Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
PLoS Pathog ; 20(5): e1011652, 2024 May.
Article in English | MEDLINE | ID: mdl-38768256

ABSTRACT

The year 2022 was marked by the mpox outbreak caused by the human monkeypox virus (MPXV), which is approximately 98% identical to the vaccinia virus (VACV) at the sequence level with regard to the proteins involved in DNA replication. We present the production in the baculovirus-insect cell system of the VACV DNA polymerase holoenzyme, which consists of the E9 polymerase in combination with its co-factor, the A20-D4 heterodimer. This led to the 3.8 Å cryo-electron microscopy (cryo-EM) structure of the DNA-free form of the holoenzyme. The model of the holoenzyme was constructed from high-resolution structures of the components of the complex and the A20 structure predicted by AlphaFold 2. The structures do not change in the context of the holoenzyme compared to the previously determined crystal and NMR structures, but the E9 thumb domain became disordered. The E9-A20-D4 structure shows the same compact arrangement with D4 folded back on E9 as observed for the recently solved MPXV holoenzyme structures in the presence and the absence of bound DNA. A conserved interface between E9 and D4 is formed by a cluster of hydrophobic residues. Small-angle X-ray scattering data show that other, more open conformations of E9-A20-D4 without the E9-D4 contact exist in solution using the flexibility of two hinge regions in A20. Biolayer interferometry (BLI) showed that the E9-D4 interaction is indeed weak and transient in the absence of DNA although it is very important, as it has not been possible to obtain viable viruses carrying mutations of key residues within the E9-D4 interface.


Subject(s)
Cryoelectron Microscopy , DNA-Directed DNA Polymerase , Vaccinia virus , Vaccinia virus/enzymology , DNA-Directed DNA Polymerase/metabolism , DNA-Directed DNA Polymerase/chemistry , Holoenzymes/chemistry , Holoenzymes/metabolism , Viral Proteins/metabolism , Viral Proteins/chemistry , Viral Proteins/genetics , Animals , Humans , Models, Molecular , Protein Conformation , Crystallography, X-Ray
2.
J Infect Dis ; 228(10): 1421-1429, 2023 11 11.
Article in English | MEDLINE | ID: mdl-37224627

ABSTRACT

BACKGROUND: On May 6, 2022, a powerful outbreak of monkeypox virus (MPXV) had been reported outside of Africa, with many continuing new cases being reported around the world. Analysis of mutations among the 2 different lineages present in the 2021 and 2022 outbreaks revealed the presence of G->A mutations occurring in the 5'GpA context, indicative of APOBEC3 cytidine deaminase activity. METHODS: By using a sensitive polymerase chain reaction (differential DNA denaturation PCR) method allowing differential amplification of AT-rich DNA, we analyzed the level of APOBEC3-induced MPXV editing in infected cells and in patients. RESULTS: We demonstrate that G->A hypermutated MPXV genomes can be recovered experimentally from APOBEC3 transfection followed by MPXV infection. Here, among the 7 human APOBEC3 cytidine deaminases (A3A-A3C, A3DE, A3F-A3H), only APOBEC3F was capable of extensively deaminating cytidine residues in MPXV genomes. Hyperedited genomes were also recovered in ∼42% of analyzed patients. Moreover, we demonstrate that substantial repair of these mutations occurs. Upon selection, corrected G->A mutations escaping drift loss contribute to the MPXV evolution observed in the current epidemic. CONCLUSIONS: Stochastic or transient overexpression of the APOBEC3F gene exposes the MPXV genome to a broad spectrum of mutations that may be modeling the mutational landscape after multiple cycles of viral replication.


Subject(s)
Cytidine Deaminase , Monkeypox virus , Humans , Monkeypox virus/genetics , Cytidine Deaminase/genetics , Mutation , Disease Outbreaks , Cytidine , Cytosine Deaminase/chemistry , Cytosine Deaminase/genetics
3.
Virologie (Montrouge) ; 28(1): 23-35, 2024 02 01.
Article in French | MEDLINE | ID: mdl-38450665

ABSTRACT

In the spring of 2022, an epidemic due to human monkeypox virus (MPXV) of unprecedented magnitude spread across all continents. Although this event was surprising in its suddenness, the resurgence of a virus from the Poxviridae family is not surprising in a world population that has been largely naïve to these viruses since the eradication of the smallpox virus in 1980 and the concomitant cessation of vaccination. Since then, a vaccine and two antiviral compounds have been developed to combat a possible return of smallpox. However, the use of these treatments during the 2022 MPXV epidemic showed certain limitations, indicating the importance of continuing to develop the therapeutic arsenal against these viruses. For several decades, efforts to understand the molecular mechanisms involved in the synthesis of the DNA genome of these viruses have been ongoing. Although many questions remain unanswered up to now, the three-dimensional structures of essential proteins, and in particular of the DNA polymerase holoenzyme in complex with DNA, make it possible to consider the development of a model for poxvirus DNA replication. In addition, these structures are valuable tools for the development of new antivirals targeting viral genome synthesis. This review will first present the molecules approved for the treatment of poxvirus infections, followed by a review of our knowledge of the replication machinery of these viruses. Finally, we will describe how these proteins could be the target of new antiviral compounds.


Subject(s)
Mpox (monkeypox) , Poxviridae , Variola virus , Humans , Poxviridae/genetics , Variola virus/genetics , DNA , DNA Replication , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use
4.
Article in English | MEDLINE | ID: mdl-31932370

ABSTRACT

Forty years after the last endemic smallpox case, variola virus (VARV) is still considered a major threat to humans due to its possible use as a bioterrorism agent. For many years, the risk of disease reemergence was thought to solely be through deliberate misuse of VARV strains kept in clandestine laboratories. However, recent experiments using synthetic biology have proven the feasibility of recreating a poxvirus de novo, implying that VARV could, in theory, be resurrected. Because of this new perspective, the WHO Advisory Committee on VARV Research released new recommendations concerning research on poxviruses that strongly encourages pursuing the development of new antiviral drugs against orthopoxviruses. In 2018, the U.S. FDA advised in favor of two molecules for smallpox treatment, tecovirimat and brincidofovir. This review highlights the difficulties to develop new drugs targeting an eradicated disease, especially as it requires working under the FDA "animal efficacy rule" with the few, and imperfect, animal models available.


Subject(s)
Antiviral Agents/pharmacology , Drug Discovery/methods , Smallpox/drug therapy , Variola virus/drug effects , Animals , Benzamides/pharmacology , Biological Warfare Agents , Biomedical Research/legislation & jurisprudence , Cytosine/analogs & derivatives , Cytosine/pharmacology , Disease Models, Animal , Isoindoles/pharmacology , Organophosphonates/pharmacology , Smallpox/virology
6.
J Virol ; 90(9): 4604-4613, 2016 May.
Article in English | MEDLINE | ID: mdl-26912611

ABSTRACT

UNLABELLED: 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.


Subject(s)
DNA Helicases/chemistry , DNA Primase/chemistry , Models, Molecular , Protein Interaction Domains and Motifs , Vaccinia virus , Viral Proteins/chemistry , Adenosine Triphosphatases/metabolism , DNA Helicases/metabolism , DNA Primase/metabolism , DNA, Viral/metabolism , Enzyme Activation , Kinetics , Microscopy, Electron , Protein Binding , Protein Multimerization , Recombinant Fusion Proteins , Viral Proteins/metabolism
7.
J Biol Chem ; 290(29): 17923-17934, 2015 Jul 17.
Article in English | MEDLINE | ID: mdl-26045555

ABSTRACT

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.


Subject(s)
DNA/metabolism , Uracil-DNA Glycosidase/chemistry , Vaccinia virus/enzymology , Amino Acid Sequence , Crystallography, X-Ray , DNA/chemistry , Humans , Molecular Docking Simulation , Molecular Sequence Data , Nucleic Acid Conformation , Protein Conformation , Sequence Alignment , Uracil-DNA Glycosidase/metabolism , Vaccinia/virology , Vaccinia virus/chemistry , Vaccinia virus/metabolism
8.
PLoS Pathog ; 10(3): e1003978, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24603707

ABSTRACT

Vaccinia virus polymerase holoenzyme is composed of the DNA polymerase E9, the uracil-DNA glycosylase D4 and A20, a protein with no known enzymatic activity. The D4/A20 heterodimer is the DNA polymerase co-factor whose function is essential for processive DNA synthesis. Genetic and biochemical data have established that residues located in the N-terminus of A20 are critical for binding to D4. However, no information regarding the residues of D4 involved in A20 binding is yet available. We expressed and purified the complex formed by D4 and the first 50 amino acids of A20 (D4/A201₋50). We showed that whereas D4 forms homodimers in solution when expressed alone, D4/A201₋50 clearly behaves as a heterodimer. The crystal structure of D4/A201₋50 solved at 1.85 Å resolution reveals that the D4/A20 interface (including residues 167 to 180 and 191 to 206 of D4) partially overlaps the previously described D4/D4 dimer interface. A201₋50 binding to D4 is mediated by an α-helical domain with important leucine residues located at the very N-terminal end of A20 and a second stretch of residues containing Trp43 involved in stacking interactions with Arg167 and Pro173 of D4. Point mutations of the latter residues disturb D4/A201₋50 formation and reduce significantly thermal stability of the complex. Interestingly, small molecule docking with anti-poxvirus inhibitors selected to interfere with D4/A20 binding could reproduce several key features of the D4/A201₋50 interaction. Finally, we propose a model of D4/A201₋50 in complex with DNA and discuss a number of mutants described in the literature, which affect DNA synthesis. Overall, our data give new insights into the assembly of the poxvirus DNA polymerase cofactor and may be useful for the design and rational improvement of antivirals targeting the D4/A20 interface.


Subject(s)
DNA-Directed DNA Polymerase/chemistry , Vaccinia virus/chemistry , Vaccinia virus/enzymology , Animals , Chromatography, Gel , Crystallography , DNA-Directed DNA Polymerase/ultrastructure , Escherichia coli , Holoenzymes/chemistry , Holoenzymes/ultrastructure , Molecular Docking Simulation , Protein Subunits/chemistry , Vaccinia virus/ultrastructure
9.
Virologie (Montrouge) ; 20(4): 218-230, 2016 Aug 01.
Article in English | MEDLINE | ID: mdl-32260034

ABSTRACT

Parapoxviruses, double-stranded DNA viruses of the Poxviridæ family, are etiologic agents of cutaneaous infectious diseases among farm animals. These highly contagious viruses are responsible for wide outbreaks among livestock. The clinical manifestations are generally mild and consist of cutaneous or mucosal lesions, which resolve spontaneously within a few weeks. However, secondary bacterial or fungal infections on the lesion sites can aggravate the symptoms. Sore lesions located within the oral cavity and on the udders can impair feeding or nursing, thus leading to death. Livestock parapoxviruses can infect humans by direct or indirect transmission and affect mainly farmers, slaughters and veterinarians. Human symptoms generally consist of small cutaneous lesions located at the inoculation points but more severe forms can occur, peculiarly in immunocompromised persons. The parapoxvirus epidemiology is poorly understood: their respective host range and ecology among wild animals are to be clarified. The identification of parapoxviruses among marine mammals suggests that the genetic diversity within the genus is still underestimated.

10.
J Virol ; 87(3): 1679-89, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23175373

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.


Subject(s)
DNA Replication , Macromolecular Substances/ultrastructure , Vaccinia virus/physiology , Vaccinia virus/ultrastructure , Virus Replication , Microscopy, Electron , Protein Interaction Mapping , Scattering, Small Angle , Viral Proteins/metabolism , Viral Proteins/ultrastructure
11.
Virologie (Montrouge) ; 16(4): 210-224, 2012 Aug 01.
Article in English | MEDLINE | ID: mdl-33065882

ABSTRACT

Poxviruses are distinguished from other DNA viruses by replicating exclusively in the cytoplasm of the infected host cell. Replication of the linear double-stranded DNA genome takes place in the perinuclear area, in cytoplasmic foci called viral factories. Poxvirus genome organization evolved in order to prevent the virus from being dependent on nuclear enzymes. Therefore, they encode most, if not all, of the proteins required for efficient replication of their genome. Some of these proteins are essential for virus growth (i.e., enzymes directly involved in DNA synthesis). In contrast, others are dispensable for virus propagation in cell culture (i.e., proteins involved in nucleotide metabolism). Most of our knowledge concerning poxvirus replication comes from studies performed on vaccinia virus, the virus used as vaccine to eradicate smallpox more than 30 years ago. This article reviews our current knowledge of the molecular mechanisms governing poxvirus genome synthesis, with a particular focus on the viral proteins involved in this process. A working model for poxvirus DNA replication is also presented.

12.
Viruses ; 14(7)2022 07 18.
Article in English | MEDLINE | ID: mdl-35891539

ABSTRACT

The vaccinia virus (VACV) was previously used as a vaccine for smallpox eradication. Nowadays, recombinant VACVs are developed as vaccine platforms for infectious disease prevention and cancer treatment. The conventional method for genome editing of the VACV is based on homologous recombination, which is poorly efficient. Recently, the use of CRISPR/Cas9 technology was shown to greatly improve the speed and efficiency of the production of recombinant VACV expressing a heterologous gene. However, the ability to rapidly recover viruses bearing single nucleotide substitutions is still challenging. Notwithstanding, ongoing studies on the VACV and its interaction with the host cell could benefit from viral gene targeted mutagenesis. Here, we present a modified version of the CRISPR/Cas9 system for the rapid selection of mutant VACV carrying point mutations. For this purpose, we introduced a silent mutation into the donor gene (which will replace the wildtype gene) that serves a double function: it is located in the PAM (NGG) sequence, which is essential for Cas9 cleavage, and it alters a restriction site. This silent mutation, once introduced into the VACV genome, allows for rapid selection and screening of mutant viruses carrying a mutation of interest in the targeted gene. As a proof of concept, we produced several recombinant VACVs, with mutations in the E9L gene, upon which, phenotypic analysis was performed.


Subject(s)
CRISPR-Cas Systems , Vaccinia virus , Base Sequence , Gene Editing/methods , Point Mutation , Vaccinia virus/genetics
13.
Viruses ; 14(2)2022 02 10.
Article in English | MEDLINE | ID: mdl-35215961

ABSTRACT

A modified SELEX (Systematic Evolution of Ligands by Exponential Enrichment) pr,otocol (referred to as PT SELEX) was used to select primer-template (P/T) sequences that bound to the vaccinia virus polymerase catalytic subunit (E9) with enhanced affinity. A single selected P/T sequence (referred to as E9-R5-12) bound in physiological salt conditions with an apparent equilibrium dissociation constant (KD,app) of 93 ± 7 nM. The dissociation rate constant (koff) and binding half-life (t1/2) for E9-R5-12 were 0.083 ± 0.019 min-1 and 8.6 ± 2.0 min, respectively. The values indicated a several-fold greater binding ability compared to controls, which bound too weakly to be accurately measured under the conditions employed. Loop-back DNA constructs with 3'-recessed termini derived from E9-R5-12 also showed enhanced binding when the hybrid region was 21 nucleotides or more. Although the sequence of E9-R5-12 matched perfectly over a 12-base-pair segment in the coding region of the virus B20 protein, there was no clear indication that this sequence plays any role in vaccinia virus biology, or a clear reason why it promotes stronger binding to E9. In addition to E9, five other polymerases (HIV-1, Moloney murine leukemia virus, and avian myeloblastosis virus reverse transcriptases (RTs), and Taq and Klenow DNA polymerases) have demonstrated strong sequence binding preferences for P/Ts and, in those cases, there was biological or potential evolutionary relevance. For the HIV-1 RT, sequence preferences were used to aid crystallization and study viral inhibitors. The results suggest that several other DNA polymerases may have P/T sequence preferences that could potentially be exploited in various protocols.


Subject(s)
DNA, Viral/biosynthesis , DNA-Directed DNA Polymerase/metabolism , Vaccinia virus/enzymology , Viral Proteins/metabolism , Avian Myeloblastosis Virus/genetics , Avian Myeloblastosis Virus/metabolism , Base Sequence , DNA-Directed DNA Polymerase/genetics , HIV Reverse Transcriptase/genetics , HIV Reverse Transcriptase/metabolism , Moloney murine leukemia virus/genetics , Moloney murine leukemia virus/metabolism , Protein Binding , SELEX Aptamer Technique , Vaccinia virus/genetics , Viral Proteins/genetics , Virus Replication
14.
Nat Microbiol ; 7(12): 1951-1955, 2022 12.
Article in English | MEDLINE | ID: mdl-36344621

ABSTRACT

The ongoing monkeypox virus (MPXV) outbreak is the largest ever recorded outside of Africa. We isolated and sequenced a virus from the first clinical MPXV case diagnosed in France (May 2022). We report that tecovirimat (ST-246), a US Food and Drug Administration approved drug, is efficacious against this isolate in vitro at nanomolar concentrations, whereas cidofovir is only effective at micromolar concentrations. Our results support the use of tecovirimat in ongoing human clinical trials.


Subject(s)
Monkeypox virus , Mpox (monkeypox) , United States , Humans , Mpox (monkeypox)/drug therapy , Isoindoles/pharmacology , Isoindoles/therapeutic use , Benzamides/pharmacology , Benzamides/therapeutic use
15.
Virol J ; 8: 249, 2011 May 21.
Article in English | MEDLINE | ID: mdl-21600011

ABSTRACT

BACKGROUND: The genus Nairovirus in the family Bunyaviridae contains 34 tick-borne viruses classified into seven serogroups. Hazara virus (HAZV) belongs to the Crimean-Congo hemorrhagic fever (CCHF) serogroup that also includes CCHF virus (CCHFV) a major pathogen for humans. HAZV is an interesting model to study CCHFV due to a close serological and phylogenetical relationship and a classification which allows handling in a BSL2 laboratory. Nairoviruses are characterized by a tripartite negative-sense single stranded RNA genome (named L, M and S segments) that encode the RNA polymerase, the Gn-Gc glycoproteins and the nucleoprotein (NP), respectively. Currently, there are neither vaccines nor effective therapies for the treatment of any bunyavirus infection in humans. In this study we report, for the first time, the use of RNA interference (RNAi) as an approach to inhibit nairovirus replication. RESULTS: Chemically synthesized siRNAs were designed to target the mRNA produced by the three genomic segments. We first demonstrated that the siRNAs targeting the NP mRNA displayed a stronger antiviral effect than those complementary to the L and M transcripts in A549 cells. We further characterized the two most efficient siRNAs showing, that the induced inhibition is specific and associated with a decrease in NP synthesis during HAZV infection. Furthermore, both siRNAs depicted an antiviral activity when used before and after HAZV infection. We next showed that HAZV was sensitive to ribavirin which is also known to inhibit CCHFV. Finally, we demonstrated the additive or synergistic antiviral effect of siRNAs used in combination with ribavirin. CONCLUSIONS: Our study highlights the interest of using RNAi (alone or in combination with ribavirin) to treat nairovirus infection. This approach has to be considered for the development of future antiviral compounds targeting CCHFV, the most pathogenic nairovirus.


Subject(s)
Antiviral Agents/pharmacology , Biological Products/pharmacology , Nairovirus/drug effects , Nairovirus/physiology , RNA, Small Interfering/pharmacology , Ribavirin/pharmacology , Virus Replication/drug effects , Animals , Cell Line , Chlorocebus aethiops , Drug Synergism , Humans , Microbial Sensitivity Tests
16.
J Mol Biol ; 433(13): 167009, 2021 06 25.
Article in English | MEDLINE | ID: mdl-33901538

ABSTRACT

Poxviruses are enveloped viruses with a linear, double-stranded DNA genome. Viral DNA synthesis is achieved by a functional DNA polymerase holoenzyme composed of three essential proteins. For vaccinia virus (VACV) these are E9, the catalytic subunit, a family B DNA polymerase, and the heterodimeric processivity factor formed by D4 and A20. The A20 protein links D4 to the catalytic subunit. High-resolution structures have been obtained for the VACV D4 protein in complex with an N-terminal fragment of A20 as well as for E9. In addition, biochemical studies provided evidence that a poxvirus-specific insertion (insert 3) in E9 interacts with the C-terminal residues of A20. Here, we provide solution structures of two different VACV A20 C-terminal constructs containing residues 304-426, fused at their C-terminus to either a BAP (Biotin Acceptor Peptide)-tag or a short peptide containing the helix of E9 insert 3. Together with results from titration studies, these structures shed light on the molecular interface between the catalytic subunit and the processivity factor component A20. The interface comprises hydrophobic residues conserved within the Chordopoxvirinae subfamily. Finally, we constructed a HADDOCK model of the VACV A20304-426-E9 complex, which is in excellent accordance with previous experimental data.


Subject(s)
DNA-Directed DNA Polymerase/chemistry , Protein Domains , Vaccinia virus/enzymology , Viral Proteins/chemistry , Amino Acid Sequence , Catalytic Domain/genetics , Crystallography, X-Ray , DNA, Viral/chemistry , DNA, Viral/genetics , DNA, Viral/metabolism , DNA-Directed DNA Polymerase/genetics , DNA-Directed DNA Polymerase/metabolism , Holoenzymes/chemistry , Holoenzymes/genetics , Holoenzymes/metabolism , Models, Molecular , Peptides/chemistry , Peptides/genetics , Peptides/metabolism , Protein Binding , Sequence Homology, Amino Acid , Solutions/chemistry , Vaccinia virus/genetics , Viral Proteins/genetics , Viral Proteins/metabolism , Virus Replication/genetics
17.
Med Sci (Paris) ; 36(8-9): 797-802, 2020.
Article in French | MEDLINE | ID: mdl-32755538

ABSTRACT

SARS-CoV-2 (severe acute respiratory syndrome-coronavirus-2, which emerged in China at the end of 2019, is responsible for a global health crisis resulting in the confinement of more than 3 billion people worldwide and the sharp decline of the world economy. In this context, a race against the clock is launched in order to develop a treatment to stop the pandemic as soon as possible. A study published in Nature by the Volker Thiel team reports the development of reverse genetics for SARS-CoV-2 allowing them to recreate the virus in just a few weeks. The perspectives of this work are very interesting since it will allow the genetic manipulation of the virus and thus the development of precious tools which will be useful to fight the infection. Even though this approach represents a technological leap that will improve our knowledge of the virus, it also carries the germ of possible misuse and the creation of the virus for malicious purposes. The advantages and disadvantages of recreating SARS-CoV-2 in this pandemic period are discussed in this mini-synthesis.


TITLE: Une course contre la montre - Création du SARS-CoV-2 en laboratoire, un mois après son émergence ! ABSTRACT: Le SARS-CoV-2 (severe acute respiratory syndrome-coronavirus-2), qui a émergé à la fin de l'année 2019 en République populaire de Chine, est responsable d'une crise sanitaire mondiale qui a entraîné le confinement de plus de 3 milliards d'individus et l'arrêt brutal de l'économie planétaire. Dans ce contexte, une course contre la montre est lancée afin de développer, dans les plus brefs délais, un traitement permettant d'enrayer la pandémie. Une étude de l'équipe de Volker Thiel, parue dans le journal Nature, rapporte la mise au point d'une technique de génétique inverse pour le SARS-CoV-2, leur ayant permis de recréer le virus en seulement quelques semaines. Les perspectives de ces travaux sont très intéressantes puisqu'elles permettent d'envisager la manipulation génétique du virus et ainsi le développement d'outils précieux qui seront utiles pour combattre l'infection. Si la technique représente également un saut technologique qui permettra d'améliorer nos connaissances sur le virus, elle porte aussi en elle le germe d'un possible mésusage et la création d'un virus à des fins malveillantes. Les avantages et inconvénients de recréer le SARS-CoV-2 dans cette période de pandémie sont discutés dans cet article.


Subject(s)
Betacoronavirus/genetics , Coronavirus Infections/virology , Organisms, Genetically Modified , Pandemics , Pneumonia, Viral/virology , Reverse Genetics/methods , Betacoronavirus/pathogenicity , Biohazard Release , COVID-19 , COVID-19 Vaccines , Chromosomes, Artificial, Yeast , Cloning, Molecular/methods , Coronaviridae/classification , Coronaviridae/genetics , Coronaviridae/pathogenicity , Coronavirus Infections/prevention & control , DNA, Complementary/genetics , Host Specificity , Humans , Organisms, Genetically Modified/genetics , Organisms, Genetically Modified/pathogenicity , Pandemics/prevention & control , Pneumonia, Viral/prevention & control , RNA, Viral/genetics , Recombination, Genetic , Risk , SARS-CoV-2 , Viral Vaccines
18.
Nat Commun ; 10(1): 1181, 2019 03 12.
Article in English | MEDLINE | ID: mdl-30862836

ABSTRACT

Attachment of human adenovirus (HAd) to the host cell is a critical step of infection. Initial attachment occurs via the adenoviral fibre knob protein and a cellular receptor. Here we report the cryo-electron microscopy (cryo-EM) structure of a <100 kDa non-symmetrical complex comprising the trimeric HAd type 3 fibre knob (HAd3K) and human desmoglein 2 (DSG2). The structure reveals a unique stoichiometry of 1:1 and 2:1 (DSG2: knob trimer) not previously observed for other HAd-receptor complexes. We demonstrate that mutating Asp261 in the fibre knob is sufficient to totally abolish receptor binding. These data shed new light on adenovirus infection strategies and provide insights for adenoviral vector development and structure-based design.


Subject(s)
Adenoviruses, Human/metabolism , Capsid Proteins/metabolism , Desmoglein 2/metabolism , Receptors, Virus/metabolism , Virus Attachment , Adenoviridae Infections/pathology , Adenoviridae Infections/virology , Adenoviruses, Human/pathogenicity , Asparagine/genetics , Capsid Proteins/ultrastructure , Cryoelectron Microscopy , Desmoglein 2/ultrastructure , HEK293 Cells , Humans , Models, Molecular , Protein Domains , Receptors, Virus/ultrastructure , Recombinant Proteins/metabolism , Recombinant Proteins/ultrastructure
19.
Sci Rep ; 8(1): 8381, 2018 05 30.
Article in English | MEDLINE | ID: mdl-29849084

ABSTRACT

High-affinity binding of the trimeric fibre protein to a cell surface primary receptor is a common feature shared by all adenovirus serotypes. Recently, a long elusive species B adenovirus receptor has been identified. Desmoglein 2 (DSG2) a component of desmosomal junction, has been reported to interact at high affinity with Human adenoviruses HAd3, HAd7, HAd11 and HAd14. Little is known with respect to the molecular interactions of adenovirus fibre with the DSG2 ectodomain. By using different DSG2 ectodomain constructs and biochemical and biophysical experiments, we report that the third extracellular cadherin domain (EC3) of DSG2 is critical for HAd3 fibre binding. Unexpectedly, stoichiometry studies using multi-angle laser light scattering (MALLS) and analytical ultra-centrifugation (AUC) revealed a non-classical 1:1 interaction (one DSG2 per trimeric fibre), thus differentiating 'DSG2-interacting' adenoviruses from other protein receptor interacting adenoviruses in their infection strategy.


Subject(s)
Adenoviridae/metabolism , Desmoglein 2/metabolism , Serogroup , Adenoviridae/genetics , Desmoglein 2/chemistry , Glycosylation , Humans , Protein Binding , Protein Domains
20.
Viruses ; 10(1)2017 12 23.
Article in English | MEDLINE | ID: mdl-29295488

ABSTRACT

Since the official declaration of smallpox eradication in 1980, the general population vaccination has ceased worldwide. Therefore, people under 40 year old are generally not vaccinated against smallpox and have no cross protection against orthopoxvirus infections. This naïve population may be exposed to natural or intentional orthopoxvirus emergences. The virology unit of the Institut de Recherche Biomédicale des Armées (France) has developed research programs on orthopoxviruses since 2000. Its missions were conceived to improve the diagnosis capabilities, to foster vaccine development, and to develop antivirals targeting specific viral proteins. The role of the virology unit was asserted in 2012 when the responsibility of the National Reference Center for the Orthopoxviruses was given to the unit. This article presents the evolution of the unit activity since 2000, and the past and current research focusing on orthopoxviruses.


Subject(s)
Communicable Disease Control/trends , Orthopoxvirus/physiology , Poxviridae Infections/prevention & control , Poxviridae Infections/virology , Research/trends , Animals , Antiviral Agents/chemical synthesis , Antiviral Agents/pharmacology , Antiviral Agents/supply & distribution , France , Humans , Orthopoxvirus/classification , Orthopoxvirus/drug effects , Orthopoxvirus/genetics , Poxviridae/classification , Poxviridae/genetics , Poxviridae Infections/diagnosis , Poxviridae Infections/pathology , Smallpox Vaccine/administration & dosage , Smallpox Vaccine/biosynthesis , Smallpox Vaccine/supply & distribution , Viral Proteins/chemistry , Viral Proteins/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL