Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 84
Filter
1.
J Virol ; 98(6): e0049424, 2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38757985

ABSTRACT

Mitochondria are energy producers in cells, which can affect viral replication by regulating the host innate immune signaling pathways, and the changes in their biological functions are inextricably linked the viral life cycle. In this study, we screened a library of 382 mitochondria-targeted compounds and identified the antiviral inhibitors of dihydroorotate dehydrogenase (DHODH), the rate-limiting enzyme in the de novo synthesis pathway of pyrimidine ribonucleotides, against classical swine fever virus (CSFV). Our data showed that the inhibitors interfered with viral RNA synthesis in a dose-dependent manner, with half-maximal effective concentrations (EC50) ranging from 0.975 to 26.635 nM. Remarkably, DHODH inhibitors obstructed CSFV replication by enhancing the innate immune response including the TBK1-IRF3-STAT1 and NF-κB signaling pathways. Furthermore, the data from a series of compound addition and supplementation trials indicated that DHODH inhibitors also inhibited CSFV replication by blocking the de novo pyrimidine synthesis. Remarkably, DHODH knockdown demonstrated that it was essential for CSFV replication. Mechanistically, confocal microscopy and immunoprecipitation assays showed that the non-structural protein 4A (NS4A) recruited and interacted with DHODH in the perinuclear. Notably, NS4A enhanced the DHODH activity and promoted the generation of UMP for efficient viral replication. Structurally, the amino acids 65-229 of DHODH and the amino acids 25-40 of NS4A were pivotal for this interaction. Taken together, our findings highlight the critical role of DHODH in the CSFV life cycle and offer a potential antiviral target for the development of novel therapeutics against CSF. IMPORTANCE: Classical swine fever remains one of the most economically important viral diseases of domestic pigs and wild boar worldwide. dihydroorotate dehydrogenase (DHODH) inhibitors have been shown to suppress the replication of several viruses in vitro and in vivo, but the effects on Pestivirus remain unknown. In this study, three specific DHODH inhibitors, including DHODH-IN-16, BAY-2402234, and Brequinar were found to strongly suppress classical swine fever virus (CSFV) replication. These inhibitors target the host DHODH, depleting the pyrimidine nucleotide pool to exert their antiviral effects. Intriguingly, we observed that the non-structural protein 4A of CSFV induced DHODH to accumulate around the nucleus in conjunction with mitochondria. Moreover, NS4A exhibited a strong interaction with DHODH, enhancing its activity to promote efficient CSFV replication. In conclusion, our findings enhance the understanding of the pyrimidine synthesis in CSFV infection and expand the novel functions of CSFV NS4A in viral replication, providing a reference for further exploration of antiviral targets against CSFV.


Subject(s)
Antiviral Agents , Classical Swine Fever Virus , Dihydroorotate Dehydrogenase , Viral Nonstructural Proteins , Virus Replication , Animals , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Cell Line , Classical Swine Fever/drug therapy , Classical Swine Fever/immunology , Classical Swine Fever/metabolism , Classical Swine Fever/virology , Classical Swine Fever Virus/drug effects , Classical Swine Fever Virus/growth & development , Classical Swine Fever Virus/immunology , Classical Swine Fever Virus/metabolism , Dihydroorotate Dehydrogenase/metabolism , Dose-Response Relationship, Drug , Immunity, Innate/drug effects , Immunity, Innate/immunology , Immunoprecipitation , Microscopy, Confocal , Mitochondria/enzymology , Mitochondria/metabolism , RNA, Viral/biosynthesis , Signal Transduction/drug effects , Swine/virology , Viral Nonstructural Proteins/metabolism , Virus Replication/drug effects
2.
J Virol ; 97(10): e0111523, 2023 10 31.
Article in English | MEDLINE | ID: mdl-37796122

ABSTRACT

IMPORTANCE: Of the flaviviruses, only CSFV and bovine viral diarrhea virus express Npro as the non-structural protein which is not essential for viral replication but functions to dampen host innate immunity. We have deciphered a novel mechanism with which CSFV uses to evade the host antiviral immunity by the N-terminal domain of its Npro to facilitate proteasomal degradation of Sp1 with subsequent reduction of HDAC1 and ISG15 expression. This is distinct from earlier findings involving Npro-mediated IRF3 degradation via the C-terminal domain. This study provides insights for further studies on how HDAC1 plays its role in antiviral immunity, and if and how other viral proteins, such as the core protein of CSFV, the nucleocapsid protein of porcine epidemic diarrhea virus, or even other coronaviruses, exert antiviral immune responses via the Sp1-HDAC1 axis. Such research may lead to a deeper understanding of viral immune evasion strategies as part of their pathogenetic mechanisms.


Subject(s)
Classical Swine Fever Virus , Classical Swine Fever , Endopeptidases , Histone Deacetylase 1 , Immunity, Innate , Proteasome Endopeptidase Complex , Sp1 Transcription Factor , Viral Proteins , Animals , Classical Swine Fever/immunology , Classical Swine Fever/metabolism , Classical Swine Fever/virology , Classical Swine Fever Virus/enzymology , Classical Swine Fever Virus/immunology , Classical Swine Fever Virus/metabolism , Classical Swine Fever Virus/pathogenicity , Endopeptidases/chemistry , Endopeptidases/metabolism , Histone Deacetylase 1/biosynthesis , Histone Deacetylase 1/metabolism , Interferon Regulatory Factor-3 , Nucleocapsid Proteins/metabolism , Proteasome Endopeptidase Complex/metabolism , Sp1 Transcription Factor/metabolism , Swine/virology , Viral Core Proteins/metabolism , Viral Proteins/chemistry , Viral Proteins/metabolism , Ubiquitins/metabolism , Cytokines/metabolism , Porcine epidemic diarrhea virus/immunology , Porcine epidemic diarrhea virus/metabolism , Protein Domains
3.
PLoS Pathog ; 18(2): e1010294, 2022 02.
Article in English | MEDLINE | ID: mdl-35120190

ABSTRACT

As the important molecular machinery for membrane protein sorting in eukaryotic cells, the endosomal sorting and transport complexes (ESCRT-0/I/II/III and VPS4) usually participate in various replication stages of enveloped viruses, such as endocytosis and budding. The main subunit of ESCRT-I, Tsg101, has been previously revealed to play a role in the entry and replication of classical swine fever virus (CSFV). However, the effect of the whole ESCRT machinery during CSFV infection has not yet been well defined. Here, we systematically determine the effects of subunits of ESCRT on entry, replication, and budding of CSFV by genetic analysis. We show that EAP20 (VPS25) (ESCRT-II), CHMP4B and CHMP7 (ESCRT-III) regulate CSFV entry and assist vesicles in transporting CSFV from Clathrin, early endosomes, late endosomes to lysosomes. Importantly, we first demonstrate that HRS (ESCRT-0), VPS28 (ESCRT-I), VPS25 (ESCRT-II) and adaptor protein ALIX play important roles in the formation of virus replication complexes (VRC) together with CHMP2B/4B/7 (ESCRT-III), and VPS4A. Further analyses reveal these subunits interact with CSFV nonstructural proteins (NS) and locate in the endoplasmic reticulum, but not Golgi, suggesting the role of ESCRT in regulating VRC assembly. In addition, we demonstrate that VPS4A is close to lipid droplets (LDs), indicating the importance of lipid metabolism in the formation of VRC and nucleic acid production. Altogether, we draw a new picture of cellular ESCRT machinery in CSFV entry and VRC formation, which could provide alternative strategies for preventing and controlling the diseases caused by CSFV or other Pestivirus.


Subject(s)
Classical Swine Fever Virus/metabolism , Classical Swine Fever/virology , Endosomal Sorting Complexes Required for Transport/metabolism , RNA/metabolism , Viral Nonstructural Proteins/metabolism , Animals , Cell Line , Classical Swine Fever Virus/genetics , Clathrin/metabolism , Endoplasmic Reticulum/metabolism , Host Microbial Interactions , Swine , Transport Vesicles , Virus Internalization , Virus Replication
4.
J Virol ; 96(2): e0176821, 2022 01 26.
Article in English | MEDLINE | ID: mdl-34730400

ABSTRACT

Classical swine fever (CSF) is an economically important disease of pigs caused by classical swine fever virus (CSFV). The live attenuated vaccine C-strain (also called HCLV strain) against CSF was produced by multiple passages of a highly virulent strain in rabbits. However, the molecular determinants for its attenuation and protection remain unclear. In this study, we identified a unique glycosylation at position 986 (986NYT988) on the E2 glycoprotein Domain IV of C-strain but not (986NYA988) the highly virulent CSFV Shimen strain. We evaluated the infectivity, virulence, and protective efficacy of the C-strain-based mutant rHCLV-T988A lacking the glycosylation and Shimen strain mutant rShimen-A988T acquiring an additional glycosylation at position 986. rShimen-A988T showed a significantly decreased viral replication ability in SK6 cells, while rHCLV-T988A exhibited a growth kinetics indistinguishable from that of C-strain. Removal of the C-strain glycosylation site does not affect viral replication in rabbits and the attenuated phenotype in pigs. However, rShimen-A988T was attenuated and protected the pigs from a lethal challenge at 14 days postinoculation. In contrast, the rHCLV-T988A-inoculated pigs showed transient fever, a few clinical signs, and pathological changes in the spleens upon challenge with the Shimen strain. Mechanistic investigations revealed that the unique glycosylation at position 986 influences viral spreading, alters the formation of E2 homodimers, and leads to increased production of neutralizing antibodies. Collectively, our data for the first time demonstrate that the unique glycosylation at position 986 on the E2 glycoprotein is responsible for viral attenuation and protection. IMPORTANCE Viral glycoproteins involve in infectivity, virulence, and host immune responses. Deglycosylation on the Erns, E1, or E2 glycoprotein of highly virulent classical swine fever virus (CSFV) attenuated viral virulence in pigs, indicating that the glycosylation contributes to the pathogenicity of the highly virulent strain. However, the effects of the glycosylation on the C-strain E2 glycoprotein on viral infectivity in cells, viral attenuation, and protection in pigs have not been elucidated. This study demonstrates the unique glycosylation at position 986 on the C-strain E2 glycoprotein. C-strain mutant removing the glycosylation at the site provides only partial protection against CSFV challenge. Remarkably, the addition of the glycan to E2 of the highly virulent Shimen strain attenuates the viral virulence and confers complete protection against the lethal challenge in pigs. Our findings provide a new insight into the contribution of the glycosylation to the virus attenuation and protection.


Subject(s)
Classical Swine Fever Virus/immunology , Classical Swine Fever Virus/pathogenicity , Classical Swine Fever/prevention & control , Viral Envelope Proteins/metabolism , Viral Vaccines/immunology , Animals , Antibodies, Neutralizing/blood , Classical Swine Fever/virology , Classical Swine Fever Virus/genetics , Classical Swine Fever Virus/metabolism , Glycosylation , Immunization/veterinary , Mutation , Protein Multimerization , Rabbits , Swine , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/genetics , Vaccines, Attenuated/immunology , Vaccines, Attenuated/metabolism , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/genetics , Viral Envelope Proteins/immunology , Viral Vaccines/administration & dosage , Viral Vaccines/genetics , Viral Vaccines/metabolism , Virulence , Virus Replication
5.
PLoS Pathog ; 17(3): e1009393, 2021 03.
Article in English | MEDLINE | ID: mdl-33684175

ABSTRACT

Classical swine fever virus (CSFV) is an important pathogen in the swine industry. Virion attachment is mediated by envelope proteins Erns and E2, and E2 is indispensable. Using a pull-down assay with soluble E2 as the bait, we demonstrated that ADAM17, a disintegrin and metalloproteinase 17, is essential for CSFV entry. Loss of ADAM17 in a permissive cell line eliminated E2 binding and viral entry, but compensation with pig ADAM17 cDNA completely rescued these phenotypes. Similarly, ADAM17 silencing in primary porcine fibroblasts significantly impaired virus infection. In addition, human and mouse ADAM17, which is highly homologous to pig ADAM17, also mediated CSFV entry. The metalloproteinase domain of ADAM17 bound directly to E2 protein in a zinc-dependent manner. A surface exposed region within this domain was mapped and shown to be critical for CSFV entry. These findings clearly demonstrate that ADAM17 serves as an essential attachment factor for CSFV.


Subject(s)
ADAM17 Protein/metabolism , Classical Swine Fever Virus/metabolism , Receptors, Virus/metabolism , Viral Envelope Proteins/metabolism , Virus Internalization , Animals , Classical Swine Fever , Classical Swine Fever Virus/pathogenicity , Humans , Swine
6.
J Virol ; 95(12)2021 05 24.
Article in English | MEDLINE | ID: mdl-33827941

ABSTRACT

The classical swine fever virus (CSFV) glycoprotein E2 is the major structural component of the virus particle. E2 is involved in several functions, such as virus adsorption to the cell, the elicitation of protective immune responses, and virus virulence in swine. Using a yeast two-hybrid system, we previously identified the swine host protein Torsin-1A, an ATPase protein residing in the endoplasmic reticulum and inner nucleus membrane of the cell, as a specific binding partner for E2. The interaction between Torsin-1A and E2 proteins was confirmed to occur in CSFV-infected swine cells using three independent methods: coimmunoprecipitation, confocal microscopy, and proximity ligation assay (PLA). Furthermore, the E2 residue critical to mediate the protein-protein interaction with Torsin-1A was identified by a reverse yeast two-hybrid assay using a randomly mutated E2 library. A recombinant CSFV E2 mutant protein with a Q316L substitution failed to bind swine Torsin-1A in the yeast two-hybrid model. In addition, a CSFV infectious clone harboring the E2 Q316L substitution, although expressing substantial levels of E2 protein, repetitively failed to produce virus progeny when the corresponding RNA was transfected into susceptible SK6 cells. Importantly, PLA analysis of the transfected cells demonstrated an abolishment of the interaction between E2 Q316L and Torsin-1A, indicating a critical role for that interaction during CSFV replication.IMPORTANCE Structural glycoprotein E2 is an important structural component of the CSFV particle. E2 is involved in several virus functions, particularly virus-host interactions. Here, we characterized the interaction between CSFV E2 and swine protein Torsin-1A during virus infection. The critical amino acid residue in E2 mediating the interaction with Torsin-1A was identified and the effect of disrupting the E2-Torsin-1A protein-protein interaction was studied using reverse genetics. It is shown that the amino acid substitution abrogating E2-Torsin-1A interaction constitutes a lethal mutation, demonstrating that this virus-host protein-protein interaction is a critical factor during CSFV replication. This highlights the potential importance of the E2-Torsin-1A protein-protein interaction during CSFV replication and provides a potential pathway toward blocking virus replication, an important step toward the potential development of novel virus countermeasures.


Subject(s)
Classical Swine Fever Virus/physiology , Molecular Chaperones/metabolism , Viral Envelope Proteins/metabolism , Amino Acid Substitution , Animals , Cell Line , Classical Swine Fever Virus/metabolism , Host-Pathogen Interactions , Molecular Chaperones/genetics , Mutation , Protein Binding , Recombinant Proteins/metabolism , Swine , Two-Hybrid System Techniques , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/genetics , Virus Replication
7.
J Virol ; 93(18)2019 09 15.
Article in English | MEDLINE | ID: mdl-31292243

ABSTRACT

For members of the Flaviviridae, it is known that, besides the structural proteins, nonstructural (NS) proteins also play a critical role in virion formation. Pestiviruses, such as bovine viral diarrhea virus (BVDV), rely on uncleaved NS2-3 for virion formation, while its cleavage product, NS3, is selectively active in RNA replication. This dogma was recently challenged by the selection of gain-of-function mutations in NS2 and NS3 which allowed virion formation in the absence of uncleaved NS2-3 in BVDV type 1 (BVDV-1) variants encoding either a ubiquitin (Ubi) (NS2-Ubi-NS3) or an internal ribosome entry site (IRES) (NS2-IRES-NS3) between NS2 and NS3. To determine whether the ability to adapt to NS2-3-independent virion morphogenesis is conserved among pestiviruses, we studied the corresponding NS2 and NS3 mutations (2/T444-V and 3/M132-A) in classical swine fever virus (CSFV). We observed that these mutations were capable of restoring low-level NS2-3-independent virion formation only for CSFV NS2-Ubi-NS3. Interestingly, a second NS2 mutation (V439-D), identified by selection, was essential for high-titer virion production. Similar to previous findings for BVDV-1, these mutations in NS2 and NS3 allowed for low-titer virion production only in CSFV NS2-IRES-NS3. For efficient virion morphogenesis, additional exchanges in NS4A (A48-T) and NS5B (D280-G) were required, indicating that these proteins cooperate in NS2-3-independent virion formation. Interestingly, both NS5B mutations, selected independently for NS2-IRES-NS3 variants of BVDV-1 and CSFV, are located in the fingertip region of the viral RNA-dependent RNA polymerase, classifying this structural element as a novel determinant for pestiviral NS2-3-independent virion formation. Together, these findings will stimulate further mechanistic studies on the genome packaging of pestiviruses.IMPORTANCE For Flaviviridae members, the nonstructural proteins are essential for virion formation and thus exert a dual role in RNA replication and virion morphogenesis. However, it remains unclear how these proteins are functionalized for either process. In wild-type pestiviruses, the NS3/4A complex is selectively active in RNA replication, while NS2-3/4A is essential for virion formation. Mutations recently identified in BVDV-1 rendered NS3/4A capable of supporting NS2-3-independent virion morphogenesis. A comparison of NS3/4A complexes incapable/capable of supporting virion morphogenesis revealed that changes in NS3/NS4A surface interactions are decisive for the gain of function. However, so far, the role of the NS2 mutations as well as the accessory mutations additionally required in the NS2-IRES-NS3 virus variant has not been clarified. To unravel the course of genome packaging, the additional sets of mutations obtained for a second pestivirus species (CSFV) are of significant importance to develop mechanistic models for this complex process.


Subject(s)
Classical Swine Fever Virus/physiology , Cysteine Endopeptidases/metabolism , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/metabolism , Amino Acid Sequence , Animals , Cell Line , Classical Swine Fever/virology , Classical Swine Fever Virus/genetics , Classical Swine Fever Virus/metabolism , Cysteine Endopeptidases/genetics , Pestivirus/genetics , Pestivirus/metabolism , RNA Helicases/metabolism , RNA, Viral/genetics , Swine , Virion/genetics , Virion/metabolism , Virus Assembly , Virus Replication
8.
J Virol ; 94(1)2019 12 12.
Article in English | MEDLINE | ID: mdl-31597779

ABSTRACT

The E2 protein in classical swine fever (CSF) virus (CSFV) is the major virus structural glycoprotein and is an essential component of the viral particle. E2 has been shown to be involved in several functions, including virus adsorption, induction of protective immunity, and virulence in swine. Using the yeast two-hybrid system, we previously identified a swine host protein, dynactin subunit 6 (DCTN6) (a component of the cell dynactin complex), as a specific binding partner for E2. We confirmed the interaction between DCTN6 and E2 proteins in CSFV-infected swine cells by using two additional independent methodologies, i.e., coimmunoprecipitation and proximity ligation assays. E2 residues critical for mediating the protein-protein interaction with DCTN6 were mapped by a reverse yeast two-hybrid approach using a randomly mutated E2 library. A recombinant CSFV mutant, E2ΔDCTN6v, harboring specific substitutions in those critical residues was developed to assess the importance of the E2-DCTN6 protein-protein interaction for virus replication and virulence in swine. CSFV E2ΔDCTN6v showed reduced replication, compared with the parental virus, in an established swine cell line (SK6) and in primary swine macrophage cultures. Remarkably, animals infected with CSFV E2ΔDCTN6v remained clinically normal during the 21-day observation period, which suggests that the ability of CSFV E2 to bind host DCTN6 protein efficiently during infection may play a role in viral virulence.IMPORTANCE Structural glycoprotein E2 is an important component of CSFV due to its involvement in many virus activities, particularly virus-host interactions. Here, we present the description and characterization of the protein-protein interaction between E2 and the swine host protein DCTN6 during virus infection. The E2 amino acid residues mediating the interaction with DCTN6 were also identified. A recombinant CSFV harboring mutations disrupting the E2-DCTN6 interaction was created. The effect of disrupting the E2-DCTN6 protein-protein interaction was studied using reverse genetics. It was shown that the same amino acid substitutions that abrogated the E2-DCTN6 interaction in vitro constituted a critical factor in viral virulence in the natural host, domestic swine. This highlights the potential importance of the E2-DCTN6 protein-protein interaction in CSFV virulence and provides possible mechanisms of virus attenuation for the development of improved CSF vaccines.


Subject(s)
Classical Swine Fever Virus/genetics , Classical Swine Fever/virology , Dynactin Complex/genetics , Gene Expression Regulation , Host-Pathogen Interactions/genetics , Viral Envelope Proteins/genetics , Animals , Binding Sites , Cell Line , Classical Swine Fever/mortality , Classical Swine Fever/pathology , Classical Swine Fever Virus/metabolism , Classical Swine Fever Virus/pathogenicity , Dynactin Complex/metabolism , Epithelial Cells/metabolism , Epithelial Cells/virology , Gene Library , Macrophages/metabolism , Macrophages/virology , Mutation , Primary Cell Culture , Protein Binding , Signal Transduction , Survival Analysis , Swine , Two-Hybrid System Techniques , Viral Envelope Proteins/metabolism , Virus Replication
9.
Protein Expr Purif ; 167: 105527, 2020 03.
Article in English | MEDLINE | ID: mdl-31678666

ABSTRACT

Precaution of classical swine fever (CSF) is an important mission for the worldwide swine industry. Glycoprotein E2 is the leading antigen candidate for subunit vaccine of classical swine fever virus (CSFV). In this study, two Spy-tagged E2 genes were synthesized in vitro and subcloned into pMCO-AOX vector for intracellular expression in Pichia pastoris after methanol induction. Western blot analysis and semi-quantitative analysis showed that the yield of recombinant E2 protein was improved 17.87 folds by using co-translocational signal peptide cSIG. After the construction of the tandem multiple copy expression vectors, further increase of E2 production was observed by repetitive transforming expression vectors into P. pastoris genome. Finally, the yeast transformants harboring 8 or 16 copies of cSIG-E2-Spy increased the E2 expression level by 27.01-fold or 30.72-fold, respectively. These results demonstrate that utilizing co-translocational signal peptide together with multi-copy integration strategy can increase the production of recombinant E2 protein efficiently.


Subject(s)
Cloning, Molecular/methods , Viral Envelope Proteins , Animals , Classical Swine Fever Virus/metabolism , Mice , Protein Sorting Signals/genetics , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Saccharomycetales/genetics , Swine , Viral Envelope Proteins/biosynthesis , Viral Envelope Proteins/genetics
10.
Protein Expr Purif ; 167: 105526, 2020 03.
Article in English | MEDLINE | ID: mdl-31689499

ABSTRACT

The E2 envelope protein is the main protective antigen of classical swine fever virus (CSFV). Importantly, gram-positive enhancer matrix (GEM) particles can work as an immunostimulant and/or carrier system to improve the immune effect of antigens. In this study, the artificially designed E2-Spy was expressed and glycosylated in Pichia pastoris, and subsequently conjugated with SpyCatcher-PA which was expressed in Escherichia coli. The conjugated E2-Spy-PA was displayed on the surface of GEM particles, generating the E2-Spy-PA-GEM complex. Blocking ELISA analysis and neutralization assays showed that both E2-Spy and E2-Spy-PA-GEM complexes induced high levels of anti-CSFV antibodies in mice. Furthermore, statistical analyses indicated that the E2-Spy-PA-GEM complex exhibited enhanced immunogenicity compared with E2-Spy alone.


Subject(s)
Antibodies, Viral/immunology , Classical Swine Fever Virus/immunology , Viral Envelope Proteins , Adaptive Immunity , Animals , Classical Swine Fever Virus/metabolism , Cloning, Molecular , Enzyme-Linked Immunosorbent Assay/methods , Escherichia coli/genetics , Mice , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Saccharomycetales/genetics , Swine , Viral Envelope Proteins/biosynthesis , Viral Envelope Proteins/genetics , Viral Envelope Proteins/immunology
11.
Nucleic Acids Res ; 46(20): 10840-10854, 2018 11 16.
Article in English | MEDLINE | ID: mdl-30239956

ABSTRACT

Typically not assisted by proofreading, the RNA-dependent RNA polymerases (RdRPs) encoded by the RNA viruses may need to independently control its fidelity to fulfill virus viability and fitness. However, the precise mechanism by which the RdRP maintains its optimal fidelity level remains largely elusive. By solving 2.1-2.5 Å resolution crystal structures of the classical swine fever virus (CSFV) NS5B, an RdRP with a unique naturally fused N-terminal domain (NTD), we identified high-resolution intra-molecular interactions between the NTD and the RdRP palm domain. In order to dissect possible regulatory functions of NTD, we designed mutations at residues Y471 and E472 to perturb key interactions at the NTD-RdRP interface. When crystallized, some of these NS5B interface mutants maintained the interface, while the others adopted an 'open' conformation that no longer retained the intra-molecular interactions. Data from multiple in vitro RdRP assays indicated that the perturbation of the NTD-RdRP interactions clearly reduced the fidelity level of the RNA synthesis, while the processivity of the NS5B elongation complex was not affected. Collectively, our work demonstrates an explicit and unique mode of polymerase fidelity modulation and provides a vivid example of co-evolution in multi-domain enzymes.


Subject(s)
Classical Swine Fever Virus/genetics , Genomic Instability/genetics , Protein Interaction Domains and Motifs , RNA, Viral/biosynthesis , RNA-Dependent RNA Polymerase/chemistry , Classical Swine Fever Virus/enzymology , Classical Swine Fever Virus/metabolism , Crystallography, X-Ray , Genome, Viral/genetics , Models, Molecular , Molecular Conformation , Protein Interaction Domains and Motifs/genetics , RNA, Viral/genetics , RNA, Viral/metabolism , RNA-Dependent RNA Polymerase/genetics , RNA-Dependent RNA Polymerase/metabolism , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/metabolism
12.
J Virol ; 92(15)2018 08 01.
Article in English | MEDLINE | ID: mdl-29769350

ABSTRACT

The members of Flaviviridae utilize several endocytic pathways to enter a variety of host cells. Our previous work showed that classical swine fever virus (CSFV) enters porcine kidney (PK-15) cells through a clathrin-dependent pathway that requires Rab5 and Rab7. The entry mechanism for CSFV into other cell lines remains unclear, for instance, porcine alveolar macrophages (3D4/21 cells). More importantly, the trafficking of CSFV within endosomes controlled by Rab GTPases is unknown in 3D4/21 cells. In this study, entry and postinternalization of CSFV were analyzed using chemical inhibitors, RNA interference, and dominant-negative (DN) mutants. Our data demonstrated that CSFV entry into 3D4/21 cells depends on caveolae, dynamin, and cholesterol but not clathrin or macropinocytosis. The effects of DN mutants and knockdown of four Rab proteins that regulate endosomal trafficking were examined on CSFV infection, respectively. The results showed that Rab5, Rab7, and Rab11, but not Rab9, regulate CSFV endocytosis. Confocal microscopy showed that virus particles colocalize with Rab5, Rab7, or Rab11 within 30 min after virus entry and further with lysosomes, suggesting that after internalization CSFV moves to early, late, and recycling endosomes and then into lysosomes before the release of the viral genome. Our findings provide insights into the life cycle of pestiviruses in macrophages.IMPORTANCE Classical swine fever, is caused by classical swine fever virus (CSFV). The disease is notifiable to World Organisation for Animal Health (OIE) in most countries and causes significant financial losses to the pig industry globally. Understanding the processes of CSFV endocytosis and postinternalization will advance our knowledge of the disease and provide potential novel drug targets against CSFV. With this objective, we used systematic approaches to dissect these processes in CSFV-infected 3D4/21 cells. The data presented here demonstrate for the first time to our knowledge that CSFV is able to enter cells via caveola-mediated endocytosis that requires Rab5, Rab7 and Rab11, in addition to the previously described classical clathrin-dependent pathway that requires Rab5 and Rab7. The characterization of CSFV entry will further promote our current understanding of Pestivirus cellular entry pathways and provide novel targets for antiviral drug development.


Subject(s)
Caveolae/metabolism , Classical Swine Fever Virus/metabolism , Endocytosis , Macrophages, Alveolar/metabolism , rab GTP-Binding Proteins/metabolism , rab5 GTP-Binding Proteins/metabolism , Animals , Caveolae/virology , Classical Swine Fever Virus/genetics , Macrophages, Alveolar/virology , Swine , rab GTP-Binding Proteins/genetics , rab5 GTP-Binding Proteins/genetics , rab7 GTP-Binding Proteins
13.
Virol J ; 16(1): 127, 2019 11 06.
Article in English | MEDLINE | ID: mdl-31694654

ABSTRACT

BACKGROUND: Capsid (C) protein plays an important role in the replication of classical swine fever virus (CSFV). The ubiquitin proteasome system (UPS) involves in replication of many viruses via modulation of viral proteins. The relationship of CSFV with UPS is poorly understood and the impact of 26S proteasome on C protein has never been reported before. METHODS: In this study, fused C protein with an EGFP tag is expressed in PK-15 and 3D4/2 cells. MG132 and 3-methyladenine (3-MA) are used to detect the roles of 26S proteasome and autophagolysosome in expression levels of C protein. Truncated and mutant C proteins are used to find the exact residues responsible for the degradation of C protein. Immunoprecipitaion is performed to find whether C protein is ubiquitinated or not. RESULTS: C-EGFP protein expresses in a cleaved form at a low level and is degraded by 26S proteasome which could be partly inhibited by MG132. C-terminal residues play more important roles in the degradation of C protein than N-terminal residues. Residues 260 to 267, especially M260 and L261, are crucial for the degradation. In addition, C-terminal residues 262 to 267 determine cleavage efficiency of C protein. CONCLUSIONS: CSFV C protein is degraded by 26S proteasome in a ubiquitin-independent manner. Last 8 residues at C-terminus of immature C protein play a major role in proteasomal degradation of CSFV C protein and determine the cleavage efficiency of C protein by signal peptide peptidase (SPP). Our findings provide valuable help for fully understanding degradation process of C protein and contribute to fully understanding the role of C protein in CSFV replication.


Subject(s)
Capsid Proteins/chemistry , Capsid Proteins/metabolism , Classical Swine Fever Virus/metabolism , Amino Acids , Animals , Capsid Proteins/genetics , Cell Line , Classical Swine Fever/virology , Gene Expression , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Mutation , Proteasome Endopeptidase Complex/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Swine
14.
J Gen Virol ; 99(1): 86-96, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29235980

ABSTRACT

The classical swine fever virus (CSFV) represents one of the most important pathogens of swine. The CSFV glycoprotein Erns is an essential structural protein and an important virulence factor. The latter is dependent on the RNase activity of this envelope protein and, most likely, its secretion from the infected cell. A further important feature with regard to its function as a virulence factor is the formation of disulfide-linked Erns homodimers that are found in virus-infected cells and virions. Mutant CSFV lacking cysteine (Cys) 171, the residue responsible for intermolecular disulfide bond formation, were found to be attenuated in pigs (Tews BA, Schürmann EM, Meyers G. J Virol 2009;83:4823-4834). In the course of an animal experiment with such a dimerization-negative CSFV mutant, viruses were reisolated from pigs that contained a mutation of serine (Ser) 209 to Cys. This mutation restored the ability to form disulphide-linked Erns homodimers. In transient expression studies Erns mutants carrying the S209C change were found to form homodimers with about wt efficiency. Also the secretion level of the mutated proteins was equivalent to that of wt Erns. Virus mutants containing the Cys171Ser/Ser209Cys configuration exhibited wt growth rates and increased virulence when compared with the Cys171Ser mutant. These results provide further support for the connection between CSFV virulence and Erns dimerization.


Subject(s)
Classical Swine Fever Virus/genetics , Classical Swine Fever Virus/pathogenicity , Classical Swine Fever/virology , Epithelial Cells/virology , Mutation , Viral Envelope Proteins/genetics , Animals , Base Sequence , Cell Line , Classical Swine Fever/pathology , Classical Swine Fever Virus/metabolism , Cricetulus , Gene Expression , Genetic Engineering , Kidney/virology , Protein Multimerization , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Swine , Viral Envelope Proteins/metabolism , Viral Load , Virulence
15.
Arch Virol ; 162(2): 391-400, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27766426

ABSTRACT

NS5A is a multifunctional non-structural protein of classical swine fever virus (CSFV) that plays an important role in viral replication, but how it exerts its functions is unknown. Here, we report the cleavage of NS5A of the vaccine C-strain, resulting in two truncated forms (b and c). Further experiments using calpain- and caspase-family-specific inhibitors, followed by a caspase-6-specific shRNAs and inhibitor, showed that the cleavage of C-strain NS5A to produce truncated form c is mediated by caspase-6, mapping to 272DTTD275, while the cleavage producing truncated form b is probably mediated by another unknown protease. shRNA-mediated downregulation of caspase-6 and blocking of enzyme activity in ST cells significantly impaired genome replication and virus production, indicating that NS5A cleavage is required for CSFV replication.


Subject(s)
Calpain/genetics , Caspase 6/genetics , Classical Swine Fever Virus/drug effects , Leydig Cells/drug effects , RNA-Dependent RNA Polymerase/chemistry , Viral Nonstructural Proteins/chemistry , Amino Acid Chloromethyl Ketones/pharmacology , Amino Acid Sequence , Animals , Calpain/antagonists & inhibitors , Calpain/metabolism , Caspase 6/metabolism , Caspase Inhibitors/pharmacology , Cell Line , Classical Swine Fever Virus/genetics , Classical Swine Fever Virus/metabolism , Leydig Cells/virology , Male , Plasmids/chemistry , Plasmids/metabolism , Protease Inhibitors/pharmacology , Proteolysis/drug effects , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , RNA-Dependent RNA Polymerase/genetics , RNA-Dependent RNA Polymerase/metabolism , Swine , Testis/drug effects , Testis/virology , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/metabolism , Virus Replication/drug effects
16.
Virol J ; 13: 101, 2016 06 14.
Article in English | MEDLINE | ID: mdl-27296632

ABSTRACT

BACKGROUND: Classical swine fever (CSF) caused by CSF virus (CSFV) is a highly contagious disease of the pigs. A number of studies have suggested that CSFV non-structural (NS) 5A protein is involved in CSFV-associated pathogenesis, but its mechanism is still uncertain. The aim of this study was to investigate the roles of NS5A protein in CSFV-associated pathogenesis in cultured porcine alveolar macrophages (PAMs). METHODS: After PAMs cultured in vitro were transfected with CSFV NS5A, the alterations in IL-1ß, IL-6 and TNF-α expression were determined by ELISA, the RIG-I signaling activity related to inflammatory cytokine secretion was investigated by Western blot and Immunofluorescent staining. RESULTS: It was suggested that, the stable expressed CSFV NS5A solely had no influence on the expressions of inflammatory cytokines IL-1ß, IL-6 and TNF-α in PAMs Moreover, NS5A protein could suppressed IL-1ß, IL-6 and TNF-α expression induced by poly(I:C). It was also showed that NS5A protein did not impair the expressions of RIG-I, MDA5, IPS-1, NF-κB and IkBα in cells without poly(I:C) stimulation. Protein expressions of RIG-I, MDA5, IPS-1, NF-κB were not disrupted by NS5A protein in poly(I:C)-stimulated cells, while poly(I:C)-induced NF-κB nuclear translocation and activity was obviously suppressed by this protein. A suppression in poly(I:C)-induced IkBα degradation in NS5A-expressing cells was also observed. CONCLUSION: These data indicated that CSFV NS5A protein could inhibit the secretion of inflammatory cytokine induced by poly(I:C) through the suppression of the NF-κB signaling pathway, indicating the participation of CSFV NS5A protein in the pathogenesis of CSFV.


Subject(s)
Classical Swine Fever Virus/metabolism , Classical Swine Fever/virology , Cytokines/metabolism , Macrophages, Alveolar/metabolism , NF-kappa B/metabolism , Viral Nonstructural Proteins/metabolism , Animals , Classical Swine Fever/genetics , Classical Swine Fever/metabolism , Classical Swine Fever Virus/genetics , Cytokines/genetics , Host-Parasite Interactions , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Interleukin-6/genetics , Interleukin-6/metabolism , NF-kappa B/genetics , Signal Transduction , Swine , Viral Nonstructural Proteins/genetics
17.
Virus Genes ; 49(1): 106-15, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24824302

ABSTRACT

Study of the classical swine fever virus (CSFV) replication is challenging because it is a BSL-3-Ag pathogen that requires specialized facilities. We developed a cell-based assay in human embryonic kidney 293T cells that can quantify the activities of NS5B, the CSFV RNA-dependent RNA polymerase. The 5BR assay uses transiently-expressed CSFV NS5B to produce RNAs that activate the RIG-I-mediated signaling pathway to result in reporter protein production. Upon co-expression of the CSFV core protein, we observed enhancement of the CSFV RdRp activity. The CSFV core and NS5B proteins could co-immunoprecipitate with each other and co-localize in cells, when visualized by confocal microscopy. Analyses of combinations of RdRps and capsid proteins from different viruses demonstrated that the CSFV core could enhance the CSFV NS5B activity in a virus species-specific manner. Studies of truncated versions of CSFV core demonstrated that the first 30 residues of core protein are dispensable for interaction with the CSFV NS5B. Purified core protein could enhance RNA synthesis by the purified NS5B in vitro, with the increase being in the synthesis of the de novo-initiated RNA. These results demonstrate that the CSFV core protein can regulate the mechanism of RNA synthesis by the CSFV RdRp.


Subject(s)
Classical Swine Fever Virus/enzymology , Classical Swine Fever Virus/metabolism , RNA/biosynthesis , Viral Core Proteins/metabolism , Viral Nonstructural Proteins/metabolism , Cell Line , DNA Mutational Analysis , Humans , Immunoprecipitation , Microscopy, Confocal , Protein Binding , Protein Interaction Mapping , Signal Transduction
18.
BMC Vet Res ; 10: 279, 2014 Dec 02.
Article in English | MEDLINE | ID: mdl-25439655

ABSTRACT

BACKGROUND: Classical swine fever virus (CSFV) infection causes significant losses of pigs, which is characterized by hemorrhage, disseminated intravascular coagulation and leucopenia. The swine vascular endothelial cell is a primary target cell for CSFV. The aim of this study was to determine the role of CSFV infection in inducing oxidative stress (OS) in vascular endothelial cells. RESULTS: We demonstrated that CSFV infection induced oxidative stress in swine umbilical vein endothelial cells (SUVECs), characterized by the induction of reactive oxygen species (ROS) production and the elevations of porcine antioxidant proteins thioredoxin (Trx), peroxiredoxin-6 (PRDX-6) and heme oxygenase-1 (HO-1) expression. Furthermore, cyclooxygenase-2 (COX-2), a pro-inflammatory protein related to oxidative stress, was up-regulated while anti-inflammatory protein peroxisome proliferator-activated receptor-γ (PPAR-γ), an important mediator in vascular functional regulation, was down-regulated in the CSFV infected cells. In addition, antioxidants showed significant inhibitory effects on the CSFV replication, indicating a close relationship between CSFV replication and OS induced in the host cells. CONCLUSIONS: Our results indicated that CSFV infection induced oxidative stress in SUVECs. These findings provide novel information on the mechanism by which CSFV can alter intracellular events associated with the viral infection.


Subject(s)
Classical Swine Fever Virus/metabolism , Endothelium, Vascular/virology , Oxidative Stress , Umbilical Veins , Animals , Classical Swine Fever/metabolism , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Flow Cytometry , Heme Oxygenase-1/metabolism , Microscopy, Fluorescence , Peroxiredoxin VI/metabolism , Reactive Oxygen Species/metabolism , Swine , Thioredoxins/metabolism
19.
J Virol ; 86(12): 6778-91, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22496228

ABSTRACT

The nonstructural protein p7 of classical swine fever virus (CSFV) is a small hydrophobic polypeptide with an apparent molecular mass of 6 to 7 kDa. The protein contains two hydrophobic stretches of amino acids interrupted by a short charged segment that are predicted to form transmembrane helices and a cytosolic loop, respectively. Using reverse genetics, partial in-frame deletions of p7 were deleterious for virus growth, demonstrating that CSFV p7 function is critical for virus production in cell cultures. A panel of recombinant mutant CSFVs was created using alanine scanning mutagenesis of the p7 gene harboring sequential three- to six-amino-acid residue substitutions spanning the entire protein. These recombinant viruses allowed the identification of the regions within p7 that are critical for virus production in vitro. In vivo, some of these viruses were partially or completely attenuated in swine relative to the highly virulent parental CSFV Brescia strain, indicating a significant role of p7 in CSFV virulence. Structure-function analyses in model membranes emulating the endoplasmic reticulum lipid composition confirmed that CSFV p7 is a pore-forming protein, and that pore-forming activity resides in the C-terminal transmembrane helix. Therefore, p7 is a viroporin which is clearly involved in the process of CSFV virulence in swine.


Subject(s)
Classical Swine Fever Virus/metabolism , Classical Swine Fever Virus/pathogenicity , Classical Swine Fever/virology , Viral Nonstructural Proteins/metabolism , Amino Acid Motifs , Animals , Cell Line , Classical Swine Fever Virus/chemistry , Classical Swine Fever Virus/genetics , Swine , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/genetics , Virulence
20.
Risk Anal ; 33(8): 1454-72, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23231448

ABSTRACT

Exotic animal diseases (EADs) are characterized by their capacity to spread global distances, causing impacts on animal health and welfare with significant economic consequences. We offer a critique of current import risk analysis approaches employed in the EAD field, focusing on their capacity to assess complex systems at a policy level. To address the shortcomings identified, we propose a novel method providing a systematic analysis of the likelihood of a disease incursion, developed by reference to the multibarrier system employed for the United Kingdom. We apply the network model to a policy-level risk assessment of classical swine fever (CSF), a notifiable animal disease caused by the CSF virus. In doing so, we document and discuss a sequence of analyses that describe system vulnerabilities and reveal the critical control points (CCPs) for intervention, reducing the likelihood of U.K. pig herds being exposed to the CSF virus.


Subject(s)
Classical Swine Fever/epidemiology , Classical Swine Fever/transmission , Risk Assessment/methods , Algorithms , Animal Husbandry , Animals , Classical Swine Fever/virology , Classical Swine Fever Virus/metabolism , Disease Outbreaks/veterinary , Health Policy , Humans , Models, Statistical , Stochastic Processes , Sus scrofa , Swine , Systems Theory , United Kingdom
SELECTION OF CITATIONS
SEARCH DETAIL