Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 89
Filter
Add more filters










Publication year range
1.
Nat Commun ; 15(1): 4607, 2024 May 30.
Article in English | MEDLINE | ID: mdl-38816407

ABSTRACT

Type II topoisomerases are ubiquitous enzymes that play a pivotal role in modulating the topological configuration of double-stranded DNA. These topoisomerases are required for DNA metabolism and have been extensively studied in both prokaryotic and eukaryotic organisms. However, our understanding of virus-encoded type II topoisomerases remains limited. One intriguing example is the African swine fever virus, which stands as the sole mammalian-infecting virus encoding a type II topoisomerase. In this work, we use several approaches including cryo-EM, X-ray crystallography, and biochemical assays to investigate the structure and function of the African swine fever virus type II topoisomerase, pP1192R. We determine the structures of pP1192R in different conformational states and confirm its enzymatic activity in vitro. Collectively, our results illustrate the basic mechanisms of viral type II topoisomerases, increasing our understanding of these enzymes and presenting a potential avenue for intervention strategies to mitigate the impact of the African swine fever virus.


Subject(s)
African Swine Fever Virus , Cryoelectron Microscopy , DNA Topoisomerases, Type II , African Swine Fever Virus/enzymology , African Swine Fever Virus/genetics , DNA Topoisomerases, Type II/metabolism , DNA Topoisomerases, Type II/chemistry , Animals , Crystallography, X-Ray , Swine , Viral Proteins/metabolism , Viral Proteins/chemistry , Viral Proteins/genetics , Models, Molecular , Protein Conformation , African Swine Fever/virology
2.
Nucleic Acids Res ; 51(17): 9475-9490, 2023 09 22.
Article in English | MEDLINE | ID: mdl-37587714

ABSTRACT

African swine fever virus (ASFV) is highly contagious and can cause lethal disease in pigs. Although it has been extensively studied in the past, no vaccine or other useful treatment against ASFV is available. The genome of ASFV encodes more than 170 proteins, but the structures and functions for the majority of the proteins remain elusive, which hindered our understanding on the life cycle of ASFV and the development of ASFV-specific inhibitors. Here, we report the structural and biochemical studies of the highly conserved C962R protein of ASFV, showing that C962R is a multidomain protein. The N-terminal AEP domain is responsible for the DNA polymerization activity, whereas the DNA unwinding activity is catalyzed by the central SF3 helicase domain. The middle PriCT2 and D5_N domains and the C-terminal Tail domain all contribute to the DNA unwinding activity of C962R. C962R preferentially works on forked DNA, and likely functions in Base-excision repair (BER) or other repair pathway in ASFV. Although it is not essential for the replication of ASFV, C962R can serve as a model and provide mechanistic insight into the replicative primase proteins from many other species, such as nitratiruptor phage NrS-1, vaccinia virus (VACV) and other viruses.


Subject(s)
African Swine Fever Virus , Viral Proteins , Animals , African Swine Fever/virology , African Swine Fever Virus/enzymology , Swine , Viral Proteins/chemistry , Viral Proteins/metabolism , DNA Topoisomerases, Type I/chemistry , DNA Replication
3.
J Biol Chem ; 299(7): 104844, 2023 07.
Article in English | MEDLINE | ID: mdl-37209818

ABSTRACT

Cytoplasmic stress granules (SGs) are generally triggered by stress-induced translation arrest for storing mRNAs. Recently, it has been shown that SGs are regulated by different stimulators including viral infection, which is involved in the antiviral activity of host cells to limit viral propagation. To survive, several viruses have been reported to execute various strategies, such as modulating SG formation, to create optimal surroundings for viral replication. African swine fever virus (ASFV) is one of the most notorious pathogens in the global pig industry. However, the interplay between ASFV infection and SG formation remains largely unknown. In this study, we found that ASFV infection inhibited SG formation. Through SG inhibitory screening, we found that several ASFV-encoded proteins are involved in inhibition of SG formation. Among them, an ASFV S273R protein (pS273R), the only cysteine protease encoded by the ASFV genome, significantly affected SG formation. ASFV pS273R interacted with G3BP1 (Ras-GTPase-activating protein [SH3 domain] binding protein 1), a vital nucleating protein of SG formation. Furthermore, we found that ASFV pS273R cleaved G3BP1 at the G140-F141 to produce two fragments (G3BP1-N1-140 and G3BP1-C141-456). Interestingly, both the pS273R-cleaved fragments of G3BP1 lost the ability to induce SG formation and antiviral activity. Taken together, our finding reveals that the proteolytic cleavage of G3BP1 by ASFV pS273R is a novel mechanism by which ASFV counteracts host stress and innate antiviral responses.


Subject(s)
African Swine Fever Virus , Stress Granules , Viral Proteins , Animals , African Swine Fever/metabolism , African Swine Fever/virology , African Swine Fever Virus/enzymology , African Swine Fever Virus/genetics , Poly-ADP-Ribose Binding Proteins/metabolism , Stress Granules/metabolism , Swine , Virus Replication/physiology , Chlorocebus aethiops , Humans , HEK293 Cells , Cells, Cultured , Macrophages, Alveolar/virology , Viral Proteins/metabolism , Proteolysis
4.
Molecules ; 28(2)2023 Jan 06.
Article in English | MEDLINE | ID: mdl-36677630

ABSTRACT

The African Swine Fever virus (ASFV) causes an infectious viral disease in pigs of all ages. The development of antiviral drugs primarily aimed at inhibition of proteases required for the proteolysis of viral polyproteins. In this study, the conformation of the pS273R protease in physiological states were investigated, virtually screened the multi-protein conformation of pS273R target proteins, combined various molecular docking scoring functions, and identified five potential drugs from the Food and Drug Administration drug library that may inhibit pS273R. Subsequent validation of the dynamic interactions of pS273R with the five putative inhibitors was achieved using molecular dynamics simulations and binding free energy calculations using the molecular mechanics/Poison-Boltzmann (Generalized Born) (MM/PB(GB)SA) surface area. These findings demonstrate that the arm domain and Thr159-Lys167 loop region of pS273R are significantly more flexible compared to the core structural domain, and the Thr159-Lys167 loop region can serve as a "gatekeeper" in the substrate channel. Leucovorin, Carboprost, Protirelin, Flavin Mononucleotide, and Lovastatin Acid all have Gibbs binding free energies with pS273R that were less than -20 Kcal/mol according to the MM/PBSA analyses. In contrast to pS273R in the free energy landscape, the inhibitor and drug complexes of pS273R showed distinct structural group distributions. These five drugs may be used as potential inhibitors of pS273R and may serve as future drug candidates for treating ASFV.


Subject(s)
African Swine Fever Virus , Antiviral Agents , Protease Inhibitors , Animals , African Swine Fever Virus/drug effects , African Swine Fever Virus/enzymology , Endopeptidases , Molecular Docking Simulation , Molecular Dynamics Simulation , Peptide Hydrolases , Protein Conformation , Swine , Protease Inhibitors/chemistry , Protease Inhibitors/pharmacology , Antiviral Agents/chemistry , Antiviral Agents/pharmacology
5.
J Virol ; 96(10): e0190521, 2022 05 25.
Article in English | MEDLINE | ID: mdl-35481780

ABSTRACT

Removal of 5' cap on cellular mRNAs by the African swine fever virus (ASFV) decapping enzyme g5R protein (g5Rp) is beneficial to viral gene expression during the early stages of infection. As the only nucleoside diphosphate-linked moiety X (Nudix) decapping enzyme encoded in the ASFV genome, g5Rp works in both the degradation of cellular mRNA and the hydrolyzation of the diphosphoinositol polyphosphates. Here, we report the structures of dimeric g5Rp and its complex with inositol hexakisphosphate (InsP6). The two g5Rp protomers interact head to head to form a dimer, and the dimeric interface is formed by extensive polar and nonpolar interactions. Each protomer is composed of a unique N-terminal helical domain and a C-terminal classic Nudix domain. As g5Rp is an mRNA-decapping enzyme, we identified key residues, including K8, K94, K95, K98, K175, R221, and K243 located on the substrate RNA binding interfaces of g5Rp which are important to RNA binding and decapping enzyme activity. Furthermore, the g5Rp-mediated mRNA decapping was inhibited by InsP6. The g5Rp-InsP6 complex structure showed that the InsP6 molecules occupy the same regions that primarily mediate g5Rp-RNA interaction, elucidating the roles of InsP6 in the regulation of the viral decapping activity of g5Rp in mRNA degradation. Collectively, these results provide the structural basis of interaction between RNA and g5Rp and highlight the inhibitory mechanism of InsP6 on mRNA decapping by g5Rp. IMPORTANCE ASF is a highly contagious hemorrhagic viral disease in domestic pigs which causes high mortality. Currently, there are still no effective vaccines or specific drugs available against this particular virus. The protein g5Rp is the only viral mRNA-decapping enzyme, playing an essential role in the machinery assembly of mRNA regulation and translation initiation. In this study, we solved the crystal structures of g5Rp dimer and complex with InsP6. Structure-based mutagenesis studies revealed critical residues involved in a candidate RNA binding region, which also play pivotal roles in complex with InsP6. Notably, InsP6 can inhibit g5Rp activity by competitively blocking the binding of substrate mRNA to the enzyme. Our structure-function studies provide the basis for potential anti-ASFV inhibitor designs targeting the critical enzyme.


Subject(s)
African Swine Fever Virus , Endoribonucleases , Phytic Acid , African Swine Fever , African Swine Fever Virus/drug effects , African Swine Fever Virus/enzymology , Animals , Endoribonucleases/genetics , Endoribonucleases/metabolism , Phytic Acid/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Swine
6.
J Biol Chem ; 298(1): 101480, 2022 01.
Article in English | MEDLINE | ID: mdl-34890644

ABSTRACT

African swine fever (ASF) is a viral hemorrhagic disease that affects domestic pigs and wild boar and is caused by the African swine fever virus (ASFV). The ASFV virion contains a long double-stranded DNA genome, which encodes more than 150 proteins. However, the immune escape mechanism and pathogenesis of ASFV remain poorly understood. Here, we report that the pyroptosis execution protein gasdermin D (GSDMD) is a new binding partner of ASFV-encoded protein S273R (pS273R), which belongs to the SUMO-1 cysteine protease family. Further experiments demonstrated that ASFV pS273R-cleaved swine GSDMD in a manner dependent on its protease activity. ASFV pS273R specifically cleaved GSDMD at G107-A108 to produce a shorter N-terminal fragment of GSDMD consisting of residues 1 to 107 (GSDMD-N1-107). Interestingly, unlike the effect of GSDMD-N1-279 fragment produced by caspase-1-mediated cleavage, the assay of LDH release, cell viability, and virus replication showed that GSDMD-N1-107 did not trigger pyroptosis or inhibit ASFV replication. Our findings reveal a previously unrecognized mechanism involved in the inhibition of ASFV infection-induced pyroptosis, which highlights an important function of pS273R in inflammatory responses and ASFV replication.


Subject(s)
African Swine Fever Virus , African Swine Fever , Cysteine Proteases , Phosphate-Binding Proteins , Pore Forming Cytotoxic Proteins , Viral Proteins , African Swine Fever/virology , African Swine Fever Virus/enzymology , African Swine Fever Virus/metabolism , Animals , Cysteine Proteases/metabolism , Phosphate-Binding Proteins/metabolism , Pore Forming Cytotoxic Proteins/metabolism , Pyroptosis , Sus scrofa , Swine , Viral Proteins/metabolism
7.
Viruses ; 13(6)2021 06 17.
Article in English | MEDLINE | ID: mdl-34204411

ABSTRACT

African swine fever virus (ASFV) is an acute and persistent swine virus with a high economic burden that encodes multiple genes to evade host immune response. In this work, we have revealed that early viral protein UBCv1, the only known conjugating enzyme encoded by a virus, modulates innate immune and inflammatory signaling. Transient overexpression of UBCv1 impaired activation of NF-κB and AP-1 transcription factors induced by several agonists of these pathways. In contrast, activation of IRF3 and ISRE signaling upon stimulation with TRIFΔRIP, cGAS/STING or RIG-I-CARD remained unaltered. Experiments aimed at mapping UBCv1 inhibitory activity indicated that this viral protein acts upstream or at the level step of IKKß. In agreement with this, UBCv1 was able to block p65 nuclear translocation upon cytokine stimulation, a key event in NF-ĸB signaling. Additionally, A549 stably transduced for UBCv1 showed a significant decrease in the levels of NF-ĸB dependent genes. Interestingly, despite the well-defined capacity of UBCv1 to conjugate ubiquitin chains, a mutant disabled for ubiquitylation activity retained similar immunomodulatory activity as the wild-type enzyme, suggesting that the two functions are segregated. Altogether these data suggest that ASFV UBCv1 manipulates the innate immune response targeting the NF-κB and AP-1 pathways and opens new questions about the multifunctionality of this enzyme.


Subject(s)
African Swine Fever Virus/enzymology , Immunity, Innate , Immunomodulation , NF-kappa B/genetics , Ubiquitin-Conjugating Enzymes/genetics , Ubiquitin-Conjugating Enzymes/immunology , A549 Cells , African Swine Fever Virus/immunology , Animals , HEK293 Cells , Humans , Interferon Type I/immunology , NF-kappa B/immunology , NF-kappa B/metabolism , Signal Transduction/immunology , Swine , Ubiquitin/metabolism , Ubiquitin-Conjugating Enzymes/metabolism
8.
Bioorg Med Chem ; 35: 116055, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33607487

ABSTRACT

African swine fever (ASF) is a viral disease in swine that results in high mortality in domestic pigs and causes considerable economic losses. Currently, there is no effective vaccine or drugs available for treatment. Identification of new anti-ASFV drugs is urgently needed. Here, the pS273R protein of the African swine fever virus (ASFV) is a specific SUMO-1-like cysteine protease that plays an important role in its replication process. To inhibit virus replication and improve treatment options, a set of small-molecule compounds, targeted inhibitors against the ASFV pS273R protease, were obtained through molecular screening by homology modeling and molecular docking based on structural information of pS273R. Our results clearly demonstrated that the 14th carbon atom of the cysteinase inhibitor E-64 could form one CS covalent bond with the Cys 232 amino acid of the pS273R protease and seven additional hydrogen bonds to maintain a stable binding state. Simultaneously, cell viability, immunophenotyping, and in vitro enzyme activity inhibition assays were performed to comprehensively evaluate E-64 characteristics. Our findings demonstrated that 4 mmol/L E-64 could effectively inhibit the enzyme activity center of the pS273R protease by preventing pS273R protease from lysing pp62, while promoting the upregulation of immune-related cytokines at the transcription level. Moreover, cell viability results revealed that 4 mmol/L E-64 was not cytotoxic. Taken together, we identified a novel strategy to potentially prevent ASFV infection in pigs by blocking the activity of pS273R protease with a small-molecule inhibitor.


Subject(s)
African Swine Fever Virus/enzymology , Cysteine Proteases/metabolism , Cysteine Proteinase Inhibitors/pharmacology , Small Molecule Libraries/pharmacology , Viral Proteins/antagonists & inhibitors , Animals , Cell Line , Cell Survival/drug effects , Cysteine Proteinase Inhibitors/chemistry , Dose-Response Relationship, Drug , Models, Molecular , Molecular Structure , Small Molecule Libraries/chemistry , Structure-Activity Relationship , Swine , Viral Proteins/metabolism , Virus Replication/drug effects
9.
Biochem Biophys Res Commun ; 534: 526-532, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33223051

ABSTRACT

The DNA polymerase from african swine fever virus (ASFV Pol X), lacking both 8 kDa and thumb domains, is the smallest enzyme featuring competence in DNA extension. Here we show that ASFV Pol X features poor filling activity of DNA gaps consisting of 15 bases, and exerts a more efficient action at the expense of DNA substrates containing a recessed end of equal length. We also show that shortening the recessed end of DNA substrates decreases the rate of DNA elongation catalysed by ASFV Pol X. Finally, by means of stopped-flow experiments we were able to determine that DNA binding is a step responsible for restraining the efficiency of ASFV Pol X catalytic action.


Subject(s)
African Swine Fever Virus/metabolism , African Swine Fever/virology , DNA, Viral/metabolism , DNA-Directed DNA Polymerase/metabolism , Viral Proteins/metabolism , African Swine Fever Virus/chemistry , African Swine Fever Virus/enzymology , Animals , DNA, Viral/chemistry , DNA-Directed DNA Polymerase/chemistry , Models, Molecular , Substrate Specificity , Swine/virology , Viral Proteins/chemistry
10.
J Biol Chem ; 296: 100015, 2021.
Article in English | MEDLINE | ID: mdl-33139328

ABSTRACT

African swine fever, caused by the African swine fever virus (ASFV), is among the most significant swine diseases. There are currently no effective treatments against ASFV. ASFV contains a gene encoding a dUTPase (E165R), which is required for viral replication in swine macrophages, making it an attractive target for inhibitor development. However, the full structural details of the ASFV dUTPase and those of the comparable swine enzyme are not available, limiting further insights. Herein, we determine the crystal structures of ASFV dUTPase and swine dUTPase in both their ligand-free and ligand-bound forms. We observe that the swine enzyme employs a classical dUTPase architecture made up of three-subunit active sites, whereas the ASFV enzyme employs a novel two-subunit active site. We then performed a comparative analysis of all dUTPase structures uploaded in the Protein Data Bank (PDB), which showed classical and non-classical types were mainly determined by the C-terminal ß-strand orientation, and the difference was mainly related to the four amino acids behind motif IV. Thus, our study not only explains the reason for the structural diversity of dUTPase but also reveals how to predict dUTPase type, which may have implications for the dUTPase family. Finally, we tested two dUTPase inhibitors developed for the Plasmodium falciparum dUTPase against the swine and ASFV enzymes. One of these compounds inhibited the ASFV dUTPase at low micromolar concentrations (Kd = 15.6 µM) and with some selectivity (∼2x) over swine dUTPase. In conclusion, our study expands our understanding of the dUTPase family and may aid in the development of specific ASFV inhibitors.


Subject(s)
African Swine Fever Virus/enzymology , Antiviral Agents/pharmacology , Enzyme Inhibitors/pharmacology , Pyrophosphatases/antagonists & inhibitors , Pyrophosphatases/chemistry , African Swine Fever Virus/drug effects , African Swine Fever Virus/physiology , Amino Acid Sequence , Animals , Antiviral Agents/chemistry , Catalytic Domain , Crystallography, X-Ray , Drug Development , Enzyme Inhibitors/chemistry , Host-Pathogen Interactions , Macrophages/virology , Plasmodium falciparum/enzymology , Protein Conformation , Swine , Virus Replication/drug effects
11.
J Enzyme Inhib Med Chem ; 35(1): 1045-1049, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32299265

ABSTRACT

African swine fever (ASF) caused by the ASF virus (ASFV) is the most hazardous swine disease. Since a huge number of pigs have been slaughtered to avoid a pandemic spread, intense studies on the disease should be followed quickly. Recent studies reported that flavonoids have various antiviral activity including ASFV. In this report, ASFV protease was selected as an antiviral target protein to cope with ASF. With a FRET (Fluorescence resonance energy transfer) method, ASFV protease was assayed with a flavonoid library which was composed of sixty-five derivatives classified based on ten different scaffolds. Of these, the flavonols scaffold contains a potential anti-ASFV protease activity. The most prominent flavonol was myricetin with IC50 of 8.4 µM. Its derivative, myricitrin, with the rhamnoside moiety was also showed the profound inhibitory effect on ASFV protease. These two flavonols apparently provide a way to develop anti-ASFV agents based on their scaffold.


Subject(s)
African Swine Fever Virus/drug effects , Antiviral Agents/pharmacology , Endopeptidases/metabolism , Flavonoids/pharmacology , Viral Proteins/antagonists & inhibitors , African Swine Fever Virus/enzymology , Antiviral Agents/chemistry , Dose-Response Relationship, Drug , Endopeptidases/genetics , Flavonoids/chemistry , Microbial Sensitivity Tests , Molecular Structure , Structure-Activity Relationship , Viral Proteins/genetics , Viral Proteins/metabolism
12.
J Virol ; 94(10)2020 05 04.
Article in English | MEDLINE | ID: mdl-32075933

ABSTRACT

African swine fever (ASF) is a highly contagious hemorrhagic viral disease of domestic and wild pigs that is responsible for serious economic and production losses. It is caused by the African swine fever virus (ASFV), a large and complex icosahedral DNA virus of the Asfarviridae family. Currently, there is no effective treatment or approved vaccine against the ASFV. pS273R, a specific SUMO-1 cysteine protease, catalyzes the maturation of the pp220 and pp62 polyprotein precursors into core-shell proteins. Here, we present the crystal structure of the ASFV pS273R protease at a resolution of 2.3 Å. The overall structure of the pS273R protease is represented by two domains named the "core domain" and the N-terminal "arm domain." The "arm domain" contains the residues from M1 to N83, and the "core domain" contains the residues from N84 to A273. A structure analysis reveals that the "core domain" shares a high degree of structural similarity with chlamydial deubiquitinating enzyme, sentrin-specific protease, and adenovirus protease, while the "arm domain" is unique to ASFV. Further, experiments indicated that the "arm domain" plays an important role in maintaining the enzyme activity of ASFV pS273R. Moreover, based on the structural information of pS273R, we designed and synthesized several peptidomimetic aldehyde compounds at a submolar 50% inhibitory concentration, which paves the way for the design of inhibitors to target this severe pathogen.IMPORTANCE African swine fever virus, a large and complex icosahedral DNA virus, causes a deadly infection in domestic pigs. In addition to Africa and Europe, countries in Asia, including China, Vietnam, and Mongolia, were negatively affected by the hazards posed by ASFV outbreaks in 2018 and 2019, at which time more than 30 million pigs were culled. Until now, there has been no vaccine for protection against ASFV infection or effective treatments to cure ASF. Here, we solved the high-resolution crystal structure of the ASFV pS273R protease. The pS273R protease has a two-domain structure that distinguishes it from other members of the SUMO protease family, while the unique "arm domain" has been proven to be essential for its hydrolytic activity. Moreover, the peptidomimetic aldehyde compounds designed to target the substrate binding pocket exert prominent inhibitory effects and can thus be used in a potential lead for anti-ASFV drug development.


Subject(s)
African Swine Fever Virus/enzymology , Cysteine Endopeptidases/chemistry , Viral Proteins/chemistry , African Swine Fever/virology , Amino Acid Sequence , Animals , Crystallography, X-Ray , Cysteine Endopeptidases/genetics , Molecular Dynamics Simulation , Polyproteins/chemistry , Protein Conformation , Protein Domains , SUMO-1 Protein , Sequence Alignment , Sus scrofa , Swine , Viral Proteins/genetics , Viral Proteins/metabolism
13.
mBio ; 10(5)2019 10 29.
Article in English | MEDLINE | ID: mdl-31662460

ABSTRACT

E165R, a highly specific dUTP nucleotidohydrolase (dUTPase) encoded by the African swine fever virus (ASFV) genome, is required for productive replication of ASFV in swine macrophages. Here, we solved the high-resolution crystal structures of E165R in its apo state and in complex with its product dUMP. Structural analysis explicitly defined the architecture of the active site of the enzyme as well as the interaction between the active site and the dUMP ligand. By comparing the ASFV E165R structure with dUTPase structures from other species, we found that the active site of E165R is highly similar to those of dUTPases from Mycobacterium tuberculosis and Plasmodium falciparum, against which small-molecule chemicals have been developed, which could be the potential drug or lead compound candidates for ASFV. Our results provide important basis for anti-ASFV drug design by targeting E165R.IMPORTANCE African swine fever virus (ASFV), an Asfivirus affecting pigs and wild boars with up to 100% case fatality rate, is currently rampaging throughout China and some other countries in Asia. There is an urgent need to develop therapeutic and preventive reagents against the virus. Our crystallographic and biochemical studies reveal that ASFV E165R is a member of trimeric dUTP nucleotidohydrolase (dUTPase) family that catalyzes the hydrolysis of dUTP into dUMP. Our apo-E165R and E165R-dUMP structures reveal the constitutive residues and the configuration of the active center of this enzyme in rich detail and give evidence that the active center of E165R is very similar to that of dUTPases from Plasmodium falciparum and Mycobacterium tuberculosis, which have already been used as targets for designing drugs. Therefore, our high-resolution structures of E165R provide useful structural information for chemotherapeutic drug design.


Subject(s)
African Swine Fever Virus/enzymology , Pyrophosphatases/chemistry , Pyrophosphatases/metabolism , African Swine Fever/virology , African Swine Fever Virus/genetics , Animals , Binding Sites , Catalytic Domain , Cloning, Molecular , Crystallography, X-Ray , Drug Delivery Systems , Drug Design , Kinetics , Models, Molecular , Mycobacterium tuberculosis/enzymology , Plasmodium falciparum , Protein Conformation , Pyrophosphatases/classification , Pyrophosphatases/genetics , Sequence Alignment , Swine
14.
Viruses ; 11(9)2019 09 17.
Article in English | MEDLINE | ID: mdl-31533244

ABSTRACT

Animal diseases constitute a continuing threat to animal health, food safety, national economy, and the environment. Among those, African swine fever (ASF) is one of the most devastating viruses affecting pigs and wild suids due to the lack of vaccine or effective treatment. ASF is endemic in countries in sub-Saharan Africa, but since its introduction to the Caucasus region in 2007, a highly virulent strain of ASF virus (ASFV) has continued to circulate and spread into Eastern Europe and Russia, and most recently into Western Europe, China, and various countries of Southeast Asia. Given the importance of this disease, this review will highlight recent discoveries in basic virology with special focus on proteomic analysis, replication cycle, and some recent data on genes involved in cycle progression and viral-host interactions, such as I215L (E2 ubiquitin-conjugating enzyme), EP402R (CD2v), A104R (histone-like protein), QP509L, and Q706L (RNA helicases) or P1192R (Topoisomerase II). Taking into consideration the large DNA genome of ASFV and its complex interactions with the host, more studies and new approaches are to be taken to understand the basic virus-host interaction for ASFV. Proteomic studies are just paving the way for future research.


Subject(s)
African Swine Fever Virus/genetics , African Swine Fever/virology , Host-Pathogen Interactions , Proteomics , RNA Helicases/genetics , African Swine Fever/epidemiology , African Swine Fever Virus/enzymology , Amino Acid Substitution , Animals , Swine , Viral Proteins/genetics
15.
Emerg Microbes Infect ; 8(1): 291-302, 2019.
Article in English | MEDLINE | ID: mdl-30866783

ABSTRACT

African swine fever virus is complex DNA virus that infects pigs with mortality rates up to 100% leading to devastating socioeconomic effected in the affected countries. There is neither a vaccine nor a treatment to control ASF. African swine fever virus genome encodes two putative SF2 RNA helicases (QP509L and Q706L). In the present study, we found that these two RNA helicases do not share a common ancestral besides sharing a sequence overlap. Although, our phylogenetic studies revealed that they are conserved among virulent and non-virulent isolates, it was possible to observe a degree of variation between isolates corresponding to different genotypes occurring in distinct geographic regions. Further experiments showed that QP509L and Q706L are actively transcribed from 4 h post infection. The immunoblot analysis revealed that both protein co-localized in the viral factories at 12 h post infection, however, QP509L was also detected in the cell nucleus. Finally, siRNA assays uncover the relevant role of these proteins during viral cycle progression, in particular, for the late transcription, genome replication, and viral progeny (a reduction of infectious particles up to 99.4% when siRNA against QP509L was used and 98.4% for siRNA against Q706L). Thus, our results suggest that both helicases are essential during viral infection, highlighting the potential use of these enzymes as target for drug and vaccine development against African swine fever.


Subject(s)
African Swine Fever Virus/physiology , Cell Nucleus/metabolism , RNA Helicases/genetics , RNA Helicases/metabolism , African Swine Fever Virus/enzymology , African Swine Fever Virus/genetics , Animals , Conserved Sequence , Gene Expression Regulation, Viral , Phylogeny , Swine , Transcription, Genetic , Viral Proteins/genetics , Viral Proteins/metabolism , Virulence , Virus Replication
16.
Nat Commun ; 10(1): 387, 2019 01 23.
Article in English | MEDLINE | ID: mdl-30674878

ABSTRACT

African swine fever virus (ASFV) is contagious and can cause highly lethal disease in pigs. ASFV DNA ligase (AsfvLIG) is one of the most error-prone ligases identified to date; it catalyzes DNA joining reaction during DNA repair process of ASFV and plays important roles in mutagenesis of the viral genome. Here, we report four AsfvLIG:DNA complex structures and demonstrate that AsfvLIG has a unique N-terminal domain (NTD) that plays critical roles in substrate binding and catalytic complex assembly. In combination with mutagenesis, in vitro binding and catalytic assays, our study reveals that four unique active site residues (Asn153 and Leu211 of the AD domain; Leu402 and Gln403 of the OB domain) are crucial for the catalytic efficiency of AsfvLIG. These unique structural features can serve as potential targets for small molecule design, which could impair genome repair in ASFV and help combat this virus in the future.


Subject(s)
African Swine Fever Virus/enzymology , Catalytic Domain , DNA Ligases/chemistry , African Swine Fever Virus/genetics , Base Sequence , DNA/metabolism , DNA Ligases/genetics , DNA Repair , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Enzyme Assays , Models, Molecular , Mutagenesis , Protein Conformation , Protein Folding , Viral Proteins/genetics
17.
J Virol ; 91(24)2017 12 15.
Article in English | MEDLINE | ID: mdl-29021398

ABSTRACT

African swine fever virus (ASFV) infection is characterized by a progressive decrease in cellular protein synthesis with a concomitant increase in viral protein synthesis, though the mechanism by which the virus achieves this is still unknown. Decrease of cellular mRNA is observed during ASFV infection, suggesting that inhibition of cellular proteins is due to an active mRNA degradation process. ASFV carries a gene (Ba71V D250R/Malawi g5R) that encodes a decapping protein (ASFV-DP) that has a Nudix hydrolase motif and decapping activity in vitro Here, we show that ASFV-DP was expressed from early times and accumulated throughout the infection with a subcellular localization typical of the endoplasmic reticulum, colocalizing with the cap structure and interacting with the ribosomal protein L23a. ASFV-DP was capable of interaction with poly(A) RNA in cultured cells, primarily mediated by the N-terminal region of the protein. ASFV-DP also interacted with viral and cellular RNAs in the context of infection, and its overexpression in infected cells resulted in decreased levels of both types of transcripts. This study points to ASFV-DP as a viral decapping enzyme involved in both the degradation of cellular mRNA and the regulation of viral transcripts.IMPORTANCE Virulent ASFV strains cause a highly infectious and lethal disease in domestic pigs for which there is no vaccine. Since 2007, an outbreak in the Caucasus region has spread to Russia, jeopardizing the European pig population and making it essential to deepen knowledge about the virus. Here, we demonstrate that ASFV-DP is a novel RNA-binding protein implicated in the regulation of mRNA metabolism during infection, making it a good target for vaccine development.


Subject(s)
African Swine Fever Virus/enzymology , Endoribonucleases/genetics , Endoribonucleases/metabolism , RNA, Messenger/metabolism , African Swine Fever Virus/genetics , African Swine Fever Virus/metabolism , Animals , Chlorocebus aethiops , Gene Deletion , Host-Pathogen Interactions , Protein Binding , Pyrophosphatases/genetics , Pyrophosphatases/metabolism , Ribosomal Proteins/metabolism , Sus scrofa , Vero Cells , Viral Proteins/genetics , Nudix Hydrolases
18.
PLoS Biol ; 15(2): e1002599, 2017 02.
Article in English | MEDLINE | ID: mdl-28245220

ABSTRACT

African swine fever virus (ASFV) can cause highly lethal disease in pigs and is becoming a global threat. ASFV DNA Polymerase X (AsfvPolX) is the most distinctive DNA polymerase identified to date; it lacks two DNA-binding domains (the thumb domain and 8-KD domain) conserved in the homologous proteins. AsfvPolX catalyzes the gap-filling reaction during the DNA repair process of the ASFV virus genome; it is highly error prone and plays an important role during the strategic mutagenesis of the viral genome. The structural basis underlying the natural substrate binding and the most frequent dG:dGTP misincorporation of AsfvPolX remain poorly understood. Here, we report eight AsfvPolX complex structures; our structures demonstrate that AsfvPolX has one unique 5'-phosphate (5'-P) binding pocket, which can favor the productive catalytic complex assembly and enhance the dGTP misincorporation efficiency. In combination with mutagenesis and in vitro catalytic assays, our study also reveals the functional roles of the platform His115-Arg127 and the hydrophobic residues Val120 and Leu123 in dG:dGTP misincorporation and can provide information for rational drug design to help combat ASFV in the future.


Subject(s)
African Swine Fever Virus/enzymology , DNA-Directed DNA Polymerase/metabolism , Deoxyguanine Nucleotides/metabolism , Amino Acids/metabolism , Base Sequence , Binding Sites , Crystallography, X-Ray , DNA, Viral/chemistry , DNA, Viral/metabolism , Kinetics , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Protein Binding , Protein Structure, Secondary , Structural Homology, Protein
19.
J Virol ; 91(11)2017 06 01.
Article in English | MEDLINE | ID: mdl-28298603

ABSTRACT

Reoviruses, like many eukaryotic viruses, contain an inverted 7-methylguanosine (m7G) cap linked to the 5' nucleotide of mRNA. The traditional functions of capping are to promote mRNA stability, protein translation, and concealment from cellular proteins that recognize foreign RNA. To address the role of mRNA capping during reovirus replication, we assessed the benefits of adding the African swine fever virus NP868R capping enzyme during reovirus rescue. C3P3, a fusion protein containing T7 RNA polymerase and NP868R, was found to increase protein expression 5- to 10-fold compared to T7 RNA polymerase alone while enhancing reovirus rescue from the current reverse genetics system by 100-fold. Surprisingly, RNA stability was not increased by C3P3, suggesting a direct effect on protein translation. A time course analysis revealed that C3P3 increased protein synthesis within the first 2 days of a reverse genetics transfection. This analysis also revealed that C3P3 enhanced processing of outer capsid µ1 protein to µ1C, a previously described hallmark of reovirus assembly. Finally, to determine the rate of infectious-RNA incorporation into new virions, we developed a new recombinant reovirus S1 gene that expressed the fluorescent protein UnaG. Following transfection of cells with UnaG and infection with wild-type virus, passage of UnaG through progeny was significantly enhanced by C3P3. These data suggest that capping provides nontraditional functions to reovirus, such as promoting assembly and infectious-RNA incorporation.IMPORTANCE Our findings expand our understanding of how viruses utilize capping, suggesting that capping provides nontraditional functions to reovirus, such as promoting assembly and infectious-RNA incorporation, in addition to enhancing protein translation. Beyond providing mechanistic insight into reovirus replication, our findings also show that reovirus reverse genetics rescue is enhanced 100-fold by the NP868R capping enzyme. Since reovirus shows promise as a cancer therapy, efficient reovirus reverse genetics rescue will accelerate production of recombinant reoviruses as candidates to enhance therapeutic potency. NP868R-assisted reovirus rescue will also expedite production of recombinant reovirus for mechanistic insights into reovirus protein function and structure.


Subject(s)
African Swine Fever Virus/enzymology , Nucleotidyltransferases/metabolism , Orthoreovirus, Mammalian/genetics , Orthoreovirus, Mammalian/physiology , RNA, Viral/metabolism , Virion/physiology , Virus Assembly , African Swine Fever Virus/genetics , Cell Line , Recombinant Fusion Proteins/metabolism , Reverse Genetics , Virion/genetics , Virus Replication
20.
Antiviral Res ; 134: 34-41, 2016 10.
Article in English | MEDLINE | ID: mdl-27568922

ABSTRACT

African swine fever virus (ASFV) is the etiological agent of a highly-contagious and fatal disease of domestic pigs, leading to serious socio-economic impact in affected countries. To date, neither a vaccine nor a selective anti-viral drug are available for prevention or treatment of African swine fever (ASF), emphasizing the need for more detailed studies at the role of ASFV proteins involved in viral DNA replication and transcription. Notably, ASFV encodes for a functional type II topoisomerase (ASFV-Topo II) and we recently showed that several fluoroquinolones (bacterial DNA topoisomerase inhibitors) fully abrogate ASFV replication in vitro. Here, we report that ASFV-Topo II gene is actively transcribed throughout infection, with transcripts being detected as early as 2 hpi and reaching a maximum peak concentration around 16 hpi, when viral DNA synthesis, transcription and translation are more active. siRNA knockdown experiments showed that ASFV-Topo II plays a critical role in viral DNA replication and gene expression, with transfected cells presenting lower viral transcripts (up to 89% decrease) and reduced cytopathic effect (-66%) when compared to the control group. Further, a significant decrease in the number of both infected cells (75.5%) and viral factories per cell and in virus yields (up to 99.7%, 2.5 log) was found only in cells transfected with siRNA targeting ASFV-Topo II. We also demonstrate that a short exposure to enrofloxacin during the late phase of infection (from 15 to 1 hpi) induces fragmentation of viral genomes, whereas no viral genomes were detected when enrofloxacin was added from the early phase of infection (from 2 to 16 hpi), suggesting that fluoroquinolones are ASFV-Topo II poisons. Altogether, our results demonstrate that ASFV-Topo II enzyme has an essential role during viral genome replication and transcription, emphasizing the idea that this enzyme can be a potential target for drug and vaccine development against ASF.


Subject(s)
African Swine Fever Virus/drug effects , DNA Topoisomerases, Type II/drug effects , Topoisomerase II Inhibitors/pharmacology , Virus Replication/drug effects , African Swine Fever/virology , African Swine Fever Virus/enzymology , African Swine Fever Virus/genetics , African Swine Fever Virus/physiology , Animals , Antiviral Agents/pharmacology , Chlorocebus aethiops , Enrofloxacin , Fluoroquinolones/pharmacology , Genome, Viral/drug effects , RNA Interference , Real-Time Polymerase Chain Reaction , Swine , Vero Cells , Virus Replication/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...