1
|
Mahendran TR, Cynthia B, Thevendran R, Maheswaran S. Prospects of Innovative Therapeutics in Combating the COVID-19 Pandemic. Mol Biotechnol 2025; 67:2598-2606. [PMID: 39085563 DOI: 10.1007/s12033-024-01240-4] [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: 01/19/2024] [Accepted: 07/03/2024] [Indexed: 08/02/2024]
Abstract
The sudden global crisis of COVID-19, driven by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), demands swift containment measures due to its rapid spread and numerous problematic mutations, which complicate the establishment of herd immunity. With escalating fatalities across various nations no foreseeable end in sight, there is a pressing need to create swiftly deployable, rapid, cost-effective detection, and treatment methods. While various steps are taken to mitigate the transmission and severity of the disease, vaccination is proven throughout mankind history as the best method to acquire immunity and circumvent the spread of infectious diseases. Nonetheless, relying solely on vaccination might not be adequate to match the relentless viral mutations observed in emerging variants of SARS-CoV-2, including alterations to their RBD domain, acquisition of escape mutations, and potential resistance to antibody binding. Beyond the immune system activation achieved through vaccination, it is crucial to develop new medications or treatment methods to either impede the infection or enhance existing treatment modalities. This review emphasizes innovative treatment strategies that aim to directly disrupt the virus's ability to replicate and spread, which could play a role in ending the SARS-CoV-2 pandemic.
Collapse
Affiliation(s)
- Thamby Rajah Mahendran
- School of Biological Sciences, Universiti Sains Malaysia, 11800, Minden, Penang, Malaysia
| | - Binsin Cynthia
- School of Biological Sciences, Universiti Sains Malaysia, 11800, Minden, Penang, Malaysia
| | - Ramesh Thevendran
- Centre of Excellence for Nanobiotechnology & Nanomedicine (CoExNano), Faculty of Applied Sciences, AIMST University, 08100, Bedong, Kedah, Malaysia
- Faculty of Applied Sciences, AIMST University, 08100, Bedong, Kedah, Malaysia
| | - Solayappan Maheswaran
- Centre of Excellence for Nanobiotechnology & Nanomedicine (CoExNano), Faculty of Applied Sciences, AIMST University, 08100, Bedong, Kedah, Malaysia.
- Faculty of Applied Sciences, AIMST University, 08100, Bedong, Kedah, Malaysia.
| |
Collapse
|
2
|
Azcarate D, Olasagasti Arsuaga F, Granizo Rodriguez E, Arana-Arri E, España PP, Intxausti M, Sancho C, García de Vicuña Meléndez A, Ibarrondo O, M de Pancorbo M. Human-genetic variants associated with susceptibility to SARS-CoV-2 infection. Gene 2025; 953:149423. [PMID: 40120867 DOI: 10.1016/j.gene.2025.149423] [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: 11/19/2024] [Revised: 03/13/2025] [Accepted: 03/15/2025] [Indexed: 03/25/2025]
Abstract
SARS-CoV-2, the third major coronavirus of the 21st century, causing COVID-19 disease, profoundly impacts public health and workforces worldwide. Identifying individuals at heightened risk of SARS-CoV-2 infection is crucial for targeted interventions and preparedness. This study investigated 35 SNVs within viral infection-associated genes in SARS-CoV-2 patients and uninfected controls from the Basque Country (March 2020-July 2021). Its primary aim was to uncover genetic markers indicative of SARS-CoV-2 susceptibility and explore genetic predispositions to infection. Association analyses revealed previously unreported associations between SNVs and susceptibility. Haplotype analyses uncovered novel links between haplotypes and susceptibility, surpassing individual SNV associations. Descriptive modelling identified key susceptibility factors, with rs11246068-CC (IFITM3), rs5742933-GG (ORMDL1), rs35337543-CG (IFIH1), and GGGCT (rs2070788, rs2298659, rs17854725, rs12329760, rs3787950) variation in TMPRSS2 emerging as main infection-susceptibility indicators for a COVID-19 pandemic situation. These findings underscore the importance of integrated SNV and haplotype analyses in delineating susceptibility to SARS-CoV-2 and informing proactive prevention strategies. The genetic markers profiled in this study offer valuable insights for future pandemic preparedness.
Collapse
Affiliation(s)
- Daniel Azcarate
- BIOMICs Research Group (BIOMICS and Microfluidics cluster), Zoology and animal cellular biology department, Faculty of Science and Technology (UPV/EHU), 48940 Leioa, Biscay (Basque Country), Spain
| | - Felix Olasagasti Arsuaga
- BIOMICs Research Group (BIOMICS and Microfluidics cluster), Biochemistry and molecular biology department, Faculty of Pharmacy (UPV/EHU), 01006 Vitoria-Gasteiz, Alava (Basque Country), Spain.
| | - Eva Granizo Rodriguez
- BIOMICs Research Group (BIOMICS and Microfluidics cluster), Zoology and animal cellular biology department, Faculty of Science and Technology (UPV/EHU), 48940 Leioa, Biscay (Basque Country), Spain
| | - Eunate Arana-Arri
- Clinical Epidemiology Unit, Cruces University Hospital, 48903 Barakaldo, Biscay (Basque Country), Spain
| | - Pedro Pablo España
- Pulmonology Service, Galdakao-Usansolo University Hospital, 48960 Galdakao, Biscay (Basque Country), Spain
| | - Maider Intxausti
- Pulmonology Service, Alava University Hospital - Txagorritxu, 01009 Vitoria-Gasteiz, Álava (Basque Country), Spain
| | - Cristina Sancho
- Department of Pneumology, Basurto University Hospital, 48013 Bilbao, Biscay (Basque Country), Spain
| | | | - Oliver Ibarrondo
- Consultant in Statistics and Health Economics Research, Debagoiena AP-OSI Research Unit, 20500 Arrasate, Gipuzkoa (Basque Country), Spain
| | - Marian M de Pancorbo
- BIOMICs Research Group (BIOMICS and Microfluidics cluster), Zoology and animal cellular biology department, Faculty of Science and Technology (UPV/EHU), 48940 Leioa, Biscay (Basque Country), Spain.
| |
Collapse
|
3
|
Anderson M, Lopez J, Wyr M, Ramirez PW. Defining diverse spike-receptor interactions involved in SARS-CoV-2 entry: Mechanisms and therapeutic opportunities. Virology 2025; 607:110507. [PMID: 40157321 DOI: 10.1016/j.virol.2025.110507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Revised: 03/15/2025] [Accepted: 03/19/2025] [Indexed: 04/01/2025]
Abstract
Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) is an enveloped RNA virus that caused the Coronavirus Disease 2019 (COVID-19) pandemic. The SARS-CoV-2 Spike glycoprotein binds to angiotensin converting enzyme 2 (ACE2) on host cells to facilitate viral entry. However, the presence of SARS-CoV-2 in nearly all human organs - including those with little or no ACE2 expression - suggests the involvement of alternative receptors. Recent studies have identified several cellular proteins and molecules that influence SARS-CoV-2 entry through ACE2-dependent, ACE2-independent, or inhibitory mechanisms. In this review, we explore how these alternative receptors were identified, their expression patterns and roles in viral entry, and their impact on SARS-CoV-2 infection. Additionally, we discuss therapeutic strategies aimed at disrupting these virus-receptor interactions to mitigate COVID-19 pathogenesis.
Collapse
Affiliation(s)
- Michael Anderson
- Department of Biological Sciences, California State University Long Beach, Long Beach, CA, USA
| | - Julian Lopez
- Department of Biological Sciences, California State University Long Beach, Long Beach, CA, USA
| | - Maya Wyr
- Department of Biological Sciences, California State University Long Beach, Long Beach, CA, USA
| | - Peter W Ramirez
- Department of Biological Sciences, California State University Long Beach, Long Beach, CA, USA.
| |
Collapse
|
4
|
Catanzaro NJ, Wu Z, Fan C, Jefferson V, Abdelgadir A, Schäfer A, Yount BL, Bjorkman PJ, Baric R, Letko M. ACE2 from Pipistrellus abramus bats is a receptor for HKU5 coronaviruses. Nat Commun 2025; 16:4932. [PMID: 40436893 PMCID: PMC12119886 DOI: 10.1038/s41467-025-60286-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Accepted: 05/20/2025] [Indexed: 06/01/2025] Open
Abstract
The merbecovirus subgenus of coronaviruses includes Middle East Respiratory Syndrome Coronavirus (MERS-CoV), a zoonotic pathogen transmitted from dromedary camels to humans that causes severe respiratory disease. Viral discovery efforts uncover hundreds of merbecoviruses in different species across multiple continents, but few are studied under laboratory conditions, leaving basic questions regarding their human threat potential unresolved. Viral entry into host cells is a critical step for transmission between hosts. Here, we develop and apply a scalable approach to assesses novel merbecovirus cell entry across the entire merbecovirus subgenus. Merbecoviruses are sorted into clades based on the receptor-binding domain of the spike glycoprotein. Receptor tropism is clade-specific, with the clade including MERS-CoV using DPP4 and multiple clades using ACE2, including HKU5 bat coronaviruses. Mutational analysis identifies possible structural limitations to HKU5 adaptability and a cryo-EM structure of the HKU5-20s spike trimer reveals only 'down' RBDs.
Collapse
Affiliation(s)
- Nicholas J Catanzaro
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Ziyan Wu
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Chengcheng Fan
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Victoria Jefferson
- Paul G. Allen School for Global Health, Washington State University, Pullman, WA, USA
| | - Anfal Abdelgadir
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Alexandra Schäfer
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Boyd L Yount
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Pamela J Bjorkman
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Ralph Baric
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Epidemiology, Gillings School of Global Public Heath, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Michael Letko
- Paul G. Allen School for Global Health, Washington State University, Pullman, WA, USA.
| |
Collapse
|
5
|
Levasseur MD. Nonviral protein cages as tools to decipher and combat viral threats. NPJ VIRUSES 2025; 3:45. [PMID: 40419646 DOI: 10.1038/s44298-025-00127-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2025] [Accepted: 05/12/2025] [Indexed: 05/28/2025]
Abstract
Zoonotic viruses rank among the greatest threats to public health, with urbanization and global warming accelerating their emergence and spread. As the risk of future pandemics grows, innovative tools are needed to deepen our understanding of viral pathogenesis and enhance pandemic preparedness. Nonviral protein cages provide a versatile platform for studying viral mechanisms, virus-host interactions, and designing next-generation therapeutic approaches, making them powerful assets in the fight against viral threats.
Collapse
|
6
|
Bi Z, Ren W, Zeng H, Zhou Y, Liu J, Chen Z, Zhang X, He X, Lu G, Wei Y, Wei X. LL-37 Inhibits TMPRSS2-Mediated S2' Site Cleavage and SARS-CoV-2 Infection but Not Omicron Variants. Cell Prolif 2025:e70060. [PMID: 40375579 DOI: 10.1111/cpr.70060] [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: 02/11/2025] [Revised: 04/10/2025] [Accepted: 04/29/2025] [Indexed: 05/18/2025] Open
Abstract
Continual evolution of SARS-CoV-2 spike drives the emergence of Omicron variants that show increased spreading and immune evasion. Understanding how the variants orientate themselves towards host immune defence is crucial for controlling future pandemics. Herein, we demonstrate that human cathelicidin LL-37, a crucial component of innate immunity, predominantly binds to the S2 subunit of SARS-CoV-2 spike protein, occupying sites where TMPRSS2 typically binds. This binding impedes TMPRSS2-mediated priming at site S2' and subsequent membrane fusion processes. The mutation N764K within S2 subunit of Omicron variants reduces affinity for LL-37 significantly, thereby diminishing binding capacity and inhibitory effects on membrane fusion. Moreover, the early humoral immune response enhanced by LL-37 is observed in mice against SARS-CoV-2 spike but not Omicron BA.4/5 spike. These findings reveal the mechanism underlying interactions amongst LL-37, TMPRSS2 and SARS-CoV-2 and VOCs, and highlight the distinct mutation for Omicron variants to evade the fusion activity inhibition by host innate immunity.
Collapse
Affiliation(s)
- Zhenfei Bi
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
- Department of Hepatobiliary Surgery, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Wenyan Ren
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Hao Zeng
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Yuanyuan Zhou
- Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Science, Shenzhen, China
| | - Jian Liu
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Zimin Chen
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Xindan Zhang
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Xuemei He
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Guangwen Lu
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Yuquan Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
7
|
Pantoja C, Acosta FM, Granatir S, Anderson M, Wyr M, Tailor J, Fuori A, Dower W, Marr HB, Ramirez PW. Electromagnetic waves destabilize the SARS-CoV-2 Spike protein and reduce SARS-CoV-2 Virus-Like particle (SC2-VLP) infectivity. Sci Rep 2025; 15:16836. [PMID: 40374718 PMCID: PMC12081674 DOI: 10.1038/s41598-025-01896-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 05/09/2025] [Indexed: 05/17/2025] Open
Abstract
Infection and transmission of Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) continues to pose a global public health concern. Using electromagnetic waves represents an alternative strategy to inactivate pathogenic viruses such as SARS-CoV-2. However, whether electromagnetic waves reduce SARS-CoV-2 infectivity is unclear. Here, we adapted a coplanar waveguide (CPW) to identify frequencies that could potentially neutralize SARS-CoV-2 virus-like particles (SC2-VLPs). Treatment of SC2-VLPs at frequencies between 2.5 and 3.5 GHz and an electric field of 413 V/m reduced infectivity. Exposure of SC2-VLPs to a frequency of 3.1 GHz -and to a lesser extent, 5.9 GHz- reduced their binding to antibodies targeting the SARS-CoV-2 Spike S1 receptor-binding domain (RBD) but did not alter the total levels of Spike, Nucleocapsid, Envelope, or Membrane proteins in virus particles. These results suggest that electromagnetic waves alter the conformation of Spike, thereby reducing viral attachment and entry. Overall, this data provides proof-of-concept in using electromagnetic waves for sanitation and prevention efforts to curb the transmission of SARS-CoV-2 and potentially other pathogenic enveloped viruses.
Collapse
Affiliation(s)
- Christina Pantoja
- Department of Biological Sciences, California State University Long Beach, Long Beach, CA, USA
| | - Francisco M Acosta
- Department of Biological Sciences, California State University Long Beach, Long Beach, CA, USA
| | | | - Michael Anderson
- Department of Biological Sciences, California State University Long Beach, Long Beach, CA, USA
| | - Maya Wyr
- Department of Biological Sciences, California State University Long Beach, Long Beach, CA, USA
| | - Johann Tailor
- Department of Biological Sciences, California State University Long Beach, Long Beach, CA, USA
| | - Angus Fuori
- Department of Biological Sciences, California State University Long Beach, Long Beach, CA, USA
| | | | | | - Peter W Ramirez
- Department of Biological Sciences, California State University Long Beach, Long Beach, CA, USA.
| |
Collapse
|
8
|
Le DTH, Yang C, Zhang Y, Zhao G, Ang MJY, Bae KH, Hui JHP, Hedrick JL, Yang YY. Replacing PEG-Lipid with Amphiphilic Polycarbonates in mRNA-Loaded Lipid Nanoparticles: Impact of Polycarbonate Structure on Physicochemical and Transfection Properties. Biomacromolecules 2025. [PMID: 40347133 DOI: 10.1021/acs.biomac.5c00088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/12/2025]
Abstract
Since the remarkable breakthrough of COVID-19 mRNA vaccines, lipid nanoparticles (LNPs) have gained substantial attention as the most cutting-edge clinical formulations for mRNA delivery. PEGylated lipid (PEG-lipid) has been regarded as an essential constituent of LNPs that helps to prolong their systemic circulation by preventing particle aggregation in the blood and sequestration by the mononuclear phagocyte system. Herein, we synthesized a series of mRNA-loaded nanoparticles by replacing ALC-0159 (a PEG-lipid used in the Comirnaty formulation) with amphiphilic PEG-polycarbonate diblock copolymers (PC-HNPs). Interestingly, variations of polycarbonate block length and structure significantly influenced mRNA encapsulation efficiency, transfection potency, colloidal stability, and PEG shedding rate of PC-HNPs. In vivo and ex vivo bioluminescence imaging revealed that upon subcutaneous administration in mice, the leading candidate PC3-HNP achieved lymph node accumulation comparable to that of the conventional ALC-0159-based LNP formulation while avoiding undesirable liver accumulation. Our findings may provide valuable information for the construction of next-generation nanocarriers for effective mRNA delivery.
Collapse
Affiliation(s)
- Dao Thi Hong Le
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, #06-01 Centros, Singapore 138668, Singapore
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 119288 Singapore
| | - Chuan Yang
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, #06-01 Centros, Singapore 138668, Singapore
| | - Yue Zhang
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, #06-01 Centros, Singapore 138668, Singapore
| | - Gui Zhao
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, #06-01 Centros, Singapore 138668, Singapore
| | - Melgious J Y Ang
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, #06-01 Centros, Singapore 138668, Singapore
| | - Ki Hyun Bae
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, #06-01 Centros, Singapore 138668, Singapore
| | - James H P Hui
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 119288 Singapore
| | - James L Hedrick
- IBM Almaden Research Center, 650 Harry Road, San Jose, California 95120, United States
| | - Yi Yan Yang
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, #06-01 Centros, Singapore 138668, Singapore
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 119288 Singapore
| |
Collapse
|
9
|
Fraser BJ, Wilson RP, Ferková S, Ilyassov O, Lac J, Dong A, Li YY, Seitova A, Li Y, Hejazi Z, Kenney TMG, Penn LZ, Edwards A, Leduc R, Boudreault PL, Morin GB, Bénard F, Arrowsmith CH. Structural basis of TMPRSS11D specificity and autocleavage activation. Nat Commun 2025; 16:4351. [PMID: 40348740 PMCID: PMC12065894 DOI: 10.1038/s41467-025-59677-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Accepted: 05/01/2025] [Indexed: 05/14/2025] Open
Abstract
Transmembrane Protease, Serine-2 (TMPRSS2) and TMPRSS11D are human proteases that enable SARS-CoV-2 and Influenza A/B virus infections, but their biochemical mechanisms for facilitating viral cell entry remain unclear. We show these proteases spontaneously and efficiently cleave their own zymogen activation motifs, activating their broader protease activity on cellular substrates. We determine TMPRSS11D co-crystal structures with a native and an engineered activation motif, revealing insights into its autocleavage activation and distinct substrate binding cleft features. Leveraging this structural data, we develop nanomolar potency peptidomimetic inhibitors of TMPRSS11D and TMPRSS2. We show that a broad serine protease inhibitor that underwent clinical trials for TMPRSS2-targeted COVID-19 therapy, nafamostat mesylate, was rapidly cleaved by TMPRSS11D and converted to low activity derivatives. In this work, we develop mechanistic insights into human protease viral tropism and highlight both the strengths and limitations of existing human serine protease inhibitors, informing future drug discovery efforts targeting these proteases.
Collapse
Affiliation(s)
- Bryan J Fraser
- Structural Genomics Consortium Toronto, Toronto, ON, Canada.
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada.
| | - Ryan P Wilson
- Structural Genomics Consortium Toronto, Toronto, ON, Canada
| | - Sára Ferková
- Department of Pharmacology and Physiology, Faculty of Medicine and Health Sciences, Institut de Pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
- Department of Radiology, University of British Columbia, Vancouver, BC, Canada
| | | | - Jackie Lac
- Structural Genomics Consortium Toronto, Toronto, ON, Canada
| | - Aiping Dong
- Structural Genomics Consortium Toronto, Toronto, ON, Canada
| | - Yen-Yen Li
- Structural Genomics Consortium Toronto, Toronto, ON, Canada
| | - Alma Seitova
- Structural Genomics Consortium Toronto, Toronto, ON, Canada
| | - Yanjun Li
- Structural Genomics Consortium Toronto, Toronto, ON, Canada
| | - Zahra Hejazi
- Structural Genomics Consortium Toronto, Toronto, ON, Canada
| | - Tristan M G Kenney
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
- Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - Linda Z Penn
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
- Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - Aled Edwards
- Structural Genomics Consortium Toronto, Toronto, ON, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Richard Leduc
- Department of Pharmacology and Physiology, Faculty of Medicine and Health Sciences, Institut de Pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
- Department of Radiology, University of British Columbia, Vancouver, BC, Canada
| | - Pierre-Luc Boudreault
- Department of Pharmacology and Physiology, Faculty of Medicine and Health Sciences, Institut de Pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
- Department of Radiology, University of British Columbia, Vancouver, BC, Canada
| | - Gregg B Morin
- Canada's Michael Smith Genome Sciences Centre, Vancouver, BC, Canada.
- British Columbia Cancer Research Institute, Vancouver, BC, Canada.
- University of British Columbia, Vancouver, BC, Canada.
| | - François Bénard
- British Columbia Cancer Research Institute, Vancouver, BC, Canada.
- University of British Columbia, Vancouver, BC, Canada.
| | - Cheryl H Arrowsmith
- Structural Genomics Consortium Toronto, Toronto, ON, Canada.
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada.
- Princess Margaret Cancer Centre, Toronto, ON, Canada.
| |
Collapse
|
10
|
Pita Dos Santos LG, da Silva Coutinho G, Rodrigues Guimarães JM, Miranda da Silva M, Francisco da Silva A, Marcelino Neto PP, Coral Rodrigues BC, Aaron de Almeida W, Carlos Alves Dos Santos AJ, Napoleão TH, Pontual EV. Advances in COVID-19 Therapeutics: Exploring the role of lectins and protease inhibitors. Microb Pathog 2025; 205:107687. [PMID: 40349995 DOI: 10.1016/j.micpath.2025.107687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 05/01/2025] [Accepted: 05/08/2025] [Indexed: 05/14/2025]
Abstract
The rapid global spread of SARS-CoV-2 has demanded innovative approaches to treatment and prevention. This article reviews the current landscape of COVID-19 therapeutics and vaccines, emphasizing the role of biotechnological products, particularly lectins and protease inhibitors. SARS-CoV-2, a single-stranded RNA virus, infects host cells via its spike (S) protein, which binds to the angiotensin-converting enzyme 2 (ACE2) receptor. This interaction is facilitated by host proteases like TMPRSS2, which are critical for viral entry. Treatments for COVID-19 primarily focus on antiviral drugs, anti-inflammatory agents, and monoclonal antibodies. Protease inhibitors that target viral enzymes like Mpro and PLpro have demonstrated potential. Additionally, vaccines, including mRNA-based, DNA-based, and those using viral vectors or inactivated viruses, are essential for preventing new infections. Lectins, proteins that bind specifically to carbohydrates, have emerged as potential antiviral agents. They can impede viral entry by binding to glycoproteins on the virus's surface or modulate immune responses. Studies indicate that lectins like cyanovirin-N and griffithsin exhibit significant antiviral activity against SARS-CoV-2. While most of the research on these biotechnological products is still in preclinical or early stages, their potential for treating and preventing COVID-19 is substantial. Further investigation and clinical trials are crucial to validate their efficacy and safety. This article underscores the need for continued exploration of novel therapeutic strategies to combat the evolving COVID-19 pandemic. However, the review is limited by the scarcity of clinical data on these products, highlighting the need for translational research.
Collapse
Affiliation(s)
| | - Gabriel da Silva Coutinho
- Departamento de Morfologia e Fisiologia Animal, Universidade Federal Rural de Pernambuco, Recife, Brazil
| | | | - Marcelo Miranda da Silva
- Departamento de Morfologia e Fisiologia Animal, Universidade Federal Rural de Pernambuco, Recife, Brazil
| | - Alex Francisco da Silva
- Departamento de Morfologia e Fisiologia Animal, Universidade Federal Rural de Pernambuco, Recife, Brazil
| | - Pedro Paulo Marcelino Neto
- Departamento de Bioquímica, Centro de Biociências, Universidade Federal de Pernambuco, Recife, Pernambuco, Brazil
| | - Beto Cherles Coral Rodrigues
- Departamento de Bioquímica, Centro de Biociências, Universidade Federal de Pernambuco, Recife, Pernambuco, Brazil
| | - Welton Aaron de Almeida
- Departamento de Bioquímica, Centro de Biociências, Universidade Federal de Pernambuco, Recife, Pernambuco, Brazil; Programa de Pós-Graduação em Ciências da Saúde, Universidade de Pernambuco, Recife, Pernambuco, Brazil
| | | | - Thiago Henrique Napoleão
- Departamento de Bioquímica, Centro de Biociências, Universidade Federal de Pernambuco, Recife, Pernambuco, Brazil
| | - Emmanuel Viana Pontual
- Departamento de Morfologia e Fisiologia Animal, Universidade Federal Rural de Pernambuco, Recife, Brazil.
| |
Collapse
|
11
|
Kakh M, Doroudchi M, Talepoor A. Induction of Regulatory T Cells After Virus Infection and Vaccination. Immunology 2025. [PMID: 40329764 DOI: 10.1111/imm.13927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 03/04/2025] [Accepted: 03/17/2025] [Indexed: 05/08/2025] Open
Abstract
Vaccines have been proven to be one of the safest and most effective ways to prevent and combat diseases. However, the main focus has been on the evaluation of the potency of effector mechanisms and the lack of adverse effects of vaccine candidates. Recently, the importance of induced regulatory mechanisms of the immune system after vaccination has come to light. With the increase in our knowledge about these regulatory mechanisms including the regulatory T cells (Tregs), we have come to understand the significance of this arm of the immune system in controlling immunopathology and/or diminishing the effectiveness of vaccines, especially viral vaccines. Tregs play a dual role during infectious diseases by limiting immune-mediated pathology and also contributing to chronic pathogen persistence by decreasing effector immunity and clearance of infection. Tregs may also affect immune responses after vaccination primarily by inhibiting antigen presenting cell function such as cytokine secretion and co-stimulatory molecule expression as well as effector T (Teff) and B cell function. In this article, we review the current knowledge on the induction of Tregs after several life-threatening virus infections and their available vaccines to bring them to the spotlight and emphasise that studying viral-induced antigen-specific Tregs will help us improve the effectiveness and decrease the immunopathology or side effects of viral vaccines. Trial Registration: ClinicalTrials.gov identifier: NCT04357444.
Collapse
Affiliation(s)
- MansourehKarimi Kakh
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mehrnoosh Doroudchi
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - AtefeGhamar Talepoor
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
- Autoimmune Diseases Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
12
|
Chen B, Farzan M, Choe H. SARS-CoV-2 spike protein: structure, viral entry and variants. Nat Rev Microbiol 2025:10.1038/s41579-025-01185-8. [PMID: 40328900 DOI: 10.1038/s41579-025-01185-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/08/2025] [Indexed: 05/08/2025]
Abstract
Coronavirus disease 2019 (COVID-19), caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has been a devastating global pandemic for 4 years and is now an endemic disease. With the emergence of new viral variants, COVID-19 is a continuing threat to public health despite the wide availability of vaccines. The virus-encoded trimeric spike protein (S protein) mediates SARS-CoV-2 entry into host cells and also induces strong immune responses, making it an important target for development of therapeutics and vaccines. In this Review, we summarize our latest understanding of the structure and function of the SARS-CoV-2 S protein, the molecular mechanism of viral entry and the emergence of new variants, and we discuss their implications for development of S protein-related intervention strategies.
Collapse
Affiliation(s)
- Bing Chen
- Division of Molecular Medicine, Boston Children's Hospital, and Department of Paediatrics, Harvard Medical School, Boston, MA, USA.
| | - Michael Farzan
- Division of Infectious Diseases, Boston Children's Hospital, and Department of Paediatrics, Harvard Medical School, Boston, MA, USA.
- Center for Integrated Solutions for Infectious Diseases (CISID), The Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| | - Hyeryun Choe
- Division of Infectious Diseases, Boston Children's Hospital, and Department of Paediatrics, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
13
|
Vanderkamp SG, Niazy M, Stegelmeier AA, Stinson KJ, Ricker N, Bridle BW. Cytokine, chemokine, and acute-phase protein profiles in plasma as correlative biomarkers of clinical outcomes for patients with COVID-19. Sci Rep 2025; 15:15397. [PMID: 40316702 PMCID: PMC12048561 DOI: 10.1038/s41598-025-99248-6] [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: 06/28/2024] [Accepted: 04/17/2025] [Indexed: 05/04/2025] Open
Abstract
Coronavirus disease identified in 2019 (COVID-19), caused by severe acute respiratory syndrome-coronavirus-2, had a global impact on human health and the economy. The aim of this study was to quantify cytokines, chemokines, and acute phase proteins in the plasma of patients with COVID-19 to elucidate potential biomarkers to inform prognostic and treatment decisions. Clustering analysis using the K-prototypes method identified underlying biological patterns in patients with COVID-19. The penalized multinomial logistic regression analysis identified two comorbidities (hypertension, congestive heart failure) and thirteen analytes as potential risk factors for COVID-19 progression with 88.2% accuracy. Based on a patient's age, high concentrations of interleukin (IL)-6, monocyte chemoattractant protein-1, and pentraxin 3 were important biomarkers for lethal COVID-19. Decreased concentrations of interferon gamma-induced protein-10, IL-10, and soluble tumor necrosis factor receptor I were found to be associated with mild COVID-19, while increasing concentrations of these analytes could be used to predict COVID-19 severity.
Collapse
Affiliation(s)
- Sierra G Vanderkamp
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - Maysa Niazy
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - Ashley A Stegelmeier
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | | | - Nicole Ricker
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, N1G 2W1, Canada.
| | - Byram W Bridle
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, N1G 2W1, Canada.
| |
Collapse
|
14
|
Costa SM, Simas MCDC, da Costa LJ, Silva R. Dynamics of SARS-CoV-2 Mutations in Wastewater Provide Insights into the Circulation of Virus Variants in the Population. Int J Mol Sci 2025; 26:4324. [PMID: 40362561 PMCID: PMC12072199 DOI: 10.3390/ijms26094324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2025] [Revised: 04/23/2025] [Accepted: 04/25/2025] [Indexed: 05/15/2025] Open
Abstract
SARS-CoV-2 high transmission and genomic mutations result in the emergence of new variants that impact COVID-19 vaccine efficacy and virus transmission by evading the host immune system. Wastewater-based epidemiology is an effective approach to monitor SARS-CoV-2 variants circulation in the population but is a challenge due to the presence of reaction inhibitors and the low concentrations of SARS-CoV-2 in this environment. Here, we aim to improve SARS-CoV-2 variant detection in wastewater by employing nested PCR followed by next-generation sequencing (NGS) of small amplicons of the S gene. Eight SARS-CoV-2 wastewater samples from Alegria Wastewater Treatment Plant, in Rio de Janeiro, Brazil, were collected monthly from February to September 2021. Samples were submitted to virus concentration, RNA extraction and nested PCR followed by NGS. The small amplicons were used to prepare libraries for sequencing without the need to perform any fragmentation step. We identified and calculated the frequencies of 29 mutations matching the Alpha, Beta, Gamma, Delta, Omicron, and P.2 variants. Omicron matching-mutations were detected before the lineage was classified as a variant of concern. SARS-CoV-2 wastewater sequences clustered with SARS-CoV-2 variants detected in clinical samples that circulated in 2021 in Rio de Janeiro. We show that sequencing of selected small amplicons of SARS-CoV-2 S gene allows the identification of SARS-CoV-2 variants matching mutations and their frequencies' calculation. This approach may be expanded using customizing primers for additional genomic regions, in order to differentiate current variants. Approaches that allow us to learn how variants emerge and how they relate to clinical outcomes are crucial for our understanding of the dynamics of virus variants circulation, providing valuable data for public health management.
Collapse
Affiliation(s)
- Sara Mesquita Costa
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil; (S.M.C.); (M.C.d.C.S.)
- Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil;
| | - Maria Clara da Costa Simas
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil; (S.M.C.); (M.C.d.C.S.)
| | - Luciana Jesus da Costa
- Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil;
| | - Rosane Silva
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil; (S.M.C.); (M.C.d.C.S.)
| |
Collapse
|
15
|
Liu F, Shang C, Zhang C, Jiang X, Lin Y, Wu F, Li J, Han L, Shan J, Xiao Z, Zhou W. Cnidii fructus and Sophorae Flavescentis Radix polysaccharides inhibit SARS-CoV-2 entry by interfering with Spike protein-mediated membrane fusion. Int J Biol Macromol 2025; 307:142233. [PMID: 40107561 DOI: 10.1016/j.ijbiomac.2025.142233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 03/09/2025] [Accepted: 03/16/2025] [Indexed: 03/22/2025]
Abstract
Polysaccharides derived from Traditional Chinese Medicine (TCM) show potential as therapeutic agents in the treatment of viral infections. In this study, seventeen polysaccharides extracted from TCM were screened for their inhibitory effects on SARS-CoV-2 pseudovirus (PsV) infection in ACE2-overexpressing HEK293T cells. The results revealed that Cnidii fructus polysaccharides (CFPs) and Sophorae Flavescentis Radix polysaccharides (SFPs) significantly inhibited PsV entry. Then, the effects of CFPs and SFPs on the authentic SARS-CoV-2 virus were studied. We found that CFPs and SFPs significantly inhibited cellular damage and virus invasion induced by both the Original SARS-CoV-2 strain and the Omicron variant in Vero E6 cells, without causing obvious cytotoxicity. In vivo studies demonstrated that CFPs and SFPs significantly protect mice against SARS-CoV-2 virus-induced mortality, along with reductions in viral load and lung injury. Time of addition (TOA) experiments indicated that CFPs and SFPs exert inhibitory effects during the pseudoviral pre-attachment and cell entry stages, with no substantial impacts after the PsV has entered the cells. Further mechanism studies showed that CFPs and SFPs inhibited syncytial formation primarily by suppressing the host cell surface membrane fusion process mediated by the Spike protein, without significantly affecting the endosome-mediated viral entry.
Collapse
Affiliation(s)
- Feng Liu
- Beijing Institute of Pharmacology & Toxicology, Beijing 100850, China; State Key Laboratory of National Security Specially Needed Drug, Beijing Institute of Pharmacology & Toxicology, Beijing 100850, China
| | - Chao Shang
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences (CASS), Changchun 130122, China
| | - Cheng Zhang
- College of Veterinary Medicine, Hebei Agricultural University, 2596 Lucky South Street, Baoding 071000, China
| | - Xuyong Jiang
- Beijing Institute of Pharmacology & Toxicology, Beijing 100850, China; State Key Laboratory of National Security Specially Needed Drug, Beijing Institute of Pharmacology & Toxicology, Beijing 100850, China
| | - Yanling Lin
- Beijing Institute of Pharmacology & Toxicology, Beijing 100850, China; State Key Laboratory of National Security Specially Needed Drug, Beijing Institute of Pharmacology & Toxicology, Beijing 100850, China
| | - Fushan Wu
- Beijing Institute of Pharmacology & Toxicology, Beijing 100850, China; State Key Laboratory of National Security Specially Needed Drug, Beijing Institute of Pharmacology & Toxicology, Beijing 100850, China
| | - Jingxuan Li
- Beijing Institute of Pharmacology & Toxicology, Beijing 100850, China; State Key Laboratory of National Security Specially Needed Drug, Beijing Institute of Pharmacology & Toxicology, Beijing 100850, China
| | - Lu Han
- Beijing Institute of Pharmacology & Toxicology, Beijing 100850, China; State Key Laboratory of National Security Specially Needed Drug, Beijing Institute of Pharmacology & Toxicology, Beijing 100850, China
| | - Junjie Shan
- Beijing Institute of Pharmacology & Toxicology, Beijing 100850, China; State Key Laboratory of National Security Specially Needed Drug, Beijing Institute of Pharmacology & Toxicology, Beijing 100850, China
| | - Zhiyong Xiao
- Beijing Institute of Pharmacology & Toxicology, Beijing 100850, China; State Key Laboratory of National Security Specially Needed Drug, Beijing Institute of Pharmacology & Toxicology, Beijing 100850, China.
| | - Wenxia Zhou
- Beijing Institute of Pharmacology & Toxicology, Beijing 100850, China; State Key Laboratory of National Security Specially Needed Drug, Beijing Institute of Pharmacology & Toxicology, Beijing 100850, China.
| |
Collapse
|
16
|
Dong D, Song Y, Wu S, Wang B, Peng C, Zhang W, Kong W, Zhang Z, Song J, Hou LH, Li S. Molecular basis of Ad5-nCoV vaccine-induced immunogenicity. Structure 2025; 33:858-868.e5. [PMID: 40112804 DOI: 10.1016/j.str.2025.02.009] [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: 07/18/2024] [Revised: 12/12/2024] [Accepted: 02/21/2025] [Indexed: 03/22/2025]
Abstract
Ad5-nCoV (Convidecia) is listed for emergency use against COVID-19 by the World Health Organization (WHO) and has been globally administered to millions of people. It utilizes human adenovirus 5 (Ad5) replication-incompetent vector to deliver the spike (S) protein gene from various SARS-CoV-2 strains. Despite promising clinical data, the molecular mechanism underlying its high immunogenicity and adverse reactions remain incompletely understood. Here, we primarily applied cryo-electron tomography (cryo-ET), fluorescence microscopy and mass spectrometry to analyze the Ad5-nCoV_Wu and Ad5-nCoV_O vaccine-induced S antigens. These antigens encode the unmodified SARS-CoV-2 Wuhan-Hu-1 S gene and the stabilized Omicron S gene, respectively. Our findings highlight the structural integrity, antigenicity, and dense distribution on cell membrane of the vaccine-induced S proteins. Ad5-nCoV_O induced S proteins exhibit improved stability and reduced syncytia formation among inoculated cells. Our work demonstrates that Ad5-nCoV is a prominent platform for antigen induction and cryo-ET can be a useful technique for vaccine characterization and development.
Collapse
Affiliation(s)
- Dongyang Dong
- Beijing Frontier Research Center for Biological Structure & Tsinghua-Peking Center for Life Sciences & State Key Laboratory of Membrane Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Yutong Song
- Beijing Frontier Research Center for Biological Structure & Tsinghua-Peking Center for Life Sciences & State Key Laboratory of Membrane Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Shipo Wu
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing 100071, China
| | - Busen Wang
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing 100071, China
| | - Cheng Peng
- Beijing Frontier Research Center for Biological Structure & Tsinghua-Peking Center for Life Sciences & State Key Laboratory of Membrane Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Weiping Zhang
- Beijing Frontier Research Center for Biological Structure & Tsinghua-Peking Center for Life Sciences & State Key Laboratory of Membrane Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Weizheng Kong
- Beijing Frontier Research Center for Biological Structure & Tsinghua-Peking Center for Life Sciences & State Key Laboratory of Membrane Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Zheyuan Zhang
- Beijing Frontier Research Center for Biological Structure & Tsinghua-Peking Center for Life Sciences & State Key Laboratory of Membrane Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Jingwen Song
- Beijing Frontier Research Center for Biological Structure & Tsinghua-Peking Center for Life Sciences & State Key Laboratory of Membrane Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Li-Hua Hou
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing 100071, China
| | - Sai Li
- Beijing Frontier Research Center for Biological Structure & Tsinghua-Peking Center for Life Sciences & State Key Laboratory of Membrane Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China.
| |
Collapse
|
17
|
Dowgier G, Hobbs A, Greenwood D, Shawe-Taylor M, Stevenson-Leggett P, Bazire J, Penn R, Harvey R, Libri V, Kassiotis G, Gamblin S, Lewis NS, Williams B, Swanton C, Gandhi S, Bauer DLV, Carr EJ, Wall EC, Wu MY. Accurate evaluation of live-virus microneutralisation for SARS-CoV-2 variant JN.1 in the assessment of vaccination and therapeutics. Vaccine 2025; 54:126960. [PMID: 40056806 DOI: 10.1016/j.vaccine.2025.126960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 02/14/2025] [Accepted: 02/23/2025] [Indexed: 03/10/2025]
Abstract
Emerging SARS-CoV-2 variants require rapid assessments of pathogenicity and evasion of existing immunity to inform policy. A crucial component of these assessments is accurate estimation of serum neutralising antibody titres using cultured live virus isolates. Here, we report a comparison of culture methods for Omicron sub-variant JN.1 and the subsequent evaluation of neutralising antibody titres (nAbTs) in recipients of BNT162b2-XBB.1.5 monovalent and the ancestral/BA.4/5 containing bivalent vaccines. We compared culture of JN.1 in either Vero V1 cells or Caco-2 cells, finding culture in Vero V1 either resulted in low-titre stocks or induced crucial mutations at the Spike furin cleavage site (FCS). Using sequence-clean culture stocks generated in Caco-2 cells, we assessed serum samples from 71 healthy adults eligible for a COVID-19 vaccination given as a 5th dose booster in the UK: all participants had detectable nAbs against JN.1 prior to vaccination, with baseline/pre-existing nAbTs between both vaccine groups comparable (p = 0.240). However, nAbTs against JN.1 post-vaccination were 2.6-fold higher for recipients of the monovalent XBB.1.5 vaccine than the BA.4/5 bivalent vaccine (p < 0.001). Further, at clinically relevant concentrations the therapeutic monoclonal antibody Sotrovimab marginally maintains neutralisation of JN.1. Regular re-appraisal of methods and policy outcomes as new variants arise is required to ensure robust data are used to underpin future severity assessments and vaccine strain selection decisions.
Collapse
Affiliation(s)
- Giulia Dowgier
- COVID Surveillance Unit, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, United Kingdom; The Francis Crick Institute, 1 Midland Road, London NW1 1AT, United Kingdom
| | - Agnieszka Hobbs
- COVID Surveillance Unit, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, United Kingdom; The Francis Crick Institute, 1 Midland Road, London NW1 1AT, United Kingdom
| | - David Greenwood
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, United Kingdom; National Institute for Health Research (NIHR) University College London Hospitals (UCLH) Biomedical Research Centre and NIHR UCLH Clinical Research Facility,United Kingdom
| | - Marianne Shawe-Taylor
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, United Kingdom; National Institute for Health Research (NIHR) University College London Hospitals (UCLH) Biomedical Research Centre and NIHR UCLH Clinical Research Facility,United Kingdom
| | - Phoebe Stevenson-Leggett
- COVID Surveillance Unit, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, United Kingdom; The Francis Crick Institute, 1 Midland Road, London NW1 1AT, United Kingdom
| | - James Bazire
- COVID Surveillance Unit, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, United Kingdom; The Francis Crick Institute, 1 Midland Road, London NW1 1AT, United Kingdom
| | - Rebecca Penn
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, United Kingdom
| | - Ruth Harvey
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, United Kingdom; Worldwide Influenza Centre, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, United Kingdom
| | - Vincenzo Libri
- National Institute for Health Research (NIHR) University College London Hospitals (UCLH) Biomedical Research Centre and NIHR UCLH Clinical Research Facility,United Kingdom
| | - George Kassiotis
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, United Kingdom; Department of Infectious Disease, St Mary's Hospital, Imperial College London, London, United Kingdom
| | - Steve Gamblin
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, United Kingdom
| | - Nicola S Lewis
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, United Kingdom; Worldwide Influenza Centre, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, United Kingdom
| | - Bryan Williams
- National Institute for Health Research (NIHR) University College London Hospitals (UCLH) Biomedical Research Centre and NIHR UCLH Clinical Research Facility,United Kingdom; University College London, London, United Kingdom
| | - Charles Swanton
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, United Kingdom; University College London, London, United Kingdom
| | - Sonia Gandhi
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, United Kingdom; University College London, London, United Kingdom
| | - David L V Bauer
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, United Kingdom; Genotype-to-Phenotype 2 Consortium (G2P2-UK), United Kingdom
| | - Edward J Carr
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, United Kingdom; University College London, London, United Kingdom
| | - Emma C Wall
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, United Kingdom; National Institute for Health Research (NIHR) University College London Hospitals (UCLH) Biomedical Research Centre and NIHR UCLH Clinical Research Facility,United Kingdom; Research Department of Infection, Division of Infection and Immunity, University College London, United Kingdom
| | - Mary Y Wu
- COVID Surveillance Unit, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, United Kingdom; The Francis Crick Institute, 1 Midland Road, London NW1 1AT, United Kingdom.
| |
Collapse
|
18
|
de Melo BP, da Silva JAM, Rodrigues MA, Palmeira JDF, Amato AA, Argañaraz GA, Argañaraz ER. SARS-CoV-2 Spike Protein and Long COVID-Part 2: Understanding the Impact of Spike Protein and Cellular Receptor Interactions on the Pathophysiology of Long COVID Syndrome. Viruses 2025; 17:619. [PMID: 40431631 PMCID: PMC12115913 DOI: 10.3390/v17050619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Revised: 04/06/2025] [Accepted: 04/23/2025] [Indexed: 05/29/2025] Open
Abstract
SARS-CoV-2 infection has had a significant impact on global health through both acute illness, referred to as coronavirus disease 2019 (COVID-19), and chronic conditions (long COVID or post-acute sequelae of COVID-19, PASC). Despite substantial advancements in preventing severe COVID-19 cases through vaccination, the rise in the prevalence of long COVID syndrome and a notable degree of genomic mutation, primarily in the S protein, underscores the necessity for a deeper understanding of the underlying pathophysiological mechanisms related to the S protein of SARS-CoV-2. In this review, the latest part of this series, we investigate the potential pathophysiological molecular mechanisms triggered by the interaction between the spike protein and cellular receptors. Therefore, this review aims to provide a differential and focused view on the mechanisms potentially activated by the binding of the spike protein to canonical and non-canonical receptors for SARS-CoV-2, together with their possible interactions and effects on the pathogenesis of long COVID.
Collapse
Affiliation(s)
- Bruno Pereira de Melo
- Laboratory of Molecular Neurovirology, Department of Pharmacy, Faculty of Health Science, University of Brasília, Brasília 70910-900, DF, Brazil
| | - Jhéssica Adriane Mello da Silva
- Laboratory of Molecular Neurovirology, Department of Pharmacy, Faculty of Health Science, University of Brasília, Brasília 70910-900, DF, Brazil
| | - Mariana Alves Rodrigues
- Laboratory of Molecular Neurovirology, Department of Pharmacy, Faculty of Health Science, University of Brasília, Brasília 70910-900, DF, Brazil
| | - Julys da Fonseca Palmeira
- Laboratory of Molecular Neurovirology, Department of Pharmacy, Faculty of Health Science, University of Brasília, Brasília 70910-900, DF, Brazil
| | - Angélica Amorim Amato
- Laboratory of Molecular Pharmacology, Faculty of Health Science, University of Brasília, Brasilia 70910-900, DF, Brazil
| | - Gustavo Adolfo Argañaraz
- Laboratory of Molecular Neurovirology, Department of Pharmacy, Faculty of Health Science, University of Brasília, Brasília 70910-900, DF, Brazil
| | - Enrique Roberto Argañaraz
- Laboratory of Molecular Neurovirology, Department of Pharmacy, Faculty of Health Science, University of Brasília, Brasília 70910-900, DF, Brazil
| |
Collapse
|
19
|
Posa A. Spike protein-related proteinopathies: A focus on the neurological side of spikeopathies. Ann Anat 2025; 260:152662. [PMID: 40254264 DOI: 10.1016/j.aanat.2025.152662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Revised: 04/07/2025] [Accepted: 04/09/2025] [Indexed: 04/22/2025]
Abstract
BACKGROUND The spike protein (SP) is an outward-projecting transmembrane glycoprotein on viral surfaces. SARS-CoV-2 (Severe Acute Respiratory Syndrome Coronavirus 2), responsible for COVID-19 (Coronavirus Disease 2019), uses SP to infect cells that express angiotensin converting enzyme 2 (ACE2) on their membrane. Remarkably, SP has the ability to cross the blood-brain barrier (BBB) into the brain and cause cerebral damage through various pathomechanisms. To combat the COVID-19 pandemic, novel gene-based products have been used worldwide to induce human body cells to produce SP to stimulate the immune system. This artificial SP also has a harmful effect on the human nervous system. STUDY DESIGN Narrative review. OBJECTIVE This narrative review presents the crucial role of SP in neurological complaints after SARS-CoV-2 infection, but also of SP derived from novel gene-based anti-SARS-CoV-2 products (ASP). METHODS Literature searches using broad terms such as "SARS-CoV-2", "spike protein", "COVID-19", "COVID-19 pandemic", "vaccines", "COVID-19 vaccines", "post-vaccination syndrome", "post-COVID-19 vaccination syndrome" and "proteinopathy" were performed using PubMed. Google Scholar was used to search for topic-specific full-text keywords. CONCLUSIONS The toxic properties of SP presented in this review provide a good explanation for many of the neurological symptoms following SARS-CoV-2 infection and after injection of SP-producing ASP. Both SP entities (from infection and injection) interfere, among others, with ACE2 and act on different cells, tissues and organs. Both SPs are able to cross the BBB and can trigger acute and chronic neurological complaints. Such SP-associated pathologies (spikeopathies) are further neurological proteinopathies with thrombogenic, neurotoxic, neuroinflammatory and neurodegenerative potential for the human nervous system, particularly the central nervous system. The potential neurotoxicity of SP from ASP needs to be critically examined, as ASPs have been administered to millions of people worldwide.
Collapse
Affiliation(s)
- Andreas Posa
- University Clinics and Outpatient Clinics for Radiology, Neuroradiology and Neurology, Martin Luther University Halle-Wittenberg, Ernst-Grube-Straße 40, Halle 06120, Germany.
| |
Collapse
|
20
|
Oliveira ASF, Kearns FL, Rosenfeld MA, Casalino L, Tulli L, Berger I, Schaffitzel C, Davidson AD, Amaro RE, Mulholland AJ. Allosteric modulation by the fatty acid site in the glycosylated SARS-CoV-2 spike. eLife 2025; 13:RP97313. [PMID: 40208235 PMCID: PMC11984958 DOI: 10.7554/elife.97313] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/11/2025] Open
Abstract
The spike protein is essential to the SARS-CoV-2 virus life cycle, facilitating virus entry and mediating viral-host membrane fusion. The spike contains a fatty acid (FA) binding site between every two neighbouring receptor-binding domains. This site is coupled to key regions in the protein, but the impact of glycans on these allosteric effects has not been investigated. Using dynamical nonequilibrium molecular dynamics (D-NEMD) simulations, we explore the allosteric effects of the FA site in the fully glycosylated spike of the SARS-CoV-2 ancestral variant. Our results identify the allosteric networks connecting the FA site to functionally important regions in the protein, including the receptor-binding motif, an antigenic supersite in the N-terminal domain, the fusion peptide region, and another allosteric site known to bind heme and biliverdin. The networks identified here highlight the complexity of the allosteric modulation in this protein and reveal a striking and unexpected link between different allosteric sites. Comparison of the FA site connections from D-NEMD in the glycosylated and non-glycosylated spike revealed that glycans do not qualitatively change the internal allosteric pathways but can facilitate the transmission of the structural changes within and between subunits.
Collapse
Affiliation(s)
- A Sofia F Oliveira
- Centre for Computational Chemistry, School of Chemistry, University of BristolBristolUnited Kingdom
- School of Chemistry, University of BristolBristolUnited Kingdom
| | - Fiona L Kearns
- Department of Chemistry and Biochemistry, University of California San DiegoLa JollaUnited States
| | - Mia A Rosenfeld
- Department of Chemistry and Biochemistry, University of California San DiegoLa JollaUnited States
| | - Lorenzo Casalino
- Department of Chemistry and Biochemistry, University of California San DiegoLa JollaUnited States
| | - Lorenzo Tulli
- Centre for Computational Chemistry, School of Chemistry, University of BristolBristolUnited Kingdom
- School of Chemistry, University of BristolBristolUnited Kingdom
| | - Imre Berger
- School of Chemistry, University of BristolBristolUnited Kingdom
- School of Biochemistry, University of BristolBristolUnited Kingdom
- Max Planck Bristol Centre for Minimal Biology, School of ChemistryBristolUnited Kingdom
| | | | - Andrew D Davidson
- School of Cellular and Molecular Medicine, University of Bristol, University WalkBristolUnited Kingdom
| | - Rommie E Amaro
- Department of Molecular Biology, University of California San DiegoLa JollaUnited States
| | - Adrian J Mulholland
- Centre for Computational Chemistry, School of Chemistry, University of BristolBristolUnited Kingdom
- School of Chemistry, University of BristolBristolUnited Kingdom
| |
Collapse
|
21
|
M Bader S, Calleja DJ, Devine SM, Kuchel NW, Lu BGC, Wu X, Birkinshaw RW, Bhandari R, Loi K, Volpe R, Khakham Y, Au AE, Blackmore TR, Mackiewicz L, Dayton M, Schaefer J, Scherer L, Stock AT, Cooney JP, Schoffer K, Maluenda A, Kleeman EA, Davidson KC, Allison CC, Ebert G, Chen G, Katneni K, Klemm TA, Nachbur U, Georgy SR, Czabotar PE, Hannan AJ, Putoczki TL, Tanzer M, Pellegrini M, Lechtenberg BC, Charman SA, Call MJ, Mitchell JP, Lowes KN, Lessene G, Doerflinger M, Komander D. A novel PLpro inhibitor improves outcomes in a pre-clinical model of long COVID. Nat Commun 2025; 16:2900. [PMID: 40180914 PMCID: PMC11969009 DOI: 10.1038/s41467-025-57905-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 03/06/2025] [Indexed: 04/05/2025] Open
Abstract
The COVID-19 pandemic caused by the coronavirus SARS-CoV-2 has highlighted the vulnerability of a globally connected population to zoonotic viruses. The FDA-approved coronavirus antiviral Paxlovid targets the essential SARS-CoV-2 main protease, Mpro. Whilst effective in the acute phase of a COVID infection, Paxlovid cannot be used by all patients, can lead to viral recurrence, and does not protect against post-acute sequelae of COVID-19 (PASC), commonly known as long COVID, an emerging significant health burden that remains poorly understood and untreated. Alternative antivirals that are addressing broader patient needs are urgently required. We here report our drug discovery efforts to target PLpro, a further essential coronaviral protease, for which we report a novel chemical scaffold that targets SARS-CoV-2 PLpro with low nanomolar activity, and which exhibits activity against PLpro of other pathogenic coronaviruses. Our lead compound shows excellent in vivo efficacy in a mouse model of severe acute disease. Importantly, our mouse model recapitulates long-term pathologies matching closely those seen in PASC patients. Our lead compound offers protection against a range of PASC symptoms in this model, prevents lung pathology and reduces brain dysfunction. This provides proof-of-principle that PLpro inhibition may have clinical relevance for PASC prevention and treatment going forward.
Collapse
Affiliation(s)
- Stefanie M Bader
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Australia
| | - Dale J Calleja
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Australia
| | - Shane M Devine
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.
- Department of Medical Biology, University of Melbourne, Melbourne, Australia.
| | - Nathan W Kuchel
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
| | - Bernadine G C Lu
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Australia
| | - Xinyu Wu
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Australia
| | - Richard W Birkinshaw
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Australia
| | - Reet Bhandari
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Australia
| | - Katie Loi
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Australia
| | - Rohan Volpe
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Australia
| | - Yelena Khakham
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
| | - Amanda E Au
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Australia
| | - Timothy R Blackmore
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Australia
| | - Liana Mackiewicz
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
| | - Merle Dayton
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
| | - Jan Schaefer
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Australia
| | - Lena Scherer
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
| | - Angus T Stock
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Australia
| | - James P Cooney
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Australia
| | - Kael Schoffer
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Australia
| | - Ana Maluenda
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
| | - Elizabeth A Kleeman
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Kathryn C Davidson
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Australia
| | - Cody C Allison
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Australia
| | - Gregor Ebert
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Australia
| | - Gong Chen
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Kasiram Katneni
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Theresa A Klemm
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Australia
| | - Ueli Nachbur
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Australia
| | - Smitha Rose Georgy
- Anatomic Pathology - Veterinary Biosciences, Melbourne Veterinary School, University of Melbourne, Werribee, VIC, Australia
| | - Peter E Czabotar
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Australia
| | - Anthony J Hannan
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
- Department of Anatomy and Physiology, University of Melbourne, Parkville, VIC, Australia
| | - Tracy L Putoczki
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Australia
- Department of Surgery, University of Melbourne, Melbourne, Australia
| | - Maria Tanzer
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Australia
| | - Marc Pellegrini
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Australia
- Centenary Institute of Cancer Medicine and Cell Biology, Camperdown, NSW, Australia
| | - Bernhard C Lechtenberg
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Australia
| | - Susan A Charman
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Melissa J Call
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Australia
| | - Jeffrey P Mitchell
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Australia
| | - Kym N Lowes
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Australia
| | - Guillaume Lessene
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.
- Department of Medical Biology, University of Melbourne, Melbourne, Australia.
- Department of Biochemistry and Pharmacology, University of Melbourne, Melbourne, Australia.
| | - Marcel Doerflinger
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.
- Department of Medical Biology, University of Melbourne, Melbourne, Australia.
| | - David Komander
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.
- Department of Medical Biology, University of Melbourne, Melbourne, Australia.
| |
Collapse
|
22
|
Dhawan M, Thakur N, Sharma M, Rabaan AA. The comprehensive insights into the B-cells-mediated immune response against COVID-19 infection amid the ongoing evolution of SARS-CoV-2. Biomed Pharmacother 2025; 185:117936. [PMID: 40056829 DOI: 10.1016/j.biopha.2025.117936] [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: 09/16/2024] [Revised: 02/08/2025] [Accepted: 02/20/2025] [Indexed: 03/10/2025] Open
Abstract
The antibody-mediated immune response is crucial for the development of protective immunity against SARS-CoV-2, the virus responsible for the COVID-19 pandemic. Understanding the interaction between SARS-CoV-2 and the immune system is critical because new variants emerge as a result of the virus's ongoing evolution. Understanding the function of B cells in the SARS-CoV-2 infection process is critical for developing effective and long-lasting vaccines against this virus. Triggered by the innate immune response, B cells transform into memory B cells (MBCs). It is fascinating to observe how MBCs provide enduring immune defence, not only eradicating the infection but also safeguarding against future reinfection. If there is a lack of B cell activation or if the B cells are not functioning properly, it can lead to a serious manifestation of the disease and make immunisation less effective. Individuals with disruptions in the B cells have shown increased production of cytokines and chemokines, resulting in a poor prognosis for the disease. Therefore, we have developed an updated review article to gain insight into the involvement of B cells in SARS-CoV-2 infection. The discussion has covered the generation, functioning, and dynamics of neutralising antibodies (nAbs). Furthermore, we have emphasised immunotherapeutics that rely on nAbs.
Collapse
Affiliation(s)
- Manish Dhawan
- Department of Microbiology, Punjab Agricultural University, Ludhiana, Punjab 141004, India; Trafford College, Altrincham, Altrincham, Manchester WA14 5PQ, UK.
| | - Nanamika Thakur
- University Institute of Biotechnology, Department of Biotechnology, Chandigarh University, Mohali 140413, India
| | - Manish Sharma
- University Institute of Biotechnology, Department of Biotechnology, Chandigarh University, Mohali 140413, India
| | - Ali A Rabaan
- Research Center, Dr. Sulaiman Alhabib Medical Group, Riyadh 13328, Saudi Arabia; Molecular Diagnostic Laboratory, Johns Hopkins Aramco Healthcare, Dhahran 31311, Saudi Arabia; Department of Public Health and Nutrition, The University of Haripur, Haripur 22610, Pakistan.
| |
Collapse
|
23
|
Matsuzawa Y, Tsujita K. Impact of SARS-CoV-2 variants on viral infectivity and the role of the renin-angiotensin-aldosterone system. Hypertens Res 2025; 48:1636-1638. [PMID: 39871005 DOI: 10.1038/s41440-025-02115-0] [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: 11/28/2024] [Revised: 12/29/2024] [Accepted: 12/30/2024] [Indexed: 01/29/2025]
Abstract
Balance between Protective vs. Exacerbating Effects of ACEIs and ARBs in Omicron Variant Infections. The spike protein on the surface of the Omicron variant has a high affinity for ACE2, making it more prone to enter cells and induce ACE2 downregulation. Therefore, the effects of ACEIs and ARBs on Omicron variant infections may differ from those of the previous variants. The current study demonstrates that ACEIs and ARBs do not aggravate or prolong COVID-19 due to Omicron variant infections.
Collapse
Affiliation(s)
- Yasushi Matsuzawa
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan.
| | - Kenichi Tsujita
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
24
|
Dong Y, Xie Z, Xu L. Receptors and host factors: key players in human metapneumovirus infection. Front Cell Infect Microbiol 2025; 15:1557880. [PMID: 40235933 PMCID: PMC11996802 DOI: 10.3389/fcimb.2025.1557880] [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: 01/09/2025] [Accepted: 03/13/2025] [Indexed: 04/17/2025] Open
Abstract
Human metapneumovirus (hMPV) is a significant global pathogen that causes acute respiratory tract infections, especially in infants, young children, the elderly, and immunocompromised individuals. Despite its increasing prevalence, there are currently no vaccines or effective treatments available for hMPV. The pathogenesis of hMPV infection is a complex process involving a multitude of host factors and viral receptors. These interactions determine the virus ability to enter host cells, replicate, and evade the immune response. This review is the first to provide a comprehensive overview of the current understanding of host-virus interactions in hMPV pathogenesis. By elucidating these mechanisms, we can identify potential targets for antiviral drugs and improve the management of hMPV infections.
Collapse
Affiliation(s)
- Yingdong Dong
- Beijing Key Laboratory of Core Technologies for the Prevention and Treatment of Emerging Infectious Diseases in Children, National Clinical Research Center for Respiratory Diseases, National Key Discipline of Pediatrics (Capital Medical University), Beijing Research Center for Respiratory Infectious Diseases, Beijing Pediatric Research Institute, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
- Research Unit of Critical Infection in Children, Chinese Academy of Medical Sciences, Beijing, China
| | - Zhengde Xie
- Beijing Key Laboratory of Core Technologies for the Prevention and Treatment of Emerging Infectious Diseases in Children, National Clinical Research Center for Respiratory Diseases, National Key Discipline of Pediatrics (Capital Medical University), Beijing Research Center for Respiratory Infectious Diseases, Beijing Pediatric Research Institute, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
- Research Unit of Critical Infection in Children, Chinese Academy of Medical Sciences, Beijing, China
| | - Lili Xu
- Beijing Key Laboratory of Core Technologies for the Prevention and Treatment of Emerging Infectious Diseases in Children, National Clinical Research Center for Respiratory Diseases, National Key Discipline of Pediatrics (Capital Medical University), Beijing Research Center for Respiratory Infectious Diseases, Beijing Pediatric Research Institute, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
- Research Unit of Critical Infection in Children, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
25
|
Letscher H, Guilligay D, Effantin G, Amen A, Sulbaran G, Burger JA, Bossevot L, Junges L, Leonec M, Morin J, Van Tilbeurgh M, Hérate C, Gallouët AS, Relouzat F, van der Werf S, Cavarelli M, Dereuddre-Bosquet N, van Gils MJ, Sanders RW, Poignard P, Le Grand R, Weissenhorn W. RBD-depleted SARS-CoV-2 spike generates protective immunity in cynomolgus macaques. NPJ Vaccines 2025; 10:63. [PMID: 40159504 PMCID: PMC11955555 DOI: 10.1038/s41541-025-01113-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 03/17/2025] [Indexed: 04/02/2025] Open
Abstract
The SARS-CoV-2 pandemic revealed the rapid evolution of circulating strains. This led to new variants carrying mostly mutations within the receptor binding domain, which is immunodominant upon immunization and infection. In order to steer the immune response away from RBD epitopes to more conserved domains, we generated S glycoprotein trimers without RBD and stabilized them by formaldehyde cross-linking. The cryoEM structure demonstrated that SΔRBD folds into the native prefusion conformation, stabilized by one specific cross-link between S2 protomers. SΔRBD was coated onto lipid vesicles, to produce synthetic virus-like particles, SΔRBD-LV, which were utilized in a heterologous prime-boost strategy. Immunization of cynomolgus macaques either three times with the mRNA Comirnaty vaccine or two times followed by SΔRBD-LV showed that the SΔRBD-LV boost induced similar antibody titers and neutralization of different variants, including omicron. Upon challenge with omicron XBB.3, both the Comirnaty only and Comirnaty/SΔRBD-LV vaccination schemes conferred similar overall protection from infection for both the Comirnaty only and Comirnaty/SΔRBD-LV vaccination schemes. However, the SΔRBD-LV boost indicated better protection against lung infection than the Comirnaty strategy alone. Together our findings indicate that SΔRBD is highly immunogenic and provides improved protection compared to a third mRNA boost indicative of superior antibody-based protection.
Collapse
Affiliation(s)
- Hélène Letscher
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT/UMR-S 1184), Fontenay-aux-Roses & Le Kremlin-Bicêtre, Paris, France.
| | - Delphine Guilligay
- Univ. Grenoble Alpes, CEA, CNRS, Institut de Biologie Structurale (IBS), Grenoble, France
| | - Gregory Effantin
- Univ. Grenoble Alpes, CEA, CNRS, Institut de Biologie Structurale (IBS), Grenoble, France
| | - Axelle Amen
- Univ. Grenoble Alpes, CEA, CNRS, Institut de Biologie Structurale (IBS), Grenoble, France
- CHU Grenoble Alpes, Grenoble, France
| | - Guidenn Sulbaran
- Univ. Grenoble Alpes, CEA, CNRS, Institut de Biologie Structurale (IBS), Grenoble, France
| | - Judith A Burger
- University of Amsterdam, Department of Medical Microbiology and Infection Prevention, Amsterdam University Medical Centers, Location AMC, Amsterdam, The Netherlands
| | - Laetitia Bossevot
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT/UMR-S 1184), Fontenay-aux-Roses & Le Kremlin-Bicêtre, Paris, France
| | - Laura Junges
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT/UMR-S 1184), Fontenay-aux-Roses & Le Kremlin-Bicêtre, Paris, France
| | - Marco Leonec
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT/UMR-S 1184), Fontenay-aux-Roses & Le Kremlin-Bicêtre, Paris, France
| | - Julie Morin
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT/UMR-S 1184), Fontenay-aux-Roses & Le Kremlin-Bicêtre, Paris, France
| | - Matthieu Van Tilbeurgh
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT/UMR-S 1184), Fontenay-aux-Roses & Le Kremlin-Bicêtre, Paris, France
| | - Cécile Hérate
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT/UMR-S 1184), Fontenay-aux-Roses & Le Kremlin-Bicêtre, Paris, France
| | - Anne-Sophie Gallouët
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT/UMR-S 1184), Fontenay-aux-Roses & Le Kremlin-Bicêtre, Paris, France
| | - Francis Relouzat
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT/UMR-S 1184), Fontenay-aux-Roses & Le Kremlin-Bicêtre, Paris, France
| | - Sylvie van der Werf
- Institut Pasteur, Molecular Genetics of RNA Viruses, Department of Virology, CNRS UMR 3569, Université de Paris, Paris, France
- Institut Pasteur, National Reference Center for Respiratory Viruses, Paris, France
| | - Mariangela Cavarelli
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT/UMR-S 1184), Fontenay-aux-Roses & Le Kremlin-Bicêtre, Paris, France
| | - Nathalie Dereuddre-Bosquet
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT/UMR-S 1184), Fontenay-aux-Roses & Le Kremlin-Bicêtre, Paris, France
| | - Marit J van Gils
- University of Amsterdam, Department of Medical Microbiology and Infection Prevention, Amsterdam University Medical Centers, Location AMC, Amsterdam, The Netherlands
| | - Rogier W Sanders
- University of Amsterdam, Department of Medical Microbiology and Infection Prevention, Amsterdam University Medical Centers, Location AMC, Amsterdam, The Netherlands
- Weill Medical College of Cornell University, Department of Microbiology and Immunology, New York, NY, USA
| | - Pascal Poignard
- Univ. Grenoble Alpes, CEA, CNRS, Institut de Biologie Structurale (IBS), Grenoble, France
- CHU Grenoble Alpes, Grenoble, France
| | - Roger Le Grand
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT/UMR-S 1184), Fontenay-aux-Roses & Le Kremlin-Bicêtre, Paris, France.
| | - Winfried Weissenhorn
- Univ. Grenoble Alpes, CEA, CNRS, Institut de Biologie Structurale (IBS), Grenoble, France.
| |
Collapse
|
26
|
Liu Y, Wang K, Wang W, Kashyap S, Jih J, Imani A, Hsiai T, Zhou ZH. Demonstration and structural basis of a therapeutic DNA aptamer for SARS-CoV-2 spike protein detection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.14.643408. [PMID: 40166151 PMCID: PMC11957045 DOI: 10.1101/2025.03.14.643408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
At the onset of the COVID-19 pandemic, the absence of a rapid and highly specific diagnostic method for the SARS-CoV-2 virus led to significant delays in detection, adversely affecting clinical outcomes. This shortfall highlights the urgent need for adaptable, scalable, and reusable diagnostic technologies to improve future pandemic responses. To address this challenge, we developed a renewable electrochemical impedance biosensor device employing a synthetic nucleotide-based therapeutic aptamer (termed 'aptasensor') targeting the SARS-CoV-2 spike (S) protein receptor-binding domain (RBD). We demonstrate that our aptasensor can detect the Omicron BA.2 S protein within one hour and possesses concentration-dependent sensitivity at biologically relevant levels. Notably, the aptasensor is reusable after regeneration by a simple pH 2 buffer treatment. Aptamer binding to the S protein was confirmed by immunogold labeling and visualization by negative-stain electron microscopy. We used cryogenic electron microscopy (cryo-EM) to resolve high-resolution maps of the S protein in both the open and closed conformations and characterized aptamer binding to the up RBD in the open conformation. Taken together, these results establish the versatility and scalability of aptamer-based biosensors, presenting them as a potential transformative diagnostic platform for emerging pathogens. This combination of rapid detection, specificity, and renewable capabilities in a single diagnostic solution marks a significant advance in pandemic preparedness.
Collapse
Affiliation(s)
- Yujun Liu
- California NanoSystems Institute, University of California at Los Angeles, Los Angeles, 90095, California, USA
- Department of Microbiology, Immunology and Molecular Genetics, University of California at Los Angeles, Los Angeles, 90095, California, USA
| | - Kaidong Wang
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California 90095, USA
- Department of Medicine, Greater Los Angeles VA Healthcare System, Los Angeles, California 90073, USA
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Science, University of California Los Angeles, Los Angeles, California 90095, USA
| | - Weiguang Wang
- California NanoSystems Institute, University of California at Los Angeles, Los Angeles, 90095, California, USA
- Department of Microbiology, Immunology and Molecular Genetics, University of California at Los Angeles, Los Angeles, 90095, California, USA
| | - Saarang Kashyap
- California NanoSystems Institute, University of California at Los Angeles, Los Angeles, 90095, California, USA
- Department of Microbiology, Immunology and Molecular Genetics, University of California at Los Angeles, Los Angeles, 90095, California, USA
| | - Jonathan Jih
- California NanoSystems Institute, University of California at Los Angeles, Los Angeles, 90095, California, USA
- Department of Microbiology, Immunology and Molecular Genetics, University of California at Los Angeles, Los Angeles, 90095, California, USA
- Molecular Biology Institute, University of California, Los Angeles (UCLA); Los Angeles, CA 90095, USA
| | - Anthony Imani
- California NanoSystems Institute, University of California at Los Angeles, Los Angeles, 90095, California, USA
- Department of Microbiology, Immunology and Molecular Genetics, University of California at Los Angeles, Los Angeles, 90095, California, USA
| | - Tzung Hsiai
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California 90095, USA
- Department of Medicine, Greater Los Angeles VA Healthcare System, Los Angeles, California 90073, USA
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Science, University of California Los Angeles, Los Angeles, California 90095, USA
| | - Z. Hong Zhou
- California NanoSystems Institute, University of California at Los Angeles, Los Angeles, 90095, California, USA
- Department of Microbiology, Immunology and Molecular Genetics, University of California at Los Angeles, Los Angeles, 90095, California, USA
- Molecular Biology Institute, University of California, Los Angeles (UCLA); Los Angeles, CA 90095, USA
| |
Collapse
|
27
|
Mani N, Suresh R, Chakraborty S. Cleaved vs. Uncleaved: How Furin Cleavage Reshapes the Conformational Landscape of SARS-CoV-2 Spike. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.12.642945. [PMID: 40161653 PMCID: PMC11952566 DOI: 10.1101/2025.03.12.642945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
The SARS-CoV-2 Spike protein is the primary target for vaccine design, with immunogens typically engineered to enhance stability by introducing proline mutations (2P) and mutating or deleting the Furin Cleavage Site (FCS). While these modifications improve structural integrity, studies suggest that furin cleavage can play a functional role in Spike protein dynamics, potentially enhancing ACE2 receptor binding. However, the impact of this cleavage on the unbound form of the Spike protein remains unclear. In this study, we use extensive all-atom molecular dynamics (MD) simulations to compare the structural and dynamic properties of cleaved and uncleaved Spike proteins in their pre-fusion, unbound state. Our results show that Furin cleavage significantly alters allosteric communication within the protein, increasing correlated motions between the Receptor Binding Domain (RBD) and N-terminal Domain (NTD), which may facilitate receptor engagement. Principal Component Analysis (PCA) reveals that the cleaved and uncleaved Spike proteins sample distinct conformational landscapes, with the cleaved form displaying enhanced flexibility and a broader range of RBD tilt angles. Additionally, Furin cleavage primes the S2 subunit by expanding the central helix, potentially influencing the transition to the post-fusion state. Glycan clustering patterns further suggest an adaptive structural response to cleavage, particularly in the NTD and RBD regions. These findings highlight the potential functional consequences of FCS deletion in immunogen design and underscore the importance of considering the native cleavage state in vaccine and therapeutic development.
Collapse
Affiliation(s)
- Natesan Mani
- Department of Chemical Engineering, Northeastern University, Boston, MA 02115; Department of Chemistry and Chemical Biology, Northeastern University, Boston, MA 02115
| | - Raghavendran Suresh
- Department of Chemical Engineering, Northeastern University, Boston, MA 02115; Department of Chemistry and Chemical Biology, Northeastern University, Boston, MA 02115
| | - Srirupa Chakraborty
- Department of Chemical Engineering, Northeastern University, Boston, MA 02115; Department of Chemistry and Chemical Biology, Northeastern University, Boston, MA 02115
| |
Collapse
|
28
|
Banerjee T, Frazier C, Koti N, Yates P, Bowie E, Liermann M, Johnson D, Willis SH, Santra S. Development of Receptor-Integrated Magnetically Labeled Liposomes for Investigating SARS-CoV-2 Fusion Interactions. Anal Chem 2025; 97:4490-4498. [PMID: 39925203 PMCID: PMC11883728 DOI: 10.1021/acs.analchem.4c05966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 01/16/2025] [Accepted: 02/04/2025] [Indexed: 02/11/2025]
Abstract
The impacts of highly pathogenic enveloped viruses, such as SARS-CoV-2, have turned scientific inquiry toward the fusion mechanisms responsible for viral pathogenesis and to seek cost-effective and adaptable strategies to mitigate future outbreaks. Current approaches for studying SARS-CoV-2 fusion include computational studies, pan-coronavirus viral inhibitors, and modified peptides and lipopeptides, along with various nanotechniques. Although these methodologies have illuminated the fusion mechanisms, they possess key limitations that prevent their widespread utility in outbreaks, including high financial or instrumental costs, operational proficiency, cytotoxicity, or viral specificity. This work measures changes in spin-spin T2 magnetic (transverse) relaxation times using a benchtop NMR instrument and introduces a bioanalytical approach to quickly quantify fusion interactions between the SARS-CoV-2 spike protein and liposome-coated iron oxide nanosensors (LIONs). Additionally, this study modifies the LION platform by appending the angiotensin-converting enzyme (ACE2) receptor, thereby creating LIONs-ACE2 that mimics the ACE2 host cell receptor targeted by SARS-CoV-2. Furthermore, SARS-CoV-2 fusion to other receptors reported to be involved is also examined. Environmental factors impacting fusion, such as calcium ion concentration, cholesterol composition, pH, neutralizing antibodies, and lower temperature, are investigated. Finally, molecular dynamics (MD) simulation studies reveal that the receptor binding domain (RBD) of the spike protein interacts more favorably with ACE2 than the lipid bilayer in the opened conformation, yet the closed conformation of RBD interacts with the bilayer with a similar energy as with ACE2. These findings reveal how the LION platform offers a customizable, fast-acting, inexpensive, and accessible mechanism for examining the fusion process of SARS-CoV-2 and other enveloped viruses.
Collapse
Affiliation(s)
- Tuhina Banerjee
- Department
of Chemistry and Biochemistry, Missouri
State University, 901 S. National Avenue, Springfield, Missouri 65897, United States of America
| | - Clayton Frazier
- Department
of Chemistry and Biochemistry, Missouri
State University, 901 S. National Avenue, Springfield, Missouri 65897, United States of America
| | - Neelima Koti
- Department
of Chemistry and Biochemistry, Missouri
State University, 901 S. National Avenue, Springfield, Missouri 65897, United States of America
| | - Paris Yates
- Department
of Chemistry and Biochemistry, Missouri
State University, 901 S. National Avenue, Springfield, Missouri 65897, United States of America
| | - Elizabeth Bowie
- Department
of Chemistry and Biochemistry, Missouri
State University, 901 S. National Avenue, Springfield, Missouri 65897, United States of America
| | - Megan Liermann
- Department
of Chemistry and Biochemistry, Missouri
State University, 901 S. National Avenue, Springfield, Missouri 65897, United States of America
| | - David Johnson
- Computational
Chemical Biology Core, University of Kansas, 2034 Becker Drive, Lawrence, Kansas 66018, United States of America
| | - Sharon H Willis
- Integral
Molecular Incorporation, One uCity Square 25 N. 38th Street, Suite 800, Philadelphia, Pennsylvania 19104, United States of America
| | - Santimukul Santra
- Department
of Chemistry and Biochemistry, Missouri
State University, 901 S. National Avenue, Springfield, Missouri 65897, United States of America
| |
Collapse
|
29
|
Xiao W, Li Z, Chen C, Shi Y, Fang P, Xiao S, Fang L. Revisiting the roles of trypsin in the productive infection of porcine deltacoronavirus in porcine-derived cells. Virology 2025; 604:110453. [PMID: 39961260 DOI: 10.1016/j.virol.2025.110453] [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: 11/29/2024] [Revised: 01/25/2025] [Accepted: 02/12/2025] [Indexed: 05/09/2025]
Abstract
Porcine deltacoronavirus (PDCoV) is an emerging enteric coronavirus with the potential for interspecies transmission. Trypsin has been shown to play a positive role in the isolation and multiplication of PDCoV in vitro, however, the functions of trypsin during PDCoV replication cycle remain controversial. In this study, we revisited the roles of trypsin for PDCoV infection by utilizing two kinds of PDCoV, PDCoVT+ and PDCoVT-, which were prepared in the presence or absence of trypsin, respectively. We found that PDCoVT+ was able to continuously proliferate in the medium containing trypsin, achieving a higher titer as the infection progress in LLC-PK1 and other tested porcine-derived cells. However, its replication was only transiently improved at 12 hours post-infection, and lower viral titers were observed under trypsin-free culture conditions. Furthermore, the trypsin-mediated enhancement of viral replication could be inhibited by trypsin inhibitor SBTI, suggesting that the second-round viral reproduction of PDCoVT+ might be impeded without trypsin. We further investigated the replication dynamics of PDCoVT- in LLC-PK1 cells in the presence or absence of trypsin. The results indicated that PDCoVT- generated lower viral titers under trypsin-free culture conditions, while the addition of trypsin reverted the infectivity of PDCoVT-. Additionally, we demonstrated that trypsin cleaved the PDCoV spike protein, activating viral attachment and internalization. Moreover, trypsin promoted viral replication and release, accelerating PDCoV maturation and facilitating second-round infection. Taken together, this study systematically revaluated and emphasized an essential role of trypsin in PDCoV infection, providing mechanistic insights into the productive infection of PDCoV in porcine-derived cells.
Collapse
Affiliation(s)
- Wenwen Xiao
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, China
| | - Zhuang Li
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, China
| | - Chaoqun Chen
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, China
| | - Yuting Shi
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, China
| | - Puxian Fang
- College of Veterinary Medicine, Shandong Agricultural University, Tai'an, 271018, China
| | - Shaobo Xiao
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, China
| | - Liurong Fang
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, China.
| |
Collapse
|
30
|
Liu S, Liu P, Lu Q, Shen Y, Zhang L, Liang Z, Yu Y, Huang W, Wang Y. The Compensatory Effect of S375F on S371F Is Vital for Maintaining the Infectivity of SARS-CoV-2 Omicron Variants. J Med Virol 2025; 97:e70242. [PMID: 40062404 PMCID: PMC11891949 DOI: 10.1002/jmv.70242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 01/14/2025] [Accepted: 02/06/2025] [Indexed: 05/13/2025]
Abstract
The emergence of Omicron variants dramatically changed the transmission rate and infection characteristics compared to previously prevalent strains, primarily due to spike protein mutations. However, the impact of individual mutations remained unclear. Here, we used virus-like particle (VLP) pseudotyped to investigate the functional contributions by 12 common mutations in the spike protein. We found that the S371F mutation in the receptor binding domain (RBD) of spike protein led to a 5- and 10-fold decrease of ACE2 utilization efficiency and viral infectivity, respectively, accompanied by a 5- to 11-fold reduction of neutralization sensitivity to monoclonal antibodies. However, the S375F mutation in the RBD had a compensatory effect, rescuing the infectivity of the S371F Omicron variant. Based on molecular dynamics simulations, we proposed a "tug of war" model to explain this compensation phenomenon. These results provide a comprehensive and dynamic perspective on the evolution of this important pandemic virus.
Collapse
Affiliation(s)
- Shuo Liu
- Changping LaboratoryBeijingChina
- Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Pan Liu
- CAS Key Laboratory of Infection and Immunity, National Laboratory of Macromolecules, Institute of Biophysics, Chinese Academy of SciencesBeijingChina
| | - Qiong Lu
- Division of HIV/AIDS and Sexually Transmitted Virus VaccinesInstitute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC)BeijingChina
| | - Yanru Shen
- Division of HIV/AIDS and Sexually Transmitted Virus VaccinesInstitute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC)BeijingChina
| | - Li Zhang
- Division of HIV/AIDS and Sexually Transmitted Virus VaccinesInstitute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC)BeijingChina
| | - Ziteng Liang
- Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- Division of HIV/AIDS and Sexually Transmitted Virus VaccinesInstitute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC)BeijingChina
| | | | - Weijin Huang
- Division of HIV/AIDS and Sexually Transmitted Virus VaccinesInstitute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC)BeijingChina
| | - Youchun Wang
- Changping LaboratoryBeijingChina
- Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| |
Collapse
|
31
|
Verhulst E, De Bruyn M, Berckmans P, Sim Y, Augustyns K, Pintelon I, Berg M, Van Wielendaele P, Lambeir A, Sterckx YG, Nelissen I, De Meester I. Human Transmembrane Serine Protease 2 (TMPRSS2) on Human Seminal Fluid Extracellular Vesicles Is Proteolytically Active. J Extracell Vesicles 2025; 14:e70061. [PMID: 40091430 PMCID: PMC11911546 DOI: 10.1002/jev2.70061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 02/03/2025] [Accepted: 02/12/2025] [Indexed: 03/19/2025] Open
Abstract
Human transmembrane serine protease 2 (TMPRSS2) has garnered substantial interest due to its clinical significance in various pathologies, notably its pivotal role in viral entry into host cells. The development of effective strategies to target TMPRSS2 is a current area of intense research and necessitates a consistent source of active TMPRSS2 with sufficient stability. Here, we comprehensively characterised human seminal-fluid extracellular vesicles (SF-EVs, also referred to as prostasomes), bearing a native source of surface-exposed, enzymatically active TMPRSS2 as demonstrated by high-sensitivity flow cytometry and a fluorometric activity assay. Additionally, we recombinantly produced human TMPRSS2 ectodomain in mammalian cells adopting a directed activation strategy. We observed comparable catalytic parameters and inhibition characteristics for both native SF-EV-associated and recombinant TMPRSS2 when exposed to serine protease inhibitor Nafamostat mesylate. Leveraging these findings, we developed a robust in vitro biochemical assay based on these SF-EVs for the screening of TMPRSS2-targeting compounds. Our results will accelerate the discovery and advancement of efficacious therapeutic approaches targeting TMPRSS2 and propel further exploration into the biological role of SF-EV-associated active TMPRSS2.
Collapse
Affiliation(s)
- Emile Verhulst
- Laboratory of Medical Biochemistry, Faculty of Pharmaceutical, Biomedical and Veterinary SciencesUniversity of AntwerpWilrijkBelgium
| | - Michelle De Bruyn
- Laboratory of Medical Biochemistry, Faculty of Pharmaceutical, Biomedical and Veterinary SciencesUniversity of AntwerpWilrijkBelgium
| | | | - Yani Sim
- Laboratory of Medical Biochemistry, Faculty of Pharmaceutical, Biomedical and Veterinary SciencesUniversity of AntwerpWilrijkBelgium
| | - Koen Augustyns
- Laboratory of Medicinal Chemistry, Faculty of Pharmaceutical, Biomedical and Veterinary SciencesUniversity of AntwerpWilrijkBelgium
- Infla‐Med Centre of ExcellenceUniversity of AntwerpWilrijkBelgium
| | - Isabel Pintelon
- Laboratory of Cell Biology and Histology, Faculty of Pharmaceutical, Biomedical and Veterinary SciencesUniversity of AntwerpWilrijkBelgium
- Antwerp Centre for Advanced Microscopy (ACAM), Faculty of Pharmaceutical, Biomedical and Veterinary SciencesUniversity of AntwerpWilrijkBelgium
| | - Maya Berg
- Infla‐Med Centre of ExcellenceUniversity of AntwerpWilrijkBelgium
| | - Pieter Van Wielendaele
- Laboratory of Medical Biochemistry, Faculty of Pharmaceutical, Biomedical and Veterinary SciencesUniversity of AntwerpWilrijkBelgium
| | - Anne‐Marie Lambeir
- Laboratory of Medical Biochemistry, Faculty of Pharmaceutical, Biomedical and Veterinary SciencesUniversity of AntwerpWilrijkBelgium
| | - Yann G.‐J. Sterckx
- Laboratory of Medical Biochemistry, Faculty of Pharmaceutical, Biomedical and Veterinary SciencesUniversity of AntwerpWilrijkBelgium
| | - Inge Nelissen
- Health UnitFlemish Institute for Technological ResearchMolBelgium
| | - Ingrid De Meester
- Laboratory of Medical Biochemistry, Faculty of Pharmaceutical, Biomedical and Veterinary SciencesUniversity of AntwerpWilrijkBelgium
- Infla‐Med Centre of ExcellenceUniversity of AntwerpWilrijkBelgium
| |
Collapse
|
32
|
Akıl C, Xu J, Shen J, Zhang P. Unveiling the Complete Spectrum of SARS-CoV-2 Fusion Stages by In Situ Cryo-ET. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.25.640151. [PMID: 40060467 PMCID: PMC11888396 DOI: 10.1101/2025.02.25.640151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/15/2025]
Abstract
SARS-CoV-2 entry into host cells is mediated by the spike protein, which drives membrane fusion. While cryo-EM has revealed stable prefusion and postfusion conformations of the spike, the transient intermediate states during the fusion process have remained poorly understood. Here, we designed a near-native viral fusion system that recapitulates SARS-CoV-2 entry and used cryo-electron tomography (cryo-ET) to capture fusion intermediates leading to complete fusion. The spike protein undergoes extensive structural rearrangements, progressing through extended, partially folded, and fully folded intermediates prior to fusion-pore formation, a process that is dependent on protease cleavage and inhibited by the WS6 S2 antibody. Upon interaction with ACE2 receptor dimer, spikes cluster at membrane interfaces and following S2' cleavage concurrently transition to postfusion conformations encircling the hemifusion and pre-fusion pores in a distinct conical arrangement. Subtomogram averaging revealed that the WS6 S2 antibody binds to the spike's stem-helix, crosslinks and clusters prefusion spikes and inhibits refolding of fusion intermediates. These findings elucidate the complete process of spike-mediated fusion and SARS-CoV-2 entry, highlighting the neutralizing mechanism of S2-targeting antibodies.
Collapse
Affiliation(s)
- Caner Akıl
- Chinese Academy of Medical Sciences Oxford Institute, University of Oxford, Oxford, OX3 7BN, UK
- Division of Structural Biology, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7BN, UK
| | - Jialu Xu
- Division of Structural Biology, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7BN, UK
| | - Juan Shen
- Division of Structural Biology, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7BN, UK
| | - Peijun Zhang
- Chinese Academy of Medical Sciences Oxford Institute, University of Oxford, Oxford, OX3 7BN, UK
- Division of Structural Biology, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7BN, UK
- Diamond Light Source, Harwell Science and Innovation Campus, Didcot, OX11 0DE, UK
| |
Collapse
|
33
|
Heffner AL, Rouault TA. A Comparison of Conserved Features in the Human Coronavirus Family Shows That Studies of Viruses Less Pathogenic than SARS-CoV-2, Such as HCoV-OC43, Are Good Model Systems for Elucidating Basic Mechanisms of Infection and Replication in Standard Laboratories. Viruses 2025; 17:256. [PMID: 40007010 PMCID: PMC11860170 DOI: 10.3390/v17020256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 02/05/2025] [Accepted: 02/06/2025] [Indexed: 02/27/2025] Open
Abstract
In 2021, at the height of the COVID-19 pandemic, coronavirus research spiked, with over 83,000 original research articles related to the word "coronavirus" added to the online resource PubMed. Just 2 years later, in 2023, only 30,900 original research articles related to the word "coronavirus" were added. While, irrefutably, the funding of coronavirus research drastically decreased, a possible explanation for the decrease in interest in coronavirus research is that projects on SARS-CoV-2, the causative agent of COVID-19, halted due to the challenge of establishing a good cellular or animal model system. Most laboratories do not have the capabilities to culture SARS-CoV-2 'in house' as this requires a Biosafety Level (BSL) 3 laboratory. Until recently, BSL 2 laboratory research on endemic coronaviruses was arduous due to the low cytopathic effect in isolated cell culture infection models and the lack of means to quantify viral loads. The purpose of this review article is to compare the human coronaviruses and provide an assessment of the latest techniques that use the endemic coronaviruses-HCoV-229E, HCoV-OC43, HCoV-NL63, and HCoV-HKU1-as lower-biosafety-risk models for the more pathogenic coronaviruses-SARS-CoV-2, SARS-CoV, and MERS-CoV.
Collapse
Affiliation(s)
- Audrey L. Heffner
- Molecular Medicine Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD 20892, USA
- Department of Biology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Tracey A. Rouault
- Molecular Medicine Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD 20892, USA
| |
Collapse
|
34
|
Suzuki Okutani M, Okamura S, Gis T, Sasaki H, Lee S, Kashiwabara A, Goto S, Matsumoto M, Yamawaki M, Miyazaki T, Nakagawa T, Ikawa M, Kamitani W, Takekawa S, Yamanishi K, Ebina H. Immunogenicity and safety of a live-attenuated SARS-CoV-2 vaccine candidate based on multiple attenuation mechanisms. eLife 2025; 13:RP97532. [PMID: 39932490 PMCID: PMC11813227 DOI: 10.7554/elife.97532] [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] [Indexed: 02/13/2025] Open
Abstract
mRNA vaccines against SARS-CoV-2 were rapidly developed and were effective during the pandemic. However, some limitations remain to be resolved, such as the short-lived induced immune response and certain adverse effects. Therefore, there is an urgent need to develop new vaccines that address these issues. While live-attenuated vaccines are a highly effective modality, they pose a risk of adverse effects, including virulence reversion. In the current study, we constructed a live-attenuated vaccine candidate, BK2102, combining naturally occurring virulence-attenuating mutations in the NSP14, NSP1, spike, and ORF7-8 coding regions. Intranasal inoculation with BK2102 induced humoral and cellular immune responses in Syrian hamsters without apparent tissue damage in the lungs, leading to protection against a SARS-CoV-2 D614G and an Omicron BA.5 strains. The neutralizing antibodies induced by BK2102 persisted for up to 364 days, which indicated that they confer long-term protection against infection. Furthermore, we confirmed the safety of BK2102 using transgenic (Tg) mice expressing human ACE2 (hACE2) that are highly susceptible to SARS-CoV-2. BK2102 did not kill the Tg mice, even when virus was administered at a dose of 106 plaque-forming units (PFUs), while 102 PFU of the D614G strain or an attenuated strain lacking the furin cleavage site of the spike was sufficient to kill mice. These results suggest that BK2102 is a promising live-vaccine candidate strain that confers long-term protection without significant virulence.
Collapse
MESH Headings
- Animals
- Vaccines, Attenuated/immunology
- Vaccines, Attenuated/adverse effects
- Vaccines, Attenuated/administration & dosage
- Vaccines, Attenuated/genetics
- SARS-CoV-2/immunology
- SARS-CoV-2/genetics
- SARS-CoV-2/pathogenicity
- COVID-19 Vaccines/immunology
- COVID-19 Vaccines/adverse effects
- COVID-19 Vaccines/administration & dosage
- COVID-19 Vaccines/genetics
- COVID-19/prevention & control
- COVID-19/immunology
- Mice
- Antibodies, Neutralizing/blood
- Antibodies, Neutralizing/immunology
- Antibodies, Viral/blood
- Antibodies, Viral/immunology
- Mesocricetus
- Humans
- Mice, Transgenic
- Immunogenicity, Vaccine
- Cricetinae
- Female
- Spike Glycoprotein, Coronavirus/genetics
- Spike Glycoprotein, Coronavirus/immunology
Collapse
Affiliation(s)
- Mie Suzuki Okutani
- The Research Foundation for Microbial Diseases of Osaka UniversitySuitaJapan
- Virus Vaccine Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Institute for Open and Transdisciplinary Research Initiatives, Osaka UniversitySuitaJapan
| | - Shinya Okamura
- The Research Foundation for Microbial Diseases of Osaka UniversitySuitaJapan
- Virus Vaccine Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Institute for Open and Transdisciplinary Research Initiatives, Osaka UniversitySuitaJapan
| | - Tang Gis
- The Research Foundation for Microbial Diseases of Osaka UniversitySuitaJapan
| | - Hitomi Sasaki
- The Research Foundation for Microbial Diseases of Osaka UniversitySuitaJapan
| | - Suni Lee
- The Research Foundation for Microbial Diseases of Osaka UniversitySuitaJapan
| | - Akiho Kashiwabara
- The Research Foundation for Microbial Diseases of Osaka UniversitySuitaJapan
- Virus Vaccine Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Institute for Open and Transdisciplinary Research Initiatives, Osaka UniversitySuitaJapan
| | - Simon Goto
- The Research Foundation for Microbial Diseases of Osaka UniversitySuitaJapan
| | - Mai Matsumoto
- The Research Foundation for Microbial Diseases of Osaka UniversitySuitaJapan
| | - Mayuko Yamawaki
- The Research Foundation for Microbial Diseases of Osaka UniversitySuitaJapan
| | - Toshiaki Miyazaki
- The Research Foundation for Microbial Diseases of Osaka UniversitySuitaJapan
| | - Tatsuya Nakagawa
- Department of Experimental Genome Research, Research Institute for Microbial Diseases, Osaka UniversitySuitaJapan
| | - Masahito Ikawa
- Department of Experimental Genome Research, Research Institute for Microbial Diseases, Osaka UniversitySuitaJapan
- Center for Advanced Modalities and DDS (CAMaD), Osaka UniversitySuitaJapan
- Center for Infectious Disease Education and Research (CiDER), Osaka UniversitySuitaJapan
| | - Wataru Kamitani
- Department of Infectious Diseases and Host Defense, Gunma University Graduate School of MedicineMaebashiJapan
| | - Shiro Takekawa
- The Research Foundation for Microbial Diseases of Osaka UniversitySuitaJapan
| | - Koichi Yamanishi
- The Research Foundation for Microbial Diseases of Osaka UniversitySuitaJapan
| | - Hirotaka Ebina
- The Research Foundation for Microbial Diseases of Osaka UniversitySuitaJapan
- Virus Vaccine Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Institute for Open and Transdisciplinary Research Initiatives, Osaka UniversitySuitaJapan
- Center for Advanced Modalities and DDS (CAMaD), Osaka UniversitySuitaJapan
- Center for Infectious Disease Education and Research (CiDER), Osaka UniversitySuitaJapan
- Virus Vaccine Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Research institute for Microbial Diseases, Osaka UniversitySuitaJapan
| |
Collapse
|
35
|
Jiang Y, Sanyal M, Hussein NA, Baghdasaryan A, Zhang M, Wang F, Ren F, Li J, Zhu G, Meng Y, Adamska JZ, Mellins E, Dai H. A SARS-CoV-2 vaccine on an NIR-II/SWIR emitting nanoparticle platform. SCIENCE ADVANCES 2025; 11:eadp5539. [PMID: 39919189 PMCID: PMC11804919 DOI: 10.1126/sciadv.adp5539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Accepted: 01/07/2025] [Indexed: 02/09/2025]
Abstract
The COVID-19 pandemic caused a global health crisis that resulted in millions of deaths. Effective vaccines have played central roles in curtailing the pandemic. Here, we developed a down-converting near-infrared IIb (NIR-IIb; 1500 to 1700 nanometers) luminescent, pure NaErF4@NaYF4 rare-earth nanoparticle (pEr) as vaccine carriers. The pEr nanoparticles were coated with three layers of cross-linked biocompatible polymers (pEr-P3; ~55 nanometers) and conjugated to the receptor binding domain (RBD) of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike protein. Upon subcutaneous injection of the pEr-P3-RBD nanovaccine in mice, in vivo NIR-IIb imaging revealed active vaccine trafficking and migration to lymph nodes through lymphatic vessels. Two doses of the adjuvant-free vaccine elicited long-lasting (>7 months) high titers of serum viral neutralization antibody and anti-RBD immunoglobulin G, along with robust RBD-specific germinal center B cells and T follicular helper cells. We devised in vivo NIR-II molecular imaging of RBD-specific cells in lymph nodes, opening noninvasive assessments of vaccine-elicited immune responses longitudinally.
Collapse
Affiliation(s)
- Yingying Jiang
- Department of Chemistry and Bio-X, Stanford University, Stanford, CA, USA
- Department of Chemistry, The University of Hong Kong, Pokfulam, Hong Kong SAR
- Materials Innovation Institute for Life Sciences and Energy (MILES), HKU-SIRI, Shenzhen, P. R. China
| | - Mrinmoy Sanyal
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA
- Department of Biochemistry, School of Medicine, Stanford University, Stanford, CA, USA
| | - Noor A. Hussein
- Department of Pediatrics, Stanford University, Stanford, CA, USA
| | - Ani Baghdasaryan
- Department of Chemistry and Bio-X, Stanford University, Stanford, CA, USA
| | - Mengzhen Zhang
- Department of Chemistry and Bio-X, Stanford University, Stanford, CA, USA
| | - Feifei Wang
- Department of Chemistry and Bio-X, Stanford University, Stanford, CA, USA
- Materials Innovation Institute for Life Sciences and Energy (MILES), HKU-SIRI, Shenzhen, P. R. China
- Department of Electrical and Electronic Engineering, The University of Hong Kong, Pokfulam, Hong Kong SAR
| | - Fuqiang Ren
- Department of Chemistry and Bio-X, Stanford University, Stanford, CA, USA
| | - Jiachen Li
- Department of Chemistry and Bio-X, Stanford University, Stanford, CA, USA
| | - Guanzhou Zhu
- Department of Chemistry and Bio-X, Stanford University, Stanford, CA, USA
| | - Yifan Meng
- Department of Chemistry and Bio-X, Stanford University, Stanford, CA, USA
| | - Julia Zofia Adamska
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford University, Stanford, CA, USA
- Department of Genetics, Stanford University, Stanford, CA, USA
| | | | - Hongjie Dai
- Department of Chemistry and Bio-X, Stanford University, Stanford, CA, USA
- Department of Chemistry, The University of Hong Kong, Pokfulam, Hong Kong SAR
- Materials Innovation Institute for Life Sciences and Energy (MILES), HKU-SIRI, Shenzhen, P. R. China
| |
Collapse
|
36
|
Shoemaker SR, Luo M, Dam KMA, Pak JE, Hoffmann MAG, Marqusee S. The Interplay of Furin Cleavage and D614G in Modulating SARS-CoV-2 Spike Protein Dynamics. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.27.635166. [PMID: 39975226 PMCID: PMC11838386 DOI: 10.1101/2025.01.27.635166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
We report a detailed analysis of the full-length SARS-CoV-2 spike dynamics within a native-like membrane environment and variants inaccessible to studies on soluble constructs by conducting hydrogen-deuterium exchange mass spectrometry (HDX-MS) on enveloped virus-like particles (eVLPs) displaying various spike constructs. We find that the previously identified open-interface trimer conformation is sampled in all eVLP-displayed spike variants studied including sequences from engineered vaccine constructs and native viral sequences. The D614G mutation, which arose early in the pandemic, favors the canonical 'closed-interface' prefusion conformation, potentially mitigating premature S1 shedding in the presence of a cleaved furin site and providing an evolutionary advantage to the virus. Remarkably, furin cleavage at the S1/S2 boundary allosterically increases the flexibility of the S2' site, which may facilitate increased TMPRSS2 processing, enhancing viral infectivity. The use of eVLPs in HDX-MS studies provides a powerful platform for studying viral and membrane proteins in near-native environments.
Collapse
|
37
|
Varghese A, Liu J, Liu B, Guo W, Dong F, Patterson TA, Hong H. Analysis of Structures of SARS-CoV-2 Papain-like Protease Bound with Ligands Unveils Structural Features for Inhibiting the Enzyme. Molecules 2025; 30:491. [PMID: 39942596 PMCID: PMC11820935 DOI: 10.3390/molecules30030491] [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: 12/17/2024] [Revised: 01/17/2025] [Accepted: 01/20/2025] [Indexed: 02/16/2025] Open
Abstract
The COVID-19 pandemic, driven by the novel coronavirus SARS-CoV-2, has drastically reshaped global health and socioeconomic landscapes. The papain-like protease (PLpro) plays a critical role in viral polyprotein cleavage and immune evasion, making it a prime target for therapeutic intervention. Numerous compounds have been identified as inhibitors of SARS-CoV-2 PLpro, with many characterized through crystallographic studies. To date, over 70 three-dimensional (3D) structures of PLpro complexed ligands have been deposited in the Protein Data Bank, offering valuable insight into ligand-binding features that could aid the discovery and development of effective COVID-19 treatments targeting PLpro. In this study, we reviewed and analyzed these 3D structures, focusing on the key residues involved in ligand interactions. Our analysis revealed that most inhibitors bind to PLpro's substrate recognition sites S3/S4 and SUb2. While these sites are highly attractive and have been extensively explored, other potential binding regions, such as SUb1 and the Zn(II) domain, are less explored and may hold untapped potential for future COVID-19 drug discovery and development. Our structural analysis provides insights into the molecular features of PLpro that could accelerate the development of novel therapeutics targeting this essential viral enzyme.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Huixiao Hong
- National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR 72079, USA; (A.V.); (J.L.); (B.L.); (W.G.); (F.D.); (T.A.P.)
| |
Collapse
|
38
|
Meurens F, Renois F, Karniychuk U. Elegant and Innovative Recoding Strategies for Advancing Vaccine Development. Vaccines (Basel) 2025; 13:78. [PMID: 39852857 PMCID: PMC11768987 DOI: 10.3390/vaccines13010078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 01/10/2025] [Accepted: 01/14/2025] [Indexed: 01/26/2025] Open
Abstract
Recoding strategies have emerged as a promising approach for developing safer and more effective vaccines by altering the genetic structure of microorganisms, such as viruses, without changing their proteins. This method enhances vaccine safety and efficacy while minimizing the risk of reversion to virulence. Recoding enhances the frequency of CpG dinucleotides, which in turn activates immune responses and ensures a strong attenuation of the pathogens. Recent advancements highlight synonymous recoding's potential, offering improved genetic stability and immunogenicity compared to traditional methods. Live vaccines attenuated using classical methods pose a risk of reversion to virulence and can be time-consuming to produce. Synonymous recoding, involving numerous codon alterations, boosts safety and vaccine stability. One challenge is balancing attenuation with yield; however, innovations like Zinc-finger antiviral protein (ZAP) knockout cell lines can enhance vaccine production. Beyond viral vaccines, recoding can apply to bacterial vaccines, as exemplified by modified Escherichia coli and Streptococcus pneumoniae strains, which show reduced virulence. Despite promising results, challenges like ensuring genetic stability, high yield, and regulatory approval remain. Briefly, ongoing research aims to harness these innovations for comprehensive improvements in vaccine design and deployment. In this commentary, we sought to further engage the community's interest in this elegant approach by briefly highlighting its main advantages, disadvantages, and future prospects.
Collapse
Affiliation(s)
- François Meurens
- Centre de Recherche en Infectiologie Porcine et Avicole, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC J2S 2M2, Canada;
- Department of Veterinary Microbiology and Immunology, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E2, Canada;
| | - Fanny Renois
- Centre de Recherche en Infectiologie Porcine et Avicole, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC J2S 2M2, Canada;
| | - Uladzimir Karniychuk
- Department of Veterinary Microbiology and Immunology, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E2, Canada;
- Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
39
|
Shi YH, Shen JX, Tao Y, Xia YL, Zhang ZB, Fu YX, Zhang KQ, Liu SQ. Dissecting the Binding Affinity of Anti-SARS-CoV-2 Compounds to Human Transmembrane Protease, Serine 2: A Computational Study. Int J Mol Sci 2025; 26:587. [PMID: 39859303 PMCID: PMC11766390 DOI: 10.3390/ijms26020587] [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: 11/30/2024] [Revised: 01/05/2025] [Accepted: 01/09/2025] [Indexed: 01/27/2025] Open
Abstract
The human transmembrane protease, serine 2 (TMPRSS2), essential for SARS-CoV-2 entry, is a key antiviral target. Here, we computationally profiled the TMPRSS2-binding affinities of 15 antiviral compounds. Molecular dynamics (MD) simulations for the docked complexes revealed that three compounds exited the substrate-binding cavity (SBC), suggesting noncompetitive inhibition. Of the remaining compounds, five charged ones exhibited reduced binding stability due to competing electrostatic interactions and increased solvent exposure, while seven neutral compounds showed stronger binding affinity driven by van der Waals (vdW) interactions compensating for unfavorable electrostatic effects (including electrostatic interactions and desolvation penalties). Positive and negative hotspot residues were identified as uncharged and charged, respectively, both lining the SBC. Despite forming diverse interactions with compounds, the burial of positive hotspots led to strong vdW interactions that overcompensated for unfavorable electrostatic effects, whereas negative hotspots incurred high desolvation penalties, negating any favorable contributions. Charged residues at the SBC's outer rim can reduce binding affinity significantly when forming hydrogen bonds or salt bridges. These findings underscore the importance of enhancing vdW interactions with uncharged residues and minimizing the unfavorable electrostatic effects of charged residues, providing valuable insights for designing effective TMPRSS2 inhibitors.
Collapse
Affiliation(s)
- Yue-Hui Shi
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan and School of Life Sciences, Yunnan University, Kunming 650091, China; (Y.-H.S.); (J.-X.S.); (Y.T.); (Y.-L.X.); (K.-Q.Z.)
| | - Jian-Xin Shen
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan and School of Life Sciences, Yunnan University, Kunming 650091, China; (Y.-H.S.); (J.-X.S.); (Y.T.); (Y.-L.X.); (K.-Q.Z.)
| | - Yan Tao
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan and School of Life Sciences, Yunnan University, Kunming 650091, China; (Y.-H.S.); (J.-X.S.); (Y.T.); (Y.-L.X.); (K.-Q.Z.)
| | - Yuan-Ling Xia
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan and School of Life Sciences, Yunnan University, Kunming 650091, China; (Y.-H.S.); (J.-X.S.); (Y.T.); (Y.-L.X.); (K.-Q.Z.)
| | - Zhi-Bi Zhang
- Yunnan Key Laboratory of Stem Cell and Regenerative Medicine, Biomedical Engineering Research Center, Kunming Medical University, Kunming 650500, China;
| | - Yun-Xin Fu
- Human Genetics Center and Department of Biostatistics and Data Science, School of Public Health, The University of Texas Health Science Center, Houston, TX 77030, USA;
| | - Ke-Qin Zhang
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan and School of Life Sciences, Yunnan University, Kunming 650091, China; (Y.-H.S.); (J.-X.S.); (Y.T.); (Y.-L.X.); (K.-Q.Z.)
| | - Shu-Qun Liu
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan and School of Life Sciences, Yunnan University, Kunming 650091, China; (Y.-H.S.); (J.-X.S.); (Y.T.); (Y.-L.X.); (K.-Q.Z.)
| |
Collapse
|
40
|
Barros de Lima G, Nencioni E, Thimoteo F, Perea C, Pinto RFA, Sasaki SD. TMPRSS2 as a Key Player in Viral Pathogenesis: Influenza and Coronaviruses. Biomolecules 2025; 15:75. [PMID: 39858469 PMCID: PMC11764435 DOI: 10.3390/biom15010075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 12/28/2024] [Accepted: 01/03/2025] [Indexed: 01/27/2025] Open
Abstract
TMPRSS2, a human transmembrane protease enzyme, plays a crucial role in the spread of certain viruses, including influenza and coronaviruses. This enzyme promotes viral infection by cleaving viral glycoproteins, which helps viruses like SARS-CoV-2 and influenza A enter cells more effectively. Genetic differences in TMPRSS2 may affect people's susceptibility to COVID-19, underscoring the need for studies that consider diverse populations. Beyond infectious diseases, TMPRSS2 has also been linked to some cancers, suggesting it could be a valuable target for drug development. This review provides a summary of TMPRSS2 inhibitors currently under study, with some already in clinical trials to test their effectiveness against viral infections. As we uncover more about TMPRSS2's role in pathogenesis, it could open new doors for therapies to combat future outbreaks.
Collapse
Affiliation(s)
| | | | | | | | | | - Sergio Daishi Sasaki
- Graduate Program of Biosystems, Centro de Ciências Naturais e Humanas, Universidade Federal do ABC (UFABC), São Bernardo do Campo, São Paulo 09606-045, Brazil; (G.B.d.L.); (E.N.); (F.T.); (C.P.); (R.F.A.P.)
| |
Collapse
|
41
|
Bermúdez-Abreut E, Fundora-Barrios T, Hernández Fernández DR, Noa Romero E, Fraga-Quintero A, Casadesús Pazos AV, Fernández-Marrero B, Plasencia Iglesias CA, Clavel Pérez M, Sosa Aguiar K, Sánchez-Ramírez B, Hernández T. Antiviral activity of an ACE2-Fc fusion protein against SARS-CoV-2 and its variants. PLoS One 2025; 20:e0312402. [PMID: 39752453 PMCID: PMC11698409 DOI: 10.1371/journal.pone.0312402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 10/04/2024] [Indexed: 01/06/2025] Open
Abstract
SARS-CoV-2 has continued spreading around the world in recent years since the initial outbreak in 2019, frequently developing into new variants with greater human infectious capacity. SARS-CoV-2 and its mutants use the angiotensin-converting enzyme 2 (ACE2) as a cellular entry receptor, which has triggered several therapeutic strategies against COVID-19 relying on the use of ACE2 recombinant proteins as decoy receptors. In this work, we propose an ACE2 silent Fc fusion protein (ACE2-hFcLALA) as a candidate therapy against COVID-19. This fusion protein was able to block the binding of SARS-CoV-2 RBD to ACE2 receptor as measured by ELISA and flow cytometry inhibition assays. Moreover, we used classical neutralization assays and a progeny neutralization assay to show that the ACE2-hFcLALA fusion protein is capable of neutralizing the authentic virus. Additionally, we found that this fusion protein was more effective in preventing in vitro infection with different variants of interest (alpha, beta, delta, and omicron) compared to the D614G strain. Our results suggest the potential of this molecule to be used in both therapeutic and preventive settings against current and emerging mutants that use ACE2 as a gateway to human cells.
Collapse
Affiliation(s)
| | - Talia Fundora-Barrios
- Immunology and Immunotherapy Division, Center of Molecular Immunology (CIM), Havana, Cuba
| | | | - Enrique Noa Romero
- National Laboratory of Civil Defense (NLCD), Jamaica Highway and National Highway, San José of Lajas, Mayabeque, Cuba
| | - Anitza Fraga-Quintero
- National Laboratory of Civil Defense (NLCD), Jamaica Highway and National Highway, San José of Lajas, Mayabeque, Cuba
| | - Ana V. Casadesús Pazos
- Immunology and Immunotherapy Division, Center of Molecular Immunology (CIM), Havana, Cuba
| | | | | | - Marilyn Clavel Pérez
- Immunology and Immunotherapy Division, Center of Molecular Immunology (CIM), Havana, Cuba
| | - Katya Sosa Aguiar
- Immunology and Immunotherapy Division, Center of Molecular Immunology (CIM), Havana, Cuba
| | | | - Tays Hernández
- Immunology and Immunotherapy Division, Center of Molecular Immunology (CIM), Havana, Cuba
| |
Collapse
|
42
|
Zhang Q, Rosa RSL, Ray A, Durlet K, Dorrazehi GM, Bernardi RC, Alsteens D. Probing SARS-CoV-2 membrane binding peptide via single-molecule AFM-based force spectroscopy. Nat Commun 2025; 16:6. [PMID: 39747000 PMCID: PMC11696146 DOI: 10.1038/s41467-024-55358-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 12/09/2024] [Indexed: 01/04/2025] Open
Abstract
The SARS-CoV-2 spike protein's membrane-binding domain bridges the viral and host cell membrane, a critical step in triggering membrane fusion. Here, we investigate how the SARS-CoV-2 spike protein interacts with host cell membranes, focusing on a membrane-binding peptide (MBP) located near the TMPRSS2 cleavage site. Through in vitro and computational studies, we examine both primed (TMPRSS2-cleaved) and unprimed versions of the MBP, as well as the influence of its conserved disulfide bridge on membrane binding. Our results show that the MBP preferentially associates with cholesterol-rich membranes, and we find that cholesterol depletion significantly reduces viral infectivity. Furthermore, we observe that the disulfide bridge stabilizes the MBP's interaction with the membrane, suggesting a structural role in viral entry. Together, these findings highlight the importance of membrane composition and peptide structure in SARS-CoV-2 infectivity and suggest that targeting the disulfide bridge could provide a therapeutic strategy against infection.
Collapse
Affiliation(s)
- Qingrong Zhang
- Louvain Institute of Biomolecular Science and Technology, Université catholique de Louvain, Croix du sud 4-5, L7.07.07, Louvain-la-Neuve, Belgium
| | - Raissa S L Rosa
- Department of Chemistry and Biochemistry, Auburn University, Auburn, AL, USA
| | - Ankita Ray
- Louvain Institute of Biomolecular Science and Technology, Université catholique de Louvain, Croix du sud 4-5, L7.07.07, Louvain-la-Neuve, Belgium
| | - Kimberley Durlet
- Louvain Institute of Biomolecular Science and Technology, Université catholique de Louvain, Croix du sud 4-5, L7.07.07, Louvain-la-Neuve, Belgium
| | - Gol Mohammad Dorrazehi
- Louvain Institute of Biomolecular Science and Technology, Université catholique de Louvain, Croix du sud 4-5, L7.07.07, Louvain-la-Neuve, Belgium
| | - Rafael C Bernardi
- Department of Chemistry and Biochemistry, Auburn University, Auburn, AL, USA.
- Department of Physics, Auburn University, Auburn, AL, USA.
| | - David Alsteens
- Louvain Institute of Biomolecular Science and Technology, Université catholique de Louvain, Croix du sud 4-5, L7.07.07, Louvain-la-Neuve, Belgium.
- WELBIO department, WEL Research Institute, Avenue Pasteur, 6, Wavre, Belgium.
| |
Collapse
|
43
|
Furnon W, Cowton VM, De Lorenzo G, Orton R, Herder V, Cantoni D, Ilia G, Mendonca DC, Kerr K, Allan J, Upfold N, Meehan GR, Bakshi S, Das UR, Molina Arias S, McElwee M, Little S, Logan N, Kwok K, Smollett K, Willett BJ, Da Silva Filipe A, Robertson DL, Grove J, Patel AH, Palmarini M. Phenotypic evolution of SARS-CoV-2 spike during the COVID-19 pandemic. Nat Microbiol 2025; 10:77-93. [PMID: 39753670 PMCID: PMC11726466 DOI: 10.1038/s41564-024-01878-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 11/11/2024] [Indexed: 01/12/2025]
Abstract
SARS-CoV-2 variants are mainly defined by mutations in their spike. It is therefore critical to understand how the evolutionary trajectories of spike affect virus phenotypes. So far, it has been challenging to comprehensively compare the many spikes that emerged during the pandemic in a single experimental platform. Here we generated a panel of recombinant viruses carrying different spike proteins from 27 variants circulating between 2020 and 2024 in the same genomic background. We then assessed several of their phenotypic traits both in vitro and in vivo. We found distinct phenotypic trajectories of spike among and between variants circulating before and after the emergence of Omicron variants. Spike of post-Omicron variants maintained enhanced tropism for the nasal epithelium and large airways but displayed, over time, several phenotypic traits typical of the pre-Omicron variants. Hence, spike with phenotypic features of both pre- and post-Omicron variants may continue to emerge in the future.
Collapse
Affiliation(s)
- Wilhelm Furnon
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK
| | - Vanessa M Cowton
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK
| | | | - Richard Orton
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK
| | - Vanessa Herder
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK
- CVR-CRUSH, MRC-University of Glasgow Centre for Virus Research, Glasgow, UK
| | - Diego Cantoni
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK
| | - Georgios Ilia
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK
| | - Diogo Correa Mendonca
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK
- CVR-CRUSH, MRC-University of Glasgow Centre for Virus Research, Glasgow, UK
| | - Karen Kerr
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK
- CVR-CRUSH, MRC-University of Glasgow Centre for Virus Research, Glasgow, UK
| | - Jay Allan
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK
| | - Nicole Upfold
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK
| | - Gavin R Meehan
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK
- CVR-CRUSH, MRC-University of Glasgow Centre for Virus Research, Glasgow, UK
| | - Siddharth Bakshi
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK
| | - Udeet Ranjan Das
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK
| | - Sergi Molina Arias
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK
- CVR-CRUSH, MRC-University of Glasgow Centre for Virus Research, Glasgow, UK
| | - Marion McElwee
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK
- CVR-CRUSH, MRC-University of Glasgow Centre for Virus Research, Glasgow, UK
| | - Sarah Little
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK
| | - Nicola Logan
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK
| | - Kirsty Kwok
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK
| | | | - Brian J Willett
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK
| | | | | | - Joe Grove
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK
| | - Arvind H Patel
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK.
- CVR-CRUSH, MRC-University of Glasgow Centre for Virus Research, Glasgow, UK.
| | - Massimo Palmarini
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK.
- CVR-CRUSH, MRC-University of Glasgow Centre for Virus Research, Glasgow, UK.
| |
Collapse
|
44
|
McCaig CD. SARS-CoV-2 Is an Electricity-Driven Virus. Rev Physiol Biochem Pharmacol 2025; 187:361-410. [PMID: 39838019 DOI: 10.1007/978-3-031-68827-0_18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2025]
Abstract
One of the most important and challenging biological events of recent times has been the pandemic caused by SARS-CoV-2. Since the underpinning argument behind this book is the ubiquity of electrical forces driving multiple disparate biological events, consideration of key aspects of the SARS-CoV-2 structural proteins is included. Electrical regulation of spike protein, nucleocapsid protein, membrane protein, and envelope protein is included, with several of their activities regulated by LLPS and the multivalent and π-cation and π-π electrical forces that drive phase separation.
Collapse
Affiliation(s)
- Colin D McCaig
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, Scotland, UK
| |
Collapse
|
45
|
Brocke SA, Reidel B, Ehre C, Rebuli ME, Robinette C, Schichlein KD, Brooks CA, Jaspers I. Profiling endogenous airway proteases and antiproteases and modeling proteolytic activation of Influenza HA using in vitro and ex vivo human airway surface liquid samples. PLoS One 2024; 19:e0306197. [PMID: 39739661 PMCID: PMC11687774 DOI: 10.1371/journal.pone.0306197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 12/09/2024] [Indexed: 01/02/2025] Open
Abstract
Imbalance of airway proteases and antiproteases has been implicated in diseases such as COPD and environmental exposures including cigarette smoke and ozone. To initiate infection, endogenous proteases are commandeered by respiratory viruses upon encountering the airway epithelium. The airway proteolytic environment likely contains redundant antiproteases and proteases with diverse catalytic mechanisms, however a proteomic profile of these enzymes and inhibitors in airway samples has not been reported. The objective of this study was to first profile extracellular proteases and antiproteases using human airway epithelial cell cultures and ex vivo nasal epithelial lining fluid (NELF) samples. Secondly, we present an optimized method for probing the proteolytic environment of airway surface liquid samples (in vitro and ex vivo) using fluorogenic peptides modeling the cleavage sites of respiratory viruses. We detected 48 proteases in the apical wash of cultured human nasal epithelial cells (HNECs) (n = 6) and 57 in NELF (n = 13) samples from healthy human subjects using mass-spectrometry based proteomics. Additionally, we detected 29 and 48 antiproteases in the HNEC apical washes and NELF, respectively. We observed large interindividual variability in rate of cleavage of an Influenza H1 peptide in the ex vivo clinical samples. Since protease and antiprotease levels have been found to be altered in the airways of smokers, we compared proteolytic cleavage in ex vivo nasal lavage samples from male/female smokers and non-smokers. There was a statistically significant increase in proteolysis of Influenza H1 in NLF from male smokers compared to female smokers. Furthermore, we measured cleavage of the S1/S2 site of SARS-CoV, SARS-CoV-2, and SARS-CoV-2 Delta peptides in various airway samples, suggesting the method could be used for other viruses of public health relevance. This assay presents a direct and efficient method of evaluating the proteolytic environment of human airway samples in assessment of therapeutic treatment, exposure, or underlying disease.
Collapse
Affiliation(s)
- Stephanie A. Brocke
- Curriculum in Toxicology and Environmental Medicine, University of North Carolina, Chapel Hill, NC, United States of America
| | - Boris Reidel
- Marsico Lung Institute, University of North Carolina, Chapel Hill, NC, United States of America
| | - Camille Ehre
- Marsico Lung Institute, University of North Carolina, Chapel Hill, NC, United States of America
| | - Meghan E. Rebuli
- Curriculum in Toxicology and Environmental Medicine, University of North Carolina, Chapel Hill, NC, United States of America
- Center for Environmental Medicine, Asthma, and Lung Biology, University of North Carolina, Chapel Hill, NC, United States of America
- Department of Pediatrics, School of Medicine, University of North Carolina, Chapel Hill, NC, United States of America
| | - Carole Robinette
- Center for Environmental Medicine, Asthma, and Lung Biology, University of North Carolina, Chapel Hill, NC, United States of America
| | - Kevin D. Schichlein
- Curriculum in Toxicology and Environmental Medicine, University of North Carolina, Chapel Hill, NC, United States of America
| | - Christian A. Brooks
- Center for Environmental Medicine, Asthma, and Lung Biology, University of North Carolina, Chapel Hill, NC, United States of America
| | - Ilona Jaspers
- Curriculum in Toxicology and Environmental Medicine, University of North Carolina, Chapel Hill, NC, United States of America
- Center for Environmental Medicine, Asthma, and Lung Biology, University of North Carolina, Chapel Hill, NC, United States of America
- Department of Pediatrics, School of Medicine, University of North Carolina, Chapel Hill, NC, United States of America
- Department of Microbiology and Immunology, School of Medicine, University of North Carolina, Chapel Hill, NC, United States of America
| |
Collapse
|
46
|
Šerý O, Dziedzinska R. Risk impact of SARS-CoV-2 coronavirus and spike protein on cardiac tissue: a comprehensive review. Physiol Res 2024; 73:S655-S669. [PMID: 39808169 PMCID: PMC11827061 DOI: 10.33549/physiolres.935476] [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: 04/25/2024] [Accepted: 08/19/2024] [Indexed: 01/18/2025] Open
Abstract
The global COVID-19 pandemic, caused by SARS-CoV-2, has led to significant morbidity and mortality, with a profound impact on cardiovascular health. This review investigates the mechanisms of SARS-CoV-2's interaction with cardiac tissue, particularly emphasizing the role of the Spike protein and ACE2 receptor in facilitating viral entry and subsequent cardiac complications. We dissect the structural features of the virus, its interactions with host cell receptors, and the resulting pathophysiological changes in the heart. Highlighting SARS-CoV-2's broad organ tropism, especially its effects on cardiomyocytes via ACE2 and TMPRSS2, the review addresses how these interactions exacerbate cardiovascular issues in patients with pre-existing conditions such as diabetes and hypertension. Additionally, we assess both direct and indirect mechanisms of virus-induced cardiac damage, including myocarditis, arrhythmias, and long-term complications such as 'long COVID'. This review underscores the complexity of SARS-CoV-2's impact on the heart, emphasizing the need for ongoing research to fully understand its long-term effects on cardiovascular health. Key words: COVID-19, Heart, ACE2, Spike protein, Cardiomyocytes, Myocarditis, Long COVID.
Collapse
Affiliation(s)
- O Šerý
- Laboratory of Neurobiology and Molecular Psychiatry, Department of Biochemistry, Faculty of Science, Masaryk University, Brno, Czech Republic.
| | | |
Collapse
|
47
|
La Rosa P, Tiberi J, Palermo E, Stefanelli R, Tiano SML, Canterini S, Cortese M, Hiscott J, Fiorenza MT. The inactivation of the Niemann Pick C1 cholesterol transporter restricts SARS-CoV-2 entry into host cells by decreasing ACE2 abundance at the plasma membrane. Cell Biosci 2024; 14:148. [PMID: 39707537 DOI: 10.1186/s13578-024-01331-4] [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: 05/07/2024] [Accepted: 11/25/2024] [Indexed: 12/23/2024] Open
Abstract
BACKGROUND The Niemann Pick C1 (NPC1) protein is an intracellular cholesterol transporter located in the late endosome/lysosome (LE/Ly) that is involved in the mobilization of endocytosed cholesterol. Loss-of-function mutations in the NPC1 gene lead to the accumulation of cholesterol and sphingolipids in LE/Ly, resulting in severe fatal NPC1 disease. Cellular alterations associated with NPC1 inactivation affect both the integrity of lipid rafts and the endocytic pathway. Because the angiotensin-converting enzyme 2 (ACE2) and type 2 serine transmembrane protease (TMPRSS2), interactors of the SARS-CoV-2 Spike protein also localize to lipid rafts, we sought to investigate the hypothesis that NPC1 inactivation would generate an intrinsically unfavorable barrier to SARS-CoV-2 entry. RESULTS In this study, we show that inhibition of the cholesterol transporter activity of NPC1 in cells that express both ACE2 and TMPRSS2, considerably reduces SARS-CoV-2 infectivity, evaluated as early as 4 h post-infection. Mechanistically, treatment with NPC1 specific inhibitor U18666A relocalizes ACE2 from the plasma membrane to the autophagosomal/lysosomal compartment, thereby reducing SARS-CoV-2 entry into treated cells. Reduction of viral entry was observed for both fully infectious SARS-CoV-2 virus and with a pseudotyped VSV-Spike-GFP virus. For instance, U18666A-treated Caco-2 cells infected with the pseudotyped VSV-Spike-GFP showed a > threefold and > 40-fold reduction in virus titer when infectivity was measured at 4 h or 24 h post-infection, respectively. A similar effect was observed in CRISP/R-Cas9-edited Caco-2 cells, which were even more resistant to SARS-CoV-2 infection as indicated by a 97% reduction of viral titers. CONCLUSION Overall, this study provides compelling evidence that the inhibition of NPC1 cholesterol transporter activity generates a cellular environment that hinders SARS-CoV-2 entry. ACE2 depletion from the plasma membrane appears to play a major role as limiting factor for viral entry.
Collapse
Affiliation(s)
- Piergiorgio La Rosa
- Division of Neuroscience, Dept. of Psychology, University La Sapienza, Via dei Sardi 70, 00185, Rome, Italy
- European Center for Brain Research, IRCCS Fondazione Santa Lucia, Via del Fosso di Fiorano 64, 00143, Rome, Italy
| | - Jessica Tiberi
- Division of Neuroscience, Dept. of Psychology, University La Sapienza, Via dei Sardi 70, 00185, Rome, Italy
- PhD Program in Behavioral Neuroscience, Sapienza University of Rome, Rome, Italy
| | - Enrico Palermo
- Istituto Pasteur Italia-Cenci Bolognetti Foundation, Viale Regina Elena 291, 00161, Rome, Italy
| | - Roberta Stefanelli
- Division of Neuroscience, Dept. of Psychology, University La Sapienza, Via dei Sardi 70, 00185, Rome, Italy
| | - Sofia Maria Luigia Tiano
- Telethon Institute of Genetics and Medicine, TIGEM, Via Campi Flegrei, 34, 80078, Pozzuoli, Italy
| | - Sonia Canterini
- Division of Neuroscience, Dept. of Psychology, University La Sapienza, Via dei Sardi 70, 00185, Rome, Italy
- European Center for Brain Research, IRCCS Fondazione Santa Lucia, Via del Fosso di Fiorano 64, 00143, Rome, Italy
| | - Mirko Cortese
- Telethon Institute of Genetics and Medicine, TIGEM, Via Campi Flegrei, 34, 80078, Pozzuoli, Italy
- Universitá della Campania Luigi Vanvitelli, Via Vivaldi, 43, 81100, Caserta, Italy
| | - John Hiscott
- Istituto Pasteur Italia-Cenci Bolognetti Foundation, Viale Regina Elena 291, 00161, Rome, Italy
| | - Maria Teresa Fiorenza
- Division of Neuroscience, Dept. of Psychology, University La Sapienza, Via dei Sardi 70, 00185, Rome, Italy.
- European Center for Brain Research, IRCCS Fondazione Santa Lucia, Via del Fosso di Fiorano 64, 00143, Rome, Italy.
| |
Collapse
|
48
|
Birtles D, Lee J. Exploring the influence of anionic lipids in the host cell membrane on viral fusion. Biochem Soc Trans 2024; 52:2593-2602. [PMID: 39700018 PMCID: PMC11668307 DOI: 10.1042/bst20240833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 11/25/2024] [Accepted: 12/02/2024] [Indexed: 12/21/2024]
Abstract
Membrane fusion is an essential component of the viral lifecycle that allows the delivery of the genetic information of the virus into the host cell. Specialized viral glycoproteins exist on the surface of mature virions where they facilitate fusion through significant conformational changes, ultimately bringing opposing membranes into proximity until they eventually coalesce. This process can be positively influenced by a number of specific cellular factors such as pH, enzymatic cleavage, divalent ions, and the composition of the host cell membrane. In this review, we have summarized how anionic lipids have come to be involved in viral fusion and how the endosomal resident anionic lipid BMP has become increasingly implicated as an important cofactor for those viruses that fuse via the endocytic pathway.
Collapse
Affiliation(s)
- Daniel Birtles
- Department of Chemistry and Biochemistry, University of Maryland, College Park, MD 20742, U.S.A
| | - Jinwoo Lee
- Department of Chemistry and Biochemistry, University of Maryland, College Park, MD 20742, U.S.A
| |
Collapse
|
49
|
Stein SC, Hansen G, Ssebyatika G, Ströh LJ, Ochulor O, Herold E, Schwarzloh B, Mutschall D, Zischke J, Cordes AK, Schneider T, Hinrichs I, Blasczyk R, Kleine-Weber H, Hoffmann M, Klein F, Kaiser FK, Gonzalez-Hernandez M, Armando F, Ciurkiewicz M, Beythien G, Pöhlmann S, Baumgärtner W, Osterhaus A, Schulz TF, Krey T. A human monoclonal antibody neutralizing SARS-CoV-2 Omicron variants containing the L452R mutation. J Virol 2024; 98:e0122324. [PMID: 39494911 DOI: 10.1128/jvi.01223-24] [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: 07/12/2024] [Accepted: 10/10/2024] [Indexed: 11/05/2024] Open
Abstract
The effectiveness of SARS-CoV-2 therapeutic antibodies targeting the spike (S) receptor-binding domain (RBD) has been hampered by the emergence of variants of concern (VOCs), which have acquired mutations to escape neutralizing antibodies (nAbs). These mutations are not evenly distributed on the RBD surface but cluster on several distinct surfaces, suggesting an influence of the targeted epitope on the capacity to neutralize a broad range of VOCs. Here, we identified a potent nAb from convalescent patients targeting the receptor-binding domain of a broad range of SARS-CoV-2 VOCs. Except for the Lambda and BA.2.86 variants, this nAb efficiently inhibited the entry of most tested VOCs, including Omicron subvariants BA.1, BA.2, XBB.1.5, and EG.5.1 and to a limited extent also BA.4/5, BA.4.6, and BQ.1.1. It bound recombinant S protein with picomolar affinity, reduced the viral load in the lung of infected hamsters, and prevented the severe lung pathology typical for SARS-CoV-2 infections. An X-ray structure of the nAb-RBD complex revealed an epitope that does not fall into any of the conventional classes and provided insights into its broad neutralization properties. Our findings highlight a conserved epitope within the SARS-CoV-2 RBD that should be preferably targeted by therapeutic antibodies and inform rational vaccine development.IMPORTANCETherapeutic antibodies are effective in preventing severe disease from SARS-CoV-2 infection and constitute an important option in pandemic preparedness, but mutations within the S protein of virus variants (e.g., a mutation of L452) confer resistance to many of such antibodies. Here, we identify a human antibody targeting the S protein receptor-binding domain (RBD) with an elevated escape barrier and characterize its interaction with the RBD functionally and structurally at the atomic level. A direct comparison with reported antibodies targeting the same epitope illustrates important differences in the interface, providing insights into the breadth of antibody binding. These findings highlight the relevance of an extended neutralization profiling in combination with biochemical and structural characterization of the antibody-RBD interaction for the selection of future therapeutic antibodies, which may accelerate the control of potential future pandemics.
Collapse
Affiliation(s)
- Saskia C Stein
- Institute of Virology, Hannover Medical School, Hannover, Germany
| | - Guido Hansen
- Institute of Biochemistry, Center of Structural and Cell Biology in Medicine, University of Lübeck, Lübeck, Germany
| | - George Ssebyatika
- Institute of Biochemistry, Center of Structural and Cell Biology in Medicine, University of Lübeck, Lübeck, Germany
| | - Luisa J Ströh
- Institute of Virology, Hannover Medical School, Hannover, Germany
| | | | - Elisabeth Herold
- Institute of Biochemistry, Center of Structural and Cell Biology in Medicine, University of Lübeck, Lübeck, Germany
| | - Britta Schwarzloh
- Institute of Biochemistry, Center of Structural and Cell Biology in Medicine, University of Lübeck, Lübeck, Germany
| | - Doris Mutschall
- Institute of Biochemistry, Center of Structural and Cell Biology in Medicine, University of Lübeck, Lübeck, Germany
| | - Jasmin Zischke
- Institute of Virology, Hannover Medical School, Hannover, Germany
- Laboratory of Experimental Immunology, Institute of Virology, University of Cologne, Cologne, Germany
| | - Anne K Cordes
- Institute of Virology, Hannover Medical School, Hannover, Germany
| | - Talia Schneider
- Institute of Virology, Hannover Medical School, Hannover, Germany
| | - Imke Hinrichs
- Institute of Biochemistry, Center of Structural and Cell Biology in Medicine, University of Lübeck, Lübeck, Germany
| | - Rainer Blasczyk
- Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, Hannover, Germany
| | - Hannah Kleine-Weber
- German Primate Center, Leibniz Institute for Primate Research, and Faculty of Biology and Psychology, University Göttingen, Göttingen, Germany
| | - Markus Hoffmann
- German Primate Center, Leibniz Institute for Primate Research, and Faculty of Biology and Psychology, University Göttingen, Göttingen, Germany
| | - Florian Klein
- Laboratory of Experimental Immunology, Institute of Virology, University of Cologne, Cologne, Germany
- German Center for Infection Research, Partner Site Bonn-Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Franziska K Kaiser
- Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
| | - Mariana Gonzalez-Hernandez
- Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
| | - Federico Armando
- Department of Pathology, University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
| | - Malgorzata Ciurkiewicz
- Department of Pathology, University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
| | - Georg Beythien
- Department of Pathology, University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
| | - Stefan Pöhlmann
- German Primate Center, Leibniz Institute for Primate Research, and Faculty of Biology and Psychology, University Göttingen, Göttingen, Germany
| | - Wolfgang Baumgärtner
- Department of Pathology, University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
| | - Albert Osterhaus
- Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
- Excellence Cluster 2155 RESIST, Hannover, Germany
- Global Virus Network, Center of Excellence, University of Veterinary Medicine, Hannover, Germany
| | - Thomas F Schulz
- Institute of Virology, Hannover Medical School, Hannover, Germany
- Excellence Cluster 2155 RESIST, Hannover, Germany
- German Center for Infection Research, Partner Site Hannover-Braunschweig, Hannover, Germany
| | - Thomas Krey
- Institute of Virology, Hannover Medical School, Hannover, Germany
- Institute of Biochemistry, Center of Structural and Cell Biology in Medicine, University of Lübeck, Lübeck, Germany
- Excellence Cluster 2155 RESIST, Hannover, Germany
- German Center for Infection Research, Partner Site Hamburg-Lübeck-Borstel-Riems, Hannover, Germany
- Centre for Structural Systems Biology, Hamburg, Germany
| |
Collapse
|
50
|
Bangaru S, Jackson AM, Copps J, Fernández-Quintero ML, Torres JL, Richey ST, Nogal B, Sewall LM, de la Peña AT, Rehman A, Guebre-Xabier M, Girard B, Das R, Corbett-Helaire KS, Seder RA, Graham BS, Edwards DK, Patel N, Smith G, Ward AB. Structural serology of polyclonal antibody responses to mRNA-1273 and NVX-CoV2373 COVID-19 vaccines. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.11.628030. [PMID: 39713412 PMCID: PMC11661243 DOI: 10.1101/2024.12.11.628030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
Current COVID-19 vaccines are largely limited in their ability to induce broad, durable immunity against emerging viral variants. Design and development of improved vaccines utilizing existing platforms requires an in-depth understanding of the antigenic and immunogenic properties of available vaccines. Here we examined the antigenicity of two of the original COVID-19 vaccines, mRNA-1273 and NVX-CoV2373, by electron microscopy-based polyclonal epitope mapping (EMPEM) of serum from immunized non-human primates (NHPs) and clinical trial donors. Both vaccines induce diverse polyclonal antibody (pAb) responses to the N-terminal domain (NTD) in addition to the receptor-binding domain (RBD) of the Spike protein, with the NTD supersite being an immunodominant epitope. High-resolution cryo-EMPEM studies revealed extensive pAb responses to and around the supersite with unique angles of approach and engagement. NTD supersite pAbs were also the most susceptible to variant mutations compared to other specificities, indicating that ongoing Spike ectodomain-based vaccine design strategies should consider immuno-masking this site to prevent induction of these strain-specific responses.
Collapse
Affiliation(s)
- Sandhya Bangaru
- Dept. of Integrative Structural and Computational Biology, The Scripps Research Institute; La Jolla, CA, 92037, USA
| | - Abigail M. Jackson
- Dept. of Integrative Structural and Computational Biology, The Scripps Research Institute; La Jolla, CA, 92037, USA
| | - Jeffrey Copps
- Dept. of Integrative Structural and Computational Biology, The Scripps Research Institute; La Jolla, CA, 92037, USA
| | - Monica L. Fernández-Quintero
- Dept. of Integrative Structural and Computational Biology, The Scripps Research Institute; La Jolla, CA, 92037, USA
| | - Jonathan L. Torres
- Dept. of Integrative Structural and Computational Biology, The Scripps Research Institute; La Jolla, CA, 92037, USA
| | - Sara T. Richey
- Dept. of Integrative Structural and Computational Biology, The Scripps Research Institute; La Jolla, CA, 92037, USA
| | - Bartek Nogal
- Dept. of Integrative Structural and Computational Biology, The Scripps Research Institute; La Jolla, CA, 92037, USA
| | - Leigh M. Sewall
- Dept. of Integrative Structural and Computational Biology, The Scripps Research Institute; La Jolla, CA, 92037, USA
| | - Alba Torrents de la Peña
- Dept. of Integrative Structural and Computational Biology, The Scripps Research Institute; La Jolla, CA, 92037, USA
| | - Asma Rehman
- Novavax, Inc; 21 Firstfield Road, Gaithersburg, MD, 20878, USA
| | | | | | | | - Kizzmekia S. Corbett-Helaire
- Vaccine Research Center; National Institutes of Allergy and Infectious Diseases; National Institutes of Health; Bethesda, Maryland, 20892, USA
- Current affiliation: Department of Immunology and Infectious Diseases; Harvard T.H. Chan School of Public Health; Boston, Massachusetts, 02115, USA
- Current affiliation: Howard Hughes Medical Institute; Chevy Chase, Maryland, 20815, USA
| | - Robert A. Seder
- Vaccine Research Center; National Institutes of Allergy and Infectious Diseases; National Institutes of Health; Bethesda, Maryland, 20892, USA
| | - Barney S. Graham
- Vaccine Research Center; National Institutes of Allergy and Infectious Diseases; National Institutes of Health; Bethesda, Maryland, 20892, USA
- Current affiliation: Department of Microbiology, Biochemistry & Immunology; Morehouse School of Medicine; Atlanta, Georgia, 30310, USA
| | | | - Nita Patel
- Novavax, Inc; 21 Firstfield Road, Gaithersburg, MD, 20878, USA
| | - Gale Smith
- Novavax, Inc; 21 Firstfield Road, Gaithersburg, MD, 20878, USA
| | - Andrew B. Ward
- Dept. of Integrative Structural and Computational Biology, The Scripps Research Institute; La Jolla, CA, 92037, USA
| |
Collapse
|