Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
PLoS Pathog ; 20(5): e1011652, 2024 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-38768256

RESUMO

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.

2.
Virologie (Montrouge) ; 28(1): 23-35, 2024 02 01.
Artigo em Francês | MEDLINE | ID: mdl-38450665

RESUMO

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.


Assuntos
Mpox , Poxviridae , Vírus da Varíola , Humanos , Poxviridae/genética , Vírus da Varíola/genética , DNA , Replicação do DNA , Antivirais/farmacologia , Antivirais/uso terapêutico
3.
Viruses ; 14(11)2022 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-36366462

RESUMO

Determining the structural organisation of viral replication complexes and unravelling the impact of infection on cellular homeostasis represent important challenges in virology. This may prove particularly useful when confronted with viruses that pose a significant threat to human health, that appear unique within their family, or for which knowledge is scarce. Among Mononegavirales, bornaviruses (family Bornaviridae) stand out due to their compact genomes and their nuclear localisation for replication. The recent recognition of the zoonotic potential of several orthobornaviruses has sparked a surge of interest in improving our knowledge on this viral family. In this work, we provide a complete analysis of the structural organisation of Borna disease virus 1 (BoDV-1) phosphoprotein (P), an important cofactor for polymerase activity. Using X-ray diffusion and diffraction experiments, we revealed that BoDV-1 P adopts a long coiled-coil α-helical structure split into two parts by an original ß-strand twist motif, which is highly conserved across the members of whole Orthobornavirus genus and may regulate viral replication. In parallel, we used BioID to determine the proximal interactome of P in living cells. We confirmed previously known interactors and identified novel proteins linked to several biological processes such as DNA repair or mRNA metabolism. Altogether, our study provides important structure/function cues, which may improve our understanding of BoDV-1 pathogenesis.


Assuntos
Vírus da Doença de Borna , Bornaviridae , Animais , Humanos , Vírus da Doença de Borna/genética , Fosfoproteínas/genética , Bornaviridae/genética , Reparo do DNA , DNA , RNA Mensageiro/genética
4.
Viruses ; 14(10)2022 10 07.
Artigo em Inglês | MEDLINE | ID: mdl-36298761

RESUMO

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.


Assuntos
DNA Primase , Vaccinia virus , DNA Primase/química , DNA Primase/genética , DNA Primase/metabolismo , Microscopia Crioeletrônica , Vaccinia virus/genética , DNA Helicases/genética , DNA , Replicação do DNA , Nucleotídeos
5.
Virologie (Montrouge) ; 26(3): 240-252, 2022 05 01.
Artigo em Francês | MEDLINE | ID: mdl-35792843

RESUMO

For the past three years, the nature and evolution of human viruses have been taught in University Grenoble-Alpes without relying on the systematic list of all virus families. A «historical¼ approach allows to define three main categories of viruses following if they have co-evolved with humans for a very long time (ancient human viruses), if they began to infect humans in the Neolithic or later (recent human viruses) or if they are still animal viruses that are transmitted to humans sporadically (zoonotic viruses). We present below the principles and some examples of this pedagogic separation which has not the pretention to replace the classical taxonomic classification based on morphological and sequence similarity (ICTV classification) or on the form and replication mode of the viral genome (Baltimore classification). It helps grouping of viruses with similar effects even if their evolution is different. We show where human viruses come from and how they can cause human diseases. This approach was tested with Biology students, and then extended to Medicine and Pharmacy students to ensure that teaching was based on the same concepts in the three Faculties. In the end, all the students were very receptive and interested in this approach. Of course, different teaching methods can work, but this way of presenting things is also more fun for teachers and promotes cooperation between speakers.


Depuis trois ans, une expérience pédagogique est menée à l'université Grenoble-Alpes pour enseigner la nature et l'évolution des virus humains, sans se baser sur la liste systématique de toutes les familles de virus. Le choix a été fait d'une approche « historique ¼ des virus chez l'homme, permettant de définir trois grandes catégories de virus selon qu'ils aient co-évolué avec l'homme pendant très longtemps (virus humains anciens), ou qu'ils l'aient infecté plus récemment au Néolithique ou plus tard (virus humains récents) ou enfin qu'ils évoluent à partir de virus animaux transmis à l'homme de manière sporadique (virus zoonotiques). Nous exposons ci-dessous les principes et quelques exemples de cette distinction pédagogique alternative qui n'a pas la prétention de remplacer les classifications taxonomiques classiques basées sur les similarités morphologiques et de séquences (classification ICTV) ou sur la forme et le mode de réplication du génome viral (classification de Baltimore). Elle permet de faciliter le regroupement de virus ayant des effets similaires même si leur divergence évolutive est importante. Nous montrons ainsi l'origine des virus humains et comment ils peuvent entraîner des maladies humaines. Cette approche a été expérimentée avec les étudiants de biologie, puis étendue aux étudiants de médecine et de pharmacie, pour que l'enseignement soit basé sur les mêmes concepts dans les trois UFR. Au final, tous les étudiants ont été très réceptifs et intéressés par cette approche. Bien sûr, différentes méthodes d'enseignement peuvent fonctionner, mais cette façon de présenter les choses est également plus ludique pour les enseignants et favorise la coopération entre les intervenants.


Assuntos
Vírus , Zoonoses , Animais , Baltimore , Genoma Viral , Humanos , Vírus/genética
6.
J Mol Biol ; 434(10): 167551, 2022 05 30.
Artigo em Inglês | MEDLINE | ID: mdl-35317998

RESUMO

To understand the dynamic interactions between the phosphoprotein (P) and the nucleoprotein (N) within the transcription/replication complex of the Paramyxoviridae and to decipher their roles in regulating viral multiplication, we characterized the structural properties of the C-terminal X domain (PXD) of Nipah (NiV) and Hendra virus (HeV) P protein. In crystals, isolated NiV PXD adopted a two-helix dimeric conformation, which was incompetent for binding its partners, but in complex with the C-terminal intrinsically disordered tail of the N protein (NTAIL), it folded into a canonical 3H bundle conformation. In solution, SEC-MALLS, SAXS and NMR spectroscopy experiments indicated that both NiV and HeV PXD were larger in size than expected for compact proteins of the same molecular mass and were in conformational exchange between a compact three-helix (3H) bundle and partially unfolded conformations, where helix α3 is detached from the other two. Some measurements also provided strong evidence for dimerization of NiV PXD in solution but not for HeV PXD. Ensemble modeling of experimental SAXS data and statistical-dynamical modeling reconciled all these data, yielding a model where NiV and HeV PXD exchanged between different conformations, and where NiV but not HeV PXD formed dimers. Finally, recombinant NiV comprising a chimeric P carrying HeV PXD was rescued and compared with parental NiV. Experiments carried out in cellula demonstrated that the replacement of PXD did not significantly affect the replication dynamics while caused a slight virus attenuation, suggesting a possible role of the dimerization of NiV PXD in viral replication.


Assuntos
Vírus Hendra , Vírus Nipah , Proteínas do Nucleocapsídeo , Fosfoproteínas , Proteínas Virais , Replicação Viral , Vírus Hendra/genética , Vírus Hendra/fisiologia , Humanos , Vírus Nipah/genética , Vírus Nipah/fisiologia , Proteínas do Nucleocapsídeo/química , Proteínas do Nucleocapsídeo/genética , Fosfoproteínas/química , Fosfoproteínas/genética , Domínios Proteicos , Dobramento de Proteína , Multimerização Proteica , Espalhamento a Baixo Ângulo , Proteínas Virais/química , Proteínas Virais/genética , Difração de Raios X
7.
Viruses ; 14(2)2022 02 10.
Artigo em Inglês | MEDLINE | ID: mdl-35215961

RESUMO

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.


Assuntos
DNA Viral/biossíntese , DNA Polimerase Dirigida por DNA/metabolismo , Vaccinia virus/enzimologia , Proteínas Virais/metabolismo , Vírus da Mieloblastose Aviária/genética , Vírus da Mieloblastose Aviária/metabolismo , Sequência de Bases , DNA Polimerase Dirigida por DNA/genética , Transcriptase Reversa do HIV/genética , Transcriptase Reversa do HIV/metabolismo , Vírus da Leucemia Murina de Moloney/genética , Vírus da Leucemia Murina de Moloney/metabolismo , Ligação Proteica , Técnica de Seleção de Aptâmeros , Vaccinia virus/genética , Proteínas Virais/genética , Replicação Viral
8.
J Mol Biol ; 433(13): 167009, 2021 06 25.
Artigo em Inglês | MEDLINE | ID: mdl-33901538

RESUMO

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.


Assuntos
DNA Polimerase Dirigida por DNA/química , Domínios Proteicos , Vaccinia virus/enzimologia , Proteínas Virais/química , Sequência de Aminoácidos , Domínio Catalítico/genética , Cristalografia por Raios X , DNA Viral/química , DNA Viral/genética , DNA Viral/metabolismo , DNA Polimerase Dirigida por DNA/genética , DNA Polimerase Dirigida por DNA/metabolismo , Holoenzimas/química , Holoenzimas/genética , Holoenzimas/metabolismo , Modelos Moleculares , Peptídeos/química , Peptídeos/genética , Peptídeos/metabolismo , Ligação Proteica , Homologia de Sequência de Aminoácidos , Soluções/química , Vaccinia virus/genética , Proteínas Virais/genética , Proteínas Virais/metabolismo , Replicação Viral/genética
9.
J Vis Exp ; (167)2021 01 28.
Artigo em Inglês | MEDLINE | ID: mdl-33586708

RESUMO

BioSAXS is a popular technique used in molecular and structural biology to determine the solution structure, particle size and shape, surface-to-volume ratio and conformational changes of macromolecules and macromolecular complexes. A high quality SAXS dataset for structural modeling must be from monodisperse, homogeneous samples and this is often only reached by a combination of inline chromatography and immediate SAXS measurement. Most commonly, size-exclusion chromatography is used to separate samples and exclude contaminants and aggregations from the particle of interest allowing SAXS measurements to be made from a well-resolved chromatographic peak of a single protein species. Still, in some cases, even inline purification is not a guarantee of monodisperse samples, either because multiple components are too close to each other in size or changes in shape induced through binding alter perceived elution time. In these cases, it may be possible to deconvolute the SAXS data of a mixture to obtain the idealized SAXS curves of individual components. Here, we show how this is achieved and the practical analysis of SEC-SAXS data is performed on ideal and difficult samples. Specifically, we show the SEC-SAXS analysis of the vaccinia E9 DNA polymerase exonuclease minus mutant.


Assuntos
Algoritmos , Cromatografia em Gel , Análise de Dados , Espalhamento a Baixo Ângulo , Difração de Raios X , DNA/química , Proteínas/química
10.
Biophys J ; 118(10): 2470-2488, 2020 05 19.
Artigo em Inglês | MEDLINE | ID: mdl-32348724

RESUMO

The structural characterization of modular proteins containing long intrinsically disordered regions intercalated with folded domains is complicated by their conformational diversity and flexibility and requires the integration of multiple experimental approaches. Nipah virus (NiV) phosphoprotein, an essential component of the viral RNA transcription/replication machine and a component of the viral arsenal that hijacks cellular components and counteracts host immune responses, is a prototypical model for such modular proteins. Curiously, the phosphoprotein of NiV is significantly longer than the corresponding protein of other paramyxoviruses. Here, we combine multiple biophysical methods, including x-ray crystallography, NMR spectroscopy, and small angle x-ray scattering, to characterize the structure of this protein and provide an atomistic representation of the full-length protein in the form of a conformational ensemble. We show that full-length NiV phosphoprotein is tetrameric, and we solve the crystal structure of its tetramerization domain. Using NMR spectroscopy and small angle x-ray scattering, we show that the long N-terminal intrinsically disordered region and the linker connecting the tetramerization domain to the C-terminal X domain exchange between multiple conformations while containing short regions of residual secondary structure. Some of these transient helices are known to interact with partners, whereas others represent putative binding sites for yet unidentified proteins. Finally, using NMR spectroscopy and isothermal titration calorimetry, we map a region of the phosphoprotein, comprising residues between 110 and 140 and common to the V and W proteins, that binds with weak affinity to STAT1 and confirm the involvement of key amino acids of the viral protein in this interaction. This provides new, to our knowledge, insights into how the phosphoprotein and the nonstructural V and W proteins of NiV perform their multiple functions.


Assuntos
Vírus Nipah , Fosfoproteínas , Conformação Proteica , Proteínas Virais , Replicação Viral
11.
Nat Commun ; 8(1): 1455, 2017 11 13.
Artigo em Inglês | MEDLINE | ID: mdl-29129932

RESUMO

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.


Assuntos
Domínio Catalítico/genética , DNA Polimerase Dirigida por DNA/ultraestrutura , Vaccinia virus/enzimologia , Cristalografia por Raios X , DNA Glicosilases/genética , Proteínas de Ligação a DNA/genética , DNA Polimerase Dirigida por DNA/genética , Nucleosídeo-Trifosfatase/genética
12.
Acta Crystallogr F Struct Biol Commun ; 72(Pt 9): 687-91, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27599859

RESUMO

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.


Assuntos
DNA Polimerase Dirigida por DNA/química , Mutação Puntual , Uracila-DNA Glicosidase/química , Vaccinia virus/química , Proteínas Virais/química , Motivos de Aminoácidos , Domínio Catalítico , Clonagem Molecular , Cristalização , Cristalografia por Raios X , DNA Polimerase Dirigida por DNA/genética , DNA Polimerase Dirigida por DNA/metabolismo , Expressão Gênica , Modelos Moleculares , Plasmídeos/química , Plasmídeos/metabolismo , Conformação Proteica , Multimerização Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Uracila-DNA Glicosidase/genética , Uracila-DNA Glicosidase/metabolismo , Vaccinia virus/metabolismo , Proteínas Virais/genética , Proteínas Virais/metabolismo , Difração de Raios X
13.
J Virol ; 90(9): 4604-4613, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26912611

RESUMO

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.


Assuntos
DNA Helicases/química , DNA Primase/química , Modelos Moleculares , Domínios e Motivos de Interação entre Proteínas , Vaccinia virus , Proteínas Virais/química , Adenosina Trifosfatases/metabolismo , DNA Helicases/metabolismo , DNA Primase/metabolismo , DNA Viral/metabolismo , Ativação Enzimática , Cinética , Microscopia Eletrônica , Ligação Proteica , Multimerização Proteica , Proteínas Recombinantes de Fusão , Proteínas Virais/metabolismo
14.
J Biol Chem ; 290(29): 17923-17934, 2015 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-26045555

RESUMO

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.


Assuntos
DNA/metabolismo , Uracila-DNA Glicosidase/química , Vaccinia virus/enzimologia , Sequência de Aminoácidos , Cristalografia por Raios X , DNA/química , Humanos , Simulação de Acoplamento Molecular , Dados de Sequência Molecular , Conformação de Ácido Nucleico , Conformação Proteica , Alinhamento de Sequência , Uracila-DNA Glicosidase/metabolismo , Vacínia/virologia , Vaccinia virus/química , Vaccinia virus/metabolismo
15.
Nat Struct Mol Biol ; 21(9): 754-9, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25108352

RESUMO

Nipah virus (NiV) is a highly pathogenic emergent paramyxovirus causing deadly encephalitis in humans. Its replication requires a constant supply of unassembled nucleoprotein (N(0)) in complex with its viral chaperone, the phosphoprotein (P). To elucidate the chaperone function of P, we reconstituted NiV the N(0)-P core complex and determined its crystal structure. The binding of the N-terminal region of P blocks the polymerization of N by interfering with subdomain exchange between N protomers and keeps N(0) in an open conformation, ready to grasp an RNA molecule. We found that a peptide derived from the N-binding region of P protects cells against viral infection and demonstrated by structure-based mutagenesis that this peptide acts by inhibiting N(0)-P formation. These results provide new insights about the assembly of N along genomic RNA and validate the N(0)-P complex as a target for drug development.


Assuntos
Infecções por Henipavirus/virologia , Vírus Nipah/fisiologia , Nucleoproteínas/metabolismo , Fosfoproteínas/metabolismo , Proteínas Virais/metabolismo , Replicação Viral , Sequência de Aminoácidos , Cristalografia por Raios X , Células HEK293 , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Vírus Nipah/química , Nucleoproteínas/química , Fosfoproteínas/química , Ligação Proteica , Conformação Proteica , Proteínas Virais/química
16.
PLoS Pathog ; 10(3): e1003978, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24603707

RESUMO

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.


Assuntos
DNA Polimerase Dirigida por DNA/química , Vaccinia virus/química , Vaccinia virus/enzimologia , Animais , Cromatografia em Gel , Cristalografia , DNA Polimerase Dirigida por DNA/ultraestrutura , Escherichia coli , Holoenzimas/química , Holoenzimas/ultraestrutura , Simulação de Acoplamento Molecular , Subunidades Proteicas/química , Vaccinia virus/ultraestrutura
17.
PLoS Pathog ; 9(9): e1003631, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24086133

RESUMO

Hendra virus (HeV) is a recently emerged severe human pathogen that belongs to the Henipavirus genus within the Paramyxoviridae family. The HeV genome is encapsidated by the nucleoprotein (N) within a helical nucleocapsid. Recruitment of the viral polymerase onto the nucleocapsid template relies on the interaction between the C-terminal domain, N(TAIL), of N and the C-terminal X domain, XD, of the polymerase co-factor phosphoprotein (P). Here, we provide an atomic resolution description of the intrinsically disordered N(TAIL) domain in its isolated state and in intact nucleocapsids using nuclear magnetic resonance (NMR) spectroscopy. Using electron microscopy, we show that HeV nucleocapsids form herringbone-like structures typical of paramyxoviruses. We also report the crystal structure of XD of P that consists of a three-helix bundle. We study the interaction between N(TAIL) and XD using NMR titration experiments and provide a detailed mapping of the reciprocal binding sites. We show that the interaction is accompanied by α-helical folding of the molecular recognition element of N(TAIL) upon binding to a hydrophobic patch on the surface of XD. Finally, using solution NMR, we investigate the interaction between intact nucleocapsids and XD. Our results indicate that monomeric XD binds to N(TAIL) without triggering an additional unwinding of the nucleocapsid template. The present results provide a structural description at the atomic level of the protein-protein interactions required for transcription and replication of HeV, and the first direct observation of the interaction between the X domain of P and intact nucleocapsids in Paramyxoviridae.


Assuntos
Vírus Hendra/química , Proteínas do Nucleocapsídeo/química , Fosfoproteínas/química , Cristalografia por Raios X , Vírus Hendra/genética , Vírus Hendra/metabolismo , Humanos , Espectroscopia de Ressonância Magnética , Microscopia Eletrônica de Transmissão , Proteínas do Nucleocapsídeo/genética , Proteínas do Nucleocapsídeo/metabolismo , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Estrutura Quaternária de Proteína , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína
18.
FEBS Lett ; 587(20): 3335-40, 2013 Oct 11.
Artigo em Inglês | MEDLINE | ID: mdl-24021643

RESUMO

The nucleotide sequence of the unique neutralizing monoclonal antibody D32.10 raised against a conserved conformational epitope shared between E1 and E2 on the serum-derived hepatitis C virus (HCV) envelope was determined. Subsequently, the recombinant single-chain Fv fragment (scFv) was cloned and expressed in Escherichia coli, and its molecular characterization was assessed using multi-angle laser light scattering. The scFv mimicked the antibody in binding to the native serum-derived HCV particles from patients, as well as to envelope E1E2 complexes and E1, E2 glycoproteins carrying the viral epitope. The scFv D32.10 competed with the parental IgG for binding to antigen, and therefore could be a promising candidate for therapeutics and diagnostics.


Assuntos
Anticorpos Monoclonais/química , Hepacivirus/metabolismo , Anticorpos de Cadeia Única/química , Proteínas Virais/imunologia , Anticorpos Monoclonais/metabolismo , Biologia Computacional , Escherichia coli , Anticorpos de Cadeia Única/metabolismo , Proteínas Virais/metabolismo
19.
J Virol ; 87(17): 9569-78, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23785215

RESUMO

Lettuce necrotic yellows virus (LNYV) is a prototype of the plant-adapted cytorhabdoviruses. Through a meta-prediction of disorder, we localized a folded C-terminal domain in the amino acid sequence of its phosphoprotein. This domain consists of an autonomous folding unit that is monomeric in solution. Its structure, solved by X-ray crystallography, reveals a lollipop-shaped structure comprising five helices. The structure is different from that of the corresponding domains of other Rhabdoviridae, Filoviridae, and Paramyxovirinae; only the overall topology of the polypeptide chain seems to be conserved, suggesting that this domain evolved under weak selective pressure and varied in size by the acquisition or loss of functional modules.


Assuntos
Fosfoproteínas/química , Vírus de Plantas/química , Rhabdoviridae/química , Proteínas Virais/química , Sequência de Aminoácidos , Cristalografia por Raios X , Evolução Molecular , Lactuca/virologia , Modelos Moleculares , Dados de Sequência Molecular , Fosfoproteínas/genética , Filogenia , Doenças das Plantas/virologia , Vírus de Plantas/classificação , Vírus de Plantas/genética , Dobramento de Proteína , Estrutura Terciária de Proteína , Rhabdoviridae/classificação , Rhabdoviridae/genética , Proteínas Virais/genética
20.
J Virol ; 87(3): 1679-89, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23175373

RESUMO

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.


Assuntos
Replicação do DNA , Substâncias Macromoleculares/ultraestrutura , Vaccinia virus/fisiologia , Vaccinia virus/ultraestrutura , Replicação Viral , Microscopia Eletrônica , Mapeamento de Interação de Proteínas , Espalhamento a Baixo Ângulo , Proteínas Virais/metabolismo , Proteínas Virais/ultraestrutura
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA