87 results on '"Galloux M"'
Search Results
2. Function of the translocation domain belt
- Author
-
Galloux, M., Araye-Guet, A., Vitrac, H., Montagner, C., Raffestin, S., Popoff, M. R., Chenal, A., Forge, V., and Gillet, D.
- Published
- 2013
- Full Text
- View/download PDF
3. Boosting subdominant neutralizing antibody responses with a computationally designed epitope-focused immunogen
- Author
-
Sesterhenn, F, primary, Galloux, M, additional, Vollers, SS, additional, Csepregi, L, additional, Yang, C, additional, Descamps, D, additional, Bonet, J, additional, Friedensohn, S, additional, Gainza, P, additional, Corthésy, P, additional, Chen, M, additional, Rosset, S, additional, Rameix-Welti, MA, additional, Eléouët, JF, additional, Reddy, ST, additional, Graham, BS, additional, Riffault, S, additional, and Correia, BE, additional
- Published
- 2018
- Full Text
- View/download PDF
4. Modélisation mathématique dans un essai clinique de phase 1 : application dans le traitement des cancers bronchiques non à petites cellules et des mésothéliomes avancés, par vinorelbine orale, selon un schéma métronomique
- Author
-
Barbolosi, M., primary, Imbs, D.C., additional, Tomasini, P., additional, Greillier, L., additional, Galloux, M., additional, Testot-Ferry, A., additional, Garcia, M., additional, Elharrar, X., additional, Pelletier, A., additional, Andre, N., additional, Mascaux, C., additional, Lacarelle, B., additional, El Cheikh, R., additional, Serre, R., additional, Ciccolini, J., additional, Barbolosi, D., additional, and Barlesi, F., additional
- Published
- 2018
- Full Text
- View/download PDF
5. Targeting human respiratory syncytial virus transcription anti-termination factor M2-1 to inhibit in vivo viral replication
- Author
-
Bailly, B., primary, Richard, C.-A., additional, Sharma, G., additional, Wang, L., additional, Johansen, L., additional, Cao, J., additional, Pendharkar, V., additional, Sharma, D.-C., additional, Galloux, M., additional, Wang, Y., additional, Cui, R., additional, Zou, G., additional, Guillon, P., additional, von Itzstein, M., additional, Eléouët, J.-F., additional, and Altmeyer, R., additional
- Published
- 2016
- Full Text
- View/download PDF
6. The respiratory syncytial virus nucleoprotein–RNA\ud complex forms a left-handed helical nucleocapsid
- Author
-
Bakker, S. E., Duquerroy, S., Galloux, M., Loney, C., Conner, E., Eleouet, J.-F., Rey, F. A., and Bhella, D.
- Subjects
viruses - Abstract
Respiratory Syncytial Virus (RSV) is an important human pathogen. Its nucleocapsid (NC), which comprises the negative sense RNA viral genome coated by the viral nucleoprotein N, is a critical assembly that serves as template for both mRNA synthesis and genome replication. We have previously described the X-ray structure of a nucleocapsid-like structure: a decameric ring formed of N-RNA that mimics one turn of the helical NC. In the absence of experimental data we had hypothesized that the NC helix would be right-handed, as the N-N contacts in the ring appeared to more easily adapt to that conformation. We now unambiguously show that the RSV NC is a left-handed helix. We further show that the contacts in the ring can be distorted to maintain key N-N protein interactions in a left-handed helix, and discuss the implications of the resulting atomic model of the helical NC for viral replication and transcription.
- Published
- 2013
7. N-terminal globular domain of the RSV Nucleoprotein in complex with the Nucleoprotein Phosphoprotein interaction inhibitor M76
- Author
-
Ouizougun-Oubari, M., primary, Pereira, N., additional, Tarus, B., additional, Galloux, M., additional, Tortorici, M.-A., additional, Hoos, S., additional, Baron, B., additional, England, P., additional, Bontems, F., additional, Rey, F.A., additional, Eleouet, J.-F., additional, Sizun, C., additional, Slama-Schwok, A., additional, and Duquerroy, S., additional
- Published
- 2015
- Full Text
- View/download PDF
8. N-terminal globular domain of the RSV Nucleoprotein
- Author
-
Ouizougun-Oubari, M., primary, Pereira, N., additional, Tarus, B., additional, Galloux, M., additional, Tortorici, M.-A., additional, Hoos, S., additional, Baron, B., additional, England, P., additional, Bontems, F., additional, Rey, F.A., additional, Eleouet, J.-F., additional, Sizun, C., additional, Slama-Schwok, A., additional, and Duquerroy, S., additional
- Published
- 2015
- Full Text
- View/download PDF
9. N-terminal globular domain of the RSV Nucleoprotein in complex with C- terminal dipeptide of the Phosphoprotein
- Author
-
Ouizougun-Oubari, M., primary, Pereira, N., additional, Tarus, B., additional, Galloux, M., additional, Tortorici, M.-A., additional, Hoos, S., additional, Baron, B., additional, England, P., additional, Bontems, F., additional, Rey, F.A., additional, Eleouet, J.-F., additional, Sizun, C., additional, Slama-Schwok, A., additional, and Duquerroy, S., additional
- Published
- 2015
- Full Text
- View/download PDF
10. N-terminal globular domain of the RSV Nucleoprotein in complex with C- terminal peptide of the Phosphoprotein
- Author
-
Ouizougun-Oubari, M., primary, Pereira, N., additional, Tarus, B., additional, Galloux, M., additional, Tortorici, M.-A., additional, Hoos, S., additional, Baron, B., additional, England, P., additional, Bontems, F., additional, Rey, F.A., additional, Eleouet, J.-F., additional, Sizun, C., additional, Slama-Schwok, A., additional, and Duquerroy, S., additional
- Published
- 2015
- Full Text
- View/download PDF
11. Esprit d'entreprise : Aspects managériaux dans le monde francophone
- Author
-
Nzisabira, J., Tamini, Z., J.-M., Roussignol, J.-H., Guilmette, Ponson, B., d'Iribarne, P., Cancade, G., Albagli, C., J.-L., Schaan, Haudeville, B., Lecointre, G., J.-P., Laporte, Doutriaux, J., Havet, J., Abdeljalil N., Ibn, Mikayoulou, D., Arellano, R., Gasse, Y., Verna, G., C.-N., Diouf, M.-B., Sall, Wade, B., Dzaka, T., Assignon, E., G.-A., Brenner, Fouda, H., J.-M., Toulouse, Mugunga, E., D.-A., Soedjede, M.-R., Amrani, Gupta, J., Causse, G., Bloy, Eddy, Jabes, J., Bekolo-Ebe, B., Lelart, M., Makaya, A., Mayoukou, C., Ossie, W., Pairault, T., Sumata, C., Abdaimi M., El, Galloux, M., N.-E., Haddab, Traimond, P., Deggis, Gilles, and Agence Universitaire de la Francophonie (AUF)
- Subjects
esprit d'entreprise ,entreprise ,francophonie ,[SHS.GESTION]Humanities and Social Sciences/Business administration ,pays en voie de développement ,[SHS.GESTION] Humanities and Social Sciences/Business administration ,développement - Abstract
ISBN : 2-7420-0003-8; Dans le contexte des pays en voie de développement, on observe des différences dans l'expression de l'esprit d'entreprise, dont la plupart sont évoquées dans cet ouvrage. Celui-ci est articulé autour de trois thèmes : l'émergence et les manifestations de l'esprit d'entreprise, le management et l'esprit d'entreprise, les circuits financiers de l'entreprise.
- Published
- 2007
12. The Respiratory Syncytial Virus nucleoprotein-RNA complex forms a left-handed helical nucleocapsid.
- Author
-
Bakker, S.E., primary, Duquerroy, S., additional, Galloux, M., additional, Loney, C., additional, Conner, E., additional, Eleouet, J.F., additional, Rey, F.A., additional, and Bhella, D., additional
- Published
- 2013
- Full Text
- View/download PDF
13. NMR structure of pep46 from the infectious bursal disease virus (IBDV) in dodecylphosphocholin (DPC).
- Author
-
Galloux, M., primary, Libersou, S., additional, Morellet, N., additional, Bouaziz, S., additional, Ouldali, M., additional, Da Costa, B., additional, Lepault, J., additional, and Delmas, B., additional
- Published
- 2007
- Full Text
- View/download PDF
14. Structural relationships among birnaviruses and other icosahedral viruses
- Author
-
Coulibaly, F., Christophe Chevalier, Galloux, M., Da Costa, B., Lepault, J., Delmas, B., Rey, F., and Chevalier, Christophe
- Subjects
[SDV.MP.VIR] Life Sciences [q-bio]/Microbiology and Parasitology/Virology ,[SDV.BBM.BS] Life Sciences [q-bio]/Biochemistry, Molecular Biology/Structural Biology [q-bio.BM] - Abstract
Les particules des virus à ARN double brin (ARNdb) sont compétentes pour la transcription et doivent traverser une membrane cellulaire pour fonctionner dans le cytoplasme de la cellule cible. Parmi ces virus, les birnavirus sont singuliers car ils possèdent une simple capside icosaédrique de triangulation T \= 13 et ne contiennent pas la capside interne caractéristique observée dans la plupart de ces virus. Nous avons récemment élucidé la structure de sous-particules virales ainsi que de particules virales complètes d’un birnavirus aviaire [1]. Nos résultats révèlent des relations structurales inattendues entre virus icosaédriques et nous permettent de proposer un lien phylogénétique entre certains virus à ARNdb et des virus à ARN de polarité positive.
15. Structural relationships among birnaviruses and other icosahedral viruses | Les relations structurales entre birnavirus et autres virus icosaédriques à ARN
- Author
-
Fasséli Coulibaly, Chevalier, C., Galloux, M., Da Costa, B., Lepault, J., Delmas, B., and Rey, F.
16. HERV-W ENV transcription in B cells predicting symptomatic COVID-19 and risk for long COVID can express a full-length protein despite stop codon in mRNA from chromosome X via a ribosome readthrough.
- Author
-
Brunel J, Paganini J, Galloux M, Charvet B, and Perron H
- Abstract
The human genome comprises 8% of endogenous retroviruses (HERVs). Though HERVS contribute to physiological functions, copies retained pathogenic potential. The HERV-W ENV protein was shown expressed in patients with worse COVID-19 symptoms and post-COVID syndrome. A significant detection of the mRNA encoding HERV-W ENV from patients with COVID-19 in B cells from RNAseq reads obtained from peripheral blond mononuclear cells. This data stratified with increased COVID-19 symptoms or with post-acute sequelae of COVID-19 (long COVID) after 3 months. The HERV-W ENV-U3R RNA was confirmed to display the best alignment with chromosome X ERVWE2 locus. However, a stop codon precluding its translation was re-addressed after recent understandings of ribosome readthrough mechanisms. Experimental results evidenced that this HERV gene can effectively express a full-length protein in the presence of molecules allowing translation via a readthrough mechanism at the ribosome level. Results not only confirm HERV-W ENV RNA origin in these patients but show for the first time how a defective HERV copy can be translated into a complete protein when specific factors make it possible at the ribosome level. The present proof of concept now requires further studies to identify the factors involved in this newly understood mechanism, following SARS-CoV-2 exposure., (Copyright © 2024. Published by Elsevier Masson SAS.)
- Published
- 2024
- Full Text
- View/download PDF
17. Nanoencapsulated deltamethrin combined with indoxacarb: An effective synergistic association against aphids.
- Author
-
Galloux M, Bastiat G, Lefrancois C, Apaire-Marchais V, and Deshayes C
- Abstract
Widespread pesticide use for decades has caused environmental damage, biodiversity loss, serious human and animal health problems, and resistance to insecticides. Innovative strategies are needed to reduce treatment doses in pest management and to overcome insecticide resistance. In the present study, combinations of indoxacarb, an oxadiazine insecticide, with sublethal concentrations of deltamethrin encapsulated in lipid nanocapsules, have been tested on the crop pest Acyrthosiphon pisum . In vivo toxicological tests on A. pisum larvae have shown a synergistic effect of nanoencapsulated deltamethrin with a low dose of indoxacarb. Furthermore, the stability of deltamethrin nanoparticles has been demonstrated in vitro under different mimicking environmental conditions. In parallel, the integrity and stability of lipid nanoparticles in the digestive system of aphid larvae over time have been observed by Förster Resonance Energy Transfer (FRET) imaging. Thus, the deltamethrin nanocapsules/indoxacarb synergistic association is promising for the development of future formulations against pest insects to reduce insecticide doses., Competing Interests: Declaration of Competing InterestThe authors declare that there is no competing interest., (© 2024 Pesticide Science Society of Japan.)
- Published
- 2024
- Full Text
- View/download PDF
18. Author Correction: Mucosal bivalent live attenuated vaccine protects against human metapneumovirus and respiratory syncytial virus in mice.
- Author
-
Ogonczyk-Makowska D, Brun P, Vacher C, Chupin C, Droillard C, Carbonneau J, Laurent E, Dulière V, Traversier A, Terrier O, Julien T, Galloux M, Paul S, Eléouët JF, Fouret J, Hamelin ME, Pizzorno A, Boivin G, Rosa-Calatrava M, and Dubois J
- Published
- 2024
- Full Text
- View/download PDF
19. Unraveling Liquid-Liquid Phase Separation (LLPS) in Viral Infections to Understand and Treat Viral Diseases.
- Author
-
Galloux M and Longhi S
- Subjects
- Humans, Virus Replication, Liquid-Liquid Extraction methods, Phase Separation, Virus Diseases virology
- Abstract
In the field of virology, liquid-liquid phase separation (LLPS) has emerged as a pivotal mechanism enabling the compartmentalization required for specific steps of the viral replication cycle [...].
- Published
- 2024
- Full Text
- View/download PDF
20. Mucosal bivalent live attenuated vaccine protects against human metapneumovirus and respiratory syncytial virus in mice.
- Author
-
Ogonczyk-Makowska D, Brun P, Vacher C, Chupin C, Droillard C, Carbonneau J, Laurent E, Dulière V, Traversier A, Terrier O, Julien T, Galloux M, Paul S, Eléouët JF, Fouret J, Hamelin ME, Pizzorno A, Boivin G, Rosa-Calatrava M, and Dubois J
- Abstract
Live-Attenuated Vaccines (LAVs) stimulate robust mucosal and cellular responses and have the potential to protect against Respiratory Syncytial Virus (RSV) and Human Metapneumovirus (HMPV), the main etiologic agents of viral bronchiolitis and pneumonia in children. We inserted the RSV-F gene into an HMPV-based LAV (Metavac®) we previously validated for the protection of mice against HMPV challenge, and rescued a replicative recombinant virus (Metavac®-RSV), exposing both RSV- and HMPV-F proteins at the virion surface and expressing them in reconstructed human airway epithelium models. When administered to BALB/c mice by the intranasal route, bivalent Metavac®-RSV demonstrated its capacity to replicate with reduced lung inflammatory score and to protect against both RSV and lethal HMPV challenges in vaccinated mice while inducing strong IgG and broad RSV and HMPV neutralizing antibody responses. Altogether, our results showed the versatility of the Metavac® platform and suggested that Metavac®-RSV is a promising mucosal bivalent LAV candidate to prevent pneumovirus-induced diseases., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
21. Engineering Tumor Stroma Morphogenesis Using Dynamic Cell-Matrix Spheroid Assembly.
- Author
-
Buckenmeyer MJ, Brooks EA, Taylor MS, Yang L, Holewinski RJ, Meyer TJ, Galloux M, Garmendia-Cedillos M, Pohida TJ, Andresson T, Croix B, and Wolf MT
- Abstract
The tumor microenvironment consists of resident tumor cells organized within a compositionally diverse, three-dimensional (3D) extracellular matrix (ECM) network that cannot be replicated in vitro using bottom-up synthesis. We report a new self-assembly system to engineer ECM-rich 3D MatriSpheres wherein tumor cells actively organize and concentrate microgram quantities of decellularized ECM dispersions which modulate cell phenotype. 3D colorectal cancer (CRC) MatriSpheres were created using decellularized small intestine submucosa (SIS) as an orthotopic ECM source that had greater proteomic homology to CRC tumor ECM than traditional ECM formulations such as Matrigel. SIS ECM was rapidly concentrated from its environment and assembled into ECM-rich 3D stroma-like regions by mouse and human CRC cell lines within 4-5 days via a mechanism that was rheologically distinct from bulk hydrogel formation. Both ECM organization and transcriptional regulation by 3D ECM cues affected programs of malignancy, lipid metabolism, and immunoregulation that corresponded with an in vivo MC38 tumor cell subpopulation identified via single cell RNA sequencing. This 3D modeling approach stimulates tumor specific tissue morphogenesis that incorporates the complexities of both cancer cell and ECM compartments in a scalable, spontaneous assembly process that may further facilitate precision medicine., Competing Interests: Competing interests: None
- Published
- 2024
- Full Text
- View/download PDF
22. Drug repurposing screen identifies lonafarnib as respiratory syncytial virus fusion protein inhibitor.
- Author
-
Sake SM, Zhang X, Rajak MK, Urbanek-Quaing M, Carpentier A, Gunesch AP, Grethe C, Matthaei A, Rückert J, Galloux M, Larcher T, Le Goffic R, Hontonnou F, Chatterjee AK, Johnson K, Morwood K, Rox K, Elgaher WAM, Huang J, Wetzke M, Hansen G, Fischer N, Eléouët JF, Rameix-Welti MA, Hirsch AKH, Herold E, Empting M, Lauber C, Schulz TF, Krey T, Haid S, and Pietschmann T
- Subjects
- Animals, Female, Mice, Drug Repositioning, Piperidines pharmacology, Piperidines therapeutic use, Viral Fusion Proteins genetics, Viral Fusion Proteins chemistry, Dibenzocycloheptenes, Pyridines, Respiratory Syncytial Virus Infections drug therapy
- Abstract
Respiratory syncytial virus (RSV) is a common cause of acute lower respiratory tract infection in infants, older adults and the immunocompromised. Effective directly acting antivirals are not yet available for clinical use. To address this, we screen the ReFRAME drug-repurposing library consisting of 12,000 small molecules against RSV. We identify 21 primary candidates including RSV F and N protein inhibitors, five HSP90 and four IMPDH inhibitors. We select lonafarnib, a licensed farnesyltransferase inhibitor, and phase III candidate for hepatitis delta virus (HDV) therapy, for further follow-up. Dose-response analyses and plaque assays confirm the antiviral activity (IC
50 : 10-118 nM). Passaging of RSV with lonafarnib selects for phenotypic resistance and fixation of mutations in the RSV fusion protein (T335I and T400A). Lentiviral pseudotypes programmed with variant RSV fusion proteins confirm that lonafarnib inhibits RSV cell entry and that these mutations confer lonafarnib resistance. Surface plasmon resonance reveals RSV fusion protein binding of lonafarnib and co-crystallography identifies the lonafarnib binding site within RSV F. Oral administration of lonafarnib dose-dependently reduces RSV virus load in a murine infection model using female mice. Collectively, this work provides an overview of RSV drug repurposing candidates and establishes lonafarnib as a bona fide fusion protein inhibitor., (© 2024. The Author(s).)- Published
- 2024
- Full Text
- View/download PDF
23. A New Derivative of Retro-2 Displays Antiviral Activity against Respiratory Syncytial Virus.
- Author
-
Le Rouzic A, Fix J, Vinck R, Kappler-Gratias S, Volmer R, Gallardo F, Eléouët JF, Keck M, Cintrat JC, Barbier J, Gillet D, and Galloux M
- Subjects
- Infant, Newborn, Adult, Child, Aged, Humans, Antibodies, Antiviral Agents pharmacology, Respiratory Syncytial Virus, Human, Respiratory Syncytial Virus Infections drug therapy, Respiratory Tract Infections
- Abstract
Human respiratory syncytial virus (hRSV) is the most common cause of bronchiolitis and pneumonia in newborns, with all children being infected before the age of two. Reinfections are very common throughout life and can cause severe respiratory infections in the elderly and immunocompromised adults. Although vaccines and preventive antibodies have recently been licensed for use in specific subpopulations of patients, there is still no therapeutic treatment commonly available for these infections. Here, we investigated the potential antiviral activity of Retro-2.2, a derivative of the cellular retrograde transport inhibitor Retro-2, against hRSV. We show that Retro-2.2 inhibits hRSV replication in cell culture and impairs the ability of hRSV to form syncytia. Our results suggest that Retro-2.2 treatment affects virus spread by disrupting the trafficking of the viral de novo synthetized F and G glycoproteins to the plasma membrane, leading to a defect in virion morphogenesis. Taken together, our data show that targeting intracellular transport may be an effective strategy against hRSV infection.
- Published
- 2023
- Full Text
- View/download PDF
24. Structure of the N-RNA/P interface indicates mode of L/P recruitment to the nucleocapsid of human metapneumovirus.
- Author
-
Whitehead JD, Decool H, Leyrat C, Carrique L, Fix J, Eléouët JF, Galloux M, and Renner M
- Subjects
- Child, Humans, Child, Preschool, Nucleocapsid metabolism, RNA, Viral genetics, RNA, Viral metabolism, Nucleoproteins metabolism, Phosphoproteins metabolism, Metapneumovirus genetics
- Abstract
Human metapneumovirus (HMPV) is a major cause of respiratory illness in young children. The HMPV polymerase (L) binds an obligate cofactor, the phosphoprotein (P). During replication and transcription, the L/P complex traverses the viral RNA genome, which is encapsidated within nucleoproteins (N). An essential interaction between N and a C-terminal region of P tethers the L/P polymerase to the template. This N-P interaction is also involved in the formation of cytoplasmic viral factories in infected cells, called inclusion bodies. To define how the polymerase component P recognizes N-encapsidated RNA (N-RNA) we employed cryogenic electron microscopy (cryo-EM) and molecular dynamics simulations, coupled to activity assays and imaging of inclusion bodies in cells. We report a 2.9 Å resolution structure of a triple-complex between multimeric N, bound to both RNA and the C-terminal region of P. Furthermore, we also present cryo-EM structures of assembled N in different oligomeric states, highlighting the plasticity of N. Combined with our functional assays, these structural data delineate in molecular detail how P attaches to N-RNA whilst retaining substantial conformational dynamics. Moreover, the N-RNA-P triple complex structure provides a molecular blueprint for the design of therapeutics to potentially disrupt the attachment of L/P to its template., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
25. Structural landscape of the respiratory syncytial virus nucleocapsids.
- Author
-
Gonnin L, Desfosses A, Bacia-Verloop M, Chevret D, Galloux M, Éléouët JF, and Gutsche I
- Subjects
- Child, Aged, Humans, Cryoelectron Microscopy, Nucleocapsid genetics, RNA, Viral genetics, Nucleoproteins genetics, Respiratory Syncytial Virus, Human genetics
- Abstract
Human Respiratory Syncytial Virus (HRSV) is a prevalent cause of severe respiratory infections in children and the elderly. The helical HRSV nucleocapsid is a template for the viral RNA synthesis and a scaffold for the virion assembly. This cryo-electron microscopy analysis reveals the non-canonical arrangement of the HRSV nucleocapsid helix, composed of 16 nucleoproteins per asymmetric unit, and the resulting systematic variations in the RNA accessibility. We demonstrate that this unique helical symmetry originates from longitudinal interactions by the C-terminal arm of the HRSV nucleoprotein. We explore the polymorphism of the nucleocapsid-like assemblies, report five structures of the full-length particles and two alternative arrangements formed by a C-terminally truncated nucleoprotein mutant, and demonstrate the functional importance of the identified longitudinal interfaces. We put all these findings in the context of the HRSV RNA synthesis machinery and delineate the structural basis for its further investigation., (© 2023. Springer Nature Limited.)
- Published
- 2023
- Full Text
- View/download PDF
26. Saturation genome editing of 11 codons and exon 13 of BRCA2 coupled with chemotherapeutic drug response accurately determines pathogenicity of variants.
- Author
-
Sahu S, Sullivan TL, Mitrophanov AY, Galloux M, Nousome D, Southon E, Caylor D, Mishra AP, Evans CN, Clapp ME, Burkett S, Malys T, Chari R, Biswas K, and Sharan SK
- Subjects
- Animals, Humans, Mice, Female, Virulence, BRCA2 Protein genetics, BRCA2 Protein metabolism, Exons genetics, Codon, Nucleotides, Genetic Predisposition to Disease, BRCA1 Protein genetics, Gene Editing, Breast Neoplasms genetics
- Abstract
The unknown pathogenicity of a significant number of variants found in cancer-related genes is attributed to limited epidemiological data, resulting in their classification as variant of uncertain significance (VUS). To date, Breast Cancer gene-2 (BRCA2) has the highest number of VUSs, which has necessitated the development of several robust functional assays to determine their functional significance. Here we report the use of a humanized-mouse embryonic stem cell (mESC) line expressing a single copy of the human BRCA2 for a CRISPR-Cas9-based high-throughput functional assay. As a proof-of-principle, we have saturated 11 codons encoded by BRCA2 exons 3, 18, 19 and all possible single-nucleotide variants in exon 13 and multiplexed these variants for their functional categorization. Specifically, we used a pool of 180-mer single-stranded donor DNA to generate all possible combination of variants. Using a high throughput sequencing-based approach, we show a significant drop in the frequency of non-functional variants, whereas functional variants are enriched in the pool of the cells. We further demonstrate the response of these variants to the DNA-damaging agents, cisplatin and olaparib, allowing us to use cellular survival and drug response as parameters for variant classification. Using this approach, we have categorized 599 BRCA2 variants including 93-single nucleotide variants (SNVs) across the 11 codons, of which 28 are reported in ClinVar. We also functionally categorized 252 SNVs from exon 13 into 188 functional and 60 non-functional variants, demonstrating that saturation genome editing (SGE) coupled with drug sensitivity assays can enhance functional annotation of BRCA2 VUS., Competing Interests: The authors have declared that no competing interests exist., (Copyright: This is an open access article, free of all copyright, and may be freely reproduced, distributed, transmitted, modified, built upon, or otherwise used by anyone for any lawful purpose. The work is made available under the Creative Commons CC0 public domain dedication.)
- Published
- 2023
- Full Text
- View/download PDF
27. Hardening of Respiratory Syncytial Virus Inclusion Bodies by Cyclopamine Proceeds through Perturbation of the Interactions of the M2-1 Protein with RNA and the P Protein.
- Author
-
Diot C, Richard CA, Risso-Ballester J, Martin D, Fix J, Eléouët JF, Sizun C, Rameix-Welti MA, and Galloux M
- Subjects
- Veratrum Alkaloids, Inclusion Bodies, RNA, Respiratory Syncytial Virus, Human
- Abstract
Respiratory syncytial virus (RSV) RNA synthesis takes place in cytoplasmic viral factories also called inclusion bodies (IBs), which are membrane-less organelles concentrating the viral RNA polymerase complex. The assembly of IBs is driven by liquid-liquid phase separation promoted by interactions between the viral nucleoprotein N and the phosphoprotein P. We recently demonstrated that cyclopamine (CPM) inhibits RSV multiplication by disorganizing and hardening IBs. Although a single mutation in the viral transcription factor M2-1 induced resistance to CPM, the mechanism of action of CPM still remains to be characterized. Here, using FRAP experiments on reconstituted pseudo-IBs both in cellula and in vitro, we first demonstrated that CPM activity depends on the presence of M2-1 together with N and P. We showed that CPM impairs the competition between P and RNA binding to M2-1. As mutations on both P and M2-1 induced resistance against CPM activity, we suggest that CPM may affect the dynamics of the M2-1-P interaction, thereby affecting the relative mobility of the proteins contained in RSV IBs. Overall, our results reveal that stabilizing viral protein-protein interactions is an attractive new antiviral approach. They pave the way for the rational chemical optimization of new specific anti-RSV molecules., Competing Interests: The authors declare no conflict of interest.
- Published
- 2023
- Full Text
- View/download PDF
28. Screening antivirals with a mCherry-expressing recombinant bovine respiratory syncytial virus: a proof of concept using cyclopamine.
- Author
-
Fix J, Descamps D, Galloux M, Ferret C, Bouguyon E, Zohari S, Näslund K, Hägglund S, Altmeyer R, Valarcher JF, Riffault S, and Eléouët JF
- Subjects
- Animals, Cattle, Humans, Antiviral Agents pharmacology, Antiviral Agents therapeutic use, Antibodies, Viral, Respiratory Syncytial Virus, Bovine, Respiratory Syncytial Virus Infections drug therapy, Respiratory Syncytial Virus Infections veterinary, Respiratory Syncytial Virus, Human metabolism, Cattle Diseases
- Abstract
Bovine respiratory syncytial virus (BRSV) is a pathogenic pneumovirus and a major cause of acute respiratory infections in calves. Although different vaccines are available against BRSV, their efficiency remains limited, and no efficient and large-scale treatment exists. Here, we developed a new reverse genetics system for BRSV expressing the red fluorescent protein mCherry, based on a field strain isolated from a sick calf in Sweden. Although this recombinant fluorescent virus replicated slightly less efficiently compared to the wild type virus, both viruses were shown to be sensitive to the natural steroidal alkaloid cyclopamine, which was previously shown to inhibit human RSV replication. Our data thus point to the potential of this recombinant fluorescent BRSV as a powerful tool in preclinical drug discovery to enable high throughput compound screening., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
29. Level of maternal antibodies against respiratory syncytial virus (RSV) nucleoprotein at birth and risk of RSV very severe lower respiratory tract infection.
- Author
-
Receveur M, Ottmann M, Reynes JM, Eleouet JF, Galloux M, Receveur A, Ploin D, Fiorini S, Rivat N, Valette M, Lina B, and Casalegno JS
- Subjects
- Infant, Pregnancy, Female, Infant, Newborn, Humans, Infant, Premature, Prospective Studies, Antibodies, Viral, Respiratory Syncytial Virus Infections epidemiology, Respiratory Syncytial Virus, Human, Respiratory Tract Infections
- Abstract
Background: The nucleoprotein (N protein) of respiratory syncytial virus (RSV) is a candidate antigen for new RSV vaccine development. The aim of the present study was to investigate the association between maternal antibody titers against the RSV N protein at birth and the newborns' risk of developing very severe lower respiratory tract infection (VS-LRTI)., Methods: In this single-center prospective cohort study, 578 infants born during the RSV epidemic season in France were included. Among these, 36 were hospitalized for RSV VS-LRTI. A generalized linear model was used to test the occurrence of a VS-LRTI in function of sex, mode of delivery, parity of the mother, type of pregnancy, date of birth in relation to the peak of the epidemic, and antibody titer against N protein., Results: All cord blood samples had detectable antibodies against N protein. The mean titers were significantly lower in newborns with risk factors for RSV severe LRTI (preterm infants, birth before the peak epidemic, multiparous mother). There was no association between antibody titer against the N protein and a protection against VS-LRTI., Conclusions: Further studies are needed to support the hypothesis that transfer of maternal antibodies against the RSV N protein can provide a significant immune protection early in infancy and that N protein candidate vaccine may be a suitable target for maternal vaccine., (© 2022 The Authors. Influenza and Other Respiratory Viruses published by John Wiley & Sons Ltd.)
- Published
- 2023
- Full Text
- View/download PDF
30. Investigation of the Fuzzy Complex between RSV Nucleoprotein and Phosphoprotein to Optimize an Inhibition Assay by Fluorescence Polarization.
- Author
-
Khodjoyan S, Morissette D, Hontonnou F, Checa Ruano L, Richard CA, Sperandio O, Eléouët JF, Galloux M, Durand P, Deville-Foillard S, and Sizun C
- Subjects
- Peptides metabolism, Phosphoproteins metabolism, Fluorescence Polarization, Nucleoproteins, Respiratory Syncytial Virus, Human metabolism
- Abstract
The interaction between Respiratory Syncytial Virus phosphoprotein P and nucleoprotein N is essential for the formation of the holo RSV polymerase that carries out replication. In vitro screening of antivirals targeting the N-P protein interaction requires a molecular interaction model, ideally consisting of a complex between N protein and a short peptide corresponding to the C-terminal tail of the P protein. However, the flexibility of C-terminal P peptides as well as their phosphorylation status play a role in binding and may bias the outcome of an inhibition assay. We therefore investigated binding affinities and dynamics of this interaction by testing two N protein constructs and P peptides of different lengths and composition, using nuclear magnetic resonance and fluorescence polarization (FP). We show that, although the last C-terminal Phe
241 residue is the main determinant for anchoring P to N, only longer peptides afford sub-micromolar affinity, despite increasing mobility towards the N-terminus. We investigated competitive binding by peptides and small compounds, including molecules used as fluorescent labels in FP. Based on these results, we draw optimized parameters for a robust RSV N-P inhibition assay and validated this assay with the M76 molecule, which displays antiviral properties, for further screening of chemical libraries.- Published
- 2022
- Full Text
- View/download PDF
31. Respiratory Syncytial Virus NS1 Protein Targets the Transactivator Binding Domain of MED25.
- Author
-
Dong J, Basse V, Bierre M, Peres de Oliveira A, Vidalain PO, Sibille P, Tangy F, Galloux M, Eleouet JF, Sizun C, and Bajorek M
- Subjects
- Chromatin chemistry, Humans, Protein Binding, Protein Domains, RNA Polymerase II metabolism, Mediator Complex chemistry, Respiratory Syncytial Virus, Human genetics, Trans-Activators chemistry, Viral Nonstructural Proteins chemistry, Viral Nonstructural Proteins genetics
- Abstract
Human RSV is the leading cause of infantile bronchiolitis in the world and one of the major causes of childhood deaths in resource-poor settings. It is a major unmet target for vaccines and anti-viral drugs. Respiratory syncytial virus has evolved a unique strategy to evade host immune response by coding for two non-structural proteins NS1 and NS2. Recently it was shown that in infected cells, nuclear NS1 could be involved in transcription regulation of host genes linked to innate immune response, via interactions with chromatin and the Mediator complex. Here we identified the MED25 Mediator subunit as an NS1 interactor in a yeast two-hybrid screen. We demonstrate that NS1 directly interacts with MED25 in vitro and in cellula, and that this interaction involves the MED25 transactivator binding ACID domain on the one hand, and the C-terminal α3 helix of NS1, with an additional contribution of the globular domain of NS1, on the other hand. By NMR we show that the NS1 α3 sequence primarily binds to the MED25 ACID H2 face, similarly to the α-helical transactivation domains (TADs) of transcription regulators such as Herpex simplex VP16 and ATF6α, a master regulator of ER stress response activated upon viral infection. Moreover, we found out that the NS1 could compete with ATF6α TAD for binding to MED25. These findings point to a mechanism of NS1 interfering with innate immune response by impairing recruitment by cellular TADs of the Mediator via MED25 and hence transcription of specific genes by RNA polymerase II., Competing Interests: Declaration of interests The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2022 The Author(s). Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
32. Importance of RNA length for in vitro encapsidation by the nucleoprotein of human respiratory syncytial virus.
- Author
-
Gonnin L, Richard CA, Gutsche I, Chevret D, Troussier J, Vasseur JJ, Debart F, Eléouët JF, and Galloux M
- Subjects
- Humans, Phosphoproteins metabolism, Recombinant Fusion Proteins chemistry, Nucleocapsid chemistry, Nucleocapsid physiology, Nucleoproteins chemistry, Nucleoproteins metabolism, RNA, Viral chemistry, RNA, Viral metabolism, Respiratory Syncytial Virus, Human chemistry, Respiratory Syncytial Virus, Human physiology, Viral Genome Packaging, Viral Structural Proteins chemistry, Viral Structural Proteins metabolism
- Abstract
Respiratory syncytial virus has a negative-sense single-stranded RNA genome constitutively encapsidated by the viral nucleoprotein N, forming a helical nucleocapsid which is the template for viral transcription and replication by the viral polymerase L. Recruitment of L onto the nucleocapsid depends on the viral phosphoprotein P, which is an essential L cofactor. A prerequisite for genome and antigenome encapsidation is the presence of the monomeric, RNA-free, neosynthesized N protein, named N
0 . Stabilization of N0 depends on the binding of the N-terminal residues of P to its surface, which prevents N oligomerization. However, the mechanism involved in the transition from N0 -P to nucleocapsid assembly, and thus in the specificity of viral genome encapsidation, is still unknown. Furthermore, the specific role of N oligomerization and RNA in the morphogenesis of viral factories, where viral transcription and replication occur, have not been elucidated although the interaction between P and N complexed to RNA has been shown to be responsible for this process. Here, using a chimeric protein comprising N and the first 40 N-terminal residues of P, we succeeded in purifying a recombinant N0 -like protein competent for RNA encapsidation in vitro. Our results showed the importance of RNA length for stable encapsidation and revealed that the nature of the 5' end of RNA does not explain the specificity of encapsidation. Finally, we showed that RNA encapsidation is crucial for the in vitro reconstitution of pseudo-viral factories. Together, our findings provide insight into respiratory syncytial virus viral genome encapsidation specificity., Competing Interests: Conflict of interest The authors declare that they have no conflicts of interest with the contents of this article., (Copyright © 2022 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2022
- Full Text
- View/download PDF
33. Revisiting metronomic vinorelbine with mathematical modelling: a Phase I trial in lung cancer.
- Author
-
Barlesi F, Deyme L, Imbs DC, Cousin E, Barbolosi M, Bonnet S, Tomasini P, Greillier L, Galloux M, Testot-Ferry A, Pelletier A, André N, Ciccolini J, and Barbolosi D
- Subjects
- Administration, Metronomic, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Humans, Models, Theoretical, Vinblastine therapeutic use, Vinorelbine adverse effects, Carcinoma, Non-Small-Cell Lung pathology, Lung Neoplasms pathology, Neutropenia chemically induced, Neutropenia drug therapy
- Abstract
Background: A phase Ia/Ib trial of metronomic oral vinorelbine (MOV) driven by a mathematical model was performed in heavily pretreated metastatic Non-Small Cell Lung Cancer or Pleural Mesothelioma patients. Disease Control Rate, progression free survival, toxicity and PK/PD were the main endpoints., Methods: Best MOV scheduling was selected using a simplified phenomenological, semi-mechanistic model with a total weekly dose of 150-mg vinorelbine. Computation of individual PK parameters was performed using population approach., Results: The mathematical model proposed the following metronomic schedule for a 150-mg weekly dose of vinorelbine: 60 mg D1, 30 mg D2, 60 mg D4. A total of 37 heavily pre-treated patients (30 evaluable) were enrolled. Grade III/IV neutropenia was observed in 30% patients. Median PFS was 11 weeks. Disease Control Rate was 73% (i.e.; 13% partial response and 60% stable disease). A large variability in drug exposure (AUC
0-24 h : 53%) and PK parameters (Cl: 83%) were observed among patients. Simulated trough levels after D2 and D4 showed similarly 56-73% variability among patients. Drug exposure was not associated with efficacy, but neutropenia was more frequent in patients with AUC > 250 ng/ml.h. Tumor burden, performance status and neutrophils-to-lymphocyte ratio (NLR) were associated with PFS, suggesting that MOV would be indicated in selected patients. We built a composite score to predict efficacy, mixing baseline tumor size and NLR showing 84% selectivity and 75% specificity., Conclusions: MOV was characterized by important variability in drug exposure among patients. However, and despite being all heavily pre-treated, 73% of disease control rate and 11 weeks PFS were achieved with manageable toxicities. PK/PD relationships yielded conflicting results depending on the initial tumor burden and BSA, suggesting that patients should be carefully selected prior to be scheduled for metronomic regimen. Possible role NLR could play as a predictive marker suggests immunomodulating features with MOV., (© 2022. The Author(s), under exclusive licence to Springer-Verlag GmbH Germany, part of Springer Nature.)- Published
- 2022
- Full Text
- View/download PDF
34. Macromolecular Viral Entry Inhibitors as Broad-Spectrum First-Line Antivirals with Activity against SARS-CoV-2.
- Author
-
Groß R, Dias Loiola LM, Issmail L, Uhlig N, Eberlein V, Conzelmann C, Olari LR, Rauch L, Lawrenz J, Weil T, Müller JA, Cardoso MB, Gilg A, Larsson O, Höglund U, Pålsson SA, Tvilum AS, Løvschall KB, Kristensen MM, Spetz AL, Hontonnou F, Galloux M, Grunwald T, Zelikin AN, and Münch J
- Subjects
- Animals, Antiviral Agents pharmacology, Antiviral Agents therapeutic use, Gold, Mice, SARS-CoV-2, Virus Internalization, Metal Nanoparticles, COVID-19 Drug Treatment
- Abstract
Inhibitors of viral cell entry based on poly(styrene sulfonate) and its core-shell nanoformulations based on gold nanoparticles are investigated against a panel of viruses, including clinical isolates of SARS-CoV-2. Macromolecular inhibitors are shown to exhibit the highly sought-after broad-spectrum antiviral activity, which covers most analyzed enveloped viruses and all of the variants of concern for SARS-CoV-2 tested. The inhibitory activity is quantified in vitro in appropriate cell culture models and for respiratory viral pathogens (respiratory syncytial virus and SARS-CoV-2) in mice. Results of this study comprise a significant step along the translational path of macromolecular inhibitors of virus cell entry, specifically against enveloped respiratory viruses., (© 2022 The Authors. Advanced Science published by Wiley-VCH GmbH.)
- Published
- 2022
- Full Text
- View/download PDF
35. Characterization of the Interaction Domains between the Phosphoprotein and the Nucleoprotein of Human Metapneumovirus.
- Author
-
Decool H, Bardiaux B, Checa Ruano L, Sperandio O, Fix J, Gutsche I, Richard CA, Bajorek M, Eléouët JF, and Galloux M
- Subjects
- Animals, Binding Sites, Cell Line, Cricetinae, Inclusion Bodies metabolism, Metapneumovirus physiology, Models, Molecular, Mutation, Nucleocapsid Proteins genetics, Nucleocapsid Proteins metabolism, Phosphoproteins genetics, Phosphoproteins metabolism, Protein Binding, Protein Interaction Domains and Motifs, RNA, Viral metabolism, RNA-Dependent RNA Polymerase metabolism, Virus Replication, Metapneumovirus chemistry, Nucleocapsid Proteins chemistry, Phosphoproteins chemistry
- Abstract
Human metapneumovirus (HMPV) causes severe respiratory diseases in young children. The HMPV RNA genome is encapsidated by the viral nucleoprotein (N), forming an RNA-N complex (N
Nuc ), which serves as the template for genome replication and mRNA transcription by the RNA-dependent RNA polymerase (RdRp). The RdRp is formed by the association of the large polymerase subunit (L), which has RNA polymerase, capping, and methyltransferase activities, and the tetrameric phosphoprotein (P). P plays a central role in the RdRp complex by binding to NNuc and L, allowing the attachment of the L polymerase to the NNuc template. During infection these proteins concentrate in cytoplasmic inclusion bodies (IBs) where viral RNA synthesis occurs. By analogy to the closely related pneumovirus respiratory syncytial virus (RSV), it is likely that the formation of IBs depends on the interaction between HMPV P and NNuc , which has not been demonstrated yet. Here, we finely characterized the binding P-NNuc interaction domains by using recombinant proteins, combined with a functional assay for the polymerase complex activity, and the study of the recruitment of these proteins to IBs by immunofluorescence. We show that the last 6 C-terminal residues of HMPV P are necessary and sufficient for binding to NNuc and that P binds to the N-terminal domain of N (NNTD ), and we identified conserved N residues critical for the interaction. Our results allowed us to propose a structural model for the HMPV P-NNuc interaction. IMPORTANCE Human metapneumovirus (HMPV) is a leading cause of severe respiratory infections in children but also affects human populations of all ages worldwide. Currently, no vaccine or efficient antiviral treatments are available for this pneumovirus. A better understanding of the molecular mechanisms involved in viral replication could help the design or discovery of specific antiviral compounds. In this work, we have investigated the interaction between two major viral proteins involved in HMPV RNA synthesis, the N and P proteins. We finely characterized their domains of interaction and identified a pocket on the surface of the N protein, a potential target of choice for the design of compounds interfering with N-P complexes and inhibiting viral replication.- Published
- 2022
- Full Text
- View/download PDF
36. Interactions between the Nucleoprotein and the Phosphoprotein of Pneumoviruses: Structural Insight for Rational Design of Antivirals.
- Author
-
Decool H, Gonnin L, Gutsche I, Sizun C, Eléouët JF, and Galloux M
- Subjects
- Animals, Humans, Metapneumovirus drug effects, Metapneumovirus genetics, Models, Molecular, Nucleocapsid Proteins chemistry, Paramyxoviridae Infections drug therapy, Paramyxoviridae Infections virology, Phosphoproteins chemistry, Protein Binding, Protein Conformation, RNA, Viral chemistry, RNA, Viral metabolism, Respiratory Syncytial Virus Infections drug therapy, Respiratory Syncytial Virus Infections virology, Respiratory Syncytial Virus, Human drug effects, Respiratory Syncytial Virus, Human genetics, Transcription, Genetic, Viral Proteins chemistry, Virus Replication, Antiviral Agents chemistry, Antiviral Agents pharmacology, Drug Design, Metapneumovirus metabolism, Nucleocapsid Proteins metabolism, Phosphoproteins metabolism, Respiratory Syncytial Virus, Human metabolism, Viral Proteins metabolism
- Abstract
Pneumoviruses include pathogenic human and animal viruses, the most known and studied being the human respiratory syncytial virus (hRSV) and the metapneumovirus (hMPV), which are the major cause of severe acute respiratory tract illness in young children worldwide, and main pathogens infecting elderly and immune-compromised people. The transcription and replication of these viruses take place in specific cytoplasmic inclusions called inclusion bodies (IBs). These activities depend on viral polymerase L, associated with its cofactor phosphoprotein P, for the recognition of the viral RNA genome encapsidated by the nucleoprotein N, forming the nucleocapsid (NC). The polymerase activities rely on diverse transient protein-protein interactions orchestrated by P playing the hub role. Among these interactions, P interacts with the NC to recruit L to the genome. The P protein also plays the role of chaperone to maintain the neosynthesized N monomeric and RNA-free (called N
0 ) before specific encapsidation of the viral genome and antigenome. This review aims at giving an overview of recent structural information obtained for hRSV and hMPV P, N, and more specifically for P-NC and N0 -P complexes that pave the way for the rational design of new antivirals against those viruses.- Published
- 2021
- Full Text
- View/download PDF
37. Depletion of TAX1BP1 Amplifies Innate Immune Responses during Respiratory Syncytial Virus Infection.
- Author
-
Descamps D, Peres de Oliveira A, Gonnin L, Madrières S, Fix J, Drajac C, Marquant Q, Bouguyon E, Pietralunga V, Iha H, Morais Ventura A, Tangy F, Vidalain PO, Eléouët JF, and Galloux M
- Subjects
- Animals, Cell Line, Cricetinae, Humans, Immunity, Innate, Mice, Mice, Knockout, Virus Replication, Intracellular Signaling Peptides and Proteins immunology, Neoplasm Proteins immunology, Nucleocapsid Proteins immunology, Respiratory Syncytial Virus Infections immunology, Respiratory Syncytial Virus, Human immunology
- Abstract
Respiratory syncytial virus (RSV) is the main cause of acute respiratory infections in young children and also has a major impact on the elderly and immunocompromised people. In the absence of a vaccine or efficient treatment, a better understanding of RSV interactions with the host antiviral response during infection is needed. Previous studies revealed that cytoplasmic inclusion bodies (IBs), where viral replication and transcription occur, could play a major role in the control of innate immunity during infection by recruiting cellular proteins involved in the host antiviral response. We recently showed that the morphogenesis of IBs relies on a liquid-liquid-phase separation mechanism depending on the interaction between viral nucleoprotein (N) and phosphoprotein (P). These scaffold proteins are expected to play a central role in the recruitment of cellular proteins to IBs. Here, we performed a yeast two-hybrid screen using RSV N protein as bait and identified the cellular protein TAX1BP1 as a potential partner of this viral protein. This interaction was validated by pulldown and immunoprecipitation assays. We showed that TAX1BP1 suppression has only a limited impact on RSV infection in cell cultures. However, RSV replication is decreased in TAX1BP1-deficient (TAX1BP1 knockout [TAX1BP1
KO ]) mice, whereas the production of inflammatory and antiviral cytokines is enhanced. In vitro infection of wild-type or TAX1BP1KO alveolar macrophages confirmed that the innate immune response to RSV infection is enhanced in the absence of TAX1BP1. Altogether, our results suggest that RSV could hijack TAX1BP1 to restrain the host immune response during infection. IMPORTANCE Respiratory syncytial virus (RSV), which is the leading cause of lower respiratory tract illness in infants, remains a medical problem in the absence of a vaccine or efficient treatment. This virus is also recognized as a main pathogen in the elderly and immunocompromised people, and the occurrence of coinfections (with other respiratory viruses and bacteria) amplifies the risks of developing respiratory distress. In this context, a better understanding of the pathogenesis associated with viral respiratory infections, which depends on both viral replication and the host immune response, is needed. The present study reveals that the cellular protein TAX1BP1, which interacts with the RSV nucleoprotein N, participates in the control of the innate immune response during RSV infection, suggesting that the N-TAX1BP1 interaction represents a new target for the development of antivirals.- Published
- 2021
- Full Text
- View/download PDF
38. Avian Cell Line DuckCelt ® -T17 Is an Efficient Production System for Live-Attenuated Human Metapneumovirus Vaccine Candidate Metavac ® .
- Author
-
Chupin C, Pizzorno A, Traversier A, Brun P, Ogonczyk-Makowska D, Padey B, Milesi C, Dulière V, Laurent E, Julien T, Galloux M, Lina B, Eléouët JF, Moreau K, Hamelin ME, Terrier O, Boivin G, Dubois J, and Rosa-Calatrava M
- Abstract
The development of a live-attenuated vaccine (LAV) for the prevention of human metapneumovirus (HMPV) infection is often hampered by the lack of highly efficient and scalable cell-based production systems that support eventual global vaccine production. Avian cell lines cultivated in suspension compete with traditional cell platforms used for viral vaccine manufacture. We investigated whether the DuckCelt
® -T17 avian cell line (Vaxxel), previously described as an efficient production system for several influenza strains, could also be used to produce a new HMPV LAV candidate (Metavac® , SH gene-deleted A1/C-85473 HMPV). To that end, we characterized the operational parameters of MOI, cell density, and trypsin addition to achieve the optimal production of Metavac® , and demonstrated that the DuckCelt® -T17 cell line is permissive and well-adapted to the production of the wild-type A1/C-85473 HMPV and the Metavac® vaccine candidate. Moreover, our results confirmed that the LAV candidate produced in DuckCelt® -T17 cells conserves its advantageous replication properties in LLC-MK2 and 3D-reconstituted human airway epithelium models, and its capacity to induce efficient neutralizing antibodies in a BALB/c mouse model. Our results suggest that the DuckCelt® -T17 avian cell line is a very promising platform for the scalable in-suspension serum-free production of the HMPV-based LAV candidate Metavac® .- Published
- 2021
- Full Text
- View/download PDF
39. A Structural and Dynamic Analysis of the Partially Disordered Polymerase-Binding Domain in RSV Phosphoprotein.
- Author
-
Cardone C, Caseau CM, Bardiaux B, Thureaux A, Galloux M, Bajorek M, Eléouët JF, Litaudon M, Bontems F, and Sizun C
- Subjects
- Hydrogen Bonding, Light, Magnetic Resonance Spectroscopy, Models, Molecular, Protein Binding, Protein Conformation, Protein Domains, Scattering, Radiation, Terpenes chemistry, X-Rays, Nucleoproteins metabolism, Phosphoproteins chemistry, Respiratory Syncytial Virus, Human chemistry, Respiratory Syncytial Virus, Human genetics, Viral Proteins chemistry
- Abstract
The phosphoprotein P of Mononegavirales ( MNV ) is an essential co-factor of the viral RNA polymerase L. Its prime function is to recruit L to the ribonucleocapsid composed of the viral genome encapsidated by the nucleoprotein N. MNV phosphoproteins often contain a high degree of disorder. In Pneumoviridae phosphoproteins, the only domain with well-defined structure is a small oligomerization domain (P
OD ). We previously characterized the differential disorder in respiratory syncytial virus (RSV) phosphoprotein by NMR. We showed that outside of RSV POD , the intrinsically disordered N-and C-terminal regions displayed a structural and dynamic diversity ranging from random coil to high helical propensity. Here we provide additional insight into the dynamic behavior of PCα , a domain that is C-terminal to POD and constitutes the RSV L-binding region together with POD . By using small phosphoprotein fragments centered on or adjacent to POD , we obtained a structural picture of the POD -PCα region in solution, at the single residue level by NMR and at lower resolution by complementary biophysical methods. We probed POD -PCα inter-domain contacts and showed that small molecules were able to modify the dynamics of PCα . These structural properties are fundamental to the peculiar binding mode of RSV phosphoprotein to L, where each of the four protomers binds to L in a different way.- Published
- 2021
- Full Text
- View/download PDF
40. A condensate-hardening drug blocks RSV replication in vivo.
- Author
-
Risso-Ballester J, Galloux M, Cao J, Le Goffic R, Hontonnou F, Jobart-Malfait A, Desquesnes A, Sake SM, Haid S, Du M, Zhang X, Zhang H, Wang Z, Rincheval V, Zhang Y, Pietschmann T, Eléouët JF, Rameix-Welti MA, and Altmeyer R
- Subjects
- Animals, Antiviral Agents pharmacology, Cell Line, Female, Humans, Inclusion Bodies, Lung virology, Mice, Mice, Inbred BALB C, Respiratory Syncytial Virus, Human physiology, Transcription Factors, Viral Proteins, Biomolecular Condensates virology, Respiratory Syncytial Virus, Human drug effects, Veratrum Alkaloids pharmacology, Virus Replication drug effects
- Abstract
Biomolecular condensates have emerged as an important subcellular organizing principle
1 . Replication of many viruses, including human respiratory syncytial virus (RSV), occurs in virus-induced compartments called inclusion bodies (IBs) or viroplasm2,3 . IBs of negative-strand RNA viruses were recently shown to be biomolecular condensates that form through phase separation4,5 . Here we report that the steroidal alkaloid cyclopamine and its chemical analogue A3E inhibit RSV replication by disorganizing and hardening IB condensates. The actions of cyclopamine and A3E were blocked by a point mutation in the RSV transcription factor M2-1. IB disorganization occurred within minutes, which suggests that these molecules directly act on the liquid properties of the IBs. A3E and cyclopamine inhibit RSV in the lungs of infected mice and are condensate-targeting drug-like small molecules that have in vivo activity. Our data show that condensate-hardening drugs may enable the pharmacological modulation of not only many previously undruggable targets in viral replication but also transcription factors at cancer-driving super-enhancers6 ., (© 2021. The Author(s), under exclusive licence to Springer Nature Limited.)- Published
- 2021
- Full Text
- View/download PDF
41. Tetramerization of Phosphoprotein is Essential for Respiratory Syncytial Virus Budding while its N Terminal Region Mediates Direct Interactions with the Matrix Protein.
- Author
-
Bajorek M, Galloux M, Richard CA, Szekely O, Rosenzweig R, Sizun C, and Eleouet JF
- Abstract
It was shown previously that the Matrix (M), Phosphoprotein (P), and the Fusion (F) proteins of Respiratory syncytial virus (RSV) are sufficient to produce virus-like particles (VLPs) that resemble the RSV infection-induced virions. However, the exact mechanism and interactions among the three proteins are not known. This work examines the interaction between P and M during RSV assembly and budding. We show that M interacts with P in the absence of other viral proteins in cells using a Split Nano Luciferase assay. By using recombinant proteins, we demonstrate a direct interaction between M and P. By using Nuclear Magnetic Resonance (NMR) we identify three novel M interaction sites on P, namely site I in the α
N2 region, site II in the 115-125 region, and the oligomerization domain (OD). We show that the OD, and likely the tetrameric structural organization of P, is required for virus-like filament formation and VLP release. Although sites I and II are not required for VLP formation, they appear to modulate P levels in RSV VLPs. Importance Human RSV is the commonest cause of infantile bronchiolitis in the developed world and of childhood deaths in resource-poor settings. It is a major unmet target for vaccines and anti-viral drugs. The lack of knowledge of RSV budding mechanism presents a continuing challenge for VLP production for vaccine purpose. We show that direct interaction between P and M modulates RSV VLP budding. This further emphasizes P as a central regulator of RSV life cycle, as an essential actor for transcription and replication early during infection and as a mediator for assembly and budding in the later stages for virus production., (Copyright © 2021 American Society for Microbiology.)- Published
- 2021
- Full Text
- View/download PDF
42. New Look at RSV Infection: Tissue Clearing and 3D Imaging of the Entire Mouse Lung at Cellular Resolution.
- Author
-
Frétaud M, Descamps D, Laubreton D, Rameix-Welti MA, Eléouët JF, Larcher T, Galloux M, and Langevin C
- Subjects
- Animals, Disease Models, Animal, Epithelial Cells virology, Inclusion Bodies, Viral pathology, Mice, Respiratory Syncytial Virus Infections pathology, Respiratory Syncytial Virus Infections virology, Virus Replication, Imaging, Three-Dimensional, Lung pathology, Lung virology, Respiratory Syncytial Virus, Human physiology
- Abstract
Background: Respiratory Syncytial Virus (RSV) is the major cause of severe acute respiratory tract illness in young children worldwide and a main pathogen for the elderly and immune-compromised people. In the absence of vaccines or effective treatments, a better characterization of the pathogenesis of RSV infection is required. To date, the pathophysiology of the disease and its diagnosis has mostly relied on chest X-ray and genome detection in nasopharyngeal swabs. The development of new imaging approaches is instrumental to further the description of RSV spread, virus-host interactions and related acute respiratory disease, at the level of the entire lung., Methods: By combining tissue clearing, 3D microscopy and image processing, we developed a novel visualization tool of RSV infection in undissected mouse lungs., Results: Whole tissue analysis allowed the identification of infected cell subtypes, based on both morphological traits and position within the cellular network. Furthermore, 3D imaging was also valuable to detect the cytoplasmic viral factories, also called inclusion bodies, a hallmark of RSV infection., Conclusions: Whole lung clearing and 3D deep imaging represents an unprecedented visualization method of infected lungs to allow insight into RSV pathophysiology and improve the 2D histology analyses.
- Published
- 2021
- Full Text
- View/download PDF
43. Single-Stranded Oligonucleotide-Mediated Inhibition of Respiratory Syncytial Virus Infection.
- Author
-
Pålsson SA, Dondalska A, Bergenstråhle J, Rolfes C, Björk A, Sedano L, Power UF, Rameix-Welti MA, Lundeberg J, Wahren-Herlenius M, Mastrangelo P, Eleouet JF, Le Goffic R, Galloux M, and Spetz AL
- Subjects
- A549 Cells, Animals, Chemokine CCL2 genetics, Chemokine CXCL10 genetics, Female, Humans, Interferons genetics, Interferons metabolism, Mice, Phosphoproteins genetics, Phosphoproteins metabolism, Protein Binding, RNA-Binding Proteins genetics, RNA-Binding Proteins metabolism, Respiratory Mucosa pathology, STAT1 Transcription Factor genetics, STAT2 Transcription Factor genetics, Virus Internalization, Nucleolin, DNA, Single-Stranded genetics, Oligonucleotides genetics, Respiratory Mucosa metabolism, Respiratory Syncytial Virus Infections therapy, Respiratory Syncytial Viruses physiology
- Abstract
Respiratory syncytial virus (RSV) is the leading cause of acute lower respiratory tract infections in young children. Currently, there is no RSV vaccine or universally accessible antiviral treatment available. Addressing the urgent need for new antiviral agents, we have investigated the capacity of a non-coding single-stranded oligonucleotide (ssON) to inhibit RSV infection. By utilizing a GFP-expressing RSV, we demonstrate that the ssON significantly reduced the proportion of RSV infected A549 cells (lung epithelial cells). Furthermore, we show that ssON's antiviral activity was length dependent and that both RNA and DNA of this class of oligonucleotides have antiviral activity. We reveal that ssON inhibited RSV infection by competing with the virus for binding to the cellular receptor nucleolin in vitro . Additionally, using a recombinant RSV that expresses luciferase we show that ssON effectively blocked RSV infection in mice. Treatment with ssON in vivo resulted in the upregulation of RSV-induced interferon stimulated genes (ISGs) such as Stat1 , Stat2 , Cxcl10 , and Ccl2. This study highlights the possibility of using oligonucleotides as therapeutic agents against RSV infection. We demonstrate that the mechanism of action of ssON is the inhibition of viral entry in vitro , likely through the binding of the receptor, nucleolin and that ssON treatment against RSV infection in vivo additionally results in the upregulation of ISGs., Competing Interests: AD and A-LS are shareholders of TIRmed Pharma in possession of intellectual properties related to ssON. A-LS is CEO of TIRmed Pharma. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2020 Pålsson, Dondalska, Bergenstråhle, Rolfes, Björk, Sedano, Power, Rameix-Welti, Lundeberg, Wahren-Herlenius, Mastrangelo, Eleouet, Le Goffic, Galloux and Spetz.)
- Published
- 2020
- Full Text
- View/download PDF
44. Minimal Elements Required for the Formation of Respiratory Syncytial Virus Cytoplasmic Inclusion Bodies In Vivo and In Vitro .
- Author
-
Galloux M, Risso-Ballester J, Richard CA, Fix J, Rameix-Welti MA, and Eléouët JF
- Subjects
- Animals, Cell Line, Cricetinae, Humans, Kidney cytology, Morphogenesis, Nucleoproteins genetics, Phosphoproteins genetics, Respiratory Syncytial Virus, Human genetics, Virus Replication, Inclusion Bodies, Viral physiology, Nucleoproteins metabolism, Phosphoproteins metabolism, Respiratory Syncytial Virus, Human physiology, Viral Proteins metabolism
- Abstract
Infection of host cells by the respiratory syncytial virus (RSV) is characterized by the formation of spherical cytoplasmic inclusion bodies (IBs). These structures, which concentrate all the proteins of the polymerase complex as well as some cellular proteins, were initially considered aggresomes formed by viral dead-end products. However, recent studies revealed that IBs are viral factories where viral RNA synthesis, i.e., replication and transcription, occurs. The analysis of IBs by electron microscopy revealed that they are membrane-less structures, and accumulated data on their structure, organization, and kinetics of formation revealed that IBs share the characteristics of cellular organelles, such as P-bodies or stress granules, suggesting that their morphogenesis depends on a liquid-liquid phase separation mechanism. It was previously shown that expression of the RSV nucleoprotein N and phosphoprotein P of the polymerase complex is sufficient to induce the formation of pseudo-IBs. Here, using a series of truncated P proteins, we identified the domains of P required for IB formation and show that the oligomeric state of N, provided it can interact with RNA, is critical for their morphogenesis. We also show that pseudo-IBs can form in vitro when recombinant N and P proteins are mixed. Finally, using fluorescence recovery after photobleaching approaches, we reveal that in cellula and in vitro IBs are liquid organelles. Our results strongly support the liquid-liquid phase separation nature of IBs and pave the way for further characterization of their dynamics. IMPORTANCE Respiratory syncytial virus (RSV) is the leading cause of lower respiratory tract illness in infants, elderly, and immunocompromised people. No vaccine or efficient antiviral treatment is available against this virus. The replication and transcription steps of the viral genome are appealing mechanisms to target for the development of new antiviral strategies. These activities take place within cytoplasmic inclusion bodies (IBs) that assemble during infection. Although expression of both the viral nucleoprotein (N) and phosphoprotein (P) allows induction of the formation of these IBs, the mechanism sustaining their assembly remains poorly characterized. Here, we identified key elements of N and P required for the scaffolding of IBs and managed for the first time to reconstitute RSV pseudo-IBs in vitro by coincubating recombinant N and P proteins. Our results provide strong evidence that the biogenesis of RSV IBs occurs through liquid-liquid phase transition mediated by N-P interactions., (Copyright © 2020 Galloux et al.)
- Published
- 2020
- Full Text
- View/download PDF
45. Targeting the Respiratory Syncytial Virus N 0 -P Complex with Constrained α-Helical Peptides in Cells and Mice.
- Author
-
Galloux M, Gsponer N, Gaillard V, Fenner B, Larcher T, Vilotte M, Rivière J, Richard CA, Eléouët JF, Le Goffic R, Mettier J, and Nyanguile O
- Subjects
- Animals, Humans, Mice, Phosphoproteins pharmacology, Respiratory Syncytial Virus Infections drug therapy, Virus Replication, Antiviral Agents pharmacology, Peptides pharmacology, Protein Conformation, alpha-Helical, Respiratory Syncytial Virus, Human
- Abstract
Respiratory syncytial virus (RSV) is the main cause of severe respiratory infection in young children worldwide, and no therapies have been approved for the treatment of RSV infection. Data from recent clinical trials of fusion or L polymerase inhibitors for the treatment of RSV-infected patients revealed the emergence of escape mutants, highlighting the need for the discovery of inhibitors with novel mechanisms of action. Here we describe stapled peptides derived from the N terminus of the phosphoprotein (P) that act as replication inhibitors. We demonstrate that these peptides inhibit RSV replication in vitro and in vivo by preventing the formation of the N
0 -P complex. The present strategy provides a novel means of targeting RSV replication with constrained macrocyclic peptides or small molecules and is broadly applicable to other viruses of the Mononegavirales order., (Copyright © 2020 Galloux et al.)- Published
- 2020
- Full Text
- View/download PDF
46. Respiratory syncytial virus tropism for olfactory sensory neurons in mice.
- Author
-
Bryche B, Frétaud M, Saint-Albin Deliot A, Galloux M, Sedano L, Langevin C, Descamps D, Rameix-Welti MA, Eléouët JF, Le Goffic R, and Meunier N
- Subjects
- Animals, Central Nervous System diagnostic imaging, Central Nervous System virology, Central Nervous System Diseases diagnostic imaging, Central Nervous System Diseases virology, Female, Head anatomy & histology, Imaging, Three-Dimensional, Mice, Mice, Inbred BALB C, Nasal Mucosa virology, Olfactory Bulb virology, Olfactory Mucosa diagnostic imaging, RNA, Viral isolation & purification, Tropism, Virus Replication, Olfactory Mucosa innervation, Olfactory Mucosa virology, Olfactory Receptor Neurons virology, Respiratory Syncytial Viruses
- Abstract
The olfactory mucosa, where the first step of odor detection occurs, is a privileged pathway for environmental toxicants and pathogens toward the central nervous system. Indeed, some pathogens can infect olfactory sensory neurons including their axons projecting to the olfactory bulb allowing them to bypass the blood-brain barrier and reach the central nervous system (CNS) through the so-called olfactory pathway. The respiratory syncytial virus (RSV) is a major respiratory tract pathogen but there is growing evidence that RSV may lead to CNS impairments. However, the mechanisms involved in RSV entering into the CNS have been poorly described. In this study, we wanted to explore the capacity of RSV to reach the CNS via the olfactory pathway and to better characterize RSV cellular tropism in the nasal cavity. We first explored the distribution of RSV infectious sites in the nasal cavity by in vivo bioluminescence imaging and a tissue clearing protocol combined with deep-tissue imaging and 3D image analyses. This whole tissue characterization was confirmed with immunohistochemistry and molecular biology approaches. Together, our results provide a novel 3D atlas of mouse nasal cavity anatomy and show that RSV can infect olfactory sensory neurons giving access to the central nervous system by entering the olfactory bulb. Cover Image for this issue: doi: 10.1111/jnc.14765., (© 2019 International Society for Neurochemistry.)
- Published
- 2020
- Full Text
- View/download PDF
47. Ventral midline thalamus is not necessary for systemic consolidation of a social memory in the rat.
- Author
-
Quet E, Cassel JC, Cosquer B, Galloux M, Pereira De Vasconcelos A, and Stéphan A
- Abstract
According to the standard theory of memory consolidation, recent memories are stored in the hippocampus before their transfer to cortical modules, a process called systemic consolidation. The ventral midline thalamus (reuniens and rhomboid nuclei, ReRh) takes part in this transfer as its lesion disrupts systemic consolidation of spatial and contextual fear memories. Here, we wondered whether ReRh lesions would also affect the systemic consolidation of another type of memory, namely an olfaction-based social memory. To address this question we focused on social transmission of food preference. Adult Long-Evans rats were subjected to N-methyl-d-aspartate-induced, fibre-sparing lesions of the ReRh nuclei or to a sham-operation, and subsequently trained in a social transmission of food preference paradigm. Retrieval was tested on the next day (recent memory, n
Sham = 10, nReRh = 12) or after a 25-day delay (remote memory, nSham = 10, nReRh = 10). All rats, whether sham-operated or subjected to ReRh lesions, learned and remembered the task normally, whatever the delay. Compared to our former results on spatial and contextual fear memories (Ali et al., 2017; Klein et al., 2019; Loureiro et al., 2012; Quet et al., 2020), the present findings indicate that the ReRh nuclei might not be part of a generic, systemic consolidation mechanism processing all kinds of memories in order to make them persistent. The difference between social transmission of food preference and spatial or contextual fear memories could be explained by the fact that social transmission of food preference is not hippocampus-dependent and that the persistence of social transmission of food preference memory relies on different circuits., Competing Interests: Declaration of conflicting interests: The author(s) declared no potential conflicts of interest with respect to the research, authorship and/or publication of this article., (© The Author(s) 2020.)- Published
- 2020
- Full Text
- View/download PDF
48. De novo protein design enables the precise induction of RSV-neutralizing antibodies.
- Author
-
Sesterhenn F, Yang C, Bonet J, Cramer JT, Wen X, Wang Y, Chiang CI, Abriata LA, Kucharska I, Castoro G, Vollers SS, Galloux M, Dheilly E, Rosset S, Corthésy P, Georgeon S, Villard M, Richard CA, Descamps D, Delgado T, Oricchio E, Rameix-Welti MA, Más V, Ervin S, Eléouët JF, Riffault S, Bates JT, Julien JP, Li Y, Jardetzky T, Krey T, and Correia BE
- Subjects
- Amino Acid Motifs, Humans, Immunodominant Epitopes immunology, Protein Conformation, Recombinant Fusion Proteins immunology, Respiratory Syncytial Virus Vaccines immunology, Single-Domain Antibodies chemistry, Single-Domain Antibodies immunology, Antibodies, Neutralizing biosynthesis, Computational Biology methods, Immunodominant Epitopes chemistry, Protein Engineering methods, Recombinant Fusion Proteins chemistry, Respiratory Syncytial Virus Vaccines chemistry, Respiratory Syncytial Virus, Human immunology
- Abstract
De novo protein design has been successful in expanding the natural protein repertoire. However, most de novo proteins lack biological function, presenting a major methodological challenge. In vaccinology, the induction of precise antibody responses remains a cornerstone for next-generation vaccines. Here, we present a protein design algorithm called TopoBuilder, with which we engineered epitope-focused immunogens displaying complex structural motifs. In both mice and nonhuman primates, cocktails of three de novo-designed immunogens induced robust neutralizing responses against the respiratory syncytial virus. Furthermore, the immunogens refocused preexisting antibody responses toward defined neutralization epitopes. Overall, our design approach opens the possibility of targeting specific epitopes for the development of vaccines and therapeutic antibodies and, more generally, will be applicable to the design of de novo proteins displaying complex functional motifs., (Copyright © 2020 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works.)
- Published
- 2020
- Full Text
- View/download PDF
49. Labyrinthopeptins as virolytic inhibitors of respiratory syncytial virus cell entry.
- Author
-
Blockus S, Sake SM, Wetzke M, Grethe C, Graalmann T, Pils M, Le Goffic R, Galloux M, Prochnow H, Rox K, Hüttel S, Rupcic Z, Wiegmann B, Dijkman R, Rameix-Welti MA, Eléouët JF, Duprex WP, Thiel V, Hansen G, Brönstrup M, Haid S, and Pietschmann T
- Subjects
- Animals, Cell Line, Cells, Cultured, Female, Humans, Lung cytology, Mice, Mice, Inbred BALB C, Respiratory Syncytial Virus, Human physiology, Antiviral Agents pharmacology, Bacteriocins pharmacology, Respiratory Syncytial Virus Infections drug therapy, Respiratory Syncytial Virus, Human drug effects, Virus Internalization drug effects
- Abstract
Acute lower respiratory tract infections (ALRI) caused by respiratory syncytial virus (RSV) are associated with a severe disease burden among infants and elderly patients. Treatment options are limited. While numerous drug candidates with different viral targets are under development, the utility of RSV entry inhibitors is challenged by a low resistance barrier and by single mutations causing cross-resistance against a wide spectrum of fusion inhibitor chemotypes. We developed a cell-based screening assay for discovery of compounds inhibiting infection with primary RSV isolates. Using this system, we identified labyrinthopeptin A1 and A2 (Laby A1/A2), lantibiotics isolated from Actinomadura namibiensis, as effective RSV cell entry inhibitors with IC
50 s of 0.39 μM and 4.97 μM, respectively, and with favourable therapeutic index (>200 and > 20, respectively). Both molecules were active against multiple RSV strains including primary isolates and their antiviral activity against RSV was confirmed in primary human airway cells ex vivo and a murine model in vivo. Laby A1/A2 were antiviral in prophylactic and therapeutic treatment regimens and displayed synergistic activity when applied in combination with each other. Mechanistic studies showed that Laby A1/A2 exert virolytic activity likely by binding to phosphatidylethanolamine moieties within the viral membrane and by disrupting virus particle membrane integrity. Probably due to its specific mode of action, Laby A1/A2 antiviral activity was not affected by common resistance mutations to known RSV entry inhibitors. Taken together, Laby A1/A2 represent promising candidates for development as RSV inhibitors. Moreover, the cell-based screening system with primary RSV isolates described here should be useful to identify further antiviral agents., Competing Interests: Declaration of competing interest The antiviral activity of Laby A1/A2 against a broad spectrum of enveloped viruses has been patented (PCT/EP2016/078143). Otherwise, no competing financial interests exist., (Copyright © 2020 The Authors. Published by Elsevier B.V. All rights reserved.)- Published
- 2020
- Full Text
- View/download PDF
50. Biochemical characterization of the respiratory syncytial virus N 0 -P complex in solution.
- Author
-
Esneau C, Raynal B, Roblin P, Brûlé S, Richard CA, Fix J, Eléouët JF, and Galloux M
- Subjects
- Binding Sites, DNA-Directed RNA Polymerases genetics, DNA-Directed RNA Polymerases metabolism, Models, Molecular, Mutation, Nucleoproteins genetics, Protein Conformation, Solutions, Surface Properties, Viral Proteins genetics, Nucleoproteins chemistry, Nucleoproteins metabolism, Respiratory Syncytial Virus, Human, Viral Proteins chemistry, Viral Proteins metabolism
- Abstract
As all the viruses belonging to the Mononegavirales order, the nonsegmented negative-strand RNA genome of respiratory syncytial virus (RSV) is encapsidated by the viral nucleoprotein N. N protein polymerizes along the genomic and anti-genomic RNAs during replication. This requires the maintenance of the neosynthesized N protein in a monomeric and RNA-free form by the viral phosphoprotein P that plays the role of a chaperone protein, forming a soluble N
0 -P complex. We have previously demonstrated that residues 1-30 of P specifically bind to N0 Here, to isolate a stable N0 -P complex suitable for structural studies, we used the N-terminal peptide of P (P40) to purify truncated forms of the N protein. We show that to purify a stable N0 -P-like complex, a deletion of the first 30 N-terminal residues of N (NΔ30 ) is required to impair N oligomerization, whereas the presence of a full-length C-arm of N is required to inhibit RNA binding. We generated structural models of the RSV N0 -P with biophysical approaches, including hydrodynamic measurements and small-angle X-ray scattering (SAXS), coupled with biochemical and functional analyses of human RSV (hRSV) NΔ30 mutants. These models suggest a strong structural homology between the hRSV and the human metapneumovirus (hMPV) N0 -P complexes. In both complexes, the P40-binding sites on N0 appear to be similar, and the C-arm of N provides a high flexibility and a propensity to interact with the N RNA groove. These findings reveal two potential sites to target on N0 -P for the development of RSV antivirals., (© 2019 Esneau et al.)- Published
- 2019
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.