Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
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
2.
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
3.
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
4.
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
5.
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
6.
Viral Immunol ; 25(6): 461-70, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23061794

RESUMO

Epstein-Barr virus (EBV), like many other persistent herpes viruses, has acquired numerous mechanisms for subverting or evading immune surveillance. This study investigates the role of secreted EBV-encoded BARF1 protein (sBARF1) in creating an immune evasive microenvironment. Wild-type consensus BARF1 was expressed in the human 293 cell line and purified. This native hexameric sBARF1 had inhibitory capacity on macrophage colony stimulating factor (M-CSF)-stimulated, and not on granulocyte macrophage-colony stimulating factor (GM-CSF)-stimulated growth and differentiation of myeloid cells. Antibodies specific to hexameric sBARF1 were able to block this effect. M-CSF was shown to interact with sBARF1 via the protruding N-terminal loops involving Val38 and Ala84. Each BARF1 hexamer was capable of binding three M-CSF dimers. Mutations in the BARF1 loops greatly affected M-CSF interaction, and showed loss of growth inhibition. Analysis of the activation state of the M-CSF receptor c-fms and its downstream kinase pathways showed that sBARF1 prevented M-CSF-induced downstream phosphorylation. Since M-CSF is an important factor in macrophage differentiation, the effect of sBARF1 on the function of monocyte-derived macrophages was evaluated. sBARF1 affected overall survival and morphology and significantly reduced expression of macrophage differentiation surface markers such as CD14, CD11b, CD16, and CD169. Macrophages differentiating in the presence of sBARF1 showed impaired responses to lipopolysaccharide and decreased oxygen radical formation as well as reduced phagocytosis of apoptotic cells. In conclusion, EBV sBARF1 protein is a potent decoy receptor for M-CSF, hampering the function and differentiation of macrophages. These results suggest that sBARF1 contributes to the modulation of immune responses in the microenvironment of EBV-positive carcinomas.


Assuntos
Herpesvirus Humano 4/patogenicidade , Interações Hospedeiro-Patógeno , Evasão da Resposta Imune , Fator Estimulador de Colônias de Macrófagos/antagonistas & inibidores , Macrófagos/imunologia , Proteínas Virais/metabolismo , Fatores de Virulência/metabolismo , Diferenciação Celular , Linhagem Celular , Proliferação de Células , Análise Mutacional de DNA , Herpesvirus Humano 4/imunologia , Humanos , Fator Estimulador de Colônias de Macrófagos/imunologia , Macrófagos/fisiologia , Macrófagos/virologia , Proteínas Mutantes/genética , Proteínas Mutantes/imunologia , Proteínas Mutantes/metabolismo , Ligação Proteica , Mapeamento de Interação de Proteínas , Transdução de Sinais , Proteínas Virais/genética , Proteínas Virais/imunologia , Fatores de Virulência/genética , Fatores de Virulência/imunologia
7.
Nat Struct Mol Biol ; 19(9): 938-47, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22902366

RESUMO

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.


Assuntos
Infecções por Vírus Epstein-Barr/metabolismo , Herpesvirus Humano 4/fisiologia , Interações Hospedeiro-Patógeno , Fator Estimulador de Colônias de Macrófagos/metabolismo , Monócitos/virologia , Proteínas Virais/metabolismo , Sequência de Aminoácidos , Animais , Sítios de Ligação , Células Cultivadas , Cristalografia por Raios X , Herpesvirus Humano 4/metabolismo , Humanos , Fator Estimulador de Colônias de Macrófagos/química , Camundongos , Camundongos Endogâmicos C57BL , Modelos Moleculares , Dados de Sequência Molecular , Monócitos/citologia , Ligação Proteica , Conformação Proteica , Multimerização Proteica , Transdução de Sinais , Proteínas Virais/química
8.
PLoS Pathog ; 7(9): e1002248, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21960769

RESUMO

Replication of non-segmented negative-strand RNA viruses requires the continuous supply of the nucleoprotein (N) in the form of a complex with the phosphoprotein (P). Here, we present the structural characterization of a soluble, heterodimeric complex between a variant of vesicular stomatitis virus N lacking its 21 N-terminal residues (N(Δ21)) and a peptide of 60 amino acids (P(60)) encompassing the molecular recognition element (MoRE) of P that binds RNA-free N (N(0)). The complex crystallized in a decameric circular form, which was solved at 3.0 Å resolution, reveals how the MoRE folds upon binding to N and competes with RNA binding and N polymerization. Small-angle X-ray scattering experiment and NMR spectroscopy on the soluble complex confirms the binding of the MoRE and indicates that its flanking regions remain flexible in the complex. The structure of this complex also suggests a mechanism for the initiation of viral RNA synthesis.


Assuntos
Complexos Multiproteicos/química , Proteínas do Nucleocapsídeo/química , Fosfoproteínas/química , Vesiculovirus/química , Proteínas Estruturais Virais/química , Cristalografia por Raios X , Complexos Multiproteicos/genética , Complexos Multiproteicos/metabolismo , Ressonância Magnética Nuclear Biomolecular , Proteínas do Nucleocapsídeo/genética , Proteínas do Nucleocapsídeo/metabolismo , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Ligação Proteica , Estrutura Quaternária de Proteína , RNA Viral/biossíntese , RNA Viral/química , RNA Viral/genética , Sequências Reguladoras de Ácido Ribonucleico/fisiologia , Vesiculovirus/genética , Vesiculovirus/metabolismo , Proteínas Estruturais Virais/genética , Proteínas Estruturais Virais/metabolismo
9.
J Biol Chem ; 284(37): 25280-9, 2009 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-19586911

RESUMO

Deoxyuridine 5'-triphosphate pyrophosphatases (dUTPases) are ubiquitous enzymes essential for hydrolysis of dUTP, thus preventing its incorporation into DNA. Although Epstein-Barr virus (EBV) dUTPase is monomeric, it has a high degree of similarity with the more frequent trimeric form of the enzyme. In both cases, the active site is composed of five conserved sequence motifs. Structural and functional studies of mutants based on the structure of EBV dUTPase gave new insight into the mechanism of the enzyme. A first mutant allowed us to exclude a role in enzymatic activity for the disulfide bridge involving the beginning of the disordered C terminus. Sequence alignments revealed two groups of dUTPases, based on the position in sequence of a conserved aspartic acid residue close to the active site. Single mutants of this residue in EBV dUTPase showed a highly impaired catalytic activity, which could be partially restored by a second mutation, making EBV dUTPase more similar to the second group of enzymes. Deletion of the flexible C-terminal tail carrying motif V resulted in a protein completely devoid of enzymatic activity, crystallizing with unhydrolyzed Mg(2+)-dUTP complex in the active site. Point mutations inside motif V highlighted the essential role of lid residue Phe(273). Magnesium appears to play a role mainly in substrate binding, since in absence of Mg(2+), the K(m) of the enzyme is reduced, whereas the k(cat) is less affected.


Assuntos
Herpesvirus Humano 4/enzimologia , Pirofosfatases/química , Motivos de Aminoácidos , Sequência de Aminoácidos , Catálise , Domínio Catalítico , Cristalização , Cristalografia por Raios X/métodos , Cinética , Magnésio/química , Dados de Sequência Molecular , Mutação , Estrutura Terciária de Proteína , Homologia de Sequência de Aminoácidos , Especificidade por Substrato
10.
J Mol Biol ; 391(4): 717-28, 2009 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-19538972

RESUMO

Epstein-Barr virus, a double-stranded DNA (dsDNA) virus, is a major human pathogen from the herpesvirus family. The nuclease is one of the lytic cycle proteins required for successful viral replication. In addition to the previously described endonuclease and exonuclease activities on single-stranded DNA and dsDNA substrates, we observed an RNase activity for Epstein-Barr virus nuclease in the presence of Mn(2+), giving a possible explanation for its role in host mRNA degradation. Its crystal structure shows a catalytic core of the D-(D/E)XK nuclease superfamily closely related to the exonuclease from bacteriophage lambda with a bridge across the active-site canyon. This bridge may reduce endonuclease activity, ensure processivity or play a role in strand separation of dsDNA substrates. As the DNA strand that is subject to cleavage is likely to make a sharp turn in front of the bridge, endonuclease activity on single-stranded DNA stretches appears to be possible, explaining the cleavage of circular substrates.


Assuntos
Desoxirribonucleases , Endonucleases , Exonucleases , Herpesvirus Humano 4/enzimologia , Conformação Proteica , Ribonucleases , Proteínas Virais , Sequência de Aminoácidos , Animais , Domínio Catalítico , Cristalografia por Raios X , DNA/química , DNA/metabolismo , Desoxirribonucleases/química , Desoxirribonucleases/genética , Desoxirribonucleases/metabolismo , Endonucleases/química , Endonucleases/genética , Endonucleases/metabolismo , Exonucleases/química , Exonucleases/genética , Exonucleases/metabolismo , Herpesvirus Humano 4/patogenicidade , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Estrutura Molecular , Ribonucleases/química , Ribonucleases/genética , Ribonucleases/metabolismo , Alinhamento de Sequência , Proteínas Virais/química , Proteínas Virais/genética , Proteínas Virais/metabolismo
11.
J Mol Biol ; 366(1): 117-31, 2007 Feb 09.
Artigo em Inglês | MEDLINE | ID: mdl-17157317

RESUMO

Epstein-Barr virus (EBV) is a human gamma-herpesvirus. Within its 86 open reading frame containing genome, two enzymes avoiding uracil incorporation into DNA can be found: uracil triphosphate hydrolase and uracil-DNA glycosylase (UNG). The latter one excises uracil bases that are due to cytosine deamination or uracil misincorporation from double-stranded DNA substrates. The EBV enzyme belongs to family 1 UNGs. We solved the three-dimensional structure of EBV UNG in complex with the uracil-DNA glycosylase inhibitor protein (Ugi) from bacteriophage PBS-2 at a resolution of 2.3 A by X-ray crystallography. The structure of EBV UNG encoded by the BKRF3 reading frame shows the excellent global structural conservation within the solved examples of family 1 enzymes. Four out of the five catalytic motifs are completely conserved, whereas the fifth one, the leucine loop, carries a seven residue insertion. Despite this insertion, catalytic constants of EBV UNG are similar to those of other UNGs. Modelling of the EBV UNG-DNA complex shows that the longer leucine loop still contacts DNA and is likely to fulfil its role of DNA binding and deformation differently than the enzymes with previously solved structures. We could show that despite the evolutionary distance of EBV UNG from the natural host protein, bacteriophage Ugi binds with an inhibitory constant of 8 nM to UNG. This is due to an excellent specificity of Ugi for conserved elements of UNG, four of them corresponding to catalytic motifs and a fifth one corresponding to an important beta-turn structuring the catalytic site.


Assuntos
Gammaherpesvirinae/enzimologia , Herpesvirus Humano 4/enzimologia , Uracila-DNA Glicosidase/química , Proteínas Virais/química , Motivos de Aminoácidos , Sequência de Aminoácidos , Sequência de Bases , Gammaherpesvirinae/química , Herpesvirus Humano 4/química , Leucina/química , Modelos Moleculares , Dados de Sequência Molecular , Estrutura Molecular , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Homologia de Sequência de Aminoácidos , Relação Estrutura-Atividade
12.
Acta Crystallogr D Biol Crystallogr ; 62(Pt 10): 1276-85, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17001105

RESUMO

Epstein-Barr virus is a herpesvirus that causes infectious mononucleosis, carcinomas and immunoproliferative disease. Its genome encodes 86 proteins, which provided targets for a structural genomics project. After updating the annotation of the genome, 23 open reading frames were chosen for expression in Escherichia coli, initially selecting for those with known enzyme activity and then supplementing this set based on a series of predicted properties, in particular secondary structure. The major obstacle turned out to be poor expression and low solubility. Surprisingly, this could not be overcome by modifications of the constructs, changes of expression temperature or strain or renaturation. Of the eight soluble proteins, five were crystallized using robotic nanolitre-drop crystallization trials, which led to four solved structures. Although these results depended on individual treatment rather than standardized protocols, a high-throughput miniaturized crystallization screening protocol was a key component of success with these difficult proteins.


Assuntos
Herpesvirus Humano 4/genética , Proteínas Virais/genética , Clonagem Molecular , Cristalização , DNA Viral/biossíntese , DNA Viral/genética , Regulação Viral da Expressão Gênica , Marcação de Genes , Genômica , Herpesviridae/genética , Modelos Moleculares , Dobramento de Proteína , Estrutura Terciária de Proteína , Proteínas Virais/biossíntese , Proteínas Virais/química
13.
J Mol Biol ; 359(3): 667-78, 2006 Jun 09.
Artigo em Inglês | MEDLINE | ID: mdl-16647084

RESUMO

The Epstein-Barr virus is a human gamma-herpesvirus that persistently infects more than 90% of the human population. It is associated with numerous epithelial cancers, principally undifferentiated nasopharyngeal carcinoma and gastric carcinoma. The BARF1 gene is expressed in a high proportion of these cancers. An oncogenic, mitogenic and immortalizing activity of the BARF1 protein has been shown. We solved the structure of the secreted BARF1 glycoprotein expressed in a human cell line by X-ray crystallography at a resolution of 2.3A. The BARF1 protein consists of two immunoglobulin (Ig)-like domains. The N-terminal domain belongs to the subfamily of variable domains whereas the C-terminal one is related to a constant Ig-domain. BARF1 shows an unusual hexamerisation involving two principal contacts, one between the C-terminal domains and one between the N-terminal domains. The C-terminal contact with an uncommonly large contact surface extends the beta-sandwich of the Ig-domain through the second molecule. The N-terminal contact involves Ig-domains with an unusual relative orientation but with a more classical contact surface with a size in the range of dimer interactions of Ig-domains. The structure of BARF1 is most closely related to CD80 or B7-1, a co-stimulatory molecule present on antigen presenting cells, from which BARF1 must have been derived during evolution. Still, domain orientation and oligomerization differ between BARF1 and CD80. It had been shown that BARF1 binds to hCSF-1, the human colony-stimulating factor 1, but this interaction has to be principally different from the one between CSF-1 and CSF-1 receptor.


Assuntos
Proteínas Virais/química , Sequência de Aminoácidos , Linhagem Celular , Cristalografia por Raios X , Dimerização , Glicosilação , Humanos , Imunoglobulinas/química , Microscopia Eletrônica de Transmissão , Modelos Moleculares , Dados de Sequência Molecular , Ligação Proteica , Conformação Proteica , Estrutura Terciária de Proteína , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Proteínas Virais/isolamento & purificação , Proteínas Virais/ultraestrutura
14.
Structure ; 13(9): 1299-310, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16154087

RESUMO

Deoxyuridine 5'-triphosphate pyrophosphatases (dUTPases) are ubiquitous enzymes cleaving dUTP into dUMP and pyrophosphate. They occur as monomeric, dimeric, or trimeric molecules. The trimeric and monomeric enzymes both contain the same five characteristic sequence motifs but in a different order, whereas the dimeric enzymes are not homologous. Monomeric dUTPases only occur in herpesviruses, such as Epstein-Barr virus (EBV). Here, we describe the crystal structures of EBV dUTPase in complex with the product dUMP and a substrate analog alpha,beta-imino-dUTP. The molecule consists of three domains forming one active site that has a structure extremely similar to one of the three active sites of trimeric dUTPases. The three domains functionally correspond to the subunits of the trimeric form. Domains I and II have the dUTPase fold, but they differ considerably in the regions that are not involved in the formation of the unique active site, whereas domain III has only little secondary structure.


Assuntos
Herpesvirus Humano 4/enzimologia , Pirofosfatases/química , Sequência de Aminoácidos , Sítios de Ligação , Catálise , Cristalografia , Dissulfetos/química , Desenho de Fármacos , Evolução Molecular , Dados de Sequência Molecular , Dobramento de Proteína , Estrutura Terciária de Proteína , Especificidade por Substrato
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA