Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
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.
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
3.
Viruses ; 14(10)2022 10 07.
Article in English | MEDLINE | ID: mdl-36298761

ABSTRACT

Poxviruses are large DNA viruses with a linear double-stranded DNA genome circularized at the extremities. The helicase-primase D5, composed of six identical 90 kDa subunits, is required for DNA replication. D5 consists of a primase fragment flexibly attached to the hexameric C-terminal polypeptide (res. 323-785) with confirmed nucleotide hydrolase and DNA-binding activity but an elusive helicase activity. We determined its structure by single-particle cryo-electron microscopy. It displays an AAA+ helicase core flanked by N- and C-terminal domains. Model building was greatly helped by the predicted structure of D5 using AlphaFold2. The 3.9 Å structure of the N-terminal domain forms a well-defined tight ring while the resolution decreases towards the C-terminus, still allowing the fit of the predicted structure. The N-terminal domain is partially present in papillomavirus E1 and polyomavirus LTA helicases, as well as in a bacteriophage NrS-1 helicase domain, which is also closely related to the AAA+ helicase domain of D5. Using the Pfam domain database, a D5_N domain followed by DUF5906 and Pox_D5 domains could be assigned to the cryo-EM structure, providing the first 3D structures for D5_N and Pox_D5 domains. The same domain organization has been identified in a family of putative helicases from large DNA viruses, bacteriophages, and selfish DNA elements.


Subject(s)
DNA Primase , Vaccinia virus , DNA Primase/chemistry , DNA Primase/genetics , DNA Primase/metabolism , Cryoelectron Microscopy , Vaccinia virus/genetics , DNA Helicases/genetics , DNA , DNA Replication , Nucleotides
4.
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
5.
Acta Crystallogr F Struct Biol Commun ; 75(Pt 12): 750-757, 2019 Dec 01.
Article in English | MEDLINE | ID: mdl-31797817

ABSTRACT

The cryo-electron microscopy (cryo-EM) structure of the complex between the trimeric human adenovirus B serotype 3 fibre knob and human desmoglein 2 fragments containing cadherin domains EC2 and EC3 has been published, showing 3:1 and 3:2 complexes. Here, the crystal structure determined at 4.5 Šresolution is presented with one EC2-EC3 desmoglein fragment bound per fibre knob monomer in the asymmetric unit, leading to an apparent 3:3 stoichiometry. However, in concentrated solution the 3:2 complex is predominant, as shown by small-angle X-ray scattering (SAXS), while cryo-EM at lower concentrations showed a majority of the 3:1 complex. Substitution of the calcium ions bound to the desmoglein domains by terbium ions allowed confirmation of the X-ray model using their anomalous scattering and shows that at least one binding site per cluster of calcium ions is intact and exchangeable and, combined with SAXS data, that the cadherin domains are folded even in the distal part that is invisible in the cryo-EM reconstruction.


Subject(s)
Adenoviruses, Human/metabolism , Cadherins/metabolism , Capsid Proteins/chemistry , Capsid Proteins/metabolism , Desmoglein 2/chemistry , Desmoglein 2/metabolism , Adenoviruses, Human/classification , Amino Acid Sequence , Cadherins/chemistry , Crystallization , Crystallography, X-Ray , Humans , Models, Molecular , Protein Binding , Protein Conformation , Protein Domains , Serogroup
6.
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
7.
Nat Commun ; 8(1): 1455, 2017 11 13.
Article in English | MEDLINE | ID: mdl-29129932

ABSTRACT

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.


Subject(s)
Catalytic Domain/genetics , DNA-Directed DNA Polymerase/ultrastructure , Vaccinia virus/enzymology , Crystallography, X-Ray , DNA Glycosylases/genetics , DNA-Binding Proteins/genetics , DNA-Directed DNA Polymerase/genetics , Nucleoside-Triphosphatase/genetics
8.
Acta Crystallogr F Struct Biol Commun ; 72(Pt 9): 687-91, 2016 09.
Article in English | MEDLINE | ID: mdl-27599859

ABSTRACT

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.


Subject(s)
DNA-Directed DNA Polymerase/chemistry , Point Mutation , Uracil-DNA Glycosidase/chemistry , Vaccinia virus/chemistry , Viral Proteins/chemistry , Amino Acid Motifs , Catalytic Domain , Cloning, Molecular , Crystallization , Crystallography, X-Ray , DNA-Directed DNA Polymerase/genetics , DNA-Directed DNA Polymerase/metabolism , Gene Expression , Models, Molecular , Plasmids/chemistry , Plasmids/metabolism , Protein Conformation , Protein Multimerization , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Uracil-DNA Glycosidase/genetics , Uracil-DNA Glycosidase/metabolism , Vaccinia virus/metabolism , Viral Proteins/genetics , Viral Proteins/metabolism , X-Ray Diffraction
9.
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
10.
Virus Genes ; 51(2): 171-81, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26292944

ABSTRACT

The helicase-primase complex is part of the lytic DNA replication machinery of herpesviruses, but up to now, almost nothing is known about its structure. For Epstein-Barr virus it consists in the helicase BBLF4, the primase BSLF1 and the accessory protein BBLF2/3. The accessory protein shows only weak sequence homology within the herpesvirus family but may be related to an inactive B-family polymerase. BSLF1 belongs to the archaeo-eukaryotic primase family, whereas the helicase BBLF4 has been related either to Dda helicases of caudovirales or to Pif1 helicases. We produced the helicase-primase complex in insect cells using a baculovirus coding for all three proteins simultaneously. The soluble monomeric helicase-primase complex containing the three proteins with 1:1:1 stoichiometry showed ATPase activity, which is strongly stimulated in the presence of ssDNA oligomers. Furthermore, we expressed BBLF2/3 as soluble monomeric protein and performed small-angle X-ray scattering experiments which yielded an envelope whose shape is compatible with B-family polymerases.


Subject(s)
Herpesvirus 4, Human/enzymology , Herpesvirus 4, Human/genetics , Viral Proteins/genetics , Viral Proteins/metabolism , Adenosine Triphosphate/metabolism , Animals , Baculoviridae/genetics , Cell Line , DNA Helicases/genetics , DNA Helicases/metabolism , Genetic Vectors , Insecta , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Scattering, Small Angle , Viral Proteins/chemistry
11.
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
12.
PLoS One ; 10(3): e0119289, 2015.
Article in English | MEDLINE | ID: mdl-25794153

ABSTRACT

The vesivirus feline calicivirus (FCV) is a positive strand RNA virus encapsidated by an icosahedral T=3 shell formed by the viral VP1 protein. Upon its expression in the insect cell - baculovirus system in the context of vaccine development, two types of virus-like particles (VLPs) were formed, a majority built of 60 subunits (T=1) and a minority probably built of 180 subunits (T=3). The structure of the small particles was determined by x-ray crystallography at 0.8 nm resolution helped by cryo-electron microscopy in order to understand their formation. Cubic crystals belonged to space group P213. Their self-rotation function showed the presence of an octahedral pseudo-symmetry similar to the one described previously by Agerbandje and co-workers for human parvovirus VLPs. The crystal structure could be solved starting from the published VP1 structure in the context of the T=3 viral capsid. In contrast to viral capsids, where the capsomers are interlocked by the exchange of the N-terminal arm (NTA) domain, this domain is disordered in the T=1 capsid of the VLPs. Furthermore it is prone to proteolytic cleavage. The relative orientation of P (protrusion) and S (shell) domains is alerted so as to fit VP1 to the smaller T=1 particle whereas the intermolecular contacts around 2-fold, 3-fold and 5-fold axes are conserved. By consequence the surface of the VLP is very similar compared to the viral capsid and suggests a similar antigenicity. The knowledge of the structure of the VLPs will help to improve their stability, in respect to a use for vaccination.


Subject(s)
Calicivirus, Feline/ultrastructure , Virion/ultrastructure , Amino Acid Sequence , Animals , Calicivirus, Feline/genetics , Capsid Proteins/chemistry , Capsid Proteins/genetics , Cats , Models, Molecular , Molecular Sequence Data , Protein Conformation , Sequence Alignment
13.
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
14.
J Antimicrob Chemother ; 69(1): 101-10, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23963236

ABSTRACT

OBJECTIVES: Francisella tularensis, a CDC class A potential bioterrorism agent, is a Gram-negative bacterium responsible for tularaemia. Understanding the mechanisms of resistance to antibiotics used as first-line treatment is of major security relevance. METHODS: We propagated the three parental reference strains Francisella tularensis subsp. holarctica live vaccine strain, Francisella novicida and Francisella philomiragia with increasing concentrations of ciprofloxacin, a fluoroquinolone used as curative and prophylactic treatment for tularaemia. This evolution procedure provided us with high-level ciprofloxacin-resistant mutants and all evolutionary intermediates towards high-level resistance. We determined the resistance levels to other fluoroquinolones (levofloxacin and moxifloxacin) and other antibiotic families (aminoglycosides, tetracyclines and macrolides) and characterized the genetic changes in the fluoroquinolone target genes encoding DNA gyrase and topoisomerase IV. RESULTS: All high-level resistant mutants shared cross-resistance to the tested fluoroquinolones, while some also revealed striking levels of cross-resistance to other clinically relevant antibiotic classes. High-level resistant mutants carried one to three mutations, including some not previously reported. We mapped all mutations onto known topoisomerase three-dimensional structures. Along the pathways towards high-level resistance, we identified complex evolutionary trajectories including polymorphic states and additional resistance mechanisms likely to be associated with efflux processes. CONCLUSIONS: Our data demonstrated the efficiency and speed of in vitro production of mutants highly resistant to fluoroquinolones in Francisella species. They emphasize the urgent need to identify all antibiotic resistance mechanisms in these species, develop molecular tools for their detection and design new therapeutic alternatives for tularaemia.


Subject(s)
Anti-Bacterial Agents/pharmacology , Drug Resistance, Bacterial , Fluoroquinolones/pharmacology , Francisella/drug effects , DNA Gyrase/genetics , DNA Mutational Analysis , DNA Topoisomerase IV/genetics , Francisella/enzymology , Francisella/genetics , Francisella/growth & development , Humans , Microbial Sensitivity Tests , Selection, Genetic , Serial Passage
15.
Eur J Cell Biol ; 92(3): 105-11, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23395486

ABSTRACT

Ubinuclein-1 is a NACos (Nuclear and Adhesion junction Complex components) protein which shuttles between the nucleus and tight junctions, but its function in the latter is not understood. Here, by co-immunoprecipitation and confocal analysis, we show that Ubinuclein-1 interacts with the 14-3-3ɛ protein both in HT29 colon cells, and AGS gastric cells. This interaction is mediated by an Ubinuclein-1 phosphoserine motif. We show that the arginine residues (R56, R60 and R132) which form the 14-3-3ɛ ligand binding site are responsible for the binding of 14-3-3ɛ to phosphorylated Ubinuclein-1. Furthermore, we demonstrate that in vitro Ubinuclein-1 can be directly phosphorylated by cAMP-dependent protein kinase A. This in vitro phosphorylation allows binding of wildtype 14-3-3ɛ. Moreover, treatment of the cells with inhibitors of the cAMP-dependent protein kinase, KT5720 or H89, modifies the subcellular localization of Ubinuclein-1. Indeed, KT5720 and H89 greatly increase the staining of Ubinuclein-1 at the tight junctions in AGS gastric cells. In the presence of the kinase inhibitor KT5720, the amount of Ubinuclein-1 in the NP40 insoluble fraction is increased, together with actin. Moreover, treatment of the cells with KT5720 or H89 induces the concentration of Ubinuclein-1 at tricellular intersections of MDCK cells. Taken together, our findings demonstrate novel cell signaling trafficking by Ubinuclein-1 via association with 14-3-3ɛ following Ubinuclein-1 phosphorylation by the cAMP-dependent protein kinase-A.


Subject(s)
14-3-3 Proteins/metabolism , Adherens Junctions/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Epithelial Cells/metabolism , Tight Junctions/metabolism , Amino Acid Sequence , Animals , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Dogs , HT29 Cells , Humans , Madin Darby Canine Kidney Cells , Mice , Molecular Sequence Data , Signal Transduction
16.
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
17.
PLoS One ; 7(9): e46075, 2012.
Article in English | MEDLINE | ID: mdl-23049939

ABSTRACT

During the viral life cycle adenoviruses produce excess capsid proteins. Human adenovirus serotype 3 (Ad3) synthesizes predominantly an excess of free pentons, the complexes of pentameric penton base and trimeric fiber proteins, which are responsible for virus penetration. In infected cells Ad3 pentons spontaneously assemble into dodecahedral virus-like nano-particles containing twelve pentons. They also form in insect cells during expression in the baculovirus system. Similarly, in the absence of fiber protein dodecahedric particles built of 12 penton base pentamers can be produced. Both kinds of dodecahedra show remarkable efficiency of intracellular penetration and can be engineered to deliver several millions of foreign cargo molecules to a single target cell. For this reason, they are of great interest as a delivery vector. In order to successfully manipulate this potential vector for drug and/or gene delivery, an understanding of the molecular basis of vector assembly and integrity is critical. Crystallographic data in conjunction with site-directed mutagenesis and biochemical analysis provide a model for the molecular determinants of dodecamer particle assembly and the requirements for stability. The 3.8 Å crystal structure of Ad3 penton base dodecamer (Dd) shows that the dodecahedric structure is stabilized by strand-swapping between neighboring penton base molecules. Such N-terminal strand-swapping does not occur for Dd of Ad2, a serotype which does not form Dd under physiological conditions. This unique stabilization of the Ad3 dodecamer is controlled by residues 59-61 located at the site of strand switching, the residues involved in putative salt bridges between pentamers and by the disordered N-terminus (residues 1-47), as confirmed by site directed mutagenesis and biochemical analysis of mutant and wild type protein. We also provide evidence that the distal N-terminal residues are externally exposed and available for attaching cargo.


Subject(s)
Adenoviruses, Human/metabolism , Capsid Proteins/chemistry , Capsid Proteins/metabolism , Amino Acid Sequence , Capsid Proteins/genetics , Crystallography, X-Ray , Humans , Molecular Sequence Data , Mutagenesis, Site-Directed , Protein Structure, Secondary , Sequence Homology, Amino Acid
18.
Nat Struct Mol Biol ; 19(9): 938-47, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22902366

ABSTRACT

Hematopoietic human colony-stimulating factor 1 (hCSF-1) is essential for innate and adaptive immunity against viral and microbial infections and cancer. The human pathogen Epstein-Barr virus secretes the lytic-cycle protein BARF1 that neutralizes hCSF-1 to achieve immunomodulation. Here we show that BARF1 binds the dimer interface of hCSF-1 with picomolar affinity, away from the cognate receptor-binding site, to establish a long-lived complex featuring three hCSF-1 at the periphery of the BARF1 toroid. BARF1 locks dimeric hCSF-1 into an inactive conformation, rendering it unable to signal via its cognate receptor on human monocytes. This reveals a new functional role for hCSF-1 cooperativity in signaling. We propose a new viral strategy paradigm featuring an allosteric decoy receptor of the competitive type, which couples efficient sequestration and inactivation of the host growth factor to abrogate cooperative assembly of the cognate signaling complex.


Subject(s)
Epstein-Barr Virus Infections/metabolism , Herpesvirus 4, Human/physiology , Host-Pathogen Interactions , Macrophage Colony-Stimulating Factor/metabolism , Monocytes/virology , Viral Proteins/metabolism , Amino Acid Sequence , Animals , Binding Sites , Cells, Cultured , Crystallography, X-Ray , Herpesvirus 4, Human/metabolism , Humans , Macrophage Colony-Stimulating Factor/chemistry , Mice , Mice, Inbred C57BL , Models, Molecular , Molecular Sequence Data , Monocytes/cytology , Protein Binding , Protein Conformation , Protein Multimerization , Signal Transduction , Viral Proteins/chemistry
19.
J Virol ; 86(17): 9175-87, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22696660

ABSTRACT

Replication of the human herpesvirus Epstein-Barr virus drastically impairs cellular protein synthesis. This shutoff phenotype results from mRNA degradation upon expression of the early lytic-phase protein BGLF5. Interestingly, BGLF5 is the viral DNase, or alkaline exonuclease, homologues of which are present throughout the herpesvirus family. During productive infection, this DNase is essential for processing and packaging of the viral genome. In contrast to this widely conserved DNase activity, shutoff is only mediated by the alkaline exonucleases of the subfamily of gammaherpesviruses. Here, we show that BGLF5 can degrade mRNAs of both cellular and viral origin, irrespective of polyadenylation. Furthermore, shutoff by BGLF5 induces nuclear relocalization of the cytosolic poly(A) binding protein. Guided by the recently resolved BGLF5 structure, mutants were generated and analyzed for functional consequences on DNase and shutoff activities. On the one hand, a point mutation destroying DNase activity also blocks RNase function, implying that both activities share a catalytic site. On the other hand, other mutations are more selective, having a more pronounced effect on either DNA degradation or shutoff. The latter results are indicative of an oligonucleotide-binding site that is partially shared by DNA and RNA. For this, the flexible "bridge" that crosses the active-site canyon of BGLF5 appears to contribute to the interaction with RNA substrates. These findings extend our understanding of the molecular basis for the shutoff function of BGLF5 that is conserved in gammaherpesviruses but not in alpha- and betaherpesviruses.


Subject(s)
Deoxyribonucleases/chemistry , Deoxyribonucleases/metabolism , Epstein-Barr Virus Infections/genetics , Epstein-Barr Virus Infections/virology , Herpesvirus 4, Human/enzymology , Herpesvirus 4, Human/physiology , Viral Proteins/chemistry , Viral Proteins/metabolism , Amino Acid Motifs , Amino Acid Sequence , Cell Line , Deoxyribonucleases/genetics , Epstein-Barr Virus Infections/metabolism , Herpesvirus 4, Human/chemistry , Herpesvirus 4, Human/genetics , Humans , Molecular Sequence Data , Protein Biosynthesis , RNA Stability , Sequence Alignment , Viral Proteins/genetics , Virus Replication
20.
Exp Cell Res ; 318(5): 509-20, 2012 Mar 10.
Article in English | MEDLINE | ID: mdl-22245583

ABSTRACT

We have previously characterized ubinuclein (Ubn-1) as a NACos (Nuclear and Adherent junction Complex components) protein which interacts with viral or cellular transcription factors and the tight junction (TJ) protein ZO-1. The purpose of the present study was to get more insights on the binding partners of Ubn-1, notably those present in the epithelial junctions. Using an in vivo assay of fluorescent protein-complementation assay (PCA), we demonstrated that the N-terminal domains of the Ubn-1 and ZO-1 proteins triggered a functional interaction inside the cell. Indeed, expression of both complementary fragments of venus fused to the N-terminal parts of Ubn-1 and ZO-1 was able to reconstitute a fluorescent venus protein. Furthermore, nuclear expression of the chimeric Ubn-1 triggered nuclear localization of the chimeric ZO-1. We could localize this interaction to the PDZ2 domain of ZO-1 using an in vitro pull-down assay. More precisely, a 184-amino acid region (from amino acids 39 to 223) at the N-terminal region of Ubn-1 was responsible for the interaction with the PDZ2 domain of ZO-1. Co-imunoprecipitation and confocal microscopy experiments also revealed the tight junction protein cingulin as a new interacting partner of Ubn-1. A proteomic approach based on mass spectrometry analysis (MS) was then undertaken to identify further binding partners of GST-Ubn-1 fusion protein in different subcellular fractions of human epithelial HT29 cells. LYRIC (Lysine-rich CEACAM1-associated protein) and RACK-1 (receptor for activated C-kinase) proteins were validated as bona fide interacting partners of Ubn-1. Altogether, these results suggest that Ubn-1 is a scaffold protein influencing protein subcellular localization and is involved in several processes such as cell-cell contact signalling or modulation of gene activity.


Subject(s)
Membrane Proteins/metabolism , Microfilament Proteins/metabolism , Nuclear Proteins/metabolism , Phosphoproteins/metabolism , Transcription Factors/metabolism , Cell Adhesion Molecules/metabolism , GTP-Binding Proteins/metabolism , HEK293 Cells , HT29 Cells , Humans , Membrane Proteins/chemistry , Neoplasm Proteins/metabolism , Nuclear Proteins/chemistry , Peptide Fragments/metabolism , Phosphoproteins/chemistry , Protein Binding , Protein Interaction Domains and Motifs , Protein Interaction Mapping , RNA-Binding Proteins , Receptors for Activated C Kinase , Receptors, Cell Surface/metabolism , Recombinant Fusion Proteins/metabolism , Tight Junctions/metabolism , Transcription Factors/chemistry , Zonula Occludens-1 Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...