Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 68
Filtrar
Mais filtros










Intervalo de ano de publicação
1.
Front Immunol ; 12: 740223, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34956175

RESUMO

Poxviruses have evolved a variety of innate immunity evasion mechanisms, some of which involve poxvirus-encoded E3 ubiquitin ligases and adaptor proteins. Based on their functional domains and ubiquitin transfer mechanisms, these poxvirus-encoded E3 ubiquitin ligases and adaptor proteins can be divided into five categories: PRANC, ANK/BC, BBK, P28/RING, and MARCH proteins. Although the substrates of many poxvirus E3 ubiquitin ligases remain to be discovered, most of the identified substrates are components of the innate immune system. In this review, we discuss the current research progress on poxvirus-encoded E3 ubiquitin ligases and adaptor proteins to provide mechanistic insights into the interplay between these viruses and their hosts.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/imunologia , Poxviridae/enzimologia , Ubiquitina-Proteína Ligases/imunologia , Proteínas Virais/imunologia , Humanos , Poxviridae/imunologia
2.
Protein Sci ; 25(12): 2113-2131, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27684934

RESUMO

Uracil-DNA glycosylases are ubiquitous enzymes, which play a key role repairing damages in DNA and in maintaining genomic integrity by catalyzing the first step in the base excision repair pathway. Within the superfamily of uracil-DNA glycosylases family I enzymes or UNGs are specific for recognizing and removing uracil from DNA. These enzymes feature conserved structural folds, active site residues and use common motifs for DNA binding, uracil recognition and catalysis. Within this family the enzymes of poxviruses are unique and most remarkable in terms of amino acid sequences, characteristic motifs and more importantly for their novel non-enzymatic function in DNA replication. UNG of vaccinia virus, also known as D4, is the most extensively characterized UNG of the poxvirus family. D4 forms an unusual heterodimeric processivity factor by attaching to a poxvirus-specific protein A20, which also binds to the DNA polymerase E9 and recruits other proteins necessary for replication. D4 is thus integrated in the DNA polymerase complex, and its DNA-binding and DNA scanning abilities couple DNA processivity and DNA base excision repair at the replication fork. The adaptations necessary for taking on the new function are reflected in the amino acid sequence and the three-dimensional structure of D4. An overview of the current state of the knowledge on the structure-function relationship of D4 is provided here.


Assuntos
Poxviridae/enzimologia , Uracila-DNA Glicosidase/metabolismo , Proteínas Virais/metabolismo , Motivos de Aminoácidos , Replicação do DNA/fisiologia , DNA Viral/biossíntese , DNA Viral/genética , Poxviridae/genética , Uracila-DNA Glicosidase/genética , Proteínas Virais/genética
3.
J Biol Chem ; 290(52): 30713-25, 2015 Dec 25.
Artigo em Inglês | MEDLINE | ID: mdl-26559969

RESUMO

In vertebrate species, the innate immune system down-regulates protein translation in response to viral infection through the action of the double-stranded RNA (dsRNA)-activated protein kinase (PKR). In some teleost species another protein kinase, Z-DNA-dependent protein kinase (PKZ), plays a similar role but instead of dsRNA binding domains, PKZ has Zα domains. These domains recognize the left-handed conformer of dsDNA and dsRNA known as Z-DNA/Z-RNA. Cyprinid herpesvirus 3 infects common and koi carp, which have PKZ, and encodes the ORF112 protein that itself bears a Zα domain, a putative competitive inhibitor of PKZ. Here we present the crystal structure of ORF112-Zα in complex with an 18-bp CpG DNA repeat, at 1.5 Å. We demonstrate that the bound DNA is in the left-handed conformation and identify key interactions for the specificity of ORF112. Localization of ORF112 protein in stress granules induced in Cyprinid herpesvirus 3-infected fish cells suggests a functional behavior similar to that of Zα domains of the interferon-regulated, nucleic acid surveillance proteins ADAR1 and DAI.


Assuntos
DNA Forma Z/metabolismo , Proteína Quinase Ativada por DNA/química , Proteína Quinase Ativada por DNA/metabolismo , Doenças dos Peixes/virologia , Vírus de RNA/enzimologia , Proteínas Virais/química , Proteínas Virais/metabolismo , Sequência de Aminoácidos , Animais , Sítios de Ligação , Carpas , Sequência Conservada , DNA Forma Z/química , DNA Forma Z/genética , Proteína Quinase Ativada por DNA/genética , Interferons/genética , Interferons/metabolismo , Modelos Moleculares , Conformação de Ácido Nucleico , Poxviridae/química , Poxviridae/enzimologia , Poxviridae/genética , Ligação Proteica , Estrutura Terciária de Proteína , Vírus de RNA/química , Vírus de RNA/genética , RNA de Cadeia Dupla/química , RNA de Cadeia Dupla/genética , RNA de Cadeia Dupla/metabolismo , Proteínas Virais/genética
4.
Cell Host Microbe ; 12(2): 200-10, 2012 Aug 16.
Artigo em Inglês | MEDLINE | ID: mdl-22901540

RESUMO

The life cycle of several viruses involves host or virally encoded small noncoding RNAs, which play important roles in posttranscriptional regulation. Small noncoding RNAs include microRNAs (miRNAs), which modulate the transcriptome, and small interfering RNAs (siRNAs), which are involved in pathogen defense in plants, worms, and insects. We show that insect and mammalian poxviruses induce the degradation of host miRNAs. The virally encoded poly(A) polymerase, which polyadenylates viral transcripts, also mediates 3' polyadenylation of host miRNAs, resulting in their degradation by the host machinery. In contrast, siRNAs, which are protected by 2'O-methylation (2'OMe), were not targeted by poxviruses. These findings suggest that poxviruses may degrade host miRNAs to promote replication and that virus-mediated small RNA degradation likely contributed to 2'OMe evolution.


Assuntos
MicroRNAs/metabolismo , Polinucleotídeo Adenililtransferase/metabolismo , Infecções por Poxviridae/metabolismo , Poxviridae/enzimologia , Proteínas Virais/metabolismo , Animais , Linhagem Celular , Drosophila , Interações Hospedeiro-Patógeno , Humanos , Metilação , Camundongos , MicroRNAs/química , MicroRNAs/genética , Mariposas , Polinucleotídeo Adenililtransferase/genética , Poxviridae/genética , Infecções por Poxviridae/genética , Infecções por Poxviridae/virologia , Estabilidade de RNA , Proteínas Virais/genética
5.
Biochemistry ; 51(13): 2940-9, 2012 Apr 03.
Artigo em Inglês | MEDLINE | ID: mdl-22417571

RESUMO

A key aspect of the reaction mechanism of type IB topoisomerases is the controlled unwinding of DNA supercoils while the enzyme is transiently bound to one strand of the DNA duplex via a phosphotyrosyl linkage. In this complex, the mobile segment of the bound DNA downstream from the site of cleavage must rotate around the helical axis, requiring that interactions with the enzyme must break and re-form multiple times during the course of removing supercoils. A crystal structure of variola virus type IB topoisomerase (vTopo) bound to DNA shows several positively charged side chains that interact with the downstream mobile and upstream rigid segments, suggesting that these groups may play a role in catalysis, including the processive unwinding of supercoils. We have mutated three such residues, R67, K35, and K271, to Ala and Glu and determined the energetic effects of these mutations at each point along the reaction coordinate of vTopo. R67 interacts with a phosphate group in the rigid DNA segment across from the site of DNA strand cleavage. The ~30-fold damaging effects of the R67A and R67E mutations were primarily on the phosphoryl transfer step, with little effect on enzyme-DNA binding, or the processivity of supercoil unwinding. Removal of the K35 interaction shows mutational effects similar to those of R67, even though this residue interacts with the mobile segment 3 bp from the cleavage site. The two mutations of K271, which interacts with the mobile region even further from the site of covalent linkage, show significant effects not only on phosphoryl transfer but also on downstream DNA strand positioning. Moreover, supercoil unwinding measurements indicate that the K271A and K271E mutations increase the average number of supercoils that are removed during the lifetime of the covalent complex, enhancing the processivity of supercoil unwinding. These measurements support the proposal that the processivity of supercoil unwinding can be regulated by electrostatic interactions between the enzyme and the mobile DNA phosphate backbone.


Assuntos
Mutação , Poxviridae/enzimologia , Inibidores da Topoisomerase/farmacologia , Modelos Moleculares
6.
Nature ; 468(7322): 452-6, 2010 Nov 18.
Artigo em Inglês | MEDLINE | ID: mdl-21085181

RESUMO

Cellular messenger RNA (mRNA) of higher eukaryotes and many viral RNAs are methylated at the N-7 and 2'-O positions of the 5' guanosine cap by specific nuclear and cytoplasmic methyltransferases (MTases), respectively. Whereas N-7 methylation is essential for RNA translation and stability, the function of 2'-O methylation has remained uncertain since its discovery 35 years ago. Here we show that a West Nile virus (WNV) mutant (E218A) that lacks 2'-O MTase activity was attenuated in wild-type primary cells and mice but was pathogenic in the absence of type I interferon (IFN) signalling. 2'-O methylation of viral RNA did not affect IFN induction in WNV-infected fibroblasts but instead modulated the antiviral effects of IFN-induced proteins with tetratricopeptide repeats (IFIT), which are interferon-stimulated genes (ISGs) implicated in regulation of protein translation. Poxvirus and coronavirus mutants that lacked 2'-O MTase activity similarly showed enhanced sensitivity to the antiviral actions of IFN and, specifically, IFIT proteins. Our results demonstrate that the 2'-O methylation of the 5' cap of viral RNA functions to subvert innate host antiviral responses through escape of IFIT-mediated suppression, and suggest an evolutionary explanation for 2'-O methylation of cellular mRNA: to distinguish self from non-self RNA. Differential methylation of cytoplasmic RNA probably serves as an example for pattern recognition and restriction of propagation of foreign viral RNA in host cells.


Assuntos
Proteínas de Transporte/metabolismo , Regulação da Expressão Gênica/imunologia , Imunidade Inata/imunologia , Interferons/imunologia , Proteínas/metabolismo , Capuzes de RNA/metabolismo , RNA Viral/metabolismo , Células 3T3 , Proteínas Adaptadoras de Transdução de Sinal , Animais , Proteínas Reguladoras de Apoptose , Proteínas de Transporte/genética , Células Cultivadas , Coronavirus/enzimologia , Coronavirus/genética , Coronavirus/imunologia , Coronavirus/fisiologia , Fibroblastos , Regulação da Expressão Gênica/genética , Humanos , Imunidade Inata/genética , Interferons/deficiência , Interferons/genética , Metilação , Metiltransferases/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Modelos Genéticos , Modelos Imunológicos , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Poxviridae/enzimologia , Poxviridae/genética , Poxviridae/imunologia , Poxviridae/fisiologia , Biossíntese de Proteínas/imunologia , Proteínas/genética , Capuzes de RNA/genética , Capuzes de RNA/imunologia , RNA Viral/genética , RNA Viral/imunologia , Proteínas de Ligação a RNA , Receptor de Interferon alfa e beta/deficiência , Receptor de Interferon alfa e beta/genética , Taxa de Sobrevida , Replicação Viral , Vírus do Nilo Ocidental/enzimologia , Vírus do Nilo Ocidental/genética , Vírus do Nilo Ocidental/imunologia , Vírus do Nilo Ocidental/fisiologia
7.
Biol Direct ; 4: 51, 2009 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-20021668

RESUMO

BACKGROUND: Eukaryotic Nucleo-Cytoplasmic Large DNA Viruses (NCLDV) encode most if not all of the enzymes involved in their DNA replication. It has been inferred that genes for these enzymes were already present in the last common ancestor of the NCLDV. However, the details of the evolution of these genes that bear on the complexity of the putative ancestral NCLDV and on the evolutionary relationships between viruses and their hosts are not well understood. RESULTS: Phylogenetic analysis of the ATP-dependent and NAD-dependent DNA ligases encoded by the NCLDV reveals an unexpectedly complex evolutionary history. The NAD-dependent ligases are encoded only by a minority of NCLDV (including mimiviruses, some iridoviruses and entomopoxviruses) but phylogenetic analysis clearly indicated that all viral NAD-dependent ligases are monophyletic. Combined with the topology of the NCLDV tree derived by consensus of trees for universally conserved genes suggests that this enzyme was represented in the ancestral NCLDV. Phylogenetic analysis of ATP-dependent ligases that are encoded by chordopoxviruses, most of the phycodnaviruses and Marseillevirus failed to demonstrate monophyly and instead revealed an unexpectedly complex evolutionary trajectory. The ligases of the majority of phycodnaviruses and Marseillevirus seem to have evolved from bacteriophage or bacterial homologs; the ligase of one phycodnavirus, Emiliana huxlei virus, belongs to the eukaryotic DNA ligase I branch; and ligases of chordopoxviruses unequivocally cluster with eukaryotic DNA ligase III. CONCLUSIONS: Examination of phyletic patterns and phylogenetic analysis of DNA ligases of the NCLDV suggest that the common ancestor of the extant NCLDV encoded an NAD-dependent ligase that most likely was acquired from a bacteriophage at the early stages of evolution of eukaryotes. By contrast, ATP-dependent ligases from different prokaryotic and eukaryotic sources displaced the ancestral NAD-dependent ligase at different stages of subsequent evolution. These findings emphasize complex routes of viral evolution that become apparent through detailed phylogenomic analysis but not necessarily in reconstructions based on phyletic patterns of genes. REVIEWERS: This article was reviewed by: Patrick Forterre, George V. Shpakovski, and Igor B. Zhulin.


Assuntos
Evolução Biológica , DNA Ligases/genética , Vírus de DNA/enzimologia , Vírus de DNA/genética , Asfarviridae/classificação , Asfarviridae/enzimologia , Asfarviridae/genética , Núcleo Celular/enzimologia , Citoplasma/virologia , DNA Ligase Dependente de ATP , DNA Ligases/metabolismo , Vírus de DNA/classificação , Eucariotos , Genoma Viral , Iridoviridae/classificação , Iridoviridae/enzimologia , Iridoviridae/genética , Phycodnaviridae/classificação , Phycodnaviridae/enzimologia , Phycodnaviridae/genética , Filogenia , Poxviridae/classificação , Poxviridae/enzimologia , Poxviridae/genética
8.
Proc Natl Acad Sci U S A ; 106(46): 19527-32, 2009 Nov 17.
Artigo em Inglês | MEDLINE | ID: mdl-19887645

RESUMO

The anaphase-promoting complex (APC), or cyclosome, is a ubiquitin ligase with major roles in cell cycle regulation. It is required for mitotic exit, but must be deactivated for the G(1)/S phase transition to occur. APC consists of at least 12 subunits with the catalytic core formed by a scaffold protein, APC2, and a RING-H2 protein, APC11. APC11 facilitates ubiquitin chain formation by recruiting ubiquitin-charged conjugating enzymes through its RING-H2 domain. We report that a small number of poxviruses encode RING-H2 proteins with sequence similarities to APC11. We show that a representative of these viral proteins mimics APC11 in its interactions with APC, but unlike APC11, the viral protein fails to promote ubiquitin chain formation. This absence of ubiquitin ligase activity is linked to a distinctive sequence variation within its RING-H2 domain. Expression of the viral protein led to cell cycle deregulation and the accumulation of APC substrates in a manner consistent with impaired APC function. Our data characterize this protein as a regulator of APC activity, and consequently, we have called it PACR (poxvirus APC/cyclosome regulator). Deletion of the PACR gene substantially reduced viral replication. Here, we report a viral mimic of an APC component and reveal an intriguing mechanism by which viruses can manipulate cell cycle progression and, thereby, promote their own replication.


Assuntos
Ciclo Celular , Poxviridae/fisiologia , Complexos Ubiquitina-Proteína Ligase/metabolismo , Proteínas Virais/metabolismo , Replicação Viral , Sequência de Aminoácidos , Ciclossomo-Complexo Promotor de Anáfase , Subunidade Apc11 do Ciclossomo-Complexo Promotor de Anáfase , Linhagem Celular , Deleção de Genes , Humanos , Dados de Sequência Molecular , Poxviridae/enzimologia , Poxviridae/genética , Complexos Ubiquitina-Proteína Ligase/genética , Proteínas Virais/genética
9.
Proc Natl Acad Sci U S A ; 104(47): 18724-9, 2007 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-18000036

RESUMO

Poxviruses are large enveloped viruses that replicate in the cytoplasm of vertebrate or invertebrate cells. At least six virus-encoded proteins are required for synthesis and processing of the double-stranded DNA genome of vaccinia virus, the prototype member of the family. One of these proteins, D5, is an NTPase that contains an N-terminal archaeoeukaryotic primase domain and a C-terminal superfamily III helicase domain. Here we report that individual conserved aspartic acid residues in the predicted primase active site were required for in vivo complementation of infectious virus formation as well as genome and plasmid replication. Furthermore, purified recombinant D5 protein synthesized oligoribonucleotides in vitro. Incorporation of label from [alpha-(32)P]CTP or [alpha-(32)P]UTP into a RNase-sensitive and DNase-resistant product was demonstrated by using single-stranded circular bacteriophage DNA templates and depended on ATP or GTP and a divalent cation. Mutagenesis studies showed that the primase and NTPase activities of the recombinant D5 protein could be independently inactivated. Highly conserved orthologs of D5 are present in all poxviruses that have been sequenced, and more diverged orthologs are found in members of all other families of nucleocytoplasmic large DNA viruses. These viral primases may have roles in initiation of DNA replication or lagging-strand synthesis and represent potential therapeutic targets.


Assuntos
DNA Primase/metabolismo , Poxviridae/enzimologia , Hidrolases Anidrido Ácido/metabolismo , Aminoácidos/genética , Aminoácidos/metabolismo , Sítios de Ligação , Catálise , DNA Primase/genética , Vetores Genéticos/genética , Genoma Viral/genética , Células HeLa , Humanos , Mutação/genética , Oligonucleotídeos/metabolismo , Plasmídeos/genética , Poxviridae/genética
10.
J Comput Aided Mol Des ; 21(10-11): 549-58, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17960327

RESUMO

Essential for viral replication and highly conserved among poxviridae, the vaccinia virus I7L ubiquitin-like proteinase (ULP) is an attractive target for development of smallpox antiviral drugs. At the same time, the I7L proteinase exemplifies several interesting challenges from the rational drug design perspective. In the absence of a published I7L X-ray structure, we have built a detailed 3D model of the I7L ligand binding site (S2-S2' pocket) based on exceptionally high structural conservation of this site in proteases of the ULP family. The accuracy and limitations of this model were assessed through comparative analysis of available X-ray structures of ULPs, as well as energy based conformational modeling. The 3D model of the I7L ligand binding site was used to perform covalent docking and VLS of a comprehensive library of about 230,000 available ketone and aldehyde compounds. Out of 456 predicted ligands, 97 inhibitors of I7L proteinase activity were confirmed in biochemical assays ( approximately 20% overall hit rate). These experimental results both validate our I7L ligand binding model and provide initial leads for rational optimization of poxvirus I7L proteinase inhibitors. Thus, fragments predicted to bind in the prime portion of the active site can be combined with fragments on non-prime side to yield compounds with improved activity and specificity.


Assuntos
Antivirais/química , Antivirais/farmacologia , Cisteína Endopeptidases/química , Inibidores de Cisteína Proteinase/química , Inibidores de Cisteína Proteinase/farmacologia , Ubiquitinas/antagonistas & inibidores , Vaccinia virus/enzimologia , Sequência de Aminoácidos , Sítios de Ligação , Simulação por Computador , Cisteína Endopeptidases/genética , Desenho de Fármacos , Avaliação Pré-Clínica de Medicamentos , Cetonas/química , Ligantes , Modelos Moleculares , Dados de Sequência Molecular , Poxviridae/efeitos dos fármacos , Poxviridae/enzimologia , Poxviridae/genética , Homologia de Sequência de Aminoácidos , Relação Estrutura-Atividade , Especificidade por Substrato , Ubiquitinas/química , Ubiquitinas/genética , Interface Usuário-Computador , Vaccinia virus/efeitos dos fármacos , Vaccinia virus/genética
11.
J Virol ; 80(15): 7706-13, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16840349

RESUMO

The possibility of the release of smallpox virus into a predominantly nonimmunized population highlights the importance of understanding poxvirus biology. Poxviruses encode a conserved pathway that is required to oxidize disulfide bonds in nascent viral proteins that fold in the reducing environment of the eukaryotic host cytoplasm. We present the structure of the last enzyme of the vaccinia virus pathway, G4, which is almost identical in smallpox virus. G4 catalyzes the formation of disulfide bonds in proteins that are critical for virus maturation and host cell infection. G4 contains a thioredoxin fold and a Cys-X-X-Cys active site. In solution, G4 monomers and dimers are observed. In the crystal, G4 is found as a dimer that buries 4,500 A(2) in the interface and occludes the active site, which could protect the reactive disulfide from reduction in the cytoplasm. The structure serves as a model for drug design targeting viral disulfide bond formation.


Assuntos
Dissulfetos/metabolismo , Oxirredutases/química , Poxviridae/enzimologia , Vírion/química , Sequência de Aminoácidos , Sítios de Ligação , Cristalografia por Raios X , Cisteína/química , Cisteína/metabolismo , Citoplasma , Dimerização , Modelos Moleculares , Dados de Sequência Molecular , Oxirredução , Oxirredutases/metabolismo , Poxviridae/genética , Conformação Proteica , Dobramento de Proteína , Homologia de Sequência de Aminoácidos , Tiorredoxinas/química , Vírion/genética , Vírion/metabolismo
12.
J Biol Chem ; 281(28): 18953-60, 2006 Jul 14.
Artigo em Inglês | MEDLINE | ID: mdl-16707499

RESUMO

The guanine-N7 methyltransferase domain of vaccinia virus mRNA capping enzyme is a heterodimer composed of a catalytic subunit vD1-(540-844) and a stimulatory subunit vD12. The poxvirus enzyme can function in vivo in Saccharomyces cerevisiae in lieu of the essential cellular cap methyltransferase Abd1. Coexpression of both poxvirus subunits is required to complement the growth of abd1delta cells. We performed a genetic screen for mutations in the catalytic subunit that bypassed the requirement for the stimulatory subunit in vivo. We thereby identified missense changes in vicinal residues Tyr-752 (to Ser, Cys, or His) and Asn-753 (to Ile), which are located in the cap guanine-binding pocket. Biochemical experiments illuminated a mechanism of intersubunit allostery, whereby the vD12 subunit enhances the affinity of the catalytic subunit for AdoMet and the cap guanine methyl acceptor by 6- and 14-fold, respectively, and increases kcat by a factor of 4. The bypass mutations elicited gains of function in both vD12-independent and vD12-dependent catalysis of cap methylation in vitro when compared with wild-type vD1-(540-844). These results highlight the power of yeast as a surrogate model for the genetic analysis of interacting poxvirus proteins and demonstrate that the activity of an RNA processing enzyme can be augmented through selection and protein engineering.


Assuntos
Metiltransferases/genética , Metiltransferases/fisiologia , Mutação , Poxviridae/enzimologia , RNA Mensageiro/metabolismo , Alelos , Sítio Alostérico , Domínio Catalítico , Dimerização , Guanina/química , Cinética , Ligação Proteica , Engenharia de Proteínas , RNA/química , Saccharomyces cerevisiae/metabolismo , Vaccinia virus/genética
13.
J Biol Chem ; 281(17): 11618-26, 2006 Apr 28.
Artigo em Inglês | MEDLINE | ID: mdl-16513635

RESUMO

Recently, poxviruses were found to encode a protein with signature motifs present in the RuvC family of Holliday junction (HJ) resolvases, which have a key role in homologous recombination in bacteria. The vaccinia virus homolog A22 specifically cleaved synthetic HJ DNA in vitro and was required for the in vivo resolution of viral DNA concatemers into unit-length genomes with hairpin telomeres. It was of interest to further characterize a poxvirus resolvase in view of the low sequence similarity with RuvC, the absence of virus-encoded RuvA and RuvB to interact with, and the different functions of the viral and bacterial resolvases. Because purified A22 aggregated severely, studies were carried out with maltose-binding protein fused to A22 as well as to RuvC. Using gel filtration, chemical cross-linking, analytical ultracentrifugation, and light scattering, we demonstrated that A22 and RuvC are homodimers in solution. Furthermore, the dimeric form of the resolvase associated with HJ DNA, presumably facilitating the symmetrical cleavage of such structures. Like RuvC, A22 symmetrically cleaved fixed HJ junctions as well as junctions allowing strand mobility. Unlike RuvC and other members of the family, however, the poxvirus enzyme exhibited little cleavage sequence specificity. Structural and enzymatic similarities of poxvirus, bacterial, and fungal mitochondrial HJ resolvases are consistent with their predicted evolutionary relationship based on sequence analysis. The absence of a homologous resolvase in mammalian cells makes these microbial enzymes excellent potential therapeutic targets.


Assuntos
DNA Cruciforme/metabolismo , Resolvases de Junção Holliday/metabolismo , Poxviridae/enzimologia , Estrutura Quaternária de Proteína , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Primers do DNA/química , DNA Cruciforme/química , DNA Cruciforme/genética , Dimerização , Resolvases de Junção Holliday/genética , Resolvases de Junção Holliday/isolamento & purificação , Proteínas Ligantes de Maltose , Modelos Moleculares , Conformação de Ácido Nucleico , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Relação Estrutura-Atividade , Especificidade por Substrato
14.
J Gen Virol ; 85(Pt 10): 2741-2754, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15448335

RESUMO

Studies of cellular biology in recent decades have highlighted the crucial roles of glycans in numerous important biological processes, raising the concept of glycomics that is now considered as important as genomics, transcriptomics and proteomics. For millions of years, viruses have been co-evolving with their hosts. Consequently, during this co-evolution process, viruses have acquired mechanisms to mimic, hijack or sabotage host processes that favour their replication, including mechanisms to modify the glycome. The importance of the glycome in the regulation of host-virus interactions has recently led to a new concept called 'glycovirology'. One fascinating aspect of glycovirology is the study of how viruses affect the glycome. Viruses reach that goal either by regulating expression of host glycosyltransferases or by expressing their own glycosyltransferases. This review describes all virally encoded glycosyltransferases and discusses their established or putative functions. The description of these enzymes illustrates several intriguing aspects of virology and provides further support for the importance of glycomics in biological processes.


Assuntos
Glicosiltransferases/fisiologia , Vírus/enzimologia , Bacteriófagos/enzimologia , Baculoviridae/enzimologia , Glicosiltransferases/genética , Herpesvirus Bovino 4/enzimologia , Phycodnaviridae/enzimologia , Poxviridae/enzimologia
15.
Methods Mol Biol ; 269: 169-86, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15114016

RESUMO

Cytoplasmic replication of poxviruses dictates the encoding of most, if not all, of the trans-acting factors required for faithful genome duplication. Several of these proteins have been identified through genetic and biochemical evaluation, including the catalytic DNA polymerase (E9), an essential and stoichiometric component of the processive polymerase (A20), a single-strand DNA-binding protein (I3), a type I topoisomerase (H6), the uracil DNA glycosylase (D4), a nucleic acid-independent nucleoside triphosphatase (D5), a serine/threonine protein kinase (B1), and a Holliday Junction resolvase (A22). All of these factors work in concert to faithfully duplicate the viral genome. Although a replication origin has not been defined for the poxviruses, cis-acting sequences found within the telomeric 200 bp have been implicated as necessary and sufficient for minichromosome replication. Replication occurs within cytoplasmic foci from approx 3 to 12 h postinfection. This chapter includes several methodologies to assay and quantitate replication in vivo, visualize replication foci microscopically, and test the integrity of central replication enzymes in vitro.


Assuntos
Replicação do DNA , DNA Viral/análise , Poxviridae/fisiologia , Linhagem Celular , DNA Glicosilases/metabolismo , DNA Viral/biossíntese , Poxviridae/enzimologia , Poxviridae/patogenicidade , Vaccinia virus/genética , Proteínas Virais/genética , Proteínas Virais/metabolismo
16.
Proc Natl Acad Sci U S A ; 100(20): 11291-6, 2003 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-12972642

RESUMO

Vaccinia virus encodes a type I DNA topoisomerase that is highly conserved in all known poxviruses. Although the structure and catalytic activity of the enzyme were well studied, little was known about its biological function. The viral topoisomerase was thought to be essential, and roles in DNA replication, recombination, concatemer resolution, and transcription were suggested. Here, we demonstrated that the topoisomerase is not essential for replication of vaccinia virus in cultured cells, although deletion mutants formed fewer and smaller plaques on cell monolayers than wild-type virus. Purified mutant virus particles were able to bind and enter cells but exhibited reduced viral early transcription and a delay in DNA replication. Infecting with a high number of virus particles increased early mRNA and accelerated viral DNA synthesis. Processing of viral DNA concatemers into unit-length genomes was unimpaired at either a low or high multiplicity of infection. The data suggest that the primary, perhaps only, role of the poxvirus topoisomerase is to increase early transcription, which takes place within virus cores in the cytoplasm of infected cells. Because the topoisomerase functions early in infection, drugs capable of penetrating the virus core and irreversibly damaging DNA by trapping nicked DNA-topoisomerase intermediates could make potent antiviral agents.


Assuntos
DNA Topoisomerases Tipo I/fisiologia , Mutação , Poxviridae/enzimologia , Transcrição Gênica , DNA Topoisomerases Tipo I/genética , DNA Viral/biossíntese , Células HeLa , Humanos , Microscopia Eletrônica , Microscopia de Fluorescência , Poxviridae/genética , Poxviridae/patogenicidade
17.
J Virol ; 77(13): 7300-7, 2003 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12805428

RESUMO

Structural differences between poxvirus and human mRNA capping enzymes recommend cap formation as a target for antipoxviral drug discovery. Genetic and pharmacologic analysis of the poxvirus capping enzymes requires in vivo assays in which the readout depends on the capacity of the viral enzyme to catalyze cap synthesis. Here we have used the budding yeast Saccharomyces cerevisiae as a genetic model for the study of poxvirus cap guanine-N7 methyltransferase. The S. cerevisiae capping system consists of separate triphosphatase (Cet1), guanylyltransferase (Ceg1), and methyltransferase (Abd1) components. All three activities are essential for cell growth. We report that the methyltransferase domain of vaccinia virus capping enzyme (composed of catalytic vD1-C and stimulatory vD12 subunits) can function in lieu of yeast Abd1. Coexpression of both vaccinia virus subunits is required for complementation of the growth of abd1Delta cells. Previously described mutations of vD1-C and vD12 that eliminate or reduce methyltransferase activity in vitro either abolish abd1Delta complementation or elicit conditional growth defects. We have used the yeast complementation assay as the primary screen in a new round of alanine scanning of the catalytic subunit. We thereby identified several new amino acids that are critical for cap methylation activity in vivo. Studies of recombinant proteins show that the lethal vD1-C mutations do not preclude heterodimerization with vD12 but either eliminate or reduce cap methyltransferase activity in vitro.


Assuntos
Metiltransferases/metabolismo , Poxviridae/enzimologia , Capuzes de RNA , Saccharomyces cerevisiae/genética , Sequência de Aminoácidos , Teste de Complementação Genética , Metiltransferases/genética , Dados de Sequência Molecular , Plasmídeos , Homologia de Sequência de Aminoácidos , Proteínas Virais/genética , Proteínas Virais/metabolismo
18.
J Virol ; 77(2): 1427-40, 2003 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-12502858

RESUMO

The genomes of several poxviruses contain open reading frames with homology to the K3 and K5 genes of Kaposi's sarcoma-associated herpesvirus (KSHV) and the K3 gene of murine gammaherpesvirus 68, which target major histocompatibility complex class I (MHC-I) as well as costimulatory molecules for proteasomal or lysosomal degradation. The homologous gene product of myxomavirus (MV), M153R, was recently shown to reduce the cell surface expression of MHC-I. In addition, normal MHC-I surface expression was observed in cells infected with MV lacking M153R (J. L. Guerin, J. Gelfi, S. Boullier, M. Delverdier, F. A. Bellanger, S. Bertagnoli, I. Drexler, G. Sutter, and F. Messud-Petit, J. Virol. 76:2912-2923, 2002). Here, we show that M153R also downregulates the T-cell coreceptor CD4 and we study the molecular mechanism by which M153R achieves the downregulation of CD4 and MHC-I. Upon M153R expression, CD4 was rapidly internalized and degraded in lysosomes, whereas deletion of M153R from the genome of MV restored CD4 expression. The downregulation of both CD4 and MHC-I was dependent on the presence of lysine residues in their cytoplasmic tails. Increased ubiquitination of CD4 was observed upon coexpression with M153R in the presence of inhibitors of lysosomal acidification. Surface expression of CD4 was restored upon overexpression of Hrs, a ubiquitin interaction motif-containing protein that sorts ubiquitinated proteins into endosomes. Moreover, the purified PHD/LAP zinc finger of M153R catalyzed the formation of multiubiquitin adducts in vitro. Our data suggest that M153R acts as a membrane-bound ubiquitin ligase that conjugates ubiquitin to the cytoplasmic domain of substrate glycoproteins, with ubiquitin serving as a lysosomal targeting signal. Since a similar mechanism was recently proposed for KSHV K5, it seems that members of the unrelated families of gamma-2 herpesviruses and poxviruses share a common immune evasion mechanism that targets host cell immune receptors.


Assuntos
Antígenos CD4/metabolismo , Endocitose , Ligases/metabolismo , Poxviridae/enzimologia , Ubiquitina/metabolismo , Sequência de Aminoácidos , Sequência de Bases , Linhagem Celular , DNA Viral , Regulação para Baixo , Endossomos/metabolismo , Complexo de Golgi/metabolismo , Humanos , Dados de Sequência Molecular
19.
J Gen Virol ; 83(Pt 10): 2339-2345, 2002 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-12237414

RESUMO

Herpesviruses and poxviruses are known to encode the DNA repair enzyme uracil-DNA glycosylase (UNG), an enzyme involved in the base excision repair pathway that specifically removes the RNA base uracil from DNA, while at least one retrovirus (human immunodeficiency virus type 1) packages cellular UNG into virus particles. In these instances, UNG is implicated as being important in virus replication. However, a clear understanding of the role(s) of UNG in virus replication remains elusive. Herpesviruses, poxviruses and some retroviruses encode dUTPase, an enzyme that can minimize the misincorporation of uracil into DNA. The encoding of dUTPase by these viruses also implies their importance in virus replication. An understanding at the molecular level of how these viruses replicate in non-dividing cells should provide clues to the biological relevance of UNG and dUTPase function in virus replication.


Assuntos
DNA Glicosilases , Replicação do DNA/fisiologia , N-Glicosil Hidrolases/metabolismo , Pirofosfatases/metabolismo , Replicação Viral/fisiologia , Animais , Produtos do Gene vpr/genética , Produtos do Gene vpr/fisiologia , HIV-1/genética , HIV-1/fisiologia , Herpesviridae/enzimologia , Herpesviridae/genética , Herpesviridae/fisiologia , Humanos , Lentivirus/enzimologia , Lentivirus/genética , Lentivirus/fisiologia , Poxviridae/enzimologia , Poxviridae/genética , Poxviridae/fisiologia , Uracila-DNA Glicosidase , Produtos do Gene vpr do Vírus da Imunodeficiência Humana
20.
Proc Natl Acad Sci U S A ; 99(4): 1853-8, 2002 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-11830640

RESUMO

We report that diverse species of bacteria encode a type IB DNA topoisomerase that resembles vaccinia virus topoisomerase. Deinococcus radiodurans topoisomerase IB (DraTopIB), an exemplary member of this family, relaxes supercoiled DNA in the absence of a divalent cation or ATP. DraTopIB has a compact size (346 aa) and is a monomer in solution. Mutational analysis shows that the active site of DraTopIB is composed of the same constellation of catalytic side chains as the vaccinia enzyme. Sequence comparisons and limited proteolysis suggest that their folds are conserved. These findings imply an intimate evolutionary relationship between the poxvirus and bacterial type IB enzymes, and they engender a scheme for the evolution of topoisomerase IB and tyrosine recombinases from a common ancestral strand transferase in the bacterial domain. Remarkably, bacteria that possess topoisomerase IB appear to lack DNA topoisomerase III.


Assuntos
Proteínas de Bactérias/química , DNA Topoisomerases Tipo I/química , Poxviridae/enzimologia , Motivos de Aminoácidos , Sequência de Aminoácidos , Sítios de Ligação , Domínio Catalítico , Centrifugação com Gradiente de Concentração , Clonagem Molecular , DNA/metabolismo , Análise Mutacional de DNA , DNA Topoisomerases Tipo I/metabolismo , DNA Super-Helicoidal/metabolismo , Glicerol/química , Dados de Sequência Molecular , Família Multigênica , Mutagênese Sítio-Dirigida , Conformação de Ácido Nucleico , Ligação Proteica , Estrutura Terciária de Proteína , Proteínas Recombinantes/metabolismo , Homologia de Sequência de Aminoácidos , Fatores de Tempo , Tirosina/química , Tirosina/metabolismo , Vaccinia virus/enzimologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...