, LabGenomics Corporation, Gyeonggi-do 13488, Korea; (S.L.); (T.K.); (K.H.C.); (T.G.L.)
| | - Taeerk Kim
- R&D Department Drug Development Division, LabGenomics Corporation, Gyeonggi-do 13488, Korea; (S.L.); (T.K.); (K.H.C.); (T.G.L.)
| | - Ki Hwan Chang
- R&D Department Drug Development Division, LabGenomics Corporation, Gyeonggi-do 13488, Korea; (S.L.); (T.K.); (K.H.C.); (T.G.L.)
| | - Tae Gyu Lee
- R&D Department Drug Development Division, LabGenomics Corporation, Gyeonggi-do 13488, Korea; (S.L.); (T.K.); (K.H.C.); (T.G.L.)
| | - In-San Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Korea;
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul 02456, Korea;
- Department of Research and Development, ShiftBio, Seoul 02751, Korea; (S.K.); (G.B.K.); (G.-H.N.)
| | - Eun Jung Lee
- Department of Chemical Engineering, Kyungpook National University, Daegu 41566, Korea; (N.K.); (Y.L.)
| |
Collapse
|
19
|
Mabrouk MT, Huang W, Martinez‐Sobrido L, Lovell JF. Advanced Materials for SARS-CoV-2 Vaccines. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2107781. [PMID: 34894000 PMCID: PMC8957524 DOI: 10.1002/adma.202107781] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 11/28/2021] [Indexed: 05/09/2023]
Abstract
The ongoing coronavirus disease 2019 (COVID-19) pandemic, caused by severe acute respiratory coronavirus 2 (SARS-CoV-2), has killed untold millions worldwide and has hurtled vaccines into the spotlight as a go-to approach to mitigate it. Advances in virology, genomics, structural biology, and vaccine technologies have enabled a rapid and unprecedented rollout of COVID-19 vaccines, although much of the developing world remains unvaccinated. Several new vaccine platforms have been developed or deployed against SARS-CoV-2, with most targeting the large viral Spike immunogen. Those that safely induce strong and durable antibody responses at low dosages are advantageous, as well are those that can be rapidly produced at a large scale. Virtually all COVID-19 vaccines and adjuvants possess nanoscale or microscale dimensions and represent diverse and unique biomaterials. Viral vector vaccine platforms, lipid nanoparticle mRNA vaccines and multimeric display technologies for subunit vaccines have received much attention. Nanoscale vaccine adjuvants have also been used in combination with other vaccines. To deal with the ongoing pandemic, and to be ready for potential future ones, advanced vaccine technologies will continue to be developed in the near future. Herein, the recent use of advanced materials used for developing COVID-19 vaccines is summarized.
Collapse
Affiliation(s)
- Moustafa T. Mabrouk
- Department of Biomedical EngineeringUniversity at BuffaloState University of New YorkBuffaloNY14260USA
| | - Wei‐Chiao Huang
- Department of Biomedical EngineeringUniversity at BuffaloState University of New YorkBuffaloNY14260USA
| | - Luis Martinez‐Sobrido
- Division of Disease Intervention and PreventionTexas Biomedical Research InstituteSan AntonioTX78227USA
| | - Jonathan F. Lovell
- Department of Biomedical EngineeringUniversity at BuffaloState University of New YorkBuffaloNY14260USA
| |
Collapse
|
20
|
Sung HD, Kim N, Lee Y, Lee EJ. Protein-Based Nanoparticle Vaccines for SARS-CoV-2. Int J Mol Sci 2021; 22:13445. [PMID: 34948241 PMCID: PMC8703262 DOI: 10.3390/ijms222413445] [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: 10/30/2021] [Revised: 12/02/2021] [Accepted: 12/10/2021] [Indexed: 12/17/2022] Open
Abstract
The pandemic caused by the severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) has upended healthcare systems and economies around the world. Rapid understanding of the structural biology and pathogenesis of SARS-CoV-2 has allowed the development of emergency use or FDA-approved vaccines and various candidate vaccines. Among the recently developed SARS-CoV-2 candidate vaccines, natural protein-based nanoparticles well suited for multivalent antigen presentation and enhanced immune stimulation to elicit potent humoral and cellular immune responses are currently being investigated. This mini-review presents recent innovations in protein-based nanoparticle vaccines against SARS-CoV-2. The design and strategy of displaying antigenic domains, including spike protein, receptor-binding domain (RBD), and other domains on the surface of various protein-based nanoparticles and the performance of the developed nanoparticle-based vaccines are highlighted. In the final part of this review, we summarize and discuss recent advances in clinical trials and provide an outlook on protein-based nanoparticle vaccines.
Collapse
Affiliation(s)
- Hyo-Dong Sung
- Department of Chemical Engineering, School of Applied Chemical Engineering, Kyungpook National University, Daegu 41566, Korea
| | - Nayeon Kim
- Department of Chemical Engineering, School of Applied Chemical Engineering, Kyungpook National University, Daegu 41566, Korea
| | - Yeram Lee
- Department of Chemical Engineering, School of Applied Chemical Engineering, Kyungpook National University, Daegu 41566, Korea
| | - Eun Jung Lee
- Department of Chemical Engineering, School of Applied Chemical Engineering, Kyungpook National University, Daegu 41566, Korea
| |
Collapse
|
21
|
Manček‐Keber M, Hafner‐Bratkovič I, Lainšček D, Benčina M, Govednik T, Orehek S, Plaper T, Jazbec V, Bergant V, Grass V, Pichlmair A, Jerala R. Disruption of disulfides within RBD of SARS-CoV-2 spike protein prevents fusion and represents a target for viral entry inhibition by registered drugs. FASEB J 2021; 35:e21651. [PMID: 34004056 PMCID: PMC8206760 DOI: 10.1096/fj.202100560r] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 04/22/2021] [Accepted: 04/23/2021] [Indexed: 12/11/2022]
Abstract
The SARS-CoV-2 pandemic imposed a large burden on health and society. Therapeutics targeting different components and processes of the viral infection replication cycle are being investigated, particularly to repurpose already approved drugs. Spike protein is an important target for both vaccines and therapeutics. Insights into the mechanisms of spike-ACE2 binding and cell fusion could support the identification of compounds with inhibitory effects. Here, we demonstrate that the integrity of disulfide bonds within the receptor-binding domain (RBD) plays an important role in the membrane fusion process although their disruption does not prevent binding of spike protein to ACE2. Several reducing agents and thiol-reactive compounds are able to inhibit viral entry. N-acetyl cysteine amide, L-ascorbic acid, JTT-705, and auranofin prevented syncytia formation, viral entry into cells, and infection in a mouse model, supporting disulfides of the RBD as a therapeutically relevant target.
Collapse
Affiliation(s)
- Mateja Manček‐Keber
- Department of Synthetic Biology and ImmunologyNational Institute of ChemistryLjubljanaSlovenia
- Centre of Excellence EN‐FISTLjubljanaSlovenia
| | - Iva Hafner‐Bratkovič
- Department of Synthetic Biology and ImmunologyNational Institute of ChemistryLjubljanaSlovenia
- Centre of Excellence EN‐FISTLjubljanaSlovenia
| | - Duško Lainšček
- Department of Synthetic Biology and ImmunologyNational Institute of ChemistryLjubljanaSlovenia
- Centre of Excellence EN‐FISTLjubljanaSlovenia
| | - Mojca Benčina
- Department of Synthetic Biology and ImmunologyNational Institute of ChemistryLjubljanaSlovenia
- Centre of Excellence EN‐FISTLjubljanaSlovenia
| | - Tea Govednik
- Department of Synthetic Biology and ImmunologyNational Institute of ChemistryLjubljanaSlovenia
- Graduate School of BiomedicineUniversity of LjubljanaLjubljanaSlovenia
| | - Sara Orehek
- Department of Synthetic Biology and ImmunologyNational Institute of ChemistryLjubljanaSlovenia
- Graduate School of BiomedicineUniversity of LjubljanaLjubljanaSlovenia
| | - Tjaša Plaper
- Department of Synthetic Biology and ImmunologyNational Institute of ChemistryLjubljanaSlovenia
- Graduate School of BiomedicineUniversity of LjubljanaLjubljanaSlovenia
| | - Vid Jazbec
- Department of Synthetic Biology and ImmunologyNational Institute of ChemistryLjubljanaSlovenia
- Graduate School of BiomedicineUniversity of LjubljanaLjubljanaSlovenia
| | - Valter Bergant
- Immunopathology of Virus Infections LaboratoryInstitute of VirologyTechnical University of MunichMunichGermany
| | - Vincent Grass
- Immunopathology of Virus Infections LaboratoryInstitute of VirologyTechnical University of MunichMunichGermany
| | - Andreas Pichlmair
- Immunopathology of Virus Infections LaboratoryInstitute of VirologyTechnical University of MunichMunichGermany
| | - Roman Jerala
- Department of Synthetic Biology and ImmunologyNational Institute of ChemistryLjubljanaSlovenia
- Centre of Excellence EN‐FISTLjubljanaSlovenia
| |
Collapse
|