Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
J Virol ; 97(1): e0161422, 2023 01 31.
Article in English | MEDLINE | ID: covidwho-2223572

ABSTRACT

Porcine epidemic diarrhea (PED) indicates the disease of the acute and highly contagious intestinal infection due to porcine epidemic diarrhea virus (PEDV), with the characteristics of watery diarrhea, vomiting, and dehydration. One of the reasons for diarrhea and death of piglets is PEDV, which leads to 100% mortality in neonatal piglets. Therefore, it is necessary to explore the interaction between virus and host to prevent and control PEDV. This study indicated that the host protein, pre-mRNA processing factor 19 (PRPF19), could be controlled by the signal transducer as well as activator of transcription 1 (STAT1). Thus, PEDV replication could be hindered through selective autophagy. Moreover, PRPF19 was found to recruit the E3 ubiquitin ligase MARCH8 to the N protein for ubiquitination. For the purpose of degradation, the ubiquitin N protein is acknowledged by the cargo receptor NDP52 and transported to autolysosomes, thus inhibiting virus proliferation. To conclude, a unique antiviral mechanism of PRPF19-mediated virus restriction was shown. Moreover, a view of the innate immune response and protein degradation against PEDV replication was provided in this study. IMPORTANCE The highly virulent porcine epidemic diarrhea virus (PEDV) emerged in 2010, and causes high mortality rates in newborn pigs. There are no effective and safe vaccines against the highly virulent PEDV. This virus has caused devastating economic losses in the pork industry worldwide. Studying the relationship between virus and host antiviral factors is important to develop the new antiviral strategies. This study identified the pre-mRNA processing factor 19 (PRPF19) as a novel antiviral protein in PEDV replication and revealed its viral restriction mechanisms for the first time. PRPF19 recruited the E3 ubiquitin ligase MARCH8 to the PEDV N protein for ubiquitination, and the ubiquitin N protein was acknowledged by the cargo receptor NDP52 and transported to autolysosomes for degradation. Our findings provide new insights in host antiviral factors PRPF19 that regulate the selective autophagy protein degradation pathway to inhibit PEDV replication.


Subject(s)
Capsid Proteins , Coronavirus Infections , Porcine epidemic diarrhea virus , Swine Diseases , Animals , Capsid Proteins/metabolism , Coronavirus Infections/immunology , Coronavirus Infections/veterinary , Coronavirus Infections/virology , Porcine epidemic diarrhea virus/physiology , Swine , Swine Diseases/immunology , Swine Diseases/virology , Ubiquitin-Protein Ligases/metabolism , Ubiquitins , Virus Replication/genetics , Nuclear Proteins/metabolism , Autophagy
2.
Sci Adv ; 9(2): eade0869, 2023 01 13.
Article in English | MEDLINE | ID: covidwho-2193381

ABSTRACT

Parvovirus B19 (B19V) infects human erythroid progenitor cells (EPCs) and causes several hematological disorders and fetal hydrops. Amino acid (aa) 5-68 of minor capsid protein VP1 (VP1u5-68aa) is the minimal receptor binding domain for B19V to enter EPCs. Here, we carried out a genome-wide CRISPR-Cas9 guide RNA screen and identified tyrosine protein kinase receptor UFO (AXL) as a proteinaceous receptor for B19V infection of EPCs. AXL gene silencing in ex vivo expanded EPCs remarkably decreased B19V internalization and replication. Additions of the recombinant AXL extracellular domain or a polyclonal antibody against it upon infection efficiently inhibited B19V infection of ex vivo expanded EPCs. Moreover, B19V VP1u interacted with the recombinant AXL extracellular domain in vitro at a relatively high affinity (KD = 103 nM). Collectively, we provide evidence that AXL is a co-receptor for B19V infection of EPCs.


Subject(s)
Axl Receptor Tyrosine Kinase , Erythema Infectiosum , Parvovirus B19, Human , Humans , Capsid Proteins/genetics , Capsid Proteins/metabolism , Erythema Infectiosum/metabolism , Parvovirus B19, Human/genetics , Parvovirus B19, Human/metabolism , Protein Binding , Axl Receptor Tyrosine Kinase/metabolism
3.
Int J Mol Sci ; 23(24)2022 Dec 10.
Article in English | MEDLINE | ID: covidwho-2155136

ABSTRACT

Capsid protein of Hepatitis E virus (HEV) is capable of self-assembly into virus-like particles (VLPs) when expressed in Nicotiana benthamiana plants. Such VLPs could be used as carriers of antigens for vaccine development. In this study, we obtained VLPs based on truncated coat protein of HEV bearing the M2e peptide of Influenza A virus or receptor-binding domain of SARS-CoV-2 spike glycoprotein (RBD). We optimized the immunogenic epitopes' presentation by inserting them into the protruding domain of HEV ORF2 at position Tyr485. The fusion proteins were expressed in Nicotiana benthamiana plants using self-replicating potato virus X (PVX)-based vector. The fusion protein HEV/M2, targeted to the cytosol, was expressed at the level of about 300-400 µg per gram of fresh leaf tissue and appeared to be soluble. The fusion protein was purified using metal affinity chromatography under native conditions with the final yield about 200 µg per gram of fresh leaf tissue. The fusion protein HEV/RBD, targeted to the endoplasmic reticulum, was expressed at about 80-100 µg per gram of fresh leaf tissue; the yield after purification was up to 20 µg per gram of fresh leaf tissue. The recombinant proteins HEV/M2 and HEV/RBD formed nanosized virus-like particles that could be recognized by antibodies against inserted epitopes. The ELISA assay showed that antibodies of COVID-19 patients can bind plant-produced HEV/RBD virus-like particles. This study shows that HEV capsid protein is a promising carrier for presentation of foreign antigen.


Subject(s)
Artificial Virus-Like Particles , Capsid Proteins , Hepatitis E virus , Humans , Capsid Proteins/metabolism , COVID-19 , Epitopes , Recombinant Proteins , SARS-CoV-2/metabolism , Tobacco , Antigen Presentation , Plants, Genetically Modified , Recombinant Fusion Proteins/biosynthesis
4.
Int J Biol Macromol ; 213: 1007-1017, 2022 Jul 31.
Article in English | MEDLINE | ID: covidwho-2121014

ABSTRACT

The COVID-19 pandemic has highlighted the need for new vaccine platforms to rapidly develop solutions against emerging pathogens. In particular, some plant viruses offer several advantages for developing subunit vaccines, such as high expression rates in E. coli, high immunogenicity and safety, and absence of pre-immunity that could interfere with the vaccine's efficacy. Cowpea chlorotic mottle virus (CCMV) is a model system that has been extensively characterized, with key advantages for its use as an epitope carrier. In the present study, three relevant epitopes from the SARS-CoV-2 Spike protein were genetically inserted into the CCMV CP and expressed in E. coli cultures, resulting in the CCMV1, CCMV2, and CCMV3 chimeras. The recombinant CP mutants were purified from the formed inclusion bodies and refolded, and their immunogenicity as a subunit vaccine was assessed in BALB/c mice. The three mutants are immunogenic as they induce high IgG antibody titers that recognize the recombinant full-length S protein. This study supports the application of CCMV CP as an attractive carrier for the clinical evaluation of vaccine candidates against SARS-CoV-2. Furthermore, it suggests that VLPs assembled from these chimeric proteins could result in antigens with better immunogenicity.


Subject(s)
Bromovirus , COVID-19 , Animals , Bromovirus/genetics , Bromovirus/metabolism , COVID-19/prevention & control , Capsid Proteins/genetics , Capsid Proteins/metabolism , Chimera/metabolism , Epitopes , Escherichia coli/metabolism , Humans , Mice , Pandemics , SARS-CoV-2/genetics , Spike Glycoprotein, Coronavirus , Vaccines, Subunit
5.
Viruses ; 14(10)2022 09 30.
Article in English | MEDLINE | ID: covidwho-2066551

ABSTRACT

Molecular therapies exploiting mRNA vectors embody enormous potential, as evidenced by the utility of this technology for the context of the COVID-19 pandemic. Nonetheless, broad implementation of these promising strategies has been restricted by the limited repertoires of delivery vehicles capable of mRNA transport. On this basis, we explored a strategy based on exploiting the well characterized entry biology of adenovirus. To this end, we studied an adenovirus-polylysine (AdpL) that embodied "piggyback" transport of the mRNA on the capsid exterior of adenovirus. We hypothesized that the efficient steps of Ad binding, receptor-mediated entry, and capsid-mediated endosome escape could provide an effective pathway for transport of mRNA to the cellular cytosol for transgene expression. Our studies confirmed that AdpL could mediate effective gene transfer of mRNA vectors in vitro and in vivo. Facets of this method may offer key utilities to actualize the promise of mRNA-based therapeutics.


Subject(s)
Adenoviridae Infections , COVID-19 , Humans , Adenoviridae/genetics , Genetic Vectors/genetics , Gene Transfer Techniques , Polylysine , RNA, Messenger/genetics , RNA, Messenger/metabolism , Pandemics , Capsid Proteins/genetics , Capsid Proteins/metabolism , Biology
6.
Toxins (Basel) ; 14(10)2022 10 04.
Article in English | MEDLINE | ID: covidwho-2066493

ABSTRACT

With the outbreak and spread of COVID-19, a deep investigation of SARS-CoV-2 is urgent. Direct usage of this virus for scientific research could provide reliable results and authenticity. However, it is strictly constrained and unrealistic due to its high pathogenicity and infectiousness. Considering its biosafety, different systems and technologies have been employed in immunology and biomedical studies. In this study, phage display technology was used to construct a nonpathogenic model for COVID-19 research. The nucleocapsid protein of SARS-CoV-2 was fused with the M13 phage capsid p3 protein and expressed on the M13 phages. After validation of its successful expression, its potential as the standard for qPCR quantification and affinity with antibodies were confirmed, which may show the possibility of using this nonpathogenic bacteriophage to replace the pathogenic virus in scientific research concerning SARS-CoV-2. In addition, the model was used to develop a system for the classification and identification of different samples using ATR-FTIR, which may provide an idea for the development and evaluation of virus monitoring equipment in the future.


Subject(s)
COVID-19 , Viruses , Humans , SARS-CoV-2/genetics , Cell Surface Display Techniques , Bacteriophage M13/genetics , Bacteriophage M13/metabolism , Capsid Proteins/genetics , Capsid Proteins/metabolism , Nucleocapsid Proteins/genetics , Nucleocapsid Proteins/metabolism
7.
J Virol ; 96(17): e0107422, 2022 09 14.
Article in English | MEDLINE | ID: covidwho-2038239

ABSTRACT

Rotavirus (RV) viroplasms are cytosolic inclusions where both virus genome replication and primary steps of virus progeny assembly take place. A stabilized microtubule cytoskeleton and lipid droplets are required for the viroplasm formation, which involves several virus proteins. The viral spike protein VP4 has not previously been shown to have a direct role in viroplasm formation. However, it is involved with virus-cell attachment, endocytic internalization, and virion morphogenesis. Moreover, VP4 interacts with actin cytoskeleton components, mainly in processes involving virus entrance and egress, and thereby may have an indirect role in viroplasm formation. In this study, we used reverse genetics to construct a recombinant RV, rRV/VP4-BAP, that contains a biotin acceptor peptide (BAP) in the K145-G150 loop of the VP4 lectin domain, permitting live monitoring. The recombinant virus was replication competent but showed a reduced fitness. We demonstrate that rRV/VP4-BAP infection, as opposed to rRV/wt infection, did not lead to a reorganized actin cytoskeleton as viroplasms formed were insensitive to drugs that depolymerize actin and inhibit myosin. Moreover, wild-type (wt) VP4, but not VP4-BAP, appeared to associate with actin filaments. Similarly, VP4 in coexpression with NSP5 and NSP2 induced a significant increase in the number of viroplasm-like structures. Interestingly, a small peptide mimicking loop K145-G150 rescued the phenotype of rRV/VP4-BAP by increasing its ability to form viroplasms and hence improve virus progeny formation. Collectively, these results provide a direct link between VP4 and the actin cytoskeleton to catalyze viroplasm assembly. IMPORTANCE The spike protein VP4 participates in diverse steps of the rotavirus (RV) life cycle, including virus-cell attachment, internalization, modulation of endocytosis, virion morphogenesis, and virus egress. Using reverse genetics, we constructed for the first time a recombinant RV, rRV/VP4-BAP, harboring a heterologous peptide in the lectin domain (loop K145-G150) of VP4. The rRV/VP4-BAP was replication competent but with reduced fitness due to a defect in the ability to reorganize the actin cytoskeleton, which affected the efficiency of viroplasm assembly. This defect was rescued by adding a permeable small-peptide mimicking the wild-type VP4 loop K145-G150. In addition to revealing a new role of VP4, our findings suggest that rRV harboring an engineered VP4 could be used as a new dual vaccination platform providing immunity against RV and additional heterologous antigens.


Subject(s)
Actin Cytoskeleton , Capsid Proteins , Rotavirus , Actin Cytoskeleton/metabolism , Capsid Proteins/metabolism , Humans , Lectins , Reverse Genetics , Rotavirus/genetics , Rotavirus/physiology , Rotavirus Infections , Viral Replication Compartments , Virus Replication
8.
Cell ; 184(25): 6037-6051.e14, 2021 12 09.
Article in English | MEDLINE | ID: covidwho-1520752

ABSTRACT

RNA viruses generate defective viral genomes (DVGs) that can interfere with replication of the parental wild-type virus. To examine their therapeutic potential, we created a DVG by deleting the capsid-coding region of poliovirus. Strikingly, intraperitoneal or intranasal administration of this genome, which we termed eTIP1, elicits an antiviral response, inhibits replication, and protects mice from several RNA viruses, including enteroviruses, influenza, and SARS-CoV-2. While eTIP1 replication following intranasal administration is limited to the nasal cavity, its antiviral action extends non-cell-autonomously to the lungs. eTIP1 broad-spectrum antiviral effects are mediated by both local and distal type I interferon responses. Importantly, while a single eTIP1 dose protects animals from SARS-CoV-2 infection, it also stimulates production of SARS-CoV-2 neutralizing antibodies that afford long-lasting protection from SARS-CoV-2 reinfection. Thus, eTIP1 is a safe and effective broad-spectrum antiviral generating short- and long-term protection against SARS-CoV-2 and other respiratory infections in animal models.


Subject(s)
Capsid Proteins/genetics , Defective Interfering Viruses/metabolism , Virus Replication/drug effects , Administration, Intranasal , Animals , Antiviral Agents/pharmacology , Broadly Neutralizing Antibodies/immunology , Broadly Neutralizing Antibodies/pharmacology , COVID-19 , Capsid Proteins/metabolism , Cell Line , Defective Interfering Viruses/pathogenicity , Disease Models, Animal , Genome, Viral/genetics , Humans , Influenza, Human , Interferons/metabolism , Male , Mice , Mice, Inbred C57BL , Poliovirus/genetics , Poliovirus/metabolism , Respiratory Tract Infections/virology , SARS-CoV-2/drug effects , SARS-CoV-2/pathogenicity
9.
J Phys Chem Lett ; 12(39): 9557-9563, 2021 Oct 07.
Article in English | MEDLINE | ID: covidwho-1439482

ABSTRACT

Lipid-enveloped viruses, such as Ebola, influenza, or coronaviruses, are a major threat to human health. Ethanol is an efficient disinfectant that is widely used to inactivate these viruses and prevent their transmission. However, the interactions between ethanol and enveloped viruses leading to their inactivation are not yet fully understood. This study demonstrates the link between ethanol-induced viral inactivation and the nanostructural and chemical transformations of the model virus Phi6, an 85 nm diameter lipid-enveloped bacterial virus that is commonly used as surrogate for human pathogenic viruses. The virus morphology was investigated using small-angle X-ray scattering and dynamic light scattering and was related to its infectivity. The Phi6's surface chemistry was characterized by cryogenic X-ray photoelectron spectroscopy, and the modifications in protein structure were assessed by circular dichroism and fluorescence spectroscopy. Ethanol-triggered structural modifications were found in the lipid envelope, detaching from the protein capsid and forming coexisting nanostructures.


Subject(s)
Bacteriophage phi 6/chemistry , Ethanol/pharmacology , Virus Inactivation/drug effects , Bacteriophage phi 6/drug effects , Bacteriophage phi 6/ultrastructure , Capsid Proteins/chemistry , Capsid Proteins/metabolism , Circular Dichroism , Dynamic Light Scattering , Ethanol/chemistry , Microscopy, Electron, Transmission , Photoelectron Spectroscopy , Scattering, Small Angle , X-Ray Diffraction
10.
Viruses ; 13(7)2021 06 24.
Article in English | MEDLINE | ID: covidwho-1389547

ABSTRACT

Adenovirus vector-based genetic vaccines have emerged as a powerful strategy against the SARS-CoV-2 health crisis. This success is not unexpected because adenoviruses combine many desirable features of a genetic vaccine. They are highly immunogenic and have a low and well characterized pathogenic profile paired with technological approachability. Ongoing efforts to improve adenovirus-vaccine vectors include the use of rare serotypes and non-human adenoviruses. In this review, we focus on the viral capsid and how the choice of genotypes influences the uptake and subsequent subcellular sorting. We describe how understanding capsid properties, such as stability during the entry process, can change the fate of the entering particles and how this translates into differences in immunity outcomes. We discuss in detail how mutating the membrane lytic capsid protein VI affects species C viruses' post-entry sorting and briefly discuss if such approaches could have a wider implication in vaccine and/or vector development.


Subject(s)
Adenoviruses, Human/immunology , Adenoviruses, Human/physiology , Capsid/metabolism , Genetic Vectors , Viral Vaccines/immunology , Virus Internalization , Adaptive Immunity , Adenoviruses, Human/genetics , Animals , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , COVID-19/immunology , Capsid/immunology , Capsid Proteins/genetics , Capsid Proteins/immunology , Capsid Proteins/metabolism , Clinical Trials as Topic , Humans , Immunity, Innate , Mice , SARS-CoV-2/immunology
11.
Ann Diagn Pathol ; 53: 151744, 2021 Aug.
Article in English | MEDLINE | ID: covidwho-1227970

ABSTRACT

OBJECTIVES: Assess the pathologic changes in the lungs of COVID-19 decedents and correlate these changes with demographic data, clinical course, therapies, and duration of illness. METHODS: Lungs of 12 consecutive COVID-19 decedents consented for autopsy were evaluated for gross and histopathologic abnormalities. A complete Ghon "en block" dissection was performed on all cases; lung weights and gross characteristics recorded. Immunohistochemical studies were performed to characterize lymphocytic infiltrates and to assess SARS-CoV-2 capsid protein. RESULTS: Two distinct patterns of pulmonary involvement were identified. Three of 12 cases demonstrated a predominance of acute alveolar damage (DAD) while 9 of 12 cases demonstrated a marked increase in intra-alveolar macrophages in a fashion resembling desquamative interstitial pneumonia or macrophage activation syndrome (DIP/MAS). Two patterns were correlated solely with a statistically significant difference in the duration of illness. The group exhibiting DAD had duration of illness of 5.7 days while the group with DIP/MAS had duration of illness of 21.5 days (t-test p = 0.014). CONCLUSIONS: The pulmonary pathology of COVID-19 patients demonstrates a biphasic pattern, an acute phase demonstrating DAD changes while the patients with a more prolonged course exhibit a different pattern that resembles DIP/MAS-like pattern. The potential mechanisms and clinical significance are discussed.


Subject(s)
COVID-19/pathology , Immunohistochemistry/methods , Lung Diseases, Interstitial/pathology , Lung/pathology , Macrophage Activation Syndrome/pathology , Adult , Aged , Aged, 80 and over , Autopsy , COVID-19/complications , COVID-19/diagnosis , COVID-19/virology , Capsid Proteins/metabolism , Comorbidity , Female , Humans , Lung/metabolism , Lung Diseases, Interstitial/etiology , Lung Diseases, Interstitial/virology , Lymphocytes/metabolism , Lymphocytes/pathology , Macrophage Activation Syndrome/etiology , Macrophage Activation Syndrome/virology , Macrophages/pathology , Male , Middle Aged , Pulmonary Alveoli/immunology , Pulmonary Alveoli/pathology , SARS-CoV-2/genetics , Sick Leave
12.
Ann Diagn Pathol ; 50: 151645, 2021 Feb.
Article in English | MEDLINE | ID: covidwho-1064802

ABSTRACT

The objective of this study was to elucidate the pathophysiology that underlies severe COVID-19 by assessing the histopathology and the in situ detection of infectious SARS-CoV-2 and viral capsid proteins along with the cellular target(s) and host response from twelve autopsies. There were three key findings: 1) high copy infectious virus was limited mostly to the alveolar macrophages and endothelial cells of the septal capillaries; 2) viral spike protein without viral RNA localized to ACE2+ endothelial cells in microvessels that were most abundant in the subcutaneous fat and brain; 3) although both infectious virus and docked viral spike protein was associated with complement activation, only the endocytosed pseudovirions induced a marked up-regulation of the key COVID-19 associated proteins IL6, TNF alpha, IL1 beta, p38, IL8, and caspase 3. Importantly, this microvasculitis was associated with characteristic findings on hematoxylin and eosin examination that included endothelial degeneration and resultant basement membrane zone disruption and reduplication. It is concluded that serious COVID-19 infection has two distinct mechanisms: 1) a microangiopathy of pulmonary capillaries associated with a high infectious viral load where endothelial cell death releases pseudovirions into the circulation, and 2) the pseudovirions dock on ACE2+ endothelial cells most prevalent in the skin/subcutaneous fat and brain that activates the complement pathway/coagulation cascade resulting in a systemic procoagulant state as well as the expression of cytokines that produce the cytokine storm. The data predicts a favorable response to therapies based on either removal of circulating viral proteins and/or blunting of the endothelial-induced response.


Subject(s)
COVID-19/physiopathology , Capsid Proteins/metabolism , SARS-CoV-2/pathogenicity , Spike Glycoprotein, Coronavirus/metabolism , Thrombotic Microangiopathies/physiopathology , Vascular Diseases/physiopathology , Adult , Aged , Aged, 80 and over , Angiotensin-Converting Enzyme 2/metabolism , Autopsy , COVID-19/virology , Capsid Proteins/genetics , Endothelial Cells/enzymology , Endothelial Cells/virology , Female , Humans , Lung/physiopathology , Lung/virology , Male , Microvessels/physiopathology , Microvessels/virology , Middle Aged , RNA, Viral/genetics , SARS-CoV-2/genetics , Spike Glycoprotein, Coronavirus/genetics , Thrombotic Microangiopathies/virology , Vascular Diseases/virology , Virion
13.
Viruses ; 13(1)2021 Jan 15.
Article in English | MEDLINE | ID: covidwho-1038679

ABSTRACT

Adenovirus (AdV) infection elicits a strong immune response with the production of neutralizing antibodies and opsonization by complement and coagulation factors. One anti-hexon neutralizing antibody, called 9C12, is known to activate the complement cascade, resulting in the deposition of complement component C4b on the capsid, and the neutralization of the virus. The mechanism of AdV neutralization by C4b is independent of downstream complement proteins and involves the blockage of the release of protein VI, which is required for viral escape from the endosome. To investigate the structural basis underlying how C4b blocks the uncoating of AdV, we built a model for the complex of human adenovirus type-5 (HAdV5) with 9C12, together with complement components C1 and C4b. This model positions C4b near the Arg-Gly-Asp (RGD) loops of the penton base. There are multiple amino acids in the RGD loop that might serve as covalent binding sites for the reactive thioester of C4b. Molecular dynamics simulations with a multimeric penton base and C4b indicated that stabilizing interactions may form between C4b and multiple RGD loops. We propose that C4b deposition on one RGD loop leads to the entanglement of C4b with additional RGD loops on the same penton base multimer and that this entanglement blocks AdV uncoating.


Subject(s)
Adenoviridae/immunology , Complement C4/chemistry , Complement C4/immunology , Models, Molecular , Antibodies, Neutralizing/chemistry , Antibodies, Neutralizing/immunology , Antibodies, Viral/chemistry , Antibodies, Viral/immunology , Binding Sites , Capsid/chemistry , Capsid/metabolism , Capsid/ultrastructure , Capsid Proteins/chemistry , Capsid Proteins/immunology , Capsid Proteins/metabolism , Capsid Proteins/ultrastructure , Humans , Immunoglobulin G/chemistry , Immunoglobulin G/immunology , Molecular Docking Simulation , Molecular Dynamics Simulation , Protein Binding , Protein Conformation , Structure-Activity Relationship
14.
Viruses ; 12(10)2020 10 09.
Article in English | MEDLINE | ID: covidwho-906169

ABSTRACT

Superimposition of protein structures is key in unravelling structural homology across proteins whose sequence similarity is lost. Structural comparison provides insights into protein function and evolution. Here, we review some of the original findings and thoughts that have led to the current established structure-based phylogeny of viruses: starting from the original observation that the major capsid proteins of plant and animal viruses possess similar folds, to the idea that each virus has an innate "self". This latter idea fueled the conceptualization of the PRD1-adenovirus lineage whose members possess a major capsid protein (innate "self") with a double jelly roll fold. Based on this approach, long-range viral evolutionary relationships can be detected allowing the virosphere to be classified in four structure-based lineages. However, this process is not without its challenges or limitations. As an example of these hurdles, we finally touch on the difficulty of establishing structural "self" traits for enveloped viruses showcasing the coronaviruses but also the power of structure-based analysis in the understanding of emerging viruses.


Subject(s)
Adenoviridae/metabolism , Capsid Proteins/metabolism , Coronavirus/metabolism , Protein Structure, Tertiary/physiology , Rhinovirus/metabolism , Adenoviridae/genetics , Coronavirus/genetics , Crystallography, X-Ray , Genome, Viral/genetics , Rhinovirus/genetics , Viral Structures/metabolism
15.
Cell Rep ; 33(1): 108234, 2020 10 06.
Article in English | MEDLINE | ID: covidwho-778591

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) replication and host immune response determine coronavirus disease 2019 (COVID-19), but studies evaluating viral evasion of immune response are lacking. Here, we use unbiased screening to identify SARS-CoV-2 proteins that antagonize type I interferon (IFN-I) response. We found three proteins that antagonize IFN-I production via distinct mechanisms: nonstructural protein 6 (nsp6) binds TANK binding kinase 1 (TBK1) to suppress interferon regulatory factor 3 (IRF3) phosphorylation, nsp13 binds and blocks TBK1 phosphorylation, and open reading frame 6 (ORF6) binds importin Karyopherin α 2 (KPNA2) to inhibit IRF3 nuclear translocation. We identify two sets of viral proteins that antagonize IFN-I signaling through blocking signal transducer and activator of transcription 1 (STAT1)/STAT2 phosphorylation or nuclear translocation. Remarkably, SARS-CoV-2 nsp1 and nsp6 suppress IFN-I signaling more efficiently than SARS-CoV and Middle East respiratory syndrome coronavirus (MERS-CoV). Thus, when treated with IFN-I, a SARS-CoV-2 replicon replicates to a higher level than chimeric replicons containing nsp1 or nsp6 from SARS-CoV or MERS-CoV. Altogether, the study provides insights on SARS-CoV-2 evasion of IFN-I response and its potential impact on viral transmission and pathogenesis.


Subject(s)
Capsid Proteins/metabolism , Coronavirus Infections/immunology , Immune Evasion , Interferon Type I/metabolism , Methyltransferases/metabolism , Pneumonia, Viral/immunology , RNA Helicases/metabolism , Viral Nonstructural Proteins/metabolism , Viral Proteins/metabolism , A549 Cells , Animals , Betacoronavirus/immunology , Betacoronavirus/pathogenicity , COVID-19 , Coronavirus Infections/virology , Coronavirus Nucleocapsid Proteins , Cricetinae , Cricetulus , HEK293 Cells , Humans , Interferon Regulatory Factor-3/metabolism , Interferon Type I/genetics , Pandemics , Pneumonia, Viral/virology , Protein Binding , Protein Serine-Threonine Kinases/metabolism , SARS-CoV-2 , STAT Transcription Factors/metabolism , alpha Karyopherins/metabolism
16.
Cells ; 9(7)2020 07 05.
Article in English | MEDLINE | ID: covidwho-636152

ABSTRACT

The SARS-CoV-2 pandemic necessitates a review of the molecular mechanisms underlying cellular infection by coronaviruses, in order to identify potential therapeutic targets against the associated new disease (COVID-19). Previous studies on its counterparts prove a complex and concomitant interaction between coronaviruses and autophagy. The precise manipulation of this pathway allows these viruses to exploit the autophagy molecular machinery while avoiding its protective apoptotic drift and cellular innate immune responses. In turn, the maneuverability margins of such hijacking appear to be so narrow that the modulation of the autophagy, regardless of whether using inducers or inhibitors (many of which are FDA-approved for the treatment of other diseases), is usually detrimental to viral replication, including SARS-CoV-2. Recent discoveries indicate that these interactions stretch into the still poorly explored noncanonical autophagy pathway, which might play a substantial role in coronavirus replication. Still, some potential therapeutic targets within this pathway, such as RAB9 and its interacting proteins, look promising considering current knowledge. Thus, the combinatory treatment of COVID-19 with drugs affecting both canonical and noncanonical autophagy pathways may be a turning point in the fight against this and other viral infections, which may also imply beneficial prospects of long-term protection.


Subject(s)
Autophagy , Coronavirus Infections/pathology , Pneumonia, Viral/pathology , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Apoptosis , Autophagy/drug effects , Autophagy-Related Proteins/antagonists & inhibitors , Autophagy-Related Proteins/metabolism , Betacoronavirus/classification , Betacoronavirus/physiology , COVID-19 , Capsid Proteins/metabolism , Coronavirus Infections/drug therapy , Coronavirus Infections/virology , Coronavirus Nucleocapsid Proteins , Humans , Pandemics , Pneumonia, Viral/drug therapy , Pneumonia, Viral/virology , SARS-CoV-2 , Virus Replication/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL