80 results on '"Hanoulle X"'
Search Results
2. VHH Z70 mutant 1 in interaction with PHF6 Tau peptide
- Author
-
Dupre, E., primary, Mortelecque, J., additional, NGuyen, M., additional, Hanoulle, X., additional, and Landrieu, I., additional
- Published
- 2023
- Full Text
- View/download PDF
3. VHH Z70 mutant 20 in interaction with PHF6 Tau peptide
- Author
-
Dupre, E., primary, Mortelecque, J., additional, NGuyen, M., additional, Hanoulle, X., additional, and Landrieu, I., additional
- Published
- 2023
- Full Text
- View/download PDF
4. SARS-CoV-2 Main Protease complexed with N-(pyridin-3-ylmethyl)thioformamide
- Author
-
Hanoulle, X., primary, Charton, J., additional, and Deprez, B., additional
- Published
- 2023
- Full Text
- View/download PDF
5. Crystal structure of the SARS-CoV-2 Main Protease complexed with N-(pyridin-3-ylmethyl)thioformamide
- Author
-
Dupre, E., primary, Villeret, V., additional, and Hanoulle, X., additional
- Published
- 2022
- Full Text
- View/download PDF
6. Crystallographic structure of human Tsg101 UEV domain in complex with a HEV ORF3 peptide
- Author
-
Moschidi, D., primary, Dupre, E., additional, Villeret, V., additional, and Hanoulle, X., additional
- Published
- 2022
- Full Text
- View/download PDF
7. Crystal structure of the SARS-CoV-2 Main Protease
- Author
-
Dupre, E., primary, Villeret, V., additional, and Hanoulle, X., additional
- Published
- 2022
- Full Text
- View/download PDF
8. Crystal structure of the SARS-CoV-2 Main Protease with a Zinc ion coordinated in the active site
- Author
-
Dupre, E., primary, Villeret, V., additional, and Hanoulle, X., additional
- Published
- 2022
- Full Text
- View/download PDF
9. CRYSTAL STRUCTURE OF THE SARS-COV-2 MAIN PROTEASE COMPLEXED WITH FRAGMENT F01
- Author
-
Hanoulle, X., primary and Moschidi, D., additional
- Published
- 2021
- Full Text
- View/download PDF
10. Crystal structure of the SARS-CoV-2 Main Protease with oxidized C145
- Author
-
Dupre, E., primary, Villeret, V., additional, and Hanoulle, X., additional
- Published
- 2021
- Full Text
- View/download PDF
11. NMR Structure of NS5A-D2 (JFH1) peptide (304-323)
- Author
-
Dujardin, M., primary, Cantrelle, F.X., additional, Lippens, G., additional, and Hanoulle, X., additional
- Published
- 2019
- Full Text
- View/download PDF
12. Control of glycokinase activity by the HCV protein NS5A increases lipogenesis
- Author
-
Jacquemin, C., primary, Panthu, B., additional, Ramière, C., additional, Hanoulle, X., additional, Perrin-Cocon, L., additional, Andre, P., additional, Lotteau, V., additional, and Diaz, O., additional
- Published
- 2018
- Full Text
- View/download PDF
13. In-cell NMR: from metabolites to macromolecules
- Author
-
Lippens, G., primary, Cahoreau, E., additional, Millard, P., additional, Charlier, C., additional, Lopez, J., additional, Hanoulle, X., additional, and Portais, J. C., additional
- Published
- 2018
- Full Text
- View/download PDF
14. HUMAN MED26 N-TERMINAL DOMAIN (1-92)
- Author
-
Lens, Z., primary, Cantrelle, F.-X., additional, Perruzini, R., additional, Dewitte, F., additional, Hanoulle, X., additional, Villeret, V., additional, Verger, A., additional, and Landrieu, I., additional
- Published
- 2017
- Full Text
- View/download PDF
15. X-Ray structure of a CypA-Alisporivir complex at 1.5 angstrom resolution
- Author
-
Dujardin, M., primary, Bouckaert, J., additional, Rucktooa, P., additional, and Hanoulle, X., additional
- Published
- 2017
- Full Text
- View/download PDF
16. SAT-380 - Control of glycokinase activity by the HCV protein NS5A increases lipogenesis
- Author
-
Jacquemin, C., Panthu, B., Ramière, C., Hanoulle, X., Perrin-Cocon, L., Andre, P., Lotteau, V., and Diaz, O.
- Published
- 2018
- Full Text
- View/download PDF
17. Ns5a308
- Author
-
Montserret, R., primary, Badillo, A., additional, Hanoulle, X., additional, Lippens, G., additional, and Penin, F., additional
- Published
- 2014
- Full Text
- View/download PDF
18. Studying Posttranslational Modifications by In-Cell NMR
- Author
-
Lippens, G., primary, Landrieu, I., additional, and Hanoulle, X., additional
- Published
- 2008
- Full Text
- View/download PDF
19. N-acylbenzimidazoles as selective Acylators of the catalytic cystein of the coronavirus 3CL protease.
- Author
-
Chaibi FZ, Brier L, Carré P, Landry V, Desmarets L, Tarricone A, Cantrelle FX, Moschidi D, Herledan A, Biela A, Bourgeois F, Ribes C, Ikherbane S, Malessan M, Dubuisson J, Belouzard S, Hanoulle X, Leroux F, Deprez B, and Charton J
- Subjects
- Structure-Activity Relationship, Humans, Cysteine Endopeptidases metabolism, Acylation, Cysteine chemistry, Cysteine pharmacology, Molecular Structure, Dose-Response Relationship, Drug, Protease Inhibitors pharmacology, Protease Inhibitors chemical synthesis, Protease Inhibitors chemistry, Models, Molecular, Drug Design, Crystallography, X-Ray, Coronavirus 3C Proteases antagonists & inhibitors, Coronavirus 3C Proteases metabolism, Antiviral Agents pharmacology, Antiviral Agents chemistry, Antiviral Agents chemical synthesis, SARS-CoV-2 drug effects, SARS-CoV-2 enzymology, Benzimidazoles pharmacology, Benzimidazoles chemistry, Benzimidazoles chemical synthesis
- Abstract
The 3CL protease (3CL
pro , Mpro ) plays a key role in the replication of the SARS-CoV-2 and was validated as therapeutic target by the development and approval of specific antiviral drugs (nirmatrelvir, ensitrelvir), inhibitors of this protease. Moreover, its high conservation within the coronavirus family renders it an attractive therapeutic target for the development of anti-coronavirus compounds with broad spectrum activity to control COVID-19 and future coronavirus diseases. Here we report on the design, synthesis and structure-activity relationships of a new series of small covalent reversible inhibitors of the SARS-CoV-2 3CLpro . As elucidated thanks to the X-Ray structure of some inhibitors with the 3CLpro , the mode of inhibition involves acylation of the thiol of the catalytic cysteine. The synthesis of 60 analogs led to the identification of compound 56 that inhibits the SARS-CoV-2 3CLpro with high potency (IC50 = 70 nM) and displays antiviral activity in cells (EC50 = 3.1 μM). Notably, compound 56 inhibits the 3CLpro of three other human coronaviruses and exhibit a good selectivity against two human cysteine proteases. These results demonstrate the potential of this electrophilic N-acylbenzimidazole series as a basis for further optimization., Competing Interests: Declaration of competing interest 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 © 2024. Published by Elsevier Masson SAS.)- Published
- 2024
- Full Text
- View/download PDF
20. A selection and optimization strategy for single-domain antibodies targeting the PHF6 linear peptide within the tau intrinsically disordered protein.
- Author
-
Mortelecque J, Zejneli O, Bégard S, Simões MC, ElHajjar L, Nguyen M, Cantrelle FX, Hanoulle X, Rain JC, Colin M, Gomes CM, Buée L, Landrieu I, Danis C, and Dupré E
- Subjects
- Humans, Epitopes chemistry, Epitopes immunology, Peptides chemistry, Peptides immunology, Intrinsically Disordered Proteins chemistry, Intrinsically Disordered Proteins immunology, Single-Domain Antibodies chemistry, Single-Domain Antibodies genetics, Single-Domain Antibodies immunology, tau Proteins chemistry, tau Proteins immunology
- Abstract
The use of variable domain of the heavy-chain of the heavy-chain-only antibodies (VHHs) as disease-modifying biomolecules in neurodegenerative disorders holds promises, including targeting of aggregation-sensitive proteins. Exploitation of their clinical values depends however on the capacity to deliver VHHs with optimal physico-chemical properties for their specific context of use. We described previously a VHH with high therapeutic potential in a family of neurodegenerative diseases called tauopathies. The activity of this promising parent VHH named Z70 relies on its binding within the central region of the tau protein. Accordingly, we carried out random mutagenesis followed by yeast two-hybrid screening to obtain optimized variants. The VHHs selected from this initial screen targeted the same epitope as VHH Z70 as shown using NMR spectroscopy and had indeed improved binding affinities according to dissociation constant values obtained by surface plasmon resonance spectroscopy. The improved affinities can be partially rationalized based on three-dimensional structures and NMR data of three complexes consisting of an optimized VHH and a peptide containing the tau epitope. Interestingly, the ability of the VHH variants to inhibit tau aggregation and seeding could not be predicted from their affinity alone. We indeed showed that the in vitro and in cellulo VHH stabilities are other limiting key factors to their efficacy. Our results demonstrate that only a complete pipeline of experiments, here described, permits a rational selection of optimized VHH variants, resulting in the selection of VHH variants with higher affinities and/or acting against tau seeding in cell models., Competing Interests: Conflict of interests J.-C. R. is the CEO of Hybrigenic services. Part of the work is included in patent WO2020120644 NEW ANTI TAU SINGLE DOMAIN ANTIBODY L. I., B. L., D. E., D. C., R. J.-C., and A. A. The other authors declare that they have no conflicts of interest with the contents of this article., (Copyright © 2024 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
21. Magnetic resonance investigation of conformational responses of tau protein to specific phosphorylation.
- Author
-
Lasorsa A, Merzougui H, Cantrelle FX, Sicoli G, Dupré E, Hanoulle X, Belle V, Smet-Nocca C, and Landrieu I
- Subjects
- Phosphorylation, Magnetic Resonance Spectroscopy, Protein Conformation, Electron Spin Resonance Spectroscopy, Nuclear Magnetic Resonance, Biomolecular, tau Proteins chemistry, Intrinsically Disordered Proteins chemistry
- Abstract
Intrinsically disordered proteins (IDPs) are known to adopt many rapidly interconverting structures, making it difficult to pinpoint the specific conformational states that are relevant for their function. Tau is an important IDP, and its conformation is known to be affected by post-translational modifications (PTMs), such as phosphorylation. To investigate the effect of specific phosphorylation on full-length Tau's dynamic global conformation, we employed a combination of nuclear magnetic resonance-based paramagnetic relaxation interference methods and electron paramagnetic resonance spectroscopy. By reproducing the AT8 epitope, comprising exclusive phosphorylation at residues S202 and T205, we were able to identify conformations specific to phosphorylated Tau, which exhibited a tendency towards less compact states. These mechanistic details are of significance to understand the path leading from soluble Tau to the ordered structure of Tau fibers. This approach proved to be successful for studying the conformational changes of (phosphorylated) full-length Tau and can potentially be extended to the study of other IDPs that undergo various PTMs., Competing Interests: Declaration of Competing Interest The authors declare the submitted work was carried out without the presence of any personal, professional or financial relationships that could potentially be construed as a conflict of interest., (Copyright © 2023 Elsevier B.V. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
22. The phosphatidylserine receptor TIM1 promotes infection of enveloped hepatitis E virus.
- Author
-
Corneillie L, Lemmens I, Montpellier C, Ferrié M, Weening K, Van Houtte F, Hanoulle X, Cocquerel L, Amara A, Tavernier J, and Meuleman P
- Subjects
- Virus Internalization, Receptors, Cell Surface metabolism, Hepatitis E virus genetics, Hepatitis E virus metabolism, Viruses
- Abstract
The hepatitis E virus (HEV) is an underestimated RNA virus of which the viral life cycle and pathogenicity remain partially understood and for which specific antivirals are lacking. The virus exists in two forms: nonenveloped HEV that is shed in feces and transmits between hosts; and membrane-associated, quasi-enveloped HEV that circulates in the blood. It is suggested that both forms employ different mechanisms for cellular entry and internalization but little is known about the exact mechanisms. Interestingly, the membrane of enveloped HEV is enriched with phosphatidylserine, a natural ligand for the T-cell immunoglobulin and mucin domain-containing protein 1 (TIM1) during apoptosis and involved in 'apoptotic mimicry', a process by which viruses hijack the apoptosis pathway to promote infection. We here investigated the role of TIM1 in the entry process of HEV. We determined that HEV infection with particles derived from culture supernatant, which are cloaked by host-derived membranes (eHEV), was significantly impaired after knockout of TIM1, whereas infection with intracellular HEV particles (iHEV) was unaffected. eHEV infection was restored upon TIM1 expression; and enhanced after ectopic TIM1 expression. The significance of TIM1 during entry was further confirmed by viral binding assay, and point mutations of the PS-binding pocket diminished eHEV infection. In addition, Annexin V, a PS-binding molecule also significantly reduced infection. Taken together, our findings support a role for TIM1 in eHEV-mediated cell entry, facilitated by the PS present on the viral membrane, a strategy HEV may use to promote viral spread throughout the infected body., (© 2023. The Author(s), under exclusive licence to Springer Nature Switzerland AG.)
- Published
- 2023
- Full Text
- View/download PDF
23. Conformation and Affinity Modulations by Multiple Phosphorylation Occurring in the BIN1 SH3 Domain Binding Site of the Tau Protein Proline-Rich Region.
- Author
-
Lasorsa A, Bera K, Malki I, Dupré E, Cantrelle FX, Merzougui H, Sinnaeve D, Hanoulle X, Hritz J, and Landrieu I
- Subjects
- Humans, Phosphorylation, src Homology Domains, Protein Binding, Peptides chemistry, Binding Sites, Proline metabolism, Nuclear Proteins metabolism, Tumor Suppressor Proteins chemistry, Adaptor Proteins, Signal Transducing metabolism, tau Proteins metabolism, Alzheimer Disease metabolism
- Abstract
An increase in phosphorylation of the Tau protein is associated with Alzheimer's disease (AD) progression through unclear molecular mechanisms. In general, phosphorylation modifies the interaction of intrinsically disordered proteins, such as Tau, with other proteins; however, elucidating the structural basis of this regulation mechanism remains challenging. The bridging integrator-1 gene is an AD genetic determinant whose gene product, BIN1, directly interacts with Tau. The proline-rich motif recognized within a Tau(210-240) peptide by the SH3 domain of BIN1 (BIN1 SH3) is defined as
216 PTPP219 , and this interaction is modulated by phosphorylation. Phosphorylation of T217 within the Tau(210-240) peptide led to a 6-fold reduction in the affinity, while single phosphorylation at either T212, T231, or S235 had no effect on the interaction. Nonetheless, combined phosphorylation of T231 and S235 led to a 3-fold reduction in the affinity, although these phosphorylations are not within the BIN1 SH3-bound region of the Tau peptide. Using nuclear magnetic resonance (NMR) spectroscopy, these phosphorylations were shown to affect the local secondary structure and dynamics of the Tau(210-240) peptide. Models of the (un)phosphorylated peptides were obtained from molecular dynamics (MD) simulation validated by experimental data and showed compaction of the phosphorylated peptide due to increased salt bridge formation. This dynamic folding might indirectly impact the BIN1 SH3 binding by a decreased accessibility of the binding site. Regulation of the binding might thus not only be due to local electrostatic or steric effects from phosphorylation but also to the modification of the conformational properties of Tau.- Published
- 2023
- Full Text
- View/download PDF
24. Backbone NMR resonance assignment of the apo human Tsg101-UEV domain.
- Author
-
Moschidi D, Cantrelle FX, Boll E, and Hanoulle X
- Subjects
- Humans, Nuclear Magnetic Resonance, Biomolecular, Endosomal Sorting Complexes Required for Transport chemistry, Endosomal Sorting Complexes Required for Transport genetics, Endosomal Sorting Complexes Required for Transport metabolism, Ubiquitin metabolism, DNA-Binding Proteins chemistry
- Abstract
The Endosomal Sorting Complex Required for Transport (ESCRT) pathway, through inverse topology membrane remodeling, is involved in many biological functions, such as ubiquitinated membrane receptor trafficking and degradation, multivesicular bodies (MVB) formation and cytokinesis. Dysfunctions in ESCRT pathway have been associated to several human pathologies, such as cancers and neurodegenerative diseases. The ESCRT machinery is also hijacked by many enveloped viruses to bud away from the plasma membrane of infected cells. Human tumor susceptibility gene 101 (Tsg101) protein is an important ESCRT-I complex component. The structure of the N-terminal ubiquitin E2 variant (UEV) domain of Tsg101 (Tsg101-UEV) comprises an ubiquitin binding pocket next to a late domain [P(S/T)AP] binding groove. These two binding sites have been shown to be involved both in the physiological roles of ESCRT-I and in the release of the viral particles, and thus are attractive targets for antivirals. The structure of the Tsg101-UEV domain has been characterized, using X-ray crystallography or NMR spectroscopy, either in its apo-state or bound to ubiquitin or late domains. In this study, we report the backbone NMR resonance assignments, including the proline signals, of the apo human Tsg101-UEV domain, that so far was not publicly available. These data, that are in good agreement with the crystallographic structure of Tsg101-UEV domain, can therefore be used for further NMR studies, including protein-protein interaction studies and drug discovery., (© 2023. The Author(s), under exclusive licence to Springer Nature B.V.)
- Published
- 2023
- Full Text
- View/download PDF
25. Novel dithiocarbamates selectively inhibit 3CL protease of SARS-CoV-2 and other coronaviruses.
- Author
-
Brier L, Hassan H, Hanoulle X, Landry V, Moschidi D, Desmarets L, Rouillé Y, Dumont J, Herledan A, Warenghem S, Piveteau C, Carré P, Ikherbane S, Cantrelle FX, Dupré E, Dubuisson J, Belouzard S, Leroux F, Deprez B, and Charton J
- Subjects
- Humans, Peptide Hydrolases, Cysteine Endopeptidases metabolism, Protease Inhibitors chemistry, Coronavirus 3C Proteases, Antiviral Agents chemistry, SARS-CoV-2 metabolism, COVID-19
- Abstract
Since end of 2019, the global and unprecedented outbreak caused by the coronavirus SARS-CoV-2 led to dramatic numbers of infections and deaths worldwide. SARS-CoV-2 produces two large viral polyproteins which are cleaved by two cysteine proteases encoded by the virus, the 3CL protease (3CL
pro ) and the papain-like protease, to generate non-structural proteins essential for the virus life cycle. Both proteases are recognized as promising drug targets for the development of anti-coronavirus chemotherapy. Aiming at identifying broad spectrum agents for the treatment of COVID-19 but also to fight emergent coronaviruses, we focused on 3CLpro that is well conserved within this viral family. Here we present a high-throughput screening of more than 89,000 small molecules that led to the identification of a new chemotype, potent inhibitor of the SARS-CoV-2 3CLpro . The mechanism of inhibition, the interaction with the protease using NMR and X-Ray, the specificity against host cysteine proteases and promising antiviral properties in cells are reported., Competing Interests: Declaration of competing interest 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 © 2023 Elsevier Masson SAS. All rights reserved.)- Published
- 2023
- Full Text
- View/download PDF
26. Exploring the Antitubercular Activity of Anthranilic Acid Derivatives: From MabA (FabG1) Inhibition to Intrabacterial Acidification.
- Author
-
Faïon L, Djaout K, Pintiala C, Piveteau C, Leroux F, Biela A, Slupek S, Antoine R, Záhorszká M, Cantrelle FX, Hanoulle X, Korduláková J, Deprez B, Willand N, Baulard AR, and Flipo M
- Abstract
Mycobacterium tuberculosis , the pathogen that causes tuberculosis, is responsible for the death of 1.5 million people each year and the number of bacteria resistant to the standard regimen is constantly increasing. This highlights the need to discover molecules that act on new M. tuberculosis targets. Mycolic acids, which are very long-chain fatty acids essential for M. tuberculosis viability, are synthesized by two types of fatty acid synthase (FAS) systems. MabA (FabG1) is an essential enzyme belonging to the FAS-II cycle. We have recently reported the discovery of anthranilic acids as MabA inhibitors. Here, the structure-activity relationships around the anthranilic acid core, the binding of a fluorinated analog to MabA by NMR experiments, the physico-chemical properties and the antimycobacterial activity of these inhibitors were explored. Further investigation of the mechanism of action in bacterio showed that these compounds affect other targets than MabA in mycobacterial cells and that their antituberculous activity is due to the carboxylic acid moiety which induces intrabacterial acidification.
- Published
- 2023
- Full Text
- View/download PDF
27. Inhibition of Tau seeding by targeting Tau nucleation core within neurons with a single domain antibody fragment.
- Author
-
Danis C, Dupré E, Zejneli O, Caillierez R, Arrial A, Bégard S, Mortelecque J, Eddarkaoui S, Loyens A, Cantrelle FX, Hanoulle X, Rain JC, Colin M, Buée L, and Landrieu I
- Subjects
- Animals, Disease Models, Animal, Mice, Neurons metabolism, Repressor Proteins, tau Proteins genetics, Alzheimer Disease metabolism, Single-Domain Antibodies, Tauopathies metabolism
- Abstract
Tau proteins aggregate into filaments in brain cells in Alzheimer's disease and related disorders referred to as tauopathies. Here, we used fragments of camelid heavy-chain-only antibodies (VHHs or single domain antibody fragments) targeting Tau as immuno-modulators of its pathologic seeding. A VHH issued from the screen against Tau of a synthetic phage-display library of humanized VHHs was selected for its capacity to bind Tau microtubule-binding domain, composing the core of Tau fibrils. This parent VHH was optimized to improve its biochemical properties and to act in the intra-cellular compartment, resulting in VHH Z70. VHH Z70 precisely binds the PHF6 sequence, known for its nucleation capacity, as shown by the crystal structure of the complex. VHH Z70 was more efficient than the parent VHH to inhibit in vitro Tau aggregation in heparin-induced assays. Expression of VHH Z70 in a cellular model of Tau seeding also decreased the aggregation-reporting fluorescence signal. Finally, intra-cellular expression of VHH Z70 in the brain of an established tauopathy mouse seeding model demonstrated its capacity to mitigate accumulation of pathological Tau. VHH Z70, by targeting Tau inside brain neurons, where most of the pathological Tau resides, provides an immunological tool to target the intra-cellular compartment in tauopathies., Competing Interests: Declaration of interests A.A. and J.-C.R. are employees of Hybrigenic services., (Copyright © 2022 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
28. Domain 2 of Hepatitis C Virus Protein NS5A Activates Glucokinase and Induces Lipogenesis in Hepatocytes.
- Author
-
Perrin-Cocon L, Kundlacz C, Jacquemin C, Hanoulle X, Aublin-Gex A, Figl M, Manteca J, André P, Vidalain PO, Lotteau V, and Diaz O
- Subjects
- Cell Line, Tumor, Gene Knockdown Techniques, Glycogen metabolism, Glycolysis, Host-Pathogen Interactions, Humans, Lipid Metabolism, Lipogenesis, Mitochondria metabolism, Protein Binding, Protein Interaction Domains and Motifs, RNA-Dependent RNA Polymerase chemistry, Viral Nonstructural Proteins chemistry, Glucokinase metabolism, Hepacivirus physiology, Hepatitis C metabolism, Hepatitis C virology, Hepatocytes metabolism, Hepatocytes virology, RNA-Dependent RNA Polymerase metabolism, Viral Nonstructural Proteins metabolism
- Abstract
Hepatitis C virus (HCV) relies on cellular lipid metabolism for its replication, and actively modulates lipogenesis and lipid trafficking in infected hepatocytes. This translates into an intracellular accumulation of triglycerides leading to liver steatosis, cirrhosis and hepatocellular carcinoma, which are hallmarks of HCV pathogenesis. While the interaction of HCV with hepatocyte metabolic pathways is patent, how viral proteins are able to redirect central carbon metabolism towards lipogenesis is unclear. Here, we report that the HCV protein NS5A activates the glucokinase (GCK) isoenzyme of hexokinases through its D2 domain (NS5A-D2). GCK is the first rate-limiting enzyme of glycolysis in normal hepatocytes whose expression is replaced by the hexokinase 2 (HK2) isoenzyme in hepatocellular carcinoma cell lines. We took advantage of a unique cellular model specifically engineered to re-express GCK instead of HK2 in the Huh7 cell line to evaluate the consequences of NS5A-D2 expression on central carbon and lipid metabolism. NS5A-D2 increased glucose consumption but decreased glycogen storage. This was accompanied by an altered mitochondrial respiration, an accumulation of intracellular triglycerides and an increased production of very-low density lipoproteins. Altogether, our results show that NS5A-D2 can reprogram central carbon metabolism towards a more energetic and glycolytic phenotype compatible with HCV needs for replication.
- Published
- 2022
- Full Text
- View/download PDF
29. Hepatitis E virus RNA-dependent RNA polymerase is involved in RNA replication and infectious particle production.
- Author
-
Oechslin N, Da Silva N, Szkolnicka D, Cantrelle FX, Hanoulle X, Moradpour D, and Gouttenoire J
- Subjects
- Hep G2 Cells, Hepatitis E virus genetics, Humans, Mutation, Protein Conformation, alpha-Helical genetics, RNA, Viral metabolism, RNA-Dependent RNA Polymerase genetics, RNA-Dependent RNA Polymerase ultrastructure, Structure-Activity Relationship, Hepatitis E virology, Hepatitis E virus pathogenicity, RNA-Dependent RNA Polymerase metabolism, Virus Replication genetics
- Abstract
Background and Aims: Hepatitis E virus (HEV) is one of the most common causes of acute hepatitis worldwide. Its positive-strand RNA genome encodes three open reading frames (ORF). ORF1 is translated into a large protein composed of multiple domains and is known as the viral replicase. The RNA-dependent RNA polymerase (RDRP) domain is responsible for the synthesis of viral RNA., Approach and Results: Here, we identified a highly conserved α-helix located in the RDRP thumb subdomain. Nuclear magnetic resonance demonstrated an amphipathic α-helix extending from amino acids 1628 to 1644 of the ORF1 protein. Functional analyses revealed a dual role of this helix in HEV RNA replication and virus production, including assembly and release. Mutations on the hydrophobic side of the amphipathic α-helix impaired RNA replication and resulted in the selection of a second-site compensatory change in the RDRP palm subdomain. Other mutations enhanced RNA replication but impaired virus assembly and/or release., Conclusions: Structure-function analyses identified a conserved amphipathic α-helix in the thumb subdomain of the HEV RDRP with a dual role in viral RNA replication and infectious particle production. This study provides structural insights into a key segment of the ORF1 protein and describes the successful use of reverse genetics in HEV, revealing functional interactions between the RDRP thumb and palm subdomains. On a broader scale, it demonstrates that the HEV replicase, similar to those of other positive-strand RNA viruses, is also involved in virus production., (© 2021 The Authors. Hepatology published by Wiley Periodicals LLC on behalf of American Association for the Study of Liver Diseases.)
- Published
- 2022
- Full Text
- View/download PDF
30. NMR Spectroscopy of the Main Protease of SARS-CoV-2 and Fragment-Based Screening Identify Three Protein Hotspots and an Antiviral Fragment.
- Author
-
Cantrelle FX, Boll E, Brier L, Moschidi D, Belouzard S, Landry V, Leroux F, Dewitte F, Landrieu I, Dubuisson J, Deprez B, Charton J, and Hanoulle X
- Subjects
- Animals, Antiviral Agents chemistry, Binding Sites, Chlorocebus aethiops, Coronavirus 3C Proteases chemistry, Cysteine Proteinase Inhibitors chemistry, Drug Evaluation, Preclinical, Microbial Sensitivity Tests, Nuclear Magnetic Resonance, Biomolecular, Protein Conformation, Protein Multimerization, SARS-CoV-2 chemistry, Small Molecule Libraries chemistry, Vero Cells, Antiviral Agents pharmacology, Coronavirus 3C Proteases antagonists & inhibitors, Cysteine Proteinase Inhibitors pharmacology, SARS-CoV-2 drug effects, Small Molecule Libraries pharmacology
- Abstract
The main protease (3CLp) of the SARS-CoV-2, the causative agent for the COVID-19 pandemic, is one of the main targets for drug development. To be active, 3CLp relies on a complex interplay between dimerization, active site flexibility, and allosteric regulation. The deciphering of these mechanisms is a crucial step to enable the search for inhibitors. In this context, using NMR spectroscopy, we studied the conformation of dimeric 3CLp from the SARS-CoV-2 and monitored ligand binding, based on NMR signal assignments. We performed a fragment-based screening that led to the identification of 38 fragment hits. Their binding sites showed three hotspots on 3CLp, two in the substrate binding pocket and one at the dimer interface. F01 is a non-covalent inhibitor of the 3CLp and has antiviral activity in SARS-CoV-2 infected cells. This study sheds light on the complex structure-function relationships of 3CLp and constitutes a strong basis to assist in developing potent 3CLp inhibitors., (© 2021 Wiley-VCH GmbH.)
- Published
- 2021
- Full Text
- View/download PDF
31. Correction to: Alzheimer's genetic risk factor FERMT2 (Kindlin-2) controls axonal growth and synaptic plasticity in an APP-dependent manner.
- Author
-
Eysert F, Coulon A, Boscher E, Vreulx AC, Flaig A, Mendes T, Hughes S, Grenier-Boley B, Hanoulle X, Demiautte F, Bauer C, Marttinen M, Takalo M, Amouyel P, Desai S, Pike I, Hiltunen M, Chécler F, Farinelli M, Delay C, Malmanche N, Hébert SS, Dumont J, Kilinc D, Lambert JC, and Chapuis J
- Published
- 2021
- Full Text
- View/download PDF
32. Alzheimer's genetic risk factor FERMT2 (Kindlin-2) controls axonal growth and synaptic plasticity in an APP-dependent manner.
- Author
-
Eysert F, Coulon A, Boscher E, Vreulx AC, Flaig A, Mendes T, Hughes S, Grenier-Boley B, Hanoulle X, Demiautte F, Bauer C, Marttinen M, Takalo M, Amouyel P, Desai S, Pike I, Hiltunen M, Chécler F, Farinelli M, Delay C, Malmanche N, Hébert SS, Dumont J, Kilinc D, Lambert JC, and Chapuis J
- Subjects
- Amyloid beta-Protein Precursor genetics, Humans, Membrane Proteins, Neoplasm Proteins, Neuronal Plasticity genetics, Neurons, Risk Factors, Alzheimer Disease genetics
- Abstract
Although APP metabolism is being intensively investigated, a large fraction of its modulators is yet to be characterized. In this context, we combined two genome-wide high-content screenings to assess the functional impact of miRNAs and genes on APP metabolism and the signaling pathways involved. This approach highlighted the involvement of FERMT2 (or Kindlin-2), a genetic risk factor of Alzheimer's disease (AD), as a potential key modulator of axon guidance, a neuronal process that depends on the regulation of APP metabolism. We found that FERMT2 directly interacts with APP to modulate its metabolism, and that FERMT2 underexpression impacts axonal growth, synaptic connectivity, and long-term potentiation in an APP-dependent manner. Last, the rs7143400-T allele, which is associated with an increased AD risk and localized within the 3'UTR of FERMT2, induced a downregulation of FERMT2 expression through binding of miR-4504 among others. This miRNA is mainly expressed in neurons and significantly overexpressed in AD brains compared to controls. Altogether, our data provide strong evidence for a detrimental effect of FERMT2 underexpression in neurons and insight into how this may influence AD pathogenesis., (© 2020. The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
33. Identification of a Potential Inhibitor of the FIV p24 Capsid Protein and Characterization of Its Binding Site.
- Author
-
Long M, Cantrelle FX, Robert X, Boll E, Sierra N, Gouet P, Hanoulle X, Alvarez GI, and Guillon C
- Subjects
- Animals, Binding Sites, Capsid metabolism, Capsid Proteins antagonists & inhibitors, Capsid Proteins metabolism, Cats, Gene Products, gag metabolism, Immunodeficiency Virus, Feline metabolism, Lead pharmacology, Protein Domains, Antiviral Agents pharmacology, Benzimidazoles pharmacology, Gene Products, gag antagonists & inhibitors, Immunodeficiency Virus, Feline drug effects
- Abstract
Feline immunodeficiency virus (FIV) is a veterinary infective agent for which there is currently no efficient drug available. Drugs targeting the lentivirus capsid are currently under development for the treatment of human immunodeficiency virus 1 (HIV-1). Here we describe a lead compound that interacts with the FIV capsid. This compound, 696 , modulates the in vitro assembly of and stabilizes the assembled capsid protein. To decipher the mechanism of binding of this compound to the protein, we performed the first nuclear magnetic resonance (NMR) assignment of the FIV p24 capsid protein. Experimental NMR chemical shift perturbations (CSPs) observed after the addition of 696 enabled the characterization of a specific binding site for 696 on p24. This site was further analyzed by molecular modeling of the protein:compound interaction, demonstrating a strong similarity with the binding sites of existing drugs targeting the HIV-1 capsid protein. Taken together, we characterized a promising capsid-interacting compound with a low cost of synthesis, for which derivatives could lead to the development of efficient treatments for FIV infection. More generally, our strategy combining the NMR assignment of FIV p24 with NMR CSPs and molecular modeling will be useful for the analysis of future compounds targeting p24 in the quest to identify an efficient treatment for FIV.
- Published
- 2021
- Full Text
- View/download PDF
34. Phosphorylated full-length Tau interacts with 14-3-3 proteins via two short phosphorylated sequences, each occupying a binding groove of 14-3-3 dimer.
- Author
-
Neves JF, Petrvalská O, Bosica F, Cantrelle FX, Merzougui H, O'Mahony G, Hanoulle X, Obšil T, and Landrieu I
- Subjects
- 14-3-3 Proteins chemistry, 14-3-3 Proteins genetics, Alzheimer Disease genetics, Alzheimer Disease pathology, Binding Sites, Cyclic AMP-Dependent Protein Kinases metabolism, Exoribonucleases chemistry, Exoribonucleases genetics, Exoribonucleases metabolism, Humans, Magnetic Resonance Spectroscopy, Mutation, Phosphorylation, Protein Binding, Protein Conformation, Serine chemistry, Serine metabolism, Surface Plasmon Resonance, tau Proteins chemistry, tau Proteins genetics, 14-3-3 Proteins metabolism, Alzheimer Disease metabolism, Protein Multimerization, tau Proteins metabolism
- Abstract
Protein-protein interactions (PPIs) remain poorly explored targets for the treatment of Alzheimer's disease. The interaction of 14-3-3 proteins with Tau was shown to be linked to Tau pathology. This PPI is therefore seen as a potential target for Alzheimer's disease. When Tau is phosphorylated by PKA (Tau-PKA), several phosphorylation sites are generated, including two known 14-3-3 binding sites, surrounding the phosphorylated serines 214 and 324 of Tau. The crystal structures of 14-3-3 in complex with peptides surrounding these Tau phosphosites show that both these motifs are anchored in the amphipathic binding groove of 14-3-3. However, in the absence of structural data with the full-length Tau protein, the stoichiometry of the complex or the interface and affinity of the partners is still unclear. In this work, we addressed these points, using a broad range of biophysical techniques. The interaction of the long and disordered Tau-PKA protein with 14-3-3σ is restricted to two short sequences, containing phosphorylated serines, which bind in the amphipathic binding groove of 14-3-3σ. Phosphorylation of Tau is fundamental for the formation of this stable complex, and the affinity of the Tau-PKA/14-3-3σ interaction is in the 1-10 micromolar range. Each monomer of the 14-3-3σ dimer binds one of two different phosphorylated peptides of Tau-PKA, suggesting a 14-3-3/Tau-PKA stoichiometry of 2 : 1, confirmed by analytical ultracentrifugation. These results contribute to a better understanding of this PPI and provide useful insights for drug discovery projects aiming at the modulation of this interaction., (© 2020 Federation of European Biochemical Societies.)
- Published
- 2021
- Full Text
- View/download PDF
35. Discovery of the first Mycobacterium tuberculosis MabA (FabG1) inhibitors through a fragment-based screening.
- Author
-
Faïon L, Djaout K, Frita R, Pintiala C, Cantrelle FX, Moune M, Vandeputte A, Bourbiaux K, Piveteau C, Herledan A, Biela A, Leroux F, Kremer L, Blaise M, Tanina A, Wintjens R, Hanoulle X, Déprez B, Willand N, Baulard AR, and Flipo M
- Subjects
- Bacterial Proteins metabolism, Dose-Response Relationship, Drug, Drug Evaluation, Preclinical, Enzyme Inhibitors chemistry, Fatty Acid Synthases metabolism, Molecular Structure, Structure-Activity Relationship, ortho-Aminobenzoates chemistry, Bacterial Proteins antagonists & inhibitors, Drug Discovery, Enzyme Inhibitors pharmacology, Fatty Acid Synthases antagonists & inhibitors, Mycobacterium tuberculosis enzymology, ortho-Aminobenzoates pharmacology
- Abstract
Mycobacterium tuberculosis (M.tb), the etiologic agent of tuberculosis, remains the leading cause of death from a single infectious agent worldwide. The emergence of drug-resistant M.tb strains stresses the need for drugs acting on new targets. Mycolic acids are very long chain fatty acids playing an essential role in the architecture and permeability of the mycobacterial cell wall. Their biosynthesis involves two fatty acid synthase (FAS) systems. Among the four enzymes (MabA, HadAB/BC, InhA and KasA/B) of the FAS-II cycle, MabA (FabG1) remains the only one for which specific inhibitors have not been reported yet. The development of a new LC-MS/MS based enzymatic assay allowed the screening of a 1280 fragment-library and led to the discovery of the first small molecules that inhibit MabA activity. A fragment from the anthranilic acid series was optimized into more potent inhibitors and their binding to MabA was confirmed by
19 F ligand-observed NMR experiments., Competing Interests: Declaration of competing interest 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 © 2020 Elsevier Masson SAS. All rights reserved.)- Published
- 2020
- Full Text
- View/download PDF
36. Single Domain Antibody Fragments as New Tools for the Detection of Neuronal Tau Protein in Cells and in Mice Studies.
- Author
-
Dupré E, Danis C, Arrial A, Hanoulle X, Homa M, Cantrelle FX, Merzougui H, Colin M, Rain JC, Buée L, and Landrieu I
- Subjects
- Alzheimer Disease genetics, Alzheimer Disease metabolism, Alzheimer Disease pathology, Amino Acid Sequence, Animals, Cell Line, Tumor, Immunoglobulin Fragments genetics, Immunoglobulin Fragments metabolism, Mice, Mice, Transgenic, Neurons pathology, Neurons metabolism, Single-Domain Antibodies genetics, Single-Domain Antibodies metabolism, tau Proteins genetics, tau Proteins metabolism
- Abstract
Tau is a neuronal protein linked to pathologies called tauopathies, including Alzheimer's disease. In Alzheimer's disease, tau aggregates into filaments, leading to the observation of intraneuronal fibrillary tangles. Molecular mechanisms resulting in tau aggregation and in tau pathology spreading through the brain regions are still not fully understood. New tools are thus needed to decipher tau pathways involved in the diseases. In this context, a family of novel single domain antibody fragments, or VHHs, directed against tau were generated and characterized. Among the selected VHHs obtained from screening of a synthetic library, a family of six VHHs shared the same CDR3 recognition loop and recognized the same epitope, located in the C-terminal domain of tau. Affinity parameters characterizing the tau/VHHs interaction were next evaluated using surface plasmon resonance spectroscopy. The equilibrium constants K
D were in the micromolar range, but despite conservation of the CDR3 loop sequence, a range of affinities was observed for this VHH family. One of these VHHs, named F8-2, was additionally shown to bind tau upon expression in a neuronal cell line model. Optimization of VHH F8-2 by yeast two-hybrid allowed the generation of an optimized VHH family characterized by lower KD than that of the F8-2 wild-type counterpart, and recognizing the same epitope. The optimized VHHs can also be used as antibodies for detecting tau in transgenic mice brain tissues. These results validate the use of these VHHs for in vitro studies, but also their potential for in-cell expression and assays in mouse models, to explore the mechanisms underlying tau physiopathology.- Published
- 2019
- Full Text
- View/download PDF
37. Cyclophilin A allows the allosteric regulation of a structural motif in the disordered domain 2 of NS5A and thereby fine-tunes HCV RNA replication.
- Author
-
Dujardin M, Madan V, Gandhi NS, Cantrelle FX, Launay H, Huvent I, Bartenschlager R, Lippens G, and Hanoulle X
- Subjects
- Allosteric Regulation, Cyclophilin A genetics, Cyclophilin A isolation & purification, Molecular Dynamics Simulation, Nuclear Magnetic Resonance, Biomolecular, RNA, Viral chemistry, Viral Nonstructural Proteins chemistry, Viral Nonstructural Proteins isolation & purification, Virus Replication, Cyclophilin A metabolism, RNA, Viral metabolism, Viral Nonstructural Proteins metabolism
- Abstract
Implicated in numerous human diseases, intrinsically disordered proteins (IDPs) are dynamic ensembles of interconverting conformers that often contain many proline residues. Whether and how proline conformation regulates the functional aspects of IDPs remains an open question, however. Here, we studied the disordered domain 2 of nonstructural protein 5A (NS5A-D2) of hepatitis C virus (HCV). NS5A-D2 comprises a short structural motif (PW-turn) embedded in a proline-rich sequence, whose interaction with the human prolyl isomerase cyclophilin A (CypA) is essential for viral RNA replication. Using NMR, we show here that the PW-turn motif exists in a conformational equilibrium between folded and disordered states. We found that the fraction of conformers in the NS5A-D2 ensemble that adopt the structured motif is allosterically modulated both by the cis / trans isomerization of the surrounding prolines that are CypA substrates and by substitutions conferring resistance to cyclophilin inhibitor. Moreover, we noted that this fraction is directly correlated with HCV RNA replication efficiency. We conclude that CypA can fine-tune the dynamic ensemble of the disordered NS5A-D2, thereby regulating viral RNA replication efficiency., (© 2019 Dujardin et al.)
- Published
- 2019
- Full Text
- View/download PDF
38. Set-up and screening of a fragment library targeting the 14-3-3 protein interface.
- Author
-
Valenti D, Neves JF, Cantrelle FX, Hristeva S, Lentini Santo D, Obšil T, Hanoulle X, Levy LM, Tzalis D, Landrieu I, and Ottmann C
- Abstract
Protein-protein interactions (PPIs) are at the core of regulation mechanisms in biological systems and consequently became an attractive target for therapeutic intervention. PPIs involving the adapter protein 14-3-3 are representative examples given the broad range of partner proteins forming a complex with one of its seven human isoforms. Given the challenges represented by the nature of these interactions, fragment-based approaches offer a valid alternative for the development of PPI modulators. After having assembled a fragment set tailored on PPIs' modulation, we started a screening campaign on the sigma isoform of 14-3-3 adapter proteins. Through the use of both mono- and bi-dimensional nuclear magnetic resonance spectroscopy measurements, coupled with differential scanning fluorimetry, three fragment hits were identified. These molecules bind the protein at two different regions distant from the usual binding groove highlighting new possibilities for selective modulation of 14-3-3 complexes., (This journal is © The Royal Society of Chemistry 2019.)
- Published
- 2019
- Full Text
- View/download PDF
39. Backbone chemical shift assignments of human 14-3-3σ.
- Author
-
Neves JF, Landrieu I, Merzougui H, Boll E, Hanoulle X, and Cantrelle FX
- Subjects
- Amino Acid Sequence, Humans, Models, Molecular, Protein Structure, Secondary, 14-3-3 Proteins chemistry, Nuclear Magnetic Resonance, Biomolecular
- Abstract
14-3-3 proteins are a group of seven dimeric adapter proteins that exert their biological function by interacting with hundreds of phosphorylated proteins, thus influencing their sub-cellular localization, activity or stability in the cell. Due to this remarkable interaction network, 14-3-3 proteins have been associated with several pathologies and the protein-protein interactions (PPIs) established with a number of partners are now considered promising drug targets. The activity of 14-3-3 proteins is often isoform specific and to our knowledge only one out of seven isoforms, 14-3-3[Formula: see text], has been assigned. Despite the availability of the crystal structures of all seven isoforms of 14-3-3, the additional NMR assignments of 14-3-3 proteins are important for both biological mechanism studies and chemical biology approaches. Herein, we present a robust backbone assignment of 14-3-3σ, which will allow advances in the discovery of potential therapeutic compounds. This assignment is now being applied to the discovery of both inhibitors and stabilizers of 14-3-3 PPIs.
- Published
- 2019
- Full Text
- View/download PDF
40. Nuclear Magnetic Resonance Spectroscopy Insights into Tau Structure in Solution: Impact of Post-translational Modifications.
- Author
-
Danis C, Dupré E, Hanoulle X, Landrieu I, Lasorsa A, Neves JF, Schneider R, and Smet-Nocca C
- Subjects
- Humans, Nuclear Magnetic Resonance, Biomolecular, Phosphorylation, Protein Structure, Secondary, Protein Processing, Post-Translational, tau Proteins chemistry, tau Proteins metabolism
- Abstract
Although Tau is an intrinsically disordered protein, some level of structure can still be defined, corresponding to short stretches of dynamic secondary structures and a preferential global fold described as an ensemble of conformations. These structures can be modified by Tau phosphorylation, and potentially other post-translational modifications. The analytical capacity of Nuclear Magnetic Resonance (NMR) spectroscopy provides the advantage of offering a residue-specific view of these modifications, allowing to link specific sites to a particular structure. The cis or trans conformation of X-Proline peptide bonds is an additional characteristic parameter of Tau structure that is targeted and modified by prolyl cis/trans isomerases. The challenge in molecular characterization of Tau lies in being able to link structural parameters to functional consequences in normal functions and dysfunctions of Tau, including potential misfolding on the path to aggregation and/or perturbation of the interactions of Tau with its many molecular partners. Phosphorylation of Ser and Thr residues has the potential to impact the local and global structure of Tau.
- Published
- 2019
- Full Text
- View/download PDF
41. Functional Study of the C-Terminal Part of the Hepatitis C Virus E1 Ectodomain.
- Author
-
Moustafa RI, Haddad JG, Linna L, Hanoulle X, Descamps V, Mesalam AA, Baumert TF, Duverlie G, Meuleman P, Dubuisson J, and Lavie M
- Subjects
- Cell Line, DNA Mutational Analysis, Hepatocytes virology, Humans, Mutant Proteins genetics, Mutant Proteins metabolism, Viral Envelope Proteins genetics, Hepacivirus physiology, Viral Envelope Proteins metabolism, Virus Assembly, Virus Internalization
- Abstract
In the hepatitis C virus (HCV) envelope glycoproteins E1 and E2, which form a heterodimer, E2 is the receptor binding protein and the major target of neutralizing antibodies, whereas the function of E1 remains less characterized. To investigate E1 functions, we generated a series of mutants in the conserved residues of the C-terminal region of the E1 ectodomain in the context of an infectious clone. We focused our analyses on two regions of interest. The first region is located in the middle of the E1 glycoprotein (between amino acid [aa] 270 and aa 291), which contains a conserved hydrophobic sequence and was proposed to constitute a putative fusion peptide. The second series of mutants was generated in the region from aa 314 to aa 342 (the aa314-342 region), which has been shown to contain two α helices (α2 and α3) by nuclear magnetic resonance studies. Of the 22 generated mutants, 20 were either attenuated or noninfectious. Several mutations modulated the virus's dependence on claudin-1 and the scavenger receptor BI coreceptors for entry. Most of the mutations in the putative fusion peptide region affected virus assembly. Conversely, mutations in the α-helix aa 315 to 324 (315-324) residues M318, W320, D321, and M322 resulted in a complete loss of infectivity without any impact on E1E2 folding and on viral assembly. Further characterization of the W320A mutant in the HCVpp model indicated that the loss of infectivity was due to a defect in viral entry. Together, these results support a role for E1 in modulating HCV interaction with its coreceptors and in HCV assembly. They also highlight the involvement of α-helix 315-324 in a late step of HCV entry. IMPORTANCE HCV is a major public health problem worldwide. The virion harbors two envelope proteins, E1 and E2, which are involved at different steps of the viral life cycle. Whereas E2 has been extensively characterized, the function of E1 remains poorly defined. We characterized here the function of the putative fusion peptide and the region containing α helices of the E1 ectodomain, which had been previously suggested to be important for virus entry. We could confirm the importance of these regions for the virus infectivity. Interestingly, we found several residues modulating the virus's dependence on several HCV receptors, thus highlighting the role of E1 in the interaction of the virus with cellular receptors. Whereas mutations in the putative fusion peptide affected HCV infectivity and morphogenesis, several mutations in the α2-helix region led to a loss of infectivity with no effect on assembly, indicating a role of this region in virus entry., (Copyright © 2018 American Society for Microbiology.)
- Published
- 2018
- Full Text
- View/download PDF
42. X-ray structure of alisporivir in complex with cyclophilin A at 1.5 Å resolution.
- Author
-
Dujardin M, Bouckaert J, Rucktooa P, and Hanoulle X
- Subjects
- Amino Acid Motifs, Binding Sites, Cloning, Molecular, Crystallography, X-Ray, Cyclophilin A genetics, Cyclophilin A metabolism, Cyclosporine metabolism, Escherichia coli genetics, Escherichia coli metabolism, Gene Expression, Genetic Vectors chemistry, Genetic Vectors metabolism, Humans, Kinetics, Models, Molecular, Protein Binding, Protein Conformation, alpha-Helical, Protein Conformation, beta-Strand, Protein Interaction Domains and Motifs, Protein Multimerization, Protein Subunits genetics, Protein Subunits metabolism, Recombinant Proteins chemistry, Recombinant Proteins genetics, Recombinant Proteins metabolism, Cyclophilin A chemistry, Cyclosporine chemistry, Protein Subunits chemistry
- Abstract
Alisporivir (ALV) is an 11-amino-acid hydrophobic cyclic peptide with N-methyl-D-alanine and N-ethyl-L-valine (NEV) residues at positions 3 and 4, respectively. ALV is a non-immunosuppressive cyclosporin A (CsA) derivative. This inhibitor targets cyclophilins (Cyps), a family of proteins with peptidyl-prolyl cis/trans isomerase enzymatic activity. Cyps act as protein chaperones and are involved in numerous cellular functions. Moreover, Cyps have been shown to be an essential cofactor for the replication of many viruses, including Hepatitis C virus and Human immunodeficiency virus, and have also been shown to be involved in mitochondrial diseases. For these reasons, cyclophilins represent an attractive drug target. The structure of ALV in complex with cyclophilin A (CypA), the most abundant Cyp in humans, has been determined at 1.5 Å resolution. This first structure of the CypA-ALV complex shows that the binding of ALV is highly similar to that of CsA. The high resolution allowed the unambiguous determination of the conformations of residues 3 and 4 in ALV when bound to its target. In particular, the side-chain conformation of NEV4 precludes the interaction of the CypA-ALV complex with calcineurin, a cellular protein phosphatase involved in the immune response, which explains the non-immunosuppressive property of ALV. This study provides detailed molecular insights into the CypA-ALV interaction.
- Published
- 2018
- Full Text
- View/download PDF
43. Glycan Shielding and Modulation of Hepatitis C Virus Neutralizing Antibodies.
- Author
-
Lavie M, Hanoulle X, and Dubuisson J
- Subjects
- Animals, Cell Line, Glycosylation, Humans, Immunity, Humoral, Mice, Polysaccharides chemistry, Viral Envelope Proteins immunology, Viral Envelope Proteins metabolism, Virus Assembly, Virus Internalization, Antibodies, Neutralizing immunology, Hepacivirus immunology, Hepatitis C Antibodies immunology, Polysaccharides immunology
- Abstract
Hepatitis C virus (HCV) envelope glycoprotein heterodimer, E1E2, plays an essential role in virus entry and assembly. Furthermore, due to their exposure at the surface of the virion, these proteins are the major targets of anti-HCV neutralizing antibodies. Their ectodomain are heavily glycosylated with up to 5 sites on E1 and up to 11 sites on E2 modified by N-linked glycans. Thus, one-third of the molecular mass of E1E2 heterodimer corresponds to glycans. Despite the high sequence variability of E1 and E2, N-glycosylation sites of these proteins are generally conserved among the seven major HCV genotypes. N-glycans have been shown to be involved in E1E2 folding and modulate different functions of the envelope glycoproteins. Indeed, site-directed mutagenesis studies have shown that specific glycans are needed for virion assembly and infectivity. They can notably affect envelope protein entry functions by modulating their affinity for HCV receptors and their fusion activity. Importantly, glycans have also been shown to play a key role in immune evasion by masking antigenic sites targeted by neutralizing antibodies. It is well known that the high mutational rate of HCV polymerase facilitates the appearance of neutralization resistant mutants, and occurrence of mutations leading to glycan shifting is one of the mechanisms used by this virus to escape host humoral immune response. As a consequence of the importance of the glycan shield for HCV immune evasion, the deletion of N-glycans also leads to an increase in E1E2 immunogenicity and can induce a more potent antibody response against HCV.
- Published
- 2018
- Full Text
- View/download PDF
44. NMR and circular dichroism data for domain 2 of the HCV NS5A protein phosphorylated by the Casein Kinase II.
- Author
-
Bessa LM, Schneider R, and Hanoulle X
- Abstract
The Hepatitis C Virus (HCV) nonstructural 5A protein (NS5A) is a phosphoprotein (Evans et al., 2004; Ross-Thriepland and Harris, 2014) [1], [2] composed of an N-terminal well-structured domain and two C-terminal intrinsically disordered domains (Moradpour et al., 2007; Bartenschlager et al., 2013; Badillo et al., 2017) [3], [4], [5]. So far, no precise molecular function has been identified for this viral protein (Ross-Thriepland and Harris, 2015) [6] which is required for viral replication (Tellinghuisen et al., 2008) [7]. In this article, we present datasets of NMR and circular dichroism analyses of the domain 2 of the HCV NS5A protein (NS5A-D2) phosphorylated in vitro by the Casein Kinase II (CKII) (Dal Pero et al., 2007; Clemens et al., 2015; Masak et al., 2014; Kim et al., 2014) [8], [9], [10], [11]. We describe the in vitro phosphorylation of the serine 288 (pS288) of NS5A-D2 by CKII and report the circular dichroism spectrum of the phosphorylated domain (NS5-D2_CKII). This data article also contains the
1 H,15 N and13 C NMR chemical shift assignments (HN, N, Cα, Cβ and C') for the phosphorylated NS5A-D2 domain, and an assigned1 H,15 N-HSQC spectrum is shown. The NMR data have been acquired on an 800 MHz spectrometer. These NMR data have been used to calculate both the1 H,15 N combined chemical shift perturbations (CSP) induced by the phosphorylation of pS288 and the secondary structural propensity (SSP) scores that describe the structural tendencies in this intrinsically disordered domain. The circular dichroism spectrum and the SSP scores of NS5A-D2_CKII have been compared with those of unphosphorylated NS5A-D2 [12,13].- Published
- 2018
- Full Text
- View/download PDF
45. Interaction study between HCV NS5A-D2 and NS5B using 19 F NMR.
- Author
-
Dujardin M, Cantrelle FX, Lippens G, and Hanoulle X
- Subjects
- Fluorine, Fluorine-19 Magnetic Resonance Imaging methods, Isotope Labeling methods, Hepacivirus chemistry, Nuclear Magnetic Resonance, Biomolecular methods, Viral Nonstructural Proteins chemistry
- Abstract
The non structural protein 5A (NS5A) regulates the replication of the hepatitis C viral RNA through a direct molecular interaction of its domain 2 (NS5A-D2) with the RNA dependent RNA polymerase NS5B. Because of conflicting data in the literature, we study here this molecular interaction using fluorinated versions of the NS5A-D2 protein derived from the JFH1 Hepatitis C Virus strain. Two methods to prepare fluorine-labelled NS5A-D2 involving the biosynthetic incorporation of a
19 F-tryptophan using 5-fluoroindole and the posttranslational introduction of fluorine by chemical conjugation of 2-iodo-N-(trifluoromethyl)acetamide with the NS5A-D2 cysteine side chains are presented. The dissociation constants (KD ) between NS5A-D2 and NS5B obtained with these two methods are in good agreement, and yield values comparable to those derived previously from a surface plasmon resonance study. We compare benefits and limitations of both labeling methods to study the interaction between an intrinsically disordered protein and a large molecular target by19 F NMR.- Published
- 2018
- Full Text
- View/download PDF
46. NMR reveals the intrinsically disordered domain 2 of NS5A protein as an allosteric regulator of the hepatitis C virus RNA polymerase NS5B.
- Author
-
Bessa LM, Launay H, Dujardin M, Cantrelle FX, Lippens G, Landrieu I, Schneider R, and Hanoulle X
- Subjects
- Allosteric Regulation drug effects, Allosteric Site drug effects, Antiviral Agents chemistry, Antiviral Agents metabolism, Antiviral Agents pharmacology, Enzyme Inhibitors chemistry, Enzyme Inhibitors metabolism, Enzyme Inhibitors pharmacology, Gene Deletion, Intrinsically Disordered Proteins chemistry, Intrinsically Disordered Proteins genetics, Intrinsically Disordered Proteins metabolism, Isoleucine chemistry, Mutagenesis, Site-Directed, Nuclear Magnetic Resonance, Biomolecular, Oligoribonucleotides chemistry, Peptide Fragments antagonists & inhibitors, Peptide Fragments chemistry, Peptide Fragments genetics, Peptide Fragments metabolism, Point Mutation, Protein Conformation, Protein Interaction Domains and Motifs, Protein Refolding drug effects, Pyrones chemistry, Pyrones metabolism, Pyrones pharmacology, RNA-Dependent RNA Polymerase antagonists & inhibitors, RNA-Dependent RNA Polymerase chemistry, RNA-Dependent RNA Polymerase genetics, Recombinant Proteins chemistry, Recombinant Proteins metabolism, Solubility, Triazoles chemistry, Triazoles metabolism, Triazoles pharmacology, Viral Nonstructural Proteins antagonists & inhibitors, Viral Nonstructural Proteins chemistry, Viral Nonstructural Proteins genetics, Hepacivirus enzymology, Models, Molecular, Oligoribonucleotides metabolism, RNA-Dependent RNA Polymerase metabolism, Viral Nonstructural Proteins metabolism
- Abstract
Non-structural protein 5B (NS5B) is the RNA-dependent RNA polymerase that catalyzes replication of the hepatitis C virus (HCV) RNA genome and therefore is central for its life cycle. NS5B interacts with the intrinsically disordered domain 2 of NS5A (NS5A-D2), another essential multifunctional HCV protein that is required for RNA replication. As a result, these two proteins represent important targets for anti-HCV chemotherapies. Despite this importance and the existence of NS5B crystal structures, our understanding of the conformational and dynamic behavior of NS5B in solution and its relationship with NS5A-D2 remains incomplete. To address these points, we report the first detailed NMR spectroscopic study of HCV NS5B lacking its membrane anchor (NS5B
Δ21 ). Analysis of constructs with selective isotope labeling of the δ1 methyl groups of isoleucine side chains demonstrates that, in solution, NS5BΔ21 is highly dynamic but predominantly adopts a closed conformation. The addition of NS5A-D2 leads to spectral changes indicative of binding to both allosteric thumb sites I and II of NS5BΔ21 and induces long-range perturbations that affect the RNA-binding properties of the polymerase. We compared these modifications with the short- and long-range effects triggered in NS5BΔ21 upon binding of filibuvir, an allosteric inhibitor. We demonstrate that filibuvir-bound NS5BΔ21 is strongly impaired in the binding of both NS5A-D2 and RNA. NS5A-D2 induces conformational and functional perturbations in NS5B similar to those triggered by filibuvir. Thus, our work highlights NS5A-D2 as an allosteric regulator of the HCV polymerase and provides new insight into the dynamics of NS5B in solution., (© 2017 by The American Society for Biochemistry and Molecular Biology, Inc.)- Published
- 2017
- Full Text
- View/download PDF
47. Solution Structure of the N-Terminal Domain of Mediator Subunit MED26 and Molecular Characterization of Its Interaction with EAF1 and TAF7.
- Author
-
Lens Z, Cantrelle FX, Peruzzini R, Hanoulle X, Dewitte F, Ferreira E, Baert JL, Monté D, Aumercier M, Villeret V, Verger A, and Landrieu I
- Subjects
- Humans, Magnetic Resonance Spectroscopy, Protein Binding, Protein Conformation, Protein Interaction Mapping, Mediator Complex chemistry, Mediator Complex metabolism, TATA-Binding Protein Associated Factors chemistry, TATA-Binding Protein Associated Factors metabolism, Transcription Factor TFIID chemistry, Transcription Factor TFIID metabolism, Transcription Factors metabolism
- Abstract
MED26 is a subunit of Mediator, a large complex central to the regulation of gene transcription by RNA Polymerase II. MED26 plays a role in the switch between the initiation and elongation phases of RNA Polymerase II-mediated transcription process. Regulation of these steps requires successive binding of MED26 N-terminal domain (NTD) to TATA-binding protein-associated factor 7 (TAF7) and Eleven-nineteen lysine-rich in leukemia-Associated Factor 1 (EAF1). In order to investigate the mechanism of regulation by MED26, MED26-NTD structure was solved by NMR, revealing a 4-helix bundle. EAF1 (239-268) and TAF7 (205-235) peptide interactions were both mapped to the same groove formed by H3 and H4 helices of MED26-NTD. Both interactions are characterized by dissociation constants in the 10-μM range. Further experiments revealed a folding-upon-binding mechanism that leads to the formation of EAF1 (N247-S260) and TAF7 (L214-S227) helices. Chemical shift perturbations and nuclear Overhauser enhancement contacts support the involvement of residues I222/F223 in anchoring TAF7 helix to a hydrophobic pocket of MED26-NTD, including residues L48, W80 and I84. In addition, Ala mutations of charged residues located in the C-terminal disordered part of TAF7 and EAF1 peptides affected the binding, with a loss of affinity characterized by a 10-time increase of dissociation constants. A structural model of MED26-NTD/TAF7 complex shows bi-partite components, combining ordered and disordered segments, as well as hydrophobic and electrostatic contributions to the binding. This study provides molecular detail that will help to decipher the mechanistic basis for the initiation to elongation switch-function mediated by MED26-NTD., (Copyright © 2017 Elsevier Ltd. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
48. Overall Structural Model of NS5A Protein from Hepatitis C Virus and Modulation by Mutations Confering Resistance of Virus Replication to Cyclosporin A.
- Author
-
Badillo A, Receveur-Brechot V, Sarrazin S, Cantrelle FX, Delolme F, Fogeron ML, Molle J, Montserret R, Bockmann A, Bartenschlager R, Lohmann V, Lippens G, Ricard-Blum S, Hanoulle X, and Penin F
- Subjects
- Hepacivirus genetics, Hepacivirus growth & development, Mass Spectrometry, Microbial Sensitivity Tests, Models, Molecular, Protein Conformation, Antiviral Agents pharmacology, Cyclosporine pharmacology, Drug Resistance, Viral drug effects, Hepacivirus drug effects, Mutation, Viral Nonstructural Proteins chemistry, Viral Nonstructural Proteins genetics, Virus Replication drug effects
- Abstract
Hepatitis C virus (HCV) nonstructural protein 5A (NS5A) is a RNA-binding phosphoprotein composed of a N-terminal membrane anchor (AH), a structured domain 1 (D1), and two intrinsically disordered domains (D2 and D3). The knowledge of the functional architecture of this multifunctional protein remains limited. We report here that NS5A-D1D2D3 produced in a wheat germ cell-free system is obtained under a highly phosphorylated state. Its NMR analysis revealed that these phosphorylations do not change the disordered nature of D2 and D3 domains but increase the number of conformers due to partial phosphorylations. By combining NMR and small angle X-ray scattering, we performed a comparative structural characterization of unphosphorylated recombinant D2 domains of JFH1 (genotype 2a) and the Con1 (genotype 1b) strains produced in Escherichia coli. These analyses highlighted a higher intrinsic folding of the latter, revealing the variability of intrinsic conformations in HCV genotypes. We also investigated the effect of D2 mutations conferring resistance of HCV replication to cyclophilin A (CypA) inhibitors on the structure of the recombinant D2 Con1 mutants and their binding to CypA. Although resistance mutations D320E and R318W could induce some local and/or global folding perturbation, which could thus affect the kinetics of conformer interconversions, they do not significantly affect the kinetics of CypA/D2 interaction measured by surface plasmon resonance (SPR). The combination of all our data led us to build a model of the overall structure of NS5A, which provides a useful template for further investigations of the structural and functional features of this enigmatic protein.
- Published
- 2017
- Full Text
- View/download PDF
49. Identification of Novel Functions for Hepatitis C Virus Envelope Glycoprotein E1 in Virus Entry and Assembly.
- Author
-
Haddad JG, Rouillé Y, Hanoulle X, Descamps V, Hamze M, Dabboussi F, Baumert TF, Duverlie G, Lavie M, and Dubuisson J
- Subjects
- Cell Line, DNA Mutational Analysis, Hepacivirus genetics, Hepatocytes virology, Humans, Mutation, Missense, Protein Folding, Protein Multimerization, Receptors, Virus metabolism, Viral Envelope Proteins genetics, Hepacivirus physiology, Viral Envelope Proteins metabolism, Virus Assembly, Virus Internalization
- Abstract
Hepatitis C virus (HCV) envelope glycoprotein complex is composed of E1 and E2 subunits. E2 is the receptor-binding protein as well as the major target of neutralizing antibodies, whereas the functions of E1 remain poorly defined. Here, we took advantage of the recently published structure of the N-terminal region of the E1 ectodomain to interrogate the functions of this glycoprotein by mutating residues within this 79-amino-acid region in the context of an infectious clone. The phenotypes of the mutants were characterized to determine the effects of the mutations on virus entry, replication, and assembly. Furthermore, biochemical approaches were also used to characterize the folding and assembly of E1E2 heterodimers. Thirteen out of 19 mutations led to viral attenuation or inactivation. Interestingly, two attenuated mutants, T213A and I262A, were less dependent on claudin-1 for cellular entry in Huh-7 cells. Instead, these viruses relied on claudin-6, indicating a shift in receptor dependence for these two mutants in the target cell line. An unexpected phenotype was also observed for mutant D263A which was no longer infectious but still showed a good level of core protein secretion. Furthermore, genomic RNA was absent from these noninfectious viral particles, indicating that the D263A mutation leads to the assembly and release of viral particles devoid of genomic RNA. Finally, a change in subcellular colocalization between HCV RNA and E1 was observed for the D263A mutant. This unique observation highlights for the first time cross talk between HCV glycoprotein E1 and the genomic RNA during HCV morphogenesis. IMPORTANCE Hepatitis C virus (HCV) infection is a major public health problem worldwide. It encodes two envelope proteins, E1 and E2, which play a major role in the life cycle of this virus. E2 has been extensively characterized, whereas E1 remains poorly understood. Here, we investigated E1 functions by using site-directed mutagenesis in the context of the viral life cycle. Our results identify unique phenotypes. Unexpectedly, two mutants clearly showed a shift in receptor dependence for cell entry, highlighting a role for E1 in modulating HCV particle interaction with a cellular receptor(s). More importantly, another mutant led to the assembly and release of viral particles devoid of genomic RNA. This unique phenotype was further characterized, and we observed a change in subcellular colocalization between HCV RNA and E1. This unique observation highlights for the first time cross talk between a viral envelope protein and genomic RNA during morphogenesis., (Copyright © 2017 American Society for Microbiology.)
- Published
- 2017
- Full Text
- View/download PDF
50. Nuclear Magnetic Resonance Spectroscopy for the Identification of Multiple Phosphorylations of Intrinsically Disordered Proteins.
- Author
-
Danis C, Despres C, Bessa LM, Malki I, Merzougui H, Huvent I, Qi H, Lippens G, Cantrelle FX, Schneider R, Hanoulle X, Smet-Nocca C, and Landrieu I
- Subjects
- Alzheimer Disease, Humans, Recombinant Proteins biosynthesis, Recombinant Proteins chemistry, Intrinsically Disordered Proteins chemistry, Magnetic Resonance Spectroscopy, Phosphorylation, tau Proteins chemistry
- Abstract
Aggregates of the neuronal Tau protein are found inside neurons of Alzheimer's disease patients. Development of the disease is accompanied by increased, abnormal phosphorylation of Tau. In the course of the molecular investigation of Tau functions and dysfunctions in the disease, nuclear magnetic resonance (NMR) spectroscopy is used to identify the multiple phosphorylations of Tau. We present here detailed protocols of recombinant production of Tau in bacteria, with isotopic enrichment for NMR studies. Purification steps that take advantage of Tau's heat stability and high isoelectric point are described. The protocol for in vitro phosphorylation of Tau by recombinant activated ERK2 allows for generating multiple phosphorylations. The protein sample is ready for data acquisition at the issue of these steps. The parameter setup to start recording on the spectrometer is considered next. Finally, the strategy to identify phosphorylation sites of modified Tau, based on NMR data, is explained. The benefit of this methodology compared to other techniques used to identify phosphorylation sites, such as immuno-detection or mass spectrometry (MS), is discussed.
- Published
- 2016
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.