Network


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

Hotspot


Dive into the research topics where Frédéric Iseni is active.

Publication


Featured researches published by Frédéric Iseni.


The EMBO Journal | 2002

Sendai virus trailer RNA binds TIAR, a cellular protein involved in virus‐induced apoptosis

Frédéric Iseni; Dominique Garcin; Machiko Nishio; Nancy Kedersha; Paul Anderson; Daniel Kolakofsky

Sendai virus (SeV) leader (le) and trailer (tr) RNAs are short transcripts generated during abortive antigenome and genome synthesis, respectively. Recom binant SeV (rSeV) that express tr‐like RNAs from the leader region are non‐cytopathic and, moreover, prevent wild‐type SeV from inducing apoptosis in mixed infections. These rSeV thus appear to have gained a function. Here we report that tr RNA binds to a cellular protein with many links to apoptosis (TIAR) via the AU‐rich sequence 5′ UUUUAAAUUUU. Duplication of this AU‐rich sequence alone within the le RNA confers TIAR binding on this le* RNA and a non‐cytopathic phenotype to these rSeV in cell culture. Transgenic overexpression of TIAR during SeV infection promotes apoptosis and reverses the anti‐apoptotic effects of le* RNA expression. More over, TIAR overexpression and SeV infection act synergistically to induce apoptosis. These short viral RNAs may act by sequestering TIAR, a multivalent RNA recognition motif (RRM) family RNA‐binding protein involved in SeV‐induced apoptosis. In this view, tr RNA is not simply a by‐product of abortive genome synthesis, but is also an antigenome transcript that modulates the cellular antiviral response.


Journal of Virology | 2001

Structure of Recombinant Rabies Virus Nucleoprotein-RNA Complex and Identification of the Phosphoprotein Binding site

Guy Schoehn; Frédéric Iseni; Manos Mavrakis; Danielle Blondel; Rob W. H. Ruigrok

ABSTRACT Rabies virus nucleoprotein (N) was produced in insect cells, in which it forms nucleoprotein-RNA (N-RNA) complexes that are biochemically and biophysically indistinguishable from rabies virus N-RNA. We selected recombinant N-RNA complexes that were bound to short insect cellular RNAs which formed small rings containing 9 to 11 N monomers. We also produced recombinant N-RNA rings and viral N-RNA that were treated with trypsin and that had lost the C-terminal quarter of the nucleoprotein. Trypsin-treated N-RNA no longer bound to recombinant rabies virus phosphoprotein (the viral polymerase cofactor), so the presence of the C-terminal part of N is needed for binding of the phosphoprotein. Both intact and trypsin-treated recombinant N-RNA rings were analyzed with cryoelectron microscopy, and three-dimensional models were calculated from single-particle image analysis combined with back projection. Nucleoprotein has a bilobed shape, and each monomer has two sites of interaction with each neighbor. Trypsin treatment cuts off part of one of the lobes without shortening the protein or changing other structural parameters. Using negative-stain electron microscopy, we visualized phosphoprotein bound to the tips of the N-RNA rings, most likely at the site that can be removed by trypsin. Based on the shape of N determined here and on structural parameters derived from electron microscopy on free rabies virus N-RNA and from nucleocapsid in virus, we propose a low-resolution model for rabies virus N-RNA in the virus.


RNA | 2000

Structure of the RNA inside the vesicular stomatitis virus nucleocapsid

Frédéric Iseni; Florence Baudin; Danielle Blondel; Rob W. H. Ruigrok

The structure of the viral RNA (vRNA) inside intact nucleocapsids of vesicular stomatitis virus was studied by chemical probing experiments. Most of the Watson-Crick positions of the nucleotide bases of vRNA in intact virus and in nucleoprotein (N)-RNA template were accessible to the chemical probes and the phosphates were protected. This suggests that the nucleoprotein binds to the sugar-phosphate backbone of the RNA and leaves the Watson-Crick positions free for the transcription and replication activities of the viral RNA-dependent RNA polymerase. The same architecture has been proposed for the influenza virus nucleocapsids. However, about 5% of the nucleotide bases were found to be relatively nonreactive towards the chemical probes and some bases were hyperreactive. The pattern of reactivities was the same for RNA inside virus and for RNA in N-RNA template that was purified over a CsCl gradient and which had more than 94% of the polymerase and phosphoprotein molecules removed. All reactivities were more or less equal on naked vRNA. This suggests that the variations in reactivity towards the chemical probes are caused by the presence of the nucleoprotein.


Journal of Virology | 2007

Suppression of the Sendai virus M protein through a novel short interfering RNA approach inhibits viral particle production but does not affect viral RNA synthesis

Geneviève Mottet-Osman; Frédéric Iseni; Thierry Pelet; Maciej Wiznerowicz; Dominique Garcin; Laurent Roux

ABSTRACT Short RNA interference is more and more widely recognized as an effective method to specifically suppress viral functions in eukaryotic cells. Here, we used an experimental system that allows suppression of the Sendai virus (SeV) M protein by using a target sequence, derived from the green fluorescent protein gene, that was introduced in the 3′ untranslated region of the M protein mRNA. Silencing of the M protein gene was eventually achieved by a small interfering RNA (siRNA) directed against this target sequence. This siRNA was constitutively expressed in a cell line constructed by transduction with an appropriate lentivirus vector. Suppression of the M protein was sufficient to diminish virus production by 50- to 100-fold. This level of suppression had no apparent effect on viral replication and transcription, supporting the lack of M involvement in SeV transcription or replication control.


Journal of Virology | 2004

A short peptide at the amino terminus of the Sendai virus C protein acts as an independent element that induces STAT1 instability.

Dominique Garcin; Jean-Baptiste Marq; Frédéric Iseni; Stephen R. Martin; Daniel Kolakofsky

ABSTRACT The Sendai virus C protein acts to dismantle the interferon-induced cellular antiviral state in an MG132-sensitive manner, in part by inducing STAT1 instability. This activity of C maps to the first 23 amino acids (C1-23) of the 204-amino-acid (aa)-long protein (C1-204). C1-23 was found to act as an independent viral element that induces STAT1 instability, since this peptide fused to green fluorescent protein (C1-23/GFP) is at least as active as C1-204 in this respect. This peptide also induces the degradation of C1-23/GFP and other proteins to which it is fused. Most of C1-204, and particularly its amino-terminal half, is predicted to be structurally disordered. C1-23 as a peptide was found to be disordered by circular dichroism, and the first 11 aa have a strong potential to form an amphipathic α-helix in low concentrations of trifluoroethanol, which is thought to mimic protein-protein interaction. The critical degradation-determining sequence of C1-23 was mapped by mutation to eight residues near its N terminus: 4FLKKILKL11. All the large hydrophobic residues of 4FLKKILKL11, plus its ability to form an amphipathic α-helix, were found to be critical for STAT1 degradation. In contrast, C1-23/GFP self-degradation did not require 8ILKL11, nor the ability to form an α-helix throughout this region. Remarkably, C1-23/GFP also stimulated C1-204 degradation, and this degradation in trans required the same peptide determinants as for STAT1. Our results suggest that C1-204 coordinates its dual activities of regulating viral RNA synthesis and counteracting the host innate antiviral response by sensing both its own intracellular concentration and that of STAT1.


Journal of Virology | 2013

Low-resolution structure of vaccinia virus DNA replication machinery.

Céleste Sèle; Frank Gabel; Irina Gutsche; Ivan Ivanov; Wim P. Burmeister; Frédéric Iseni; Nicolas Tarbouriech

ABSTRACT Smallpox caused by the poxvirus variola virus is a highly lethal disease that marked human history and was eradicated in 1979 thanks to a worldwide mass vaccination campaign. This virus remains a significant threat for public health due to its potential use as a bioterrorism agent and requires further development of antiviral drugs. The viral genome replication machinery appears to be an ideal target, although very little is known about its structure. Vaccinia virus is the prototypic virus of the Orthopoxvirus genus and shares more than 97% amino acid sequence identity with variola virus. Here we studied four essential viral proteins of the replication machinery: the DNA polymerase E9, the processivity factor A20, the uracil-DNA glycosylase D4, and the helicase-primase D5. We present the recombinant expression and biochemical and biophysical characterizations of these proteins and the complexes they form. We show that the A20D4 polymerase cofactor binds to E9 with high affinity, leading to the formation of the A20D4E9 holoenzyme. Small-angle X-ray scattering yielded envelopes for E9, A20D4, and A20D4E9. They showed the elongated shape of the A20D4 cofactor, leading to a 150-Å separation between the polymerase active site of E9 and the DNA-binding site of D4. Electron microscopy showed a 6-fold rotational symmetry of the helicase-primase D5, as observed for other SF3 helicases. These results favor a rolling-circle mechanism of vaccinia virus genome replication similar to the one suggested for tailed bacteriophages.


Journal of Biological Chemistry | 2015

Crystal Structure of the Vaccinia Virus Uracil-DNA Glycosylase in Complex with DNA

Wim P. Burmeister; Nicolas Tarbouriech; Pascal Fender; Céline Contesto-Richefeu; Christophe N. Peyrefitte; Frédéric Iseni

Background: D4 is a uracil-DNA glycosylase (UNG) and an essential component of the vaccinia virus DNA polymerase holoenzyme. Results: The crystal structure of D4 in complex with DNA is presented. Conclusion: The D4·DNA contacts exhibit major differences compared with the human UNG·DNA complex. Significance: This work allows a better understanding of the structural determinants required for UNG function. Vaccinia virus polymerase holoenzyme is composed of the DNA polymerase catalytic subunit E9 associated with its heterodimeric co-factor A20·D4 required for processive genome synthesis. Although A20 has no known enzymatic activity, D4 is an active uracil-DNA glycosylase (UNG). The presence of a repair enzyme as a component of the viral replication machinery suggests that, for poxviruses, DNA synthesis and base excision repair is coupled. We present the 2.7 Å crystal structure of the complex formed by D4 and the first 50 amino acids of A20 (D4·A201–50) bound to a 10-mer DNA duplex containing an abasic site resulting from the cleavage of a uracil base. Comparison of the viral complex with its human counterpart revealed major divergences in the contacts between protein and DNA and in the enzyme orientation on the DNA. However, the conformation of the dsDNA within both structures is very similar, suggesting a dominant role of the DNA conformation for UNG function. In contrast to human UNG, D4 appears rigid, and we do not observe a conformational change upon DNA binding. We also studied the interaction of D4·A201–50 with different DNA oligomers by surface plasmon resonance. D4 binds weakly to nonspecific DNA and to uracil-containing substrates but binds abasic sites with a Kd of <1.4 μm. This second DNA complex structure of a family I UNG gives new insight into the role of D4 as a co-factor of vaccinia virus DNA polymerase and allows a better understanding of the structural determinants required for UNG action.


Journal of Virology | 2016

Domain organization of vaccinia virus helicase-primase D5.

Stephanie Hutin; Wai Li Ling; Adam Round; Grégory Effantin; Stefan Reich; Frédéric Iseni; Nicolas Tarbouriech; Guy Schoehn; Wim P. Burmeister

ABSTRACT Poxviridae are viruses with a large linear double-stranded DNA genome coding for up to 250 open reading frames and a fully cytoplasmic replication. The double-stranded DNA genome is covalently circularized at both ends. Similar structures of covalently linked extremities of the linear DNA genome are found in the African swine fever virus (asfarvirus) and in the Phycodnaviridae. We are studying the machinery which replicates this peculiar genome structure. From our work with vaccinia virus, we give first insights into the overall structure and function of the essential poxvirus virus helicase-primase D5 and show that the active helicase domain of D5 builds a hexameric ring structure. This hexamer has ATPase and, more generally, nucleoside triphosphatase activities that are indistinguishable from the activities of full-length D5 and that are independent of the nature of the base. In addition, hexameric helicase domains bind tightly to single- and double-stranded DNA. Still, the monomeric D5 helicase construct truncated within the D5N domain leads to a well-defined structure, but it does not have ATPase or DNA-binding activity. This shows that the full D5N domain has to be present for hexamerization. This allowed us to assign a function to the D5N domain which is present not only in D5 but also in other viruses of the nucleocytoplasmic large DNA virus (NCLDV) clade. The primase domain and the helicase domain were structurally analyzed via a combination of small-angle X-ray scattering and, when appropriate, electron microscopy, leading to consistent low-resolution models of the different proteins. IMPORTANCE Since the beginning of the 1980s, research on the vaccinia virus replication mechanism has basically stalled due to the absence of structural information. As a result, this important class of pathogens is less well understood than most other viruses. This lack of information concerns in general viruses of the NCLDV clade, which use a superfamily 3 helicase for replication, as do poxviruses. Here we provide for the first time information about the domain structure and DNA-binding activity of D5, the poxvirus helicase-primase. This result not only refines the current model of the poxvirus replication fork but also will lead in the long run to a structural basis for antiviral drug design.


Acta Crystallographica Section F-structural Biology and Crystallization Communications | 2016

Structural analysis of point mutations at the Vaccinia virus A20/D4 interface.

Céline Contesto-Richefeu; Nicolas Tarbouriech; Xavier Brazzolotto; Wim P. Burmeister; Christophe N. Peyrefitte; Frédéric Iseni

The Vaccinia virus polymerase holoenzyme is composed of three subunits: E9, the catalytic DNA polymerase subunit; D4, a uracil-DNA glycosylase; and A20, a protein with no known enzymatic activity. The D4/A20 heterodimer is the DNA polymerase cofactor, the function of which is essential for processive DNA synthesis. The recent crystal structure of D4 bound to the first 50 amino acids of A20 (D4/A201-50) revealed the importance of three residues, forming a cation-π interaction at the dimerization interface, for complex formation. These are Arg167 and Pro173 of D4 and Trp43 of A20. Here, the crystal structures of the three mutants D4-R167A/A201-50, D4-P173G/A201-50 and D4/A201-50-W43A are presented. The D4/A20 interface of the three structures has been analysed for atomic solvation parameters and cation-π interactions. This study confirms previous biochemical data and also points out the importance for stability of the restrained conformational space of Pro173. Moreover, these new structures will be useful for the design and rational improvement of known molecules targeting the D4/A20 interface.


Nature Communications | 2017

The vaccinia virus DNA polymerase structure provides insights into the mode of processivity factor binding

Nicolas Tarbouriech; Corinne Ducournau; Stephanie Hutin; Philippe J. Mas; Petr Man; Eric Forest; Darren J. Hart; Christophe N. Peyrefitte; Wim P. Burmeister; Frédéric Iseni

Vaccinia virus (VACV), the prototype member of the Poxviridae, replicates in the cytoplasm of an infected cell. The catalytic subunit of the DNA polymerase E9 binds the heterodimeric processivity factor A20/D4 to form the functional polymerase holoenzyme. Here we present the crystal structure of full-length E9 at 2.7 Å resolution that permits identification of important poxvirus-specific structural insertions. One insertion in the palm domain interacts with C-terminal residues of A20 and thus serves as the processivity factor-binding site. This is in strong contrast to all other family B polymerases that bind their co-factors at the C terminus of the thumb domain. The VACV E9 structure also permits rationalization of polymerase inhibitor resistance mutations when compared with the closely related eukaryotic polymerase delta–DNA complex.The catalytic subunit E9 of the vaccinia virus DNA polymerase forms a functional polymerase holoenzyme by interacting with the heterodimeric processivity factor A20/D4. Here the authors present the structure of full-length E9 and show that an insertion within its palm domain binds A20, in a mode different from other family B polymerases.

Collaboration


Dive into the Frédéric Iseni's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Nicolas Tarbouriech

Centre national de la recherche scientifique

View shared research outputs
Top Co-Authors

Avatar

Wim P. Burmeister

Centre national de la recherche scientifique

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Rob W. H. Ruigrok

Centre national de la recherche scientifique

View shared research outputs
Top Co-Authors

Avatar

Guy Schoehn

Centre national de la recherche scientifique

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Corinne Ducournau

Centre national de la recherche scientifique

View shared research outputs
Top Co-Authors

Avatar

Danielle Blondel

Centre national de la recherche scientifique

View shared research outputs
Researchain Logo
Decentralizing Knowledge