Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 115
Filter
1.
Elife ; 132024 Jun 28.
Article in English | MEDLINE | ID: mdl-38941233

ABSTRACT

A new study reveals how naturally occurring mutations affect the biophysical properties of nucleocapsid proteins in SARS-CoV-2.


Subject(s)
COVID-19 , Mutation , SARS-CoV-2 , SARS-CoV-2/genetics , COVID-19/virology , Humans , Coronavirus Nucleocapsid Proteins/genetics , Coronavirus Nucleocapsid Proteins/metabolism , Phosphoproteins/genetics , Phosphoproteins/metabolism
2.
Front Immunol ; 15: 1419165, 2024.
Article in English | MEDLINE | ID: mdl-38911852

ABSTRACT

Complement activation is considered to contribute to the pathogenesis of severe SARS-CoV-2 infection, mainly by generating potent immune effector mechanisms including a strong inflammatory response. Involvement of the lectin complement pathway, a major actor of the innate immune anti-viral defense, has been reported previously. It is initiated by recognition of the viral surface Spike glycoprotein by mannose-binding lectin (MBL), which induces activation of the MBL-associated protease MASP-2 and triggers the proteolytic complement cascade. A role for the viral nucleoprotein (N) has also been reported, through binding to MASP-2, leading to protease overactivation and potentiation of the lectin pathway. In the present study, we reinvestigated the interactions of the SARS-CoV-2 N protein, produced either in bacteria or secreted by mammalian cells, with full-length MASP-2 or its catalytic domain, in either active or proenzyme form. We could not confirm the interaction of the N protein with the catalytic domain of MASP-2 but observed N protein binding to proenzyme MASP-2. We did not find a role of the N protein in MBL-mediated activation of the lectin pathway. Finally, we showed that incubation of the N protein with MASP-2 results in proteolysis of the viral protein, an observation that requires further investigation to understand a potential functional significance in infected patients.


Subject(s)
COVID-19 , Complement Pathway, Mannose-Binding Lectin , Mannose-Binding Protein-Associated Serine Proteases , SARS-CoV-2 , Mannose-Binding Protein-Associated Serine Proteases/metabolism , Mannose-Binding Protein-Associated Serine Proteases/immunology , Humans , SARS-CoV-2/immunology , Complement Pathway, Mannose-Binding Lectin/immunology , COVID-19/immunology , COVID-19/virology , Protein Binding , Coronavirus Nucleocapsid Proteins/immunology , Coronavirus Nucleocapsid Proteins/metabolism , Complement Activation/immunology , Mannose-Binding Lectin/metabolism , Mannose-Binding Lectin/immunology , Phosphoproteins
3.
Elife ; 132024 Jun 28.
Article in English | MEDLINE | ID: mdl-38941236

ABSTRACT

Genetic diversity is a hallmark of RNA viruses and the basis for their evolutionary success. Taking advantage of the uniquely large genomic database of SARS-CoV-2, we examine the impact of mutations across the spectrum of viable amino acid sequences on the biophysical phenotypes of the highly expressed and multifunctional nucleocapsid protein. We find variation in the physicochemical parameters of its extended intrinsically disordered regions (IDRs) sufficient to allow local plasticity, but also observe functional constraints that similarly occur in related coronaviruses. In biophysical experiments with several N-protein species carrying mutations associated with major variants, we find that point mutations in the IDRs can have nonlocal impact and modulate thermodynamic stability, secondary structure, protein oligomeric state, particle formation, and liquid-liquid phase separation. In the Omicron variant, distant mutations in different IDRs have compensatory effects in shifting a delicate balance of interactions controlling protein assembly properties, and include the creation of a new protein-protein interaction interface in the N-terminal IDR through the defining P13L mutation. A picture emerges where genetic diversity is accompanied by significant variation in biophysical characteristics of functional N-protein species, in particular in the IDRs.


Like other types of RNA viruses, the genetic material of SARS-CoV-2 (the agent responsible for COVID-19) is formed of an RNA molecule which is prone to accumulating mutations. This gives SARS-CoV-2 the ability to evolve quickly, and often to remain one step ahead of treatments. Understanding how these mutations shape the behavior of RNA viruses is therefore crucial to keep diseases such as COVID-19 under control. The gene that codes for the protein that 'packages' the genetic information inside SARS-CoV-2 is particularly prone to mutations. This nucleocapsid (N) protein participates in many key processes during the life cycle of the virus, including potentially interfering with the immune response. Exactly how the physical properties of the N-Protein are impacted by the mutations in its genetic sequence remains unclear. To investigate this question, Nguyen et al. predicted the various biophysical properties of different regions of the N-protein based on a computer-based analysis of SARS-CoV-2 genetic databases. This allowed them to determine if specific protein regions were positively or negatively charged in different mutants. The analyses showed that some domains exhibited great variability in their charge between protein variants ­ reflecting the fact that the corresponding genetic sequences showed high levels of plasticity. Other regions remained conserved, however, including across related coronaviruses. Nguyen et al. also conducted biochemical experiments on a range of N-proteins obtained from clinically relevant SARS-CoV-2 variants. Their results highlighted the importance of protein segments with no fixed three-dimensional structure. Mutations in the related sequences created high levels of variation in the physical properties of these 'intrinsically disordered' regions, which had wide-ranging consequences. Some of these genetic changes even gave individual N-proteins the ability to interact with each other in a completely new way. These results shed new light on the relationship between genetic mutations and the variable physical properties of RNA virus proteins. Nguyen et al. hope that this knowledge will eventually help to develop more effective treatments for viral infections.


Subject(s)
Coronavirus Nucleocapsid Proteins , Mutation , SARS-CoV-2 , SARS-CoV-2/genetics , SARS-CoV-2/chemistry , SARS-CoV-2/metabolism , Coronavirus Nucleocapsid Proteins/genetics , Coronavirus Nucleocapsid Proteins/chemistry , Coronavirus Nucleocapsid Proteins/metabolism , COVID-19/virology , COVID-19/genetics , Humans , Intrinsically Disordered Proteins/chemistry , Intrinsically Disordered Proteins/genetics , Intrinsically Disordered Proteins/metabolism , Phosphoproteins/chemistry , Phosphoproteins/genetics , Phosphoproteins/metabolism , Nucleocapsid Proteins/genetics , Nucleocapsid Proteins/metabolism , Nucleocapsid Proteins/chemistry , Thermodynamics , Protein Stability
4.
Int J Biol Macromol ; 273(Pt 2): 133167, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38885868

ABSTRACT

The Nucleocapsid (N) protein of SARS-CoV-2 plays a crucial role in viral replication and pathogenesis, making it an attractive target for developing antiviral therapeutics. In this study, we used differential scanning fluorimetry to establish a high-throughput screening method for identifying high-affinity ligands of N-terminal domain of the N protein (N-NTD). We screened an FDA-approved drug library of 1813 compounds and identified 102 compounds interacting with N-NTD. The screened compounds were further investigated for their ability to inhibit the nucleic-acid binding activity of the N protein using electrophoretic mobility-shift assays. We have identified three inhibitors, Ceftazidime, Sennoside A, and Tannic acid, that disrupt the N protein's interaction with RNA probe. Ceftazidime and Sennoside A exhibited nano-molar range binding affinities with N protein, determined through surface plasmon resonance. The binding sites of Ceftazidime and Sennoside A were investigated using [1H, 15N]-heteronuclear single quantum coherence (HSQC) NMR spectroscopy. Ceftazidime and Sennoside A bind to the putative RNA binding site of the N protein, thus providing insights into the inhibitory mechanism of these compounds. These findings will contribute to the development of novel antiviral agents targeting the N protein of SARS-CoV-2.


Subject(s)
Antiviral Agents , Coronavirus Nucleocapsid Proteins , SARS-CoV-2 , Antiviral Agents/pharmacology , Antiviral Agents/chemistry , SARS-CoV-2/drug effects , SARS-CoV-2/metabolism , Coronavirus Nucleocapsid Proteins/chemistry , Coronavirus Nucleocapsid Proteins/antagonists & inhibitors , Coronavirus Nucleocapsid Proteins/metabolism , Binding Sites , Humans , Protein Binding , Phosphoproteins/metabolism , Phosphoproteins/chemistry , Phosphoproteins/antagonists & inhibitors , Tannins/chemistry , Tannins/pharmacology , COVID-19 Drug Treatment , Nucleocapsid Proteins/chemistry , Nucleocapsid Proteins/antagonists & inhibitors , Nucleocapsid Proteins/metabolism
5.
Int J Mol Sci ; 25(11)2024 May 25.
Article in English | MEDLINE | ID: mdl-38891942

ABSTRACT

While considerable attention has been devoted to respiratory manifestations, such as pneumonia and acute respiratory distress syndrome (ARDS), emerging evidence underlines the significance of extrapulmonary involvement. In this study, we examined 15 hospitalized patients who succumbed to severe complications following SARS-CoV-2 infection. These patients were admitted to the Sibiu County Clinical Emergency Hospital in Sibiu, Romania, between March and October 2021. All patients were ethnic Romanians. Conducted within a COVID-19-restricted environment and adhering to national safety protocols, autopsies provided a comprehensive understanding of the disease's multisystemic impact. Detailed macroscopic evaluations and histopathological analyses of myocardial, renal, hepatic, splenic, and gastrointestinal tissues were performed. Additionally, reverse transcription-quantitative polymerase chain reaction (rt-qPCR) assays and immunohistochemical staining were employed to detect the viral genome and nucleocapsid within the tissues. Myocardial lesions, including ischemic microstructural changes and inflammatory infiltrates, were prevalent, indicative of COVID-19's cardiac implications, while renal pathology revealed the chronic alterations, acute tubular necrosis, and inflammatory infiltrates most evident. Hepatic examination identified hepatocellular necroinflammatory changes and hepatocytic cytopathy, highlighting the hepatic involvement of SARS-CoV-2 infection. Splenic parenchymal disorganization was prominent, indicating systemic immune dysregulation. Furthermore, gastrointestinal examinations unveiled nonspecific changes. Molecular analyses detected viral genes in various organs, with immunohistochemical assays confirming viral presence predominantly in macrophages and fibroblasts. These findings highlighted the systemic nature of SARS-CoV-2 infection, emphasizing the need for comprehensive clinical management strategies and targeted therapeutic approaches beyond respiratory systems.


Subject(s)
COVID-19 , Genome, Viral , SARS-CoV-2 , Humans , SARS-CoV-2/genetics , COVID-19/virology , COVID-19/genetics , COVID-19/pathology , Male , Female , Middle Aged , Aged , Kidney/virology , Kidney/pathology , Kidney/metabolism , Liver/virology , Liver/pathology , Liver/metabolism , Adult , Spleen/virology , Spleen/pathology , Spleen/metabolism , Romania , Nucleocapsid/genetics , Nucleocapsid/metabolism , Myocardium/pathology , Myocardium/metabolism , Autopsy , Aged, 80 and over , Coronavirus Nucleocapsid Proteins/genetics , Coronavirus Nucleocapsid Proteins/metabolism
6.
Front Cell Infect Microbiol ; 14: 1415885, 2024.
Article in English | MEDLINE | ID: mdl-38846351

ABSTRACT

Corona Virus Disease 2019 (COVID-19) is a highly prevalent and potent infectious disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Until now, the world is still endeavoring to develop new ways to diagnose and treat COVID-19. At present, the clinical prevention and treatment of COVID-19 mainly targets the spike protein on the surface of SRAS-CoV-2. However, with the continuous emergence of SARS-CoV-2 Variants of concern (VOC), targeting the spike protein therapy shows a high degree of limitation. The Nucleocapsid Protein (N protein) of SARS-CoV-2 is highly conserved in virus evolution and is involved in the key process of viral infection and assembly. It is the most expressed viral structural protein after SARS-CoV-2 infection in humans and has high immunogenicity. Therefore, N protein as the key factor of virus infection and replication in basic research and clinical application has great potential research value. This article reviews the research progress on the structure and biological function of SARS-CoV-2 N protein, the diagnosis and drug research of targeting N protein, in order to promote researchers' further understanding of SARS-CoV-2 N protein, and lay a theoretical foundation for the possible outbreak of new and sudden coronavirus infectious diseases in the future.


Subject(s)
COVID-19 , Coronavirus Nucleocapsid Proteins , Phosphoproteins , SARS-CoV-2 , SARS-CoV-2/genetics , Humans , Coronavirus Nucleocapsid Proteins/genetics , Coronavirus Nucleocapsid Proteins/chemistry , Coronavirus Nucleocapsid Proteins/metabolism , COVID-19/virology , COVID-19/diagnosis , Phosphoproteins/metabolism , Spike Glycoprotein, Coronavirus/genetics , Spike Glycoprotein, Coronavirus/metabolism , Spike Glycoprotein, Coronavirus/chemistry , Nucleocapsid Proteins/metabolism , Nucleocapsid Proteins/genetics
7.
J Biol Chem ; 300(6): 107354, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38718862

ABSTRACT

The nucleocapsid protein (N) of SARS-CoV-2 is essential for virus replication, genome packaging, evading host immunity, and virus maturation. N is a multidomain protein composed of an independently folded monomeric N-terminal domain that is the primary site for RNA binding and a dimeric C-terminal domain that is essential for efficient phase separation and condensate formation with RNA. The domains are separated by a disordered Ser/Arg-rich region preceding a self-associating Leu-rich helix. Phosphorylation in the Ser/Arg region in infected cells decreases the viscosity of N:RNA condensates promoting viral replication and host immune evasion. The molecular level effect of phosphorylation, however, is missing from our current understanding. Using NMR spectroscopy and analytical ultracentrifugation, we show that phosphorylation destabilizes the self-associating Leu-rich helix 30 amino-acids distant from the phosphorylation site. NMR and gel shift assays demonstrate that RNA binding by the linker is dampened by phosphorylation, whereas RNA binding to the full-length protein is not significantly affected presumably due to retained strong interactions with the primary RNA-binding domain. Introducing a switchable self-associating domain to replace the Leu-rich helix confirms the importance of linker self-association to droplet formation and suggests that phosphorylation not only increases solubility of the positively charged elongated Ser/Arg region as observed in other RNA-binding proteins but can also inhibit self-association of the Leu-rich helix. These data highlight the effect of phosphorylation both at local sites and at a distant self-associating hydrophobic helix in regulating liquid-liquid phase separation of the entire protein.


Subject(s)
Coronavirus Nucleocapsid Proteins , SARS-CoV-2 , SARS-CoV-2/metabolism , SARS-CoV-2/chemistry , Phosphorylation , Coronavirus Nucleocapsid Proteins/metabolism , Coronavirus Nucleocapsid Proteins/chemistry , Coronavirus Nucleocapsid Proteins/genetics , Humans , RNA, Viral/metabolism , RNA, Viral/chemistry , RNA, Viral/genetics , Phosphoproteins/metabolism , Phosphoproteins/chemistry , Phosphoproteins/genetics , Serine/metabolism , Serine/chemistry , Nucleocapsid Proteins/metabolism , Nucleocapsid Proteins/chemistry , COVID-19/virology , COVID-19/metabolism , Arginine/chemistry , Arginine/metabolism , Protein Binding , Nucleocapsid/metabolism , Nucleocapsid/chemistry , Magnetic Resonance Spectroscopy , Phase Separation
8.
Anal Chem ; 96(23): 9551-9560, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38787915

ABSTRACT

The discovery and identification of broad-spectrum antiviral drugs are of great significance for blocking the spread of pathogenic viruses and corresponding variants of concern. Herein, we proposed a plasmonic imaging-based strategy for assessing the efficacy of potential broad-spectrum antiviral drugs targeting the N-terminal domain of a nucleocapsid protein (NTD) and nucleic acid (NA) interactions. With NTD and NA conjugated gold nanoparticles as core and satellite nanoprobes, respectively, we found that the multivalent binding interactions could drive the formation of core-satellite nanostructures with enhanced scattering brightness due to the plasmonic coupling effect. The core-satellite assembly can be suppressed in the presence of antiviral drugs targeting the NTD-NA interactions, allowing the drug efficacy analysis by detecting the dose-dependent changes in the scattering brightness by plasmonic imaging. By quantifying the changes in the scattering brightness of plasmonic nanoprobes, we uncovered that the constructed multivalent weak interactions displayed a 500-fold enhancement in affinity as compared with the monovalent NTD-NA interactions. We demonstrated the plasmonic imaging-based strategy for evaluating the efficacy of a potential broad-spectrum drug, PJ34, that can target the NTD-NA interactions, with the IC50 as 24.35 and 14.64 µM for SARS-CoV-2 and SARS-CoV, respectively. Moreover, we discovered that ceftazidime holds the potential as a candidate drug to inhibit the NTD-NA interactions with an IC50 of 22.08 µM from molecular docking and plasmonic imaging-based drug analysis. Finally, we validated that the potential antiviral drug, 5-benzyloxygramine, which can induce the abnormal dimerization of nucleocapsid proteins, is effective for SARS-CoV-2, but not effective against SARS-CoV. All these demonstrations indicated that the plasmonic imaging-based strategy is robust and can be used as a powerful strategy for the discovery and identification of broad-spectrum drugs targeting the evolutionarily conserved viral proteins.


Subject(s)
Antiviral Agents , Gold , Metal Nanoparticles , SARS-CoV-2 , Antiviral Agents/pharmacology , Antiviral Agents/chemistry , Gold/chemistry , Metal Nanoparticles/chemistry , SARS-CoV-2/drug effects , SARS-CoV-2/chemistry , Humans , Coronavirus Nucleocapsid Proteins/chemistry , Coronavirus Nucleocapsid Proteins/metabolism , Nucleic Acids/chemistry , Nucleic Acids/metabolism , COVID-19 Drug Treatment , Protein Domains , Phosphoproteins
9.
Virol J ; 21(1): 109, 2024 05 11.
Article in English | MEDLINE | ID: mdl-38734674

ABSTRACT

BACKGROUND: Syndrome coronavirus-2 (SARS-CoV-2) has developed various strategies to evade the antiviral impact of type I IFN. Non-structural proteins and auxiliary proteins have been extensively researched on their role in immune escape. Nevertheless, the detailed mechanisms of structural protein-induced immune evasion have not been well elucidated. METHODS: Human alveolar basal epithelial carcinoma cell line (A549) was stimulated with polyinosinic-polycytidylic acid (PIC) and independently transfected with four structural proteins expression plasmids, including nucleocapsid (N), spike (S), membrane (M) and envelope (E) proteins. By RT-qPCR and ELISA, the structural protein with the most pronounced inhibitory effects on IFN-ß induction was screened. RNA-sequencing (RNA-Seq) and two differential analysis strategies were used to obtain differentially expressed genes associated with N protein inhibition of IFN-ß induction. Based on DIANA-LncBase and StarBase databases, the interactive competitive endogenous RNA (ceRNA) network for N protein-associated genes was constructed. By combining single-cell sequencing data (GSE158055), lncRNA-miRNA-mRNA axis was further determined. Finally, RT-qPCR was utilized to illustrate the regulatory functions among components of the ceRNA axis. RESULTS: SARS-CoV-2 N protein inhibited IFN-ß induction in human alveolar epithelial cells most significantly compared with other structural proteins. RNA-Seq data analysis revealed genes related to N protein inhibiting IFNs induction. The obtained 858 differentially expressed genes formed the reliable ceRNA network. The function of LINC01002-miR-4324-FRMD8 axis in the IFN-dominated immune evasion was further demonstrated through integrating single-cell sequencing data. Moreover, we validated that N protein could reverse the effect of PIC on LINC01002, FRMD8 and miR-4324 expression, and subsequently on IFN-ß expression level. And LINC01002 could regulate the production of FRMD8 by inhibiting miR-4324. CONCLUSION: SARS-CoV-2 N protein suppressed the induction of IFN-ß by regulating LINC01002 which was as a ceRNA, sponging miR-4324 and participating in the regulation of FRMD8 mRNA. Our discovery provides new insights into early intervention therapy and drug development on SARS-CoV-2 infection.


Subject(s)
COVID-19 , MicroRNAs , RNA, Long Noncoding , SARS-CoV-2 , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , COVID-19/virology , COVID-19/immunology , SARS-CoV-2/genetics , A549 Cells , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Interferon-beta/genetics , Interferon-beta/metabolism , Immune Evasion , Coronavirus Nucleocapsid Proteins/genetics , Coronavirus Nucleocapsid Proteins/metabolism , RNA, Competitive Endogenous , Phosphoproteins
10.
Cell Commun Signal ; 22(1): 296, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38807115

ABSTRACT

BACKGROUND: The SARS-CoV-2 virus causes severe COVID-19 in one-fifth of patients. In addition to high mortality, infection may induce respiratory failure and cardiovascular complications associated with inflammation. Acute or prolonged inflammation results in organ fibrosis, the cause of which might be endothelial disorders arising during the endothelial-mesenchymal transition (EndMT). METHODS: HUVECs and HMEC-1 cells were stimulated with SARS-CoV-2 S (Spike) and N (Nucleocapsid) proteins, and EndMT induction was evaluated by studying specific protein markers via Western blotting. Wound healing and tube formation assays were employed to assess the potential of SARS-CoV-2 to stimulate changes in cell behaviour. MRTF nuclear translocation, ROS generation, TLR4 inhibitors, TGF-ß-neutralizing antibodies, and inhibitors of the TGF-ß-dependent pathway were used to investigate the role of the TGF-ß-MRTF signalling axis in SARS-CoV-2-dependent EndMT stimulation. RESULTS: Both viral proteins stimulate myofibroblast trans-differentiation. However, the N protein is more effective at EndMT induction. The TGF-ß-MRTF pathway plays a critical role in this process. The N protein preferentially favours action through TGF-ß2, whose secretion is induced through TLR4-ROS action. TGF-ß2 stimulates MRTF-A and MRTF-B nuclear translocation and strongly regulates EndMT. In contrast, the Spike protein stimulates TGF-ß1 secretion as a result of ACE2 downregulation. TGF-ß1 induces only MRTF-B, which, in turn, weakly regulates EndMT. Furthermore, aspirin, a common nonsteroidal anti-inflammatory drug, might prevent and reverse SARS-CoV-2-dependent EndMT induction through TGF-ß-MRTF pathway deregulation. CONCLUSION: The reported study revealed that SARS-CoV-2 infection induces EndMT. Moreover, it was demonstrated for the first time at the molecular level that the intensity of the EndMT triggered by SARS-CoV-2 infection may vary and depend on the viral protein involved. The N protein acts through TLR4-ROS-TGF-ß2-MRTF-A/B, whereas the S protein acts through ACE2-TGF-ß1-MRTF-B. Furthermore, we identified aspirin as a potential anti-fibrotic drug for treating patients with SARS-CoV-2 infection.


Subject(s)
Aspirin , COVID-19 , Coronavirus Nucleocapsid Proteins , Epithelial-Mesenchymal Transition , SARS-CoV-2 , Signal Transduction , Spike Glycoprotein, Coronavirus , Transforming Growth Factor beta , Humans , Spike Glycoprotein, Coronavirus/metabolism , SARS-CoV-2/drug effects , SARS-CoV-2/physiology , Transforming Growth Factor beta/metabolism , COVID-19/metabolism , COVID-19/virology , Coronavirus Nucleocapsid Proteins/metabolism , Aspirin/pharmacology , Signal Transduction/drug effects , Epithelial-Mesenchymal Transition/drug effects , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Transcription Factors/metabolism , Toll-Like Receptor 4/metabolism , Cell Line , Endothelial-Mesenchymal Transition , Phosphoproteins
11.
Biochem J ; 481(11): 669-682, 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38713013

ABSTRACT

The fundamental biology of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) nucleocapsid protein (Ncap), its use in diagnostic assays and its potential application as a vaccine component have received considerable attention since the outbreak of the Covid19 pandemic in late 2019. Here we report the scalable expression and purification of soluble, immunologically active, SARS-CoV-2 Ncap in Escherichia coli. Codon-optimised synthetic genes encoding the original Ncap sequence and four common variants with an N-terminal 6His affinity tag (sequence MHHHHHHG) were cloned into an inducible expression vector carrying a regulated bacteriophage T5 synthetic promoter controlled by lac operator binding sites. The constructs were used to express Ncap proteins and protocols developed which allow efficient production of purified Ncap with yields of over 200 mg per litre of culture media. These proteins were deployed in ELISA assays to allow comparison of their responses to human sera. Our results suggest that there was no detectable difference between the 6His-tagged and untagged original Ncap proteins but there may be a slight loss of sensitivity of sera to other Ncap isolates.


Subject(s)
COVID-19 , Coronavirus Nucleocapsid Proteins , Escherichia coli , SARS-CoV-2 , Escherichia coli/genetics , Escherichia coli/metabolism , Coronavirus Nucleocapsid Proteins/genetics , Coronavirus Nucleocapsid Proteins/metabolism , Coronavirus Nucleocapsid Proteins/biosynthesis , Coronavirus Nucleocapsid Proteins/isolation & purification , Coronavirus Nucleocapsid Proteins/chemistry , SARS-CoV-2/genetics , SARS-CoV-2/metabolism , Humans , COVID-19/virology , Phosphoproteins/genetics , Phosphoproteins/isolation & purification , Phosphoproteins/metabolism
12.
Nucleic Acids Res ; 52(11): 6647-6661, 2024 Jun 24.
Article in English | MEDLINE | ID: mdl-38587193

ABSTRACT

The viral genome of SARS-CoV-2 is packaged by the nucleocapsid (N-)protein into ribonucleoprotein particles (RNPs), 38 ± 10 of which are contained in each virion. Their architecture has remained unclear due to the pleomorphism of RNPs, the high flexibility of N-protein intrinsically disordered regions, and highly multivalent interactions between viral RNA and N-protein binding sites in both N-terminal (NTD) and C-terminal domain (CTD). Here we explore critical interaction motifs of RNPs by applying a combination of biophysical techniques to ancestral and mutant proteins binding different nucleic acids in an in vitro assay for RNP formation, and by examining nucleocapsid protein variants in a viral assembly assay. We find that nucleic acid-bound N-protein dimers oligomerize via a recently described protein-protein interface presented by a transient helix in its long disordered linker region between NTD and CTD. The resulting hexameric complexes are stabilized by multivalent protein-nucleic acid interactions that establish crosslinks between dimeric subunits. Assemblies are stabilized by the dimeric CTD of N-protein offering more than one binding site for stem-loop RNA. Our study suggests a model for RNP assembly where N-protein scaffolding at high density on viral RNA is followed by cooperative multimerization through protein-protein interactions in the disordered linker.


Subject(s)
Coronavirus Nucleocapsid Proteins , Protein Multimerization , RNA, Viral , SARS-CoV-2 , SARS-CoV-2/genetics , SARS-CoV-2/metabolism , SARS-CoV-2/chemistry , Coronavirus Nucleocapsid Proteins/chemistry , Coronavirus Nucleocapsid Proteins/metabolism , Coronavirus Nucleocapsid Proteins/genetics , RNA, Viral/metabolism , RNA, Viral/chemistry , RNA, Viral/genetics , Protein Binding , Binding Sites , Ribonucleoproteins/metabolism , Ribonucleoproteins/chemistry , Ribonucleoproteins/genetics , Virus Assembly/genetics , Humans , Nucleocapsid Proteins/chemistry , Nucleocapsid Proteins/metabolism , Nucleocapsid Proteins/genetics , Models, Molecular , Phosphoproteins/chemistry , Phosphoproteins/metabolism , Phosphoproteins/genetics , COVID-19/virology
13.
J Med Virol ; 96(3): e29531, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38515377

ABSTRACT

The Nucleocapsid Protein (NP) of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is not only the core structural protein required for viral packaging, but also participates in the regulation of viral replication, and its post-translational modifications such as phosphorylation have been shown to be an important strategy for regulating virus proliferation. Our previous work identified NP could be ubiquitinated, as confirmed by two independent studies. But the function of NP ubiquitination is currently unknown. In this study, we first pinpointed TRIM6 as the E3 ubiquitin ligase responsible for NP ubiquitination, binding to NP's CTD via its RING and B-box-CCD domains. TRIM6 promotes the K29-typed polyubiquitination of NP at K102, K347, and K361 residues, increasing its binding to viral genomic RNA. Consistently, functional experiments such as the use of the reverse genetic tool trVLP model and gene knockout of TRIM6 further confirmed that blocking the ubiquitination of NP by TRIM6 significantly inhibited the proliferation of SARS-CoV-2. Notably, the NP of coronavirus is relatively conserved, and the NP of SARS-CoV can also be ubiquitinated by TRIM6, indicating that NP could be a broad-spectrum anti-coronavirus target. These findings shed light on the intricate interaction between SARS-CoV-2 and the host, potentially opening new opportunities for COVID-19 therapeutic development.


Subject(s)
COVID-19 , Genome, Viral , SARS-CoV-2 , Ubiquitin-Protein Ligases , Humans , Cell Proliferation , COVID-19/genetics , COVID-19/virology , Nucleocapsid Proteins/genetics , RNA, Viral/genetics , SARS-CoV-2/genetics , SARS-CoV-2/metabolism , Tripartite Motif Proteins/genetics , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitination , Coronavirus Nucleocapsid Proteins/genetics , Coronavirus Nucleocapsid Proteins/metabolism
14.
Chemistry ; 30(29): e202400048, 2024 May 23.
Article in English | MEDLINE | ID: mdl-38483823

ABSTRACT

Recently, the discovery of antimicrobial peptides (AMPs) as excellent candidates for overcoming antibiotic resistance has attracted significant attention. AMPs are short peptides active against bacteria, cancer cells, and viruses. It has been shown that the SARS-CoV-2 nucleocapsid protein (N-P) undergoes liquid-liquid phase separation in the presence of RNA, resulting in biocondensate formation. These biocondensates are crucial for viral replication as they concentrate the viral RNA with the host cell's protein machinery required for viral protein expression. Thus, N-P biocondensates are promising targets to block or slow down viral RNA transcription and consequently virion assembly. We investigated the ability of three AMPs to interfere with N-P/RNA condensates. Using microscopy techniques, supported by biophysical characterization, we found that the AMP LL-III partitions into the condensate, leading to clustering. Instead, the AMP CrACP1 partitions into the droplets without affecting their morphology but reducing their dynamics. Conversely, GKY20 leads to the formation of fibrillar structures after partitioning. It can be expected that such morphological transformation severely impairs the normal functionality of the N-P droplets and thus virion assembly. These results could pave the way for the development of a new class of AMP-based antiviral agents targeting biocondensates.


Subject(s)
Antimicrobial Peptides , Coronavirus Nucleocapsid Proteins , SARS-CoV-2 , SARS-CoV-2/drug effects , SARS-CoV-2/metabolism , Antimicrobial Peptides/chemistry , Antimicrobial Peptides/pharmacology , Coronavirus Nucleocapsid Proteins/chemistry , Coronavirus Nucleocapsid Proteins/metabolism , Humans , RNA, Viral/metabolism , RNA, Viral/chemistry , Phosphoproteins/chemistry , Phosphoproteins/metabolism , Antiviral Agents/pharmacology , Antiviral Agents/chemistry , Virus Replication/drug effects
15.
Nucleic Acids Res ; 52(5): 2609-2624, 2024 Mar 21.
Article in English | MEDLINE | ID: mdl-38153183

ABSTRACT

The SARS-CoV-2 Nucleocapsid (N) protein is responsible for condensation of the viral genome. Characterizing the mechanisms controlling nucleic acid binding is a key step in understanding how condensation is realized. Here, we focus on the role of the RNA binding domain (RBD) and its flanking disordered N-terminal domain (NTD) tail, using single-molecule Förster Resonance Energy Transfer and coarse-grained simulations. We quantified contact site size and binding affinity for nucleic acids and concomitant conformational changes occurring in the disordered region. We found that the disordered NTD increases the affinity of the RBD for RNA by about 50-fold. Binding of both nonspecific and specific RNA results in a modulation of the tail configurations, which respond in an RNA length-dependent manner. Not only does the disordered NTD increase affinity for RNA, but mutations that occur in the Omicron variant modulate the interactions, indicating a functional role of the disordered tail. Finally, we found that the NTD-RBD preferentially interacts with single-stranded RNA and that the resulting protein:RNA complexes are flexible and dynamic. We speculate that this mechanism of interaction enables the Nucleocapsid protein to search the viral genome for and bind to high-affinity motifs.


Subject(s)
Coronavirus Nucleocapsid Proteins , RNA, Viral , SARS-CoV-2 , Humans , Coronavirus Nucleocapsid Proteins/chemistry , Coronavirus Nucleocapsid Proteins/metabolism , COVID-19/virology , Nucleocapsid Proteins/chemistry , Protein Binding , RNA, Viral/metabolism , SARS-CoV-2/genetics , SARS-CoV-2/metabolism
16.
J Virol ; 97(11): e0147023, 2023 Nov 30.
Article in English | MEDLINE | ID: mdl-37882521

ABSTRACT

IMPORTANCE: As a member of the δ-coronavirus family, porcine deltacoronavirus (PDCoV) is a vital reason for diarrhea in piglets, which can contribute to high morbidity and mortality rates. Initially identified in Hong Kong in 2012, the virus has rapidly spread worldwide. During PDCoV infection, the virus employs evasion mechanisms to evade host surveillance, while the host mounts corresponding responses to impede viral replication. Our research has revealed that PDCoV infection down-regulates the expression of PGAM5 to promote virus replication. In contrast, PGAM5 degrades PDCoV N through autophagy by interacting with the cargo receptor P62 and the E3 ubiquitination ligase STUB1. Additionally, PGAM5 interacts with MyD88 and TRAF3 to activate the IFN signal pathway, resulting in the inhibition of viral replication.


Subject(s)
Coronavirus Infections , Coronavirus Nucleocapsid Proteins , Deltacoronavirus , Interferon Type I , Mitochondrial Proteins , Phosphoprotein Phosphatases , Proteolysis , Swine Diseases , Swine , Virus Replication , Animals , Coronavirus Infections/immunology , Coronavirus Infections/veterinary , Coronavirus Infections/virology , Interferon Type I/immunology , Signal Transduction , Swine/virology , Swine Diseases/virology , Ubiquitin-Protein Ligases/metabolism , Virus Replication/immunology , Coronavirus Nucleocapsid Proteins/metabolism , Deltacoronavirus/immunology , Deltacoronavirus/metabolism , Phosphoprotein Phosphatases/metabolism , Mitochondrial Proteins/metabolism , Down-Regulation , Immune Evasion , RNA-Binding Proteins/metabolism
17.
J Med Virol ; 95(4): e28719, 2023 04.
Article in English | MEDLINE | ID: mdl-37185839

ABSTRACT

The innate immune response is the first line of host defense against viral infections, but its role in immunity against SARS-CoV-2 remains unclear. By using immunoprecipitation coupled with mass spectroscopy, we observed that the E3 ubiquitin ligase TRIM21 interacted with the SARS-CoV-2 nucleocapsid (N) protein and ubiquitinated it at Lys375 . Upon determining the topology of the TRIM21-mediated polyubiquitination chain on N protein, we then found that polyubiquitination led to tagging of the N protein for degradation by the host cell proteasome. Furthermore, TRIM21 also ubiquitinated the N proteins of SARS-CoV-2 variants of concern, including Alpha, Beta, Gamma, Delta, and Omicron together with SARS-CoV and MERS-CoV variants. Herein, we propose that ubiquitylation and degradation of the SARS-CoV-2 N protein inhibited SARS-CoV-2 viral particle assembly, by which it probably involved in preventing cytokine storm. Eventually, our study has fully revealed the association between the host innate immune system and SARS-CoV-2 N protein, which may aid in developing novel SARS-CoV-2 treatment strategies.


Subject(s)
COVID-19 , SARS-CoV-2 , Humans , Immunity, Innate , SARS-CoV-2/metabolism , Ubiquitin/metabolism , Ubiquitination , Coronavirus Nucleocapsid Proteins/metabolism
18.
Nucleic Acids Res ; 51(9): 4555-4571, 2023 05 22.
Article in English | MEDLINE | ID: mdl-36928389

ABSTRACT

The pandemic caused by SARS-CoV-2 has called for concerted efforts to generate new insights into the biology of betacoronaviruses to inform drug screening and development. Here, we establish a workflow to determine the RNA recognition and druggability of the nucleocapsid N-protein of SARS-CoV-2, a highly abundant protein crucial for the viral life cycle. We use a synergistic method that combines NMR spectroscopy and protein-RNA cross-linking coupled to mass spectrometry to quickly determine the RNA binding of two RNA recognition domains of the N-protein. Finally, we explore the druggability of these domains by performing an NMR fragment screening. This workflow identified small molecule chemotypes that bind to RNA binding interfaces and that have promising properties for further fragment expansion and drug development.


Subject(s)
COVID-19 Drug Treatment , COVID-19 , Coronavirus Nucleocapsid Proteins , Drug Development , SARS-CoV-2 , Humans , COVID-19/virology , RNA, Viral/metabolism , SARS-CoV-2/drug effects , SARS-CoV-2/metabolism , Coronavirus Nucleocapsid Proteins/antagonists & inhibitors , Coronavirus Nucleocapsid Proteins/chemistry , Coronavirus Nucleocapsid Proteins/metabolism , Nuclear Magnetic Resonance, Biomolecular , Mass Spectrometry , Workflow , Protein Binding
19.
Nature ; 615(7950): 143-150, 2023 03.
Article in English | MEDLINE | ID: mdl-36630998

ABSTRACT

The SARS-CoV-2 Omicron variant is more immune evasive and less virulent than other major viral variants that have so far been recognized1-12. The Omicron spike (S) protein, which has an unusually large number of mutations, is considered to be the main driver of these phenotypes. Here we generated chimeric recombinant SARS-CoV-2 encoding the S gene of Omicron (BA.1 lineage) in the backbone of an ancestral SARS-CoV-2 isolate, and compared this virus with the naturally circulating Omicron variant. The Omicron S-bearing virus robustly escaped vaccine-induced humoral immunity, mainly owing to mutations in the receptor-binding motif; however, unlike naturally occurring Omicron, it efficiently replicated in cell lines and primary-like distal lung cells. Similarly, in K18-hACE2 mice, although virus bearing Omicron S caused less severe disease than the ancestral virus, its virulence was not attenuated to the level of Omicron. Further investigation showed that mutating non-structural protein 6 (nsp6) in addition to the S protein was sufficient to recapitulate the attenuated phenotype of Omicron. This indicates that although the vaccine escape of Omicron is driven by mutations in S, the pathogenicity of Omicron is determined by mutations both in and outside of the S protein.


Subject(s)
COVID-19 , Coronavirus Nucleocapsid Proteins , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , Virulence Factors , Virulence , Animals , Mice , Cell Line , Immune Evasion , SARS-CoV-2/immunology , SARS-CoV-2/metabolism , SARS-CoV-2/pathogenicity , Spike Glycoprotein, Coronavirus/genetics , Spike Glycoprotein, Coronavirus/metabolism , Coronavirus Nucleocapsid Proteins/genetics , Coronavirus Nucleocapsid Proteins/metabolism , Virulence Factors/genetics , Virulence Factors/metabolism , Humans , COVID-19 Vaccines/immunology , Lung/cytology , Lung/virology , Virus Replication , Mutation
20.
Nucleic Acids Res ; 51(1): 290-303, 2023 01 11.
Article in English | MEDLINE | ID: mdl-36533523

ABSTRACT

The SARS-CoV-2 nucleocapsid (N) protein performs several functions including binding, compacting, and packaging the ∼30 kb viral genome into the viral particle. N protein consists of two ordered domains, with the N terminal domain (NTD) primarily associated with RNA binding and the C terminal domain (CTD) primarily associated with dimerization/oligomerization, and three intrinsically disordered regions, an N-arm, a C-tail, and a linker that connects the NTD and CTD. We utilize an optical tweezers system to isolate a long single-stranded nucleic acid substrate to measure directly the binding and packaging function of N protein at a single molecule level in real time. We find that N protein binds the nucleic acid substrate with high affinity before oligomerizing and forming a highly compact structure. By comparing the activities of truncated protein variants missing the NTD, CTD, and/or linker, we attribute specific steps in this process to the structural domains of N protein, with the NTD driving initial binding to the substrate and ensuring high localized protein density that triggers interprotein interactions mediated by the CTD, which forms a compact and stable protein-nucleic acid complex suitable for packaging into the virion.


Subject(s)
COVID-19 , Coronavirus Nucleocapsid Proteins , RNA, Viral , SARS-CoV-2 , Humans , COVID-19/virology , Protein Domains , RNA, Viral/metabolism , SARS-CoV-2/genetics , SARS-CoV-2/metabolism , Coronavirus Nucleocapsid Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...