1
|
Zhang DW, Xu XS, Xie L, Xu L, Fu Z, Li Y, Xu X. Natural product sennoside B disrupts liquid-liquid phase separation of SARS-CoV-2 nucleocapsid protein by inhibiting its RNA-binding activity. J Enzyme Inhib Med Chem 2025; 40:2501743. [PMID: 40371698 PMCID: PMC12082725 DOI: 10.1080/14756366.2025.2501743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 04/02/2025] [Accepted: 04/29/2025] [Indexed: 05/16/2025] Open
Abstract
The nucleocapsid protein (NP) of SARS-CoV-2, an RNA-binding protein, is capable of undergoing liquid-liquid phase separation (LLPS) during viral infection, which plays a crucial role in virus assembly, replication, and immune regulation. In this study, we developed a homogeneous time-resolved fluorescence (HTRF) method for identifying inhibitors of the SARS-CoV-2 NP-RNA interaction. Using this HTRF-based approach, we identified two natural products, sennoside A and sennoside B, as effective blockers of this interaction. Bio-layer interferometry assays confirmed that both sennosides directly bind to the NP, with binding sites located within the C-terminal domain. Additionally, fluorescence recovery after photobleaching (FRAP) experiments revealed that sennoside B significantly inhibited RNA-induced LLPS of the NP, while sennoside A displayed comparatively weaker activity. Thus, the developed HTRF-based assay is a valuable tool for identifying novel compounds that disrupt the RNA-binding activity and LLPS of the SARS-CoV-2 NP. Our findings may facilitate the development of antiviral drugs targeting SARS-CoV-2 NP.
Collapse
Affiliation(s)
- Da-Wei Zhang
- Institute of Bioinformatics and Medical Engineering, School of Electrical and Information Engineering, Jiangsu University of Technology, Changzhou, China
| | - Xiao-Shuang Xu
- Institute of Bioinformatics and Medical Engineering, School of Electrical and Information Engineering, Jiangsu University of Technology, Changzhou, China
| | - Liangxu Xie
- Institute of Bioinformatics and Medical Engineering, School of Electrical and Information Engineering, Jiangsu University of Technology, Changzhou, China
| | - Lei Xu
- Institute of Bioinformatics and Medical Engineering, School of Electrical and Information Engineering, Jiangsu University of Technology, Changzhou, China
| | - Zhiguo Fu
- Department of Orthopedics, Changzhou Hospital of Traditional Chinese Medicine, Changzhou, China
| | - Yimin Li
- College of Pharmacy and Key Laboratory for Research and Development of "Qin Medicine" of Shaanxi Administration of Chinese Medicine, Shaanxi University of Chinese Medicine, Xixian New District, China
| | - Xiaojun Xu
- Institute of Bioinformatics and Medical Engineering, School of Electrical and Information Engineering, Jiangsu University of Technology, Changzhou, China
| |
Collapse
|
2
|
Chen SC, Xu CT, Chang CF, Yang CS, Lin PH, Liu WM, Chen Y, Yu CH. Characterization of the binding features between SARS-CoV-2 5'-proximal transcripts of genomic RNA and nucleocapsid proteins. RNA Biol 2025; 22:1-16. [PMID: 40077853 PMCID: PMC11913385 DOI: 10.1080/15476286.2025.2471643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Revised: 02/01/2025] [Accepted: 02/19/2025] [Indexed: 03/14/2025] Open
Abstract
Packaging signals (PSs) of coronaviruses (CoVs) are specific RNA elements recognized by nucleocapsid (N) proteins that direct the selective packaging of genomic RNAs (gRNAs). These signals have been identified in the coding regions of the nonstructural protein 15 (Nsp 15) in CoVs classified under Embecovirus, a subgenus of betacoronaviruses (beta-CoVs). The PSs in other alpha- and beta-CoVs have been proposed to reside in the 5'-proximal regions of gRNAs, supported by comprehensive phylogenetic evidence. However, experimental data remain limited. In this study, we investigated the interactions between Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) 5'-proximal gRNA transcripts and N proteins using electrophoretic mobility shift assays (EMSAs). Our findings revealed that the in vitro synthesized 5'-proximal gRNA transcripts of CoVs can shift from a major conformation to alternative conformations. We also observed that the conformer comprising multiple stem-loops (SLs) is preferentially bound by N proteins. Deletions of the 5'-proximal structural elements of CoV gRNA transcripts, SL1 and SL5a/b/c in particular, were found to promote the formation of alternative conformations. Furthermore, we identified RNA-binding peptides from a pool derived from SARS-CoV N protein. These RNA-interacting peptides were shown to preferentially bind to wild-type SL5a RNA. In addition, our observations of N protein condensate formation in vitro demonstrated that liquid-liquid phase separation (LLPS) of N proteins with CoV-5'-UTR transcripts was influenced by the presence of SL5a/b/c. In conclusion, these results collectively reveal previously uncharacterized binding features between the 5'-proximal transcripts of CoV gRNAs and N proteins.
Collapse
Affiliation(s)
- Shih-Cheng Chen
- National Institute of Cancer Research, National Health Research Institutes, Tainan City, Taiwan
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan City, Taiwan
| | - Cui-Ting Xu
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan City, Taiwan
| | - Chuan-Fu Chang
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan City, Taiwan
| | - Chia-Shin Yang
- Institute of Translational Medicine and New Drug Development, College of Medicine, China Medical University, Taichung City, Taiwan
| | - Pin-Han Lin
- Department of Chemistry, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Wei-Min Liu
- Department of Chemistry, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Yeh Chen
- Institute of Translational Medicine and New Drug Development, College of Medicine, China Medical University, Taichung City, Taiwan
| | - Chien-Hung Yu
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan City, Taiwan
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan City, Taiwan
| |
Collapse
|
3
|
Wu JS, Kan JY, Chang YS, Le UNP, Su WC, Lai HC, Lin CW. Developing Zika virus-transduced hACE2 expression models for severe acute respiratory syndrome coronavirus 2 infection in vitro and in vivo. J Virol Methods 2025; 336:115166. [PMID: 40239870 DOI: 10.1016/j.jviromet.2025.115166] [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: 10/05/2024] [Revised: 04/10/2025] [Accepted: 04/11/2025] [Indexed: 04/18/2025]
Abstract
To address the human ACE2 dependence for SARS-CoV-2 infection, this study presents a novel strategy for generating ZIKV-hACE2 single-round infectious particles (SRIPs) by incorporating the hACE2 gene into a Zika virus (ZIKV) mini-replicon. SARS-CoV-2 SRIP infection was significantly enhanced in HEK293T cells pre-infected with ZIKV-hACE2, as evidenced by increased cytopathic effects and elevated mRNA and protein levels of the SARS-CoV-2 nucleocapsid (N) protein. A mouse model was also developed with this approach to investigate SARS-CoV-2 infection. Immunohistochemical and real-time RT-PCR analyses confirmed the presence of the SARS-CoV-2 N protein in the lungs of mice injected with ZIKV-hACE2 SRIPs, indicating successful infection. The mouse model displayed COVID-19-like pathological changes, including increased macrophages in BALF, severe lung damage, and elevated pro-inflammatory cytokines (IL-6 and IL-1β). These features mimic severe COVID-19 cases in humans. Additionally, treatment with nirmatrelvir resulted in a 6.2-fold reduction in viral load and a marked decrease in N protein levels. Overall, this ZIKV mini-replicon-mediated hACE2 expression model, both in vitro and in vivo, is a valuable tool for studying SARS-CoV-2 infection and evaluating therapeutic interventions. The mouse model's pathological features further underscore its relevance for in vivo research on SARS-CoV-2.
Collapse
Affiliation(s)
- Joh-Sin Wu
- PhD Program for Health Science and Industry, China Medical University, Taichung 404394, Taiwan; Department of Medical Laboratory Science and Biotechnology, China Medical University, Taichung 404394, Taiwan
| | - Ju-Ying Kan
- Department of Medical Laboratory Science and Biotechnology, China Medical University, Taichung 404394, Taiwan; The PhD program of Biotechnology and Biomedical Industry, China Medical University, Taichung 404394, Taiwan
| | - Young-Sheng Chang
- Department of Medical Laboratory Science and Biotechnology, China Medical University, Taichung 404394, Taiwan; Graduate Institute of Biomedical Sciences, China Medical University, Taichung 404394, Taiwan
| | - Uyen Nguyen Phuong Le
- Department of Medical Laboratory Science and Biotechnology, China Medical University, Taichung 404394, Taiwan; Department of Biological Science and Technology, China Medical University, Taichung 406040, Taiwan
| | - Wen-Chi Su
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 404394, Taiwan; International Master's Program of Biomedical Sciences, China Medical University, Taichung 404394, Taiwan
| | - Hsueh-Chou Lai
- Division of Hepato-Gastroenterology, Department of Internal Medicine, China Medical University Hospital, Taichung 404332, Taiwan
| | - Cheng-Wen Lin
- PhD Program for Health Science and Industry, China Medical University, Taichung 404394, Taiwan; Department of Medical Laboratory Science and Biotechnology, China Medical University, Taichung 404394, Taiwan; The PhD program of Biotechnology and Biomedical Industry, China Medical University, Taichung 404394, Taiwan; Graduate Institute of Biomedical Sciences, China Medical University, Taichung 404394, Taiwan; Division of Hepato-Gastroenterology, Department of Internal Medicine, China Medical University Hospital, Taichung 404332, Taiwan; Department of Medical Laboratory Science and Biotechnology, Asia University, Wufeng, Taichung 413305, Taiwan.
| |
Collapse
|
4
|
Lan HC, Hou BY, Chang ST, Kuo CY, Wang WC, Yao YL, Wu HY, Lai CC, Yang WM. Distinct Roles of SARS-CoV-2 N Protein and NFP in Host Cell Response Modulation. J Mol Biol 2025; 437:169094. [PMID: 40107650 DOI: 10.1016/j.jmb.2025.169094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 03/08/2025] [Accepted: 03/13/2025] [Indexed: 03/22/2025]
Abstract
The SARS-CoV-2 nucleocapsid (N) protein is crucial for viral replication and modulation of host cell responses. Here, we identify and characterize a novel N-fusion protein, designated NFP. NFP is derived from an alternative open reading frame spanning the N gene and the non-structural protein 1 (NSP1) sequence. While NFP shares some functional domains with the canonical N protein, it exhibits distinct structural features and protein interactions. NFP retains the ability to dimerize and bind RNA but lacks the formation of biomolecular condensates associated with N. Notably, NFP can dominantly interfere with N's condensate formation capacity when co-expressed. Functionally, NFP partially suppresses stress granule (SG) formation through a G3BP1-independent mechanism but gains the ability to interact with G3BP1 in the presence of N, potentially through N-NFP heterodimer formation. Post-translational modifications, particularly ubiquitination of specific lysine residues (K374 in N and K502 in NFP), differentially regulate the subcellular localization, SG inhibition, and cell cycle regulation activities of N and NFP. Our findings establish NFP as a distinct viral effector protein that modulates host cellular environments through both conserved and unique mechanisms compared to the canonical N protein, providing insights into SARS-CoV-2 pathogenesis and potential therapeutic targets.
Collapse
Affiliation(s)
- Hsin-Chi Lan
- Institute of Molecular Biology, National Chung Hsing University, Taichung 40227, Taiwan
| | - Bo-Yi Hou
- Institute of Molecular Biology, National Chung Hsing University, Taichung 40227, Taiwan
| | - Shu-Ting Chang
- Institute of Molecular Biology, National Chung Hsing University, Taichung 40227, Taiwan
| | - Cheng-Yu Kuo
- Institute of Molecular Biology, National Chung Hsing University, Taichung 40227, Taiwan
| | - Wei-Chen Wang
- Institute of Molecular Biology, National Chung Hsing University, Taichung 40227, Taiwan
| | - Ya-Li Yao
- Department of Medical Laboratory Science and Biotechnology, Asia University, Taichung 41354, Taiwan
| | - Hung-Yi Wu
- Graduate Institute of Veterinary Pathobiology, College of Veterinary Medicine, National Chung Hsing University, Taichung 40227, Taiwan
| | - Chien-Chen Lai
- Institute of Molecular Biology, National Chung Hsing University, Taichung 40227, Taiwan; Ph.D. Program in Translational Medicine, National Chung Hsing University, Taichung 40227, Taiwan
| | - Wen-Ming Yang
- Institute of Molecular Biology, National Chung Hsing University, Taichung 40227, Taiwan; Ph.D. Program in Translational Medicine, National Chung Hsing University, Taichung 40227, Taiwan.
| |
Collapse
|
5
|
Carra S, Fabian B, Taghavi H, Milanetti E, Giliberti V, Ruocco G, Shepherd J, Vendruscolo M, Fuxreiter M. Virus-like particles of retroviral origin in protein aggregation and neurodegenerative diseases. Mol Aspects Med 2025; 103:101369. [PMID: 40398193 DOI: 10.1016/j.mam.2025.101369] [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: 02/28/2025] [Revised: 05/06/2025] [Accepted: 05/10/2025] [Indexed: 05/23/2025]
Abstract
A wide range of human diseases are associated with protein misfolding and amyloid aggregates. Recent studies suggest that in certain neurological disorders, including Amyotrophic Lateral Sclerosis (ALS), Frontotemporal Dementia (FTD) and various tauopathies, protein aggregation may be promoted by virus-like particles (VLPs) formed by endogenous retroviruses (ERVs). The molecular mechanisms by which these VLPs contribute to protein aggregation, however, remain enigmatic. Here, we discuss possible molecular mechanisms of ERV-derived VLPs in the formation and spread of protein aggregates. An intriguing possibility is that liquid-like condensates may facilitate the formation of both protein aggregates and ERV-derived VLPs. We also describe how RNA chaperoning, and the encapsulation and trafficking of misfolded proteins, may contribute to protein homeostasis through the elimination of protein aggregates from cells. Based on these insights, we discuss future potential therapeutic opportunities.
Collapse
Affiliation(s)
- Serena Carra
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Balazs Fabian
- Department of Theoretical Biophysics, Max Planck Institute of Biophysics, Frankfurt, Germany
| | - Hamed Taghavi
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Edoardo Milanetti
- Department of Physics, Sapienza University, Rome, Italy; Center for Life Nano & Neuro Science, Istituto Italiano di Tecnologia, Rome, Italy
| | - Valeria Giliberti
- Department of Physics, Sapienza University, Rome, Italy; Center for Life Nano & Neuro Science, Istituto Italiano di Tecnologia, Rome, Italy
| | - Giancarlo Ruocco
- Department of Physics, Sapienza University, Rome, Italy; Center for Life Nano & Neuro Science, Istituto Italiano di Tecnologia, Rome, Italy
| | | | - Michele Vendruscolo
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, UK.
| | - Monika Fuxreiter
- Department of Biomedical Sciences, University of Padova, Padova, Italy; Department of Physics and Astronomy, University of Padova, Padova, Italy.
| |
Collapse
|
6
|
Peralta‐Martinez R, Visentin A, Salgueiro M, Borkosky SS, Aleixo MAA, Portugal RV, Sanchez IE, Prat‐Gay G. Protein-RNA condensation kinetics via filamentous nanoclusters. Protein Sci 2025; 34:e70136. [PMID: 40411406 PMCID: PMC12102730 DOI: 10.1002/pro.70136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 03/14/2025] [Accepted: 04/08/2025] [Indexed: 05/26/2025]
Abstract
Protein-RNA phase separation is at the center of membraneless biomolecular condensates governing cell physiology and pathology. Using an archetypical viral protein-RNA condensation model, we determined the sequence of events that starts with sub-second formation of a protomer with two RNAs per protein dimer. Association of additional RNA molecules to weaker secondary binding sites in this protomer kickstarts crystallization-like assembly of a molecular condensate. Primary nucleation is faster than the sum of secondary nucleation and growth, which is a multistep process. Protein-RNA nuclei grow over hundreds of seconds into filaments and subsequently into nanoclusters with approximately 600 nm diameter. Cryoelectron microscopy reveals an internal structure formed by incoming layers of protein-RNA filaments made of ribonucleoprotein oligomers, reminiscent of genome packing of a nucleocapsid. These nanoclusters progress to liquid condensate droplets that undergo further partial coalescence to yield typical hydrogel-like protein-RNA coacervates that may represent the scaffold of large viral factory condensates in infected cells. Our integrated experimental kinetic investigation exposes rate-limiting steps and structures along a key biological multistep pathway present across life kingdoms.
Collapse
Affiliation(s)
| | - Araceli Visentin
- Fundación Instituto Leloir, IIB‐BA, CONICETBuenos AiresArgentina
| | | | | | | | - Rodrigo Villares Portugal
- Brazilian Nanotechnology National Laboratory, CNPEMCampinasBrazil
- Biotechnoscience Program, Federal University of ABCSanto AndréBrazil
- Bioimaging and Biostructure Technologies Program, Federal University of Rio de JaneiroRio de JaneiroBrazil
| | - Ignacio Enrique Sanchez
- Laboratorio de Fisiología de Proteínas, Instituto de Química Biológica‐Facultad de Ciencias Exactas y Naturales IQUIBICEN‐Conicet, Universidad de Buenos AiresBuenos AiresArgentina
| | - Gonzalo Prat‐Gay
- Fundación Instituto Leloir, IIB‐BA, CONICETBuenos AiresArgentina
| |
Collapse
|
7
|
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
|
8
|
Amodeo D, Marchi S, Fiaschi L, Raucci L, Biba C, Salvestroni V, Trombetta CM, Manini I, Zazzi M, Montomoli E, Vicenti I, Cevenini G, Messina G. Analysis of the SARS-CoV-2 inactivation mechanism using violet-blue light (405 nm). Appl Environ Microbiol 2025:e0040325. [PMID: 40366184 DOI: 10.1128/aem.00403-25] [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: 02/18/2025] [Accepted: 04/15/2025] [Indexed: 05/15/2025] Open
Abstract
The study evaluated the effects of violet-blue light (VBL) on cell viability and replication, carbonylation of three structural proteins (S, E, and N) and one non-structural protein (NSP13), and direct damage to the RNA of SARS-CoV-2. The virus was exposed to increasing doses of VBL along with influenza A and B viruses to compare their susceptibility. At the highest dose (21.6 J/cm2), SARS-CoV-2 was significantly more susceptible to VBL than the influenza viruses, with a reduction in viral titer of 2.33 log10. Viral RNA did not show significant changes after exposure to VBL, as demonstrated by next-generation sequencing and real-time PCR quantification, suggesting that the inactivation process does not involve direct nucleic acid damage. To exclude the role of the culture suspension in the inactivation process, virus viability experiments were performed using different dilutions of Dulbecco's modified Eagle's medium (DMEM) in phosphate-buffered saline (PBS). The results indicated that the suspension medium played a secondary role in virus inactivation, as viability did not increase with increasing DMEM dilution. Subsequent tests with three different antioxidants (NAC, AsA, and SOD) at different concentrations prevented viral inactivation, from 99.99% to 85.43% (with SOD 0.003 mM). Carbonylation of S and E proteins was more pronounced when viruses were suspended in DMEM rather than PBS, although the tests demonstrated that the intrinsic properties of the viral membrane were a crucial element to consider in relation to its susceptibility to VBL.IMPORTANCELight-based disinfection methods are often used in combination with other cleaning methods due to their non-invasive nature, versatility, and environmental benefits. VBL is an effective approach as it induces the production of reactive oxygen species that reduce microbial viability. In this study, lipid peroxidation was identified as an important factor affecting the structural integrity and function of the viral envelope, reducing its ability to interact with host cells and consequently its ability to be infectious. The lipid envelope of SARS-CoV-2, composed mainly of glycerophospholipids and lacking cholesterol and sphingolipids, appears to be the critical factor in its susceptibility, distinguishing it from influenza viruses, which have a lipid profile richer in components that protect against oxidative stress.
Collapse
Affiliation(s)
- Davide Amodeo
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Serena Marchi
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Lia Fiaschi
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Luisa Raucci
- Department of Biotechnology, chemistry and pharmacy, University of Siena, Siena, Italy
| | - Camilla Biba
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Valentina Salvestroni
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | | | - Ilaria Manini
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Maurizio Zazzi
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Emanuele Montomoli
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
- VisMederi srl, Siena, Italy
| | - Ilaria Vicenti
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Gabriele Cevenini
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Gabriele Messina
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| |
Collapse
|
9
|
Fahoum J, Billan M, Varga JK, Padawer D, Britan-Rosich Y, Elgrably-Weiss M, Basu P, Stolovich-Rain M, Baraz L, Cohen-Kfir E, Kumari S, Oiknine-Djian E, Kumar M, Zelig O, Mayer G, Isupov MN, Wolf DG, Altuvia S, Wiener R, Schueler-Furman O, Rouvinski A. Transfer of SARS-CoV-2 nucleocapsid protein to uninfected epithelial cells induces antibody-mediated complement deposition. Cell Rep 2025; 44:115512. [PMID: 40343796 DOI: 10.1016/j.celrep.2025.115512] [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: 03/18/2024] [Revised: 12/28/2024] [Accepted: 03/12/2025] [Indexed: 05/11/2025] Open
Abstract
SARS-CoV-2 infection triggers a strong antibody response toward nucleocapsid protein (NP), suggesting its extracellular presence beyond intravirion RNA binding. Our co-culture experiments show NP decorates infected and proximal uninfected cell surfaces. We propose a mechanism whereby extracellular NP on uninfected cells contributes to COVID-19 pathogenicity. We show that NP binds to cell-surface sulfated glycosaminoglycans using its RNA-binding sites, facilitated by the flexible, positively charged linker. Coating uninfected lung-derived cells with NP attracted anti-NP IgG from lung fluids and sera of COVID-19 patients. Immune recognition was significantly higher in moderate versus mild COVID-19. Binding of anti-NP IgG in sera generated clusters, triggering C3b deposition via the classical complement pathway on SARS-CoV-2 non-susceptible cells co-cultured with infected cells. The heparin analog enoxaparin outcompeted NP binding, rescuing cells from anti-NP IgG-mediated complement deposition. Our findings reveal how extracellular NP may exacerbate COVID-19 damage and suggest preventative therapy avenues.
Collapse
Affiliation(s)
- Jamal Fahoum
- Department of Biochemistry, Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, The Hebrew University of Jerusalem, Jerusalem, Israel; Department of Microbiology and Molecular Genetics, Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Maria Billan
- Department of Microbiology and Molecular Genetics, Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, The Hebrew University of Jerusalem, Jerusalem, Israel; The Kuvin Center for the Study of Infectious and Tropical Diseases, The Hebrew University-Hadassah Medical School, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Julia K Varga
- Department of Microbiology and Molecular Genetics, Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Dan Padawer
- Department of Microbiology and Molecular Genetics, Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, The Hebrew University of Jerusalem, Jerusalem, Israel; Institute of Pulmonary Medicine, Hebrew University-Hadassah Medical Center, Jerusalem, Israel
| | - Yelena Britan-Rosich
- Barry Skolnick Biosafety Level 3 (BSL3) Unit, Core Research Facility, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Maya Elgrably-Weiss
- Department of Microbiology and Molecular Genetics, Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Pallabi Basu
- Department of Microbiology and Molecular Genetics, Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, The Hebrew University of Jerusalem, Jerusalem, Israel; Dove Laboratory, Boston Children's Hospital, Boston, MA 02115, USA
| | - Miri Stolovich-Rain
- Department of Microbiology and Molecular Genetics, Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, The Hebrew University of Jerusalem, Jerusalem, Israel; The Kuvin Center for the Study of Infectious and Tropical Diseases, The Hebrew University-Hadassah Medical School, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Leah Baraz
- Department of Microbiology and Molecular Genetics, Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, The Hebrew University of Jerusalem, Jerusalem, Israel; The Kuvin Center for the Study of Infectious and Tropical Diseases, The Hebrew University-Hadassah Medical School, The Hebrew University of Jerusalem, Jerusalem, Israel; Department of Medical Laboratory Sciences, Jerusalem Multidisciplinary College, Jerusalem, Israel
| | - Einav Cohen-Kfir
- Department of Biochemistry, Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Sujata Kumari
- Department of Biochemistry, Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, The Hebrew University of Jerusalem, Jerusalem, Israel; Department of Microbiology and Molecular Genetics, Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, The Hebrew University of Jerusalem, Jerusalem, Israel; The Kuvin Center for the Study of Infectious and Tropical Diseases, The Hebrew University-Hadassah Medical School, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Esther Oiknine-Djian
- Clinical Virology Unit, Hadassah Hebrew University Medical Center, Israel Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | - Manoj Kumar
- Department of Biochemistry, Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Orly Zelig
- Blood Bank, Hebrew University-Hadassah Medical Center, Jerusalem, Israel
| | - Guy Mayer
- The Institute of Chemistry, The Hebrew University of Jerusalem, Edmond J. Safra Campus, Jerusalem, Israel
| | - Michail N Isupov
- The Henry Wellcome Building for Biocatalysis, Biosciences, University of Exeter, Exeter, UK
| | - Dana G Wolf
- Clinical Virology Unit, Hadassah Hebrew University Medical Center, Israel Hadassah Hebrew University Medical Center, Jerusalem, Israel; Lautenberg Centre for Immunology and Cancer Research, The Institute for Medical Research Israel-Canada (IMRIC), Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Shoshy Altuvia
- Department of Microbiology and Molecular Genetics, Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Reuven Wiener
- Department of Biochemistry, Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, The Hebrew University of Jerusalem, Jerusalem, Israel.
| | - Ora Schueler-Furman
- Department of Microbiology and Molecular Genetics, Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, The Hebrew University of Jerusalem, Jerusalem, Israel.
| | - Alexander Rouvinski
- Department of Microbiology and Molecular Genetics, Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, The Hebrew University of Jerusalem, Jerusalem, Israel; The Kuvin Center for the Study of Infectious and Tropical Diseases, The Hebrew University-Hadassah Medical School, The Hebrew University of Jerusalem, Jerusalem, Israel.
| |
Collapse
|
10
|
Lugano D, Mwangi K, Mware B, Kibet G, Osiany S, Kiritu E, Dobi P, Muli C, Njeru R, de Oliveira T, Njenga MK, Routh A, Oyola SO. Characterization of SARS-CoV-2 intrahost genetic evolution in vaccinated and non-vaccinated patients from the Kenyan population. J Virol 2025:e0048225. [PMID: 40326760 DOI: 10.1128/jvi.00482-25] [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: 03/17/2025] [Accepted: 03/21/2025] [Indexed: 05/07/2025] Open
Abstract
Vaccination is a key control measure of coronavirus disease 2019 by preventing severe effects of disease outcomes, reducing hospitalization rates and death, and increasing immunity. However, vaccination can affect the evolution and adaptation of SARS-CoV-2 largely through vaccine-induced immune pressure. Here, we investigated intrahost recombination and single nucleotide variations (iSNVs) on the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) genome in non-vaccinated and vaccinated sequences from the Kenyan population to profile intrahost viral genetic evolution and adaptations driven by vaccine-induced immune pressure. We identified recombination hotspots in the S, N, and ORF1a/b genes and showed the genetic evolution landscape of SARS-CoV-2 by comparing within- and inter-wave recombination events from the beginning of the pandemic (June 2020 to December 2022) in Kenya. We further reveal differential expression of recombinant RNA species between vaccinated and non-vaccinated individuals and perform an in-depth analysis of iSNVs to identify and characterize the functional properties of non-synonymous mutations found in ORF-1 a/b, S, and N genes. Lastly, we detected a minority variant in non-vaccinated patients in Kenya, with an immune escape mutation S255F of the spike gene, and showed differential recombinant RNA species. Overall, this work identified unique in vivo mutations and intrahost recombination patterns in SARS-CoV-2, which could have significant implications for virus evolution, virulence, and immune escape.IMPORTANCEThe impact of vaccination on severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) genetic diversity in Kenya and much of Africa remains unknown. This can be attributed to lower sequencing rates; however, this information is relevant to improvement in vaccine and antiviral research. In this study, we investigated how vaccination and SARS-CoV-2 transmission waves affect intrahost non-homologous recombination and single nucleotide variations (iSNVs). We identified unique in vivo mutations and intrahost recombination patterns in SARS-CoV-2, which could have significant implications for virus evolution, virulence, and immune escape. We also demonstrate a methodology for studying genetic changes in a pathogen by a simultaneous analysis of both intrahost single nucleotide variations and recombination events. The study reveals the diversity of SARS-CoV-2 in Kenya and highlights the need for sustained genomic surveillance in Kenya and Africa to better understand how the virus evolves. Such surveillance ensures detection of drifts in evolution, allowing information for updates in vaccines, policy making, and containment of future variants of SARS-CoV-2.
Collapse
Affiliation(s)
- Doreen Lugano
- International Livestock Research Institute, Nairobi, Kenya
- Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch, Galveston, Texas, USA
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Kennedy Mwangi
- International Livestock Research Institute, Nairobi, Kenya
| | - Bernard Mware
- International Livestock Research Institute, Nairobi, Kenya
| | - Gilbert Kibet
- International Livestock Research Institute, Nairobi, Kenya
| | - Shebbar Osiany
- International Livestock Research Institute, Nairobi, Kenya
| | - Edward Kiritu
- International Livestock Research Institute, Nairobi, Kenya
| | - Paul Dobi
- International Livestock Research Institute, Nairobi, Kenya
| | - Collins Muli
- International Livestock Research Institute, Nairobi, Kenya
| | - Regina Njeru
- International Livestock Research Institute, Nairobi, Kenya
| | - Tulio de Oliveira
- Centre for Epidemic Response and Innovation (CERI), School of Data Science and Computational Thinking, Stellenbosch University, Stellenbosch, South Africa
| | - M Kariuki Njenga
- Washington State Global Health Program-Kenya, Washington State University, Pullman, Washington, USA
- Paul G. Allen School for Global Health, Washington State University, Pullman, Washington, USA
| | - Andrew Routh
- Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch, Galveston, Texas, USA
- Department of Immunology and Microbiology, Scripps Research, La Jolla, California, USA
| | - Samuel O Oyola
- International Livestock Research Institute, Nairobi, Kenya
| |
Collapse
|
11
|
Breznik JA, Cowbrough B, Bilaver L, Dushoff M, Stacey HD, Ang J, Clare R, Kennedy A, Costa AP, Nazy I, Loeb M, Verschoor CP, Bramson J, Miller MS, Bowdish DME. Minimal Impact of Prior Common Cold Coronavirus Exposure on Immune Responses to Severe Acute Respiratory Syndrome Coronavirus 2 Vaccination or Infection Risk in Older Adults in Congregate Care. Open Forum Infect Dis 2025; 12:ofaf178. [PMID: 40313478 PMCID: PMC12044602 DOI: 10.1093/ofid/ofaf178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 03/19/2025] [Indexed: 05/03/2025] Open
Abstract
Background Common cold coronaviruses were a frequent cause of respiratory infections in older adults living in congregate care homes before the coronavirus disease 2019 pandemic, which may influence immune responses to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccination and infection. We investigated humoral and cellular immune responses to prior common cold coronaviruses and SARS-CoV-2, how they are affected by SARS-CoV-2 vaccination and infection, and their associations with Omicron BA.1 SARS-CoV-2 infections in residents of long-term care and retirement homes. Methods In SARS-CoV-2 infection-naive residents with 3 monovalent messenger RNA SARS-CoV-2 vaccinations, we measured serum anti-receptor binding domain (RBD) immunoglobulin (Ig) G and IgA antibody titers against SARS-CoV-2 and common cold human coronavirus (HCoV) NL63, HCoV-OC43, and HCoV-229E; ancestral and Omicron BA.1 neutralizing antibodies; and CD4+ and CD8+ T-cell activation responses to membrane, nucleocapsid, and spike proteins. We examined the relationships of common cold coronavirus and SARS-CoV-2 humoral immune responses, whether antibody and T-cell responses changed after SARS-CoV-2 messenger RNA vaccination or infection, and their associations with Omicron BA.1 infection. Results Anti-RBD IgG HCoV-OC43 titers were positively correlated with SARS-CoV-2 anti-RBD IgG and neutralizing antibody titers. Common cold coronavirus anti-RBD IgA titers, but not anti-RBD IgG titers, increased after SARS-CoV-2 vaccination or infection, and many residents had cross-reactive T cells. Common cold coronavirus humoral immunity was similar in residents without and those with subsequent Omicron BA.1 infection. Conclusions Despite frequent exposure, and associations of common cold coronavirus and vaccine-induced SARS-CoV-2 humoral immunity, preexisting common cold coronavirus immunity was not associated with Omicron BA.1 infection in residents of long-term care and retirement communities.
Collapse
Affiliation(s)
- Jessica A Breznik
- Firestone Institute for Respiratory Health, St Joseph's Healthcare, Hamilton, Ontario, Canada
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
- M.G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
- McMaster Institute for Research on Aging, McMaster University, Hamilton, Ontario, Canada
| | - Braeden Cowbrough
- Firestone Institute for Respiratory Health, St Joseph's Healthcare, Hamilton, Ontario, Canada
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
- M.G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| | - Lucas Bilaver
- Firestone Institute for Respiratory Health, St Joseph's Healthcare, Hamilton, Ontario, Canada
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
- M.G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| | - Miriam Dushoff
- Firestone Institute for Respiratory Health, St Joseph's Healthcare, Hamilton, Ontario, Canada
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
- M.G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| | - Hannah D Stacey
- M.G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Jann Ang
- M.G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Rumi Clare
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- McMaster Centre for Transfusion Research, McMaster University, Hamilton, Ontario, Canada
- Thrombosis and Atherosclerosis Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Allison Kennedy
- Firestone Institute for Respiratory Health, St Joseph's Healthcare, Hamilton, Ontario, Canada
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
- M.G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| | - Andrew P Costa
- McMaster Institute for Research on Aging, McMaster University, Hamilton, Ontario, Canada
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, Ontario, Canada
| | - Ishac Nazy
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- McMaster Centre for Transfusion Research, McMaster University, Hamilton, Ontario, Canada
- Thrombosis and Atherosclerosis Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Mark Loeb
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
- M.G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| | - Chris P Verschoor
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
- Health Sciences North Research Institute, Health Sciences North, Sudbury, Ontario, Canada
| | - Jonathan Bramson
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Matthew S Miller
- M.G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Dawn M E Bowdish
- Firestone Institute for Respiratory Health, St Joseph's Healthcare, Hamilton, Ontario, Canada
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
- M.G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
- McMaster Institute for Research on Aging, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
12
|
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
|
13
|
Lazaro-Alfaro A, Nicholas SLN, Sanabria H. FRET-FCS: Advancing comprehensive insights into complex biological systems. Biophys J 2025:S0006-3495(25)00242-5. [PMID: 40241306 DOI: 10.1016/j.bpj.2025.04.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Revised: 03/02/2025] [Accepted: 04/14/2025] [Indexed: 04/18/2025] Open
Abstract
Förster resonance energy transfer (FRET) is a short-range distance-dependent photophysical phenomenon that allows the measurement of intra- and intermolecular distances through fluorescence detection. FRET measurements are sensitive to the movements of fluorescently labeled molecules as they produce fluorescence fluctuations. Fluorescence correlation spectroscopy (FCS) analyzes these fluctuations at faster and broader timescales (from picoseconds to seconds) compared with other techniques, unraveling the thermodynamic and kinetic properties of the system under study. Therefore, the combination of FRET and FCS (FRET-FCS) facilitates the analysis of molecular dynamics. Since its introduction, FRET-FCS has evolved into studying more sophisticated systems, requiring improvements in data acquisition and analysis. In this review, we discuss applications in the field of FRET-FCS that propose novel alternatives to overcome the inherent limitations of experimental setups. This work aims to promote using and enhancing FRET-FCS techniques to develop a comprehensive understanding of biological systems.
Collapse
Affiliation(s)
- Anay Lazaro-Alfaro
- Department of Physics and Astronomy, Clemson University, Clemson, South Carolina
| | | | - Hugo Sanabria
- Department of Physics and Astronomy, Clemson University, Clemson, South Carolina.
| |
Collapse
|
14
|
Gu L, Wang ZJ, Zhang XR, Liu Y, Zhao M, Jiang SZ, Pan J, Yuan Y, Cai H, Zhou T, Li T, Li TT, Xue W. Targeting the liquid-liquid phase separation of nucleocapsid broadly inhibits the replication of SARS-CoV-2 strains. Biochem Biophys Res Commun 2025; 756:151594. [PMID: 40086356 DOI: 10.1016/j.bbrc.2025.151594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Accepted: 03/05/2025] [Indexed: 03/16/2025]
Abstract
The COVID-19 pandemic, caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has led to a global public health crisis. The nucleocapsid (N) protein plays a pivotal role in a variety of biological processes in the life cycle of SARS-CoV-2, such as viral assembly. In this study, we investigated the liquid-liquid phase separation (LLPS) capacity of the N protein of seven SARS-CoV-2 strains, including the variants of concern (VOC) and interest (VOI), and its impact on viral replication. Using bioinformatic tools, we analyzed 11,433,558 complete genomes of SARS-CoV-2 and revealed a high degree of sequence conservation of N gene. While all the seven N proteins could undergo LLPS with RNA, the mutations in N impair its capacity of LLPS. With a SARS-CoV-2 trans-complementation system, we showed that SARS-CoV-2 variants carrying mutated N proteins exhibit impaired replication, highlighting the importance of LLPS of N in viral replication. We further demonstrated that (-)-gallocatechin gallate (GCG) efficiently inhibits the LLPS of N proteins and significantly suppresses the replication of different SARS-CoV-2 strains. Thus, our findings indicate that targeting the N-LLPS could be a viable strategy for the development of antiviral treatments against various SARS-CoV-2 strains, including those yet to emerge.
Collapse
Affiliation(s)
- Lin Gu
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100850, China
| | - Zheng-Jie Wang
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100850, China
| | - Xin-Ran Zhang
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100850, China
| | - Yu Liu
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100850, China
| | - Ming Zhao
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100850, China
| | - Shao-Zhen Jiang
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100850, China; School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Jie Pan
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100850, China
| | - Ying Yuan
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100850, China; Institute of Translational Medicine, Zhejiang University, Hangzhou, 310029, China
| | - Hong Cai
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100850, China
| | - Tao Zhou
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100850, China; Institute of Translational Medicine, Zhejiang University, Hangzhou, 310029, China
| | - Tao Li
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100850, China; School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China; Institute of Translational Medicine, Zhejiang University, Hangzhou, 310029, China
| | - Ting-Ting Li
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100850, China.
| | - Wen Xue
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100850, China.
| |
Collapse
|
15
|
Kubinski HC, Despres HW, Johnson BA, Schmidt MM, Jaffrani SA, Turner AH, Fanuele CD, Mills MG, Lokugamage KG, Dumas CM, Shirley DJ, Estes LK, Pekosz A, Crothers JW, Roychoudhury P, Greninger AL, Jerome KR, Di Genova BM, Walker DH, Ballif BA, Ladinsky MS, Bjorkman PJ, Menachery VD, Bruce EA. Variant mutation G215C in SARS-CoV-2 nucleocapsid enhances viral infection via altered genomic encapsidation. PLoS Biol 2025; 23:e3003115. [PMID: 40299982 PMCID: PMC12040272 DOI: 10.1371/journal.pbio.3003115] [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: 02/19/2025] [Accepted: 03/12/2025] [Indexed: 05/01/2025] Open
Abstract
The evolution of SARS-CoV-2 variants and their respective phenotypes represents an important set of tools to understand basic coronavirus biology as well as the public health implications of individual mutations in variants of concern. While mutations outside of spike are not well studied, the entire viral genome is undergoing evolutionary selection, with several variants containing mutations in the central disordered linker region of the nucleocapsid (N) protein. Here, we identify a mutation (G215C), characteristic of the Delta variant, that introduces a novel cysteine into this linker domain, which results in the formation of a more stable N-N dimer. Using reverse genetics, we determined that this cysteine residue is necessary and sufficient for stable dimer formation in a WA1 SARS-CoV-2 background, where it results in significantly increased viral growth both in vitro and in vivo. Mechanistically, we show that the N:G215C mutant has more encapsidation as measured by increased RNA binding to N, N incorporation into virions, and electron microscopy showing that individual virions are larger, with elongated morphologies.
Collapse
Affiliation(s)
- Hannah C. Kubinski
- Department of Microbiology and Molecular Genetics, Robert Larner, M.D. College of Medicine, University of Vermont, Burlington, Vermont, United States of America
| | - Hannah W. Despres
- Department of Microbiology and Molecular Genetics, Robert Larner, M.D. College of Medicine, University of Vermont, Burlington, Vermont, United States of America
| | - Bryan A. Johnson
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
- Institute for Human Infection and Immunity, University of Texas Medical Branch, Galveston, Texas, United States of America
- Center for Tropical Diseases, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Madaline M. Schmidt
- Department of Microbiology and Molecular Genetics, Robert Larner, M.D. College of Medicine, University of Vermont, Burlington, Vermont, United States of America
| | - Sara A. Jaffrani
- Department of Microbiology and Molecular Genetics, Robert Larner, M.D. College of Medicine, University of Vermont, Burlington, Vermont, United States of America
| | - Allyson H. Turner
- Department of Microbiology and Molecular Genetics, Robert Larner, M.D. College of Medicine, University of Vermont, Burlington, Vermont, United States of America
| | - Conor D. Fanuele
- Department of Microbiology and Molecular Genetics, Robert Larner, M.D. College of Medicine, University of Vermont, Burlington, Vermont, United States of America
| | - Margaret G. Mills
- Virology Division, Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, United States of America
| | - Kumari G. Lokugamage
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Caroline M. Dumas
- Department of Biology, University of Vermont, Burlington, Vermont, United States of America
| | - David J. Shirley
- Faraday, Inc. Data Science Department, Burlington, Vermont, United States of America
| | - Leah K. Estes
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Andrew Pekosz
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Jessica W. Crothers
- Department of Pathology and Laboratory Medicine, Robert Larner, MD College of Medicine, University of Vermont, Burlington, Vermont, United States of America
| | - Pavitra Roychoudhury
- Virology Division, Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, United States of America
| | - Alexander L. Greninger
- Virology Division, Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, United States of America
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle Washington, United States of America
| | - Keith R. Jerome
- Virology Division, Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, United States of America
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle Washington, United States of America
| | - Bruno Martorelli Di Genova
- Department of Microbiology and Molecular Genetics, Robert Larner, M.D. College of Medicine, University of Vermont, Burlington, Vermont, United States of America
| | - David H. Walker
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
- Institute for Human Infection and Immunity, University of Texas Medical Branch, Galveston, Texas, United States of America
- Center for Tropical Diseases, University of Texas Medical Branch, Galveston, Texas, United States of America
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Bryan A. Ballif
- Department of Biology, University of Vermont, Burlington, Vermont, United States of America
| | - Mark S. Ladinsky
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California, United States of America
| | - Pamela J. Bjorkman
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California, United States of America
| | - Vineet D. Menachery
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
- World Reference Center of Emerging Viruses and Arboviruses, University of Texas Medical Branch, Galveston, Texas, United States of America
- Center for Biodefense and Emerging Infectious Diseases, University of Texas Medical Branch, Galveston, Texas, United States of America
- Department of Pediatrics and Emory Vaccine Center, Emory University, Atlanta, Georgia, United States of America
| | - Emily A. Bruce
- Department of Microbiology and Molecular Genetics, Robert Larner, M.D. College of Medicine, University of Vermont, Burlington, Vermont, United States of America
| |
Collapse
|
16
|
Fang JL, Shrestha L, Beland FA. Flow cytometric analysis of the SARS coronavirus 2 antibodies in human plasma. Sci Rep 2025; 15:10300. [PMID: 40133428 PMCID: PMC11937374 DOI: 10.1038/s41598-025-92389-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 02/27/2025] [Indexed: 03/27/2025] Open
Abstract
COVID-19 is an infectious disease caused by the severe acute respiratory syndrome coronavirus (SARS-CoV-2). Anti-SARS-CoV-2 antibodies can provide information on patient immunity, identify asymptomatic patients, and track the spread of COVID-19. Efforts have been made to develop methods to detect anti-SARS-CoV-2 antibodies in humans. Here, we describe a flow cytometric assay for the simultaneous detection of anti-SARS-CoV-2 IgG and IgM in human plasma. To assess the antibody response against the different SARS-CoV-2 structural proteins, five viral recombinant proteins, including spike protein subunit 1 (S1), N-terminal domain of S1 (S1A), spike receptor-binding domain (RBD), spike protein subunit 2 (S2), and nucleocapsid protein (N), were generated. A comparison of the antibody profiles detected by the assay with plasma from 100 healthy blood donors collected prior to the COVID-19 pandemic and plasma from 100 virologically confirmed COVID-19 patients demonstrated a clear discrimination between the two groups. Among the COVID-19 patients, the antibody responses for the viral proteins, as determined by their prevalence, were anti-RBD IgG = anti-N IgG > anti-S1 IgG > anti-S1A IgG > anti-S2 IgG, and anti-RBD IgM > anti-S1 IgM > anti-N IgM > anti-S2 IgM. The prevalence of anti-SARS-CoV-2 IgG and IgM was not associated with sex, age, race, days after the onset of symptoms, or severity of illness, except for a higher prevalence of anti-S2 IgG being observed in men than in women. The levels of anti-RBD IgG were higher in patients 65 years and older and in patients who had severe symptoms. Similarly, patients who had severe symptoms exhibited higher levels of anti-S1 and anti-S1A IgG than patients who had mild or moderate symptoms. The levels of anti-RBD IgM tended to be higher in men but did not differ among age, race, days after the onset of symptoms, or severity of illness. Our study indicates that the flow cytometric assay, especially using RBD as target antigen, can be used to detect simultaneously anti-SARS-CoV-2 IgG and IgM antibodies in human plasma.
Collapse
Affiliation(s)
- Jia-Long Fang
- Division of Biochemical Toxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR, 72079, USA.
| | - Leeza Shrestha
- Division of Biochemical Toxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR, 72079, USA
| | - Frederick A Beland
- Division of Biochemical Toxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR, 72079, USA
| |
Collapse
|
17
|
Lotthammer JM, Holehouse AS. Disentangling Folding from Energetic Traps in Simulations of Disordered Proteins. J Chem Inf Model 2025; 65:2897-2910. [PMID: 40042172 DOI: 10.1021/acs.jcim.4c02005] [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: 03/25/2025]
Abstract
Protein conformational heterogeneity plays an essential role in a myriad of different biological processes. Extensive conformational heterogeneity is especially characteristic of intrinsically disordered proteins and protein regions (collectively IDRs), which lack a well-defined three-dimensional structure and instead rapidly exchange between a diverse ensemble of configurations. An emerging paradigm recognizes that the conformational biases encoded in IDR ensembles can play a central role in their biological function, necessitating understanding these sequence-ensemble relations. All-atom simulations have provided critical insight into our modern understanding of the solution behavior of IDRs. However, effectively exploring the accessible conformational space associated with large, heterogeneous ensembles is challenging. In particular, identifying poorly sampled or energetically trapped regions of disordered proteins in simulations often relies on qualitative assessment based on visual inspection of simulations and/or analysis data. These approaches, while convenient, run the risk of masking poorly sampled simulations. In this work, we present an algorithm for quantifying per-residue local conformational heterogeneity in protein simulations. Our work builds on prior work and compares the similarity between backbone dihedral angle distributions generated from molecular simulations in a limiting polymer model and across independent all-atom simulations. In this regime, the polymer model serves as a statistical reference model for extensive conformational heterogeneity in a real chain. Quantitative comparisons of probability vectors generated from these simulations reveal the extent of conformational sampling in a simulation, enabling us to distinguish between situations in which protein regions are well-sampled, poorly sampled, or folded. To demonstrate the effectiveness of this approach, we apply our algorithm to several toy, synthetic, and biological systems. Accurately assessing local conformational sampling in simulations of IDRs will help better quantify new enhanced sampling methods, ensure force field comparisons are equivalent, and provide confidence that conclusions drawn from simulations are robust.
Collapse
Affiliation(s)
- Jeffrey M Lotthammer
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, Missouri 63110, United States
- Center for Biomolecular Condensates (CBC), Washington University in St. Louis, St. Louis, Missouri 63110, United States
| | - Alex S Holehouse
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, Missouri 63110, United States
- Center for Biomolecular Condensates (CBC), Washington University in St. Louis, St. Louis, Missouri 63110, United States
| |
Collapse
|
18
|
Dhaka P, Mahto JK, Singh A, Kumar P, Tomar S. Structural insights into the RNA binding inhibitors of the C-terminal domain of the SARS-CoV-2 nucleocapsid. J Struct Biol 2025; 217:108197. [PMID: 40113149 DOI: 10.1016/j.jsb.2025.108197] [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: 10/09/2024] [Revised: 01/26/2025] [Accepted: 03/13/2025] [Indexed: 03/22/2025]
Abstract
The SARS-CoV-2 nucleocapsid (N) protein is an essential structural element of the virion, playing a crucial role in enclosing the viral genome into a ribonucleoprotein (RNP) assembly, as well as viral replication and transmission. The C-terminal domain of the N-protein (N-CTD) is essential for encapsidation, contributing to the stabilization of the RNP complex. In a previous study, three inhibitors (ceftriaxone, cefuroxime, and ampicillin) were screened for their potential to disrupt the RNA packaging process by targeting the N-protein. However, the binding efficacy, mechanism of RNA binding inhibition, and molecular insights of binding with N-CTD remain unclear. In this study, we evaluated the binding efficacy of these inhibitors using isothermal titration calorimetry (ITC), revealing the affinity of ceftriaxone (18 ± 1.3 μM), cefuroxime (55 ± 4.2 μM), and ampicillin (28 ± 1.2 μM) with the N-CTD. Further inhibition assay and fluorescence polarisation assay demonstrated RNA binding inhibition, with IC50 ranging from ∼ 12 to 18 μM and KD values between 24 μM to 32 μM for the inhibitors, respectively. Additionally, we also determined the inhibitor-bound complex crystal structures of N-CTD-Ceftriaxone (2.0 Å) and N-CTD-Ampicillin (2.2 Å), along with the structure of apo N-CTD (1.4 Å). These crystal structures revealed previously unobserved interaction sites involving residues K261, K266, R293, Q294, and W301 at the oligomerization interface and the predicted RNA-binding region of N-CTD. These findings provide valuable molecular insights into the inhibition of N-CTD, highlighting its potential as an underexplored but promising target for the development of novel antiviral agents against coronaviruses.
Collapse
Affiliation(s)
- Preeti Dhaka
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand 247667, India
| | - Jai Krishna Mahto
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand 247667, India
| | - Ankur Singh
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand 247667, India
| | - Pravindra Kumar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand 247667, India.
| | - Shailly Tomar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand 247667, India.
| |
Collapse
|
19
|
López-Muñoz AD, Yewdell JW. Chemokines simultaneously bind SARS-CoV-2 nucleocapsid protein RNA-binding and dimerization domains. Virol J 2025; 22:77. [PMID: 40097964 PMCID: PMC11912793 DOI: 10.1186/s12985-025-02658-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 02/11/2025] [Indexed: 03/19/2025] Open
Abstract
Viruses express chemokine (CHK)-binding proteins to interfere with the host CHK network and thereby modulate leukocyte migration. SARS-CoV-2 Nucleocapsid (N) protein binds a subset of human CHKs with high affinity, inhibiting their chemoattractant properties. Here, we report that both N's RNA-binding and dimerization domains participate individually in CHK binding. CHKs typically possess independent sites for binding glycosaminoglycans (GAG) and their receptor proteins. We show that the interaction with the N protein occurs through the CHK GAG-binding site, pointing the way to developing compounds that block this interaction for potential anti-coronavirus therapeutics.
Collapse
Affiliation(s)
| | - Jonathan W Yewdell
- Cellular Biology Section, Laboratory of Viral Diseases, NIAID (NIH), Bethesda, MD, USA.
| |
Collapse
|
20
|
Liustrovaite V, Ratautaite V, Ramanaviciene A, Ramanavicius A. Detection of the SARS-CoV-2 nucleoprotein by electrochemical biosensor based on molecularly imprinted polypyrrole formed on self-assembled monolayer. Biosens Bioelectron 2025; 272:117092. [PMID: 39787822 DOI: 10.1016/j.bios.2024.117092] [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: 06/18/2024] [Revised: 12/12/2024] [Accepted: 12/21/2024] [Indexed: 01/12/2025]
Abstract
Herein, we report the development and characterisation of an electrochemical biosensor with a polypyrrole (Ppy)-based molecularly imprinted polymer (MIP) for the serological detection of the recombinant nucleocapsid protein of SARS-CoV-2 (rN). The electrochemical biosensor utilises a Ppy-based MIP formed on a self-assembled monolayer (SAM) at the gold interface to enhance Ppy layer stability on the screen-printed electrode (SPE). Electrochemical impedance spectroscopy (EIS) and square wave voltammetry (SWV) were employed for the electrochemical characterisation of screen-printed gold electrodes (SPGEs) modified with MIP or non-imprinted polymer (NIP) layers. Removing the rN protein template from the MIP layer increased electron transfer and decreased impedance, indicating the specificity of molecular imprinting. The electrochemical biosensor with a Ppy-based MIP exhibited higher sensitivity than the NIP counterpart, demonstrating its potential for selective rN protein detection. The limit of detection 0.4 nM and 0.2 nM and the limit of quantification 1.3 nM and 0.66 nM values obtained through SWV and EIS, respectively, highlight the biosensor's ability to detect low target protein concentrations. The specificity test confirmed minimal nonspecific binding, reinforcing the reliability of the novel electrochemical sensor with a Ppy-based MIP.
Collapse
Affiliation(s)
- Viktorija Liustrovaite
- NanoTechnas - Center of Nanotechnology and Materials Science, Institute of Chemistry, Faculty of Chemistry and Geosciences, Vilnius University (VU), Naugarduko St. 24, LT-03225, Vilnius, Lithuania; Department of Physical Chemistry, Institute of Chemistry, Faculty of Chemistry and Geosciences, Vilnius University (VU), Naugarduko St. 24, LT-03225, Vilnius, Lithuania
| | - Vilma Ratautaite
- Department of Physical Chemistry, Institute of Chemistry, Faculty of Chemistry and Geosciences, Vilnius University (VU), Naugarduko St. 24, LT-03225, Vilnius, Lithuania; Department of Nanotechnology, State Research Institute Center for Physical and Technological Sciences (FTMC), Sauletekio Ave. 3, LT-10257, Vilnius, Lithuania
| | - Almira Ramanaviciene
- NanoTechnas - Center of Nanotechnology and Materials Science, Institute of Chemistry, Faculty of Chemistry and Geosciences, Vilnius University (VU), Naugarduko St. 24, LT-03225, Vilnius, Lithuania
| | - Arunas Ramanavicius
- Department of Physical Chemistry, Institute of Chemistry, Faculty of Chemistry and Geosciences, Vilnius University (VU), Naugarduko St. 24, LT-03225, Vilnius, Lithuania; Department of Nanotechnology, State Research Institute Center for Physical and Technological Sciences (FTMC), Sauletekio Ave. 3, LT-10257, Vilnius, Lithuania.
| |
Collapse
|
21
|
Lugano D, Mwangi K, Mware B, Kibet G, Osiany S, Kiritu E, Dobi P, Muli C, Njeru R, de Oliveira T, Njenga MK, Routh A, Oyola SO. Characterization of SARS-CoV-2 intrahost genetic evolution in vaccinated and non-vaccinated patients from the Kenyan population. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.03.03.25323296. [PMID: 40093225 PMCID: PMC11908290 DOI: 10.1101/2025.03.03.25323296] [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: 03/19/2025]
Abstract
Vaccination is a key control measure of COVID-19 by preventing severe effects of disease outcomes, reducing hospitalization rates and death, and increasing immunity. However, vaccination can affect the evolution and adaptation of SARS-CoV-2, largely through vaccine-induced immune pressure. Here we investigated intrahost recombination and single nucleotide variations (iSNVs) on the SARS-CoV-2 genome in non-vaccinated and vaccinated sequences from the Kenyan population to profile intrahost viral genetic evolution and adaptations driven by vaccine-induced immune pressure. We identified recombination hotspots in the S, N, and ORF1a/b genes and showed the genetic evolution landscape of SARS-CoV-2 by comparing within-wave and inter-wave recombination events from the beginning of the pandemic (June 2020) to (December 2022) in Kenya. We further reveal differential expression of recombinant RNA species between vaccinated and non-vaccinated individuals and perform an in-depth analysis of iSNVs to identify and characterize the functional properties of non-synonymous mutations found in ORF-1 a/b, S, and N genes. Lastly, we detected a minority variant in non-vaccinated patients in Kenya, with an immune escape mutation S255F of the spike gene and showed differential recombinant RNA species. Overall, this work identified unique in vivo mutations and intrahost recombination patterns in SARS-CoV-2 which could have significant implications for virus evolution, virulence, and immune escape.
Collapse
Affiliation(s)
- Doreen Lugano
- International Livestock Research Institute, P.O. Box 30709, 00100 GPO, Uthiru, Naivasha road, Nairobi-Kenya
- Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch, Galveston, Texas, 77550, USA
- KEMRI-Wellcome Trust Research Programme, P.O. Box 230 Kilifi, Kenya
| | - Kennedy Mwangi
- International Livestock Research Institute, P.O. Box 30709, 00100 GPO, Uthiru, Naivasha road, Nairobi-Kenya
| | - Bernard Mware
- International Livestock Research Institute, P.O. Box 30709, 00100 GPO, Uthiru, Naivasha road, Nairobi-Kenya
| | - Gilbert Kibet
- International Livestock Research Institute, P.O. Box 30709, 00100 GPO, Uthiru, Naivasha road, Nairobi-Kenya
| | - Shebbar Osiany
- International Livestock Research Institute, P.O. Box 30709, 00100 GPO, Uthiru, Naivasha road, Nairobi-Kenya
| | - Edward Kiritu
- International Livestock Research Institute, P.O. Box 30709, 00100 GPO, Uthiru, Naivasha road, Nairobi-Kenya
| | - Paul Dobi
- International Livestock Research Institute, P.O. Box 30709, 00100 GPO, Uthiru, Naivasha road, Nairobi-Kenya
| | - Collins Muli
- International Livestock Research Institute, P.O. Box 30709, 00100 GPO, Uthiru, Naivasha road, Nairobi-Kenya
| | - Regina Njeru
- International Livestock Research Institute, P.O. Box 30709, 00100 GPO, Uthiru, Naivasha road, Nairobi-Kenya
| | - Tulio de Oliveira
- Centre for Epidemic Response and Innovation (CERI), School of Data Science and Computational Thinking, Stellenbosch University, Stellenbosch, South Africa
| | - M. Kariuki Njenga
- Washington State Global Health Program-Kenya, Washington State University, Nairobi 00200, Kenya
- Paul G. Allen School for Global Health, Washington State University, Pullman, WA 99164, USA
| | - Andrew Routh
- Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch, Galveston, Texas, 77550, USA
- Dept Immunology and Microbiology, Scripps Research, La Jolla, CA, 92037
| | - Samuel O. Oyola
- International Livestock Research Institute, P.O. Box 30709, 00100 GPO, Uthiru, Naivasha road, Nairobi-Kenya
| |
Collapse
|
22
|
Firdaus MER, Dukhno E, Kapoor R, Gerlach P. Two Birds With One Stone: RNA Virus Strategies to Manipulate G3BP1 and Other Stress Granule Components. WILEY INTERDISCIPLINARY REVIEWS. RNA 2025; 16:e70005. [PMID: 40170442 PMCID: PMC11962251 DOI: 10.1002/wrna.70005] [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] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 01/29/2025] [Accepted: 01/30/2025] [Indexed: 04/03/2025]
Abstract
Stress granules (SGs) are membrane-less organelles forming in the cytoplasm in response to various types of stress, including viral infection. SGs and SG-associated proteins can play either a proviral role, by facilitating viral replication, or an antiviral role, by limiting the translation capacity, sequestering viral RNA, or contributing to the innate immune response of the cell. Consequently, viruses frequently target stress granules while counteracting cellular translation shut-off and the antiviral response. One strategy is to sequester SG components, not only to impair their assembly but also to repurpose and incorporate them into viral replication sites. G3BP1 is a key SG protein, driving its nucleation through protein-protein and protein-RNA interactions. Many cellular proteins, including other SG components, interact with G3BP1 via their ΦxFG motifs. Notably, SARS-CoV N proteins and alphaviral nsP3 proteins contain similar motifs, allowing them to compete for G3BP1. Several SG proteins have been shown to interact with the flaviviral capsid protein, which is primarily responsible for anchoring the viral genome inside the virion. There are also numerous examples of structured elements within coronaviral and flaviviral RNAs recruiting or sponging SG proteins. Despite these insights, the structural and biochemical details of SG-virus interactions remain largely unexplored and are known only for a handful of cases. Exploring their molecular relevance for infection and discovering new examples of direct SG-virus contacts is highly important, as advances in this area will open new possibilities for the design of targeted therapies and potentially broad-spectrum antivirals.
Collapse
Affiliation(s)
- Moh Egy Rahman Firdaus
- IMol Polish Academy of SciencesWarsawPoland
- ReMedy International Research Agenda UnitIMol Polish Academy of SciencesWarsawPoland
| | - Eliana Dukhno
- IMol Polish Academy of SciencesWarsawPoland
- ReMedy International Research Agenda UnitIMol Polish Academy of SciencesWarsawPoland
| | | | - Piotr Gerlach
- IMol Polish Academy of SciencesWarsawPoland
- ReMedy International Research Agenda UnitIMol Polish Academy of SciencesWarsawPoland
| |
Collapse
|
23
|
Quek RT, Shirazinejad CR, Young CL, Hardy KS, Lim S, Elms PJ, McSwiggen DT, Mitchison TJ, Silver PA. Comparative evaluation of cell-based assay technologies for scoring drug-induced condensation of SARS-CoV-2 nucleocapsid protein. SLAS DISCOVERY : ADVANCING LIFE SCIENCES R & D 2025; 31:100220. [PMID: 39894078 DOI: 10.1016/j.slasd.2025.100220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 01/23/2025] [Accepted: 01/30/2025] [Indexed: 02/04/2025]
Abstract
Protein-nucleic acid phase separation has been implicated in many diseases such as viral infections, neurodegeneration, and cancer. There is great interest in identifying condensate modulators (CMODs), which are small molecules that alter the dynamics and functions of phase-separated condensates, as a potential therapeutic modality. Most CMODs were identified in cellular high-content screens (HCS) where micron-scale condensates were characterized by fluorescence microscopy. These approaches lack information on protein dynamics, are limited by microscope resolution, and are insensitive to subtle condensation phenotypes missed by overfit analysis pipelines. Here, we evaluate two alternative cell-based assays: high-throughput single molecule tracking (htSMT) and proximity-based condensate biosensors using NanoBIT (split luciferase) and NanoBRET (bioluminescence resonance energy transfer) technologies. We applied these methods to evaluate condensation of the SARS-CoV-2 nucleocapsid (N) protein under GSK3 inhibitor treatment, which we had previously identified in our HCS campaign to induce condensation with well-defined structure-activity relationships (SAR). Using htSMT, we observed robust changes in N protein diffusion as early as 3 h post GSK3 inhibition. Proximity-based N biosensors also reliably reported on condensation, enabling the rapid assaying of large compound libraries with a readout independent of imaging. Both htSMT and proximity-based biosensors performed well in a screening format and provided information on CMOD activity that was complementary to HCS. We expect that this expanded toolkit for interrogating phase-separated proteins will accelerate the identification of CMODs for important therapeutic targets.
Collapse
Affiliation(s)
- Rui Tong Quek
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | | | | | - Kierra S Hardy
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Samuel Lim
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | | | | | | | - Pamela A Silver
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA.
| |
Collapse
|
24
|
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
|
25
|
Wu Y, Lv B, Ni X, Zhu S, Li D. "All-U-Want" Strand Displacement Amplification: A Versatile Signal Amplification Method for Nucleic Acid Biosensing. ACS Sens 2025; 10:965-976. [PMID: 39951690 DOI: 10.1021/acssensors.4c02765] [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: 02/16/2025]
Abstract
Strand displacement amplification (SDA) is an isothermal DNA amplification technique. Herein, we developed a novel SDA system, designated All-U-Want SDA (AUW-SDA), which was used as a signal amplification strategy for the construction of nucleic acid detection biosensors. AUW-SDA is capable of target turnover and can be utilized for detection of nucleic acid sequences without available 3'-ends. Of particular significance is the ability of AUW-SDA to generate a substantial number of programmable sequences in accordance with the specifications of the sensor signal output methods, irrespective of the sequence of the target nucleic acid. We used the N gene of SARS-CoV-2 as a model target to develop a sensing platform with dual signal outputs. The colorimetric signals were generated by the G-quadruplex/hemin DNAzyme, in which the G-rich sequences were produced by AUW-SDA with a C-rich primer. On the other hand, by altering the sequence within the replaceable region of the primer, an activator sequence was obtained from AUW-SDA, which could trigger the activity of CRISPR/Cas12a, cleaving the probes modified with a fluorophore and quencher at each end and subsequently yielding the fluorescent signals. After the DNA sequences and reaction conditions were optimized, the limit of detection (LOD) values of the fluorescent and colorimetric assays were estimated to be 0.672 fM and 13.3 fM, respectively. The biosensors were utilized for biological sample detection. The reliability of the proposed method was validated against RT-qPCR results. In addition, a portable scanner-assisted high-throughput RGB analysis (PSHRA) method was developed. This method was applied to our biosensor for multilocus detection of SARS-CoV-2. The results obtained were satisfactory, indicating the potential of this approach for field testing or point-of-care (POC) diagnostics.
Collapse
Affiliation(s)
- Yapeng Wu
- Key Laboratory of State Forestry and Grassland Administration on Subtropical Forest Biodiversity Conservation, College of Life Sciences, Nanjing Forestry University, Nanjing 210037, China
| | - Bei Lv
- Key Lab of Innovative Applications of Bioresources and Functional Molecules of Jiangsu Province, College of Life Science and Chemistry, Jiangsu Second Normal University, Nanjing 210013, China
| | - Xintian Ni
- Key Lab of Innovative Applications of Bioresources and Functional Molecules of Jiangsu Province, College of Life Science and Chemistry, Jiangsu Second Normal University, Nanjing 210013, China
| | - Sheng Zhu
- Key Laboratory of State Forestry and Grassland Administration on Subtropical Forest Biodiversity Conservation, College of Life Sciences, Nanjing Forestry University, Nanjing 210037, China
| | - Dawei Li
- Key Laboratory of State Forestry and Grassland Administration on Subtropical Forest Biodiversity Conservation, College of Life Sciences, Nanjing Forestry University, Nanjing 210037, China
| |
Collapse
|
26
|
Hnasko RM, Lin AV, McGarvey JA, Jackson ES. Immunoassay Detection of SARS-CoV-2 Using Monoclonal Antibody Binding to Viral Nucleocapsid Protein. Microb Biotechnol 2025; 18:e70117. [PMID: 39989430 PMCID: PMC11848557 DOI: 10.1111/1751-7915.70117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 02/07/2025] [Accepted: 02/10/2025] [Indexed: 02/25/2025] Open
Abstract
Immunoassays represent sensitive, easy-to-use, and cost-effective tests useful for the detection of the SARS-CoV-2 virus. In this manuscript, we report on the binding specificity of a pair of novel monoclonal antibodies (MAbs) generated against the SARS-CoV-2 nucleocapsid protein (NP) and their development into sensitive sandwich enzyme-linked immunosorbent assays (sELISA) and a lateral flow immunoassay (LFIA). Binding of these MAbs to hCoVs is limited to variants of SARS-CoV-2 and SARS-CoV NP. Chemiluminescent and absorbance spectroscopy sELISAs report a limit of detection (LOD) for the SARS-CoV-2 B.1.1.529 NP variant at 15 pg/mL, and the LFIA using a red-dyed 200 nm particle at 10 ng/mL. The sELISA exhibits broad SARS-CoV-2 viral variant detection with assay LOD for SARS-CoV-2 B.1.1.529 virus at 1.4 × 105 genome copies per mL (p ≤ 0.001). The availability of these MAbs should facilitate continued investment in the commercial development of immunoassays to increase global SARS-CoV-2 detection technologies.
Collapse
Affiliation(s)
- Robert M. Hnasko
- United States Department of Agriculture, Agricultural Research ServiceProduce Safety and Microbiology UnitAlbanyCaliforniaUSA
| | - Alice V. Lin
- United States Department of Agriculture, Agricultural Research ServiceProduce Safety and Microbiology UnitAlbanyCaliforniaUSA
| | - Jeffery A. McGarvey
- United States Department of Agriculture, Agricultural Research ServiceFoodborne Toxin Detection and Prevention UnitAlbanyCaliforniaUSA
| | - Eric S. Jackson
- United States Department of Agriculture, Agricultural Research ServiceFoodborne Toxin Detection and Prevention UnitAlbanyCaliforniaUSA
| |
Collapse
|
27
|
Nuccetelli V, Mghezzi-Habellah M, Deymier S, Roisin A, Gérard-Baraggia F, Rocchi C, Coureux PD, Gouet P, Cimarelli A, Mocquet V, Fiorini F. The SARS-CoV-2 nucleocapsid protein interferes with the full enzymatic activation of UPF1 and its interaction with UPF2. Nucleic Acids Res 2025; 53:gkaf010. [PMID: 39831305 PMCID: PMC11744187 DOI: 10.1093/nar/gkaf010] [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: 07/17/2024] [Revised: 01/01/2025] [Accepted: 01/06/2025] [Indexed: 01/22/2025] Open
Abstract
The nonsense-mediated mRNA decay (NMD) pathway triggers the degradation of defective mRNAs and governs the expression of mRNAs with specific characteristics. Current understanding indicates that NMD is often significantly suppressed during viral infections to protect the viral genome. In numerous viruses, this inhibition is achieved through direct or indirect interference with the RNA helicase UPF1, thereby promoting viral replication and enhancing pathogenesis. In this study, we employed biochemical, biophysical assays and cellular investigations to explore the interplay between UPF1 and the nucleocapsid (Np) protein of SARS-CoV-2. We evaluated their direct interaction and its impact on inhibiting cellular NMD. Furthermore, we characterized how this interaction affects UPF1's enzymatic function. Our findings demonstrate that Np inhibits the unwinding activity of UPF1 by physically obstructing its access to structured nucleic acid substrates. Additionally, we showed that Np binds directly to UPF2, disrupting the formation of the UPF1/UPF2 complex essential for NMD progression. Intriguingly, our research also uncovered a surprising pro-viral role of UPF1 and an antiviral function of UPF2. These results unveil a novel, multi-faceted mechanism by which SARS-CoV-2 evades the host's defenses and manipulates cellular components. This underscores the potential therapeutic strategy of targeting Np-UPF1/UPF2 interactions to treat COVID-19.
Collapse
Affiliation(s)
- Veronica Nuccetelli
- Molecular Microbiology and Structural Biochemistry, MMSB-IBCP, CNRS UMR 5086 , Université Claude Bernard Lyon 1, F-69367 Lyon, France
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, I-00185 Rome, Italy
| | - Makram Mghezzi-Habellah
- Laboratoire de Biologie et Modélisation de la Cellule, Ecole Normale Supérieure de Lyon, CNRS UMR 5239, Inserm U1293, Université Claude Bernard Lyon 1, F-69364 Lyon, France
| | - Séverine Deymier
- Centre International de Recherche en Infectiologie (CIRI), Ecole Normale Supérieure de Lyon, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR5308, F-69364 Lyon, France
| | - Armelle Roisin
- Laboratoire de Biologie et Modélisation de la Cellule, Ecole Normale Supérieure de Lyon, CNRS UMR 5239, Inserm U1293, Université Claude Bernard Lyon 1, F-69364 Lyon, France
| | - Francine Gérard-Baraggia
- Molecular Microbiology and Structural Biochemistry, MMSB-IBCP, CNRS UMR 5086 , Université Claude Bernard Lyon 1, F-69367 Lyon, France
| | - Cecilia Rocchi
- Molecular Microbiology and Structural Biochemistry, MMSB-IBCP, CNRS UMR 5086 , Université Claude Bernard Lyon 1, F-69367 Lyon, France
- Division of Structural Biology - Nuffield Department of Medicine, University of Oxford, OX3 7BN Oxford, England
| | - Pierre-Damien Coureux
- Molecular Microbiology and Structural Biochemistry, MMSB-IBCP, CNRS UMR 5086 , Université Claude Bernard Lyon 1, F-69367 Lyon, France
| | - Patrice Gouet
- Molecular Microbiology and Structural Biochemistry, MMSB-IBCP, CNRS UMR 5086 , Université Claude Bernard Lyon 1, F-69367 Lyon, France
| | - Andrea Cimarelli
- Centre International de Recherche en Infectiologie (CIRI), Ecole Normale Supérieure de Lyon, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR5308, F-69364 Lyon, France
| | - Vincent Mocquet
- Laboratoire de Biologie et Modélisation de la Cellule, Ecole Normale Supérieure de Lyon, CNRS UMR 5239, Inserm U1293, Université Claude Bernard Lyon 1, F-69364 Lyon, France
| | - Francesca Fiorini
- Molecular Microbiology and Structural Biochemistry, MMSB-IBCP, CNRS UMR 5086 , Université Claude Bernard Lyon 1, F-69367 Lyon, France
| |
Collapse
|
28
|
Ivancová I, Quirante TS, Ondruš M, Pohl R, Vlková M, Žilecká E, Bouřa E, Hocek M. Enzymatic synthesis of reactive RNA probes containing squaramate-linked cytidine or adenosine for bioconjugations and cross-linking with lysine-containing peptides and proteins. Commun Chem 2025; 8:1. [PMID: 39748090 PMCID: PMC11696893 DOI: 10.1038/s42004-024-01399-6] [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: 07/08/2024] [Accepted: 12/18/2024] [Indexed: 01/04/2025] Open
Abstract
Protein-RNA interactions play important biological roles and hence reactive RNA probes for cross-linking with proteins are important tools in their identification and study. To this end, we designed and synthesized 5'-O-triphosphates bearing a reactive squaramate group attached to position 5 of cytidine or position 7 of 7-deazaadenosine and used them as substrates for polymerase synthesis of modified RNA. In vitro transcription with T7 RNA polymerase or primer extension using TGK polymerase was used for synthesis of squaramate-modified RNA probes which underwent covalent bioconjugations with amine-linked fluorophore and lysine-containing peptides and proteins including several viral RNA polymerases or HIV reverse transcriptase. Inhibition of RNA-depending RNA polymerases from Japanese Encephalitis virus was observed through formation of covalent cross-link which was partially identified by MS/MS analysis. Thus, the squaramate-linked NTP analogs are useful building blocks for the synthesis of reactive RNA probes for bioconjugations with primary amines and cross-linking with lysine residues.
Collapse
Affiliation(s)
- Ivana Ivancová
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Flemingovo nam. 2, CZ-16000 Prague 6, Prague, Czech Republic
| | - Tania Sánchez Quirante
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Flemingovo nam. 2, CZ-16000 Prague 6, Prague, Czech Republic
- Department of Organic Chemistry, Faculty of Science, Charles University, Hlavova 8, CZ-12843 Prague 2, Prague, Czech Republic
| | - Marek Ondruš
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Flemingovo nam. 2, CZ-16000 Prague 6, Prague, Czech Republic
| | - Radek Pohl
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Flemingovo nam. 2, CZ-16000 Prague 6, Prague, Czech Republic
| | - Marta Vlková
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Flemingovo nam. 2, CZ-16000 Prague 6, Prague, Czech Republic
| | - Eva Žilecká
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Flemingovo nam. 2, CZ-16000 Prague 6, Prague, Czech Republic
| | - Evžen Bouřa
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Flemingovo nam. 2, CZ-16000 Prague 6, Prague, Czech Republic
| | - Michal Hocek
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Flemingovo nam. 2, CZ-16000 Prague 6, Prague, Czech Republic.
- Department of Organic Chemistry, Faculty of Science, Charles University, Hlavova 8, CZ-12843 Prague 2, Prague, Czech Republic.
| |
Collapse
|
29
|
Etibor TA, Paixão T, Amorim MJ. Fluorescence Loss After Photoactivation (FLAPh): A Pulse-Chase Cellular Assay for Understanding Kinetics and Dynamics of Viral Inclusions. Methods Mol Biol 2025; 2890:125-140. [PMID: 39890724 DOI: 10.1007/978-1-0716-4326-6_6] [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: 02/03/2025]
Abstract
Influenza A virus (IAV) relies on host cellular machinery for replication. Upon infection, the eight genomic segments, independently packed as viral ribonucleoproteins (vRNPs), are released into the cytosol before nuclear import for viral replication. After nucleocytoplasmic transport, the resulting progeny vRNPs reach the cytosol, accumulating in highly mobile and dynamic viral inclusions that display liquid properties. Being sites postulated to support IAV genome assembly, the biophysical properties of IAV inclusions may be critical for function. In agreement, imposing liquid-to-solid transitions was demonstrated to impact viral replication negatively. Therefore, screening for host factors or compounds able to alter the material properties may provide the molecular basis for how influenza genomic complex forms as well as identify novel antivirals. Conventional techniques employed to investigate biomolecular condensates' material properties include fluorescence correlation spectroscopy, raster image correlation spectroscopy, single molecule or microrheology particle tracking, and Fluorescence Recovery After Photobleaching (FRAP). These approaches allow measuring molecular dynamics in systems that do not move very much. However, the analysis of highly mobile intracellular condensates, such as IAV inclusions, poses significant challenges as these structures not only constantly move within the cell but also exchange material, fusing, and dividing, rendering the quantitation of internal rearrangements and diffusion coefficients of molecules within condensates inaccurate. As an alternative, we opted for measuring the kinetics and the exchange of material between IAV inclusions using the Fluorescence Loss After Photoactivation (FLAPh) technique. It involves pulse photoactivation of individual or pools of viral inclusions in the cell, and chasing over time in photoactivated and non-photoactivated regions. This approach is suitable for quantifying the movement and spatial distribution of components within inclusions over time, enabling the determination of both the distance and speed from a specific cellular location. As a result, this method allows the quantification of decay profiles, half-lives, decay constant rate, and mobile and immobile fractions in viral inclusions. It, therefore, enables high throughput screenings for compounds or host factors that affect this dynamism and indirectly allows assessing the material properties of IAV inclusions.
Collapse
Affiliation(s)
- Temitope Akhigbe Etibor
- Cell Biology of Viral Infection Lab (CBV), Instituto Gulbenkian de Ciência (IGC) - Fundação Calouste Gulbenkian, Oeiras, Portugal
- Molecular Medicine Lab, Institute of Pharmaceutical Chemistry (IPW), ETH Zurich, Zurich, Switzerland
| | - Tiago Paixão
- Cell Biology of Viral Infection Lab (CBV), Instituto Gulbenkian de Ciência (IGC) - Fundação Calouste Gulbenkian, Oeiras, Portugal
| | - Maria João Amorim
- Cell Biology of Viral Infection Lab (CBV), Instituto Gulbenkian de Ciência (IGC) - Fundação Calouste Gulbenkian, Oeiras, Portugal.
- Cell Biology of Viral Infection Lab (CBV), Católica Biomedical Research Centre (CBR), Católica Medical School - Universidade Católica Portuguesa, Lisbon, Portugal.
| |
Collapse
|
30
|
Ay S, Burlaud-Gaillard J, Gazi A, Tatirovsky Y, Cuche C, Diana JS, Scoca V, Di Santo JP, Roingeard P, Mammano F, Di Nunzio F. In vivo HIV-1 nuclear condensates safeguard against cGAS and license reverse transcription. EMBO J 2025; 44:166-199. [PMID: 39623137 PMCID: PMC11697293 DOI: 10.1038/s44318-024-00316-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 11/05/2024] [Accepted: 11/07/2024] [Indexed: 01/04/2025] Open
Abstract
Entry of viral capsids into the nucleus induces the formation of biomolecular condensates called HIV-1 membraneless organelles (HIV-1-MLOs). Several questions remain about their persistence, in vivo formation, composition, and function. Our study reveals that HIV-1-MLOs persisted for several weeks in infected cells, and their abundance correlated with viral infectivity. Using an appropriate animal model, we show that HIV-1-MLOs were formed in vivo during acute infection. To explore the viral structures present within these biomolecular condensates, we used a combination of double immunogold labeling, electron microscopy and tomography, and unveiled a diverse array of viral core structures. Our functional analyses showed that HIV-1-MLOs remained stable during treatment with a reverse transcriptase inhibitor, maintaining the virus in a dormant state. Drug withdrawal restored reverse transcription, promoting efficient virus replication akin to that observed in latently infected patients on antiretroviral therapy. However, when HIV-1 MLOs were deliberately disassembled by pharmacological treatment, we observed a complete loss of viral infectivity. Our findings show that HIV-1 MLOs shield the final reverse transcription product from host immune detection.
Collapse
Affiliation(s)
- Selen Ay
- Institut Pasteur, Advanced Molecular Virology Unit, Department of Virology, Université Paris Cité, 75015, Paris, France
| | - Julien Burlaud-Gaillard
- Inserm U1259 MAVIVHe, Université de Tours and CHRU de Tours, Tours, France
- Plate-Forme IBiSA de Microscopie Electronique, Université de Tours and CHRU de Tours, Tours, France
| | - Anastasia Gazi
- Institut Pasteur, Université Paris Cité, Ultrastructural BioImaging Facility, 75015, Paris, France
| | - Yevgeniy Tatirovsky
- Innate Immunity Unit, Institut Pasteur, Université Paris Cité, Inserm U1223, Paris, France
- Vaccine Research Institute, Université Paris Est, Inserm U955, Créteil, France
| | - Celine Cuche
- Institut Pasteur, Advanced Molecular Virology Unit, Department of Virology, Université Paris Cité, 75015, Paris, France
| | - Jean-Sebastien Diana
- Institut Pasteur, Advanced Molecular Virology Unit, Department of Virology, Université Paris Cité, 75015, Paris, France
| | - Viviana Scoca
- Institut Pasteur, Advanced Molecular Virology Unit, Department of Virology, Université Paris Cité, 75015, Paris, France
| | - James P Di Santo
- Innate Immunity Unit, Institut Pasteur, Université Paris Cité, Inserm U1223, Paris, France
| | - Philippe Roingeard
- Inserm U1259 MAVIVHe, Université de Tours and CHRU de Tours, Tours, France
- Plate-Forme IBiSA de Microscopie Electronique, Université de Tours and CHRU de Tours, Tours, France
| | - Fabrizio Mammano
- Inserm U1259 MAVIVHe, Université de Tours and CHRU de Tours, Tours, France
| | - Francesca Di Nunzio
- Institut Pasteur, Advanced Molecular Virology Unit, Department of Virology, Université Paris Cité, 75015, Paris, France.
| |
Collapse
|
31
|
McCaig CD. SARS-CoV-2 Is an Electricity-Driven Virus. Rev Physiol Biochem Pharmacol 2025; 187:361-410. [PMID: 39838019 DOI: 10.1007/978-3-031-68827-0_18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2025]
Abstract
One of the most important and challenging biological events of recent times has been the pandemic caused by SARS-CoV-2. Since the underpinning argument behind this book is the ubiquity of electrical forces driving multiple disparate biological events, consideration of key aspects of the SARS-CoV-2 structural proteins is included. Electrical regulation of spike protein, nucleocapsid protein, membrane protein, and envelope protein is included, with several of their activities regulated by LLPS and the multivalent and π-cation and π-π electrical forces that drive phase separation.
Collapse
Affiliation(s)
- Colin D McCaig
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, Scotland, UK
| |
Collapse
|
32
|
Wu T, Yu JC, Suresh A, Gale-Day ZJ, Alteen MG, Woo AS, Millbern Z, Johnson OT, Carroll EC, Partch CL, Fourches D, Vinueza NR, Vocadlo DJ, Gestwicki JE. Protein-adaptive differential scanning fluorimetry using conformationally responsive dyes. Nat Biotechnol 2025; 43:106-113. [PMID: 38744946 DOI: 10.1038/s41587-024-02158-7] [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: 02/14/2023] [Accepted: 01/31/2024] [Indexed: 05/16/2024]
Abstract
Differential scanning fluorimetry (DSF) is a technique that reports protein thermal stability via the selective recognition of unfolded states by fluorogenic dyes. However, DSF applications remain limited by protein incompatibilities with existing DSF dyes. Here we overcome this obstacle with the development of a protein-adaptive DSF platform (paDSF) that combines a dye library 'Aurora' with a streamlined procedure to identify protein-dye pairs on demand. paDSF was successfully applied to 94% (66 of 70) of proteins, tripling the previous compatibility and delivering assays for 66 functionally and biochemically diverse proteins, including 10 from severe acute respiratory syndrome coronavirus 2. We find that paDSF can be used to monitor biological processes that were previously inaccessible, demonstrated for the interdomain allostery of O-GlcNAc transferase. The chemical diversity and varied selectivities of Aurora dyes suggest that paDSF functionality may be readily extended. paDSF is a generalizable tool to interrogate protein stability, dynamics and ligand binding.
Collapse
Affiliation(s)
- Taiasean Wu
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA, USA
- Institute for Neurodegenerative Diseases, University of California, San Francisco, CA, USA
| | - Joshua C Yu
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA, USA
| | - Arundhati Suresh
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA, USA
| | - Zachary J Gale-Day
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA, USA
| | - Matthew G Alteen
- Department of Chemistry, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Amanda S Woo
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA, USA
| | - Zoe Millbern
- Department of Textile Engineering, North Carolina State University, Raleigh, NC, USA
| | - Oleta T Johnson
- Institute for Neurodegenerative Diseases, University of California, San Francisco, CA, USA
| | - Emma C Carroll
- Institute for Neurodegenerative Diseases, University of California, San Francisco, CA, USA
| | - Carrie L Partch
- Department of Chemistry, University of California, Santa Cruz, CA, USA
| | - Denis Fourches
- Department of Textile Engineering, North Carolina State University, Raleigh, NC, USA
| | - Nelson R Vinueza
- Department of Textile Engineering, North Carolina State University, Raleigh, NC, USA
| | - David J Vocadlo
- Department of Chemistry, Simon Fraser University, Burnaby, British Columbia, Canada
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Jason E Gestwicki
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA, USA.
- Institute for Neurodegenerative Diseases, University of California, San Francisco, CA, USA.
| |
Collapse
|
33
|
Wang K, Hu G, Wu Z, Kurgan L. Accurate and Fast Prediction of Intrinsic Disorder Using flDPnn. Methods Mol Biol 2025; 2867:201-218. [PMID: 39576583 DOI: 10.1007/978-1-0716-4196-5_12] [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: 11/24/2024]
Abstract
Intrinsically disordered proteins (IDPs) that include one or more intrinsically disordered regions (IDRs) are abundant across all domains of life and viruses and play numerous functional roles in various cellular processes. Due to a relatively low throughput and high cost of experimental techniques for identifying IDRs, there is a growing need for fast and accurate computational algorithms that accurately predict IDRs/IDPs from protein sequences. We describe one of the leading disorder predictors, flDPnn. Results from a recent community-organized Critical Assessment of Intrinsic Disorder (CAID) experiment show that flDPnn provides fast and state-of-the-art predictions of disorder, which are supplemented with the predictions of several major disorder functions. This chapter provides a practical guide to flDPnn, which includes a brief explanation of its predictive model, descriptions of its web server and standalone versions, and a case study that showcases how to read and understand flDPnn's predictions.
Collapse
Affiliation(s)
- Kui Wang
- NITFID, School of Statistics and Data Science, LPMC and KLMDASR, Nankai University, Tianjin, China
| | - Gang Hu
- NITFID, School of Statistics and Data Science, LPMC and KLMDASR, Nankai University, Tianjin, China
| | - Zhonghua Wu
- School of Mathematical Sciences and LPMC, Nankai University, Tianjin, China
| | - Lukasz Kurgan
- Department of Computer Science, Virginia Commonwealth University, Richmond, VA, USA.
| |
Collapse
|
34
|
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
|
35
|
Samad A, Hamza A, Imam MA, Ahmad Chaudhary A, Alawam AS, Abdullah Almuqri E, Islam A, Parveen S. pH induced structural and conformational changes in nucleocapsid protein leads to intermediate like conformation: a biophysical and computational approach. J Biomol Struct Dyn 2024:1-12. [PMID: 39718618 DOI: 10.1080/07391102.2024.2442791] [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: 02/20/2024] [Accepted: 04/29/2024] [Indexed: 12/25/2024]
Abstract
Nucleocapsid protein (N) of SARS-CoV-2 is a multivalent protein, which is responsible for viral replication, assembly, packaging and modulates host immune response. In this study, we report conformational measurements of N protein at different pH by observing transition in secondary and tertiary structural contents by biophysical and computational approaches. Spectroscopic measurements revealed that N protein loses its secondary and tertiary structure at extreme acidic pH while maintaining its native conformation at mild acidic and alkaline pH. Molecular dynamics simulation studies validated spectroscopic findings. Secondary structure estimation confirmed circular dichroism (CD) findings that participation of total number of average residues in formation of native structure is higher at physiological pH, and coil percentage is higher at acidic pH. In molten globule (MG) state, secondary structure is conserved but here, CD data reveal more random structure at low pH. In pre-MG, ANS (8-anilino-1-napthalene sulfonate) binds weakly to protein as compared to MG but here, ANS binds strongly to protein. All the above-mentioned findings suggested formation of intermediary like state at low pH, which can be attributed to an off-pathway species. Unravelling structural characteristics of N protein will help understand phase-separation, protein-protein interaction and host-immune response modulation behaviour, which will eventually help in designing novel therapeutic target against COVID-19.
Collapse
Affiliation(s)
- Abdus Samad
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| | - Abu Hamza
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, VA, USA
| | - Md Ali Imam
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| | - Anis Ahmad Chaudhary
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, Saudi Arabia
| | - Abdullah S Alawam
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, Saudi Arabia
| | - Eman Abdullah Almuqri
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, Saudi Arabia
| | - Asimul Islam
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| | - Shama Parveen
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| |
Collapse
|
36
|
Tikhomirova MA, Kuzmenko OL, Arifulin EA, Shirokova OM, Musinova YR, Sheval EV. The Nucleocapsid (N) Proteins of Different Human Coronaviruses Demonstrate a Variable Capacity to Induce the Formation of Cytoplasmic Condensates. Int J Mol Sci 2024; 25:13162. [PMID: 39684875 DOI: 10.3390/ijms252313162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 12/02/2024] [Accepted: 12/04/2024] [Indexed: 12/18/2024] Open
Abstract
To date, seven human coronaviruses (HCoVs) have been identified. Four of these viruses typically manifest as a mild respiratory disease, whereas the remaining three can cause severe conditions that often result in death. The reasons for these differences remain poorly understood, but they may be related to the properties of individual viral proteins. The nucleocapsid (N) protein plays a crucial role in the packaging of viral genomic RNA and the modification of host cells during infection, in part due to its capacity to form dynamic biological condensates via liquid-liquid phase separation (LLPS). In this study, we investigated the capacity of N proteins derived from all HCoVs to form condensates when transiently expressed in cultured human cells. Some of the transfected cells were observed to contain cytoplasmic granules in which most of the N proteins were accumulated. Notably, the N proteins of SARS-CoV and SARS-CoV-2 showed a significantly reduced tendency to form cytoplasmic condensates. The condensate formation was not a consequence of overexpression of N proteins, as is typical for LLPS-inducing proteins. These condensates contained components of stress granules (SGs), indicating that the expression of N proteins caused the formation of SGs, which integrate N proteins. Thus, the N proteins of two closely related viruses, SARS-CoV and SARS-CoV-2, have the capacity to antagonize SG induction and/or assembly, in contrast to all other known HCoVs.
Collapse
Affiliation(s)
- Maria A Tikhomirova
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, 119334 Moscow, Russia
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119991 Moscow, Russia
| | - Oleg L Kuzmenko
- School of Bioengineering and Bioinformatics, Lomonosov Moscow State University, 119234 Moscow, Russia
| | - Eugene A Arifulin
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119991 Moscow, Russia
| | - Olesya M Shirokova
- Central Scientific Research Laboratory, Institute of Fundamental Medicine, Privolzhsky Research Medical University, 603950 Nizhny Novgorod, Russia
| | - Yana R Musinova
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, 119334 Moscow, Russia
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119991 Moscow, Russia
| | - Eugene V Sheval
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119991 Moscow, Russia
- Department of Cell Biology and Histology, School of Biology, Lomonosov Moscow State University, 119991 Moscow, Russia
| |
Collapse
|
37
|
Minigulov N, Boranbayev K, Bekbossynova A, Gadilgereyeva B, Filchakova O. Structural proteins of human coronaviruses: what makes them different? Front Cell Infect Microbiol 2024; 14:1458383. [PMID: 39711780 PMCID: PMC11659265 DOI: 10.3389/fcimb.2024.1458383] [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: 07/04/2024] [Accepted: 10/17/2024] [Indexed: 12/24/2024] Open
Abstract
Following COVID-19 outbreak with its unprecedented effect on the entire world, the interest to the coronaviruses increased. The causative agent of the COVID-19, severe acute respiratory syndrome coronavirus - 2 (SARS-CoV-2) is one of seven coronaviruses that is pathogenic to humans. Others include SARS-CoV, MERS-CoV, HCoV-HKU1, HCoV-OC43, HCoV-NL63 and HCoV-229E. The viruses differ in their pathogenicity. SARS-CoV, MERS-CoV, and SARS-CoV-2 are capable to spread rapidly and cause epidemic, while HCoV-HKU1, HCoV-OC43, HCoV-NL63 and HCoV-229E cause mild respiratory disease. The difference in the viral behavior is due to structural and functional differences. All seven human coronaviruses possess four structural proteins: spike, envelope, membrane, and nucleocapsid. Spike protein with its receptor binding domain is crucial for the entry to the host cell, where different receptors on the host cell are recruited by different viruses. Envelope protein plays important role in viral assembly, and following cellular entry, contributes to immune response. Membrane protein is an abundant viral protein, contributing to the assembly and pathogenicity of the virus. Nucleocapsid protein encompasses the viral RNA into ribonucleocapsid, playing important role in viral replication. The present review provides detailed summary of structural and functional characteristics of structural proteins from seven human coronaviruses, and could serve as a practical reference when pathogenic human coronaviruses are compared, and novel treatments are proposed.
Collapse
Affiliation(s)
| | | | | | | | - Olena Filchakova
- Biology Department, School of Sciences and Humanities, Nazarbayev
University, Astana, Kazakhstan
| |
Collapse
|
38
|
Mitra R, Usher ET, Dedeoğlu S, Crotteau MJ, Fraser OA, Yennawar NH, Gadkari VV, Ruotolo BT, Holehouse AS, Salmon L, Showalter SA, Bardwell JCA. Molecular insights into the interaction between a disordered protein and a folded RNA. Proc Natl Acad Sci U S A 2024; 121:e2409139121. [PMID: 39589885 PMCID: PMC11626198 DOI: 10.1073/pnas.2409139121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 10/16/2024] [Indexed: 11/28/2024] Open
Abstract
Intrinsically disordered protein regions (IDRs) are well established as contributors to intermolecular interactions and the formation of biomolecular condensates. In particular, RNA-binding proteins (RBPs) often harbor IDRs in addition to folded RNA-binding domains that contribute to RBP function. To understand the dynamic interactions of an IDR-RNA complex, we characterized the RNA-binding features of a small (68 residues), positively charged IDR-containing protein, Small ERDK-Rich Factor (SERF). At high concentrations, SERF and RNA undergo charge-driven associative phase separation to form a protein- and RNA-rich dense phase. A key advantage of this model system is that this threshold for demixing is sufficiently high that we could use solution-state biophysical methods to interrogate the stoichiometric complexes of SERF with RNA in the one-phase regime. Herein, we describe our comprehensive characterization of SERF alone and in complex with a small fragment of the HIV-1 Trans-Activation Response (TAR) RNA with complementary biophysical methods and molecular simulations. We find that this binding event is not accompanied by the acquisition of structure by either molecule; however, we see evidence for a modest global compaction of the SERF ensemble when bound to RNA. This behavior likely reflects attenuated charge repulsion within SERF via binding to the polyanionic RNA and provides a rationale for the higher-order assembly of SERF in the context of RNA. We envision that the SERF-RNA system will lower the barrier to accessing the details that support IDR-RNA interactions and likewise deepen our understanding of the role of IDR-RNA contacts in complex formation and liquid-liquid phase separation.
Collapse
Affiliation(s)
- Rishav Mitra
- HHMI, University of Michigan, Ann Arbor, MI48109
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI48109
| | - Emery T. Usher
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO63110
- Center for Biomolecular Condensates, Washington University in St. Louis, St. Louis, MO63110
| | - Selin Dedeoğlu
- Centre de Résonance Magnétique Nucléaire à Très Hauts Champs, UMR 5082, CNRS, Ecole Normale Supérieure de Lyon, Université Claude Bernard Lyon 1, Université de Lyon, Villeurbanne69100, France
| | - Matthew J. Crotteau
- HHMI, University of Michigan, Ann Arbor, MI48109
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI48109
| | - Olivia A. Fraser
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, PA16802
| | - Neela H. Yennawar
- The Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA16802
| | - Varun V. Gadkari
- Department of Chemistry, University of Michigan, Ann Arbor, MI48109
| | | | - Alex S. Holehouse
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO63110
- Center for Biomolecular Condensates, Washington University in St. Louis, St. Louis, MO63110
| | - Loïc Salmon
- Centre de Résonance Magnétique Nucléaire à Très Hauts Champs, UMR 5082, CNRS, Ecole Normale Supérieure de Lyon, Université Claude Bernard Lyon 1, Université de Lyon, Villeurbanne69100, France
| | - Scott A. Showalter
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, PA16802
- Department of Chemistry, The Pennsylvania State University, University Park, PA16802
| | - James C. A. Bardwell
- HHMI, University of Michigan, Ann Arbor, MI48109
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI48109
| |
Collapse
|
39
|
Wang M, Valadez-Ingersoll M, Gilmore TD. Control of nuclear localization of the nucleocapsid protein of SARS-CoV-2. Virology 2024; 600:110232. [PMID: 39265446 DOI: 10.1016/j.virol.2024.110232] [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: 06/24/2024] [Revised: 08/29/2024] [Accepted: 09/04/2024] [Indexed: 09/14/2024]
Abstract
The nucleocapsid (N) protein of coronaviruses is a structural protein that binds viral RNA for assembly into the mature virion, a process that occurs in the cytoplasm. Several coronavirus N proteins also localize to the nucleus. Herein, we identify that two sequences (NLSs) are required for nuclear localization of the SARS-CoV-2 N protein. Deletion or mutation of these two sequences creates an N protein that does not localize to the nucleus in HEK293T cells. Overexpression of both wild-type and NLS-mutated N proteins dysregulate a largely overlapping set of mRNAs in HEK293T cells, suggesting that these N proteins do not have direct nuclear effects on transcription. Consistent with that hypothesis, both N proteins induce nuclear localization of NF-κB p65 and dysregulate a set of previously identified NF-κB-dependent genes. The effects of N on nuclear properties are proposed to alter host cell functions that contribute to viral pathogenesis or replication.
Collapse
Affiliation(s)
- Mengrui Wang
- Department of Biology, Boston University, Boston, MA, 02215, USA
| | | | - Thomas D Gilmore
- Department of Biology, Boston University, Boston, MA, 02215, USA.
| |
Collapse
|
40
|
Esler M, Belica C, Rollie J, Brown W, Moghadasi SA, Shi K, Harki D, Harris R, Aihara H. A compact stem-loop DNA aptamer targets a uracil-binding pocket in the SARS-CoV-2 nucleocapsid RNA-binding domain. Nucleic Acids Res 2024; 52:13138-13151. [PMID: 39380503 PMCID: PMC11602162 DOI: 10.1093/nar/gkae874] [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: 01/27/2024] [Revised: 08/29/2024] [Accepted: 09/24/2024] [Indexed: 10/10/2024] Open
Abstract
SARS-CoV-2 nucleocapsid (N) protein is a structural component of the virus with essential roles in the replication and packaging of the viral RNA genome. The N protein is also an important target of COVID-19 antigen tests and a promising vaccine candidate along with the spike protein. Here, we report a compact stem-loop DNA aptamer that binds tightly to the N-terminal RNA-binding domain of SARS-CoV-2 N protein. Crystallographic analysis shows that a hexanucleotide DNA motif (5'-TCGGAT-3') of the aptamer fits into a positively charged concave surface of N-NTD and engages essential RNA-binding residues including Tyr109, which mediates a sequence-specific interaction in a uracil-binding pocket. Avid binding of the DNA aptamer allows isolation and sensitive detection of full-length N protein from crude cell lysates, demonstrating its selectivity and utility in biochemical applications. We further designed a chemically modified DNA aptamer and used it as a probe to examine the interaction of N-NTD with various RNA motifs, which revealed a strong preference for uridine-rich sequences. Our studies provide a high-affinity chemical probe for the SARS-CoV-2 N protein RNA-binding domain, which may be useful for diagnostic applications and investigating novel antiviral agents.
Collapse
Affiliation(s)
- Morgan A Esler
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN 55455, USA
- Institute for Molecular Virology, University of Minnesota, Minneapolis, MN 55455, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
| | - Christopher A Belica
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN 55455, USA
- Institute for Molecular Virology, University of Minnesota, Minneapolis, MN 55455, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
| | - Joseph A Rollie
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN 55455, USA
- Institute for Molecular Virology, University of Minnesota, Minneapolis, MN 55455, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
| | - William L Brown
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN 55455, USA
- Institute for Molecular Virology, University of Minnesota, Minneapolis, MN 55455, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
| | - Seyed Arad Moghadasi
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN 55455, USA
- Institute for Molecular Virology, University of Minnesota, Minneapolis, MN 55455, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
| | - Ke Shi
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN 55455, USA
- Institute for Molecular Virology, University of Minnesota, Minneapolis, MN 55455, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
| | - Daniel A Harki
- Institute for Molecular Virology, University of Minnesota, Minneapolis, MN 55455, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, MN 55455, USA
| | - Reuben S Harris
- Department of Biochemistry and Structural Biology, University of Texas Health San Antonio, San Antonio, TX 78229, USA
- Howard Hughes Medical Institute, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| | - Hideki Aihara
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN 55455, USA
- Institute for Molecular Virology, University of Minnesota, Minneapolis, MN 55455, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
| |
Collapse
|
41
|
Mallick M, Boehm V, Xue G, Blackstone M, Gehring N, Chakrabarti S. Modulation of UPF1 catalytic activity upon interaction of SARS-CoV-2 Nucleocapsid protein with factors involved in nonsense mediated-mRNA decay. Nucleic Acids Res 2024; 52:13325-13339. [PMID: 39360627 PMCID: PMC11602160 DOI: 10.1093/nar/gkae829] [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: 03/05/2024] [Revised: 08/09/2024] [Accepted: 09/14/2024] [Indexed: 10/04/2024] Open
Abstract
The RNA genome of the SARS-CoV-2 virus encodes for four structural proteins, 16 non-structural proteins and nine putative accessory factors. A high throughput analysis of interactions between human and SARS-CoV-2 proteins identified multiple interactions of the structural Nucleocapsid (N) protein with RNA processing factors. The N-protein, which is responsible for packaging of the viral genomic RNA was found to interact with two RNA helicases, UPF1 and MOV10 that are involved in nonsense-mediated mRNA decay (NMD). Using a combination of biochemical and biophysical methods, we investigated the interaction of the SARS-CoV-2 N-protein with NMD factors at a molecular level. Our studies led us to identify the core NMD factor, UPF2, as an interactor of N. The viral N-protein engages UPF2 in multipartite interactions and can negate the stimulatory effect of UPF2 on UPF1 catalytic activity. N also inhibits UPF1 ATPase and unwinding activities by competing in binding to the RNA substrate. We further investigate the functional implications of inhibition of UPF1 catalytic activity by N in mammalian cells. The interplay of SARS-CoV-2 N with human UPF1 and UPF2 does not affect decay of host cell NMD targets but might play a role in stabilizing the viral RNA genome.
Collapse
Affiliation(s)
- Megha Mallick
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr. 6, D-14195 Berlin, Germany
| | - Volker Boehm
- Institute for Genetics, University of Cologne, Cologne 50674, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne 50931, Germany
| | - Guangpu Xue
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr. 6, D-14195 Berlin, Germany
| | - Mark Blackstone
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr. 6, D-14195 Berlin, Germany
| | - Niels H Gehring
- Institute for Genetics, University of Cologne, Cologne 50674, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne 50931, Germany
| | - Sutapa Chakrabarti
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr. 6, D-14195 Berlin, Germany
| |
Collapse
|
42
|
Han Y, Zhou H, Liu C, Wang W, Qin Y, Chen M. SARS-CoV-2 N protein coordinates viral particle assembly through multiple domains. J Virol 2024; 98:e0103624. [PMID: 39412257 PMCID: PMC11575404 DOI: 10.1128/jvi.01036-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 09/09/2024] [Indexed: 11/20/2024] Open
Abstract
Increasing evidence suggests that mutations in the nucleocapsid (N) protein of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) may enhance viral replication by modulating the assembly process. However, the mechanisms governing the selective packaging of viral genomic RNA by the N protein, along with the assembly and budding processes, remain poorly understood. Utilizing a virus-like particles (VLPs) system, we have identified that the C-terminal domain (CTD) of the N protein is essential for its interaction with the membrane (M) protein during budding, crucial for binding and packaging genomic RNA. Notably, the isolated CTD lacks M protein interaction capacity and budding ability. Yet, upon fusion with the N-terminal domain (NTD) or the linker region (LKR), the resulting NTD/CTD and LKR/CTD acquire RNA-dependent interactions with the M protein and acquire budding capabilities. Furthermore, the presence of the C-tail is vital for efficient genomic RNA encapsidation by the N protein, possibly regulated by interactions with the M protein. Remarkably, the NTD of the N protein appears dispensable for virus particle assembly, offering the virus adaptive advantages. The emergence of N* (NΔN209) in the SARS-CoV-2 B.1.1 lineage corroborates our findings and hints at the potential evolution of a more streamlined N protein by the SARS-CoV-2 virus to facilitate the assembly process. Comparable observations have been noted with the N proteins of SARS-CoV and HCoV-OC43 viruses. In essence, these findings propose that β-coronaviruses may augment their replication by fine-tuning the assembly process.IMPORTANCEAs a highly transmissible zoonotic virus, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) continues to evolve. Adaptive mutations in the nucleocapsid (N) protein highlight the critical role of N protein-based assembly in the virus's replication and evolutionary dynamics. However, the precise molecular mechanisms of N protein-mediated viral assembly remain inadequately understood. Our study elucidates the intricate interactions between the N protein, membrane (M) protein, and genomic RNA, revealing a C-terminal domain (CTD)-based assembly mechanism common among β-coronaviruses. The appearance of the N* variant within the SARS-CoV-2 B.1.1 lineage supports our conclusion that the N-terminal domain (NTD) of the N protein is not essential for viral assembly. This work not only enhances our understanding of coronavirus assembly mechanisms but also provides new insights for developing antiviral drugs targeting these conserved processes.
Collapse
Affiliation(s)
- Yuewen Han
- State Key Laboratory of Virology and Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China
| | - Haiwu Zhou
- State Key Laboratory of Virology and Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China
| | - Cong Liu
- State Key Laboratory of Virology and Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China
| | - Weiwei Wang
- State Key Laboratory of Virology and Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China
| | - Yali Qin
- School of Life Sciences, Hubei University, Wuhan, China
| | - Mingzhou Chen
- State Key Laboratory of Virology and Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China
- School of Life Sciences, Hubei University, Wuhan, China
- Hubei Jiangxia Laboratory, Wuhan, China
| |
Collapse
|
43
|
Chen Y, Hour MJ, Lin CS, Chang YS, Chen ZY, Koval'skaya AV, Su WC, Tsypysheva IP, Lin CW. Assessing the inhibitory effects of some secondary amines, thioureas and 1,3-dimethyluracil conjugates of (-)-cytisine and thermopsine on the RNA-dependent RNA polymerase of SARS-CoV-1 and SARS-CoV-2. Bioorg Med Chem Lett 2024; 113:129950. [PMID: 39251111 DOI: 10.1016/j.bmcl.2024.129950] [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: 07/13/2024] [Revised: 08/27/2024] [Accepted: 09/03/2024] [Indexed: 09/11/2024]
Abstract
SARS-CoV-2 causes COVID-19, with symptoms ranging from mild to severe, including pneumonia and death. This beta coronavirus has a 30-kilobase RNA genome and shares about 80 % of its nucleotide sequence with SARS-CoV-1. The replication/transcription complex, essential for viral RNA synthesis, includes RNA-dependent RNA polymerase (RdRp, nsp12) enhanced by nsp7 and nsp8. Antivirals like molnupiravir and remdesivir, which are RdRp inhibitors, treat severe COVID-19 but have limitations, highlighting the need for new therapies. This study assessed (-)-cytisine, methylcytisine, and thermopsine derivatives against SARS-CoV-1 and SARS-CoV-2 in vitro, focusing on their RdRp inhibition. Selected compounds from a previous study were evaluated using a SARS-CoV-2 RNA polymerase assay kit to investigate their structure-activity relationships. Compound 17 (1,3-dimethyluracil conjugate with (-)-cytisine and thermopsine) emerged as a potent inhibitor of SARS-CoV-1 and SARS-CoV-2 RdRp, with an IC50 value of 7.8 μM against SARS-CoV-2 RdRp. It showed a dose-dependent reduction in cytopathic effects in cells infected with SARS-CoV-1 and SARS-CoV-2 replicon-based single-round infectious particles (SRIPs) and significantly inhibited SARS-CoV N protein expression, with EC50 values of 0.12 µM for SARS-CoV-1 and 1.47 µM for SARS-CoV-2 SRIPs. Additionally, compound 17 reduced viral subgenomic RNA levels in a concentration-dependent manner in SRIP-infected cells. The structure-activity relationships of compound 17 with SARS-CoV-1 and SARS-CoV-2 RdRp were also investigated, highlighting it as a promising lead for developing antiviral agents against SARS and COVID-19.
Collapse
Affiliation(s)
- Yeh Chen
- Department of Food Science and Biotechnology of National Chung Hsing University, Taichung, Taiwan
| | - Mann-Jen Hour
- School of Pharmacy, China Medical University, Taichung 40402, Taiwan
| | - Chen-Sheng Lin
- Division of Gastroenterology, Kuang Tien General Hospital, No. 117, Shatian Rd, Shalu District, Taichung City 433, Taiwan
| | - Young-Sheng Chang
- Department of Medical Laboratory Science and Biotechnology, China Medical University, Taichung 404394, Taiwan; Graduate Institute of Biomedical Sciences, China Medical University, Taichung 404394, Taiwan
| | - Zan-Yu Chen
- Department of Medical Laboratory Science and Biotechnology, China Medical University, Taichung 404394, Taiwan
| | - Alena V Koval'skaya
- Ufa Institute of Chemistry, Ufa Federal Research Centre of the Russian Academy of Sciences, 71 prosp. Oktyabrya, 450054 Ufa, Russian Federation
| | - Wen-Chi Su
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 404394, Taiwan; International Master's Program of Biomedical Sciences, China Medical University, Taichung 404394, Taiwan
| | - Inna P Tsypysheva
- Ufa Institute of Chemistry, Ufa Federal Research Centre of the Russian Academy of Sciences, 71 prosp. Oktyabrya, 450054 Ufa, Russian Federation.
| | - Cheng-Wen Lin
- Department of Medical Laboratory Science and Biotechnology, China Medical University, Taichung 404394, Taiwan; Graduate Institute of Biomedical Sciences, China Medical University, Taichung 404394, Taiwan; Department of Medical Laboratory Science and Biotechnology, Asia University, Wufeng, Taichung 413305, Taiwan.
| |
Collapse
|
44
|
Xu Y, Shen Y. The Assembly of Miniaturized Droplets toward Functional Architectures. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2404366. [PMID: 39380419 DOI: 10.1002/smll.202404366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 07/19/2024] [Indexed: 10/10/2024]
Abstract
Recent explorations of bioengineering have generated new concepts and strategies for the processing of soft and functional materials. Droplet assembly techniques can address problems in the construction of extremely soft architectures by expanding the manufacturing capabilities using droplets containing liquid or hydrogels including weak hydrogels. This Perspective sets out to provide a brief overview of this growing field, and discusses the challenges and opportunities ahead. The study highlights the recent key advances of materials and architectures from hitherto effective droplet-assembly technologies, as well as the applications in biomedical and bioengineering fields from artificial tissues to bioreactors. It is envisaged that these assembled architectures, as nature-inspired models, will stimulate the discovery of biomaterials and miniaturized platforms for interdisciplinary research in health, biotechnology, and sustainability.
Collapse
Affiliation(s)
- Yufan Xu
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW, UK
| | - Yi Shen
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, CB2 1EW, UK
- School of Chemical and Biomolecular Engineering, The University of Sydney, Sydney, NSW, 2006, Australia
- The University of Sydney Nano Institute, The University of Sydney, Sydney, NSW, 2006, Australia
| |
Collapse
|
45
|
Yang Y, Tan J, Wang F, Sun W, Shi H, Cheng Z, Xie Y, Zhou X. Preconcentration and detection of SARS-CoV-2 in wastewater: A comprehensive review. Biosens Bioelectron 2024; 263:116617. [PMID: 39094290 DOI: 10.1016/j.bios.2024.116617] [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: 05/22/2024] [Revised: 07/17/2024] [Accepted: 07/29/2024] [Indexed: 08/04/2024]
Abstract
Severe acute respiratory syndrome coronaviruses 2 (SARS-CoV-2) causing coronavirus disease 2019 (COVID-19) affected the health of human beings and the global economy. The patients with SARS-CoV-2 infection had viral RNA or live infectious viruses in feces. Thus, the possible transmission of SARS-CoV-2 through wastewater received great attentions. Moreover, SARS-CoV-2 in wastewater can serve as an early indicator of the infection within communities. We summarized the preconcentration and detection technology of SARS-CoV-2 in wastewater aiming at the complex matrices of wastewater and low virus concentration and compared their performance characteristics. We described the emerging tests that would be possible to realize the rapid detection of SARS-CoV-2 in fields and encourage academics to advance their technologies beyond conception. We concluded with a brief discussion on the outlook for integrating preconcentration and the detection of SARS-CoV-2 with emerging technologies.
Collapse
Affiliation(s)
- Yihan Yang
- School of Environment, Tsinghua University, Beijing, 100084, China
| | - Jisui Tan
- School of Environment, Tsinghua University, Beijing, 100084, China
| | - Fan Wang
- School of Environment, Tsinghua University, Beijing, 100084, China
| | - Weiming Sun
- School of Environment, Tsinghua University, Beijing, 100084, China
| | - Hanchang Shi
- School of Environment, Tsinghua University, Beijing, 100084, China
| | - Zhao Cheng
- School of Environment, Tsinghua University, Beijing, 100084, China
| | - Yangcun Xie
- Chinese Academy of Environmental Planning, Beijing, 100043, China.
| | - Xiaohong Zhou
- School of Environment, Tsinghua University, Beijing, 100084, China.
| |
Collapse
|
46
|
Syed AM, Ciling A, Chen IP, Carlson CR, Adly AN, Martin HS, Taha TY, Khalid MM, Price N, Bouhaddou M, Ummadi MR, Moen JM, Krogan NJ, Morgan DO, Ott M, Doudna JA. SARS-CoV-2 evolution balances conflicting roles of N protein phosphorylation. PLoS Pathog 2024; 20:e1012741. [PMID: 39571001 PMCID: PMC11620656 DOI: 10.1371/journal.ppat.1012741] [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: 05/15/2024] [Revised: 12/05/2024] [Accepted: 11/11/2024] [Indexed: 12/07/2024] Open
Abstract
All lineages of SARS-CoV-2, the coronavirus responsible for the COVID-19 pandemic, contain mutations between amino acids 199 and 205 in the nucleocapsid (N) protein that are associated with increased infectivity. The effects of these mutations have been difficult to determine because N protein contributes to both viral replication and viral particle assembly during infection. Here, we used single-cycle infection and virus-like particle assays to show that N protein phosphorylation has opposing effects on viral assembly and genome replication. Ancestral SARS-CoV-2 N protein is densely phosphorylated, leading to higher levels of genome replication but 10-fold lower particle assembly compared to evolved variants with low N protein phosphorylation, such as Delta (N:R203M), Iota (N:S202R), and B.1.2 (N:P199L). A new open reading frame encoding a truncated N protein called N*, which occurs in the B.1.1 lineage and subsequent lineages of the Alpha, Gamma, and Omicron variants, supports high levels of both assembly and replication. Our findings help explain the enhanced fitness of viral variants of concern and a potential avenue for continued viral selection.
Collapse
Affiliation(s)
- Abdullah M. Syed
- Gladstone Institutes, San Francisco, California, United States of America
| | - Alison Ciling
- Gladstone Institutes, San Francisco, California, United States of America
| | - Irene P. Chen
- Gladstone Institutes, San Francisco, California, United States of America
- Department of Medicine, University of California, San Francisco, California, United States of America
| | - Christopher R. Carlson
- Department of Physiology, University of California, San Francisco, California, United States of America
| | - Armin N. Adly
- Department of Physiology, University of California, San Francisco, California, United States of America
| | - Hannah S. Martin
- Department of Chemistry, University of California, Berkeley, California, United States of America
| | - Taha Y. Taha
- Gladstone Institutes, San Francisco, California, United States of America
| | - Mir M. Khalid
- Gladstone Institutes, San Francisco, California, United States of America
| | - Nathan Price
- Gladstone Institutes, San Francisco, California, United States of America
| | - Mehdi Bouhaddou
- Gladstone Institutes, San Francisco, California, United States of America
- Department of Microbiology, Immunology, and Molecular Genetics (MIMG), UCLA, Los Angeles, California, United States of America
- Institute for Quantitative and Computational Biosciences (QCBio), UCLA, Los Angeles, California, United States of America
- Quantitative Biosciences Institute (QBI), University of California, San Francisco, California, United States of America
- Quantitative Biosciences Institute (QBI) COVID-19 Research Group (QCRG), San Francisco, California, United States of America
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, California, United States of America
| | - Manisha R. Ummadi
- Gladstone Institutes, San Francisco, California, United States of America
- Quantitative Biosciences Institute (QBI), University of California, San Francisco, California, United States of America
- Quantitative Biosciences Institute (QBI) COVID-19 Research Group (QCRG), San Francisco, California, United States of America
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, California, United States of America
| | - Jack M. Moen
- Gladstone Institutes, San Francisco, California, United States of America
- Quantitative Biosciences Institute (QBI), University of California, San Francisco, California, United States of America
- Quantitative Biosciences Institute (QBI) COVID-19 Research Group (QCRG), San Francisco, California, United States of America
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, California, United States of America
| | - Nevan J. Krogan
- Gladstone Institutes, San Francisco, California, United States of America
- Quantitative Biosciences Institute (QBI), University of California, San Francisco, California, United States of America
- Quantitative Biosciences Institute (QBI) COVID-19 Research Group (QCRG), San Francisco, California, United States of America
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, California, United States of America
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, California, United States of America
| | - David O. Morgan
- Department of Physiology, University of California, San Francisco, California, United States of America
| | - Melanie Ott
- Gladstone Institutes, San Francisco, California, United States of America
- Department of Medicine, University of California, San Francisco, California, United States of America
| | - Jennifer A. Doudna
- Gladstone Institutes, San Francisco, California, United States of America
- Department of Molecular and Cell Biology, University of California, Berkeley, California, United States of America
- Howard Hughes Medical Institute, University of California, Berkeley, California, United States of America
- Innovative Genomics Institute, University of California, Berkeley, California, United States of America
- California Institute for Quantitative Biosciences (QB3), University of California, Berkeley, California, United States of America
- MBIB Division, Lawrence Berkeley National Laboratory, Berkeley, California, United States of America
| |
Collapse
|
47
|
Gomez-Gonzalez A, Burkhardt P, Bauer M, Suomalainen M, Mateos JM, Loehr MO, Luedtke NW, Greber UF. Stepwise virus assembly in the cell nucleus revealed by spatiotemporal click chemistry of DNA replication. SCIENCE ADVANCES 2024; 10:eadq7483. [PMID: 39454009 PMCID: PMC11506174 DOI: 10.1126/sciadv.adq7483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 09/23/2024] [Indexed: 10/27/2024]
Abstract
Biomolecular assemblies are fundamental to life and viral disease. The spatiotemporal coordination of viral replication and assembly is largely unknown. Here, we developed a dual-color click chemistry procedure for imaging adenovirus DNA (vDNA) replication in the cell nucleus. Late- but not early-replicated vDNA was packaged into virions. Early-replicated vDNA segregated from the viral replication compartment (VRC). Single object tracking, superresolution microscopy, fluorescence recovery after photobleaching, and correlative light-electron microscopy revealed a stepwise assembly program involving vDNA and capsid intermediates. Depending on replication and the scaffolding protein 52K, late-replicated vDNA with rapidly exchanging green fluorescent protein-tagged capsid linchpin protein V and incomplete virions emerged from the VRC periphery. These nanogel-like puncta exhibited restricted movements and were located with the capsid proteins hexon, VI, and virions in the nuclear periphery, suggestive of sites for virion formation. Our findings identify VRC dynamics and assembly intermediates, essential for stepwise productive adenovirus morphogenesis.
Collapse
Affiliation(s)
| | - Patricia Burkhardt
- Department of Molecular Life Sciences, University of Zurich (UZH), Zurich, Switzerland
| | - Michael Bauer
- Department of Molecular Life Sciences, University of Zurich (UZH), Zurich, Switzerland
| | - Maarit Suomalainen
- Department of Molecular Life Sciences, University of Zurich (UZH), Zurich, Switzerland
| | - José María Mateos
- Center for Microscopy and Image Analyses, University of Zurich (UZH), Zurich, Switzerland
| | - Morten O. Loehr
- Department of Chemistry, McGill University, Montréal, QC, Canada
| | | | - Urs F. Greber
- Department of Molecular Life Sciences, University of Zurich (UZH), Zurich, Switzerland
| |
Collapse
|
48
|
Katiyar H, Arduini A, Li Y, Liang C. SARS-CoV-2 Assembly: Gaining Infectivity and Beyond. Viruses 2024; 16:1648. [PMID: 39599763 PMCID: PMC11598957 DOI: 10.3390/v16111648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 10/12/2024] [Accepted: 10/21/2024] [Indexed: 11/29/2024] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) was responsible for causing the COVID-19 pandemic. Intensive research has illuminated the complex biology of SARS-CoV-2 and its continuous evolution during and after the COVID-19 pandemic. While much attention has been paid to the structure and functions of the viral spike protein and the entry step of viral infection, partly because these are targets for neutralizing antibodies and COVID-19 vaccines, the later stages of SARS-CoV-2 replication, including the assembly and egress of viral progenies, remain poorly characterized. This includes insight into how the activities of the viral structural proteins are orchestrated spatially and temporally, which cellular proteins are assimilated by the virus to assist viral assembly, and how SARS-CoV-2 counters and evades the cellular mechanisms antagonizing virus assembly. In addition to becoming infectious, SARS-CoV-2 progenies also need to survive the hostile innate and adaptive immune mechanisms, such as recognition by neutralizing antibodies. This review offers an updated summary of the roles of SARS-CoV-2 structural proteins in viral assembly, the regulation of assembly by viral and cellular factors, and the cellular mechanisms that restrict this process. Knowledge of these key events often reveals the vulnerabilities of SARS-CoV-2 and aids in the development of effective antiviral therapeutics.
Collapse
Affiliation(s)
- Harshita Katiyar
- Lady Davis Institute, Jewish General Hospital, Montreal, QC H3T 1E2, Canada; (H.K.); (A.A.); (Y.L.)
- Department of Microbiology and Immunology, McGill University, Montreal, QC H3A 2B4, Canada
| | - Ariana Arduini
- Lady Davis Institute, Jewish General Hospital, Montreal, QC H3T 1E2, Canada; (H.K.); (A.A.); (Y.L.)
- Department of Medicine, McGill University, Montreal, QC H3G 2M1, Canada
| | - Yichen Li
- Lady Davis Institute, Jewish General Hospital, Montreal, QC H3T 1E2, Canada; (H.K.); (A.A.); (Y.L.)
- Department of Microbiology and Immunology, McGill University, Montreal, QC H3A 2B4, Canada
| | - Chen Liang
- Lady Davis Institute, Jewish General Hospital, Montreal, QC H3T 1E2, Canada; (H.K.); (A.A.); (Y.L.)
- Department of Microbiology and Immunology, McGill University, Montreal, QC H3A 2B4, Canada
- Department of Medicine, McGill University, Montreal, QC H3G 2M1, Canada
| |
Collapse
|
49
|
Jang S, Bedwell G, Singh S, Yu H, Arnarson B, Singh P, Radhakrishnan R, Douglas A, Ingram Z, Freniere C, Akkermans O, Sarafianos S, Ambrose Z, Xiong Y, Anekal P, Montero Llopis P, KewalRamani V, Francis A, Engelman A. HIV-1 usurps mixed-charge domain-dependent CPSF6 phase separation for higher-order capsid binding, nuclear entry and viral DNA integration. Nucleic Acids Res 2024; 52:11060-11082. [PMID: 39258548 PMCID: PMC11472059 DOI: 10.1093/nar/gkae769] [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: 05/07/2024] [Revised: 08/13/2024] [Accepted: 08/25/2024] [Indexed: 09/12/2024] Open
Abstract
HIV-1 integration favors nuclear speckle (NS)-proximal chromatin and viral infection induces the formation of capsid-dependent CPSF6 condensates that colocalize with nuclear speckles (NSs). Although CPSF6 displays liquid-liquid phase separation (LLPS) activity in vitro, the contributions of its different intrinsically disordered regions, which includes a central prion-like domain (PrLD) with capsid binding FG motif and C-terminal mixed-charge domain (MCD), to LLPS activity and to HIV-1 infection remain unclear. Herein, we determined that the PrLD and MCD both contribute to CPSF6 LLPS activity in vitro. Akin to FG mutant CPSF6, infection of cells expressing MCD-deleted CPSF6 uncharacteristically arrested at the nuclear rim. While heterologous MCDs effectively substituted for CPSF6 MCD function during HIV-1 infection, Arg-Ser domains from related SR proteins were largely ineffective. While MCD-deleted and wildtype CPSF6 proteins displayed similar capsid binding affinities, the MCD imparted LLPS-dependent higher-order binding and co-aggregation with capsids in vitro and in cellulo. NS depletion reduced CPSF6 puncta formation without significantly affecting integration into NS-proximal chromatin, and appending the MCD onto a heterologous capsid binding protein partially restored virus nuclear penetration and integration targeting in CPSF6 knockout cells. We conclude that MCD-dependent CPSF6 condensation with capsids underlies post-nuclear incursion for viral DNA integration and HIV-1 pathogenesis.
Collapse
Affiliation(s)
- Sooin Jang
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Gregory J Bedwell
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Satya P Singh
- Institute of Molecular Biophysics, Department of Biological Sciences, Florida State University, Tallahassee, FL 32304, USA
| | - Hyun Jae Yu
- Model Development Section, Cancer Innovation Laboratory, National Cancer Institute, Frederick, MD 21702, USA
| | - Bjarki Arnarson
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Parmit K Singh
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Rajalingam Radhakrishnan
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - AidanDarian W Douglas
- Institute of Molecular Biophysics, Department of Biological Sciences, Florida State University, Tallahassee, FL 32304, USA
| | - Zachary M Ingram
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA
| | - Christian Freniere
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06511, USA
| | - Onno Akkermans
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Stefan G Sarafianos
- Center for ViroScience and Cure, Laboratory of Biochemical Pharmacology, Department of Pediatrics, School of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Zandrea Ambrose
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA
| | - Yong Xiong
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06511, USA
| | - Praju V Anekal
- MicRoN Core, Harvard Medical School, Boston, MA 02215, USA
| | | | - Vineet N KewalRamani
- Model Development Section, Cancer Innovation Laboratory, National Cancer Institute, Frederick, MD 21702, USA
| | - Ashwanth C Francis
- Institute of Molecular Biophysics, Department of Biological Sciences, Florida State University, Tallahassee, FL 32304, USA
| | - Alan N Engelman
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
50
|
Swain BC, Sarkis P, Ung V, Rousseau S, Fernandez L, Meltonyan A, Aho VE, Mercadante D, Mackereth CD, Aznauryan M. Disordered regions of human eIF4B orchestrate a dynamic self-association landscape. Nat Commun 2024; 15:8766. [PMID: 39384813 PMCID: PMC11464913 DOI: 10.1038/s41467-024-53136-1] [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: 06/21/2024] [Accepted: 10/03/2024] [Indexed: 10/11/2024] Open
Abstract
Eukaryotic translation initiation factor eIF4B is required for efficient cap-dependent translation, it is overexpressed in cancer cells, and may influence stress granule formation. Due to the high degree of intrinsic disorder, eIF4B is rarely observed in cryo-EM structures of translation complexes and only ever by its single structured RNA recognition motif domain, leaving the molecular details of its large intrinsically disordered region (IDR) unknown. By integrating experiments and simulations we demonstrate that eIF4B IDR orchestrates and fine-tunes an intricate transition from monomers to a condensed phase, in which large-size dynamic oligomers form before mesoscopic phase separation. Single-molecule spectroscopy combined with molecular simulations enabled us to characterize the conformational ensembles and underlying intra- and intermolecular dynamics across the oligomerization transition. The observed sensitivity to ionic strength and molecular crowding in the self-association landscape suggests potential regulation of eIF4B nanoscopic and mesoscopic behaviors such as driven by protein modifications, binding partners or changes to the cellular environment.
Collapse
Affiliation(s)
- Bikash Chandra Swain
- University of Bordeaux, Inserm, CNRS, ARNA Laboratory, U1212, UMR 5320, Institut Européen de Chimie et Biologie, F-33600, Pessac, France
| | - Pascale Sarkis
- University of Bordeaux, Inserm, CNRS, ARNA Laboratory, U1212, UMR 5320, Institut Européen de Chimie et Biologie, F-33600, Pessac, France
| | - Vanessa Ung
- School of Chemical Sciences, University of Auckland, Auckland, New Zealand
| | - Sabrina Rousseau
- University of Bordeaux, Inserm, CNRS, ARNA Laboratory, U1212, UMR 5320, Institut Européen de Chimie et Biologie, F-33600, Pessac, France
| | - Laurent Fernandez
- University of Bordeaux, Inserm, CNRS, ARNA Laboratory, U1212, UMR 5320, Institut Européen de Chimie et Biologie, F-33600, Pessac, France
| | - Ani Meltonyan
- University of Bordeaux, Inserm, CNRS, ARNA Laboratory, U1212, UMR 5320, Institut Européen de Chimie et Biologie, F-33600, Pessac, France
| | - V Esperance Aho
- University of Bordeaux, Inserm, CNRS, ARNA Laboratory, U1212, UMR 5320, Institut Européen de Chimie et Biologie, F-33600, Pessac, France
- Institut de Biologie Structurale (IBS), UMR 5075, F-38044, Grenoble, France
| | - Davide Mercadante
- School of Chemical Sciences, University of Auckland, Auckland, New Zealand
| | - Cameron D Mackereth
- University of Bordeaux, Inserm, CNRS, ARNA Laboratory, U1212, UMR 5320, F-33000, Bordeaux, France.
| | - Mikayel Aznauryan
- University of Bordeaux, Inserm, CNRS, ARNA Laboratory, U1212, UMR 5320, Institut Européen de Chimie et Biologie, F-33600, Pessac, France.
| |
Collapse
|