1
|
Qiao X, Cui M, Yu Z, Ma L, Liu H, Yang X, Chen Y, Li D, Che J, Zhao L, Su R, Ren X, Cen S, Lin B, He X. Thiol esters as chemical warheads of SARS-CoV-2 main protease (3CLpro) peptide-like inhibitors. Eur J Med Chem 2025; 293:117709. [PMID: 40344734 DOI: 10.1016/j.ejmech.2025.117709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 04/27/2025] [Accepted: 04/28/2025] [Indexed: 05/11/2025]
Abstract
Peptide-like 3CLpro covalent binding inhibitors are the most effective antiviral drugs for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. Their covalent warheads were designed based on the addition reaction activity of the aldehyde (ketone) carbonyl or its derivative structures. These addition reactions between the warheads and the thiol of the 3CLpro are reversible, and the resulting hemimonothioacetals are chemically unstable. Herein, after DFT calculation, we designed thiol ester warheads using the principle of ester exchange reaction. Then, the warhead fluorescence probe binding experiment suggested these adducts of thiol ester warheads and 3CLpro protein are more stable than the hemimonothioacetals mentioned earlier. Therefore, new 3CLpro inhibitors were subsequently designed through a structure-based drug design method employing those thiol ester warheads. Those 3CLpro inhibitors demonstrated potent 3CLpro inhibitory activities and anti-coronavirus HCoV-OC43 activities. Among them, B16 stands out as the most promising, demonstrating not only the strongest anti-coronavirus HCoV-OC43 activity but also being a moderate inhibitor of CYP3A4, suggesting that B16 does not require co-administration with ritonavir in the treatment of SARS-CoV-2 infection. This work demonstrates the significant potential of thiol esters as novel chemical warheads in designing covalent binding inhibitors for 3CLpro and beyond.
Collapse
Affiliation(s)
- Xuehong Qiao
- Shenyang Pharmaceutical University, Wenhua Road 103#, Shenyang, 110016, China; Beijing Institute of Pharmacology and Toxicology, Taiping Road 27#, Haidian District, Beijing, 100850, China.
| | - Menghan Cui
- Shenyang Pharmaceutical University, Wenhua Road 103#, Shenyang, 110016, China; Beijing Institute of Pharmacology and Toxicology, Taiping Road 27#, Haidian District, Beijing, 100850, China.
| | - Zhiwei Yu
- Shenyang Pharmaceutical University, Wenhua Road 103#, Shenyang, 110016, China.
| | - Ling Ma
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Science, Tiantan Xili 1#, Dongcheng District, Beijing, 100050, China.
| | - Hailong Liu
- Shenyang Pharmaceutical University, Wenhua Road 103#, Shenyang, 110016, China; Beijing Institute of Pharmacology and Toxicology, Taiping Road 27#, Haidian District, Beijing, 100850, China.
| | - Xingxing Yang
- Shenyang Pharmaceutical University, Wenhua Road 103#, Shenyang, 110016, China; Beijing Institute of Pharmacology and Toxicology, Taiping Road 27#, Haidian District, Beijing, 100850, China.
| | - Yuan Chen
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100039, China.
| | - Dahong Li
- Shenyang Pharmaceutical University, Wenhua Road 103#, Shenyang, 110016, China.
| | - Jinjing Che
- Beijing Institute of Pharmacology and Toxicology, Taiping Road 27#, Haidian District, Beijing, 100850, China.
| | - Linxiang Zhao
- Shenyang Pharmaceutical University, Wenhua Road 103#, Shenyang, 110016, China.
| | - Ruibin Su
- Beijing Institute of Pharmacology and Toxicology, Taiping Road 27#, Haidian District, Beijing, 100850, China.
| | - Xuhong Ren
- Shenyang Pharmaceutical University, Wenhua Road 103#, Shenyang, 110016, China.
| | - Shan Cen
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Science, Tiantan Xili 1#, Dongcheng District, Beijing, 100050, China.
| | - Bin Lin
- Shenyang Pharmaceutical University, Wenhua Road 103#, Shenyang, 110016, China
| | - Xinhua He
- Beijing Institute of Pharmacology and Toxicology, Taiping Road 27#, Haidian District, Beijing, 100850, China; Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100039, China.
| |
Collapse
|
2
|
Singh S, Liu Y, Burke M, Rayaprolu V, Stein SE, Hasan SS. Production and cryo-electron microscopy structure of an internally tagged SARS-CoV-2 spike ecto-domain construct. J Struct Biol X 2025; 11:100123. [PMID: 40046771 PMCID: PMC11880631 DOI: 10.1016/j.yjsbx.2025.100123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 02/10/2025] [Accepted: 02/10/2025] [Indexed: 03/09/2025] Open
Abstract
The SARS-CoV-2 spike protein is synthesized in the endoplasmic reticulum of host cells, from where it undergoes export to the Golgi and the plasma membrane or retrieval from the Golgi to the endoplasmic reticulum. Elucidating the fundamental principles of this bidirectional secretion are pivotal to understanding virus assembly and designing the next generation of spike genetic vaccine with enhanced export properties. However, the widely used strategy of C-terminal affinity tagging of the spike cytosolic tail interferes with proper bidirectional trafficking. Hence, the structural and biophysical investigations of spike protein trafficking have been hindered by a lack of appropriate spike constructs. Here we describe a strategy for the internal tagging of the spike protein. Using sequence analyses and AlphaFold modeling, we identified a site down-stream of the signal sequence for the insertion of a twin-strep-tag, which facilitates purification of an ecto-domain construct from the extra-cellular medium of mammalian Expi293F cells. Mass spectrometry analyses show that the internal tag has minimal impact on N-glycan modifications, which are pivotal for spike-host interactions. Single particle cryo-electron microscopy reconstructions of the spike ecto-domain reveal conformational states compatible for ACE2 receptor interactions, further solidifying the feasibility of the internal tagging strategy. Collectively, these results present a substantial advance towards reagent development for the investigations of spike protein trafficking during coronavirus infection and genetic vaccination.
Collapse
Affiliation(s)
- Suruchi Singh
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore MD 21201, USA
| | - Yi Liu
- Mass Spectrometry Data Center, Biomolecular Measurement Division, National Institute of Standards and Technology, Gaithersburg MD 20899, USA
| | - Meghan Burke
- Mass Spectrometry Data Center, Biomolecular Measurement Division, National Institute of Standards and Technology, Gaithersburg MD 20899, USA
| | - Vamseedhar Rayaprolu
- Pacific Northwest Cryo-EM Center, Oregon Health and Sciences University, Portland, OR 97201, USA
| | - Stephen E. Stein
- Mass Spectrometry Data Center, Biomolecular Measurement Division, National Institute of Standards and Technology, Gaithersburg MD 20899, USA
| | - S. Saif Hasan
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore MD 21201, USA
- University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore MD 21201, USA
- Center for Biomolecular Therapeutics, University of Maryland School of Medicine, Rockville MD 20850, USA
| |
Collapse
|
3
|
Zheng L, Pan T, Wang H, He Z, Tian J. Integrin β3 N125 glycosylation is essential for human cytomegalovirus entry into fibroblasts. Int J Biol Macromol 2025; 313:144322. [PMID: 40383337 DOI: 10.1016/j.ijbiomac.2025.144322] [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: 03/11/2025] [Revised: 04/25/2025] [Accepted: 05/15/2025] [Indexed: 05/20/2025]
Abstract
Human cytomegalovirus (hCMV) infection is highly prevalent worldwide. N-glycosylation of viral receptors is a key factor in early viral infection. Integrin β3 functions as an entry receptor for hCMV infection in fibroblasts; however, the role of integrin β3 N-glycosylation in hCMV entry remains unclear. This study aims to investigate the involvement and mechanism of integrin β3 N-glycosylation in hCMV early infection. The N-glycopeptide profile of recombinant integrin β3 was examined using LC-MS/MS. To assess the effects of specific N-glycosite mutations, viral infection, attachment, and internalization in MRC-5 cells were evaluated through various virological techniques. Moreover, the role of integrin β3 N-glycosylation in receptor-ligand interactions and downstream viral entry signaling pathways was analyzed. Glycomics analysis revealed that integrin β3 N125 mainly contained complex-type glycans, with A2S1G1 as the major glycoform. The N125 mutation in integrin β3 led to a marked reduction in hCMV-induced cytopathic effects, viral DNA load, expression of immediate-early (IE) proteins, and the production of new hCMV particles. Further analysis revealed that this inhibitory effect occurred during the viral entry phase, as the N125 mutation significantly disrupted internalization without affecting viral attachment. Furthermore, the N125 mutation suppressed hCMV glycoprotein H (gH) binding to integrin β3 and inhibited activation of the integrin/Src and RhoA/cofilin signaling pathways. These findings demonstrate that integrin β3 N125 glycosylation is essential for hCMV entry into fibroblasts. More importantly, this study establishes a correlation between hCMV ligand-receptor glycosylation and viral entry signaling pathways, providing novel insights into glycobiological targets for hCMV internalization and potential strategies for antiviral drug and vaccine development.
Collapse
Affiliation(s)
- Luping Zheng
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian 116044, China.
| | - Taowen Pan
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian 116044, China
| | - Huiyi Wang
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian 116044, China
| | - Zeyi He
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian 116044, China
| | - Jiaxu Tian
- The first affiliated hospital of Dalian Medical University, Dalian 116044, China
| |
Collapse
|
4
|
Fang H, Gong T, Su Y, Xiong S, Yao M, Guo Q, Tong W, Gan T, Zhang P, Liu Q, Tan Y, Zhang C, Huang X, Xiong Y. PBS-DLS: A Novel Ultrasensitive Dynamic Light Scattering Immunoassay. ACS APPLIED MATERIALS & INTERFACES 2025; 17:29305-29317. [PMID: 40327829 DOI: 10.1021/acsami.5c03548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2025]
Abstract
Despite significant advances in ultrasensitive detection, current methodologies are often hindered by the need for sophisticated equipment, complex signal amplification processes, and specialized operation. Here, we have developed a novel strategy by universal polyvalent biotin-streptavidin cross-linking aggregation coupled with dynamic light scattering (PBS-DLS) that effectively transduces and amplifies undetected molecular recognition events at low target concentrations, demonstrating its potential application as an ultrasensitive immunoassay. The controllability in the size and quantity of the DLS nanoprobe enables this advanced design to achieve tunable sensitivity down to attomolar levels and a broad detection range spanning six orders of magnitude. By reducing the detection time to approximately 15 min, our PBS-DLS emerges as a promising tool for point-of-care (POC) testing. Moreover, this PBS-DLS immunosensor has been validated through its rapid and ultrasensitive detection of the SARS-CoV-2 nucleocapsid (N) protein (a macromolecular model target) and malachite green (MG, a small molecule model target) in complex sample matrices, outperforming conventional immunoassays and other testing methods. The exceptional sensitivity, simplicity, and speed of this novel approach position it as a highly promising platform for the development of various bioanalytical methods and POC assays.
Collapse
Affiliation(s)
- Hao Fang
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, Nanchang University, Nanchang 330047, P.R. China
| | - Tian Gong
- Jiangxi Medical Academy of Nutrition and Health Management, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, P.R. China
- Center for Molecular Diagnosis and Precision Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330209, P.R. China
- School of Public Health, Jiangxi Provincial Key Laboratory of Preventive Medicine, Nanchang University, Nanchang, Jiangxi 330031, P.R. China
| | - Yu Su
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, Nanchang University, Nanchang 330047, P.R. China
| | - Sicheng Xiong
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, Nanchang University, Nanchang 330047, P.R. China
| | - Mingjian Yao
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, Nanchang University, Nanchang 330047, P.R. China
| | - Qian Guo
- Jiangxi Province Centre for Disease Control and Prevention, Youth Science and Technology Innovation Research Team, Nanchang 330029, P.R. China
| | - Weipeng Tong
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, Nanchang University, Nanchang 330047, P.R. China
| | - Tingting Gan
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, Nanchang University, Nanchang 330047, P.R. China
| | - Peng Zhang
- Jiangxi Medical Academy of Nutrition and Health Management, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, P.R. China
- Center for Molecular Diagnosis and Precision Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330209, P.R. China
- School of Public Health, Jiangxi Provincial Key Laboratory of Preventive Medicine, Nanchang University, Nanchang, Jiangxi 330031, P.R. China
| | - Qiong Liu
- Jiangxi Medical Academy of Nutrition and Health Management, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, P.R. China
- Center for Molecular Diagnosis and Precision Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330209, P.R. China
- School of Public Health, Jiangxi Provincial Key Laboratory of Preventive Medicine, Nanchang University, Nanchang, Jiangxi 330031, P.R. China
| | - Youwen Tan
- Jiangxi Medical Academy of Nutrition and Health Management, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, P.R. China
| | - Chengsheng Zhang
- Jiangxi Medical Academy of Nutrition and Health Management, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, P.R. China
- Center for Molecular Diagnosis and Precision Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330209, P.R. China
- School of Public Health, Jiangxi Provincial Key Laboratory of Preventive Medicine, Nanchang University, Nanchang, Jiangxi 330031, P.R. China
| | - Xiaolin Huang
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, Nanchang University, Nanchang 330047, P.R. China
- Jiangxi Medical Academy of Nutrition and Health Management, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, P.R. China
| | - Yonghua Xiong
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, Nanchang University, Nanchang 330047, P.R. China
- Jiangxi Medical Academy of Nutrition and Health Management, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, P.R. China
- Jiangxi-OAI Joint Research Institute, Nanchang University, Nanchang 330047, P.R. China
| |
Collapse
|
5
|
Tian Y, Yang F, Zargar M, Liu YG, Chen MX, Zhu FY. Integration of structural study and machine learning to elucidate the RNA-SFs interaction atlas in eukaryotic cells. Biotechnol Adv 2025:108608. [PMID: 40398644 DOI: 10.1016/j.biotechadv.2025.108608] [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: 12/29/2024] [Revised: 04/15/2025] [Accepted: 05/18/2025] [Indexed: 05/23/2025]
Abstract
Alternative splicing (AS) occupies a central position in plant growth and development, stress response, and animal growth and disease processes. Mutations in SF (splicing factor) trigger aberrant AS activities that disrupt these fine biological processes. Although cryo electron microscopy (cryoEM) technology has successfully revealed the fine structure of multiple spliceosomes, the dynamic and complex network of RNA-SFs remains to be fully resolved. This review summarizes the binding patterns of RNA and SFs through machine learning's powerful computational capabilities, the deep structural analysis using cryoEM, and experimental validation of RNA protein binding. Connect RNA protein interaction experiments, high-resolution imaging capabilities of cryoEM, and powerful analytical capabilities of machine learning to jointly construct a detailed RNA-SFs interaction map, forming a powerful toolkit. These knowledge help us better understand the complexity and working mechanisms of biological systems. This article not only has profound significance in revealing the molecular mechanisms of diseases and developing multi-target efficient drugs but also provides in-depth insights into molecular breeding and plant resistance enhancement.
Collapse
Affiliation(s)
- Yuan Tian
- Co-Innovation Center for Sustainable Forestry in Southern China, College of Life Sciences, Nanjing Forestry University, Nanjing, China.
| | - Feng Yang
- Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, China
| | - Meisam Zargar
- Department of Agrobiotechnology, Institute of Agriculture, RUDN University, Moscow 117198, Russia
| | - Ying-Gao Liu
- Co-Innovation Center for Sustainable Forestry in Southern China, College of Life Sciences, Nanjing Forestry University, Nanjing, China
| | - Mo-Xian Chen
- Co-Innovation Center for Sustainable Forestry in Southern China, College of Life Sciences, Nanjing Forestry University, Nanjing, China; Department of Agrobiotechnology, Institute of Agriculture, RUDN University, Moscow 117198, Russia
| | - Fu-Yuan Zhu
- Co-Innovation Center for Sustainable Forestry in Southern China, College of Life Sciences, Nanjing Forestry University, Nanjing, China.
| |
Collapse
|
6
|
Loonen S, van Steenis L, Bauer M, Šoštarić N. Phosphorylation Changes SARS-CoV-2 Nucleocapsid Protein's Structural Dynamics and Its Interaction With RNA. Proteins 2025. [PMID: 40375582 DOI: 10.1002/prot.26842] [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: 11/05/2024] [Revised: 05/01/2025] [Accepted: 05/07/2025] [Indexed: 05/18/2025]
Abstract
The SARS-CoV-2 nucleocapsid protein, or N-protein, is a structural protein that plays an important role in the SARS-CoV-2 life cycle. The N-protein takes part in the regulation of viral RNA replication and drives highly specific packaging of full-length genomic RNA prior to virion formation. One regulatory mechanism that is proposed to drive the switch between these two operating modes is the phosphorylation state of the N-protein. Here, we assess the dynamic behavior of non-phosphorylated and phosphorylated versions of the N-protein homodimer through atomistic molecular dynamics simulations. We show that the introduction of phosphorylation yields a more dynamic protein structure and decreases the binding affinity between the N-protein and RNA. Furthermore, we find that secondary structure is essential for the preferential binding of particular RNA elements from the 5' UTR of the viral genome to the N-terminal domain of the N-protein. Altogether, we provide detailed molecular insights into N-protein dynamics, N-protein:RNA interactions, and phosphorylation. Our results corroborate the hypothesis that phosphorylation of the N-protein serves as a regulatory mechanism that determines N-protein function.
Collapse
Affiliation(s)
- Stefan Loonen
- Department of Bionanoscience, Kavli Institute of Nanoscience Delft, Delft University of Technology, Delft, HZ, the Netherlands
| | - Lina van Steenis
- Department of Bionanoscience, Kavli Institute of Nanoscience Delft, Delft University of Technology, Delft, HZ, the Netherlands
| | - Marianne Bauer
- Department of Bionanoscience, Kavli Institute of Nanoscience Delft, Delft University of Technology, Delft, HZ, the Netherlands
| | - Nikolina Šoštarić
- Department of Bionanoscience, Kavli Institute of Nanoscience Delft, Delft University of Technology, Delft, HZ, the Netherlands
| |
Collapse
|
7
|
Aliyari SR, Xie G, Xia X, Wang L, Zhou ZH, Cheng G. Infectivity and structure of SARS-CoV-2 after hydrogen peroxide treatment. mBio 2025; 16:e0399424. [PMID: 40257280 PMCID: PMC12077155 DOI: 10.1128/mbio.03994-24] [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: 01/15/2025] [Accepted: 02/18/2025] [Indexed: 04/22/2025] Open
Abstract
Hydrogen peroxide (H2O2) exhibits broad-spectrum antiviral activity and is commonly used as an over-the-counter disinfecting agent. However, its potential activities against SARS-CoV-2 have not been systematically evaluated, and mechanisms of action are not well understood. In this study, we investigate H2O2's antiviral activity against SARS-CoV-2 infection and its impact on the virion's structural integrity as compared to the commonly used fixative agent paraformaldehyde (PFA). We show that H2O2 rapidly and directly inactivates SARS-CoV-2 with a half-maximal inhibitory concentration (IC50) of 0.0015%. Cryogenic electron tomography (cryo-ET) with subtomogram averaging reveals that treatment with PFA induced the viral trimeric spike protein (S) to adopt a post-fusion conformation, and treatment of viral particles with H2O2 locked S in its pre-fusion conformation. Therefore, H2O2 treatment likely has induced modifications, such as oxidation of cysteine residues within the S subunits of the spike trimer that locked them in their pre-fusion conformation. Locking of the meta-stable pre-fusion trimer prevents its transition to the post-fusion conformation, a process essential for viral fusion with host cells and entry into host cells. Together, our cellular, biochemical, and structural studies established that hydrogen peroxide can inactivate SARS-CoV-2 in tissue culture and uncovered its underlying molecular mechanism.IMPORTANCEHydrogen peroxide (H2O2) is the commonly used, over-the-counter antiseptic solution available in pharmacies, but its effect against the SARS-CoV-2 virus has not been evaluated systematically. In this study, we show that H2O2 inactivates the SARS-CoV-2 infectivity and establish the effective concentration of this activity. Cryogenic electron tomography and sub-tomogram averaging reveal a detailed structural understanding of how H2O2 affects the SARS-CoV-2 spike in comparison with that of the commonly used fixative PFA under identical conditions. We found that PFA promoted a post-fusion conformation of the viral spike protein, while H2O2 could potentially lock the spike in its pre-fusion state. Our findings not only substantiate the disinfectant efficacy of H2O2 as a potent agent against SARS-CoV-2 but also lay the groundwork for future investigations into targeted antiviral therapies that may leverage the virus' structural susceptibilities. In addition, this study may have significant implications for developing new antiviral strategies and improving existing disinfection protocols.
Collapse
Affiliation(s)
- Saba R. Aliyari
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles (UCLA), Los Angeles, California, USA
| | - Guodong Xie
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles (UCLA), Los Angeles, California, USA
- California NanoSystems Institute, UCLA, Los Angeles, California, USA
| | - Xian Xia
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles (UCLA), Los Angeles, California, USA
- California NanoSystems Institute, UCLA, Los Angeles, California, USA
| | - Lulan Wang
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles (UCLA), Los Angeles, California, USA
| | - Z. Hong Zhou
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles (UCLA), Los Angeles, California, USA
- California NanoSystems Institute, UCLA, Los Angeles, California, USA
| | - Genhong Cheng
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles (UCLA), Los Angeles, California, USA
| |
Collapse
|
8
|
Andrew EJ, Davis K, Harris DH, Torres-Díaz I. Topological DLVO Interaction of a Spiky Particle with a Wall. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2025; 41:11515-11525. [PMID: 40310982 DOI: 10.1021/acs.langmuir.5c00592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2025]
Abstract
We report a model to quantify the effects of position and orientation on the Derjaguin-Landau-Verwey-Overbeek (DLVO) interactions between a spiky particle and a planar wall. We model DLVO interaction energy, force, and torque as a function of spike distribution, aspect ratio, particle-wall separation distance, and particle orientation. The results show a topological correlation between the energy tiling and the tessellated orientational space. Furthermore, the particles with small spikes show a divergence in the adhesion energy and force from that at the tessellated boundaries in the orientational space. However, the maximum energy, force, and torque are at orientations corresponding to the tessellated orientational space in large spiked particles. Additionally, our results show that spiky particles have a significant adhesion torque over a planar wall compared to smooth spheres or ellipsoids, notably enhancing their interactions with a planar wall.
Collapse
Affiliation(s)
- Elizabeth J Andrew
- Department of Chemical and Materials Engineering, The University of Alabama in Huntsville, Huntsville, Alabama 35899, United States
| | - KaiLian Davis
- Department of Chemical and Materials Engineering, The University of Alabama in Huntsville, Huntsville, Alabama 35899, United States
| | - David H Harris
- Department of Chemical and Materials Engineering, The University of Alabama in Huntsville, Huntsville, Alabama 35899, United States
| | - Isaac Torres-Díaz
- Department of Chemical and Materials Engineering, The University of Alabama in Huntsville, Huntsville, Alabama 35899, United States
| |
Collapse
|
9
|
Hartmann S, Radochonski L, Ye C, Martinez-Sobrido L, Chen J. SARS-CoV-2 ORF3a drives dynamic dense body formation for optimal viral infectivity. Nat Commun 2025; 16:4393. [PMID: 40355429 PMCID: PMC12069715 DOI: 10.1038/s41467-025-59475-x] [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: 05/08/2024] [Accepted: 04/24/2025] [Indexed: 05/14/2025] Open
Abstract
SARS-CoV-2 hijacks multiple organelles for virion assembly, of which the mechanisms have not been fully understood. Here, we identified a SARS-CoV-2-driven membrane structure named the 3a dense body (3DB). 3DBs are unusual electron-dense and dynamic structures driven by the accessory protein ORF3a via remodeling a specific subset of the trans-Golgi network (TGN) and early endosomal membrane. 3DB formation is conserved in related bat and pangolin coronaviruses but was lost during the evolution to SARS-CoV. During SARS-CoV-2 infection, 3DB recruits the viral structural proteins spike (S) and membrane (M) and undergoes dynamic fusion/fission to maintain the optimal unprocessed-to-processed ratio of S on assembled virions. Disruption of 3DB formation resulted in virions assembled with an abnormal S processing rate, leading to a dramatic reduction in viral entry efficiency. Our study uncovers the crucial role of 3DB in maintaining maximal SARS-CoV-2 infectivity and highlights its potential as a target for COVID-19 prophylactics and therapeutics.
Collapse
Affiliation(s)
- Stella Hartmann
- Department of Microbiology, University of Chicago, Chicago, IL, USA
- Howard Taylor Ricketts Laboratory, University of Chicago, Lemont, IL, USA
| | - Lisa Radochonski
- Department of Microbiology, University of Chicago, Chicago, IL, USA
- Howard Taylor Ricketts Laboratory, University of Chicago, Lemont, IL, USA
| | - Chengjin Ye
- Texas Biomedical Research Institute, San Antonio, TX, USA
| | | | - Jueqi Chen
- Department of Microbiology, University of Chicago, Chicago, IL, USA.
- Howard Taylor Ricketts Laboratory, University of Chicago, Lemont, IL, USA.
| |
Collapse
|
10
|
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
|
11
|
Hachimi A, El-Mansoury B, Merzouki M. Incidence, pathophysiology, risk factors, histopathology, and outcomes of COVID-19-induced acute kidney injury: A narrative review. Microb Pathog 2025; 202:107360. [PMID: 39894232 DOI: 10.1016/j.micpath.2025.107360] [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: 09/03/2024] [Revised: 01/28/2025] [Accepted: 01/30/2025] [Indexed: 02/04/2025]
Abstract
The COVID-19 pandemic, caused by the SARS-CoV-2 virus, has led to a significant burden on global healthcare systems. COVID-19-induced acute kidney injury (AKI) is among one of the complications, that has emerged as a critical and frequent condition in COVID-19 patients. This AKI among COVID-19 patients is associated with poor outcomes, and high mortality rates, especially in those with severe AKI or requiring renal replacement therapy. COVID-19-induced AKI represents a significant complication with complex pathophysiology and multifactorial risk factors. Indeed, several pathophysiological mechanisms, including direct viral invasion of renal cells, systemic inflammation, endothelial and thrombotic abnormalities as well as nephrotoxic drugs and rhabdomyolysis are believed to underlie this condition. Moreover, histopathological and immunohistopathological findings commonly observed in postmortem studies include acute tubular necrosis, glomerular injury, and the presence of viral particles within renal tissue and urine. Identified risk factors for developing AKI vary among studies, depending on regions, underlying conditions, and the severity of the disease. Moreover, histopathological and immunohistopathological findings commonly observed in postmortem studies include show acute tubular necrosis, glomerular injury, and viral particles within renal tissue and urine. While, identified risk factors for developing AKI vary among studies, according to regions, underlying conditions, and the gravity of the disease. This narrative review aims to synthesize current knowledge on the incidence, pathophysiology, risk factors, histopathology, and outcomes of AKI induced by COVID-19.
Collapse
Affiliation(s)
- Abdelhamid Hachimi
- Medical ICU, Mohammed VI(th) University Hospital of Marrakech, Marrakech, Morocco; Morpho-Science Research Laboratory, Faculty of Medicine and Pharmacy, Cadi Ayyad University, Marrakech, Morocco; Life Sciences Department, Bioengineering Laboratory, Faculty of Sciences and Technics, Sultan Moulay Slimane University, Beni Mellal, Morocco
| | - Bilal El-Mansoury
- Nutritional Physiopathologies, Neuroscience and Toxicology Team, Laboratory of Anthropogenic, Biotechnology and Health, Faculty of Sciences, Chouaib Doukkali University, El Jadida, Morocco
| | - Mohamed Merzouki
- Life Sciences Department, Bioengineering Laboratory, Faculty of Sciences and Technics, Sultan Moulay Slimane University, Beni Mellal, Morocco.
| |
Collapse
|
12
|
Liu C, Ke Z. Cryo-ET unravels the mystery of Ad5-nCoV vaccines. Structure 2025; 33:836-837. [PMID: 40315818 DOI: 10.1016/j.str.2025.04.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2025] [Revised: 04/06/2025] [Accepted: 04/07/2025] [Indexed: 05/04/2025]
Abstract
The Ad5-nCoV vaccine (Convidecia) against COVID-19 showed promising clinical results. However, the molecular mechanisms underlying its high immunogenicity and potential adverse reactions have remained elusive. In this issue of Structure, Dong et al.1 employed cryo-electron tomography as a powerful technique to show that abundant prefusion spike protein formation is induced by Ad5-nCoV vaccines.
Collapse
Affiliation(s)
- Chang Liu
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX 78712, USA
| | - Zunlong Ke
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX 78712, USA; LaMontagne Center for Infectious Disease, The University of Texas at Austin, Austin, TX 78712, USA.
| |
Collapse
|
13
|
Eltayeb A, Rubio-Casillas A, Uversky VN, Redwan EM. Intrinsic Factors Behind Long COVID: VI. Combined Impact of G3BPs and SARS-CoV-2 Nucleocapsid Protein on the Viral Persistence and Long COVID. J Cell Biochem 2025; 126:e70038. [PMID: 40415285 DOI: 10.1002/jcb.70038] [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: 04/27/2025] [Revised: 04/27/2025] [Accepted: 05/11/2025] [Indexed: 05/27/2025]
Abstract
The efficient transmission of SARS-CoV-2 caused the COVID-19 pandemic, which affected millions of people around the globe. Despite extensive efforts, specific therapeutic interventions and preventive measures against COVID-19 and its consequences, such as long COVID, have not yet been identified due to the lack of a comprehensive knowledge of the SARS-CoV-2 biology. Therefore, a deeper understanding of the sophisticated strategies employed by SARS-CoV-2 to bypass the host antiviral defense systems is needed. One of these strategies is the inhibition of the Ras GTPase-activating protein-binding protein (GAP SH3-binding protein or G3BP)-dependent host immune response by the SARS-CoV-2 nucleocapsid (N) protein. This inhibition disrupts the formation of stress granules (SGs), which are crucial for antiviral defense. By preventing SG formation, the virus enhances its replication and evades the host's immune response, leading to increased disease severity. Given the involvement of G3BP1 in SG formation and its ability to interact with viral proteins, along with the crucial role of the N protein in the replication of the virus, we hypothesize that these proteins may have a potential role in the pathogenesis of long COVID. Despite the current lack of direct evidence linking these proteins to long COVID, their interactions and functions suggest a possible connection that warrants further investigation.
Collapse
Affiliation(s)
- Ahmed Eltayeb
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Alberto Rubio-Casillas
- Autlan Regional Hospital, Jalisco Health Services, Autlan, Jalisco, Mexico
- Biology Laboratory, Autlan Regional Preparatory School, University of Guadalajara, Autlan, Jalisco, Mexico
| | - Vladimir N Uversky
- Department of Molecular Medicine and USF Health Byrd Alzheimer's Research Institute, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| | - Elrashdy M Redwan
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
- Centre of Excellence in Bionanoscience Research, King Abdulaziz University, Jeddah, Saudi Arabia
- Protein Research Department, Therapeutic and Protective Proteins Laboratory, Genetic Engineering and Biotechnology Research Institute, City for Scientific Research and Technology Applications, New Borg EL-Arab, Alexandria, Egypt
| |
Collapse
|
14
|
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
|
15
|
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
|
16
|
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
|
17
|
Laporte M, Jochmans D, Bardiot D, Desmarets L, Debski-Antoniak OJ, Mizzon G, Abdelnabi R, Leyssen P, Chiu W, Zhang Z, Nomura N, Boland S, Ohto U, Stahl Y, Wuyts J, De Jonghe S, Stevaert A, van Hemert MJ, Bontes BW, Wanningen P, Groenewold GJM, Zegar A, Owczarek K, Joshi S, Koukni M, Arzel P, Klaassen H, Vanherck JC, Vandecaetsbeek I, Cremers N, Donckers K, Francken T, Van Buyten T, Rymenants J, Schepers J, Pyrc K, Hilgenfeld R, Dubuisson J, Bosch BJ, Van Kuppeveld F, Eydoux C, Decroly E, Canard B, Naesens L, Weynand B, Snijder EJ, Belouzard S, Shimizu T, Bartenschlager R, Hurdiss DL, Marchand A, Chaltin P, Neyts J. A coronavirus assembly inhibitor that targets the viral membrane protein. Nature 2025; 640:514-523. [PMID: 40140569 PMCID: PMC11981944 DOI: 10.1038/s41586-025-08773-x] [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] [Received: 04/06/2024] [Accepted: 02/11/2025] [Indexed: 03/28/2025]
Abstract
The coronavirus membrane protein (M) is the main organizer of coronavirus assembly1-3. Here, we report on an M-targeting molecule, CIM-834, that blocks the assembly of SARS-CoV-2. CIM-834 was obtained through high-throughput phenotypic antiviral screening followed by medicinal-chemistry efforts and target elucidation. CIM-834 inhibits the replication of SARS-CoV-2 (including a broad panel of variants) and SARS-CoV. In SCID mice and Syrian hamsters intranasally infected with SARS-CoV-2, oral treatment reduced lung viral titres to nearly undetectable levels, even (as shown in mice) when treatment was delayed until 24 h before the end point. Treatment of infected hamsters prevented transmission to untreated sentinels. Transmission electron microscopy studies show that virion assembly is completely absent in cells treated with CIM-834. Single-particle cryo-electron microscopy reveals that CIM-834 binds and stabilizes the M protein in its short form, thereby preventing the conformational switch to the long form, which is required for successful particle assembly. In conclusion, we have discovered a new druggable target in the replication cycle of coronaviruses and a small molecule that potently inhibits it.
Collapse
Affiliation(s)
- Manon Laporte
- Virology, Antiviral Drug & Vaccine Research Group, Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven, Belgium
| | - Dirk Jochmans
- Virology, Antiviral Drug & Vaccine Research Group, Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven, Belgium
| | | | - Lowiese Desmarets
- CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019-UMR 9017-CIIL-Center for Infection and Immunity of Lille, Université de Lille, Lille, France
| | - Oliver J Debski-Antoniak
- Virology Section, Infectious Diseases and Immunology Division, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Giulia Mizzon
- Department of Infectious Diseases, Molecular Virology, Medical Faculty Heidelberg, University of Heidelberg, Heidelberg, Germany
- German Center for Infection Research (DZIF), Heidelberg Partner Site, Heidelberg, Germany
| | - Rana Abdelnabi
- Virology, Antiviral Drug & Vaccine Research Group, Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven, Belgium
- Department of Microbiology, Immunology and Transplantation, VirusBank Platform, KU Leuven, Leuven, Belgium
| | - Pieter Leyssen
- Virology, Antiviral Drug & Vaccine Research Group, Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven, Belgium
| | - Winston Chiu
- Virology, Antiviral Drug & Vaccine Research Group, Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven, Belgium
| | - Zhikuan Zhang
- Graduate School of Pharmaceutical Sciences, University of Tokyo, Tokyo, Japan
| | - Norimichi Nomura
- Department of Cell Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | | | - Umeharu Ohto
- Graduate School of Pharmaceutical Sciences, University of Tokyo, Tokyo, Japan
| | - Yannick Stahl
- Department of Infectious Diseases, Molecular Virology, Medical Faculty Heidelberg, University of Heidelberg, Heidelberg, Germany
| | | | - Steven De Jonghe
- Structural and Translational Virology Research Group, Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven, Belgium
| | - Annelies Stevaert
- Structural and Translational Virology Research Group, Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven, Belgium
| | - Martijn J van Hemert
- Molecular Virology Laboratory, Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Brenda W Bontes
- Molecular Virology Laboratory, Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Patrick Wanningen
- Molecular Virology Laboratory, Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - G J Mirjam Groenewold
- Molecular Virology Laboratory, Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Aneta Zegar
- Virogenetics Laboratory of Virology, Malopolska Centre of Biotechnology, Jagiellonian University, Krakow, Poland
| | - Katarzyna Owczarek
- Virogenetics Laboratory of Virology, Malopolska Centre of Biotechnology, Jagiellonian University, Krakow, Poland
| | - Sanjata Joshi
- Institute of Molecular Medicine and German Center for Infection Research (DZIF), University of Lübeck, Lübeck, Germany
| | | | | | | | | | | | - Niels Cremers
- Virology, Antiviral Drug & Vaccine Research Group, Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven, Belgium
| | - Kim Donckers
- Virology, Antiviral Drug & Vaccine Research Group, Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven, Belgium
| | - Thibault Francken
- Virology, Antiviral Drug & Vaccine Research Group, Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven, Belgium
| | - Tina Van Buyten
- Virology, Antiviral Drug & Vaccine Research Group, Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven, Belgium
| | - Jasper Rymenants
- Virology, Antiviral Drug & Vaccine Research Group, Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven, Belgium
| | - Joost Schepers
- Virology, Antiviral Drug & Vaccine Research Group, Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven, Belgium
| | - Krzysztof Pyrc
- Virogenetics Laboratory of Virology, Malopolska Centre of Biotechnology, Jagiellonian University, Krakow, Poland
| | - Rolf Hilgenfeld
- Institute of Molecular Medicine and German Center for Infection Research (DZIF), University of Lübeck, Lübeck, Germany
| | - Jean Dubuisson
- CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019-UMR 9017-CIIL-Center for Infection and Immunity of Lille, Université de Lille, Lille, France
| | - Berend-Jan Bosch
- Virology Section, Infectious Diseases and Immunology Division, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Frank Van Kuppeveld
- Virology Section, Infectious Diseases and Immunology Division, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Cecilia Eydoux
- Architecture et Fonction des Macromolécules Biologiques (AFMB), Aix-Marseille Univ., CNRS, Faculté des Sciences Campus Luminy, Marseille, France
| | - Etienne Decroly
- Architecture et Fonction des Macromolécules Biologiques (AFMB), Aix-Marseille Univ., CNRS, Faculté des Sciences Campus Luminy, Marseille, France
| | - Bruno Canard
- Architecture et Fonction des Macromolécules Biologiques (AFMB), Aix-Marseille Univ., CNRS, Faculté des Sciences Campus Luminy, Marseille, France
| | - Lieve Naesens
- Structural and Translational Virology Research Group, Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven, Belgium
| | - Birgit Weynand
- Department of Imaging and Pathology, Division of Translational Cell and Tissue Research, KU Leuven, Leuven, Belgium
| | - Eric J Snijder
- Molecular Virology Laboratory, Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Sandrine Belouzard
- CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019-UMR 9017-CIIL-Center for Infection and Immunity of Lille, Université de Lille, Lille, France
| | - Toshiyuki Shimizu
- Graduate School of Pharmaceutical Sciences, University of Tokyo, Tokyo, Japan
| | - Ralf Bartenschlager
- Department of Infectious Diseases, Molecular Virology, Medical Faculty Heidelberg, University of Heidelberg, Heidelberg, Germany
- German Center for Infection Research (DZIF), Heidelberg Partner Site, Heidelberg, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Daniel L Hurdiss
- Virology Section, Infectious Diseases and Immunology Division, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | | | - Patrick Chaltin
- CISTIM Leuven vzw, Leuven, Belgium
- Centre for Drug Design and Discovery (CD3), KU Leuven, Leuven, Belgium
| | - Johan Neyts
- Virology, Antiviral Drug & Vaccine Research Group, Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven, Belgium.
| |
Collapse
|
18
|
Latham AP, Rožič M, Webb BM, Sali A. Tutorial on integrative spatiotemporal modeling by integrative modeling platform. Protein Sci 2025; 34:e70107. [PMID: 40130765 PMCID: PMC11934212 DOI: 10.1002/pro.70107] [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: 12/05/2024] [Revised: 02/26/2025] [Accepted: 03/09/2025] [Indexed: 03/26/2025]
Abstract
Cells function through dynamic interactions between macromolecules. Detailed characterization of the dynamics of large biomolecular systems is often not feasible by individual biophysical methods. In such cases, it may be possible to compute useful models by integrating multiple sources of information. We have previously developed an integrative method to model dynamic processes by computing biomolecular heterogeneity at fixed time points, then generating static integrative structural modes for each of these heterogeneity models, and finally connecting these static models to produce a scored trajectory model that depicts the process. Here, we demonstrate how to compute, score, and assess these integrative spatiotemporal models using our open-source Integrative Modeling Platform (IMP) program (https://integrativemodeling.org/).
Collapse
Affiliation(s)
- Andrew P. Latham
- Quantitative Biosciences InstituteUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
- Department of Bioengineering and Therapeutic SciencesUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
- Department of Pharmaceutical ChemistryUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
| | - Miha Rožič
- Quantitative Biosciences InstituteUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
- Department of Bioengineering and Therapeutic SciencesUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
- Department of Pharmaceutical ChemistryUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
| | - Benjamin M. Webb
- Quantitative Biosciences InstituteUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
- Department of Bioengineering and Therapeutic SciencesUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
- Department of Pharmaceutical ChemistryUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
| | - Andrej Sali
- Quantitative Biosciences InstituteUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
- Department of Bioengineering and Therapeutic SciencesUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
- Department of Pharmaceutical ChemistryUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
| |
Collapse
|
19
|
Zhang D, Han B, Chen XF, Zhao S, Li WX, Zhang H, Zhang ML, Huo MQ, Qiu YS, Ren YJ, Zhang YD, Ren XQ, Wang W, Tang JF. A Biolayer Interferometry-Based SARS-COV-2 Mpro-Targeted Active Ingredients Recognition System: Construction and Application in Ligand Screening From Herbal Medicines. PHYTOCHEMICAL ANALYSIS : PCA 2025; 36:718-731. [PMID: 39568228 DOI: 10.1002/pca.3462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 10/03/2024] [Accepted: 10/05/2024] [Indexed: 11/22/2024]
Abstract
INTRODUCTION Drug discovery research targeting SARS-CoV-2 and other emerging pathogens remains critically important. Active compounds derived from plants frequently serve as lead compounds for further drug discovery; however, numerous unrelated chemical constituents in crude extracts may obscure the effective ingredients in LC-MS analysis. OBJECTIVE The aim of this study is to construct a biolayer interferometry (BLI)-based system for recognizing active ingredients that inhibit the main protease (Mpro) of SARS-CoV-2 and to identify the active chemical components binding to Mpro from herbal medicines. METHODOLOGY We developed a novel FRET fluorogenic probe by linking the amino acid sequences of the fluorescent proteins Lssmorange and mKate2 (Ls-mK). The interaction between traditional Chinese medicine and Mpro was analyzed using BLI. Ultrahigh performance liquid chromatography coupled with quadrupole time-of-flight mass spectrometry (UPLC-QTOF-MS) was employed to analyze the composition of herbal medicines. RESULTS Fluorescence detection and spectroscopy confirmed the successful construction of an Mpro inhibitor screening system. Lanqin Oral Liquid (LQL) and Gardeniae fructus exhibited strong inhibitory effects on Mpro. Ten compounds were identified from G. fructus extracts; among them, deacetyl asperulosidic acid methyl ester (DAAME) and Gardoside were found to strongly bind to Mpro, with dissociation constants (KD) of 3.41 μM and 801 nM, respectively. The half-maximal inhibitory concentrations (IC50) of DAAME and Gardoside for Mpro were 27.46 and 13.7 μM, respectively. CONCLUSION This study established a functional Mpro inhibitor screening system. Among the 10 components identified from G. fructus that bind to Mpro, DAAME and Gardoside displayed strong binding and inhibitory activity, indicating their potential as lead compounds for inhibiting SARS-CoV-2 viral replication.
Collapse
Affiliation(s)
- Dai Zhang
- Department of Laboratory Medicine, First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, China
| | - Bing Han
- First Affiliated Hospital Henan Provincial Center for Clinical Application, Evaluation, and Transformation of Traditional Chinese Medicine Henan Provincial Center for Safety Evaluation and Risk Control of Traditional Chinese Medicine Henan Provincial Key Laboratory of Traditional Chinese Medicine Clinical Pharmacy, Henan University of Traditional Chinese Medicine, Zhengzhou, China
| | - Xiao-Fei Chen
- First Affiliated Hospital Henan Provincial Center for Clinical Application, Evaluation, and Transformation of Traditional Chinese Medicine Henan Provincial Center for Safety Evaluation and Risk Control of Traditional Chinese Medicine Henan Provincial Key Laboratory of Traditional Chinese Medicine Clinical Pharmacy, Henan University of Traditional Chinese Medicine, Zhengzhou, China
| | - Shuai Zhao
- Henan Provincial Key Laboratory of Pediatric Genes and Metabolic Diseases, Zhengzhou University Affiliated Children's Hospital, Zhengzhou, China
| | - Wei-Xia Li
- First Affiliated Hospital Henan Provincial Center for Clinical Application, Evaluation, and Transformation of Traditional Chinese Medicine Henan Provincial Center for Safety Evaluation and Risk Control of Traditional Chinese Medicine Henan Provincial Key Laboratory of Traditional Chinese Medicine Clinical Pharmacy, Henan University of Traditional Chinese Medicine, Zhengzhou, China
- Department of Pharmacy, First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Hui Zhang
- First Affiliated Hospital Henan Provincial Center for Clinical Application, Evaluation, and Transformation of Traditional Chinese Medicine Henan Provincial Center for Safety Evaluation and Risk Control of Traditional Chinese Medicine Henan Provincial Key Laboratory of Traditional Chinese Medicine Clinical Pharmacy, Henan University of Traditional Chinese Medicine, Zhengzhou, China
| | - Ming-Liang Zhang
- First Affiliated Hospital Henan Provincial Center for Clinical Application, Evaluation, and Transformation of Traditional Chinese Medicine Henan Provincial Center for Safety Evaluation and Risk Control of Traditional Chinese Medicine Henan Provincial Key Laboratory of Traditional Chinese Medicine Clinical Pharmacy, Henan University of Traditional Chinese Medicine, Zhengzhou, China
| | - Meng-Qi Huo
- First Affiliated Hospital Henan Provincial Center for Clinical Application, Evaluation, and Transformation of Traditional Chinese Medicine Henan Provincial Center for Safety Evaluation and Risk Control of Traditional Chinese Medicine Henan Provincial Key Laboratory of Traditional Chinese Medicine Clinical Pharmacy, Henan University of Traditional Chinese Medicine, Zhengzhou, China
| | - Yong-Sheng Qiu
- Department of Anesthesiology, Zhengzhou University Affiliated Children's Hospital, Zhengzhou, China
| | - Ying-Jie Ren
- First Affiliated Hospital Henan Provincial Center for Clinical Application, Evaluation, and Transformation of Traditional Chinese Medicine Henan Provincial Center for Safety Evaluation and Risk Control of Traditional Chinese Medicine Henan Provincial Key Laboratory of Traditional Chinese Medicine Clinical Pharmacy, Henan University of Traditional Chinese Medicine, Zhengzhou, China
| | - Yao-Dong Zhang
- Henan Provincial Key Laboratory of Pediatric Genes and Metabolic Diseases, Zhengzhou University Affiliated Children's Hospital, Zhengzhou, China
| | - Xian-Qing Ren
- Department of Pediatrics, Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Wei Wang
- Department of Infection, First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, China
| | - Jin-Fa Tang
- First Affiliated Hospital Henan Provincial Center for Clinical Application, Evaluation, and Transformation of Traditional Chinese Medicine Henan Provincial Center for Safety Evaluation and Risk Control of Traditional Chinese Medicine Henan Provincial Key Laboratory of Traditional Chinese Medicine Clinical Pharmacy, Henan University of Traditional Chinese Medicine, Zhengzhou, China
- Department of Pharmacy, First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| |
Collapse
|
20
|
Meng S, Hara T, Miura Y, Arao Y, Saito Y, Inoue K, Hirotsu T, Vecchione A, Satoh T, Ishii H. In Vivo Engineered CAR-T Cell Therapy: Lessons Built from COVID-19 mRNA Vaccines. Int J Mol Sci 2025; 26:3119. [PMID: 40243757 PMCID: PMC11988490 DOI: 10.3390/ijms26073119] [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: 02/11/2025] [Revised: 03/13/2025] [Accepted: 03/26/2025] [Indexed: 04/18/2025] Open
Abstract
Chimeric antigen receptor T cell (CAR-T) therapy has revolutionized cancer immunotherapy but continues to face significant challenges that limit its broader application, such as antigen targeting, the tumor microenvironment, and cell persistence, especially in solid tumors. Meanwhile, the global implementation of mRNA vaccines during the COVID-19 pandemic has highlighted the transformative potential of mRNA and lipid nanoparticle (LNP) technologies. These innovations, characterized by their swift development timelines, precise antigen design, and efficient delivery mechanisms, provide a promising framework to address some limitations of CAR-T therapy. Recent advancements, including mRNA-based CAR engineering and optimized LNP delivery, have demonstrated the capacity to enhance CAR-T efficacy, particularly in the context of solid tumors. This review explores how mRNA-LNP technology can drive the development of in vivo engineered CAR-T therapies to address current limitations and discusses future directions, including advancements in mRNA design, LNP optimization, and strategies for improving in vivo CAR-T functionality and safety. By bridging these technological insights, CAR-T therapy may evolve into a versatile and accessible treatment paradigm across diverse oncological landscapes.
Collapse
Grants
- grant nos. 19K22658, 20H00541, 21K19526, 22H03146, 22K19559, 23K19505, 23K18313, 23KK0153, 24K22144, and 16H06279 (PAGS) Ministry of Education, Culture, Sports, Science and Technology
- grant nos. JP23ym0126809 and JP24ym0126809 Japan Agency for Medical Research and Development
- 23-255001 Princess Takamatsu Cancer Research Fund
- G-2024-3-00 IFO Research Communications
- 2024 Oceanic Wellness Foundation
- 2024 Suzuken Memorial Foundation
Collapse
Affiliation(s)
- Sikun Meng
- Department of Medical Data Science, Center of Medical Innovation and Translational Research, Osaka University Graduate School of Medicine, Yamadaoka 2-2, Suita, Osaka 565-0871, Japan
| | - Tomoaki Hara
- Department of Medical Data Science, Center of Medical Innovation and Translational Research, Osaka University Graduate School of Medicine, Yamadaoka 2-2, Suita, Osaka 565-0871, Japan
| | - Yutaka Miura
- Laboratory for Chemistry and Life Science, Institute of Integrated Research, Institute of Science Tokyo, 4259 Nagatsutacho, Midori-ku, Yokohama 226-8501, Japan
| | - Yasuko Arao
- Department of Medical Data Science, Center of Medical Innovation and Translational Research, Osaka University Graduate School of Medicine, Yamadaoka 2-2, Suita, Osaka 565-0871, Japan
| | - Yoshiko Saito
- Department of Medical Data Science, Center of Medical Innovation and Translational Research, Osaka University Graduate School of Medicine, Yamadaoka 2-2, Suita, Osaka 565-0871, Japan
| | - Kana Inoue
- Department of Medical Data Science, Center of Medical Innovation and Translational Research, Osaka University Graduate School of Medicine, Yamadaoka 2-2, Suita, Osaka 565-0871, Japan
| | | | - Andrea Vecchione
- Department of Clinical and Molecular Medicine, University of Rome “Sapienza”, Santo Andrea Hospital, Via di Grottarossa, 1035, 00189 Rome, Italy
| | - Taroh Satoh
- Center for Cancer Genomics and Precision Medicine, Osaka University Hospital, Yamadaoka 2-2, Suita, Osaka 565-0871, Japan
| | - Hideshi Ishii
- Department of Medical Data Science, Center of Medical Innovation and Translational Research, Osaka University Graduate School of Medicine, Yamadaoka 2-2, Suita, Osaka 565-0871, Japan
| |
Collapse
|
21
|
Latham AP, Zhang W, Tempkin JOB, Otsuka S, Ellenberg J, Sali A. Integrative spatiotemporal modeling of biomolecular processes: Application to the assembly of the nuclear pore complex. Proc Natl Acad Sci U S A 2025; 122:e2415674122. [PMID: 40085653 PMCID: PMC11929490 DOI: 10.1073/pnas.2415674122] [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: 08/08/2024] [Accepted: 02/06/2025] [Indexed: 03/16/2025] Open
Abstract
Dynamic processes involving biomolecules are essential for the function of the cell. Here, we introduce an integrative method for computing models of these processes based on multiple heterogeneous sources of information, including time-resolved experimental data and physical models of dynamic processes. First, for each time point, a set of coarse models of compositional and structural heterogeneity is computed (heterogeneity models). Second, for each heterogeneity model, a set of static integrative structure models is computed (a snapshot model). Finally, these snapshot models are selected and connected into a series of trajectories that optimize the likelihood of both the snapshot models and transitions between them (a trajectory model). The method is demonstrated by application to the assembly process of the human nuclear pore complex in the context of the reforming nuclear envelope during mitotic cell division, based on live-cell correlated electron tomography, bulk fluorescence correlation spectroscopy-calibrated quantitative live imaging, and a structural model of the fully assembled nuclear pore complex. Modeling of the assembly process improves the model precision over static integrative structure modeling alone. The method is applicable to a wide range of time-dependent systems in cell biology and is available to the broader scientific community through an implementation in the open source Integrative Modeling Platform (IMP) software.
Collapse
Affiliation(s)
- Andrew P. Latham
- Department of Bioengineering and Therapeutic Sciences, Quantitative Biosciences Institute, University of California, San Francisco, CA94143
- Department of Pharmaceutical Chemistry, Quantitative Biosciences Institute, University of California, San Francisco, CA94143
| | - Wanlu Zhang
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, Heidelberg69117, Germany
| | - Jeremy O. B. Tempkin
- Department of Bioengineering and Therapeutic Sciences, Quantitative Biosciences Institute, University of California, San Francisco, CA94143
- Department of Pharmaceutical Chemistry, Quantitative Biosciences Institute, University of California, San Francisco, CA94143
| | - Shotaro Otsuka
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, Heidelberg69117, Germany
| | - Jan Ellenberg
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, Heidelberg69117, Germany
| | - Andrej Sali
- Department of Bioengineering and Therapeutic Sciences, Quantitative Biosciences Institute, University of California, San Francisco, CA94143
- Department of Pharmaceutical Chemistry, Quantitative Biosciences Institute, University of California, San Francisco, CA94143
| |
Collapse
|
22
|
Conca DV, Bano F, Graul M, von Wirén J, Scherrer L, Pace H, Sharma H, Svirelis J, Thorsteinsson K, Dahlin A, Bally M. Variant-Specific Interactions at the Plasma Membrane: Heparan Sulfate's Impact on SARS-CoV-2 Binding Kinetics. Anal Chem 2025; 97:4318-4328. [PMID: 39976108 PMCID: PMC11883730 DOI: 10.1021/acs.analchem.4c04283] [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: 08/12/2024] [Revised: 01/16/2025] [Accepted: 02/10/2025] [Indexed: 02/21/2025]
Abstract
The spread of SARS-CoV-2 led to the emergence of several variants of concern (VOCs). The spike glycoprotein, responsible for engaging the viral receptor, exhibits the highest density of mutations, suggesting an ongoing evolution to optimize viral entry. This study characterizes the bond formed by virion mimics carrying the SARS-CoV-2 spike protein and the plasma membrane of host cells in the early stages of virus entry. Contrary to the traditional analysis of isolated ligand-receptor pairs, we utilized well-defined biomimetic models and biochemical and biophysical techniques to characterize the multivalent interaction of VOCs with the complex cell membrane. We observed an overall increase in the binding affinity for newer VOCs. By progressively reducing the system complexity, we identify heparan sulfate (HS) as a main driver of this variation, with a 10-fold increase in affinity for Omicron BA.1 over that of the original strain. These results demonstrate the essential role of coreceptors, particularly HS, in the modulation of SARS-CoV-2 infection and highlight the importance of multiscale biophysical and biochemical assays that account for membrane complexity to fully characterize and understand the role of molecular components and their synergy in viral attachment and entry.
Collapse
Affiliation(s)
- Dario Valter Conca
- Department
of Clinical Microbiology, Umeå University, Umeå 901 87, Sweden
- Wallenberg
Centre for Molecular Medicine (WCMM), Umeå
University, Umeå 901 87, Sweden
- Umeå
Centre for Microbial Research (UCMR), Umeå
University, Umeå 901 87, Sweden
| | - Fouzia Bano
- Department
of Clinical Microbiology, Umeå University, Umeå 901 87, Sweden
- Wallenberg
Centre for Molecular Medicine (WCMM), Umeå
University, Umeå 901 87, Sweden
- Umeå
Centre for Microbial Research (UCMR), Umeå
University, Umeå 901 87, Sweden
| | - Małgorzata Graul
- Department
of Clinical Microbiology, Umeå University, Umeå 901 87, Sweden
- Wallenberg
Centre for Molecular Medicine (WCMM), Umeå
University, Umeå 901 87, Sweden
- Umeå
Centre for Microbial Research (UCMR), Umeå
University, Umeå 901 87, Sweden
| | - Julius von Wirén
- Department
of Clinical Microbiology, Umeå University, Umeå 901 87, Sweden
- Wallenberg
Centre for Molecular Medicine (WCMM), Umeå
University, Umeå 901 87, Sweden
- Umeå
Centre for Microbial Research (UCMR), Umeå
University, Umeå 901 87, Sweden
| | - Lauriane Scherrer
- Department
of Clinical Microbiology, Umeå University, Umeå 901 87, Sweden
- Wallenberg
Centre for Molecular Medicine (WCMM), Umeå
University, Umeå 901 87, Sweden
- Umeå
Centre for Microbial Research (UCMR), Umeå
University, Umeå 901 87, Sweden
| | - Hudson Pace
- Department
of Clinical Microbiology, Umeå University, Umeå 901 87, Sweden
- Wallenberg
Centre for Molecular Medicine (WCMM), Umeå
University, Umeå 901 87, Sweden
- Umeå
Centre for Microbial Research (UCMR), Umeå
University, Umeå 901 87, Sweden
| | - Himanshu Sharma
- Wallenberg
Centre for Molecular Medicine (WCMM), Umeå
University, Umeå 901 87, Sweden
- Umeå
Centre for Microbial Research (UCMR), Umeå
University, Umeå 901 87, Sweden
- Department
of Medical Biochemistry and Biophysics, Umeå University, Umeå 901 87, Sweden
- Laboratory
for Molecular Infection Medicine Sweden (MIMS), Umeå University, Umeå 901 87, Sweden
| | - Justas Svirelis
- Department
of Chemistry and Chemical Engineering, Chalmers
University of Technology, Gothenburg 412 96, Sweden
| | - Konrad Thorsteinsson
- Department
of Clinical Microbiology, Umeå University, Umeå 901 87, Sweden
- Wallenberg
Centre for Molecular Medicine (WCMM), Umeå
University, Umeå 901 87, Sweden
- Umeå
Centre for Microbial Research (UCMR), Umeå
University, Umeå 901 87, Sweden
| | - Andreas Dahlin
- Department
of Chemistry and Chemical Engineering, Chalmers
University of Technology, Gothenburg 412 96, Sweden
| | - Marta Bally
- Department
of Clinical Microbiology, Umeå University, Umeå 901 87, Sweden
- Wallenberg
Centre for Molecular Medicine (WCMM), Umeå
University, Umeå 901 87, Sweden
- Umeå
Centre for Microbial Research (UCMR), Umeå
University, Umeå 901 87, Sweden
| |
Collapse
|
23
|
Cortés Rodríguez FJ, Frattini G, Phloi-Montri S, Pinto Meireles FT, Terrien DA, Cruz-León S, Dal Peraro M, Schier E, Lindorff-Larsen K, Limpanuparb T, Moreno DM, Abriata LA. MolecularWebXR: Multiuser discussions in chemistry and biology through immersive and inclusive augmented and virtual reality. J Mol Graph Model 2025; 135:108932. [PMID: 39719805 DOI: 10.1016/j.jmgm.2024.108932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 12/14/2024] [Accepted: 12/19/2024] [Indexed: 12/26/2024]
Abstract
MolecularWebXR is a new web-based platform for education, science communication and scientific peer discussion in chemistry and biology, based on modern web-based Virtual Reality (VR) and Augmented Reality (AR). With no installs as it is all web-served, MolecularWebXR enables multiple users to simultaneously explore, communicate and discuss concepts about chemistry and biology in immersive 3D environments, by manipulating and passing around objects with their bare hands and pointing at different elements with natural hand gestures. Users may either be present in the same physical space or distributed around the world, in the latter case talking naturally with each other thanks to built-in audio. While MolecularWebXR offers the most immersive experience on high-end AR/VR headsets, its WebXR core also supports participation on consumer devices such as smartphones (with optional cardboard goggles for enhanced immersion), computers, and tablets. MolecularWebXR includes preset VR rooms covering topics in general, inorganic, and organic chemistry, as well as biophysics, structural biology, and general biology. Users can also add new content via the PDB2AR tool. We demonstrate MolecularWebXR's versatility and ease of use across a wide age range (12-80) in fully virtual and mixed real-virtual sessions at science outreach events, undergraduate and graduate courses, scientific collaborations, and conference presentations. MolecularWebXR is available for free use without registration at https://molecularwebxr.org. A blog post version of this preprint with embedded videos is available at https://go.epfl.ch/molecularwebxr-blog-post.
Collapse
Affiliation(s)
| | - Gianfranco Frattini
- Instituto de Química Rosario (IQUIR, CONICET-UNR) and Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Rosario, Santa Fe, Argentina
| | - Sittha Phloi-Montri
- Mahidol University International College, Mahidol University, Salaya, 73170, Thailand
| | | | - Danaé A Terrien
- School of Life Sciences, École Polytechnique Fédérale de Lausanne, CH-1015, Switzerland
| | - Sergio Cruz-León
- Department of Theoretical Biophysics, Max Planck Institute of Biophysics, Frankfurt am Main, Germany
| | - Matteo Dal Peraro
- School of Life Sciences, École Polytechnique Fédérale de Lausanne, CH-1015, Switzerland
| | - Eva Schier
- School of Life Sciences, École Polytechnique Fédérale de Lausanne, CH-1015, Switzerland
| | - Kresten Lindorff-Larsen
- Linderstrøm-Lang Centre for Protein Science, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Taweetham Limpanuparb
- Mahidol University International College, Mahidol University, Salaya, 73170, Thailand
| | - Diego M Moreno
- Instituto de Química Rosario (IQUIR, CONICET-UNR) and Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Rosario, Santa Fe, Argentina
| | - Luciano A Abriata
- School of Life Sciences, École Polytechnique Fédérale de Lausanne, CH-1015, Switzerland.
| |
Collapse
|
24
|
Xu M, Zhang Z, Sun Y, Mai H, Liu S, Liu S, Lv K, Yu F, Wang Y, Yue X, Zhang J, Cai X, Zhao R, Lu H, Liu L, Luo H, Zhao H, Wang Y, Gong P, Chen S, Jing X, Zhao J, Chen YQ. IgA class switching enhances neutralizing potency against SARS-CoV-2 by increased antibody hinge flexibility. Antiviral Res 2025; 235:106082. [PMID: 39828085 DOI: 10.1016/j.antiviral.2025.106082] [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: 09/29/2024] [Revised: 01/14/2025] [Accepted: 01/14/2025] [Indexed: 01/22/2025]
Abstract
IgA antibodies are critical components of the mucosal immune barrier, providing essential first-line defense against viral infections. In this study, we investigated the impact of antibody class switching on neutralization efficacy by engineering recombinant antibodies of different isotypes (IgA1, IgG1) with identical variable regions from SARS-CoV-2 convalescent patients. A potent, broad-spectrum neutralizing monoclonal antibody CAV-C65 exhibited a ten-fold increase in neutralization potency upon switching from IgG1 to IgA1 monomer. Structural analysis revealed that this antibody binds to two adjacent receptor binding domains on the spike protein. Enhanced neutralization by IgA1 was attributed to the combined effects of increased affinity, unique hinge region properties, and potential cross-linking of viral particles. Inhaled CAV-C65 IgA1 demonstrated prophylactic efficacy against lethal SARS-CoV-2 infection in hACE2 mice. These findings highlight the pivotal role of IgA in antiviral immunity and inform the development of IgA-based therapeutics.
Collapse
MESH Headings
- Antibodies, Neutralizing/immunology
- Antibodies, Neutralizing/chemistry
- Antibodies, Neutralizing/genetics
- Animals
- SARS-CoV-2/immunology
- Immunoglobulin A/immunology
- Immunoglobulin A/genetics
- Immunoglobulin A/chemistry
- Antibodies, Viral/immunology
- Antibodies, Viral/chemistry
- Antibodies, Viral/genetics
- Humans
- COVID-19/immunology
- COVID-19/prevention & control
- Mice
- Spike Glycoprotein, Coronavirus/immunology
- Spike Glycoprotein, Coronavirus/chemistry
- Immunoglobulin G/immunology
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal/chemistry
- Immunoglobulin Class Switching/immunology
- Female
- Neutralization Tests
- Mice, Inbred BALB C
Collapse
Affiliation(s)
- Mengxin Xu
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen, China
| | - Zhaoyong Zhang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yuzhu Sun
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen, China
| | - Haoting Mai
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen, China
| | - Siqi Liu
- Molecular Imaging Center, the Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, the Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong, 519000, China
| | - Shuning Liu
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen, China
| | - Kexin Lv
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen, China
| | - Feiyang Yu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, Hubei, China
| | - Yuanyuan Wang
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen, China
| | - Xinyu Yue
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen, China
| | - Jiayi Zhang
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen, China
| | - Xiaoyu Cai
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen, China
| | - Ruixin Zhao
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen, China
| | - Hongjie Lu
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen, China
| | - Lin Liu
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen, China
| | - Huanle Luo
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen, China
| | - Haiyan Zhao
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, Hubei, China
| | - Yanqun Wang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China; GMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China; Clinical Laboratory Medicine Department, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Peng Gong
- Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, No.262 Jin Long Street, Wuhan, Hubei 430207, China
| | - Shoudeng Chen
- Molecular Imaging Center, the Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, the Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong, 519000, China; Central Laboratory, the Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong, 519000, China.
| | - Xuping Jing
- Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, No.262 Jin Long Street, Wuhan, Hubei 430207, China.
| | - Jincun Zhao
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China; Shanghai Institute for Advanced Immunochemical Studies, School of Life Science and Technology, ShanghaiTech University, Shanghai, China; Guangzhou National Laboratory, Bio-Island, Guangzhou, China; Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital, the Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, China.
| | - Yao-Qing Chen
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Shenzhen, China; Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, China; Shenzhen Key Laboratory of Pathogenic Microbes and Biosafety, Shenzhen, China.
| |
Collapse
|
25
|
Farci D, Graça AT, Hall M, Haniewicz P, Kereïche S, Faull P, Kirkpatrick J, Tramontano E, Schröder WP, Piano D. Characterization of SARS-CoV-2 nucleocapsid protein oligomers. J Struct Biol 2025; 217:108162. [PMID: 39675446 DOI: 10.1016/j.jsb.2024.108162] [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: 08/25/2024] [Revised: 11/05/2024] [Accepted: 12/12/2024] [Indexed: 12/17/2024]
Abstract
Oligomers of the SARS-CoV-2 nucleocapsid (N) protein are characterized by pronounced instability resulting in fast degradation. This property likely relates to two contrasting behaviors of the N protein: genome stabilization through a compact nucleocapsid during cell evasion and genome release by nucleocapsid disassembling during infection. In vivo, the N protein forms rounded complexes of high molecular mass from its interaction with the viral genome. To study the N protein and understand its instability, we analyzed degradation profiles under different conditions by size-exclusion chromatography and characterized samples by mass spectrometry and cryo-electron microscopy. We identified self-cleavage properties of the N protein based on specific Proprotein convertases activities, with Cl- playing a key role in modulating stability and degradation. These findings allowed isolation of a stable oligomeric complex of N, for which we report the 3D structure at ∼6.8 Å resolution. Findings are discussed considering available knowledge about the coronaviruses' infection cycle.
Collapse
Affiliation(s)
- Domenica Farci
- Department of Plant Physiology, Institute of Biology, Warsaw University of Life Sciences - SGGW, Warsaw, Poland; Department of Chemistry, Umeå University, Umeå, Sweden; Laboratory of Plant Physiology and Photobiology, Department of Life and Environmental Sciences, University of Cagliari, Cagliari, Italy; ReGenFix Laboratories, R&D Department, Sardara, Italy.
| | - André T Graça
- Department of Chemistry, Umeå University, Umeå, Sweden
| | - Michael Hall
- Department of Chemistry, Umeå University, Umeå, Sweden
| | - Patrycja Haniewicz
- Department of Plant Physiology, Institute of Biology, Warsaw University of Life Sciences - SGGW, Warsaw, Poland
| | - Sami Kereïche
- Institute of Biology and Medical Genetics, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Peter Faull
- The Francis Crick Institute, London, United Kingdom; Proteomics Facility, University of Texas at Austin, Austin, USA
| | | | - Enzo Tramontano
- Laboratory of Molecular Virology, Department of Life and Environmental Sciences, University of Cagliari, Cagliari, Italy
| | | | - Dario Piano
- Department of Plant Physiology, Institute of Biology, Warsaw University of Life Sciences - SGGW, Warsaw, Poland; Laboratory of Plant Physiology and Photobiology, Department of Life and Environmental Sciences, University of Cagliari, Cagliari, Italy; ReGenFix Laboratories, R&D Department, Sardara, Italy.
| |
Collapse
|
26
|
Wang J, Qian Q, Jiang Y, Liang Z, Peng Y, Zhao W, Yang Y, Shen C. Design and Characterization of Bispecific and Trispecific Antibodies Targeting SARS-CoV-2. Vaccines (Basel) 2025; 13:255. [PMID: 40266148 PMCID: PMC11946630 DOI: 10.3390/vaccines13030255] [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: 01/16/2025] [Revised: 02/24/2025] [Accepted: 02/25/2025] [Indexed: 04/24/2025] Open
Abstract
BACKGROUND/OBJECTIVES COVID-19, caused by SARS-CoV-2, has emerged as a global pandemic since its outbreak in 2019. As an increasing number of variants have emerged, especially concerning variants such as Omicron BA.1, BA.2, XBB.1, EG.5, which can escape the immune system and cause repeated infections, they have exerted significant pressure on monoclonal antibodies and the treatment approaches for COVID-19. Broad spectrum antiviral medication was urgently needed. In this study, we developed several bispecific antibodies based on the IgG-scFv format and one trispecific antibody containing Fab fragments with different anti-virus mechanisms studied previously. The Fab fragments are from h11B11, S2P6, and S309 respectively. METHOD all recombinant antibodies were expressed by HEK 293. The pseudoviruses' neutralization assay and the virus challenge to BALB/c mice were deployed to assess the efficiency of recombinant antibodies in vitro and in vivo. RESULTS the bispecific antibodies exhibited a favorable pseudoviruses neutralization activity, with IC50 values ranging from 8 to 591 ng/mL. The trispecific antibody performed even better, with IC50 values ranging from 5 to 27 ng/mL. Furthermore, the virus challenge to mice confirmed that the bispecific antibodies, including the trispecific antibody, had decent therapeutic efficacy. CONCLUSIONS our study provided several supplements to the therapeutic measures of COVID-19 based on multispecific antibodies, supporting the great potential of the multispecific antibodies strategy in dealing with emerging pathogens.
Collapse
Affiliation(s)
- Jiayang Wang
- BSL-3 Laboratory (Guangdong), Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou 510515, China; (J.W.); (Y.J.); (Z.L.); (W.Z.)
| | - Qi Qian
- National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, Second Hospital Affiliated to Southern University of Science and Technology, Shenzhen 518112, China; (Q.Q.); (Y.P.)
| | - Yushan Jiang
- BSL-3 Laboratory (Guangdong), Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou 510515, China; (J.W.); (Y.J.); (Z.L.); (W.Z.)
| | - Zuxin Liang
- BSL-3 Laboratory (Guangdong), Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou 510515, China; (J.W.); (Y.J.); (Z.L.); (W.Z.)
| | - Yun Peng
- National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, Second Hospital Affiliated to Southern University of Science and Technology, Shenzhen 518112, China; (Q.Q.); (Y.P.)
| | - Wei Zhao
- BSL-3 Laboratory (Guangdong), Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou 510515, China; (J.W.); (Y.J.); (Z.L.); (W.Z.)
| | - Yang Yang
- National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, Second Hospital Affiliated to Southern University of Science and Technology, Shenzhen 518112, China; (Q.Q.); (Y.P.)
| | - Chenguang Shen
- BSL-3 Laboratory (Guangdong), Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou 510515, China; (J.W.); (Y.J.); (Z.L.); (W.Z.)
- Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510515, China
- Key Laboratory of Infectious Diseases Research in South China, Southern Medical University, Ministry of Education, Guangzhou 510515, China
| |
Collapse
|
27
|
Tsai YX, Chien YC, Hsu MF, Khoo KH, Hsu STD. Molecular basis of host recognition of human coronavirus 229E. Nat Commun 2025; 16:2045. [PMID: 40016196 PMCID: PMC11868633 DOI: 10.1038/s41467-025-57359-8] [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: 10/28/2024] [Accepted: 02/20/2025] [Indexed: 03/01/2025] Open
Abstract
Human coronavirus 229E (HCoV-229E) is the earliest CoV found to infect humans. It binds to the human aminopeptidase N (hAPN) through the receptor binding domain (RBD) of its spike (S) protein to achieve host recognition. We present the cryo-electron microscopy structure of two HCoV-229E S protein in complex with a dimeric hAPN to provide structural insights on how the HCoV-229E S protein opens up its RBD to engage with its host receptor, information that is currently missing among alphacoronaviruses to which HCoV-229E belong. We quantitatively profile the glycosylation of HCoV-229E S protein and hAPN to deduce the glyco-shielding effects pertinent to antigenicity and host recognition. Finally, we present an atomic model of fully glycosylated HCoV-229E S in complex with hAPN anchored on their respective membrane bilayers to recapitulate the structural basis of the first step of host infection by HCoV-229E.
Collapse
Affiliation(s)
- Yu-Xi Tsai
- Institute of Biological Chemistry, Academia Sinica, Taipei, 11529, Taiwan
- Institute of Biochemical Sciences, National Taiwan University, Taipei, 10617, Taiwan
| | - Yu-Chun Chien
- Institute of Biological Chemistry, Academia Sinica, Taipei, 11529, Taiwan
- Institute of Biochemical Sciences, National Taiwan University, Taipei, 10617, Taiwan
| | - Min-Feng Hsu
- Institute of Biological Chemistry, Academia Sinica, Taipei, 11529, Taiwan
| | - Kay-Hooi Khoo
- Institute of Biological Chemistry, Academia Sinica, Taipei, 11529, Taiwan
- Institute of Biochemical Sciences, National Taiwan University, Taipei, 10617, Taiwan
| | - Shang-Te Danny Hsu
- Institute of Biological Chemistry, Academia Sinica, Taipei, 11529, Taiwan.
- Institute of Biochemical Sciences, National Taiwan University, Taipei, 10617, Taiwan.
- International Institute for Sustainability with Knotted Chiral Meta Matter (WPI-SKCM²), Hiroshima University, 1-3-1 Kagamiyama, Higashi-Hiroshima, Hiroshima, 739-8526, Japan.
| |
Collapse
|
28
|
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
|
29
|
Wang Y, Xia B, Gao Z. A comprehensive review of current insights into the virulence factors of SARS-CoV-2. J Virol 2025; 99:e0204924. [PMID: 39878471 PMCID: PMC11852741 DOI: 10.1128/jvi.02049-24] [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] [Indexed: 01/31/2025] Open
Abstract
The evolution of SARS-CoV-2 pathogenicity has been a major focus of attention. However, the determinants of pathogenicity are still unclear. Various hypotheses have attempted to elucidate the mechanisms underlying the evolution of viral pathogenicity, but a definitive conclusion has yet to be reached. Here, we review the potential impact of all proteins in SARS-CoV-2 on the viral pathogenic process and analyze the effects of their mutations on pathogenicity evolution. We aim to summarize which virus-encoded proteins are crucial in influencing viral pathogenicity, defined as disease severity following infection. Mutations in these key proteins, which are the virulence factors in SARS-CoV-2, may be the driving forces behind the evolution of viral pathogenicity. Mutations in the S protein can impact viral entry and fusogenicity. Mutations in proteins such as NSP2, NSP5, NSP14, and ORF7a can alter the virus's ability to suppress host protein synthesis and innate immunity. Mutations in NSP3, NSP4, NSP6, N protein, NSP5, and NSP12 may alter viral replication efficiency. The combined effects of mutations in the S protein and NSP6 can significantly reduce viral replication. In addition, various viral proteins, including ORF3a, ORF8, NSP4, Spike protein, N protein, and E protein, directly participate in the inflammatory process. Mutations in these proteins can modulate the levels of inflammation following infection. Collectively, these viral protein mutations can influence SARS-CoV-2 pathogenicity by impacting viral immune evasion, replication capacity, and the level of inflammation mediated by infection. In conclusion, the evolution of SARS-CoV-2 pathogenicity is likely determined by multiple virulence factors.
Collapse
Affiliation(s)
- Yi Wang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Bingqing Xia
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Zhaobing Gao
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
30
|
Li D, Zhai J, Wang K, Shen Y, Huang X. Three-Dimensional Reconstruction-Characterization of Polymeric Membranes: A Review. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2025; 59:2891-2916. [PMID: 39913944 DOI: 10.1021/acs.est.4c09734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/19/2025]
Abstract
Polymeric membranes serve as vital separation materials in diverse energy and environmental applications. A comprehensive understanding of three-dimensional (3D) structures of membranes is critical to performance evaluation and future design. Such quantitative 3D structural information is beyond the limit of most employed conventional two-dimentional characterization techniques such as scanning electron microscopy. In this review, we summarize eight types of 3D reconstruction-characterization techniques for membrane materials. Originated from life and materials science, these techniques have been optimized to reveal the 3D structures of membrane materials in the separation field. We systematically introduce the theories of each technique, summarize the sample preparation procedures developed for membrane materials, and demonstrate step-by-step data processing, including 3D model reconstruction and subsequent characterization. Representative case studies are introduced to show the progress of this field and how technical challenges have been overcome over the years. In the end, we share our perspectives and believe that this review can serve as a useful reference for 3D reconstruction-characterization techniques developed for membrane materials.
Collapse
Affiliation(s)
- Danyang Li
- State Key Joint Laboratory of Environment Simulation and Pollution Control, School of Environment and International Joint Laboratory on Low Carbon Clean Energy Innovation, Tsinghua University, Beijing 100084, China
| | - Juan Zhai
- Department of Civil, Environmental, and Construction Engineering, Texas Tech University, Lubbock, Texas 79409, United States
| | - Kunpeng Wang
- State Key Joint Laboratory of Environment Simulation and Pollution Control, School of Environment and International Joint Laboratory on Low Carbon Clean Energy Innovation, Tsinghua University, Beijing 100084, China
| | - Yuexiao Shen
- Department of Civil, Environmental, and Construction Engineering, Texas Tech University, Lubbock, Texas 79409, United States
| | - Xia Huang
- State Key Joint Laboratory of Environment Simulation and Pollution Control, School of Environment and International Joint Laboratory on Low Carbon Clean Energy Innovation, Tsinghua University, Beijing 100084, China
- Research and Application Center for Membrane Technology, School of Environment, Tsinghua University, Beijing 100084, China
| |
Collapse
|
31
|
Xia Q, Zhou M, Jiao K, Li B, Guo L, Wang L, Li J. Recent Advances in DNA-Templated Protein Patterning. SMALL METHODS 2025:e2401835. [PMID: 39895184 DOI: 10.1002/smtd.202401835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 01/13/2025] [Indexed: 02/04/2025]
Abstract
In recent decades, the advancement of DNA nanotechnology enables precise nanoscale organization of diverse functional materials with DNA templates. Particularly, a variety of DNA-templated protein patterns are constructed as powerful tools for programming biomimetic protein complexes. In this review, recent progress in DNA-templated protein patterning, including cutting-edge methods for arranging proteins with DNA templates, and protein patterns across varying dimensions are briefly summarized. Representative applications in biological analysis and biomedicine are discussed. DNA-protein patterns with programmable dynamics, which hold promise in precision diagnosis and therapeutics are highlighted. Finally, current challenges and opportunities in the fabrication and application of DNA-templated protein pattering are discussed.
Collapse
Affiliation(s)
- Qinglin Xia
- Division of Physical Biology, CAS Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai, 201800, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Mo Zhou
- Division of Physical Biology, CAS Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai, 201800, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
- Zhangjiang Laboratory, 100 Haike Road, Shanghai, 201210, China
| | - Kai Jiao
- Institute of Materiobiology, College of Sciences, Shanghai University, Shanghai, 200444, China
| | - Bin Li
- Division of Physical Biology, CAS Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai, 201800, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Linjie Guo
- Institute of Materiobiology, College of Sciences, Shanghai University, Shanghai, 200444, China
| | - Lihua Wang
- Institute of Materiobiology, College of Sciences, Shanghai University, Shanghai, 200444, China
| | - Jiang Li
- Institute of Materiobiology, College of Sciences, Shanghai University, Shanghai, 200444, China
| |
Collapse
|
32
|
Zheng L, Wang S. Recent advances in solid-state nuclear magnetic resonance studies on membrane fusion proteins. FEBS J 2025; 292:483-499. [PMID: 39552293 DOI: 10.1111/febs.17313] [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: 07/30/2024] [Revised: 09/13/2024] [Accepted: 10/17/2024] [Indexed: 11/19/2024]
Abstract
Membrane fusion is an essential biological process that merges two separate lipid bilayers into a whole one. Membrane fusion proteins facilitate this process by bringing lipid bilayers in close proximity to reduce the repulsive energy between membranes. Along with their interactions with membranes, the structures and dynamics of membrane fusion proteins are key to elucidating the mechanisms of membrane fusion. Solid-state NMR (SSNMR) spectroscopy has unique advantages in determining the structures and dynamics of membrane fusion proteins in their membrane-bound states. It has been extensively applied to reveal conformational changes in intermediate states of viral membrane fusion proteins and to characterize the critical lipid-membrane interactions that drive the fusion process. In this review, we summarize recent advancements in SSNMR techniques for studying membrane fusion proteins and their applications in elucidating the mechanisms of membrane fusion.
Collapse
Affiliation(s)
- Lifen Zheng
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
| | - Shenlin Wang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
| |
Collapse
|
33
|
Nankivell BJ, P'ng C, Tran T, Draper J, Ko D, Luu I, Basile K, Kable K, Sciberras F, Wong G, Kok J. The Effects of COVID-19 in Kidney Transplantation: Evidence From Tissue Pathology. Transplantation 2025; 109:352-361. [PMID: 39020461 DOI: 10.1097/tp.0000000000005121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/19/2024]
Abstract
BACKGROUND The biological effects of SARS-CoV-2 infection in transplanted kidneys are uncertain with little pathological information. METHODS This single-center, prospective observational study evaluated kidney transplant biopsies from recipients of deceased donors with COVID-19, current recipients contracting SARS-CoV-2 Omicron variant in 2022, against prior BK virus (BKV) infection and uninfected (without SARS-CoV-2 or BKV) samples, as respective positive and negative comparators (n = 503 samples). RESULTS We demonstrated nonvirus tubular injury in implanted tissue from infected donors and prevalent recipients with mild acute COVID-19 and acute kidney injury, excluding direct viral infection as a cause of kidney damage. COVID particles were absent in 4116 ultrastructural images of 295 renal tubules from 4 patients with acute COVID-19. No viral cytopathic effect, viral allograft nephropathy, or SARS-CoV-2 RNA was detected in acute tissues, nor in 128 sequential samples from infected donors or recipients with COVID-19. Following recipient COVID-19 (mean 16.8 ± 12.0 wk post-infection), the biopsy-prevalence of rejection was 33.0% (n = 100 biopsies) versus 13.4% for contemporaneous uninfected controls (n = 337; P < 0.001). Prior COVID-19 was an independent risk factor for incident rejection using multivariable generalized estimating equation adjusted for competing risks (odds ratio, 2.195; 95% confidence interval, 1.189-4.052; P = 0.012). Landmark and matched-pair analyses confirmed an association of SARS-CoV-2 with subsequent transplant rejection, with a similar pattern following BKV infection. CONCLUSIONS Transplantation from COVID-19+ deceased donors yielded good recipient outcomes without evidence of viral tissue transmission. Acute kidney injury during COVID-19 was mediated by archetypical tubular injury and infection correlated with an increased risk of subsequent rejection.
Collapse
Affiliation(s)
- Brian J Nankivell
- Department of Renal Medicine, Westmead Hospital, Westmead, NSW, Australia
| | - Chow P'ng
- Tissue Pathology and Diagnostic Oncology and Electron Microscopy Units, Westmead Hospital, Westmead, NSW, Australia
| | - Thomas Tran
- Tissue Pathology and Diagnostic Oncology and Electron Microscopy Units, Westmead Hospital, Westmead, NSW, Australia
| | - Jenny Draper
- Centre for Infectious Diseases & Microbiology Laboratory Services, NSW Health Pathology-Institute of Clinical Pathology and Medical Research, Westmead Hospital, Westmead, NSW, Australia
| | - Danny Ko
- Centre for Infectious Diseases & Microbiology Laboratory Services, NSW Health Pathology-Institute of Clinical Pathology and Medical Research, Westmead Hospital, Westmead, NSW, Australia
| | - Ivan Luu
- Centre for Infectious Diseases & Microbiology Laboratory Services, NSW Health Pathology-Institute of Clinical Pathology and Medical Research, Westmead Hospital, Westmead, NSW, Australia
| | - Kerri Basile
- Centre for Infectious Diseases & Microbiology Laboratory Services, NSW Health Pathology-Institute of Clinical Pathology and Medical Research, Westmead Hospital, Westmead, NSW, Australia
| | - Kathy Kable
- Department of Renal Medicine, Westmead Hospital, Westmead, NSW, Australia
| | | | - Germaine Wong
- Department of Renal Medicine, Westmead Hospital, Westmead, NSW, Australia
| | - Jen Kok
- Centre for Infectious Diseases & Microbiology Laboratory Services, NSW Health Pathology-Institute of Clinical Pathology and Medical Research, Westmead Hospital, Westmead, NSW, Australia
| |
Collapse
|
34
|
Mycroft-West CJ, Leanca MA, Wu L. Structural glycobiology - from enzymes to organelles. Biochem Soc Trans 2025; 53:BST20241119. [PMID: 39889286 DOI: 10.1042/bst20241119] [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: 11/01/2024] [Revised: 12/19/2024] [Accepted: 12/23/2024] [Indexed: 02/02/2025]
Abstract
Biological carbohydrate polymers represent some of the most complex molecules in life, enabling their participation in a huge range of physiological functions. The complexity of biological carbohydrates arises from an extensive enzymatic repertoire involved in their construction, deconstruction and modification. Over the past decades, structural studies of carbohydrate processing enzymes have driven major insights into their mechanisms, supporting associated applications across medicine and biotechnology. Despite these successes, our understanding of how multienzyme networks function to create complex polysaccharides is still limited. Emerging techniques such as super-resolution microscopy and cryo-electron tomography are now enabling the investigation of native biological systems at near molecular resolutions. Here, we review insights from classical in vitro studies of carbohydrate processing, alongside recent in situ studies of glycosylation-related processes. While considerable technical challenges remain, the integration of molecular mechanisms with true biological context promises to transform our understanding of carbohydrate regulation, shining light upon the processes driving functional complexity in these essential biomolecules.
Collapse
Affiliation(s)
| | - Miron A Leanca
- The Rosalind Franklin Institute, Harwell Science & Innovation Campus, OX11 0QX, Didcot, UK
| | - Liang Wu
- The Rosalind Franklin Institute, Harwell Science & Innovation Campus, OX11 0QX, Didcot, UK
- Division of Structural Biology, Nuffield Department of Medicine, University of Oxford, OX3 7BN, Oxford, UK
| |
Collapse
|
35
|
Li J, Mao X, Zhao T, Fang W, Jin Y, Liu M, Fan C, Tian Y. Tetrahedral DNA Framework-Based Spherical Nucleic Acids for Efficient siRNA Delivery. Angew Chem Int Ed Engl 2025; 64:e202416988. [PMID: 39497620 DOI: 10.1002/anie.202416988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Indexed: 11/20/2024]
Abstract
Spherical nucleic acids (SNAs) hold substantial therapeutic potential for the delivery of small interfering RNAs (siRNAs). Nevertheless, their potential remains largely untapped due to the challenges of cytosolic delivery. Inspired by the dynamic, spiky architecture of coronavirus, an interface engineering approach based on a tetrahedral DNA framework (tDF) is demonstrated for the development of coronavirus-mimicking SNAs. By exploiting their robustness and precise construction, tDFs are evenly arranged on the surface of core nanoparticles (NPs) with flexible conformations, generating a dynamic, spiky architecture. This spiky architecture in tetrahedral DNA framework-based SNAs (tDF-SNAs) substantially improve siRNAs duplex efficiency from 20 % to 95 %. Meanwhile, tDF-SNAs changed the endocytosis pathway to clathrin-independent cellular engulfment pathway and enhanced the cellular uptake efficiency. Due to these advances, the delivery efficiency of siRNA molecules by tDF-SNAs is 1-2 orders of magnitude higher than that of SNAs, resulting in a 2-fold increase in gene silencing efficacy. These results show promise in the development of bioinspired siRNAs delivery systems for intracellular applications.
Collapse
Affiliation(s)
- Jie Li
- School of Chemistry and Molecular Engineering, Shanghai Key Laboratory of Green Chemistry and Chemical Processes, East China Normal University, Shanghai, 200241, China
| | - Xiuhai Mao
- Shanghai Key Laboratory for Nucleic Acids Chemistry and Nanomedicine, Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Tiantian Zhao
- School of Chemistry and Molecular Engineering, Shanghai Key Laboratory of Green Chemistry and Chemical Processes, East China Normal University, Shanghai, 200241, China
| | - Weina Fang
- School of Chemistry and Molecular Engineering, Shanghai Key Laboratory of Green Chemistry and Chemical Processes, East China Normal University, Shanghai, 200241, China
| | - Yangyang Jin
- Shanghai Key Laboratory for Nucleic Acids Chemistry and Nanomedicine, Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Mengmeng Liu
- School of Chemistry and Molecular Engineering, Shanghai Key Laboratory of Green Chemistry and Chemical Processes, East China Normal University, Shanghai, 200241, China
| | - Chunhai Fan
- Frontiers Science Center for Transformative Molecules, School of Chemistry and Chemical Engineering, Institute of Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Yang Tian
- School of Chemistry and Molecular Engineering, Shanghai Key Laboratory of Green Chemistry and Chemical Processes, East China Normal University, Shanghai, 200241, China
| |
Collapse
|
36
|
Sekine R, Takeda K, Suenaga T, Tsuno S, Kaiya T, Kiso M, Yamayoshi S, Takaku Y, Ohno S, Yamaguchi Y, Nishizawa S, Sumitomo K, Ikuta K, Kanda T, Kawaoka Y, Nishimura H, Kuge S. G-quadruplex-forming small RNA inhibits coronavirus and influenza A virus replication. Commun Biol 2025; 8:27. [PMID: 39815031 PMCID: PMC11735773 DOI: 10.1038/s42003-024-07351-7] [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: 02/27/2024] [Accepted: 12/03/2024] [Indexed: 01/18/2025] Open
Abstract
Future pandemic threats may be caused by novel coronaviruses and influenza A viruses. Here we show that when directly added to a cell culture, 12mer guanine RNA (G12) and its phosphorothioate-linked derivatives (G12(S)), rapidly entered cytoplasm and suppressed the propagation of human coronaviruses and influenza A viruses to between 1/100 and nearly 1/1000 of normal virus infectivity without cellular toxicity and induction of innate immunity. Moreover, G12(S) alleviated the weight loss caused by coronavirus infection in mice. G12(S) might exhibit a stable G-tetrad with left-handed parallel-stranded G-quadruplex, and inhibit the replication process by impeding interaction between viral nucleoproteins and viral RNA in the cytoplasm. Unlike previous antiviral strategies that target the G-quadruplexes of the viral genome, we now show that excess exogenous G-quadruplex-forming small RNA displaces genomic RNA from ribonucleoprotein, effectively inhibiting viral replication. The approach has the potential to facilitate the creation of versatile middle-molecule antivirals featuring lipid nanoparticle-free delivery.
Collapse
Affiliation(s)
- Ryoya Sekine
- Division of Microbiology, Faculty of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University, 4-4-1, Komatsuhima, Aoba-ku, Sendai, Miyagi, 981-8558, Japan
| | - Kouki Takeda
- Division of Microbiology, Faculty of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University, 4-4-1, Komatsuhima, Aoba-ku, Sendai, Miyagi, 981-8558, Japan
| | - Tsukasa Suenaga
- Division of Microbiology, Faculty of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University, 4-4-1, Komatsuhima, Aoba-ku, Sendai, Miyagi, 981-8558, Japan
| | - Satsuki Tsuno
- Division of Microbiology, Faculty of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University, 4-4-1, Komatsuhima, Aoba-ku, Sendai, Miyagi, 981-8558, Japan
| | - Takumi Kaiya
- Division of Microbiology, Faculty of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University, 4-4-1, Komatsuhima, Aoba-ku, Sendai, Miyagi, 981-8558, Japan
| | - Maki Kiso
- Division of Virology, Institute of Medical Sciences, The University of Tokyo, 4-6-1 Shiroganedai, Minato-ku, Tokyo, 108-8639, Japan
- The University of Tokyo, Pandemic Preparedness, Infection, and Advanced Research Center, Tokyo, Japan
| | - Seiya Yamayoshi
- Division of Virology, Institute of Medical Sciences, The University of Tokyo, 4-6-1 Shiroganedai, Minato-ku, Tokyo, 108-8639, Japan
- The University of Tokyo, Pandemic Preparedness, Infection, and Advanced Research Center, Tokyo, Japan
- The Research Center for Global Viral Diseases, National Center for Global Health and Medicine Research Institute, Tokyo, Japan
| | - Yoshihide Takaku
- Department of Chemistry, Graduate School of Science, Tohoku University, 6-3 Azaaoba, Aoba-ku, Sendai, Miyagi, 980-8578, Japan
| | - Shiho Ohno
- Division of Structural Glycobiology, Faculty of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University, 4-4-1, Komatsuhima, Aoba-ku, Sendai, Miyagi, 981-8558, Japan
| | - Yoshiki Yamaguchi
- Division of Structural Glycobiology, Faculty of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University, 4-4-1, Komatsuhima, Aoba-ku, Sendai, Miyagi, 981-8558, Japan
| | - Seiichi Nishizawa
- Department of Chemistry, Graduate School of Science, Tohoku University, 6-3 Azaaoba, Aoba-ku, Sendai, Miyagi, 980-8578, Japan
| | - Kazuhiro Sumitomo
- Division of Geriatric and Community Medicine, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, 1-15-1, Fukumuro, Miyagino-ku, Sendai, Miyagi, 983-8536, Japan
| | - Kazufumi Ikuta
- Division of Microbiology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, 1-15-1, Fukumuro, Miyagino-ku, Sendai, Miyagi, 983-8536, Japan
| | - Teru Kanda
- Division of Microbiology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, 1-15-1, Fukumuro, Miyagino-ku, Sendai, Miyagi, 983-8536, Japan
| | - Yoshihiro Kawaoka
- Division of Virology, Institute of Medical Sciences, The University of Tokyo, 4-6-1 Shiroganedai, Minato-ku, Tokyo, 108-8639, Japan
- The University of Tokyo, Pandemic Preparedness, Infection, and Advanced Research Center, Tokyo, Japan
- The Research Center for Global Viral Diseases, National Center for Global Health and Medicine Research Institute, Tokyo, Japan
| | - Hidekazu Nishimura
- Virus Research Center, Clinical Research Division, National Hospital Organization Sendai Medical Center, 2-1-12, Miyagino, Miyagino-ku, Sendai, Miyagi, 983-8520, Japan
| | - Shusuke Kuge
- Division of Microbiology, Faculty of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University, 4-4-1, Komatsuhima, Aoba-ku, Sendai, Miyagi, 981-8558, Japan.
| |
Collapse
|
37
|
Jing L, Zhao F, Zheng L, Meng B, Gao S, Laporte M, Jochmans D, De Jonghe S, Neyts J, Zhan P, Kang D, Liu X. Optimization of SARS-CoV-2 M pro Inhibitors by a Structure-Based Multilevel Virtual Screening Method. Int J Mol Sci 2025; 26:670. [PMID: 39859382 PMCID: PMC11765572 DOI: 10.3390/ijms26020670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 01/04/2025] [Accepted: 01/12/2025] [Indexed: 01/30/2025] Open
Abstract
With the aim of developing novel anti-SARS-CoV-2 drugs to address the ongoing evolution and emergence of drug-resistant strains, the reported SARS-CoV-2 Mpro inhibitor WU-04 was selected as a lead to find novel, highly potent, and broad-spectrum inhibitors. Using a fragment-based multilevel virtual screening strategy, 15 hit compounds were identified and subsequently synthesized. Among them, A5 (IC50 = 1.05 μM), A6 (IC50 = 1.08 μM), and A9 (IC50 = 0.154 μM) demonstrated potent SARS-CoV-2 Mpro inhibition comparable to or slightly weaker than WU-04. Antiviral activity evaluations revealed that compound A9 exhibited the strongest antiviral activity with an EC50 value of 0.18 μM, quite comparable to the marketed drug Nirmatrelvir (EC50 = 0.123 μM) and inferior to WU-04 (EC50 = 0.042 μM). Molecular dynamics simulations elucidated the key interactions between compounds A5, A6, A9, and the binding pocket of SARS-CoV-2 Mpro, providing valuable insights into their mechanisms of action. These findings identify compound A9 as a promising lead for anti-SARS-CoV-2 drug development.
Collapse
Affiliation(s)
- Lanlan Jing
- Key Laboratory of Chemical Biology (Ministry of Education), Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, Jinan 250012, China
| | - Fabao Zhao
- Key Laboratory of Chemical Biology (Ministry of Education), Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, Jinan 250012, China
| | - Lin Zheng
- Key Laboratory of Chemical Biology (Ministry of Education), Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, Jinan 250012, China
| | - Bairu Meng
- Key Laboratory of Chemical Biology (Ministry of Education), Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, Jinan 250012, China
| | - Shenghua Gao
- Key Laboratory of Chemical Biology (Ministry of Education), Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, Jinan 250012, China
| | - Manon Laporte
- Antiviral Drug & Vaccine Research Group, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Herestraat 49, B-3000 Leuven, Belgium
| | - Dirk Jochmans
- Antiviral Drug & Vaccine Research Group, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Herestraat 49, B-3000 Leuven, Belgium
| | - Steven De Jonghe
- Molecular, Structural and Translational Virology Research Group, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Herestraat 49, B-3000 Leuven, Belgium
| | - Johan Neyts
- Antiviral Drug & Vaccine Research Group, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Herestraat 49, B-3000 Leuven, Belgium
| | - Peng Zhan
- Key Laboratory of Chemical Biology (Ministry of Education), Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, Jinan 250012, China
| | - Dongwei Kang
- Key Laboratory of Chemical Biology (Ministry of Education), Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, Jinan 250012, China
- China-Belgium Collaborative Research Center for Innovative Antiviral Drugs of Shandong Province, Shandong University, 44 West Culture Road, Jinan 250012, China
| | - Xinyong Liu
- Key Laboratory of Chemical Biology (Ministry of Education), Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, Jinan 250012, China
- China-Belgium Collaborative Research Center for Innovative Antiviral Drugs of Shandong Province, Shandong University, 44 West Culture Road, Jinan 250012, China
| |
Collapse
|
38
|
Lall S, Balaram P, Mathew MK, Gosavi S. Sequence of the SARS-CoV-2 Spike Transmembrane Domain Encodes Conformational Dynamics. J Phys Chem B 2025; 129:194-209. [PMID: 39692154 DOI: 10.1021/acs.jpcb.4c05270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2024]
Abstract
The homotrimeric SARS-CoV-2 spike protein enables viral infection by undergoing a large conformational transition, which facilitates the fusion of the viral envelope with the host cell membrane. The spike protein is anchored to the SARS-CoV-2 envelope by its transmembrane domain (TMD), composed of three TM helices, each contributed by one of the protomers of spike. Although the TMD is known to be important for viral fusion, whether it is a passive anchor of the spike or actively promotes fusion remains unknown. Specifically, it is unclear if the TMD and its dynamics facilitate the prefusion to postfusion conformational transition of the spike. Here, we computationally study the dynamics and self-assembly of the SARS-CoV-2 spike TMD in homogeneous POPC and cholesterol containing membranes. Atomistic simulations of a long TM helix-containing protomer segment show that the membrane-embedded segment bobs, tilts and gains and loses helicity, locally thinning the membrane. Coarse-grained multimerization simulations using representative TM helix structures from the atomistic simulations exhibit diverse trimer populations whose architecture depends on the structure of the TM helix protomer. While a symmetric conformation reflects the symmetry of the resting spike, an asymmetric TMD conformation could promote membrane fusion through the stabilization of a fusion intermediate. Together, our simulations demonstrate that the sequence and length of the SARS-CoV-2 spike TM segment make it inherently dynamic, that trimerization does not abrogate these dynamics and that the various observed TMD conformations may enable viral fusion.
Collapse
Affiliation(s)
- Sahil Lall
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bangalore 560065, India
- Simons Centre for the Study of Living Machines, National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bangalore 560065, India
| | - Padmanabhan Balaram
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bangalore 560065, India
| | - M K Mathew
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bangalore 560065, India
| | - Shachi Gosavi
- Simons Centre for the Study of Living Machines, National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bangalore 560065, India
| |
Collapse
|
39
|
Chen J, Mu J, Zhou K, Zhang Y, Zhang J, Shu T, Shang W, Ren Y, Xu XQ, Zhang L, Yuan S, Zhang D, Cai K, Qiu Y, Zhou X. Targeting viral suppressor of RNAi confers anti-coronaviral activity. Mol Ther 2025; 33:201-214. [PMID: 39663700 PMCID: PMC11764073 DOI: 10.1016/j.ymthe.2024.12.009] [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: 09/03/2024] [Revised: 11/02/2024] [Accepted: 12/06/2024] [Indexed: 12/13/2024] Open
Abstract
Infections caused by coronaviruses are persistent threats to human health in recent decades, necessitating the development of innovative anti-coronaviral therapies. RNA interference (RNAi) is a conserved cell-intrinsic antiviral mechanism in diverse eukaryotic organisms, including mammals. To counteract, many viruses encode viral suppressors of RNAi (VSRs) to evade antiviral RNAi, implying that targeting VSRs could be a promising strategy to develop antiviral therapies. Here, we designed a series of peptides specifically targeting the SARS-CoV-2-encoded VSR, nucleocapsid (N) protein. Among these peptides, one designated GL directly interacts with N protein and inactivates its VSR activity, which unlocks a potent RNAi response and effectively inhibits severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) replication. Moreover, GL exhibited RNAi-dependent antiviral effects not only against various SARS-CoV-2 variants, including Delta, Omicron BA.5, XBB, and JN.1, but also against other coronaviruses such as human coronavirus (HCoV)-229E, HCoV-OC43, and mouse hepatitis virus. The in vivo anti-coronaviral activity of GL was also confirmed. Our findings indicate that the VSR-targeting peptide GL has the potential to be further developed as a broad-spectrum anti-coronaviral treatment, highlighting the functional importance and therapeutic potential of antiviral RNAi.
Collapse
Affiliation(s)
- Jiyao Chen
- Joint Laboratory of Infectious Diseases and Health, Wuhan Institute of Virology & Wuhan Jinyintan Hospital, Wuhan Jinyintan Hospital, Wuhan, Hubei 430023, China; State Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences (CAS), Wuhan 430071, China; State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
| | - JingFang Mu
- State Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences (CAS), Wuhan 430071, China; State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
| | - Kangping Zhou
- Institute of Health Inspection and Testing, Hubei Provincial Center for Disease Control and Prevention, Wuhan, China
| | - Yuming Zhang
- State Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences (CAS), Wuhan 430071, China; State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
| | - Jieling Zhang
- State Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences (CAS), Wuhan 430071, China; State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China; School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China
| | - Ting Shu
- State Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences (CAS), Wuhan 430071, China; State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
| | - Weijuan Shang
- State Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences (CAS), Wuhan 430071, China; State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
| | - Yujie Ren
- State Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences (CAS), Wuhan 430071, China; State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
| | - Xi-Qiu Xu
- State Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences (CAS), Wuhan 430071, China; State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
| | - Leike Zhang
- State Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences (CAS), Wuhan 430071, China; State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
| | - Shuai Yuan
- State Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences (CAS), Wuhan 430071, China; State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China; School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China
| | - Dingyu Zhang
- Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei 430023, China.
| | - Kun Cai
- Institute of Health Inspection and Testing, Hubei Provincial Center for Disease Control and Prevention, Wuhan, China.
| | - Yang Qiu
- State Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences (CAS), Wuhan 430071, China; State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China.
| | - Xi Zhou
- State Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences (CAS), Wuhan 430071, China; State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China.
| |
Collapse
|
40
|
Márquez-Moñino MÁ, Santiveri CM, de León P, Camero S, Campos-Olivas R, Jiménez MÁ, Sáiz M, González B, Pérez-Cañadillas JM. The ALS drug riluzole binds to the C-terminal domain of SARS-CoV-2 nucleocapsid protein and has antiviral activity. Structure 2025; 33:39-50.e6. [PMID: 39541975 DOI: 10.1016/j.str.2024.10.025] [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: 07/01/2024] [Revised: 09/18/2024] [Accepted: 10/21/2024] [Indexed: 11/17/2024]
Abstract
Nucleoproteins (N) play an essential role in virus assembly and are less prone to mutation than other viral structural proteins, making them attractive targets for drug discovery. Using an NMR fragment-based drug discovery approach, we identified the 1,3-benzothiazol-2-amine (BZT) group as a scaffold to develop potential antivirals for SARS-CoV-2 nucleocapsid (N) protein. A thorough characterization of BZT derivatives using NMR, X-ray crystallography, antiviral activity assays, and intrinsic fluorescence measurements revealed their binding in the C-terminal domain (CTD) domain of the N protein, to residues Arg 259, Trp 330, and Lys 338, coinciding with the nucleotide binding site. Our most effective compound exhibits a slightly better affinity than GTP and the ALS drug riluzole, also identified during the screening, and displays notable viral inhibition activity. A virtual screening of 218 BZT-based compounds revealed a potential extended binding site that could be exploited for the future development of new SARS-CoV-2 antivirals.
Collapse
Affiliation(s)
| | - Clara M Santiveri
- Spectroscopy and Nuclear Magnetic Resonance Unit, Structural Biology Programme, Spanish National Cancer Research Centre, C/ Melchor Fernández Almagro 3, 28029 Madrid, Spain
| | - Patricia de León
- Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), Universidad Autónoma de Madrid, c/ Nicolás Cabrera nº 1, 28049 Madrid, Spain
| | - Sergio Camero
- Institute of Physical-Chemistry "Blas Cabrera", CSIC, C/ Serrano 119, 28006 Madrid, Spain
| | - Ramón Campos-Olivas
- Spectroscopy and Nuclear Magnetic Resonance Unit, Structural Biology Programme, Spanish National Cancer Research Centre, C/ Melchor Fernández Almagro 3, 28029 Madrid, Spain
| | - M Ángeles Jiménez
- Institute of Physical-Chemistry "Blas Cabrera", CSIC, C/ Serrano 119, 28006 Madrid, Spain
| | - Margarita Sáiz
- Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), Universidad Autónoma de Madrid, c/ Nicolás Cabrera nº 1, 28049 Madrid, Spain
| | - Beatriz González
- Institute of Physical-Chemistry "Blas Cabrera", CSIC, C/ Serrano 119, 28006 Madrid, Spain.
| | | |
Collapse
|
41
|
Zhu R, Martínez-Roque MA, Figueroa-Miranda G, Hu Z, Acunzo A, Li H, Hu Q, Bednar J, Gensch T, Ingebrandt S, Offenhäusser A, Mayer D. Plasmon-enhanced fluorescence and electrochemical aptasensor for SARS-CoV-2 Spike protein detection. Talanta 2025; 281:126760. [PMID: 39226699 DOI: 10.1016/j.talanta.2024.126760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 08/20/2024] [Accepted: 08/22/2024] [Indexed: 09/05/2024]
Abstract
In this work, we combined plasmon-enhanced fluorescence and electrochemical (PEF-EC) transduction mechanisms to realize a highly sensitive dual-transducer aptasensor. To implement two traducers in one biosensor, a novel large-scale nanoimprint lithography process was introduced to fabricate gold nanopit arrays (AuNpA) with unique fringe structures. Light transmitting through the AuNpA samples exhibited a surface plasmon polariton peak overlapping with the excitation peak of the C7 aptamer-associated fluorophore methylene blue (MB). We observed a five and seven-times higher average fluorescence intensity over the AuNpA and fringe structure, respectively, in comparison to a plane Au film. Furthermore, the MB fluorophore was simultaneously utilized as a redox probe for electrochemical investigations and is described here as a dual transduction label for the first time. The novel dual transducer system was deployed for the detection of SARS-CoV-2 Spike protein via a C7 aptamer in combination with a strand displacement protocol. The PEF transducer exhibited a detection range from 1 fg/mL to 10 ng/mL with a detection limit of 0.07 fg/mL, while the EC traducer showed an extended dynamic range from 1 fg/mL to 100 ng/mL with a detection limit of 0.15 fg/mL. This work provides insights into an easy-to-perform, large-scale fabrication process for nanostructures enabling plasmon-enhanced fluorescence, and the development of an advanced but universal aptasensor platform.
Collapse
Affiliation(s)
- Ruifeng Zhu
- Institute of Biological Information Processing, Bioelectronics (IBI-3), Forschungszentrum Jülich GmbH, 52428, Jülich, Germany; Institute of Materials in Electrical Engineering 1, RWTH Aachen University, 52074, Aachen, Germany
| | - Mateo Alejandro Martínez-Roque
- Institute of Biological Information Processing, Bioelectronics (IBI-3), Forschungszentrum Jülich GmbH, 52428, Jülich, Germany
| | - Gabriela Figueroa-Miranda
- Institute of Biological Information Processing, Bioelectronics (IBI-3), Forschungszentrum Jülich GmbH, 52428, Jülich, Germany
| | - Ziheng Hu
- Institute of Biological Information Processing, Bioelectronics (IBI-3), Forschungszentrum Jülich GmbH, 52428, Jülich, Germany
| | - Adriano Acunzo
- Institute of Biological Information Processing, Bioelectronics (IBI-3), Forschungszentrum Jülich GmbH, 52428, Jülich, Germany; Department of Physics, University of Naples "Federico II", Via Cintia 26, Naples, 80126, Italy
| | - Hangyu Li
- Institute of Biological Information Processing, Bioelectronics (IBI-3), Forschungszentrum Jülich GmbH, 52428, Jülich, Germany
| | - Qinyu Hu
- Institute of Biological Information Processing, Bioelectronics (IBI-3), Forschungszentrum Jülich GmbH, 52428, Jülich, Germany
| | - Justus Bednar
- Institute of Biological Information Processing, Bioelectronics (IBI-3), Forschungszentrum Jülich GmbH, 52428, Jülich, Germany; Fakultät für Mathematik, Informatik und Naturwissenschaften, RWTH Aachen University, 52074, Aachen, Germany
| | - Thomas Gensch
- Institute of Biological Information Processing, Molecular and Cellular Physiology (IBI-1), Forschungszentrum Jülich GmbH, 52428, Jülich, Germany
| | - Sven Ingebrandt
- Institute of Materials in Electrical Engineering 1, RWTH Aachen University, 52074, Aachen, Germany
| | - Andreas Offenhäusser
- Institute of Biological Information Processing, Bioelectronics (IBI-3), Forschungszentrum Jülich GmbH, 52428, Jülich, Germany
| | - Dirk Mayer
- Institute of Biological Information Processing, Bioelectronics (IBI-3), Forschungszentrum Jülich GmbH, 52428, Jülich, Germany.
| |
Collapse
|
42
|
da Silva Santos I, Magalhaes LO, Marra RKF, da Silva Lima CH, Hamerski L, Albuquerque MG, da Silva BV. Natural and Synthetic Coumarins as Potential Drug Candidates against SARS-CoV-2/COVID-19. Curr Med Chem 2025; 32:539-562. [PMID: 38243979 DOI: 10.2174/0109298673285609231220111556] [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: 10/05/2023] [Revised: 11/27/2023] [Accepted: 11/30/2023] [Indexed: 01/22/2024]
Abstract
COVID-19, an airborne disease caused by a betacoronavirus named SARS-- CoV-2, was officially declared a pandemic in early 2020, resulting in more than 770 million confirmed cases and over 6.9 million deaths by September 2023. Although the introduction of vaccines in late 2020 helped reduce the number of deaths, the global effort to fight COVID-19 is far from over. While significant progress has been made in a short period, the fight against SARS-CoV-2/COVID-19 and other potential pandemic threats continues. Like AIDS and hepatitis C epidemics, controlling the spread of COVID-19 will require the development of multiple drugs to weaken the virus's resistance to different drug treatments. Therefore, it is essential to continue developing new drug candidates derived from natural or synthetic small molecules. Coumarins are a promising drug design and development scaffold due to their synthetic versatility and unique physicochemical properties. Numerous examples reported in scientific literature, mainly by in silico prospection, demonstrate their potential contribution to the rapid development of drugs against SARS-CoV-2/COVID-19 and other emergent and reemergent viruses.
Collapse
Affiliation(s)
- Iara da Silva Santos
- Department of Organic Chemistry, Instituto de Pesquisas de Produtos Naturais Walter Mors, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Leticia Oliveira Magalhaes
- Department of Organic Chemistry, Universidade Federal do Rio de Janeiro, Instituto de Química, Rio de Janeiro, Brazil
| | - Roberta Katlen Fusco Marra
- Department of Organic Chemistry, Universidade Federal do Rio de Janeiro, Instituto de Química, Rio de Janeiro, Brazil
| | - Camilo Henrique da Silva Lima
- Department of Organic Chemistry, Universidade Federal do Rio de Janeiro, Instituto de Química, Rio de Janeiro, Brazil
| | - Lidilhone Hamerski
- Department of Organic Chemistry, Instituto de Pesquisas de Produtos Naturais Walter Mors, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Magaly Girao Albuquerque
- Department of Organic Chemistry, Universidade Federal do Rio de Janeiro, Instituto de Química, Rio de Janeiro, Brazil
| | - Barbara Vasconcellos da Silva
- Department of Organic Chemistry, Universidade Federal do Rio de Janeiro, Instituto de Química, Rio de Janeiro, Brazil
| |
Collapse
|
43
|
Sultana R, Stahelin RV. Strengths and limitations of SARS-CoV-2 virus-like particle systems. Virology 2025; 601:110285. [PMID: 39536645 PMCID: PMC11624109 DOI: 10.1016/j.virol.2024.110285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 10/19/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024]
Abstract
Virus-like particles (VLPs) resemble the parent virus but lack the viral genome, providing a safe and efficient platform for the analysis of virus assembly and budding as well as the development of vaccines and drugs. During the COVID-19 pandemic, caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the formation of SARS-CoV-2 VLPs was investigated as an alternative to authentic virions because the latter requires biosafety level 3 (BSL-3) facilities. This allowed researchers to model its assembly and budding processes, examine the role of mutations in variants of concern, and determine how the structural proteins interact with each other. Also, the absence of viral genome in VLPs circumvents worries of gains in infectivity via mutagenesis. This review summarizes the strengths and limitations of several SARS-CoV-2 VLP systems and details some of the strides that have been made in using these systems to study virus assembly and budding, viral entry, and antibody and vaccine development.
Collapse
Affiliation(s)
- Rokaia Sultana
- Borch Department of Medicinal Chemistry and Molecular Pharmacology and The Purdue Institute of Inflammation, Immunology and Infectious Disease, Purdue University, 47907, West Lafayette, IN, USA
| | - Robert V Stahelin
- Borch Department of Medicinal Chemistry and Molecular Pharmacology and The Purdue Institute of Inflammation, Immunology and Infectious Disease, Purdue University, 47907, West Lafayette, IN, USA.
| |
Collapse
|
44
|
Song JH, Jang S, Choi JW, Hwang S, Kim KH, Kim HY, Park SC, Lee W, Lee JY. Characterization of Site-Specific N- and O-Glycopeptides from Recombinant Spike and ACE2 Glycoproteins Using LC-MS/MS Analysis. Int J Mol Sci 2024; 25:13649. [PMID: 39769415 PMCID: PMC11678118 DOI: 10.3390/ijms252413649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 12/13/2024] [Accepted: 12/18/2024] [Indexed: 01/11/2025] Open
Abstract
The COVID-19 pandemic, caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has resulted in hundreds of millions of infections and millions of deaths globally. Although vaccination campaigns are mitigating the pandemic, emerging viral variants continue to pose challenges. The spike (S) protein of SARS-CoV-2 plays a critical role in viral entry by binding to the angiotensin-converting enzyme 2 (ACE2) receptor, making both proteins essential targets for therapeutic and vaccine development. The glycosylation of these proteins influences their structure and function. This underscores the need for detailed site-specific glycoproteomic analysis. In this study, we characterized the N- or O-glycosylation profiles of the recombinant receptor-binding domain (RBD) of spike protein and ACE2 proteins expressed from Expi293F cells, as well as the S2 subunit of spike protein expressed in plant (N. benthamiana) cells. Using a high-resolution Orbitrap Eclipse Tribrid mass spectrometer equipped with the Ultimate 3000 RSLCnano and I-GPA (Integrated GlycoProteome Analyzer) developed in a previous study, 148 N- and 28 O-glycopeptides from RBD, 71 N-glycopeptides from the S2 subunit, and 139 N-glycopeptides from ACE2 were characterized. In addition, we report post-translational modifications (PTMs) of glycan, including mannose-6-phosphate (M6P) and GlcNAc-1-phosphate-6-O-mannose in N-glycan of RBD and ACE2, and O-acetylation in O-glycan of RBD, identified for the first time in these recombinant proteins. The relative abundance distribution according to glycosites and glycan types were analyzed by quantified site-specific N- and O (only from RBD)-glycopeptides from RBD, S2, and ACE2 using I-GPA. Asn331 for RBD, Asn1098 for S2, and Asn103 for ACE2 were majorly N-glycosylated, and dominant glycan-type was complex from RBD and ACE2 and high-mannose from S2. These findings will provide valuable insights into the glycosylation patterns that influence protein function and immunogenicity and offer new perspectives for the development of vaccines and antibody-based therapies against COVID-19.
Collapse
Affiliation(s)
- Ju Hwan Song
- Digital Omics Research Center, Korea Basic Science Institute, Ochang 28119, Republic of Korea; (J.H.S.)
- Department of Biotechnology, Graduate School, Korea University, Seoul 02841, Republic of Korea
| | - Sangeun Jang
- Digital Omics Research Center, Korea Basic Science Institute, Ochang 28119, Republic of Korea; (J.H.S.)
| | - Jin-Woong Choi
- Digital Omics Research Center, Korea Basic Science Institute, Ochang 28119, Republic of Korea; (J.H.S.)
| | - Seoyoung Hwang
- Digital Omics Research Center, Korea Basic Science Institute, Ochang 28119, Republic of Korea; (J.H.S.)
| | - Kyoung Heon Kim
- Department of Biotechnology, Graduate School, Korea University, Seoul 02841, Republic of Korea
| | - Hye-Yeon Kim
- Biopharmaceutical Research Center, Korea Basic Science Institute, Cheongju 28119, Republic of Korea
| | - Sun Cheol Park
- Biopharmaceutical Research Center, Korea Basic Science Institute, Cheongju 28119, Republic of Korea
| | - Wonbin Lee
- Biopharmaceutical Research Center, Korea Basic Science Institute, Cheongju 28119, Republic of Korea
| | - Ju Yeon Lee
- Digital Omics Research Center, Korea Basic Science Institute, Ochang 28119, Republic of Korea; (J.H.S.)
- Department of Bio-Analytical Science, University of Science and Technology, Daejeon 34113, Republic of Korea
- Critical Diseases Diagnostics Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Republic of Korea
| |
Collapse
|
45
|
Luo L, Li J, Shen X, Li X, Peng C, Li S, Kuai R. A Bioinspired Nanovaccine for Personalized Cancer Immunotherapy. NANO LETTERS 2024; 24:15758-15766. [PMID: 39585971 DOI: 10.1021/acs.nanolett.4c04557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2024]
Abstract
Poly I:C (pIC) can act on endosomal and cytosolic pathogen recognition receptors to enhance T cell immunity. However, the poor cytosolic delivery of pIC and lack of facile methods for codelivery with antigens limit its efficacy. Inspired by the structure of a virus, we developed a liponanogel (LNG) consisting of a nanogel core and lipid shell to address these challenges. An LNG-based vaccine increases the endosomal membrane permeability in a nanogel core-dependent manner, thus enhancing cytosolic sensing of pIC. LNG induces 44.9-fold stronger CD8+ T cell responses than soluble pIC or Hiltonol adjuvanted vaccines and even induces stronger CD8+ T cell responses than state-of-the-art lipid nanoparticle adjuvanted vaccines. Remarkably, the LNG vaccine regresses 100% TC1 tumors and even regresses 60% aggressive B16F10 tumors upon combination with αPD-L1. Our study provides a safe and effective strategy for enhancing T cell immunity and may inspire new approaches for cancer immunotherapy.
Collapse
Affiliation(s)
- Lanqing Luo
- School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China
- Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
| | - Junyao Li
- School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China
- Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
| | - Xueying Shen
- School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China
- Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
| | - Xinyan Li
- School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China
- Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
| | - Cheng Peng
- Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
- School of Life Sciences, Tsinghua University, Beijing 100084, China
- Frontier Research Center for Biological Structure & State Key Laboratory of Membrane Biology, Beijing 100084, China
| | - Sai Li
- Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
- School of Life Sciences, Tsinghua University, Beijing 100084, China
- Frontier Research Center for Biological Structure & State Key Laboratory of Membrane Biology, Beijing 100084, China
| | - Rui Kuai
- School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China
- Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
| |
Collapse
|
46
|
Hoffmann T, Michel J, Nitsche A, Mache C, Schulze J, Wolff T, Laue M. Electron microscopy images and morphometric data of SARS-CoV-2 variants in ultrathin plastic sections. Sci Data 2024; 11:1322. [PMID: 39632915 PMCID: PMC11618623 DOI: 10.1038/s41597-024-04182-3] [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: 08/12/2024] [Accepted: 11/28/2024] [Indexed: 12/07/2024] Open
Abstract
Conventional thin section electron microscopy of viral pathogens, such as the pandemic SARS-CoV-2, can provide structural information on the virus particle phenotype and its evolution. We recorded about 900 transmission electron microscopy images of different SARS-CoV-2 variants, including Alpha (B.1.1.7), Beta (B.1.351), Delta (B.1.617.2) and Omicron BA.2 (B.1.1.529) and determined various morphometric parameters, such as maximal diameter and spike number, using a previously published measurement method. The datasets of the evolved virus variants were supplemented with images and measurements of the early SARS-CoV-2 isolates Munich929 and Italy-INMI1 to allow direct comparison. Infected Vero cell cultures were cultivated under comparable conditions to produce the viruses for imaging and morphometric analysis. The images and measurements can be used as a basis to analyse the morphometric changes of further evolving viruses at the particle level or for developing automated image processing workflows and analysis.
Collapse
Affiliation(s)
- Tobias Hoffmann
- Advanced Light and Electron Microscopy, Centre for Biological Threats and Special Pathogens 4 (ZBS 4), Robert Koch Institute, Berlin, Germany
| | - Janine Michel
- Highly Pathogenic Viruses, Centre for Biological Threats and Special Pathogens 1 (ZBS 1), Robert Koch Institute, Berlin, Germany
| | - Andreas Nitsche
- Highly Pathogenic Viruses, Centre for Biological Threats and Special Pathogens 1 (ZBS 1), Robert Koch Institute, Berlin, Germany
| | - Christin Mache
- Influenza and Other Respiratory Viruses (Unit 17), Robert Koch Institute, Berlin, Germany
| | - Jessica Schulze
- Influenza and Other Respiratory Viruses (Unit 17), Robert Koch Institute, Berlin, Germany
| | - Thorsten Wolff
- Influenza and Other Respiratory Viruses (Unit 17), Robert Koch Institute, Berlin, Germany
| | - Michael Laue
- Advanced Light and Electron Microscopy, Centre for Biological Threats and Special Pathogens 4 (ZBS 4), Robert Koch Institute, Berlin, Germany.
| |
Collapse
|
47
|
Ke Z, Peacock TP, Brown JC, Sheppard CM, Croll TI, Kotecha A, Goldhill DH, Barclay WS, Briggs JAG. Virion morphology and on-virus spike protein structures of diverse SARS-CoV-2 variants. EMBO J 2024; 43:6469-6495. [PMID: 39543395 PMCID: PMC11649927 DOI: 10.1038/s44318-024-00303-1] [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: 07/25/2024] [Revised: 10/23/2024] [Accepted: 10/25/2024] [Indexed: 11/17/2024] Open
Abstract
The evolution of SARS-CoV-2 variants with increased fitness has been accompanied by structural changes in the spike (S) proteins, which are the major target for the adaptive immune response. Single-particle cryo-EM analysis of soluble S protein from SARS-CoV-2 variants has revealed this structural adaptation at high resolution. The analysis of S trimers in situ on intact virions has the potential to provide more functionally relevant insights into S structure and virion morphology. Here, we characterized B.1, Alpha, Beta, Gamma, Delta, Kappa, and Mu variants by cryo-electron microscopy and tomography, assessing S cleavage, virion morphology, S incorporation, "in-situ" high-resolution S structures, and the range of S conformational states. We found no evidence for adaptive changes in virion morphology, but describe multiple different positions in the S protein where amino acid changes alter local protein structure. Taken together, our data are consistent with a model where amino acid changes at multiple positions from the top to the base of the spike cause structural changes that can modulate the conformational dynamics of the S protein.
Collapse
Affiliation(s)
- Zunlong Ke
- Department of Cell and Virus Structure, Max Planck Institute of Biochemistry, Martinsried, Germany
- Structural Studies Division, Medical Research Council Laboratory of Molecular Biology, Cambridge, UK
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, USA
| | - Thomas P Peacock
- Department of Infectious Disease, Imperial College London, London, UK
- The Pirbright Institute, Woking, UK
| | - Jonathan C Brown
- Department of Infectious Disease, Imperial College London, London, UK
| | - Carol M Sheppard
- Department of Infectious Disease, Imperial College London, London, UK
| | - Tristan I Croll
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK
- Altos Labs, Cambridge, UK
| | - Abhay Kotecha
- Materials and Structural Analysis, Thermo Fisher Scientific, Eindhoven, The Netherlands
| | - Daniel H Goldhill
- Department of Infectious Disease, Imperial College London, London, UK
- Department of Pathobiology and Population Sciences, Royal Veterinary College, London, UK
| | - Wendy S Barclay
- Department of Infectious Disease, Imperial College London, London, UK
| | - John A G Briggs
- Department of Cell and Virus Structure, Max Planck Institute of Biochemistry, Martinsried, Germany.
- Structural Studies Division, Medical Research Council Laboratory of Molecular Biology, Cambridge, UK.
| |
Collapse
|
48
|
Megha KB, Reshma S, Amir S, Krishnan MJA, Shimona A, Alka R, Mohanan PV. Comprehensive Risk Assessment of Infection Induced by SARS-CoV-2. Mol Neurobiol 2024; 61:9851-9872. [PMID: 37817031 DOI: 10.1007/s12035-023-03682-4] [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: 06/19/2023] [Accepted: 09/28/2023] [Indexed: 10/12/2023]
Abstract
The pandemic COVID-19 (coronavirus disease 2019) is caused by the severe acute respiratory syndrome corona virus 2 (SARS-CoV-2), which devastated the global economy and healthcare system. The infection caused an unforeseen rise in COVID-19 patients and increased the mortality rate globally. This study gives an overall idea about host-pathogen interaction, immune responses to COVID-19, recovery status of infection, targeted organs and complications associated, and comparison of post-infection immunity in convalescent subjects and non-infected individuals. The emergence of the variants and episodes of COVID-19 infections made the situation worsen. The timely introduction of vaccines and precautionary measures helped control the infection's severity. Later, the population that recovered from COVID-19 grew significantly. However, understanding the impact of healthcare issues resulting after infection is paramount for improving an individual's health status. It is now recognised that COVID-19 infection affects multiple organs and exhibits a broad range of clinical manifestations. So, post COVID-19 infection creates a high risk in individuals with already prevailing health complications. The identification of post-COVID-19-related health issues and their appropriate management is of greater importance to improving patient's quality of life. The persistence, sequelae and other medical complications that normally last from weeks to months after the recovery of the initial infection are involved with COVID-19. A multi-disciplinary approach is necessary for the development of preventive measures, techniques for rehabilitation and strategies for clinical management when it comes to long-term care.
Collapse
Affiliation(s)
- K B Megha
- Toxicology Division, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology (Govt. of India), Poojapura, Trivandrum, Kerala, 695 012, India
| | - S Reshma
- Toxicology Division, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology (Govt. of India), Poojapura, Trivandrum, Kerala, 695 012, India
| | - S Amir
- Toxicology Division, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology (Govt. of India), Poojapura, Trivandrum, Kerala, 695 012, India
| | - M J Ajai Krishnan
- Toxicology Division, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology (Govt. of India), Poojapura, Trivandrum, Kerala, 695 012, India
| | - A Shimona
- CSIR-Institute of Microbial Technology, Sector 39-A, Chandigarh, 160036, India
- Academy of Scientific and Innovation Research (AcSIR), Ghaziabad, 201002, India
| | - Rao Alka
- CSIR-Institute of Microbial Technology, Sector 39-A, Chandigarh, 160036, India
- Academy of Scientific and Innovation Research (AcSIR), Ghaziabad, 201002, India
| | - P V Mohanan
- Toxicology Division, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology (Govt. of India), Poojapura, Trivandrum, Kerala, 695 012, India.
| |
Collapse
|
49
|
Mizuma K, Hashizume M, Urata S, Shindo K, Takashima A, Mizuta S, Iwasaki M. U-73122, a phospholipase C inhibitor, impairs lymphocytic choriomeningitis virus virion infectivity. J Gen Virol 2024; 105. [PMID: 39688895 DOI: 10.1099/jgv.0.002060] [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: 12/18/2024] Open
Abstract
Lassa virus (LASV) is an Old World (OW) mammarenavirus that causes Lassa fever, a life-threatening acute febrile disease endemic in West Africa. Lymphocytic choriomeningitis virus (LCMV) is a worldwide-distributed, prototypic OW mammarenavirus of clinical significance that has been largely neglected as a human pathogen. No licensed OW mammarenavirus vaccines are available, and the current therapeutic option is limited to the off-label use of ribavirin, which offers only partial efficacy. This situation underscores the urgent need to develop novel antivirals against human pathogenic mammarenaviruses. Previously, we showed that afatinib, a pan-ErbB tyrosine kinase inhibitor, inhibited multiple steps of the life cycles of OW LASV and LCMV, as well as the New World Junín virus vaccine strain Candid#1. In the present study, we investigated the inhibitory effect of U-73122, a phospholipase C inhibitor that acts downstream of ErbB signalling, on LCMV multiplication. U-73122 inhibited WT recombinant (r) LCMV multiplication in cultured cells. Preincubation of cell-free LCMV virions with U-73122 resulted in impaired virion infectivity. U-73122 also inhibited the infection of rLCMVs expressing heterologous viral glycoproteins, including the vesicular stomatitis Indiana virus (VSIV) glycoprotein, whereas WT VSIV infection was not affected by U-73122 treatment. Our results show the novel bioactivity of U-73122 as an LCMV inhibitor and indicate the presence of a virion-associated molecule that is necessary for virion infectivity and can be exploited as a potential antiviral drug target against human pathogenic mammarenavirus infections.
Collapse
Affiliation(s)
- Keita Mizuma
- Laboratory of Emerging Viral Diseases, International Research Center for Infectious Diseases, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
- Present address: Division of Risk Analysis and Management, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Mei Hashizume
- Laboratory of Emerging Viral Diseases, International Research Center for Infectious Diseases, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Shuzo Urata
- National Research Center for the Control and Prevention of Infectious Diseases, Nagasaki University, Nagasaki, Japan
| | - Keiko Shindo
- Laboratory of Emerging Viral Diseases, International Research Center for Infectious Diseases, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Ayako Takashima
- Laboratory of Emerging Viral Diseases, International Research Center for Infectious Diseases, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Satoshi Mizuta
- Center for Bioinformatics and Molecular Medicine, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Masaharu Iwasaki
- Laboratory of Emerging Viral Diseases, International Research Center for Infectious Diseases, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
- Center for Infectious Disease Education and Research, Osaka University, Suita, Osaka, Japan
- Center for Advanced Modalities and Drug Delivery System, Osaka University, Suita, Osaka, Japan
- RNA Frontier Science Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita, Osaka, Japan
| |
Collapse
|
50
|
Ma J, Huang Y, Jia G, Dong X, Shi Q, Sun Y. Discovery of broad-spectrum high-affinity peptide ligands of spike protein for the vaccine purification of SARS-CoV-2 and Omicron variants. Int J Biol Macromol 2024; 283:137059. [PMID: 39500432 DOI: 10.1016/j.ijbiomac.2024.137059] [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: 09/13/2024] [Revised: 10/23/2024] [Accepted: 10/28/2024] [Indexed: 11/14/2024]
Abstract
To combat with emerging SARS-CoV-2 variants of concern (VOCs), we report the identification of a set of unique HWK-motif peptide ligands for the receptor-binding domain (RBD) of the SARS-CoV-2 spike (S) protein from a phage-displayed peptide library. These HWK-motif peptides exhibited nanomolar affinity for RBD. Among them, the peptide, HWKAVNWLKPWT (SP-HWK), had not only the highest affinities for RBD and trimer S protein, but also broad-spectrum affinities for RBDs from VOCs. Molecular dynamics simulations and competitive ELISA revealed a conserved pocket between the cryptic and the outer faces of RBD for SP-HWK binding, distinct from the human angiotensin-converting enzyme 2 receptor binding site. By coupling SP-HWK to agarose gel, the as-prepared affinity gel could efficiently capture RBD and trimer S from the ancestral strain and the Omicron variant, and the bound targets could be recovered by mild elution at pH 6.0. More importantly, the affinity gel presented excellent and stable chromatographic performance in the purification of inactivated SARS-CoV-2 and Omicron vaccines, affording high yields and purities, and strong HCP reduction. The results demonstrated the potential of SP-HWK as a broad-spectrum peptide ligand for developing a universal platform for the vaccine purification of SARS-CoV-2 and VOCs.
Collapse
Affiliation(s)
- Jing Ma
- Department of Biochemical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China
| | - Yongdong Huang
- State Key Laboratory of Biochemical Engineering and Key Laboratory of Biopharmaceutical Preparation and Delivery, Chinese Academy of Sciences, Beijing 100190, China
| | | | - Xiaoyan Dong
- Department of Biochemical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China; Key Laboratory of Systems Bioengineering and Frontiers Science Center for Synthetic Biology (Ministry of Education), Tianjin University, Tianjin 300350, China
| | - Qinghong Shi
- Department of Biochemical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China; Key Laboratory of Systems Bioengineering and Frontiers Science Center for Synthetic Biology (Ministry of Education), Tianjin University, Tianjin 300350, China.
| | - Yan Sun
- Department of Biochemical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China; Key Laboratory of Systems Bioengineering and Frontiers Science Center for Synthetic Biology (Ministry of Education), Tianjin University, Tianjin 300350, China.
| |
Collapse
|