Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
J Immunol ; 208(3): 753-761, 2022 02 01.
Article in English | MEDLINE | ID: covidwho-1614089

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which causes coronavirus disease 2019 (COVID-19), has seriously threatened global public health. Severe COVID-19 has been reported to be associated with an impaired IFN response. However, the mechanisms of how SARS-CoV-2 antagonizes the host IFN response are poorly understood. In this study, we report that SARS-CoV-2 helicase NSP13 inhibits type I IFN production by directly targeting TANK-binding kinase 1 (TBK1) for degradation. Interestingly, inhibition of autophagy by genetic knockout of Beclin1 or pharmacological inhibition can rescue NSP13-mediated TBK1 degradation in HEK-293T cells. Subsequent studies revealed that NSP13 recruits TBK1 to p62, and the absence of p62 can also inhibit TBK1 degradation in HEK-293T and HeLa cells. Finally, TBK1 and p62 degradation and p62 aggregation were observed during SARS-CoV-2 infection in HeLa-ACE2 and Calu3 cells. Overall, our study shows that NSP13 inhibits type I IFN production by recruiting TBK1 to p62 for autophagic degradation, enabling it to evade the host innate immune response, which provides new insights into the transmission and pathogenesis of SARS-CoV-2 infection.


Subject(s)
Autophagy , COVID-19/immunology , Coronavirus RNA-Dependent RNA Polymerase/physiology , Interferon Type I/biosynthesis , Methyltransferases/physiology , Protein Serine-Threonine Kinases/metabolism , RNA Helicases/physiology , SARS-CoV-2/physiology , Sequestosome-1 Protein/metabolism , Viral Nonstructural Proteins/physiology , Beclin-1/antagonists & inhibitors , Cell Line , Down-Regulation , Humans , Immune Evasion , Immunity, Innate , Immunoprecipitation , Interferon Type I/genetics , Multiprotein Complexes , Protein Aggregates , Protein Interaction Mapping
2.
RNA Biol ; 18(sup2): 804-817, 2021 11 12.
Article in English | MEDLINE | ID: covidwho-1522048

ABSTRACT

Nsp1 of SARS-CoV-2 regulates the translation of host and viral mRNAs in cells. Nsp1 inhibits host translation initiation by occluding the entry channel of the 40S ribosome subunit. The structural study of the Nsp1-ribosomal complexes reported post-termination 80S complex containing Nsp1, eRF1 and ABCE1. Considering the presence of Nsp1 in the post-termination 80S ribosomal complex, we hypothesized that Nsp1 may be involved in translation termination. Using a cell-free translation system and reconstituted in vitro translation system, we show that Nsp1 stimulates peptide release and formation of termination complexes. Detailed analysis of Nsp1 activity during translation termination stages reveals that Nsp1 facilitates stop codon recognition. We demonstrate that Nsp1 stimulation targets eRF1 and does not affect eRF3. Moreover, Nsp1 increases amount of the termination complexes at all three stop codons. The activity of Nsp1 in translation termination is provided by its N-terminal domain and the minimal required part of eRF1 is NM domain. We assume that the biological meaning of Nsp1 activity in translation termination is binding with the 80S ribosomes translating host mRNAs and remove them from the pool of the active ribosomes.


Subject(s)
Protein Biosynthesis , SARS-CoV-2 , Viral Nonstructural Proteins/physiology , Animals , Cell-Free System , Codon, Terminator/metabolism , GTP Phosphohydrolases/metabolism , HeLa Cells , Humans , Mutation , Peptide Chain Termination, Translational , Peptide Termination Factors/chemistry , Peptide Termination Factors/metabolism , Peptides/chemistry , Protein Binding , Protein Conformation , Protein Domains , RNA, Messenger/metabolism , Rabbits , Ribosomes/metabolism
3.
Genomics Proteomics Bioinformatics ; 19(5): 707-726, 2021 10.
Article in English | MEDLINE | ID: covidwho-1509803

ABSTRACT

The coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection is currently a global pandemic. Extensive investigations have been performed to study the clinical and cellular effects of SARS-CoV-2 infection. Mass spectrometry-based proteomics studies have revealed the cellular changes due to the infection and identified a plethora of interactors for all SARS-CoV-2 components, except for the longest non-structural protein 3 (NSP3). Here, we expressed the full-length NSP3 proteins of SARS-CoV and SARS-CoV-2 to investigate their unique and shared functions using multi-omics methods. We conducted interactome, phosphoproteome, ubiquitylome, transcriptome, and proteome analyses of NSP3-expressing cells. We found that NSP3 plays essential roles in cellular functions such as RNA metabolism and immune response (e.g., NF-κB signal transduction). Interestingly, we showed that SARS-CoV-2 NSP3 has both endoplasmic reticulum and mitochondrial localizations. In addition, SARS-CoV-2 NSP3 is more closely related to mitochondrial ribosomal proteins, whereas SARS-CoV NSP3 is related to the cytosolic ribosomal proteins. In summary, our integrative multi-omics study of NSP3 improves the understanding of the functions of NSP3 and offers potential targets for the development of anti-SARS strategies.


Subject(s)
Coronavirus Papain-Like Proteases/physiology , RNA-Dependent RNA Polymerase/physiology , SARS-CoV-2/physiology , Severe acute respiratory syndrome-related coronavirus/physiology , Viral Nonstructural Proteins/physiology , COVID-19 , Humans , Proteome , Ribosomal Proteins
4.
Cell Rep ; 36(9): 109650, 2021 08 31.
Article in English | MEDLINE | ID: covidwho-1363915

ABSTRACT

Coronaviruses have evolved elaborate multisubunit machines to replicate and transcribe their genomes. Central to these machines are the RNA-dependent RNA polymerase subunit (nsp12) and its intimately associated cofactors (nsp7 and nsp8). We use a high-throughput magnetic-tweezers approach to develop a mechanochemical description of this core polymerase. The core polymerase exists in at least three catalytically distinct conformations, one being kinetically consistent with incorporation of incorrect nucleotides. We provide evidence that the RNA-dependent RNA polymerase (RdRp) uses a thermal ratchet instead of a power stroke to transition from the pre- to post-translocated state. Ultra-stable magnetic tweezers enable the direct observation of coronavirus polymerase deep and long-lived backtracking that is strongly stimulated by secondary structures in the template. The framework we present here elucidates one of the most important structure-dynamics-function relationships in human health today and will form the grounds for understanding the regulation of this complex.


Subject(s)
COVID-19/virology , Coronavirus RNA-Dependent RNA Polymerase/physiology , Nucleotides/metabolism , RNA, Viral/biosynthesis , SARS-CoV-2/physiology , Coronavirus RNA-Dependent RNA Polymerase/chemistry , High-Throughput Screening Assays , Humans , Models, Molecular , Molecular Conformation , Nucleotides/chemistry , RNA, Viral/chemistry , Single Molecule Imaging , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/physiology
5.
Cell Rep ; 36(5): 109482, 2021 08 03.
Article in English | MEDLINE | ID: covidwho-1312984

ABSTRACT

Bearing a relatively large single-stranded RNA genome in nature, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) utilizes sophisticated replication/transcription complexes (RTCs), mainly composed of a network of nonstructural proteins and nucleocapsid protein, to establish efficient infection. In this study, we develop an innovative interaction screening strategy based on phase separation in cellulo, namely compartmentalization of protein-protein interactions in cells (CoPIC). Utilizing CoPIC screening, we map the interaction network among RTC-related viral proteins. We identify a total of 47 binary interactions among 14 proteins governing replication, discontinuous transcription, and translation of coronaviruses. Further exploration via CoPIC leads to the discovery of extensive ternary complexes composed of these components, which infer potential higher-order complexes. Taken together, our results present an efficient and robust interaction screening strategy, and they indicate the existence of a complex interaction network among RTC-related factors, thus opening up opportunities to understand SARS-CoV-2 biology and develop therapeutic interventions for COVID-19.


Subject(s)
COVID-19/virology , Protein Interaction Mapping/methods , Proteome , SARS-CoV-2/pathogenicity , Viral Nonstructural Proteins/physiology , Animals , Caco-2 Cells , Cell Compartmentation , Cell Line , Chlorocebus aethiops , HEK293 Cells , Humans , Protein Interaction Maps , Vero Cells , Virus Replication
6.
Bosn J Basic Med Sci ; 21(5): 515-527, 2021 Oct 01.
Article in English | MEDLINE | ID: covidwho-1134549

ABSTRACT

Coronavirus disease-19 (COVID-19) is an extremely infectious disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) that has become a major global health concern. The induction of a coordinated immune response is crucial to the elimination of any pathogenic infection. However, SARS-CoV-2 can modulate the host immune system to favor viral adaptation and persistence within the host. The virus can counteract type I interferon (IFN-I) production, attenuating IFN-I signaling pathway activation and disrupting antigen presentation. Simultaneously, SARS-CoV-2 infection can enhance apoptosis and the production of inflammatory mediators, which ultimately results in increased disease severity. SARS-CoV-2 produces an array of effector molecules, including nonstructural proteins (NSPs) and open-reading frames (ORFs) accessory proteins. We describe the complex molecular interplay of SARS-CoV-2 NSPs and accessory proteins with the host's signaling mediating immune evasion in the current review. In addition, the crucial role played by immunomodulation therapy to address immune evasion is discussed. Thus, the current review can provide new directions for the development of vaccines and specific therapies.


Subject(s)
COVID-19/immunology , Immune Evasion/physiology , Immunity, Innate/physiology , SARS-CoV-2/pathogenicity , Viral Nonstructural Proteins/physiology , Viral Regulatory and Accessory Proteins/physiology , Humans
7.
Curr Mol Med ; 22(1): 50-66, 2022.
Article in English | MEDLINE | ID: covidwho-1099962

ABSTRACT

The proteins of coronavirus are classified as non-structural, structural, and accessory. There are 16 non-structural viral proteins besides their precursors (1a and 1ab polyproteins). The non-structural proteins are named nsp1 to nsp16, and they act as enzymes, coenzymes, and binding proteins to facilitate the replication, transcription, and translation of the virus. The structural proteins are bound to the RNA in the nucleocapsid (N- protein) or to the lipid bilayer membrane of the viral envelope. The lipid bilayer proteins include the membrane protein (M), an envelope protein (E), and spike protein (S). Besides their role as structural proteins, they are essential for the host cells' binding and invasion. The SARS-CoV-2 contains six accessory proteins which participate in the viral replication, assembly and virus-host interactions. The SARS-CoV-2 accessory proteins are orf3a, orf6, orf7a, orf7b, orf8, and orf10. The functions of the SARS-CoV-2 are not well known, while the functions of their corresponding proteins in SARS-CoV are either well known or poorly studied. Recently, the Oxford University and Astrazeneca, Pfizer and BioNTech have made SARS-CoV-2 vaccines by targeting the spike protein gene. The US Food and Drug Administration (FDA) and the health authorities of the United Kingdom have approved and started conducting vaccinations using the Pfizer and BioNTech mRNA vaccine. Also, The FDA of the USA has approved the use of two monoclonal antibodies produced by Regeneron pharmaceuticals to target the spike protein for treating COVID-19. The SARS-CoV-2 proteins can be used for the diagnosis, as drug targets and in vaccination trials for COVID-19. In future COVID-19 research, more efforts should be made to elaborate the functions and structure of the SARS-CoV- 2 proteins so as to use them as targets for COVID-19 drugs and vaccines. Special attention should be paid to extensive research on the SARS-CoV-2 nsp3, orf8, and orf10.


Subject(s)
Antiviral Agents/pharmacology , COVID-19 Vaccines , COVID-19/prevention & control , SARS-CoV-2/chemistry , Viral Proteins/drug effects , Viral Proteins/immunology , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/therapeutic use , Antibodies, Viral/immunology , Antibodies, Viral/therapeutic use , Antigens, Viral/immunology , COVID-19/immunology , Drug Design , Humans , Immunotherapy , SARS-CoV-2/drug effects , SARS-CoV-2/immunology , Spike Glycoprotein, Coronavirus/immunology , Vaccine Development , Viral Nonstructural Proteins/drug effects , Viral Nonstructural Proteins/immunology , Viral Nonstructural Proteins/physiology , Viral Proteins/physiology , Viral Regulatory and Accessory Proteins/drug effects , Viral Regulatory and Accessory Proteins/immunology , Viral Regulatory and Accessory Proteins/physiology , Viral Structural Proteins/drug effects , Viral Structural Proteins/immunology , Viral Structural Proteins/physiology , mRNA Vaccines , COVID-19 Drug Treatment
8.
Expert Opin Ther Pat ; 31(4): 339-350, 2021 Apr.
Article in English | MEDLINE | ID: covidwho-1087605

ABSTRACT

Introduction: Coronaviruses encode a helicase that is essential for viral replication and represents an excellent antiviral target. However, only a few coronavirus helicase inhibitors have been patented. These patents include drug-like compound SSYA10-001, aryl diketo acids (ADK), and dihydroxychromones. Additionally, adamantane-derived bananins, natural flavonoids, one acrylamide derivative [(E)-3-(furan-2-yl)-N-(4-sulfamoylphenyl)acrylamide], a purine derivative (7-ethyl-8-mercapto-3-methyl-3,7-dihydro-1 H-purine-2,6-dione), and a few bismuth complexes. The IC50 of patented inhibitors ranges between 0.82 µM and 8.95 µM, depending upon the assays used. Considering the urgency of clinical interventions against Coronavirus Disease-19 (COVID-19), it is important to consider developing antiviral portfolios consisting of small molecules.Areas covered: This review examines coronavirus helicases as antiviral targets, and the potential of previously patented and experimental compounds to inhibit the Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2) helicase.Expert opinion: Small molecule coronavirus helicase inhibitors represent attractive pharmacological modalities for the treatment of coronaviruses such as SARS-CoV and SARS-CoV-2. Rightfully so, the current emphasis is focused upon the development of vaccines. However, vaccines may not work for everyone and broad-based adoption of vaccinations is an increasingly challenging societal endeavor. Therefore, it is important to develop additional pharmacological antivirals against the highly conserved coronavirus helicases to broadly protect against this and subsequent coronavirus epidemics.


Subject(s)
Antiviral Agents/pharmacology , COVID-19 Drug Treatment , Drug Development , Methyltransferases/antagonists & inhibitors , RNA Helicases/antagonists & inhibitors , SARS-CoV-2/drug effects , Viral Nonstructural Proteins/antagonists & inhibitors , Humans , Methyltransferases/chemistry , Methyltransferases/physiology , Patents as Topic , RNA Helicases/chemistry , RNA Helicases/physiology , Triazoles/pharmacology , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/physiology
9.
Cells ; 10(2)2021 02 02.
Article in English | MEDLINE | ID: covidwho-1060037

ABSTRACT

Many viruses disrupt host gene expression by degrading host mRNAs and/or manipulating translation activities to create a cellular environment favorable for viral replication. Often, virus-induced suppression of host gene expression, including those involved in antiviral responses, contributes to viral pathogenicity. Accordingly, clarifying the mechanisms of virus-induced disruption of host gene expression is important for understanding virus-host cell interactions and virus pathogenesis. Three highly pathogenic human coronaviruses (CoVs), including severe acute respiratory syndrome (SARS)-CoV, Middle East respiratory syndrome (MERS)-CoV, and SARS-CoV-2, have emerged in the past two decades. All of them encode nonstructural protein 1 (nsp1) in their genomes. Nsp1 of SARS-CoV and MERS-CoV exhibit common biological functions for inducing endonucleolytic cleavage of host mRNAs and inhibition of host translation, while viral mRNAs evade the nsp1-induced mRNA cleavage. SARS-CoV nsp1 is a major pathogenic determinant for this virus, supporting the notion that a viral protein that suppresses host gene expression can be a virulence factor, and further suggesting the possibility that SARS-CoV-2 nsp1, which has high amino acid identity with SARS-CoV nsp1, may serve as a major virulence factor. This review summarizes the gene expression suppression functions of nsp1 of CoVs, with a primary focus on SARS-CoV nsp1 and MERS-CoV nsp1.


Subject(s)
Betacoronavirus , Coronavirus Infections/virology , RNA-Dependent RNA Polymerase/physiology , Viral Nonstructural Proteins/physiology , Animals , Betacoronavirus/pathogenicity , Betacoronavirus/physiology , Gene Expression Regulation , Host Microbial Interactions , Humans , Mice , RNA, Messenger/genetics , Virus Replication
10.
J Med Virol ; 92(4): 418-423, 2020 04.
Article in English | MEDLINE | ID: covidwho-989

ABSTRACT

The recent emergence of a novel coronavirus (2019-nCoV), which is causing an outbreak of unusual viral pneumonia in patients in Wuhan, a central city in China, is another warning of the risk of CoVs posed to public health. In this minireview, we provide a brief introduction of the general features of CoVs and describe diseases caused by different CoVs in humans and animals. This review will help understand the biology and potential risk of CoVs that exist in richness in wildlife such as bats.


Subject(s)
Communicable Diseases, Emerging/virology , Coronavirus Infections/virology , Coronavirus/physiology , Coronavirus/pathogenicity , Animals , Betacoronavirus/pathogenicity , Betacoronavirus/physiology , COVID-19 , Coronavirus/genetics , Coronavirus Infections/veterinary , Genome, Viral , Host Specificity , Humans , Pneumonia, Viral/virology , SARS-CoV-2 , Viral Nonstructural Proteins/physiology , Viral Structural Proteins/physiology , Virus Replication
SELECTION OF CITATIONS
SEARCH DETAIL