1
|
Kim J, Yang J, Heo S, Poo H. Evaluation of mRNA Transfection Reagents for mRNA Delivery and Vaccine Efficacy via Intramuscular Injection in Mice. ACS APPLIED BIO MATERIALS 2025; 8:4315-4324. [PMID: 40263125 DOI: 10.1021/acsabm.5c00424] [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: 04/24/2025]
Abstract
The selection of an effective delivery carrier is crucial to assessing mRNA-based vaccines and therapeutics in vivo. Although lipid nanoparticles (LNPs) are commonly used for mRNA delivery, the LNP-mRNA formulation process is laborious and time-consuming and requires a high-cost microfluidic device. Instead, mixing with commercial reagents may simplify mRNA transfection into cells. However, their potential as in vivo carriers in intramuscular vaccination in mouse models remains unclear. In this study, we used three types of commercial RNA transfection reagents, MessengerMAX (MAX; liposome), TransIT-mRNA (IT; cationic polymer), and Invivofectamine (IVF; LNP), to produce nanoparticles directly by pipetting. The particle characteristics and mRNA delivery efficacy of the mRNA-transfection reagent mixtures were analyzed. Additionally, immune responses to vaccine efficacy and protective immunity of the mRNA mixtures as vaccine antigens were evaluated in a mouse model. Although MAX and IT showed high in vitro transfection efficiencies, their in vivo performances were limited. In contrast, IVF exhibited notable particle stability and homogeneity, making it a promising delivery carrier. Intramuscular IVF injection significantly enhanced both innate and adaptive immune responses with a robust systemic protein expression. Notably, when using SARS-CoV-2 Spike mRNA, IVF showed robust humoral immune responses, including production of IgG and neutralizing antibodies, thereby resulting in complete protection against SARS-CoV-2 infection. Therefore, these findings position IVF as an accessible and efficient mRNA carrier for evaluating mRNA vaccines and therapeutic efficacy in basic research.
Collapse
Affiliation(s)
- Jungho Kim
- Department of Biomedical Science and Engineering, Konkuk University, Seoul 05029, Republic of Korea
| | - Jihyun Yang
- Infectious Disease Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea
| | - Suhyeon Heo
- Infectious Disease Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea
| | - Haryoung Poo
- Department of Biomedical Science and Engineering, Konkuk University, Seoul 05029, Republic of Korea
| |
Collapse
|
2
|
Tursi NJ, Tiwari S, Bedanova N, Kannan T, Parzych E, Okba N, Liaw K, Sárközy A, Livingston C, Trullen MI, Gary EN, Vadovics M, Laenger N, Londregan J, Khan MS, Omo-Lamai S, Muramatsu H, Blatney K, Hojecki C, Machado V, Maricic I, Smith TRF, Humeau LM, Patel A, Kossenkov A, Brenner JS, Allman D, Krammer F, Pardi N, Weiner DB. Modulation of lipid nanoparticle-formulated plasmid DNA drives innate immune activation promoting adaptive immunity. Cell Rep Med 2025; 6:102035. [PMID: 40120578 PMCID: PMC12047470 DOI: 10.1016/j.xcrm.2025.102035] [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: 07/09/2024] [Revised: 11/20/2024] [Accepted: 02/25/2025] [Indexed: 03/25/2025]
Abstract
Nucleic acid vaccines have grown in importance over the past several years, with the development of new approaches remaining a focus. We describe a lipid nanoparticle-formulated DNA (DNA-LNP) formulation which induces robust innate and adaptive immunity with similar serological potency to mRNA-LNPs and adjuvanted protein. Using an influenza hemagglutinin (HA)-encoding construct, we show that priming with our HA DNA-LNP demonstrated stimulator of interferon genes (STING)-dependent upregulation and activation of migratory dendritic cell (DC) subpopulations. HA DNA-LNP induced superior antigen-specific CD8+ T cell responses relative to mRNA-LNPs or adjuvanted protein, with memory responses persisting beyond one year. In rabbits immunized with HA DNA-LNP, we observed immune responses comparable or superior to mRNA-LNPs at the same dose. In an additional model, a SARS-CoV-2 spike-encoding DNA-LNP elicited protective efficacy comparable to spike mRNA-LNPs. Our study identifies a platform-specific priming mechanism for DNA-LNPs divergent from mRNA-LNPs or adjuvanted protein, suggesting avenues for this approach in prophylactic and therapeutic vaccine development.
Collapse
Affiliation(s)
- Nicholas J Tursi
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA 19104, USA; Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sachchidanand Tiwari
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Nicole Bedanova
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Toshitha Kannan
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Elizabeth Parzych
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Nisreen Okba
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Kevin Liaw
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA 19104, USA
| | - András Sárközy
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Cory Livingston
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Maria Ibanez Trullen
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Ebony N Gary
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Máté Vadovics
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Niklas Laenger
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA 19104, USA; Biology Department, Saint Joseph's University, Philadelphia, PA 19131, USA
| | - Jennifer Londregan
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Mohammad Suhail Khan
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Serena Omo-Lamai
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Hiromi Muramatsu
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kerry Blatney
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Casey Hojecki
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA 19104, USA
| | | | - Igor Maricic
- Inovio Pharmaceuticals, Plymouth Meeting, PA 19462, USA
| | | | | | - Ami Patel
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Andrew Kossenkov
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Jacob S Brenner
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - David Allman
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Ignaz Semmelweis Institute, Interuniversity Institute for Infection Research, Medical University of Vienna, Vienna, Austria
| | - Norbert Pardi
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - David B Weiner
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA 19104, USA.
| |
Collapse
|
3
|
Kalyoncu S, Sayili D, Kuyucu AZ, Soyturk H, Gullu S, Ersayan B, Tarman IO, Avci ME, Mert O, Haskok U, Tekin E, Akinturk H, Orkut R, Demirtas A, Tilmensagir I, Ulker C, Gungor B, Inan M. Development of a Recombinant Omicron BA.1 Subunit Vaccine Candidate in Pichia pastoris. Microb Biotechnol 2025; 18:e70077. [PMID: 39815667 PMCID: PMC11735458 DOI: 10.1111/1751-7915.70077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 12/05/2024] [Accepted: 12/23/2024] [Indexed: 01/18/2025] Open
Abstract
Low-cost and safe vaccines are needed to fill the vaccine inequity gap for future pandemics. Pichia pastoris is an ideal expression system for recombinant protein production due to its cost-effective and easy-to-scale-up process. Here, we developed a next-generation SARS-CoV2 Omicron BA.1-based recombinant vaccine candidate expressed in P. pastoris. The receptor binding domain of Omicron BA.1 spike protein (RBD-Omicron) was produced at 0.35 g/L in supernatant. With a 60% recovery after two-step purification, RBD-Omicron showed 99% purity. After in vitro characterisation of purified RBD-Omicron via chromatography, mass spectrometry, calorimetry and surface plasmon resonance-based methods, it was injected into mice for immunization studies. Three different doses of Alum and CpG adjuvanted RBD-Omicron were investigated and 10 μg RBD-Omicron gave the highest antigenicity. After two doses of vaccination, IgG titers in mice serum reached to more than 106. These serum antibodies also recognized earlier (Delta Plus: B.1.617.2) and later (Eris: EG.5, Pirola: BA.2.86) SARS-CoV2 variants. The long-term immunological response in mice was measured by analyzing serum antibody titers and T-cell response of splenocytes after 60 weeks. Interestingly, IgG titers and Th1 response were significantly high even after a year. Omicron subvariants are dominantly circulating in the world, so Omicron sub-lineage-based vaccines can be used for future pandemics. The RBD-Omicron-based vaccine candidate developed in this study is suitable for technology transfer and transition into the clinic.
Collapse
Affiliation(s)
| | - Dogu Sayili
- Izmir Biomedicine and Genome CenterIzmirTurkey
| | | | - Hakan Soyturk
- Izmir Biomedicine and Genome CenterIzmirTurkey
- Faculty of Biology and CeBiTecBielefeld UniversityBielefeldGermany
| | - Seyda Gullu
- Izmir Biomedicine and Genome CenterIzmirTurkey
| | | | | | - Mehmet Ender Avci
- Izmir Biomedicine and Genome CenterIzmirTurkey
- Dokuz Eylul UniversityIzmirTurkey
| | - Olcay Mert
- Izmir Biomedicine and Genome CenterIzmirTurkey
| | - Umut Haskok
- Izmir Biomedicine and Genome CenterIzmirTurkey
- Dokuz Eylul University, Izmir International Biomedicine and Genome InstituteIzmirTurkey
| | - Ege Tekin
- Izmir Biomedicine and Genome CenterIzmirTurkey
- Dokuz Eylul University, Izmir International Biomedicine and Genome InstituteIzmirTurkey
| | | | | | | | | | - Ceren Ulker
- Izmir Biomedicine and Genome CenterIzmirTurkey
| | | | - Mehmet Inan
- Izmir Biomedicine and Genome CenterIzmirTurkey
- Akdeniz UniversityAntalyaTurkey
| |
Collapse
|
4
|
Wang N, Wang C, Wei C, Chen M, Gao Y, Zhang Y, Wang T. Constructing the cGAMP-Aluminum Nanoparticles as a Vaccine Adjuvant-Delivery System (VADS) for Developing the Efficient Pulmonary COVID-19 Subunit Vaccines. Adv Healthc Mater 2024; 13:e2401650. [PMID: 39319481 DOI: 10.1002/adhm.202401650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Revised: 09/05/2024] [Indexed: 09/26/2024]
Abstract
The cGAMP-aluminum nanoparticles (CAN) are engineered as a vaccine adjuvant-delivery system to carry mixed RBD (receptor-binding domain) of the original severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and its new variant for developing bivalent pulmonary coronavirus disease 2019 (COVID-19) vaccines (biRBD-CAN). High phosphophilicity/adsorptivity made intrapulmonary CAN instantly form the pulmonary ingredient-coated CAN (piCAN) to possess biomimetic features enhancing biocompatibility. In vitro biRBD-CAN sparked APCs (antigen-presenting cells) to mature and make extra reactive oxygen species, engendered lysosome escape effects and enhanced proteasome activities. Through activating the intracellular stimulator of interferon genes (STING) and nucleotide-binding domain and leucine-rich repeat and pyrin domain containing proteins 3 (NALP3) inflammasome pathways to exert synergy between cGAMP and AN, biRBD-CAN stimulated APCs to secret cytokines favoring mixed Th1/Th2 immunoresponses. Mice bearing twice intrapulmonary biRBD-CAN produced high levels of mucosal antibodies, the long-lasting systemic antibodies, and potent cytotoxic T lymphocytes which efficiently erased cells displaying cognate epitopes. Notably, biRBD-CAN existed in mouse lungs and different lymph nodes for at least 48 h, unveiling their sustained immunostimulatory activity as the main mechanism underlying the long-lasting immunity and memory. Hamsters bearing twice intrapulmonary biRBD-CAN developed high resistance to pseudoviral challenges performed using different recombinant strains including the ones with distinct SARS-CoV-2-spike mutations. Thus, biRBD-CAN as a broad-spectrum pulmonary COVID-19 vaccine candidate may provide a tool for controlling the emerging SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Ning Wang
- School of Food and Bioengineering, Hefei University of Technology, 420 Jade Road, Hefei, Anhui Province, 230601, China
- School of Pharmacy, Anhui Medical University, 81 Plum Hill Road, Hefei, Anhui Province, 230032, China
| | - Can Wang
- School of Pharmacy, Anhui Medical University, 81 Plum Hill Road, Hefei, Anhui Province, 230032, China
- Department of Pharmacy, The Second People's Hospital of Lianyungang, 41 Hailian East Road, Lianyungang, Jiangsu Province, 222006, China
| | - Chunliu Wei
- School of Pharmacy, Anhui Medical University, 81 Plum Hill Road, Hefei, Anhui Province, 230032, China
| | - Minnan Chen
- School of Pharmacy, Anhui Medical University, 81 Plum Hill Road, Hefei, Anhui Province, 230032, China
| | - Yuhao Gao
- School of Pharmacy, Anhui Medical University, 81 Plum Hill Road, Hefei, Anhui Province, 230032, China
| | - Yuxi Zhang
- School of Pharmacy, Anhui Medical University, 81 Plum Hill Road, Hefei, Anhui Province, 230032, China
| | - Ting Wang
- School of Pharmacy, Anhui Medical University, 81 Plum Hill Road, Hefei, Anhui Province, 230032, China
| |
Collapse
|
5
|
Li H, Yang C, Yin L, Liu W, Zhang Z, Liu B, Sun X, Liu W, Lin Z, Liu Z, He P, Feng Y, Wang C, Wang W, Guan S, Wang Q, Chen L, Li P. Comparative immunogenicity of monovalent and bivalent adenovirus vaccines carrying spikes of early and late SARS-CoV-2 variants. Emerg Microbes Infect 2024; 13:2387447. [PMID: 39082740 PMCID: PMC11334748 DOI: 10.1080/22221751.2024.2387447] [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: 05/06/2024] [Revised: 07/08/2024] [Accepted: 07/30/2024] [Indexed: 08/13/2024]
Abstract
The continuous emergence of highly immune-evasive SARS-CoV-2 variants has challenged vaccine efficacy. A vaccine that can provide broad protection is desirable. We evaluated the immunogenicity of a series of monovalent and bivalent adenovirus-vectored vaccines containing the spikes of Wildtype (WT), Beta, Delta, Omicron subvariants BA.1, BA.2, BA.2.12.1, BA.2.13, BA.3, BA.5, BQ.1.1, and XBB. Vaccination in mice using monovalent vaccines elicited the highest neutralizing titers against each self-matched strain, but against other variants were reduced 2- to 73-fold. A bivalent vaccine consisting of WT and BA.5 broadened the neutralizing breadth against pre-Omicron and Omicron subvariants except XBB. Among bivalent vaccines based on the strains before the emergence of XBB, a bivalent vaccine consisting of BA.2 and BA.5 elicited the most potent neutralizing antibodies against Omicron subvariants, including XBB. In mice primed with injected WT vaccine, intranasal booster with a bivalent vaccine containing XBB and BA.5 could elicit broad serum and respiratory mucosal neutralizing antibodies against all late Omicron subvariants, including XBB. In mice that had been sequentially vaccinated with WT and BA.5, intranasal booster with a monovalent XBB vaccine elicited greater serum and mucosal XBB neutralizing antibodies than bivalent vaccines containing XBB. Both monovalent and bivalent XBB vaccines induced neutralizing antibodies against EG.5. Unlike the antibody response, which is highly variant-specific, mice receiving either monovalent or bivalent vaccines elicited comparable T-cell responses against all variants. Furthermore, intranasal but not intramuscular booster induced antigen-specific lung resident T cells. This study provides insights into the design of the COVID-19 vaccine and vaccination strategies.
Collapse
Affiliation(s)
- Hengchun Li
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, People’s Republic of China
- Guangzhou National Laboratory, Guangzhou, People’s Republic of China
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, People’s Republic of China
| | - Chenchen Yang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, People’s Republic of China
- Guangzhou National Laboratory, Guangzhou, People’s Republic of China
| | - Li Yin
- State Key Laboratory of Respiratory Disease, Guangdong Laboratory of Computational Biomedicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, People’s Republic of China
- University of Chinese Academy of Sciences, Beijing, People’s Republic of China
| | - Wenming Liu
- State Key Laboratory of Respiratory Disease, Guangdong Laboratory of Computational Biomedicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, People’s Republic of China
| | - Zhengyuan Zhang
- State Key Laboratory of Respiratory Disease, Guangdong Laboratory of Computational Biomedicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, People’s Republic of China
- University of Chinese Academy of Sciences, Beijing, People’s Republic of China
| | - Bo Liu
- Guangzhou National Laboratory, Guangzhou, People’s Republic of China
| | - Xinxin Sun
- Guangzhou National Laboratory, Guangzhou, People’s Republic of China
| | - Wenhao Liu
- School of Life Sciences, Jilin University, Changchun, People’s Republic of China
| | - Zihan Lin
- State Key Laboratory of Respiratory Disease, Guangdong Laboratory of Computational Biomedicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, People’s Republic of China
- University of Chinese Academy of Sciences, Beijing, People’s Republic of China
| | - Zijian Liu
- State Key Laboratory of Respiratory Disease, Guangdong Laboratory of Computational Biomedicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, People’s Republic of China
- University of Chinese Academy of Sciences, Beijing, People’s Republic of China
| | - Ping He
- Guangzhou National Laboratory, Guangzhou, People’s Republic of China
| | - Ying Feng
- Guangzhou National Laboratory, Guangzhou, People’s Republic of China
| | - Chunhua Wang
- Guangzhou nBiomed Ltd., Guangzhou, People’s Republic of China
| | - Wei Wang
- Guangzhou Bio-island Laboratory, Guangzhou, People’s Republic of China
| | - Suhua Guan
- Guangzhou nBiomed Ltd., Guangzhou, People’s Republic of China
| | - Qian Wang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, People’s Republic of China
- Guangzhou National Laboratory, Guangzhou, People’s Republic of China
| | - Ling Chen
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, People’s Republic of China
- Guangzhou National Laboratory, Guangzhou, People’s Republic of China
- State Key Laboratory of Respiratory Disease, Guangdong Laboratory of Computational Biomedicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, People’s Republic of China
| | - Pingchao Li
- State Key Laboratory of Respiratory Disease, Guangdong Laboratory of Computational Biomedicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, People’s Republic of China
| |
Collapse
|
6
|
Palmer CS, Perdios C, Abdel-Mohsen M, Mudd J, Datta PK, Maness NJ, Lehmicke G, Golden N, Hellmers L, Coyne C, Moore Green K, Midkiff C, Williams K, Tiburcio R, Fahlberg M, Boykin K, Kenway C, Russell-Lodrigue K, Birnbaum A, Bohm R, Blair R, Dufour JP, Fischer T, Saied AA, Rappaport J. Non-human primate model of long-COVID identifies immune associates of hyperglycemia. Nat Commun 2024; 15:6664. [PMID: 39164284 PMCID: PMC11335872 DOI: 10.1038/s41467-024-50339-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 07/08/2024] [Indexed: 08/22/2024] Open
Abstract
Hyperglycemia, and exacerbation of pre-existing deficits in glucose metabolism, are manifestations of the post-acute sequelae of SARS-CoV-2. Our understanding of metabolic decline after acute COVID-19 remains unclear due to the lack of animal models. Here, we report a non-human primate model of metabolic post-acute sequelae of SARS-CoV-2 using SARS-CoV-2 infected African green monkeys. Using this model, we identify a dysregulated blood chemokine signature during acute COVID-19 that correlates with elevated and persistent hyperglycemia four months post-infection. Hyperglycemia also correlates with liver glycogen levels, but there is no evidence of substantial long-term SARS-CoV-2 replication in the liver and pancreas. Finally, we report a favorable glycemic effect of the SARS-CoV-2 mRNA vaccine, administered on day 4 post-infection. Together, these data suggest that the African green monkey model exhibits important similarities to humans and can be utilized to assess therapeutic candidates to combat COVID-related metabolic defects.
Collapse
Affiliation(s)
- Clovis S Palmer
- Tulane National Primate Research Center, Covington, LA, USA.
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA, USA.
| | - Chrysostomos Perdios
- Tulane National Primate Research Center, Covington, LA, USA
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA, USA
| | | | - Joseph Mudd
- Tulane National Primate Research Center, Covington, LA, USA
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Prasun K Datta
- Tulane National Primate Research Center, Covington, LA, USA
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Nicholas J Maness
- Tulane National Primate Research Center, Covington, LA, USA
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA, USA
| | | | - Nadia Golden
- Tulane National Primate Research Center, Covington, LA, USA
| | - Linh Hellmers
- Tulane National Primate Research Center, Covington, LA, USA
| | - Carol Coyne
- Tulane National Primate Research Center, Covington, LA, USA
| | | | - Cecily Midkiff
- Tulane National Primate Research Center, Covington, LA, USA
| | | | - Rafael Tiburcio
- Division of Experimental Medicine, Department of Medicine, University of San Francisco, CA, USA
| | | | - Kyndal Boykin
- Tulane National Primate Research Center, Covington, LA, USA
| | - Carys Kenway
- Tulane National Primate Research Center, Covington, LA, USA
| | - Kasi Russell-Lodrigue
- Tulane National Primate Research Center, Covington, LA, USA
- Department of Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | | | - Rudolf Bohm
- Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR, USA
| | - Robert Blair
- Tulane National Primate Research Center, Covington, LA, USA
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | - Jason P Dufour
- Tulane National Primate Research Center, Covington, LA, USA
- Department of Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | - Tracy Fischer
- Tulane National Primate Research Center, Covington, LA, USA
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Ahmad A Saied
- Tulane National Primate Research Center, Covington, LA, USA
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | - Jay Rappaport
- Tulane National Primate Research Center, Covington, LA, USA.
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA, USA.
| |
Collapse
|
7
|
Vander Straeten A, Sarmadi M, Daristotle JL, Kanelli M, Tostanoski LH, Collins J, Pardeshi A, Han J, Varshney D, Eshaghi B, Garcia J, Forster TA, Li G, Menon N, Pyon SL, Zhang L, Jacob-Dolan C, Powers OC, Hall K, Alsaiari SK, Wolf M, Tibbitt MW, Farra R, Barouch DH, Langer R, Jaklenec A. A microneedle vaccine printer for thermostable COVID-19 mRNA vaccines. Nat Biotechnol 2024; 42:510-517. [PMID: 37095347 PMCID: PMC10593912 DOI: 10.1038/s41587-023-01774-z] [Citation(s) in RCA: 57] [Impact Index Per Article: 57.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 03/30/2023] [Indexed: 04/26/2023]
Abstract
Decentralized manufacture of thermostable mRNA vaccines in a microneedle patch (MNP) format could enhance vaccine access in low-resource communities by eliminating the need for a cold chain and trained healthcare personnel. Here we describe an automated process for printing MNP Coronavirus Disease 2019 (COVID-19) mRNA vaccines in a standalone device. The vaccine ink is composed of lipid nanoparticles loaded with mRNA and a dissolvable polymer blend that was optimized for high bioactivity by screening formulations in vitro. We demonstrate that the resulting MNPs are shelf stable for at least 6 months at room temperature when assessed using a model mRNA construct. Vaccine loading efficiency and microneedle dissolution suggest that efficacious, microgram-scale doses of mRNA encapsulated in lipid nanoparticles could be delivered with a single patch. Immunizations in mice using manually produced MNPs with mRNA encoding severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike protein receptor-binding domain stimulate long-term immune responses similar to those of intramuscular administration.
Collapse
Affiliation(s)
- Aurélien Vander Straeten
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Morteza Sarmadi
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - John L Daristotle
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Maria Kanelli
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Lisa H Tostanoski
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Joe Collins
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Apurva Pardeshi
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Jooli Han
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Dhruv Varshney
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Behnaz Eshaghi
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Johnny Garcia
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Timothy A Forster
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Gary Li
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Nandita Menon
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Sydney L Pyon
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Linzixuan Zhang
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Catherine Jacob-Dolan
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | - Olivia C Powers
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Kevin Hall
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Shahad K Alsaiari
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Morris Wolf
- Macromolecular Engineering Laboratory, Department of Mechanical and Process Engineering, ETH Zurich, Zurich, Switzerland
| | - Mark W Tibbitt
- Macromolecular Engineering Laboratory, Department of Mechanical and Process Engineering, ETH Zurich, Zurich, Switzerland
| | | | - Dan H Barouch
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | - Robert Langer
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
| | - Ana Jaklenec
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
8
|
Collins CP, Longo DL, Murphy WJ. The immunobiology of SARS-CoV-2 infection and vaccine responses: potential influences of cross-reactive memory responses and aging on efficacy and off-target effects. Front Immunol 2024; 15:1345499. [PMID: 38469293 PMCID: PMC10925677 DOI: 10.3389/fimmu.2024.1345499] [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: 11/27/2023] [Accepted: 02/12/2024] [Indexed: 03/13/2024] Open
Abstract
Immune responses to both SARS-CoV-2 infection and its associated vaccines have been highly variable within the general population. The increasing evidence of long-lasting symptoms after resolution of infection, called post-acute sequelae of COVID-19 (PASC) or "Long COVID," suggests that immune-mediated mechanisms are at play. Closely related endemic common human coronaviruses (hCoV) can induce pre-existing and potentially cross-reactive immunity, which can then affect primary SARS-CoV-2 infection, as well as vaccination responses. The influence of pre-existing immunity from these hCoVs, as well as responses generated from original CoV2 strains or vaccines on the development of new high-affinity responses to CoV2 antigenic viral variants, needs to be better understood given the need for continuous vaccine adaptation and application in the population. Due in part to thymic involution, normal aging is associated with reduced naïve T cell compartments and impaired primary antigen responsiveness, resulting in a reliance on the pre-existing cross-reactive memory cell pool which may be of lower affinity, restricted in diversity, or of shorter duration. These effects can also be mediated by the presence of down-regulatory anti-idiotype responses which also increase in aging. Given the tremendous heterogeneity of clinical data, utilization of preclinical models offers the greatest ability to assess immune responses under a controlled setting. These models should now involve prior antigen/viral exposure combined with incorporation of modifying factors such as age on immune responses and effects. This will also allow for mechanistic dissection and understanding of the different immune pathways involved in both SARS-CoV-2 pathogen and potential vaccine responses over time and how pre-existing memory responses, including potential anti-idiotype responses, can affect efficacy as well as potential off-target effects in different tissues as well as modeling PASC.
Collapse
Affiliation(s)
- Craig P. Collins
- Graduate Program in Immunology, University of California (UC) Davis, Davis, CA, United States
| | - Dan L. Longo
- Harvard Medical School, Brigham and Women’s Hospital, Boston, MA, United States
| | - William J. Murphy
- Departments of Dermatology and Internal Medicine (Hematology/Oncology), University of California (UC) Davis School of Medicine, Sacramento, CA, United States
| |
Collapse
|
9
|
Naveed M, Ali U, Aziz T, Jabeen K, Arif MH, Alharbi M, Alasmari AF, Albekairi TH. Development and immunological evaluation of an mRNA-based vaccine targeting Naegleria fowleri for the treatment of primary amoebic meningoencephalitis. Sci Rep 2024; 14:767. [PMID: 38191579 PMCID: PMC10774437 DOI: 10.1038/s41598-023-51127-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 12/31/2023] [Indexed: 01/10/2024] Open
Abstract
More than 95% of patients fall victim to primary amoebic meningoencephalitis (PAM), a fatal disease attacking the central nervous system. Naegleria fowleri, a brain-eating microorganism, is PAM's most well-known pathogenic ameboflagellate. Despite the use of antibiotics, the fatality rate continues to rise as no clinical trials have been conducted against this disease. To address this, we mined the UniProt database for pathogenic proteins and selected assumed epitopes to create an mRNA-based vaccine. We identified thirty B-cell and T-cell epitopes for the vaccine candidate. These epitopes, secretion boosters, subcellular trafficking structures, and linkers were used to construct the vaccine candidate. Through predictive modeling and confirmation via the Ramachandran plot (with a quality factor of 92.22), we assessed secondary and 3D structures. The adjuvant RpfE was incorporated to enhance the vaccine construct's immunogenicity (GRAVY index: 0.394, instability index: 38.99, antigenicity: 0.8). The theoretical model of immunological simulations indicated favorable responses from both innate and adaptive immune cells, with memory cells expected to remain active for up to 350 days post-vaccination, while the antigen was eliminated from the body within 24 h. Notably, strong interactions were observed between the vaccine construct and TLR-4 (- 11.9 kcal/mol) and TLR-3 (- 18.2 kcal/mol).
Collapse
Affiliation(s)
- Muhammad Naveed
- Department of Biotechnology, Faculty of Science and Technology, University of Central Punjab, Lahore, 54590, Punjab, Pakistan.
| | - Urooj Ali
- Department of Biotechnology, Quaid-I-Azam University Islamabad, Islamabad, 45320, Pakistan
| | - Tariq Aziz
- Laboratory of Animal Health, Food Hygiene and Quality, Department of Agriculture, University of Ioannina, 47132, Arta, Greece.
| | - Khizra Jabeen
- Department of Biotechnology, Faculty of Science and Technology, University of Central Punjab, Lahore, 54590, Punjab, Pakistan
| | - Muhammad Hammad Arif
- Department of Biotechnology, Faculty of Science and Technology, University of Central Punjab, Lahore, 54590, Punjab, Pakistan
| | - Metab Alharbi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Post Box 2455, 11451, Riyadh, Saudi Arabia
| | - Abdullah F Alasmari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Post Box 2455, 11451, Riyadh, Saudi Arabia
| | - Thamer H Albekairi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Post Box 2455, 11451, Riyadh, Saudi Arabia
| |
Collapse
|
10
|
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: 12] [Impact Index Per Article: 6.0] [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
|
11
|
Kingstad-Bakke B, Cleven T, Bussan H, Yount BL, Uraki R, Iwatsuki-Horimoto K, Koga M, Yamamoto S, Yotsuyanagi H, Park H, Mishra JS, Kumar S, Baric RS, Halfmann PJ, Kawaoka Y, Suresh M. Airway surveillance and lung viral control by memory T cells induced by COVID-19 mRNA vaccine. JCI Insight 2023; 8:e172510. [PMID: 37796612 PMCID: PMC10721330 DOI: 10.1172/jci.insight.172510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 10/02/2023] [Indexed: 10/07/2023] Open
Abstract
Although SARS-CoV-2 evolution seeds a continuous stream of antibody-evasive viral variants, COVID-19 mRNA vaccines provide robust protection against severe disease and hospitalization. Here, we asked whether mRNA vaccine-induced memory T cells limit lung SARS-CoV-2 replication and severe disease. We show that mice and humans receiving booster BioNTech mRNA vaccine developed potent CD8 T cell responses and showed similar kinetics of expansion and contraction of granzyme B/perforin-expressing effector CD8 T cells. Both monovalent and bivalent mRNA vaccines elicited strong expansion of a heterogeneous pool of terminal effectors and memory precursor effector CD8 T cells in spleen, inguinal and mediastinal lymph nodes, pulmonary vasculature, and most surprisingly in the airways, suggestive of systemic and regional surveillance. Furthermore, we document that: (a) CD8 T cell memory persists in multiple tissues for > 200 days; (b) following challenge with pathogenic SARS-CoV-2, circulating memory CD8 T cells rapidly extravasate to the lungs and promote expeditious viral clearance, by mechanisms that require CD4 T cell help; and (c) adoptively transferred splenic memory CD8 T cells traffic to the airways and promote lung SARS-CoV-2 clearance. These findings provide insights into the critical role of memory T cells in preventing severe lung disease following breakthrough infections with antibody-evasive SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Brock Kingstad-Bakke
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Thomas Cleven
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Hailey Bussan
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Boyd L. Yount
- Department of Microbiology and Immunology, University of North Carolina-Chapel Hill, Chapel Hill, North Carolina, USA
| | - Ryuta Uraki
- Division of Virology, Institute of Medical Science, University of Tokyo, Tokyo, Japan
- The Research Center for Global Viral Diseases, National Center for Global Health and Medicine Research Institute, Tokyo, Japan
| | | | - Michiko Koga
- Department of Infectious Diseases and Applied Immunology, IMSUT Hospital of The Institute of Medical Science, and
- Division of Infectious Diseases, Advanced Clinical Research Center, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Shinya Yamamoto
- Division of Virology, Institute of Medical Science, University of Tokyo, Tokyo, Japan
- Division of Infectious Diseases, Advanced Clinical Research Center, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Hiroshi Yotsuyanagi
- Department of Infectious Diseases and Applied Immunology, IMSUT Hospital of The Institute of Medical Science, and
- Division of Infectious Diseases, Advanced Clinical Research Center, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Hongtae Park
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Jay S. Mishra
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Sathish Kumar
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Ralph S. Baric
- Department of Microbiology and Immunology, University of North Carolina-Chapel Hill, Chapel Hill, North Carolina, USA
| | - Peter J. Halfmann
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Yoshihiro Kawaoka
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Division of Virology, Institute of Medical Science, University of Tokyo, Tokyo, Japan
- The Research Center for Global Viral Diseases, National Center for Global Health and Medicine Research Institute, Tokyo, Japan
- The University of Tokyo, Pandemic Preparedness, Infection and Advanced Research Center (UTOPIA), Tokyo, Japan
| | - M. Suresh
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, Wisconsin, USA
| |
Collapse
|
12
|
Lee M, Suzuki H, Ogiwara K, Aoki R, Kato R, Nakayama M, Fukao Y, Nihei Y, Kano T, Makita Y, Muto M, Yamada K, Suzuki Y. The nucleotide-sensing Toll-Like Receptor 9/Toll-Like Receptor 7 system is a potential therapeutic target for IgA nephropathy. Kidney Int 2023; 104:943-955. [PMID: 37648155 DOI: 10.1016/j.kint.2023.08.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 07/26/2023] [Accepted: 08/07/2023] [Indexed: 09/01/2023]
Abstract
The progression determinants of IgA nephropathy (IgAN) are still not fully elucidated. We have previously demonstrated that the mucosal activation of toll-like receptor (TLR) 9, which senses microbial unmethylated CpG DNA, influences progression by producing aberrantly glycosylated IgA. However, numerous recent reports of patients with IgAN presenting with gross hematuria after the mRNA vaccination for coronavirus disease 2019 suggest that the RNA-sensing system also exacerbates IgAN. Here, we investigated whether TLR7, which recognizes microbial RNA, is also involved in IgAN progression using a murine model and tonsil tissue from 53 patients with IgAN compared to samples from 40 patients with chronic tonsillitis and 12 patients with sleep apnea syndrome as controls. We nasally administered imiquimod, the ligand of TLR7, to IgAN-prone ddY mice and found that TLR7 stimulation elevated the serum levels of aberrantly glycosylated IgA and induced glomerular IgA depositions and proteinuria. Co-administered hydroxychloroquine, which inhibits TLRs, canceled the kidney injuries. In vitro, stimulating splenocytes from ddY mice with imiquimod increased interleukin-6 and aberrantly glycosylated IgA levels. The expression of TLR7 in the tonsils was elevated in patients with IgAN and positively correlated with that of a proliferation-inducing ligand (APRIL) involved in the production of aberrantly glycosylated IgA. Mechanistically, TLR7 stimulation enhanced the synthesis of aberrantly glycosylated IgA through the modulation of enzymes involved in the glycosylation of IgA. Thus, our findings suggest that nucleotide-sensing TLR9 and TLR7 play a crucial role in the pathogenesis of IgAN. Hence, nucleotide-sensing TLRs could be reasonably strong candidates for disease-specific therapeutic targets in IgAN.
Collapse
Affiliation(s)
- Mingfeng Lee
- Department of Nephrology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Hitoshi Suzuki
- Department of Nephrology, Juntendo University Faculty of Medicine, Tokyo, Japan; Department of Nephrology, Juntendo University Urayasu Hospital, Chiba, Japan.
| | - Kei Ogiwara
- Department of Nephrology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Ryosuke Aoki
- Department of Nephrology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Rina Kato
- Department of Nephrology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Maiko Nakayama
- Department of Nephrology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Yusuke Fukao
- Department of Nephrology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Yoshihito Nihei
- Department of Nephrology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Toshiki Kano
- Department of Nephrology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Yuko Makita
- Department of Nephrology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Masahiro Muto
- Department of Nephrology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Koshi Yamada
- Department of Nephrology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Yusuke Suzuki
- Department of Nephrology, Juntendo University Faculty of Medicine, Tokyo, Japan.
| |
Collapse
|
13
|
Kumagai T, Koyama S, Yorozu H, Kokita A, Shimizu N, Suganuma Y, Goto T. Severe hyponatremia with consciousness disturbance after receiving SARS-CoV-2 mRNA vaccination. Endocrinol Diabetes Metab Case Rep 2023; 2023:23-0004. [PMID: 37458570 PMCID: PMC10388647 DOI: 10.1530/edm-23-0004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 06/08/2023] [Indexed: 08/02/2023] Open
Abstract
Summary There are very few reports of syndrome of inappropriate antidiuresis hormone secretion (SIADH) after receiving severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) mRNA vaccine. Herein, we present the case of an 84-year-old woman who developed severe hyponatremia following the second administration of the SARS-CoV-2 mRNA vaccine. The patient presented with nausea, vomiting, and headache. Laboratory tests showed a plasma sodium level of 119 mmol/L. After receiving 500 mL of intravenous saline over a 2-h period, her plasma sodium level raised to 121 mmol/L, but her symptoms persisted. Considering that rapid plasma sodium correction was necessary, we started 3% saline solution overnight. Her plasma sodium level raised to 132 mmol/L and her symptoms completely resolved. Clinical and laboratory findings were consistent with a diagnosis of SIADH. In the absence of any other triggering factors, we concluded that the condition was likely associated with the vaccination. Clinicians should be aware of the potential for hyponatremia, particularly SIADH, associated with SARS-CoV-2 mRNA vaccination. Learning points Reports of syndrome of inappropriate antidiuresis hormone secretion after receiving severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccination are limited. If nausea, headache, and confusion are observed immediately after SARS-CoV-2 vaccination, clinicians should consider the presence of hyponatremia. As similar case reports to date have presented with severe hyponatremia, prompt treatment may be required.
Collapse
Affiliation(s)
- Takuya Kumagai
- Postgraduate Clinical Training Center, Akita Red Cross Hospital, Akita, Japan
| | - Syohei Koyama
- Department of Metabolism, Akita Red Cross Hospital, Akita, Japan
| | - Haruka Yorozu
- Department of Gastroenterology, Akita Red Cross Hospital, Akita, Japan
| | - Ayaka Kokita
- Department of Metabolism, Akita Red Cross Hospital, Akita, Japan
| | - Naoko Shimizu
- Department of Metabolism, Akita Red Cross Hospital, Akita, Japan
| | - Yumi Suganuma
- Department of Metabolism, Akita Red Cross Hospital, Akita, Japan
| | - Takashi Goto
- Department of Metabolism, Akita Red Cross Hospital, Akita, Japan
| |
Collapse
|
14
|
Hellgren F, Cagigi A, Arcoverde Cerveira R, Ols S, Kern T, Lin A, Eriksson B, Dodds MG, Jasny E, Schwendt K, Freuling C, Müller T, Corcoran M, Karlsson Hedestam GB, Petsch B, Loré K. Unmodified rabies mRNA vaccine elicits high cross-neutralizing antibody titers and diverse B cell memory responses. Nat Commun 2023; 14:3713. [PMID: 37349310 PMCID: PMC10287699 DOI: 10.1038/s41467-023-39421-5] [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: 07/23/2022] [Accepted: 06/13/2023] [Indexed: 06/24/2023] Open
Abstract
Licensed rabies virus vaccines based on whole inactivated virus are effective in humans. However, there is a lack of detailed investigations of the elicited immune response, and whether responses can be improved using novel vaccine platforms. Here we show that two doses of a lipid nanoparticle-formulated unmodified mRNA vaccine encoding the rabies virus glycoprotein (RABV-G) induces higher levels of RABV-G specific plasmablasts and T cells in blood, and plasma cells in the bone marrow compared to two doses of Rabipur in non-human primates. The mRNA vaccine also generates higher RABV-G binding and neutralizing antibody titers than Rabipur, while the degree of somatic hypermutation and clonal diversity of the response are similar for the two vaccines. The higher overall antibody titers induced by the mRNA vaccine translates into improved cross-neutralization of related lyssavirus strains, suggesting that this platform has potential for the development of a broadly protective vaccine against these viruses.
Collapse
Affiliation(s)
- Fredrika Hellgren
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
- Center of Molecular Medicine, Stockholm, Sweden
| | - Alberto Cagigi
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
- Center of Molecular Medicine, Stockholm, Sweden
- Nykode Therapeutics, Oslo, Norway
| | - Rodrigo Arcoverde Cerveira
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
- Center of Molecular Medicine, Stockholm, Sweden
| | - Sebastian Ols
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
- Center of Molecular Medicine, Stockholm, Sweden
| | - Theresa Kern
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
- Center of Molecular Medicine, Stockholm, Sweden
| | - Ang Lin
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
- Center of Molecular Medicine, Stockholm, Sweden
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Bengt Eriksson
- Astrid Fagraeus Laboratory, Comparative Medicine, Karolinska Institutet, Stockholm, Sweden
| | | | | | | | - Conrad Freuling
- Institute for Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, Greifswald-Insel Riems, Greifswald, Germany
| | - Thomas Müller
- Institute for Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, Greifswald-Insel Riems, Greifswald, Germany
| | - Martin Corcoran
- Department of Microbiology and Tumor Biology, Karolinska Institutet, Stockholm, Sweden
| | | | | | - Karin Loré
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden.
- Center of Molecular Medicine, Stockholm, Sweden.
| |
Collapse
|
15
|
Horr B, Kurtz R, Pandey A, Hoffstrom BG, Schock E, LaBonne C, Alfandari D. Production and characterization of monoclonal antibodies to Xenopus proteins. Development 2023; 150:dev201309. [PMID: 36789951 PMCID: PMC10112901 DOI: 10.1242/dev.201309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 01/24/2023] [Indexed: 02/15/2023]
Abstract
Monoclonal antibodies are powerful and versatile tools that enable the study of proteins in diverse contexts. They are often utilized to assist with identification of subcellular localization and characterization of the function of target proteins of interest. However, because there can be considerable sequence diversity between orthologous proteins in Xenopus and mammals, antibodies produced against mouse or human proteins often do not recognize Xenopus counterparts. To address this issue, we refined existing mouse monoclonal antibody production protocols to generate antibodies against Xenopus proteins of interest. Here, we describe several approaches for the generation of useful mouse anti-Xenopus antibodies to multiple Xenopus proteins and their validation in various experimental approaches. These novel antibodies are now available to the research community through the Developmental Study Hybridoma Bank (DSHB).
Collapse
Affiliation(s)
- Brett Horr
- The University of Massachusetts Amherst, Department of Veterinary and Animal Sciences, Amherst, MA 01003, USA
| | - Ryan Kurtz
- The University of Massachusetts Amherst, Department of Veterinary and Animal Sciences, Amherst, MA 01003, USA
| | - Ankit Pandey
- The University of Massachusetts Amherst, Department of Veterinary and Animal Sciences, Amherst, MA 01003, USA
| | - Benjamin G. Hoffstrom
- Antibody Technology Resource, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Elizabeth Schock
- Department of Molecular Biosciences, Northwestern University, Evanston, IL 60208, USA
| | - Carole LaBonne
- Department of Molecular Biosciences, Northwestern University, Evanston, IL 60208, USA
| | - Dominique Alfandari
- The University of Massachusetts Amherst, Department of Veterinary and Animal Sciences, Amherst, MA 01003, USA
| |
Collapse
|
16
|
Desselberger U. 14th International dsRNA Virus Symposium, Banff, Alberta, Canada, 10-14 October 2022. Virus Res 2023; 324:199032. [PMID: 36584760 PMCID: PMC10242350 DOI: 10.1016/j.virusres.2022.199032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/23/2022] [Accepted: 12/24/2022] [Indexed: 12/29/2022]
Abstract
This triennial International dsRNA Virus Symposium covered original data which have accrued during the most recent five years. In detail, the genomic diversity of these viruses continued to be explored; various structure-function studies were carried out using reverse genetics and biophysical techniques; intestinal organoids proved to be very suitable for special pathogenesis studies; and the potential of next generation rotavirus vaccines including use of rotavirus recombinants as vectored vaccine candidates was explored. 'Non-lytic release of enteric viruses in cloaked vesicles' was the topic of the keynote lecture by Nihal Altan-Bonnet, NIH, Bethesda, USA. The Jean Cohen lecturer of this meeting was Polly Roy, London School of Hygiene and Tropical Medicine, who spoke on aspects of the replication cycle of bluetongue viruses, and how some of the data are similar to details of rotavirus replication.
Collapse
Affiliation(s)
- Ulrich Desselberger
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge CB2 0QQ, U.K..
| |
Collapse
|