Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Bogdan Tarus is active.

Publication


Featured researches published by Bogdan Tarus.


Antimicrobial Agents and Chemotherapy | 2013

Structure-Based Discovery of the Novel Antiviral Properties of Naproxen against the Nucleoprotein of Influenza A Virus

Nathalie Lejal; Bogdan Tarus; Edwige Bouguyon; Sylvie Chenavas; Nicolas Bertho; Bernard Delmas; Rob W.H. Ruigrok; Carmelo Di Primo; Anny Slama-Schwok

ABSTRACT The nucleoprotein (NP) binds the viral RNA genome and associates with the polymerase in a ribonucleoprotein complex (RNP) required for transcription and replication of influenza A virus. NP has no cellular counterpart, and the NP sequence is highly conserved, which led to considering NP a hot target in the search for antivirals. We report here that monomeric nucleoprotein can be inhibited by a small molecule binding in its RNA binding groove, resulting in a novel antiviral against influenza A virus. We identified naproxen, an anti-inflammatory drug that targeted the nucleoprotein to inhibit NP-RNA association required for NP function, by virtual screening. Further docking and molecular dynamics (MD) simulations identified in the RNA groove two NP-naproxen complexes of similar levels of interaction energy. The predicted naproxen binding sites were tested using the Y148A, R152A, R355A, and R361A proteins carrying single-point mutations. Surface plasmon resonance, fluorescence, and other in vitro experiments supported the notion that naproxen binds at a site identified by MD simulations and showed that naproxen competed with RNA binding to wild-type (WT) NP and protected active monomers of the nucleoprotein against proteolytic cleavage. Naproxen protected Madin-Darby canine kidney (MDCK) cells against viral challenges with the H1N1 and H3N2 viral strains and was much more effective than other cyclooxygenase inhibitors in decreasing viral titers of MDCK cells. In a mouse model of intranasal infection, naproxen treatment decreased the viral titers in mice lungs. In conclusion, naproxen is a promising lead compound for novel antivirals against influenza A virus that targets the nucleoprotein in its RNA binding groove.


Biochimie | 2012

Oligomerization paths of the nucleoprotein of influenza A virus

Bogdan Tarus; O. Bakowiez; Sylvie Chenavas; L. Duchemin; L.F. Estrozi; C. Bourdieu; Nathalie Lejal; J. Bernard; M. Moudjou; Christophe Chevalier; Bernard Delmas; Rob W.H. Ruigrok; C. Di Primo; Anny Slama-Schwok

The influenza viruses contain a segmented, negative strand RNA genome. Each RNA segment is covered by multiple copies of the nucleoprotein (NP) and is associated with the polymerase complex into ribonucleoprotein (RNP) particles. Despite its importance in the virus life cycle, the interactions between the NP and the genome are not well understood. Here, we studied the assembly process of NP-RNA oligomers and analyzed how the oligomeric/monomeric status of RNA-free NP affects RNA binding and oligomerization. Recombinant wild-type NP purified in low salt concentrations and a derived mutant engineered for oligomerization deficiency (R416A) were mainly monomeric in RNA-free solutions as shown by biochemical and electron microscopy techniques. NP monomer formed with RNA a fast 1/1 complex characterized by surface plasmon resonance. In a subsequent and slow process that depended on the RNA length, oligomerization of NP was mediated by RNA binding. In contrast, preparations of wild-type NP purified in high salt concentrations as well as mutant Y148A engineered for deficiency in nucleic acid binding were partly or totally oligomeric in RNA-free solutions. These trimer/tetramer NP oligomers bind directly as oligomers to RNA with a higher affinity than that of the monomers. Both oligomerization routes we characterized could be exploited by cellular or viral factors to modulate or control viral RNA encapsidation by NP.


PLOS ONE | 2012

Molecular Dynamics Studies of the Nucleoprotein of Influenza A Virus: Role of the Protein Flexibility in RNA Binding

Bogdan Tarus; Christophe Chevalier; Charles-Adrien Richard; Bernard Delmas; Carmelo Di Primo; Anny Slama-Schwok

The influenza viruses contain a segmented, negative stranded RNA genome. Each RNA segment is covered by multiple copies of the nucleoprotein (NP). X-ray structures have shown that NP contains well-structured domains juxtaposed with regions of missing electron densities corresponding to loops. In this study, we tested if these flexible loops gated or promoted RNA binding and RNA-induced oligomerization of NP. We first performed molecular dynamics simulations of wt NP monomer and trimer in comparison with the R361A protein mutated in the RNA binding groove, using the H1N1 NP as the initial structure. Calculation of the root-mean-square fluctuations highlighted the presence of two flexible loops in NP trimer: loop 1 (73–90), loop 2 (200–214). In NP, loops 1 and 2 formed a 10–15 Å-wide pinch giving access to the RNA binding groove. Loop 1 was stabilized by interactions with K113 of the adjacent β-sheet 1 (91–112) that interacted with the RNA grove (linker 360–373) via multiple hydrophobic contacts. In R361A, a salt bridge formed between E80 of loop 1 and R208 of loop 2 driven by hydrophobic contacts between L79 and W207, due to a decreased flexibility of loop 2 and loop 1 unfolding. Thus, RNA could not access its binding groove in R361A; accordingly, R361A had a much lower affinity for RNA than NP. Disruption of the E80-R208 interaction in the triple mutant R361A-E80A-E81A increased its RNA binding affinity and restored its oligomerization back to wt levels in contrast with impaired levels of R361A. Our data suggest that the flexibility of loops 1 and 2 is required for RNA sampling and binding which likely involve conformational change(s) of the nucleoprotein.


Journal of Virology | 2012

Characterization of a Viral Phosphoprotein Binding Site on the Surface of the Respiratory Syncytial Nucleoprotein

Marie Galloux; Bogdan Tarus; Ilfad Blazevic; Jenna Fix; Stéphane Duquerroy; Jean-François Eléouët

ABSTRACT The human respiratory syncytial virus (HRSV) genome is composed of a negative-sense single-stranded RNA that is tightly associated with the nucleoprotein (N). This ribonucleoprotein (RNP) complex is the template for replication and transcription by the viral RNA-dependent RNA polymerase. RNP recognition by the viral polymerase involves a specific interaction between the C-terminal domain of the phosphoprotein (P) (PCTD) and N. However, the P binding region on N remains to be identified. In this study, glutathione S-transferase (GST) pulldown assays were used to identify the N-terminal core domain of HRSV N (NNTD) as a P binding domain. A biochemical characterization of the PCTD and molecular modeling of the NNTD allowed us to define four potential candidate pockets on N (pocket I [PI] to PIV) as hydrophobic sites surrounded by positively charged regions, which could constitute sites complementary to the PCTD interaction domain. The role of selected amino acids in the recognition of the N-RNA complex by P was first screened for by site-directed mutagenesis using a polymerase activity assay, based on an HRSV minigenome containing a luciferase reporter gene. When changed to Ala, most of the residues of PI were found to be critical for viral RNA synthesis, with the R132A mutant having the strongest effect. These mutations also reduced or abolished in vitro and in vivo P-N interactions, as determined by GST pulldown and immunoprecipitation experiments. The pocket formed by these residues is critical for P binding to the N-RNA complex, is specific for pneumovirus N proteins, and is clearly distinct from the P binding sites identified so far for other nonsegmented negative-strand viruses.


ACS Chemical Neuroscience | 2017

Why Is Research on Amyloid-β Failing to Give New Drugs for Alzheimer’s Disease?

Andrew J. Doig; Maria P. del Castillo-Frias; Olivia Berthoumieu; Bogdan Tarus; Jessica Nasica-Labouze; Fabio Sterpone; Phuong H. Nguyen; Nigel M. Hooper; Peter Faller; Philippe Derreumaux

The two hallmarks of Alzheimers disease (AD) are the presence of neurofibrillary tangles (NFT) made of aggregates of the hyperphosphorylated tau protein and of amyloid plaques composed of amyloid-β (Aβ) peptides, primarily Aβ1-40 and Aβ1-42. Targeting the production, aggregation, and toxicity of Aβ with small molecule drugs or antibodies is an active area of AD research due to the general acceptance of the amyloid cascade hypothesis, but thus far all drugs targeting Aβ have failed. From a review of the recent literature and our own experience based on in vitro, in silico, and in vivo studies, we present some reasons to explain this repetitive failure.


Journal of Virology | 2015

A Druggable Pocket at the Nucleocapsid/Phosphoprotein Interaction Site of Human Respiratory Syncytial Virus.

Mohamed Ouizougun-Oubari; Nelson Pereira; Bogdan Tarus; Marie Galloux; Safa Lassoued; Jenna Fix; M. Alejandra Tortorici; Sylviane Hoos; Bruno Baron; Patrick England; Didier Desmaële; Patrick Couvreur; François Bontems; Félix A. Rey; Jean-François Eléouët; Christina Sizun; Anny Slama-Schwok; Stéphane Duquerroy

ABSTRACT Presently, respiratory syncytial virus (RSV), the main cause of severe respiratory infections in infants, cannot be treated efficiently with antivirals. However, its RNA-dependent polymerase complex offers potential targets for RSV-specific drugs. This includes the recognition of its template, the ribonucleoprotein complex (RNP), consisting of genomic RNA encapsidated by the RSV nucleoprotein, N. This recognition proceeds via interaction between the phosphoprotein P, which is the main polymerase cofactor, and N. The determinant role of the C terminus of P, and more particularly of the last residue, F241, in RNP binding and viral RNA synthesis has been assessed previously. Here, we provide detailed structural insight into this crucial interaction for RSV polymerase activity. We solved the crystallographic structures of complexes between the N-terminal domain of N (N-NTD) and C-terminal peptides of P and characterized binding by biophysical approaches. Our results provide a rationale for the pivotal role of F241, which inserts into a well-defined N-NTD pocket. This primary binding site is completed by transient contacts with upstream P residues outside the pocket. Based on the structural information of the N-NTD:P complex, we identified inhibitors of this interaction, selected by in silico screening of small compounds, that efficiently bind to N and compete with P in vitro. One of the compounds displayed inhibitory activity on RSV replication, thereby strengthening the relevance of N-NTD for structure-based design of RSV-specific antivirals. IMPORTANCE Respiratory syncytial virus (RSV) is a widespread pathogen that is a leading cause of acute lower respiratory infections in infants worldwide. RSV cannot be treated efficiently with antivirals, and no vaccine is presently available, with the development of pediatric vaccines being particularly challenging. Therefore, there is a need for new therapeutic strategies that specifically target RSV. The interaction between the RSV phosphoprotein P and the ribonucleoprotein complex is critical for viral replication. In this study, we identified the main structural determinants of this interaction, and we used them to screen potential inhibitors in silico. We found a family of molecules that were efficient competitors of P in vitro and showed inhibitory activity on RSV replication in cellular assays. These compounds provide a basis for a pharmacophore model that must be improved but that holds promises for the design of new RSV-specific antivirals.


Expert Opinion on Drug Discovery | 2015

Learning from structure-based drug design and new antivirals targeting the ribonucleoprotein complex for the treatment of influenza

Alexandre Monod; Christopher Swale; Bogdan Tarus; Alice Tissot; Bernard Delmas; Rob Wh Ruigrok; Thibaut Crépin; Anny Slama-Schwok

Introduction: Influenza viruses are a threat to human health. There are presently only two methods for treating influenza: vaccines, which require yearly updates, and two classes of antivirals that suffer with the problem of resistance by current human influenza viruses; this is especially the case with amantadine and rimantadine. Consequently, there is an urgent need for the development of new antivirals with new mechanisms of action. Areas covered: In this review, the authors focus on viral protein domains, their associated activity and their inhibition by small molecules defined by a structure-based design with a special emphasis on the ribonucleoprotein complex and its inhibitors. Several new classes of antiviral candidates targeting viral replication through individual domains of the polymerase and the nucleoprotein (NP) have been developed through structure-based design. Expert opinion: To date, the antivirals targeting neuraminidase are by far the most developed and potent. Antiviral candidates targeting the NP and polymerase domains are in the pipeline but their pharmacokinetics needs further studies. The recently published structures of the polymerase expand the possibilities for development of new antivirals. Combination therapies targeting conserved viral targets and new cellular proteins or exploiting drug promiscuity hold promises to fight against the emergence of resistance.


Proceedings of the National Academy of Sciences of the United States of America | 2012

Rational design of a fluorescent NADPH derivative imaging constitutive nitric-oxide synthases upon two-photon excitation.

Yun Li; Huan Wang; Bogdan Tarus; Miguel Romero Perez; Laurence Morellato; Etienne Henry; Vladimir Berka; Ah Lim Tsai; Booma Ramassamy; Hamid Dhimane; Chantal Dessy; Patrick Tauc; Jean Luc Boucher; Eric Deprez; Anny Slama-Schwok

We report the structure-based design and synthesis of a unique NOS inhibitor, called nanoshutter NS1, with two-photon absorption properties. NS1 targets the NADPH site of NOS by a nucleotide moiety mimicking NADPH linked to a conjugated push–pull chromophore with nonlinear absorption properties. Because NS1 could not provide reducing equivalents to the protein and competed with NADPH binding, it efficiently inhibited NOS catalysis. NS1 became fluorescent once bound to NOS with an excellent signal-to-noise ratio because of two-photon excitation avoiding interference from the flavin–autofluorescence and because free NS1 was not fluorescent in aqueous solutions. NS1 fluorescence enhancement was selective for constitutive NOS in vitro, in particular for endothelial NOS (eNOS). Molecular dynamics simulations suggested that two variable residues among NOS isoforms induced differences in binding of NS1 and in local solvation around NS1 nitro group, consistent with changes of NS1 fluorescence yield. NS1 colocalized with eNOS in living human umbilical vein endothelial cells. Thus, NS1 constitutes a unique class of eNOS probe with two-photon excitation in the 800–950-nm range, with great perspectives for eNOS imaging in living tissues.


Virus Research | 2011

Picobirnaviruses encode a protein with repeats of the ExxRxNxxxE motif

Bruno Da Costa; Stéphane Duquerroy; Bogdan Tarus; Bernard Delmas

Picobirnaviruses possess a bisegmented double-stranded RNA genome. While the segment 2 encodes the RNA-dependent RNA polymerase, the segment 1 displays two open reading frames (ORFs). ORF2 was recently shown to code the capsid precursor and ORF1 product has not been characterized. In this study, we show that the three ORF1 sequences available in databases and representing three phylogenetically distant picobirnaviruses (two from human and one from rabbit hosts) encode proteins of various sizes (106-224 residues and without proline and cysteine) harbouring a particular sequence motif (ExxRxNxxxE) repeated four to ten times, depending on the virus species. Several algorithms predicted the three proteins to be mainly unfolded in the domains containing the repeats. The glycine-rich 25-40 amino acid long C-terminal domains containing hydrophobic residues with a periodicity of 3-4 residues are predicted structurally different of the upstream domains containing the motif repetitions. The ExxRxNxxxE sequence was not previously identified as a short linear motif in eukaryotic and prokaryotic proteins. Its function remains elusive.


Journal of Molecular Recognition | 2009

Selective probing of a NADPH site controlled light-induced enzymatic catalysis.

Jean-Christophe Lambry; Edward Beaumont; Bogdan Tarus; Mireille Blanchard-Desce; Anny Slama-Schwok

Achieving molecular recognition of NADPH binding sites is a compelling strategy to control many redox biological processes. The NADPH sites recognize the ubiquitous NADPH cofactor via highly conserved binding interactions, despite differences in the regulation of the hydride transfer in redox active proteins. We recently developed a photoactive NADPH substitute, called nanotrigger NT synchronizing the initiation of enzymatic catalysis of the endothelial NO‐synthase (eNOS) with a laser pulse. Spatial and temporal control of enzymatic activity by such a designed light‐driven activator would benefit from achieving molecular selectivity, i.e. activation of a single NADPH‐mediated enzyme.

Collaboration


Dive into the Bogdan Tarus's collaboration.

Top Co-Authors

Avatar

Anny Slama-Schwok

Institut national de la recherche agronomique

View shared research outputs
Top Co-Authors

Avatar

Bernard Delmas

Institut national de la recherche agronomique

View shared research outputs
Top Co-Authors

Avatar

Nathalie Lejal

Institut national de la recherche agronomique

View shared research outputs
Top Co-Authors

Avatar

Etienne Henry

École normale supérieure de Cachan

View shared research outputs
Top Co-Authors

Avatar

Yun Li

Paris Descartes University

View shared research outputs
Top Co-Authors

Avatar

Stéphane Quideau

Centre national de la recherche scientifique

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Booma Ramassamy

Paris Descartes University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Christophe Chevalier

Institut national de la recherche agronomique

View shared research outputs
Researchain Logo
Decentralizing Knowledge