Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.477
Filter
1.
Plant Cell Rep ; 43(10): 237, 2024 Sep 24.
Article in English | MEDLINE | ID: mdl-39313743

ABSTRACT

KEY MESSAGE: Enhanced recombinant protein expression was achieved in Salinas lettuce and commercial lettuce by designing a unique RNAi that knockdown the gene-silencing mechanism in transient assays. Improved yields of recombinant proteins (RP) are necessary for protein-production efficiency and ease of purification. Achieving high yield in non-tobacco plants will enable diverse plants to be used as hosts in transient protein-expression systems. With improved protein yield, lettuce (Lactuca sativa) could take the lead as a plant host for RP production. Therefore, this study aimed to improve RP production in lettuce var. Salinas by designing a single RNA interference (RNAi) construct targeting LsRDR1 and LsRDR6 using the Tsukuba system vector. Two RNAi constructs, RNAi-1 and RNAi-2, targeting common regions of LsRDR1 and LsRDR6 with 75% and 76% similarity, respectively, were employed to evaluate simultaneous gene silencing. Quantitative transcription analysis demonstrated that both RNAi constructs effectively knocked down LsRDR6 and LsRDR1, but not LsRDR2, at both 3 and 5 days post-infiltration (dpi), with RNAi-1 exhibited slightly higher efficiency. Based on the protein yield, co-expression of RNAi-1 with enhanced green fluorescent protein (EGFP) increased EGFP expression by approximately 4.9-fold and 3.7-fold at 3 dpi and 5 dpi, respectively, compared to control. A similar but slightly lower increase (2.4-fold and 2.33-fold) was observed in commercial lettuce at 3 and 5 dpi, respectively. To confirm these results, co-infiltration with Bet v 1, a major allergen from birch pollen, resulted in a 2.5-fold increase in expression in Salinas lettuce at 5 dpi. This study marks a significant advancement in enhancing transient protein production in lettuce, elevating its potential as a host for recombinant protein production.


Subject(s)
Gene Expression Regulation, Plant , Lactuca , Plant Proteins , RNA Interference , Recombinant Proteins , Lactuca/genetics , Lactuca/metabolism , Plant Proteins/genetics , Plant Proteins/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Plants, Genetically Modified/genetics , RNA-Dependent RNA Polymerase/genetics , RNA-Dependent RNA Polymerase/metabolism
2.
Viruses ; 16(9)2024 Aug 31.
Article in English | MEDLINE | ID: mdl-39339871

ABSTRACT

RNA virus polymerases carry out multiple functions necessary for successful genome replication and transcription. A key tool for molecular studies of viral RNA-dependent RNA polymerases (RdRps) is a 'minigenome' or 'minireplicon' assay, in which viral RdRps are reconstituted in cells in the absence of full virus infection. Typically, plasmids expressing the viral polymerase protein(s) and other co-factors are co-transfected, along with a plasmid expressing an RNA encoding a fluorescent or luminescent reporter gene flanked by viral untranslated regions containing cis-acting elements required for viral RdRp recognition. This reconstitutes the viral transcription/replication machinery and allows the viral RdRp activity to be measured as a correlate of the reporter protein signal. Here, we report on the development of a 'first-generation' plasmid-based minigenome assay for species A rotavirus using a firefly luciferase reporter gene.


Subject(s)
Genes, Reporter , Genome, Viral , Rotavirus , Rotavirus/genetics , Plasmids/genetics , RNA-Dependent RNA Polymerase/genetics , RNA-Dependent RNA Polymerase/metabolism , RNA, Viral/genetics , Virus Replication , Humans , Animals , Luciferases, Firefly/genetics , Luciferases, Firefly/metabolism
3.
Structure ; 32(9): 1301-1321, 2024 Sep 05.
Article in English | MEDLINE | ID: mdl-39241763

ABSTRACT

The coronavirus disease 2019 (COVID-19), the disease caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), represents the most disastrous infectious disease pandemic of the past century. As a member of the Betacoronavirus genus, the SARS-CoV-2 genome encodes a total of 29 proteins. The spike protein, RNA-dependent RNA polymerase, and proteases play crucial roles in the virus replication process and are promising targets for drug development. In recent years, structural studies of these viral proteins and of their complexes with antibodies and inhibitors have provided valuable insights into their functions and laid a solid foundation for drug development. In this review, we summarize the structural features of these proteins and discuss recent progress in research regarding therapeutic development, highlighting mechanistically representative molecules and those that have already been approved or are under clinical investigation.


Subject(s)
Antiviral Agents , SARS-CoV-2 , SARS-CoV-2/drug effects , SARS-CoV-2/metabolism , Antiviral Agents/pharmacology , Antiviral Agents/chemistry , Humans , Spike Glycoprotein, Coronavirus/chemistry , Spike Glycoprotein, Coronavirus/metabolism , COVID-19 Drug Treatment , Coronavirus RNA-Dependent RNA Polymerase/metabolism , Coronavirus RNA-Dependent RNA Polymerase/chemistry , Coronavirus RNA-Dependent RNA Polymerase/antagonists & inhibitors , Models, Molecular , COVID-19/virology , COVID-19/metabolism , RNA-Dependent RNA Polymerase/chemistry , RNA-Dependent RNA Polymerase/metabolism
4.
Antiviral Res ; 230: 105980, 2024 10.
Article in English | MEDLINE | ID: mdl-39117284

ABSTRACT

In search of novel therapeutic options to treat influenza virus (IV) infections, we previously identified a series of inhibitors that act by disrupting the interactions between the PA and PB1 subunits of the viral RNA polymerase. These compounds showed broad-spectrum antiviral activity against human influenza A and B viruses and a high barrier to the induction of drug resistance in vitro. In this short communication, we investigated the effects of combinations of the PA-PB1 interaction inhibitor 54 with oseltamivir carboxylate (OSC), zanamivir (ZA), favipiravir (FPV), and baloxavir marboxil (BXM) on the inhibition of influenza A and B virus replication in vitro. We observed a synergistic effect of the 54/OSC and 54/ZA combinations and an antagonistic effect when 54 was combined with either FPV or BXM. Moreover, we demonstrated the efficacy of 54 against highly pathogenic avian influenza viruses (HPAIVs) both in cell culture and in the embryonated chicken eggs model. Finally, we observed that 54 enhances OSC protective effect against HPAIV replication in the embryonated eggs model. Our findings represent an advance in the development of alternative therapeutic strategies against both human and avian IV infections.


Subject(s)
Antiviral Agents , Drug Synergism , Influenza A virus , Oseltamivir , Pyrazines , Viral Proteins , Virus Replication , Oseltamivir/pharmacology , Oseltamivir/analogs & derivatives , Animals , Antiviral Agents/pharmacology , Humans , Virus Replication/drug effects , Pyrazines/pharmacology , Influenza A virus/drug effects , Chick Embryo , Viral Proteins/metabolism , Viral Proteins/antagonists & inhibitors , Amides/pharmacology , Dibenzothiepins/pharmacology , Influenza B virus/drug effects , Influenza B virus/physiology , Zanamivir/pharmacology , Triazines/pharmacology , Pyridones/pharmacology , Influenza in Birds/drug therapy , Influenza in Birds/virology , Morpholines/pharmacology , Influenza, Human/drug therapy , Influenza, Human/virology , Dogs , DNA-Directed RNA Polymerases/antagonists & inhibitors , DNA-Directed RNA Polymerases/metabolism , RNA-Dependent RNA Polymerase/antagonists & inhibitors , RNA-Dependent RNA Polymerase/metabolism , Cell Line , Madin Darby Canine Kidney Cells
5.
Nat Commun ; 15(1): 6910, 2024 Aug 19.
Article in English | MEDLINE | ID: mdl-39160148

ABSTRACT

Replication of influenza viral RNA depends on at least two viral polymerases, a parental replicase and an encapsidase, and cellular factor ANP32. ANP32 comprises an LRR domain and a long C-terminal low complexity acidic region (LCAR). Here we present evidence suggesting that ANP32 is recruited to the replication complex as an electrostatic chaperone that stabilises the encapsidase moiety within apo-polymerase symmetric dimers that are distinct for influenza A and B polymerases. The ANP32 bound encapsidase, then forms the asymmetric replication complex with the replicase, which is embedded in a parental ribonucleoprotein particle (RNP). Cryo-EM structures reveal the architecture of the influenza A and B replication complexes and the likely trajectory of the nascent RNA product into the encapsidase. The cryo-EM map of the FluB replication complex shows extra density attributable to the ANP32 LCAR wrapping around and stabilising the apo-encapsidase conformation. These structures give new insight into the various mutations that adapt avian strain polymerases to use the distinct ANP32 in mammalian cells.


Subject(s)
Cryoelectron Microscopy , Influenza A virus , Molecular Chaperones , RNA-Binding Proteins , Static Electricity , Virus Replication , Humans , Animals , Molecular Chaperones/metabolism , Molecular Chaperones/chemistry , Molecular Chaperones/genetics , Influenza A virus/metabolism , Influenza A virus/genetics , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/genetics , Nuclear Proteins/metabolism , Nuclear Proteins/chemistry , RNA, Viral/metabolism , RNA, Viral/chemistry , RNA, Viral/genetics , Birds/virology , RNA-Dependent RNA Polymerase/metabolism , RNA-Dependent RNA Polymerase/chemistry , RNA-Dependent RNA Polymerase/genetics , Influenza in Birds/virology , Influenza in Birds/metabolism , Models, Molecular , Influenza, Human/virology
6.
J Virol ; 98(9): e0097524, 2024 Sep 17.
Article in English | MEDLINE | ID: mdl-39194242

ABSTRACT

Rotaviruses (RVs) are classified into nine species, A-D and F-J, with species A being the most studied. In rotavirus of species A (RVA), replication occurs in viroplasms, which are cytosolic globular inclusions composed of main building block proteins NSP5, NSP2, and VP2. The co-expression of NSP5 with either NSP2 or VP2 in uninfected cells leads to the formation of viroplasm-like structures (VLSs). Although morphologically identical to viroplasms, VLSs do not produce viral progeny but serve as excellent tools for studying complex viroplasms. A knowledge gap exists regarding non-RVA viroplasms due to the lack of specific antibodies and suitable cell culture systems. In this study, we explored the ability of NSP5 and NSP2 from non-RVA species to form VLSs. The co-expression of these two proteins led to globular VLSs in RV species A, B, D, F, G, and I, while RVC formed filamentous VLSs. The co-expression of NSP5 and NSP2 of RV species H and J did not result in VLS formation. Interestingly, NSP5 of all RV species self-oligomerizes, with the ordered C-terminal region, termed the tail, being necessary for self-oligomerization of RV species A-C and G-J. Except for NSP5 from RVJ, all NSP5 interacted with their cognate NSP2. We also found that interspecies VLS are formed between closely related RV species B with G and D with F. Additionally, VLS from RVH and RVJ formed when the tail of NSP5 RVH and RVJ was replaced by the tail of NSP5 from RVA and co-expressed with their respective NSP2. IMPORTANCE: Rotaviruses (RVs) are classified into nine species, A-D and F-J, infecting mammals and birds. Due to the lack of research tools, all cumulative knowledge on RV replication is based on RV species A (RVA). The RV replication compartments are globular cytosolic structures named viroplasms, which have only been identified in RV species A. In this study, we examined the formation of viroplasm-like structures (VLSs) by the co-expression of NSP5 with NSP2 across RV species A to J. Globular VLSs formed for RV species A, B, D, F, G, and I, while RV species C formed filamentous structures. The RV species H and J did not form VLS with their cognates NSP5 and NSP2. Similar to RVA, NSP5 self-oligomerizes in all RV species, which is required for VLS formation. This study provides basic knowledge of the non-RVA replication mechanisms, which could help develop strategies to halt virus infection across RV species.


Subject(s)
Rotavirus , Viral Nonstructural Proteins , Virus Replication , Rotavirus/genetics , Rotavirus/metabolism , Viral Nonstructural Proteins/metabolism , Viral Nonstructural Proteins/genetics , Animals , Humans , Capsid Proteins/genetics , Capsid Proteins/metabolism , Cell Line , RNA-Dependent RNA Polymerase/metabolism , RNA-Dependent RNA Polymerase/genetics , Rotavirus Infections/virology , RNA-Binding Proteins
7.
Sci Adv ; 10(34): eadq3087, 2024 Aug 23.
Article in English | MEDLINE | ID: mdl-39178250

ABSTRACT

RNA polymerase IV (Pol IV) forms a complex with RNA-directed RNA polymerase 2 (RDR2) to produce double-stranded RNA (dsRNA) precursors essential for plant gene silencing. In the "backtracking-triggered RNA channeling" model, Pol IV backtracks and delivers its transcript's 3' terminus to RDR2, which synthesizes dsRNA. However, the mechanisms underlying Pol IV backtracking and RNA protection from cleavage are unclear. Here, we determined cryo-electron microscopy structures of Pol IV elongation complexes at four states of its nucleotide addition cycle (NAC): posttranslocation, guanosine triphosphate-bound, pretranslocation, and backtracked states. The structures reveal that Pol IV maintains an open DNA cleft and kinked bridge helix in all NAC states, loosely interacts with the nucleoside triphosphate substrate, and barely contacts proximal backtracked nucleotides. Biochemical data indicate that Pol IV is inefficient in forward translocation and RNA cleavage. These findings suggest that Pol IV transcription elongation is prone to backtracking and incapable of RNA hydrolysis, ensuring efficient dsRNA production by Pol IV-RDR2.


Subject(s)
Cryoelectron Microscopy , DNA-Directed RNA Polymerases , DNA-Directed RNA Polymerases/metabolism , DNA-Directed RNA Polymerases/genetics , DNA-Directed RNA Polymerases/chemistry , Models, Molecular , Arabidopsis/genetics , Arabidopsis/metabolism , Arabidopsis Proteins/metabolism , Arabidopsis Proteins/genetics , Arabidopsis Proteins/chemistry , Transcription Elongation, Genetic , RNA-Dependent RNA Polymerase/metabolism , RNA-Dependent RNA Polymerase/genetics , RNA-Dependent RNA Polymerase/chemistry , RNA, Double-Stranded/metabolism , Protein Binding , Transcription, Genetic
8.
Eur J Med Chem ; 277: 116737, 2024 Nov 05.
Article in English | MEDLINE | ID: mdl-39153334

ABSTRACT

Influenza viruses (IV) are single-stranded RNA viruses with a negative-sense genome and have the potential to cause pandemics. While vaccines exist for influenza, their protection is only partial. Additionally, there is only a limited number of approved anti-IV drugs, which are associated to emergence of drug resistance. To address these issues, for years we have focused on the development of small-molecules that can interfere with the heterodimerization of PA and PB1 subunits of the IV RNA-dependent RNA polymerase (RdRP). In this study, starting from a cycloheptathiophene-3-carboxamide compound that we recently identified, we performed iterative cycles of medicinal chemistry optimization that led to the identification of compounds 43 and 45 with activity in the nanomolar range against circulating A and B strains of IV. Mechanistic studies demonstrated the ability of 43 and 45 to interfere with viral RdRP activity by disrupting PA-PB1 subunits heterodimerization and to bind to the PA C-terminal domain through biophysical assays. Most important, ADME studies of 45 also showed an improvement in the pharmacokinetic profile with respect to the starting hit.


Subject(s)
Antiviral Agents , RNA-Dependent RNA Polymerase , Antiviral Agents/pharmacology , Antiviral Agents/chemistry , Antiviral Agents/chemical synthesis , RNA-Dependent RNA Polymerase/antagonists & inhibitors , RNA-Dependent RNA Polymerase/metabolism , Humans , Animals , Structure-Activity Relationship , Viral Proteins/antagonists & inhibitors , Viral Proteins/metabolism , Viral Proteins/chemistry , Molecular Structure , Protein Multimerization/drug effects , Dose-Response Relationship, Drug , Influenza A virus/drug effects , Influenza A virus/enzymology , Microbial Sensitivity Tests , Dogs
9.
Viruses ; 16(8)2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39205212

ABSTRACT

Infections with Flaviviridae viruses, such as hepatitis C (HCV), dengue (DENV), and yellow fever (YFV) viruses, are major public health problems worldwide. In the case of HCV, treatment is associated with drug resistance and high costs, while there is no clinically approved therapy for DENV and YFV. Consequently, there is still a need for new chemotherapies with alternative modes of action. We have previously identified novel 2-hydroxypyrazino[1,2-a]indole-1,3(2H,4H)-diones as metal-chelating inhibitors targeting HCV RNA replication. Here, by utilizing a structure-based approach, we rationally designed a second series of compounds by introducing various substituents at the indole core structure and at the imidic nitrogen, to improve specificity against the RNA-dependent RNA polymerase (RdRp). The resulting derivatives were evaluated for their potency against HCV genotype 1b, DENV2, and YFV-17D using stable replicon cell lines. The most favorable substitution was nitro at position 6 of the indole ring (compound 36), conferring EC50 1.6 µM against HCV 1b and 2.57 µΜ against HCV 1a, with a high selectivity index. Compound 52, carrying the acetohydroxamic acid functionality (-CH2CONHOH) on the imidic nitrogen, and compound 78, the methyl-substituted molecule at the position 4 indolediketopiperazine counterpart, were the most effective against DENV and YFV, respectively. Interestingly, compound 36 had a high genetic barrier to resistance and only one resistance mutation was detected, T181I in NS5B, suggesting that the compound target HCV RdRp is in accordance with our predicted model.


Subject(s)
Antiviral Agents , Hepacivirus , Indoles , Virus Replication , Virus Replication/drug effects , Antiviral Agents/pharmacology , Antiviral Agents/chemistry , Humans , Hepacivirus/drug effects , Hepacivirus/genetics , Hepacivirus/physiology , Indoles/pharmacology , Indoles/chemistry , RNA-Dependent RNA Polymerase/antagonists & inhibitors , RNA-Dependent RNA Polymerase/metabolism , Viral Nonstructural Proteins/metabolism , Viral Nonstructural Proteins/antagonists & inhibitors , Viral Nonstructural Proteins/genetics , Cell Line , Flaviviridae/drug effects , Flaviviridae/genetics , Structure-Activity Relationship , Dengue Virus/drug effects , Dengue Virus/genetics , Yellow fever virus/drug effects , Yellow fever virus/genetics
10.
PLoS One ; 19(8): e0307615, 2024.
Article in English | MEDLINE | ID: mdl-39102385

ABSTRACT

Viral diseases pose a serious global health threat due to their rapid transmission and widespread impact. The RNA-dependent RNA polymerase (RdRp) participates in the synthesis, transcription, and replication of viral RNA in host. The current study investigates the antiviral potential of secondary metabolites particularly those derived from bacteria, fungi, and plants to develop novel medicines. Using a virtual screening approach that combines molecular docking and molecular dynamics (MD) simulations, we aimed to discover compounds with strong interactions with RdRp of five different retroviruses. The top five compounds were selected for each viral RdRp based on their docking scores, binding patterns, molecular interactions, and drug-likeness properties. The molecular docking study uncovered several metabolites with antiviral activity against RdRp. For instance, cytochalasin Z8 had the lowest docking score of -8.9 (kcal/mol) against RdRp of SARS-CoV-2, aspulvinone D (-9.2 kcal/mol) against HIV-1, talaromyolide D (-9.9 kcal/mol) for hepatitis C, aspulvinone D (-9.9 kcal/mol) against Ebola and talaromyolide D also maintained the lowest docking score of -9.2 kcal/mol against RdRp enzyme of dengue virus. These compounds showed remarkable antiviral potential comparable to standard drug (remdesivir -7.4 kcal/mol) approved to target RdRp and possess no significant toxicity. The molecular dynamics simulation confirmed that the best selected ligands were firmly bound to their respective target proteins for a simulation time of 200 ns. The identified lead compounds possess distinctive pharmacological characteristics, making them potential candidates for repurposing as antiviral drugs against SARS-CoV-2. Further experimental evaluation and investigation are recommended to ascertain their efficacy and potential.


Subject(s)
Antiviral Agents , Molecular Docking Simulation , Molecular Dynamics Simulation , RNA-Dependent RNA Polymerase , Antiviral Agents/pharmacology , Antiviral Agents/chemistry , Antiviral Agents/metabolism , RNA-Dependent RNA Polymerase/metabolism , RNA-Dependent RNA Polymerase/chemistry , RNA-Dependent RNA Polymerase/antagonists & inhibitors , SARS-CoV-2/drug effects , SARS-CoV-2/enzymology , Humans , HIV-1/drug effects , HIV-1/enzymology , Secondary Metabolism , Biological Products/chemistry , Biological Products/pharmacology , Biological Products/metabolism
11.
Virology ; 598: 110189, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39089051

ABSTRACT

Bovine viral diarrhea virus (BVDV) is a widespread pathogen of cattle and other mammals that causes major economic losses in the livestock industry. N4-TSC and 6NO2-TSC are two thiosemicarbazones derived from 1-indanone that exhibit anti-BVDV activity in vitro. These compounds selectively inhibit BVDV and are effective against both cytopathic and non-cytopathic BVDV-1 and BVDV-2 strains. We confirmed that N4-TSC acts at the onset of viral RNA synthesis, as previously reported for 6NO2-TSC. Moreover, resistance selection and characterization showed that N4-TSCR mutants were highly resistant to N4-TSC but remained susceptible to 6NO2-TSC. In contrast, 6NO2-TSCR mutants were resistant to both compounds. Additionally, mutations N264D and A392E were found in the viral RNA-dependent RNA polymerase (RdRp) of N4-TSCR mutants, whereas I261 M was found in 6NO2-TSCR mutants. These mutations lay in a hydrophobic pocket within the fingertips region of BVDV RdRp that has been described as a "hot spot" for BVDV non-nucleoside inhibitors.


Subject(s)
Antiviral Agents , Drug Resistance, Viral , Genotype , Indans , Thiosemicarbazones , Antiviral Agents/pharmacology , Antiviral Agents/chemistry , Animals , Cattle , Thiosemicarbazones/pharmacology , Thiosemicarbazones/chemistry , Indans/pharmacology , Indans/chemistry , Drug Resistance, Viral/genetics , Diarrhea Virus 1, Bovine Viral/drug effects , Diarrhea Virus 1, Bovine Viral/genetics , Diarrhea Viruses, Bovine Viral/drug effects , Diarrhea Viruses, Bovine Viral/genetics , Cell Line , RNA-Dependent RNA Polymerase/genetics , RNA-Dependent RNA Polymerase/antagonists & inhibitors , RNA-Dependent RNA Polymerase/metabolism , Diarrhea Virus 2, Bovine Viral/genetics , Diarrhea Virus 2, Bovine Viral/drug effects , Virus Replication/drug effects , Mutation , RNA, Viral/genetics
12.
Emerg Microbes Infect ; 13(1): 2387439, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39139051

ABSTRACT

Avian influenza viruses (AIVs) are the origin of multiple mammal influenza viruses. The genetic determinants of AIVs adapted to humans have been widely elucidated, however, the molecular mechanism of cross-species transmission and adaptation of AIVs to canines are still poorly understood. In this study, two H3N2 influenza viruses isolated from a live poultry market (A/environment/Guangxi/13431/2018, GX13431) and a swab sample from a canine (A/canine/Guangdong/0601/2019, GD0601) were used to investigate the possible molecular basis that determined H3N2 AIV adapting to canine. We found that GD0601 exhibited more robust polymerase activity in cells and higher pathogenicity in mice compared with its evolution ancestor H3N2 AIV GX13431. A series of reassortments of the ribonucleoprotein (RNP) complex showed that the PB2 subunit was the crucial factor that conferred high polymerase activity of GD0601, and the substitution of I714S in the PB2 subunit of GD0601 attenuated the replication and pathogenicity in mammal cells and the mouse model. Mechanistically, the reverse mutation of I714S in the PB2 polymerase subunit which was identified in AIV GX13431 reduced the nuclear import efficiency of PB2 protein and interfered with the interactions of PB2-PA/NP that affected the assembly of the viral RNP complex. Our study reveals amino acid mutation at the position of 714 in the nuclear localization signal (NLS) area in PB2 plays an important role in overcoming the barrier from poultry to mammals of the H3N2 canine influenza virus and provides clues for further study of mammalian adaptation mechanism of AIVs.


Subject(s)
Influenza A Virus, H3N2 Subtype , Orthomyxoviridae Infections , RNA-Dependent RNA Polymerase , Ribonucleoproteins , Viral Proteins , Animals , Dogs , Influenza A Virus, H3N2 Subtype/genetics , Influenza A Virus, H3N2 Subtype/physiology , Mice , Viral Proteins/genetics , Viral Proteins/metabolism , RNA-Dependent RNA Polymerase/genetics , RNA-Dependent RNA Polymerase/metabolism , Orthomyxoviridae Infections/virology , Humans , Ribonucleoproteins/genetics , Ribonucleoproteins/metabolism , Active Transport, Cell Nucleus , Virus Replication , Mutation , Madin Darby Canine Kidney Cells , Dog Diseases/virology , Mice, Inbred BALB C , HEK293 Cells , Reassortant Viruses/genetics
13.
Protein Sci ; 33(9): e5103, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39145418

ABSTRACT

Since the emergence of SARS-CoV-2, mutations in all subunits of the RNA-dependent RNA polymerase (RdRp) of the virus have been repeatedly reported. Although RdRp represents a primary target for antiviral drugs, experimental studies exploring the phenotypic effect of these mutations have been limited. This study focuses on the phenotypic effects of substitutions in the three RdRp subunits: nsp7, nsp8, and nsp12, selected based on their occurrence rate and potential impact. We employed nano-differential scanning fluorimetry and microscale thermophoresis to examine the impact of these mutations on protein stability and RdRp complex assembly. We observed diverse impacts; notably, a single mutation in nsp8 significantly increased its stability as evidenced by a 13°C increase in melting temperature, whereas certain mutations in nsp7 and nsp8 reduced their binding affinity to nsp12 during RdRp complex formation. Using a fluorometric enzymatic assay, we assessed the overall effect on RNA polymerase activity. We found that most of the examined mutations altered the polymerase activity, often as a direct result of changes in stability or affinity to the other components of the RdRp complex. Intriguingly, a combination of nsp8 A21V and nsp12 P323L mutations resulted in a 50% increase in polymerase activity. To our knowledge, this is the first biochemical study to demonstrate the impact of amino acid mutations across all components constituting the RdRp complex in emerging SARS-CoV-2 subvariants.


Subject(s)
Coronavirus RNA-Dependent RNA Polymerase , Mutation , SARS-CoV-2 , Viral Nonstructural Proteins , SARS-CoV-2/genetics , SARS-CoV-2/enzymology , Coronavirus RNA-Dependent RNA Polymerase/genetics , Coronavirus RNA-Dependent RNA Polymerase/metabolism , Coronavirus RNA-Dependent RNA Polymerase/chemistry , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/metabolism , Humans , COVID-19/virology , RNA-Dependent RNA Polymerase/genetics , RNA-Dependent RNA Polymerase/chemistry , RNA-Dependent RNA Polymerase/metabolism , Protein Stability , Protein Binding
14.
Nucleic Acids Res ; 52(16): 9727-9744, 2024 Sep 09.
Article in English | MEDLINE | ID: mdl-39051569

ABSTRACT

Chikungunya virus (CHIKV) is a rapidly spreading re-emergent virus transmitted from mosquitoes to humans. The emergence of epidemic variants has been associated with changes in the viral genome, such as the duplication of repeated sequences in the 3' untranslated region (UTR). Indeed, blocks of repeated sequences seemingly favor RNA recombination, providing the virus with a unique ability to continuously change the 3'UTR architecture during host switching. In this work, we provide experimental data on the molecular mechanism of RNA recombination and describe specific sequence and structural elements in the viral 3'UTR that favor template switching of the viral RNA-dependent RNA polymerase on the 3'UTR. Furthermore, we found that a 3'UTR deletion mutant that exhibits markedly delayed replication in mosquito cells and impaired transmission in vivo, recombines in reference laboratory strains of mosquitoes. Altogether, our data provide novel experimental evidence indicating that RNA recombination can act as a nucleic acid repair mechanism to add repeated sequences that are associated to high viral fitness in mosquito during chikungunya virus replication.


Subject(s)
3' Untranslated Regions , Chikungunya virus , Genome, Viral , RNA, Viral , Recombination, Genetic , Virus Replication , Chikungunya virus/genetics , 3' Untranslated Regions/genetics , RNA, Viral/genetics , RNA, Viral/metabolism , Animals , Virus Replication/genetics , Chikungunya Fever/virology , Chikungunya Fever/genetics , Chikungunya Fever/transmission , Humans , Aedes/virology , Aedes/genetics , RNA-Dependent RNA Polymerase/genetics , RNA-Dependent RNA Polymerase/metabolism , Cell Line
15.
Nucleic Acids Res ; 52(16): 9803-9820, 2024 Sep 09.
Article in English | MEDLINE | ID: mdl-39077935

ABSTRACT

Non-segmented negative-strand (NNS) RNA viruses, such as rabies, Nipah and Ebola, produce 5'-capped and 3'-polyadenylated mRNAs resembling higher eukaryotic mRNAs. Here, we developed a transcription elongation-coupled pre-mRNA capping system for vesicular stomatitis virus (VSV, a prototypic NNS RNA virus). Using this system, we demonstrate that the single-polypeptide RNA-dependent RNA polymerase (RdRp) large protein (L) catalyzes all pre-mRNA modifications co-transcriptionally in the following order: (i) 5'-capping (polyribonucleotidylation of GDP) to form a GpppA cap core structure, (ii) 2'-O-methylation of GpppA into GpppAm, (iii) guanine-N7-methylation of GpppAm into m7GpppAm (cap 1), (iv) 3'-polyadenylation to yield a poly(A) tail. The GDP polyribonucleotidyltransferase (PRNTase) domain of L generated capped pre-mRNAs of 18 nucleotides or longer via the formation of covalent enzyme-pre-mRNA intermediates. The single methyltransferase domain of L sequentially methylated the cap structure only when pre-mRNAs of 40 nucleotides or longer were associated with elongation complexes. These results suggest that the formation of pre-mRNA closed loop structures in elongation complexes via the RdRp and PRNTase domains followed by the RdRp and MTase domains on the same polypeptide is required for the cap 1 formation during transcription. Taken together, our findings indicate that NNS RNA virus L acts as an all-in-one viral mRNA assembly machinery.


Subject(s)
RNA Caps , RNA, Messenger , RNA, Viral , Viral Proteins , RNA, Messenger/metabolism , RNA, Messenger/genetics , RNA Caps/metabolism , RNA Caps/genetics , RNA, Viral/metabolism , RNA, Viral/genetics , RNA, Viral/chemistry , Viral Proteins/metabolism , Viral Proteins/genetics , RNA-Dependent RNA Polymerase/metabolism , RNA-Dependent RNA Polymerase/genetics , Methylation , Vesiculovirus/genetics , RNA Precursors/metabolism , RNA Precursors/genetics , Transcription, Genetic , RNA Processing, Post-Transcriptional , Animals , Polyadenylation , Transcription Elongation, Genetic
16.
Eur J Med Chem ; 276: 116671, 2024 Oct 05.
Article in English | MEDLINE | ID: mdl-39004019

ABSTRACT

COVID-19 caused major worldwide problems. The spread of variants and limited treatment encouraged the design of novel anti-SARS-CoV-2 compounds. A series of compounds RH1-23 were designed to dually target RNA-dependent RNA polymerase (RdRp) and transmembrane serine protease 2 (TMPRSS2). Compared to remdesivir, in vitro screening indicated the highest selectivity and potent activity of RH11-13 with half maximum inhibitory concentration (IC50) 3.9, 5.7, and 19.72 nM, respectively. RH11-12 showed superior inhibition activity against TMPRSS2 and RdRP with IC50 (1.7 and 4.2), and (6.1 and 4.42) nM, respectively. WaterMap analysis and molecular dynamics studies demonstrated the superior enzyme binding activity of RH11 and RH12. On Vero-E6 cells, RH11 and RH12 significantly inhibited the viral replication with 66 % and 63.2 %, and viral adsorption with 44 % and 65 %, alongside virucidal effect with 51.40 % and 90.5 %, respectively. Furthermore, the potent activity of RH12 was tested on TMPRSS2-expressing cells (Calu-3) compared to camostat. RH12 exhibited selectivity index (26.05) similar to camostat (28.01) and comparable to its SI on Vero-E6 cells (22.6). RH12 demonstrated also a significant inhibition of the viral adsorption on Calu-3 cells with 60 % inhibition at 30 nM. The designed compounds exhibited good physiochemical properties. These findings indicate a broad-spectrum antiviral efficacy of the designed compounds, particularly RH12, with a promise for further development.


Subject(s)
Antiviral Agents , SARS-CoV-2 , Serine Endopeptidases , Serine Endopeptidases/metabolism , Humans , SARS-CoV-2/drug effects , SARS-CoV-2/enzymology , Antiviral Agents/pharmacology , Antiviral Agents/chemistry , Antiviral Agents/chemical synthesis , Chlorocebus aethiops , Animals , Vero Cells , Virus Replication/drug effects , COVID-19 Drug Treatment , Structure-Activity Relationship , RNA-Dependent RNA Polymerase/antagonists & inhibitors , RNA-Dependent RNA Polymerase/metabolism , Dose-Response Relationship, Drug , Molecular Structure , Molecular Dynamics Simulation
17.
J Virol ; 98(8): e0033224, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-39078194

ABSTRACT

The Mononegavirales, or non-segmented negative-sense RNA viruses (nsNSVs), includes significant human pathogens, such as respiratory syncytial virus, parainfluenza virus, measles virus, Ebola virus, and rabies virus. Although these viruses differ widely in their pathogenic properties, they are united by each having a genome consisting of a single strand of negative-sense RNA. Consistent with their shared genome structure, the nsNSVs have evolved similar ways to transcribe their genome into mRNAs and replicate it to produce new genomes. Importantly, both mRNA transcription and genome replication are performed by a single virus-encoded polymerase. A fundamental and intriguing question is: how does the nsNSV polymerase commit to being either an mRNA transcriptase or a replicase? The polymerase must become committed to one process or the other either before it interacts with the genome template or in its initial interactions with the promoter sequence at the 3´ end of the genomic RNA. This review examines the biochemical, molecular biology, and structural biology data regarding the first steps of transcription and RNA replication that have been gathered over several decades for different families of nsNSVs. These findings are discussed in relation to possible models that could explain how an nsNSV polymerase initiates and commits to either transcription or genome replication.


Subject(s)
Genome, Viral , RNA, Viral , Virus Replication , Humans , RNA, Viral/genetics , RNA, Viral/metabolism , Viral Transcription/genetics , Negative-Sense RNA Viruses/genetics , Negative-Sense RNA Viruses/metabolism , Transcription, Genetic , Viral Proteins/metabolism , Viral Proteins/genetics , RNA-Dependent RNA Polymerase/metabolism , RNA-Dependent RNA Polymerase/genetics
18.
Nat Commun ; 15(1): 5799, 2024 Jul 10.
Article in English | MEDLINE | ID: mdl-38987544

ABSTRACT

Germ granules are biomolecular condensates present in most animal germ cells. One function of germ granules is to help maintain germ cell totipotency by organizing mRNA regulatory machinery, including small RNA-based gene regulatory pathways. The C. elegans germ granule is compartmentalized into multiple subcompartments whose biological functions are largely unknown. Here, we identify an uncharted subcompartment of the C. elegans germ granule, which we term the E granule. The E granule is nonrandomly positioned within the germ granule. We identify five proteins that localize to the E granule, including the RNA-dependent RNA polymerase (RdRP) EGO-1, the Dicer-related helicase DRH-3, the Tudor domain-containing protein EKL-1, and two intrinsically disordered proteins, EGC-1 and ELLI-1. Localization of EGO-1 to the E granule enables synthesis of a specialized class of 22G RNAs, which derive exclusively from 5' regions of a subset of germline-expressed mRNAs. Defects in E granule assembly elicit disordered production of endogenous siRNAs, which disturbs fertility and the RNAi response. Our results define a distinct subcompartment of the C. elegans germ granule and suggest that one function of germ granule compartmentalization is to facilitate the localized production of specialized classes of small regulatory RNAs.


Subject(s)
Caenorhabditis elegans Proteins , Caenorhabditis elegans , Cytoplasmic Granules , Germ Cells , Caenorhabditis elegans/metabolism , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans Proteins/genetics , Animals , Germ Cells/metabolism , Cytoplasmic Granules/metabolism , RNA, Messenger/metabolism , RNA, Messenger/genetics , DEAD-box RNA Helicases/metabolism , DEAD-box RNA Helicases/genetics , RNA-Dependent RNA Polymerase/metabolism , RNA-Dependent RNA Polymerase/genetics , Intrinsically Disordered Proteins/metabolism , Intrinsically Disordered Proteins/genetics
19.
Viruses ; 16(7)2024 Jul 14.
Article in English | MEDLINE | ID: mdl-39066290

ABSTRACT

Foot-and-mouth disease virus (FMDV) belongs to the Picornaviridae family and is an important pathogen affecting cloven-hoof livestock. However, neither effective vaccines covering all serotypes nor specific antivirals against FMDV infections are currently available. In this study, we employed virtual screening to screen for secondary metabolite terpenoids targeting the RNA-dependent RNA polymerase (RdRp), or 3Dpol, of FMDV. Subsequently, we identified the potential antiviral activity of the 32 top-ranked terpenoids, revealing that continentalic acid, dehydroabietic acid (abietic diterpenoids), brusatol, bruceine D, and bruceine E (tetracyclic triterpenoids) significantly reduced cytopathic effects and viral infection in the terpenoid-treated, FMDV-infected BHK-21 cells in a dose-dependent manner, with nanomolar to low micromolar levels. The FMDV minigenome assay demonstrated that brusatol and bruceine D, in particular, effectively blocked FMDV 3Dpol activity, exhibiting IC50 values in the range of 0.37-0.39 µM and surpassing the efficacy of the antiviral drug control, ribavirin. Continentalic acid and bruceine E exhibited moderate inhibition of FMDV 3Dpol. The predicted protein-ligand interaction confirmed that these potential terpenoids interacted with the main catalytic and bystander residues of FMDV 3Dpol. Additionally, brusatol and bruceine D exhibited additive effects when combined with ribavirin. In conclusion, terpenoids from natural resources show promise for the development of anti-FMD agents.


Subject(s)
Antiviral Agents , Foot-and-Mouth Disease Virus , Terpenes , Foot-and-Mouth Disease Virus/drug effects , Antiviral Agents/pharmacology , Antiviral Agents/chemistry , Animals , Terpenes/pharmacology , Terpenes/chemistry , Cell Line , Virus Replication/drug effects , Computer Simulation , RNA-Dependent RNA Polymerase/metabolism , RNA-Dependent RNA Polymerase/antagonists & inhibitors , Cricetinae , Molecular Docking Simulation , Foot-and-Mouth Disease/virology , Foot-and-Mouth Disease/drug therapy , Diterpenes/pharmacology , Diterpenes/chemistry
20.
Proc Natl Acad Sci U S A ; 121(29): e2317977121, 2024 Jul 16.
Article in English | MEDLINE | ID: mdl-38990941

ABSTRACT

In a recent characterization of the Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) variability present in 30 diagnostic samples from patients of the first COVID-19 pandemic wave, 41 amino acid substitutions were documented in the RNA-dependent RNA polymerase (RdRp) nsp12. Eight substitutions were selected in this work to determine whether they had an impact on the RdRp activity of the SARS-CoV-2 nsp12-nsp8-nsp7 replication complex. Three of these substitutions were found around the polymerase central cavity, in the template entry channel (D499G and M668V), and within the motif B (V560A), and they showed polymerization rates similar to the wild type RdRp. The remaining five mutations (P323L, L372F, L372P, V373A, and L527H) were placed near the nsp12-nsp8F contact surface; residues L372, V373, and L527 participated in a large hydrophobic cluster involving contacts between two helices in the nsp12 fingers and the long α-helix of nsp8F. The presence of any of these five amino acid substitutions resulted in important alterations in the RNA polymerization activity. Comparative primer elongation assays showed different behavior depending on the hydrophobicity of their side chains. The substitution of L by the bulkier F side chain at position 372 slightly promoted RdRp activity. However, this activity was dramatically reduced with the L372P, and L527H mutations, and to a lesser extent with V373A, all of which weaken the hydrophobic interactions within the cluster. Additional mutations, specifically designed to disrupt the nsp12-nsp8F interactions (nsp12-V330S, nsp12-V341S, and nsp8-R111A/D112A), also resulted in an impaired RdRp activity, further illustrating the importance of this contact interface in the regulation of RNA synthesis.


Subject(s)
Point Mutation , RNA, Viral , SARS-CoV-2 , Viral Nonstructural Proteins , SARS-CoV-2/genetics , SARS-CoV-2/metabolism , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/metabolism , Viral Nonstructural Proteins/chemistry , RNA, Viral/genetics , RNA, Viral/metabolism , Humans , Coronavirus RNA-Dependent RNA Polymerase/genetics , Coronavirus RNA-Dependent RNA Polymerase/metabolism , Polymerization , COVID-19/virology , Amino Acid Substitution , RNA-Dependent RNA Polymerase/genetics , RNA-Dependent RNA Polymerase/metabolism , Models, Molecular
SELECTION OF CITATIONS
SEARCH DETAIL