34 results on '"Querat G"'
Search Results
2. Identification and subcellular localization of the Q gene product of visna virus
- Author
-
Audoly, G., primary, Sauze, N., additional, Harkiss, G., additional, Vitu, C., additional, Russo, P., additional, Querat, G., additional, Suzan, M., additional, and Vigne, R., additional
- Published
- 1992
- Full Text
- View/download PDF
3. Nucleotide sequence of the jaagsiekte retrovirus, an exogenous and endogenous type D and B retrovirus of sheep and goats
- Author
-
York, D F, primary, Vigne, R, additional, Verwoerd, D W, additional, and Querat, G, additional
- Published
- 1992
- Full Text
- View/download PDF
4. Isolation, identification, and partial cDNA cloning of genomic RNA of jaagsiekte retrovirus, the etiological agent of sheep pulmonary adenomatosis
- Author
-
York, D F, primary, Vigne, R, additional, Verwoerd, D W, additional, and Querat, G, additional
- Published
- 1991
- Full Text
- View/download PDF
5. Induction of NF-KB during monocyte differentiation by HIV type 1 infection.
- Author
-
Suzan, M, primary, Salaun, D, additional, Neuveut, C, additional, Spire, B, additional, Hirsch, I, additional, Le Bouteiller, P, additional, Querat, G, additional, and Sire, J, additional
- Published
- 1991
- Full Text
- View/download PDF
6. Nucleotide sequence analysis of SA-OMVV, a visna-related ovine lentivirus: phylogenetic history of lentiviruses
- Author
-
Querat, G., primary, Audoly, G., additional, Sonigo, P., additional, and Vigne, R., additional
- Published
- 1990
- Full Text
- View/download PDF
7. Induction of NF-KB during monocyte differentiation by HIV type 1 infection
- Author
-
Suzan, M., Salaun, D., Neuveut, C., Spire, B., Ivan Hirsch, Le Bouteiller, P., Querat, G., and Sire, J.
- Subjects
Immunology ,Immunology and Allergy - Abstract
The production of human immunodeficiency virus type 1 (HIV-1) progeny was followed in the U937 promonocytic cell line after stimulation either with retinoic acid or PMA, and in purified human monocytes and macrophages. Electrophoretic mobility shift assays and Southwestern blotting experiments were used to detect the binding of cellular transactivation factor NF-KB to the double repeat-KB enhancer sequence located in the long terminal repeat. PMA treatment, and not retinoic acid treatment of the U937 cells acts in inducing NF-KB expression in the nuclei. In nuclear extracts from monocytes or macrophages, induction of NF-KB occurred only if the cells were previously infected with HIV-1. When U937 cells were infected with HIV-1, no induction of NF-KB factor was detected, whereas high level of progeny virions was produced, suggesting that this factor was not required for viral replication. These results indicate that in monocytic cell lineage, HIV-1 could mimic some differentiation/activation stimuli allowing nuclear NF-KB expression.
8. Une technique ELISA pour la detection des anticorps anti-virus maedi-visna. Etude comparative avec l'immunodiffusion en gelose et la fixation du complement
- Author
-
Vitu, C, primary, Russo, P, additional, Filippi, P, additional, Vigne, R, additional, Querat, G, additional, and Giauffret, A, additional
- Published
- 1982
- Full Text
- View/download PDF
9. Uracil within DNA: an actor of antiviral immunity
- Author
-
Priet Stéphane, Esnault Cécile, Quérat Gilles, and Sire Joséphine
- Subjects
Immunologic diseases. Allergy ,RC581-607 - Abstract
Abstract Uracil is a natural base of RNA but may appear in DNA through two different pathways including cytosine deamination or misincorporation of deoxyuridine 5'-triphosphate nucleotide (dUTP) during DNA replication and constitutes one of the most frequent DNA lesions. In cellular organisms, such lesions are faithfully cleared out through several universal DNA repair mechanisms, thus preventing genome injury. However, several recent studies have brought some pieces of evidence that introduction of uracil bases in viral genomic DNA intermediates during genome replication might be a way of innate immune defence against some viruses. As part of countermeasures, numerous viruses have developed powerful strategies to prevent emergence of uracilated viral genomes and/or to eliminate uracils already incorporated into DNA. This review will present the current knowledge about the cellular and viral countermeasures against uracils in DNA and the implications of these uracils as weapons against viruses.
- Published
- 2008
- Full Text
- View/download PDF
10. HIV1 infection of human monocytes and macrophages promotes induction or translocation of NF-KB-related factors
- Author
-
Neuveut, C, Suzan, M, Querat, G, and Sire, J
- Published
- 1991
- Full Text
- View/download PDF
11. Characteristics of a novel lentivirus derived from South African sheep with pulmonary adenocarcinoma (jaagsiekte)
- Author
-
Quérat, G., Barban, V., Sauze, N., Vigne, R., Payne, A., York, D., de Villiers, E.M., and Verwoerd, D.W.
- Published
- 1987
- Full Text
- View/download PDF
12. Transcription of visna virus during its lytic cycle: Evidence for a sequential early and late gene expression
- Author
-
Vigne, R., Barban, V., Quérat, G., Mazarin, V., Gourdou, I., and Sauze, N.
- Published
- 1987
- Full Text
- View/download PDF
13. Broad spectrum anti-flavivirus pyridobenzothiazolones leading to less infective virions
- Author
-
Rolando Cannalire, Mario Milani, Giuseppe Manfroni, Géraldine Piorkowski, Serena Massari, Violetta Cecchetti, Maria Letizia Barreca, Tommaso Felicetti, Gilles Querat, Delia Tarantino, Tea Carletti, Alessandro Marcello, Stefano Sabatini, Oriana Tabarrini, Eloise Mastrangelo, Cannalire, Rolando, Tarantino, D., Piorkowski, G., Carletti, T., Massari, S., Felicetti, T., Barreca, M. L., Sabatini, S., Tabarrini, O., Marcello, A., Milani, M., Cecchetti, V., Mastrangelo, E., Manfroni, G., Querat, G., Università degli Studi di Perugia = University of Perugia (UNIPG), Sezione di Fisiologia e Biochimica delle Piante, Dipartimento di Biologia, Università degli Studi di Milano = University of Milan (UNIMI), Unité des Virus Emergents (UVE), Institut de Recherche pour le Développement (IRD)-Aix Marseille Université (AMU)-Institut National de la Santé et de la Recherche Médicale (INSERM), Laboratory of Molecular Virology, International Centre for Genetic Engineering and Biotechnology (ICGEB), Dipartimento di Biotecnologie e Bioscienze (Università di Milano-Bicocca), Université de Milan, Istituto di Biofisica del CNR, Emergence des Pathologies Virales (EPV), Institut de Recherche pour le Développement (IRD)-Aix Marseille Université (AMU)-Assistance Publique - Hôpitaux de Marseille (APHM)-Institut National de la Santé et de la Recherche Médicale (INSERM), Università degli Studi di Perugia (UNIPG), Università degli Studi di Milano [Milano] (UNIMI), Aix Marseille Université (AMU)-Institut de Recherche pour le Développement (IRD)-Institut National de la Santé et de la Recherche Médicale (INSERM), Institut National de la Santé et de la Recherche Médicale (INSERM)-Institut de Recherche pour le Développement (IRD)-Aix Marseille Université (AMU)-Assistance Publique - Hôpitaux de Marseille (APHM), and HAL AMU, Administrateur
- Subjects
0301 basic medicine ,Pyridines ,viruses ,[SDV]Life Sciences [q-bio] ,Flavivirus replication ,Virus Replication ,Dengue fever ,Zika virus ,Antiviral small molecules ,Flavivirus inhibitor ,Flavivirus inhibitors ,ComputingMilieux_MISCELLANEOUS ,[SDV.MP.VIR] Life Sciences [q-bio]/Microbiology and Parasitology/Virology ,Infectivity ,biology ,Yellow fever ,Antivirals ,3. Good health ,[SDV] Life Sciences [q-bio] ,Flavivirus ,Antiviral small molecule ,[SDV.MP]Life Sciences [q-bio]/Microbiology and Parasitology ,Flavivirus inhibitors Antiviral small molecules Flavivirus replication Antivirals Dengue inhibitors Zika virus ,[SDV.MP.VIR]Life Sciences [q-bio]/Microbiology and Parasitology/Virology ,RNA, Viral ,Yellow fever virus ,Dengue inhibitor ,West Nile virus ,Dengue inhibitors ,030106 microbiology ,Antiviral Agents ,Encephalitis Viruses, Tick-Borne ,03 medical and health sciences ,Virology ,medicine ,Encephalitis Viruses ,Animals ,Humans ,Antiviral ,[SDV.MP] Life Sciences [q-bio]/Microbiology and Parasitology ,Virus classification ,Oxazocines ,Pharmacology ,Flaviviridae ,Virion ,Dengue Virus ,Japanese encephalitis ,medicine.disease ,biology.organism_classification ,030104 developmental biology - Abstract
We report the design, synthesis, and biological evaluation of a class of 1H-pyrido[2,1-b][1,3]benzothiazol-1-ones originated from compound 1, previously identified as anti-flavivirus agent. Some of the new compounds showed activity in low ?M range with reasonable selectivity against Dengue 2, Yellow fever (Bolivia strain), and West Nile viruses. One of the most interesting molecules, compound 16, showed broad antiviral activity against additional flaviviruses such as Dengue 1, 3 and 4, Zika, Japanese encephalitis, several strains of Yellow fever, and tick-borne encephalitis viruses. Compound 16 did not exert any effect on alphaviruses and phleboviruses and its activity was maintained in YFV infected cells from different species. The activity of 16 appears specific for flavivirus with respect to other virus families, suggesting, but not proving, that it might be targeting a viral factor. We demonstrated that the antiviral effect of 16 is not related to reduced viral RNA synthesis or virion release. On the contrary, viral particles grown in the presence of 16 showed reduced infectivity, being unable to perform a second round of infection. The chemical class herein presented thus emerges as suitable to provide pan-flavivirus inhibitors.
- Published
- 2019
- Full Text
- View/download PDF
14. Blocking NS3-NS4B interaction inhibits dengue virus in non-human primates.
- Author
-
Goethals O, Kaptein SJF, Kesteleyn B, Bonfanti JF, Van Wesenbeeck L, Bardiot D, Verschoor EJ, Verstrepen BE, Fagrouch Z, Putnak JR, Kiemel D, Ackaert O, Straetemans R, Lachau-Durand S, Geluykens P, Crabbe M, Thys K, Stoops B, Lenz O, Tambuyzer L, De Meyer S, Dallmeier K, McCracken MK, Gromowski GD, Rutvisuttinunt W, Jarman RG, Karasavvas N, Touret F, Querat G, de Lamballerie X, Chatel-Chaix L, Milligan GN, Beasley DWC, Bourne N, Barrett ADT, Marchand A, Jonckers THM, Raboisson P, Simmen K, Chaltin P, Bartenschlager R, Bogers WM, Neyts J, and Van Loock M
- Subjects
- Animals, Humans, Mice, Clinical Trials, Phase I as Topic, Dose-Response Relationship, Drug, Drug Resistance, Viral, In Vitro Techniques, Molecular Targeted Therapy, Protein Binding drug effects, Virus Replication, Antiviral Agents adverse effects, Antiviral Agents pharmacology, Antiviral Agents therapeutic use, Dengue drug therapy, Dengue prevention & control, Dengue virology, Dengue Virus classification, Dengue Virus drug effects, Primates virology, Viral Nonstructural Proteins antagonists & inhibitors, Viral Nonstructural Proteins metabolism
- Abstract
Dengue is a major health threat and the number of symptomatic infections caused by the four dengue serotypes is estimated to be 96 million
1 with annually around 10,000 deaths2 . However, no antiviral drugs are available for the treatment or prophylaxis of dengue. We recently described the interaction between non-structural proteins NS3 and NS4B as a promising target for the development of pan-serotype dengue virus (DENV) inhibitors3 . Here we present JNJ-1802-a highly potent DENV inhibitor that blocks the NS3-NS4B interaction within the viral replication complex. JNJ-1802 exerts picomolar to low nanomolar in vitro antiviral activity, a high barrier to resistance and potent in vivo efficacy in mice against infection with any of the four DENV serotypes. Finally, we demonstrate that the small-molecule inhibitor JNJ-1802 is highly effective against viral infection with DENV-1 or DENV-2 in non-human primates. JNJ-1802 has successfully completed a phase I first-in-human clinical study in healthy volunteers and was found to be safe and well tolerated4 . These findings support the further clinical development of JNJ-1802, a first-in-class antiviral agent against dengue, which is now progressing in clinical studies for the prevention and treatment of dengue., (© 2023. The Author(s).)- Published
- 2023
- Full Text
- View/download PDF
15. Publisher Correction: A pan-serotype dengue virus inhibitor targeting the NS3-NS4B interaction.
- Author
-
Kaptein SJF, Goethals O, Kiemel D, Marchand A, Kesteleyn B, Bonfanti JF, Bardiot D, Stoops B, Jonckers THM, Dallmeier K, Geluykens P, Thys K, Crabbe M, Chatel-Chaix L, Münster M, Querat G, Touret F, de Lamballerie X, Raboisson P, Simmen K, Chaltin P, Bartenschlager R, Van Loock M, and Neyts J
- Published
- 2021
- Full Text
- View/download PDF
16. A pan-serotype dengue virus inhibitor targeting the NS3-NS4B interaction.
- Author
-
Kaptein SJF, Goethals O, Kiemel D, Marchand A, Kesteleyn B, Bonfanti JF, Bardiot D, Stoops B, Jonckers THM, Dallmeier K, Geluykens P, Thys K, Crabbe M, Chatel-Chaix L, Münster M, Querat G, Touret F, de Lamballerie X, Raboisson P, Simmen K, Chaltin P, Bartenschlager R, Van Loock M, and Neyts J
- Subjects
- Animals, Antiviral Agents pharmacokinetics, Antiviral Agents therapeutic use, Dengue drug therapy, Dengue Virus genetics, Dengue Virus metabolism, Disease Models, Animal, Female, Male, Membrane Proteins antagonists & inhibitors, Mice, RNA Helicases antagonists & inhibitors, RNA Helicases metabolism, Serine Endopeptidases metabolism, Viral Load drug effects, Viral Nonstructural Proteins antagonists & inhibitors, Viremia drug therapy, Viremia virology, Virus Replication drug effects, Antiviral Agents pharmacology, Dengue virology, Dengue Virus classification, Dengue Virus drug effects, Membrane Proteins metabolism, Viral Nonstructural Proteins metabolism
- Abstract
Dengue virus causes approximately 96 million symptomatic infections annually, manifesting as dengue fever or occasionally as severe dengue
1,2 . There are no antiviral agents available to prevent or treat dengue. Here, we describe a highly potent dengue virus inhibitor (JNJ-A07) that exerts nanomolar to picomolar activity against a panel of 21 clinical isolates that represent the natural genetic diversity of known genotypes and serotypes. The molecule has a high barrier to resistance and prevents the formation of the viral replication complex by blocking the interaction between two viral proteins (NS3 and NS4B), thus revealing a previously undescribed mechanism of antiviral action. JNJ-A07 has a favourable pharmacokinetic profile that results in outstanding efficacy against dengue virus infection in mouse infection models. Delaying start of treatment until peak viraemia results in a rapid and significant reduction in viral load. An analogue is currently in further development., (© 2021. The Author(s), under exclusive licence to Springer Nature Limited.)- Published
- 2021
- Full Text
- View/download PDF
17. Phylogenetically based establishment of a dengue virus panel, representing all available genotypes, as a tool in dengue drug discovery.
- Author
-
Touret F, Baronti C, Goethals O, Van Loock M, de Lamballerie X, and Querat G
- Subjects
- Dengue Virus classification, Drug Evaluation, Preclinical, Genetic Variation, Genome, Viral genetics, Genotype, Humans, Phylogeny, Serogroup, Viral Proteins antagonists & inhibitors, Viral Proteins genetics, Antiviral Agents pharmacology, Dengue Virus drug effects, Dengue Virus genetics
- Abstract
Dengue fever is the most widespread of the human arbovirus diseases, with approximately one third of the world's population at risk of infection. Dengue viruses are members of the genus Flavivirus (family Flaviviridae) and, antigenically, they separate as four closely related serotypes (1-4) that share 60-75% amino acid homology. This genetic diversity complicates the process of antiviral drug discovery. Thus, currently no approved dengue-specific therapeutic treatments are available. With the aim of providing an efficient tool for dengue virus drug discovery, a collection of nineteen dengue viruses, representing the genotypic diversity within the four serotypes, was developed. After phylogenetic analysis of the full-length genomes, we selected relevant strains from the EVAg collection at Aix-Marseille University and completed the virus collection, using a reverse genetic system based on the infectious sub-genomic amplicons technique. Finally, we evaluated this dengue virus collection against three published dengue inhibitory compounds. NITD008, which targets the highly conserved active site of the viral NS5 polymerase enzyme, exhibited similar antiviral potencies against each of the different dengue genotypes in the panel. Compounds targeting less conserved protein subdomains, such as the capsid inhibitor ST-148, or SDM25N, a ∂ opioid receptor antagonist which indirectly targets NS4B, exhibited larger differences in potency against the various genotypes of dengue viruses. These results illustrate the importance of a phylogenetically based dengue virus reference panel for dengue antiviral research. The collection developed in this study, which includes such representative dengue viruses, has been made available to the scientific community through the European Virus Archive to evaluate novel DENV antiviral candidates., (Copyright © 2019 The Authors. Published by Elsevier B.V. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
18. Broad spectrum anti-flavivirus pyridobenzothiazolones leading to less infective virions.
- Author
-
Cannalire R, Tarantino D, Piorkowski G, Carletti T, Massari S, Felicetti T, Barreca ML, Sabatini S, Tabarrini O, Marcello A, Milani M, Cecchetti V, Mastrangelo E, Manfroni G, and Querat G
- Subjects
- Animals, Dengue Virus drug effects, Encephalitis Viruses, Tick-Borne drug effects, Humans, RNA, Viral drug effects, Virion drug effects, Virus Replication drug effects, West Nile virus drug effects, Yellow fever virus drug effects, Zika Virus drug effects, Antiviral Agents chemical synthesis, Antiviral Agents pharmacology, Flaviviridae drug effects, Oxazocines chemical synthesis, Oxazocines pharmacology, Pyridines chemical synthesis, Pyridines pharmacology
- Abstract
We report the design, synthesis, and biological evaluation of a class of 1H-pyrido[2,1-b][1,3]benzothiazol-1-ones originated from compound 1, previously identified as anti-flavivirus agent. Some of the new compounds showed activity in low μM range with reasonable selectivity against Dengue 2, Yellow fever (Bolivia strain), and West Nile viruses. One of the most interesting molecules, compound 16, showed broad antiviral activity against additional flaviviruses such as Dengue 1, 3 and 4, Zika, Japanese encephalitis, several strains of Yellow fever, and tick-borne encephalitis viruses. Compound 16 did not exert any effect on alphaviruses and phleboviruses and its activity was maintained in YFV infected cells from different species. The activity of 16 appears specific for flavivirus with respect to other virus families, suggesting, but not proving, that it might be targeting a viral factor. We demonstrated that the antiviral effect of 16 is not related to reduced viral RNA synthesis or virion release. On the contrary, viral particles grown in the presence of 16 showed reduced infectivity, being unable to perform a second round of infection. The chemical class herein presented thus emerges as suitable to provide pan-flavivirus inhibitors., (Copyright © 2019 Elsevier B.V. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
19. Optimization of a fragment linking hit toward Dengue and Zika virus NS5 methyltransferases inhibitors.
- Author
-
Hernandez J, Hoffer L, Coutard B, Querat G, Roche P, Morelli X, Decroly E, and Barral K
- Subjects
- Antiviral Agents chemical synthesis, Antiviral Agents chemistry, Crystallography, X-Ray, Dengue Virus enzymology, Dose-Response Relationship, Drug, Enzyme Inhibitors chemical synthesis, Enzyme Inhibitors chemistry, Methyltransferases metabolism, Microbial Sensitivity Tests, Models, Molecular, Molecular Structure, Structure-Activity Relationship, Zika Virus enzymology, Antiviral Agents pharmacology, Dengue Virus drug effects, Enzyme Inhibitors pharmacology, Methyltransferases antagonists & inhibitors, Zika Virus drug effects
- Abstract
No antiviral drugs to treat or prevent life-threatening flavivirus infections such as those caused by mosquito-borne Dengue (DENV) and more recently Zika (ZIKV) viruses are yet available. We aim to develop, through a structure-based drug design approach, novel inhibitors targeting the NS5 AdoMet-dependent mRNA methyltransferase (MTase), a viral protein involved in the RNA capping process essential for flaviviruses replication. Herein, we describe the optimization of a hit (5) identified using fragment-based and structure-guided linking techniques, which binds to a proximal site of the AdoMet binding pocket. X-ray crystallographic structures and computational docking were used to guide our optimization process and lead to compounds 30 and 33 (DENV IC
50 = 26 μM and 23 μM; ZIKV IC50 = 28 μM and 19 μM, respectively), two representatives of novel non-nucleoside inhibitors of flavivirus MTases., (Copyright © 2018 Elsevier Masson SAS. All rights reserved.)- Published
- 2019
- Full Text
- View/download PDF
20. Inhibition of the Replication of Different Strains of Chikungunya Virus by 3-Aryl-[1,2,3]triazolo[4,5- d]pyrimidin-7(6 H)-ones.
- Author
-
Gómez-SanJuan A, Gamo AM, Delang L, Pérez-Sánchez A, Amrun SN, Abdelnabi R, Jacobs S, Priego EM, Camarasa MJ, Jochmans D, Leyssen P, Ng LFP, Querat G, Neyts J, and Pérez-Pérez MJ
- Subjects
- Animals, Antiviral Agents chemical synthesis, Antiviral Agents chemistry, Cell Line, Chemical Phenomena, Chikungunya virus physiology, Humans, Microbial Sensitivity Tests, Molecular Structure, Antiviral Agents isolation & purification, Chikungunya virus drug effects, Virus Replication drug effects
- Abstract
The re-emergence of chikungunya virus (CHIKV) is a serious global health threat. CHIKV is an alphavirus that is transmitted to humans by Aedes mosquitoes; therefore, their wide distribution significantly contributes to the globalization of the disease. Unfortunately, no effective antiviral drugs are available. We have identified a series of 3-aryl-[1,2,3]triazolo[4,5- d]pyrimidin-7(6 H)-ones as selective inhibitors of CHIKV replication. New series of compounds have now been synthesized with the aim to improve their physicochemical properties and to potentiate the inhibitory activity against different CHIKV strains. Among these newly synthesized compounds modified at position 3 of the aryl ring, tetrahydropyranyl and N- t-butylpiperidine carboxamide derivatives have shown to elicit potent antiviral activity against different clinically relevant CHIKV isolates with 50% effective concentration (EC
50 ) values ranging from 0.30 to 4.5 μM in Vero cells, as well as anti-CHIKV activity in human skin fibroblasts (EC50 = 0.1 μM), a clinically relevant cell system for CHIKV infection.- Published
- 2018
- Full Text
- View/download PDF
21. Functionalized 2,1-benzothiazine 2,2-dioxides as new inhibitors of Dengue NS5 RNA-dependent RNA polymerase.
- Author
-
Cannalire R, Tarantino D, Astolfi A, Barreca ML, Sabatini S, Massari S, Tabarrini O, Milani M, Querat G, Mastrangelo E, Manfroni G, and Cecchetti V
- Subjects
- Animals, Antiviral Agents chemical synthesis, Antiviral Agents chemistry, Caco-2 Cells, Cell Survival drug effects, Chlorocebus aethiops, Dengue Virus enzymology, Dose-Response Relationship, Drug, Enzyme Inhibitors chemical synthesis, Enzyme Inhibitors chemistry, Humans, Microbial Sensitivity Tests, Models, Molecular, Molecular Structure, RNA-Dependent RNA Polymerase metabolism, Structure-Activity Relationship, Vero Cells, Antiviral Agents pharmacology, Dengue Virus drug effects, Enzyme Inhibitors pharmacology, RNA-Dependent RNA Polymerase antagonists & inhibitors
- Abstract
Over recent years, many RNA viruses have been "re-discovered", including life-threatening flaviviruses, such as Dengue, Zika, and several encephalitis viruses. Since no specific inhibitors are currently available to treat these infections, there is a pressing need for new therapeutics. Among the flaviviral proteins, NS5 RNA-dependent RNA polymerase (RdRp) represents a validated target being essential for viral replication and it has no human analog. To date, few NS5 RdRp inhibitor chemotypes have been reported and no inhibitors are currently in clinical development. In this context, after an in vitro screening against Dengue 3 NS5 RdRp of our in-house HCV NS5B inhibitors focused library, we found that 2,1-benzothiazine 2,2-dioxides are promising non-nucleoside inhibitors of flaviviral RdRp with compounds 8 and 10 showing IC
50 of 0.6 and 0.9 μM, respectively. Preliminary structure-activity relationships indicated a key role for the C-4 benzoyl group and the importance of a properly functionalized C-6 phenoxy moiety to modulate potency. Compound 8 acts as non-competitive inhibitor and its proposed pose in the so-called N pocket of the RdRp thumb domain allowed to explain the key contribution of the benzoyl and the phenoxy moieties for the ligand binding., (Copyright © 2017 Elsevier Masson SAS. All rights reserved.)- Published
- 2018
- Full Text
- View/download PDF
22. Toscana virus cap-snatching and initiation of transcription.
- Author
-
Amroun A, Priet S, and Querat G
- Subjects
- Cells, Cultured, Epithelial Cells virology, Humans, RNA, Messenger chemistry, RNA, Messenger genetics, RNA, Viral chemistry, RNA, Viral genetics, RNA, Messenger biosynthesis, RNA, Viral biosynthesis, RNA-Dependent RNA Polymerase metabolism, Sandfly fever Naples virus enzymology, Sandfly fever Naples virus genetics, Transcription, Genetic
- Abstract
Toscana virus (TOSV) is an arthropod-borne phlebovirus within the family Phenuiviridae in the order Bunyavirales. It seems to be an important agent of human meningoencephalitis in the warm season in the Mediterranean area. Because the polymerase of Bunyavirales lacks a capping activity, it cleaves short-capped RNA leaders derived from the host cell, and uses them to initiate viral mRNA synthesis. To determine the size and nucleotide composition of the host-derived RNA leaders, and to elucidate the first steps of TOSV transcription initiation, we performed a high-throughput sequencing of the 5' end of TOSV mRNAs in infected cells at different times post-infection. Our results indicated that the viral polymerase cleaved the host-capped RNA leaders within a window of 11-16 nucleotides. A single population of cellular mRNAs could be cleaved at different sites to prime the synthesis of several viral mRNA species. The majority of the mRNA resulted from direct priming, but we observed mRNAs resulting from several rounds of prime-and-realign events. Our data suggest that the different rounds of the prime-and-realign mechanism result from the blocking of the template strand in a static position in the active site, leading to the slippage of the nascent strand by two nucleotides when the growing duplex is sorted out from the active site. To minimize this rate-limiting step, TOSV polymerase cleaves preferentially capped RNA leaders after GC, so as to greatly reduce the number of cycles of priming and realignment, and facilitate the separation of the growing duplex.
- Published
- 2017
- Full Text
- View/download PDF
23. Antiviral activity of [1,2,3]triazolo[4,5-d]pyrimidin-7(6H)-ones against chikungunya virus targeting the viral capping nsP1.
- Author
-
Gigante A, Gómez-SanJuan A, Delang L, Li C, Bueno O, Gamo AM, Priego EM, Camarasa MJ, Jochmans D, Leyssen P, Decroly E, Coutard B, Querat G, Neyts J, and Pérez-Pérez MJ
- Subjects
- Animals, Chlorocebus aethiops, Microbial Sensitivity Tests, Vero Cells, Antiviral Agents pharmacology, Chikungunya virus drug effects, Enzyme Inhibitors pharmacology, Viral Nonstructural Proteins antagonists & inhibitors
- Abstract
Chikungunya virus (CHIKV) is a re-emerging alphavirus transmitted to humans by Aedes mosquitoes. Since 2005, CHIKV has been spreading worldwide resulting in epidemics in Africa, the Indian Ocean islands, Asia and more recently in the Americas. CHIKV is thus considered as a global health concern. There is no specific vaccine or drug available for the treatment of this incapacitating viral infection. We previously identified 3-aryl-[1,2,3]triazolo[4,5-d]pyrimidin-7(6H)-ones as selective inhibitors of CHIKV replication and proposed the viral capping enzyme nsP1 as a target. This work describes the synthesis of novel series of related compounds carrying at the aryl moiety a methylketone and related oximes combined with an ethyl or an ethyl-mimic at 5-position of the triazolopyrimidinone. These compounds have shown antiviral activity against different CHIKV isolates in the very low μM range based on both virus yield reduction and virus-induced cell-killing inhibition assays. Moreover, these antivirals inhibit the in vitro guanylylation of alphavirus nsP1, as determined by Western blot using an anti-cap antibody. Thus, the data obtained seem to indicate that the anti-CHIKV activity might be related to the inhibition of this crucial step in the viral RNA capping machinery., (Copyright © 2017 The Authors. Published by Elsevier B.V. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
24. Discovery of novel dengue virus NS5 methyltransferase non-nucleoside inhibitors by fragment-based drug design.
- Author
-
Benmansour F, Trist I, Coutard B, Decroly E, Querat G, Brancale A, and Barral K
- Subjects
- Antiviral Agents pharmacology, Binding Sites, Crystallography, X-Ray, Drug Discovery, Humans, Ligands, Methyltransferases antagonists & inhibitors, Sulfates pharmacology, Sulfonamides pharmacology, Antiviral Agents chemistry, Dengue Virus enzymology, Enzyme Inhibitors chemistry, Viral Nonstructural Proteins antagonists & inhibitors
- Abstract
With the aim to help drug discovery against dengue virus (DENV), a fragment-based drug design approach was applied to identify ligands targeting a main component of DENV replication complex: the NS5 AdoMet-dependent mRNA methyltransferase (MTase) domain, playing an essential role in the RNA capping process. Herein, we describe the identification of new inhibitors developed using fragment-based, structure-guided linking and optimization techniques. Thermal-shift assay followed by a fragment-based X-ray crystallographic screening lead to the identification of three fragment hits binding DENV MTase. We considered linking two of them, which bind to proximal sites of the AdoMet binding pocket, in order to improve their potency. X-ray crystallographic structures and computational docking were used to guide the fragment linking, ultimately leading to novel series of non-nucleoside inhibitors of flavivirus MTase, respectively N-phenyl-[(phenylcarbamoyl)amino]benzene-1-sulfonamide and phenyl [(phenylcarbamoyl)amino]benzene-1-sulfonate derivatives, that show a 10-100-fold stronger inhibition of 2'-O-MTase activity compared to the initial fragments., (Copyright © 2016 Elsevier Masson SAS. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
25. Targeting flavivirus RNA dependent RNA polymerase through a pyridobenzothiazole inhibitor.
- Author
-
Tarantino D, Cannalire R, Mastrangelo E, Croci R, Querat G, Barreca ML, Bolognesi M, Manfroni G, Cecchetti V, and Milani M
- Subjects
- Antiviral Agents pharmacology, Benzothiazoles chemistry, Binding Sites, Catalytic Domain, Crystallization, Dengue Virus drug effects, Dengue Virus enzymology, Drug Discovery, Flavivirus drug effects, Kinetics, Models, Molecular, Molecular Docking Simulation, Mutation, RNA-Dependent RNA Polymerase genetics, RNA-Dependent RNA Polymerase metabolism, West Nile virus drug effects, West Nile virus enzymology, Benzothiazoles pharmacology, Flavivirus enzymology, RNA-Dependent RNA Polymerase antagonists & inhibitors, RNA-Dependent RNA Polymerase drug effects
- Abstract
RNA dependent RNA polymerases (RdRp) are essential enzymes for flavivirus replication. Starting from an in silico docking analysis we identified a pyridobenzothiazole compound, HeE1-2Tyr, able to inhibit West Nile and Dengue RdRps activity in vitro, which proved effective against different flaviviruses in cell culture. Crystallographic data show that HeE1-2Tyr binds between the fingers domain and the priming loop of Dengue virus RdRp (Site 1). Conversely, enzyme kinetics, binding studies and mutational analyses suggest that, during the catalytic cycle and assembly of the RdRp-RNA complex, HeE1-2Tyr might be hosted in a distinct binding site (Site 2). RdRp mutational studies, driven by in silico docking analysis, allowed us to locate the inhibition Site 2 in the thumb domain. Taken together, our results provide innovative concepts for optimization of a new class of anti-flavivirus compounds., (Copyright © 2016 The Authors. Published by Elsevier B.V. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
26. Novel 2-phenyl-5-[(E)-2-(thiophen-2-yl)ethenyl]-1,3,4-oxadiazole and 3-phenyl-5-[(E)-2-(thiophen-2-yl)ethenyl]-1,2,4-oxadiazole derivatives as dengue virus inhibitors targeting NS5 polymerase.
- Author
-
Benmansour F, Eydoux C, Querat G, de Lamballerie X, Canard B, Alvarez K, Guillemot JC, and Barral K
- Subjects
- Animals, Antiviral Agents chemistry, Cell Line, Dengue virology, Dengue Virus enzymology, Humans, Oxadiazoles chemistry, RNA-Dependent RNA Polymerase metabolism, Thiophenes chemistry, Thiophenes pharmacology, Viral Nonstructural Proteins metabolism, Antiviral Agents pharmacology, Dengue drug therapy, Dengue Virus drug effects, Oxadiazoles pharmacology, RNA-Dependent RNA Polymerase antagonists & inhibitors, Viral Nonstructural Proteins antagonists & inhibitors
- Abstract
Using a functional high-throughput screening (HTS) and subsequent SAR studies, we have discovered a novel series of non-nucleoside dengue viral polymerase inhibitors. We report the elaboration of SAR around hit compound 1 as well as the synthesis and antiviral evaluation of 3-phenyl-5-[(E)-2-(thiophen-2-yl)ethenyl]-1,2,4-oxadiazole and 5-phenyl-2-[2-(2-thienyl)ethenyl]-1,3,4-oxadiazole analogues derived from a rapid and easily accessible chemical pathway. A large number of compounds prepared by this method were shown to possess in vitro activity against the polymerase of dengue virus. The most potent inhibitors were tested against Dengue virus clinical isolates on infected cells model and exhibit submicromolar activity on the four dengue virus serotypes., (Copyright © 2016 Elsevier Masson SAS. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
27. Single-stranded positive-sense RNA viruses generated in days using infectious subgenomic amplicons.
- Author
-
Aubry F, Nougairède A, de Fabritus L, Querat G, Gould EA, and de Lamballerie X
- Subjects
- Animals, Cell Line, Cricetinae, DNA, Complementary genetics, DNA, Viral genetics, Flavivirus genetics, Flavivirus physiology, Genome, Viral, Humans, Molecular Sequence Data, RNA, Viral genetics, Virus Replication genetics, Virus Replication physiology, RNA Viruses genetics, RNA Viruses physiology, Reverse Genetics methods
- Abstract
Reverse genetics is a key methodology for producing genetically modified RNA viruses and deciphering cellular and viral biological properties, but methods based on the preparation of plasmid-based complete viral genomes are laborious and unpredictable. Here, both wild-type and genetically modified infectious RNA viruses were generated in days using the newly described ISA (infectious-subgenomic-amplicons) method. This new versatile and simple procedure may enhance our capacity to obtain infectious RNA viruses from PCR-amplified genetic material., (© 2014 The Authors.)
- Published
- 2014
- Full Text
- View/download PDF
28. Identification of [1,2,3]triazolo[4,5-d]pyrimidin-7(6H)-ones as novel inhibitors of Chikungunya virus replication.
- Author
-
Gigante A, Canela MD, Delang L, Priego EM, Camarasa MJ, Querat G, Neyts J, Leyssen P, and Pérez-Pérez MJ
- Subjects
- Animals, Antiviral Agents pharmacology, Chikungunya virus physiology, Chlorocebus aethiops, Pyrimidinones pharmacology, Structure-Activity Relationship, Triazoles pharmacology, Vero Cells, Antiviral Agents chemical synthesis, Chikungunya virus drug effects, Pyrimidinones chemical synthesis, Triazoles chemical synthesis, Virus Replication drug effects
- Abstract
Chikungunya virus (CHIKV) is a re-emerging Alphavirus that is transmitted to humans by Aedes mosquitoes. Currently, there are still no drugs or vaccines available for the treatment or prevention of this disease. Although traditionally restricted to Africa and Asia, the adaptation of the virus to Aedes albopictus, a mosquito species with an almost worldwide distribution, has contributed to the geographical spread of this virus in the past decade. Here, we report on a new family of compounds named [1,2,3]triazolo[4,5-d]pyrimidin-7(6H)-ones that inhibit CHIKV replication in the low micromolar range with no toxicity to the host (Vero) cells. The most potent compound in this series has an EC50 value below 1 μM with no cytotoxicity detected up to 668 μM, therefore affording a selectivity index greater than 600. Interestingly, the compounds have little or no antiviral activity on the replication of other members of the Togaviridae family. The exploration and study of this class of selective inhibitors of CHIKV replication will contribute to deeper insights into the CHIKV life cycle and may be a first step toward the development of a clinical drug candidate.
- Published
- 2014
- Full Text
- View/download PDF
29. Functional role of HIV-1 virion-associated uracil DNA glycosylase 2 in the correction of G:U mispairs to G:C pairs.
- Author
-
Priet S, Navarro JM, Gros N, Querat G, and Sire J
- Subjects
- Base Pair Mismatch, DNA Repair, HIV Infections virology, HIV-1 physiology, Humans, Macrophages enzymology, Macrophages virology, N-Glycosyl Hydrolases genetics, Uracil-DNA Glycosidase, Virion enzymology, Virion physiology, Virus Replication, DNA Glycosylases, HIV Infections enzymology, HIV-1 enzymology, N-Glycosyl Hydrolases metabolism
- Abstract
Human monocytes/macrophages are target cells for HIV-1 infection. As other non-dividing cells, they are characterized by low and imbalanced intracellular dNTP pool levels and an excess of dUTP. The replication of HIV-1 in this cellular context favors misincorporation of uracil residues into viral DNA because of the use of dUTP in place of dCTP. We have previously reported that the host uracil DNA glycosylase enzyme UNG2 is packaged into HIV-1 viral particles via a specific association with the integrase domain of the Gag-Pol precursor. In this study, we investigated whether virion-associated UNG2 plays a role similar to that of its cellular counterpart. We show that the L172A mutation of integrase impaired the packaging of UNG2 into viral particles. Using a primer-template DNA substrate containing G:U mispairs, we demonstrate that wild-type viral lysate has the ability to repair G:U mismatched pairs to G:C matched pairs, in contrast to UNG2-deficient viral lysate. Moreover, no correction of G:T mispairs by wild-type HIV-1 viral lysate was observed, which argues for the specificity of the repair process. We also show that UNG2 physically associates with the viral reverse transcriptase enzyme. Altogether our data indicate for the first time that a uracil repair pathway is specifically associated with HIV-1 viral particles. However, the molecular mechanism of this process remains to be characterized further.
- Published
- 2003
- Full Text
- View/download PDF
30. Lack of functional receptors is the only barrier that prevents caprine arthritis-encephalitis virus from infecting human cells.
- Author
-
Mselli-Lakhal L, Favier C, Leung K, Guiguen F, Grezel D, Miossec P, Mornex JF, Narayan O, Querat G, and Chebloune Y
- Subjects
- Animals, Arthritis-Encephalitis Virus, Caprine physiology, Cells, Cultured, Goats, Humans, Precipitin Tests, Receptors, Virus genetics, Reverse Transcriptase Polymerase Chain Reaction, Species Specificity, Transfection, Vesicular stomatitis Indiana virus genetics, Viral Envelope Proteins genetics, Viral Envelope Proteins metabolism, Arthritis-Encephalitis Virus, Caprine pathogenicity, Membrane Glycoproteins, Receptors, Virus metabolism, Virus Replication
- Abstract
Barriers to replication of viruses in potential host cells may occur at several levels. Lack of suitable and functional receptors on the host cell surface, thereby precluding entry of the virus, is a frequent reason for noninfectivity, as long as no alternative way of entry (e.g., pinocytosis, antibody-dependent adsorption) can be exploited by the virus. Other barriers can intervene at later stages of the virus life cycle, with restrictions on transcription of the viral genome, incorrect translation and posttranslational processing of viral proteins, inefficient viral assembly, and release or efficient early induction of apoptosis in the infected cell. The data we present here demonstrate that replication of caprine arthritis-encephalitis virus (CAEV) is restricted in a variety of human cell lines and primary tissue cultures. This barrier was efficiently overcome by transfection of a novel infectious complete-proviral CAEV construct into the same cells. The successful infection of human cells with a vesicular stomatitis virus (VSV) G-pseudotyped Env-defective CAEV confirmed that viral entry is the major obstacle to CAEV infection of human cells. The fully efficient productive infection obtained with the VSV-G-protein-pseudotyped infectious CAEV strengthened the evidence that lack of viral entry is the only practical barrier to CAEV replication in human cells. The virus thus produced retained its original host cell specificity and acquired no propensity to propagate further in human cultures.
- Published
- 2000
- Full Text
- View/download PDF
31. Nucleotide sequence and transcriptional analysis of molecular clones of CAEV which generate infectious virus.
- Author
-
Saltarelli M, Querat G, Konings DA, Vigne R, and Clements JE
- Subjects
- Amino Acid Sequence, Animals, Arthritis-Encephalitis Virus, Caprine pathogenicity, Base Sequence, Blotting, Northern, Cells, Cultured, Cloning, Molecular, Gene Products, gag genetics, Gene Products, pol genetics, Gene Products, tat, Genes, Regulator, Goats, Models, Molecular, Molecular Sequence Data, Nucleic Acid Conformation, Polymerase Chain Reaction, RNA, Messenger genetics, RNA, Messenger isolation & purification, RNA, Viral genetics, RNA, Viral isolation & purification, Sequence Homology, Nucleic Acid, Species Specificity, Synovial Membrane cytology, Transfection, Viral Envelope Proteins genetics, Arthritis-Encephalitis Virus, Caprine genetics, Genes, Viral, Transcription, Genetic
- Abstract
The lentivirus caprine arthritis-encephalitis virus (CAEV) is closely related by nucleotide sequence homology to visna virus and other sheep lentiviruses and shows less similarity to the other animal and human lentiviruses. The genomic organization of CAEV is very similar to that of visna virus and the South African ovine maedi visna virus (SA-OMVV) as well as to those of other primate lentiviruses. The CAEV genome includes the small open reading frames (ORF) between pol and env which are the hallmarks of the lentivirus genomes. The most striking difference in the organization of CAEV is in the env gene. The Env polyproteins of visna virus and the related SA-OMVV contain 20 amino acids between the translational start and the signal peptide not present in CAEV. In addition to nucleotide sequence analysis, the transcriptional products of CAEV were determined by Northern analysis. The viral mRNA present in cells transfected with the infectious clone reveal a pattern characteristic of the mRNAs observed in other lentivirus infections. The putative tat ORF of CAEV could be identified by genomic location and amino acid homology to the visna virus tat gene. However, the CAEV rev gene could not be identified in a similar fashion. Thus, to determine the location of the rev ORF cDNA clones were obtained by PCR amplification of the mRNA from infected cells. To determine if a Rev response element was contained in the CAEV genome, secondary structural analysis of the viral RNA was performed. A stable stem loop structure which is similar in location, stability, and configuration to that determined for the Rev response element of HIV was found.
- Published
- 1990
- Full Text
- View/download PDF
32. Subcellular localization of rev-gene product in visna virus-infected cells.
- Author
-
Mazarin V, Gourdou I, Querat G, Sauze N, Audoly G, Vitu C, Russo P, Rousselot C, Filippi P, and Vigne R
- Subjects
- Animals, Fluorescent Antibody Technique, Kinetics, Lysogeny, Pneumonia, Progressive Interstitial, of Sheep immunology, Pneumonia, Progressive Interstitial, of Sheep pathology, Precipitin Tests, Rabbits, Sheep, Virion genetics, Gene Products, rev genetics, Trans-Activators genetics, Virus Replication genetics, Visna-maedi virus genetics
- Abstract
The 1.4-kb mRNA of visna lentivirus is expressed early during the lytic infection of sheep choroid plexus cell cultures. It encodes for visna early gene 1 (VEG1) product, since renamed rev gene product (or Rev), based on significant amino acid sequence homologies between this protein and the proteins of simian immunodeficiency virus of macaque and human immunodeficiency virus type 2. In this report, we examined the subcellular localization and time course appearance of the Rev protein in visna virus-infected cells. Immunoprecipitation assays of [35S]methionine-labeled cell lysates with antisera raised against the Rev protein revealed a polypeptide of 19 kDa (p19rev). This protein was predominant early in the viral replication cycle and accumulated preferentially in the cytoplasmic/membrane fraction of infected cells. Indirect immunofluorescence staining of infected cells confirmed the cytoplasmic location of visna Rev protein and could reveal in some stained cells a higher concentration of Rev at the cellular plasma membrane. The regulating protein, still present late in the viral lytic cycle, is packaged into mature viral particles along with the structural gag and env gene products.
- Published
- 1990
- Full Text
- View/download PDF
33. Regulatory genes of visna virus.
- Author
-
Vigne R, Gourdou I, Mazarin V, Querat G, Sauze N, Audoly G, Filippi P, Rousselot C, Vitu C, and Russo P
- Subjects
- Amino Acid Sequence, Animals, Antibodies, Viral biosynthesis, Blotting, Northern, Blotting, Western, Cell Line, Fluorescent Antibody Technique, Gene Expression Regulation, Viral, Gene Products, rev chemistry, Gene Products, rev immunology, Gene Products, tat chemistry, Gene Products, tat genetics, Molecular Sequence Data, Precipitin Tests, RNA, Messenger genetics, RNA, Viral genetics, Radioimmunoprecipitation Assay, Transfection, Gene Products, rev genetics, Genes, Regulator, Genes, rev, Visna-maedi virus genetics
- Published
- 1990
34. [An ELISA test for detection of maedi-visna antibodies. Comparative study with gel immunodiffusion and complement-fixation test].
- Author
-
Vitu C, Russo P, Filippi P, Vigne R, Querat G, and Giauffret A
- Subjects
- Animals, Goats immunology, Sheep immunology, Antibodies, Viral analysis, Complement Fixation Tests, Enzyme-Linked Immunosorbent Assay, Immunodiffusion, Immunoenzyme Techniques, Visna-maedi virus immunology
- Abstract
An indirect microELISA test was performed for detection of maedi-visna antibodies in ovine and caprine species. The antigen consisted in viral particles, highly purified by successive ultracentrifugations. By comparative testing of 934 sera in ELISA and gel immunodiffusion, we found a good correlation between these two tests, and moreover, ELISA revealed another 11.3% of positive samples. The precocity of this ELISA was shown by experimental infection of sheep with different strains of maedi-visna: positive sera were detected 7 weeks post-infection, instead 4-5 months with gel immunodiffusion. The complement fixation test was compared with gel immunodiffusion and was found the less sensitive. This ELISA test appeared to be satisfactory, and may be used for early diagnosis of maedi-visna infection.
- Published
- 1982
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.