Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Antimicrob Agents Chemother ; 49(9): 3816-24, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16127058

RESUMO

Substitution of leucine for isoleucine at residue 50 (I50L) of human immunodeficiency virus (HIV) protease is the signature substitution for atazanavir (ATV) resistance. A unique phenotypic profile has been associated with viruses containing the I50L substitution, which produces ATV-specific resistance and increased susceptibility to most other approved HIV protease inhibitors (PIs). The basis for this unique phenotype has not been clearly elucidated. In this report, a direct effect of I50L on the susceptibility to the PI class is described. Cell-based protease assays using wild-type and PI-resistant proteases from laboratory and clinical isolates and in vitro antiviral assays were used to demonstrate a strong concordance between changes in PI susceptibility at the level of protease inhibition and changes in susceptibility observed at the level of virus infection. The results show that the induction of ATV resistance and increased susceptibility to other PIs by the I50L substitution is likely determined at the level of protease inhibition. Moreover, the I50L substitution functions to increase PI susceptibility even in the presence of other primary and secondary PI resistance substitutions. These findings may have implications regarding the optimal sequencing of PI therapies necessary to preserve PI treatment options of patients with ATV-resistant HIV infections.


Assuntos
Inibidores da Protease de HIV/farmacologia , HIV-1/efeitos dos fármacos , HIV-1/genética , Oligopeptídeos/farmacologia , Piridinas/farmacologia , Substituição de Aminoácidos , Sulfato de Atazanavir , Western Blotting , Linhagem Celular , Relação Dose-Resposta a Droga , Farmacorresistência Viral , Genes Virais/genética , Vetores Genéticos , Proteína do Núcleo p24 do HIV/genética , Proteína do Núcleo p24 do HIV/metabolismo , Infecções por HIV/virologia , Protease de HIV/genética , Protease de HIV/metabolismo , Humanos , Lamivudina/farmacologia , Mutação/genética , Fenótipo , Inibidores da Transcriptase Reversa/farmacologia
2.
Antimicrob Agents Chemother ; 48(9): 3498-507, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15328117

RESUMO

Entecavir (ETV) exhibits potent antiviral activity in patients chronically infected with wild-type or lamivudine (3TC)-resistant (3TC(r)) hepatitis B virus (HBV). Among the patients treated in phase II ETV clinical trials, two patients for whom previous therapies had failed exhibited virologic breakthrough while on ETV. Isolates from these patients (arbitrarily designated patients A and B) were analyzed genotypically for emergent substitutions in HBV reverse transcriptase (RT) and phenotypically for reduced susceptibility in cultures and in HBV polymerase assays. After 54 weeks of 3TC therapy, patient A (AI463901-A) received 0.5 mg of ETV for 52 weeks followed by a combination of ETV and 100 mg of 3TC for 89 weeks. Viral rebound occurred at 133 weeks after ETV was started. The 3TC(r) RT substitutions rtV173L, rtL180M, and rtM204V were present at study entry, and the additional substitutions rtI169T and rtM250V emerged during ETV-3TC combination treatment. Reduced ETV susceptibility in vitro required the rtM250V substitution in addition to the 3TC(r) substitutions. For liver transplant patient B (AI463015-B), previous famciclovir, ganciclovir, foscarnet, and 3TC therapies had failed, and RT changes rtS78S/T, rtV173L, rtL180M, rtT184S, and rtM204V were present at study entry. Viral rebound occurred after 76 weeks of therapy with ETV at 1.0 mg, with the emergence of rtT184G, rtI169T, and rtS202I substitutions within the preexisting 3TC(r) background. Reduced susceptibility in vitro was highest when both the rtT184G and the rtS202I changes were combined with the 3TC(r) substitutions. In summary, infrequent ETV resistance can emerge during prolonged therapy, with selection of additional RT substitutions within a 3TC(r) HBV background, leading to reduced ETV susceptibility and treatment failure.


Assuntos
Antivirais/farmacologia , Guanina/análogos & derivados , Guanina/farmacologia , Vírus da Hepatite B/efeitos dos fármacos , Vírus da Hepatite B/genética , Lamivudina/farmacologia , Inibidores da Transcriptase Reversa/farmacologia , Adulto , Substituição de Aminoácidos/genética , Antivirais/uso terapêutico , Proteínas do Capsídeo/biossíntese , Proteínas do Capsídeo/genética , Linhagem Celular Tumoral , Células Cultivadas , DNA Polimerase Dirigida por DNA/genética , Farmacorresistência Viral , Genótipo , Guanina/uso terapêutico , Hepatite B/tratamento farmacológico , Hepatite B/virologia , Vírus da Hepatite B/enzimologia , Humanos , Lamivudina/uso terapêutico , Masculino , Pessoa de Meia-Idade , Mutação , Fenótipo , DNA Polimerase Dirigida por RNA/genética , Inibidores da Transcriptase Reversa/uso terapêutico , Falha de Tratamento , Replicação Viral/efeitos dos fármacos , Replicação Viral/genética
3.
Antimicrob Agents Chemother ; 46(8): 2525-32, 2002 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12121928

RESUMO

Entecavir (ETV) is a potent and selective inhibitor of hepatitis B virus (HBV) replication in vitro and in vivo that is currently in clinical trials for the treatment of chronic HBV infections. A major limitation of the current HBV antiviral therapy, lamivudine (3TC), is the emergence of drug-resistant HBV in a majority of treated patients due to specific mutations in the nucleotide binding site of HBV DNA polymerase (HBV Pol). To determine the effects of 3TC resistance mutations on inhibition by ETV triphosphate (ETV-TP), a series of in vitro studies were performed. The inhibition of wild-type and 3TC-resistant HBV Pol by ETV-TP was measured using recombinant HBV nucleocapsids, and compared to that of 3TC-TP. These enzyme inhibition studies demonstrated that ETV-TP is a highly potent inhibitor of wild-type HBV Pol and is 100- to 300-fold more potent than 3TC-TP against 3TC-resistant HBV Pol. Cell culture assays were used to gauge the potential for antiviral cross-resistance of 3TC-resistant mutants to ETV. Results demonstrated that ETV inhibited the replication of 3TC-resistant HBV, but 20- to 30-fold higher concentrations were required. To gain further perspective regarding the potential therapeutic use of ETV, its phosphorylation was examined in hepatoma cells treated with extracellular concentrations representative of drug levels in plasma in ETV-treated patients. At these concentrations, intracellular ETV-TP accumulated to levels expected to inhibit the enzyme activity of both wild-type and 3TC-resistant HBV Pol. These findings are predictive of potent antiviral activity of ETV against both wild-type and 3TC-resistant HBV.


Assuntos
Antivirais/farmacologia , DNA Polimerase Dirigida por DNA/biossíntese , Guanina/análogos & derivados , Guanina/farmacologia , Vírus da Hepatite B/efeitos dos fármacos , Lamivudina/farmacologia , Replicação Viral/efeitos dos fármacos , Antivirais/metabolismo , Southern Blotting , Proteínas do Capsídeo/metabolismo , Células Cultivadas , DNA Polimerase Dirigida por DNA/genética , Resistência Microbiana a Medicamentos , Genes Virais/genética , Genoma Viral , Guanina/metabolismo , Humanos , Cinética , Fosforilação , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
4.
Bioorg Med Chem Lett ; 11(17): 2355-9, 2001 Sep 03.
Artigo em Inglês | MEDLINE | ID: mdl-11527730

RESUMO

A survey of isosteric replacements of the phosphonoalanine side chain coupled with a process of conformational constraint of a bisbenzimidazole-based, Zn(2+)-dependent inhibitor of hepatitis C virus (HCV) NS3 serine protease resulted in the identification of novel series of active compounds with extended side chains. However, Zn(2+)-dependent HCV NS3 inhibition was relatively insensitive to the structural variations examined but dependent on the presence of negatively charged functionality. This result was interpreted in the context of an initial electrostatic interaction between protease and inhibitor that is subsequently consolidated by Zn(2+), with binding facilitated by the featureless active site and proximal regions of the HCV NS3 protein.


Assuntos
Antivirais/química , Antivirais/farmacologia , Benzimidazóis/química , Benzimidazóis/farmacologia , Hepacivirus/enzimologia , Organofosfonatos/química , Organofosfonatos/farmacologia , Inibidores de Serina Proteinase/química , Inibidores de Serina Proteinase/farmacologia , Proteínas não Estruturais Virais/antagonistas & inibidores , Zinco/metabolismo , Antivirais/síntese química , Benzimidazóis/síntese química , Bioquímica/métodos , Avaliação Pré-Clínica de Medicamentos , Concentração Inibidora 50 , Estrutura Molecular , Organofosfonatos/síntese química , Inibidores de Serina Proteinase/síntese química , Relação Estrutura-Atividade , Proteínas não Estruturais Virais/química
5.
Virology ; 249(2): 460-70, 1998 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-9791036

RESUMO

The herpes simplex virus type 1 UL12 gene product, alkaline nuclease (AN), appears to be involved in viral DNA processing and capsid egress from the nucleus (Shao, L., Rapp, L. M., and Weller, S. K., Virology 196, 146-162, 1993). Although the HSV-1 AN is not absolutely essential for viral replication in tissue culture, conservation of the AN gene in all herpesviruses suggests an important role in the life cycle of herpesviruses. The counterpart of HSV-1 AN for human cytomegalovirus (HCMV) is the UL98 gene product. To examine whether the HCMV AN could substitute for HSV-1 AN, we performed trans-complementation experiments using a HSV-1 amplicon plasmid carrying the HCMV UL98 gene. Our results indicate (i) HCMV AN can complement the growth of the HSV-1 AN deletion mutant UL12lacZ virus in trans; (ii) a new recombinant virus, UL12laZcUL98/99, appears to be generated by the integration of the HCMV UL98 gene into the HSV-1 UL12lacZ viral genome; (iii) in contrast to its parental HSV-1 UL12lacZ virus, capsids formed in UL12lacZUL98/99-infected Vero cells were able to transport from the nucleus to the cytoplasm and mature into infectious viruses. Our results demonstrate a functional conservation of AN between HSV-1 and HCMV.


Assuntos
Citomegalovirus/enzimologia , Herpesvirus Humano 1/enzimologia , Ribonucleases/metabolismo , Animais , Chlorocebus aethiops , Mapeamento Cromossômico , Citomegalovirus/genética , Citomegalovirus/crescimento & desenvolvimento , Evolução Molecular , Deleção de Genes , Genes Virais , Teste de Complementação Genética , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/crescimento & desenvolvimento , Humanos , Óperon Lac , Microscopia Eletrônica , Mutação , Ribonucleases/genética , Especificidade da Espécie , Células Vero
6.
Virology ; 236(2): 338-47, 1997 Sep 29.
Artigo em Inglês | MEDLINE | ID: mdl-9325241

RESUMO

The method of substrate phage display was used to select a preferred substrate from three monovalent display libraries using the HSV-1 protease. The display libraries consisted of four random amino acids, six random amino acids, and a biased library containing four amino acids from the P side of the HSV-1 maturation site followed by four random amino acids. A series of consensus peptides was synthesized based upon the results from these screens and tested in peptide cleavage assays. An eight amino acids consensus peptide (LVLASSSF) derived from the phage results was cleaved as efficiently as a 20-mer maturation site peptide. The selected amino acid sequences also allowed the design of a four amino acid paranitroanilide substrate for continuous assay of HSV-1 protease. Similar to HCMV protease, these results define P4 to P1 as a minimal substrate recognition domain for the HSV-1 protease and suggest that P4 to P1 is the minimal substrate domain which all herpesvirus proteases recognize.


Assuntos
Capsídeo/metabolismo , Herpesvirus Humano 1/enzimologia , Serina Endopeptidases/metabolismo , Proteínas Virais , Sequência de Aminoácidos , Bacteriófago M13/genética , Sequência de Bases , Sítios de Ligação , Capsídeo/genética , Sequência Consenso , Primers do DNA/genética , Escherichia coli/genética , Herpesvirus Humano 1/genética , Cinética , Dados de Sequência Molecular , Oligopeptídeos/genética , Oligopeptídeos/metabolismo , Biblioteca de Peptídeos , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Serina Endopeptidases/genética , Especificidade por Substrato
7.
J Gen Virol ; 78 ( Pt 11): 2953-61, 1997 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-9367383

RESUMO

The human cytomegalovirus (HCMV) UL98 gene is predicted to encode a homologue of the conserved herpesvirus alkaline nuclease. To determine if the HCMV UL98 gene product is functionally homologous to other herpesvirus alkaline nucleases, the HCMV UL98 protein was purified and its activity characterized in vitro. Extracts of HCMV-infected cells were fractionated using Q Sepharose, phosphocellulose and native DNA cellulose chromatography. UL98 immunoreactivity copurified with alkaline pH-dependent nuclease activity. The purified protein migrated at its predicted size of approximately 65 kDa in denaturing polyacrylamide gels, and displayed nuclease activity in an activity gel assay. Enzyme activity was characterized by a high pH optimum, an absolute requirement for divalent cation, salt sensitivity, and 5' to 3' exonuclease activity. DNA digestion resulted in 5' monophosphoryl mono- and oligodeoxyribonucleotides. Kinetic analyses revealed a turnover rate of greater than 200 per min, and similar apparent affinity and rate constants on single- and double-stranded DNA. These results indicate that a functional alkaline nuclease activity is conserved among distant members of the herpesvirus family, and are consistent with a highly conserved role in the virus life cycle.


Assuntos
Citomegalovirus/genética , Genes Virais , Ribonucleases/genética , Citomegalovirus/enzimologia , Herpesviridae/enzimologia , Humanos , Cinética , Ribonucleases/análise , Ribonucleases/metabolismo , Especificidade por Substrato
8.
EMBO J ; 15(10): 2575-81, 1996 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-8665865

RESUMO

Herpes simplex virus (HSV) virions contain two regulatory proteins that facilitate the onset of the lytic cycle: VP16 activates transcription of the viral immediate-early genes, and vhs triggers shutoff of host protein synthesis and accelerated turnover of cellular and viral mRNAs. VP16 and vhs form a complex in infected cells, raising the possibility of a regulatory link between them. Here we show that viral protein synthesis and mRNA levels undergo a severe decline at intermediate times after infection with a VP16 null mutant, culminating in virtually complete translational arrest. This phenotype was rescued by a transcriptionally incompetent derivative of VP16 that retains vhs binding activity, and was eliminated by inactivating the vhs gene. These results indicate that VP16 dampens vhs activity, allowing HSV mRNAs to persist in infected cells. Further evidence supporting this hypothesis came from the demonstration that a stably transfected cell line expressing VP16 was resistant to host shutoff induced by superinfecting HSV virions. Thus, in addition to its well known function as a transcriptional activator, VP16 stimulates viral gene expression at a post-transcriptional level, by sparing viral mRNAs from degradation by one of the virus-induced host shutoff mechanisms.


Assuntos
Regulação Viral da Expressão Gênica , Proteína Vmw65 do Vírus do Herpes Simples/fisiologia , RNA Mensageiro/metabolismo , RNA Viral/metabolismo , Simplexvirus/fisiologia , Proteínas Virais/metabolismo , Replicação Viral/fisiologia , Animais , Sequência de Bases , Chlorocebus aethiops , Genes Precoces , Genes Virais , Substâncias Macromoleculares , Dados de Sequência Molecular , RNA Mensageiro/genética , RNA Viral/genética , Ribonucleases , Simplexvirus/genética , Transcrição Gênica , Células Vero , Proteínas Virais/biossíntese , Proteínas Virais/genética
9.
J Biol Chem ; 270(50): 30168-72, 1995 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-8530425

RESUMO

The herpes simplex virus type 1 protease is expressed as an 80,000-dalton polypeptide, encoded within the 635-amino acid open reading frame of the UL26 gene. The two known protein substrates for this enzyme are the protease itself and the capsid assembly protein ICP35 (Liu, F., and Roizman, B. (1991) J. Virol. 65, 5149-5156). In this report we describe the use of a rapid and quantitative assay for characterizing the protease. The assay uses a glutathione S-transferase fusion protein containing the COOH-terminal cleavage site of ICP35 as the substrate (GST-56). The protease consists of N0, the NH2-terminal 247 amino acid catalytic domain of the UL26 gene product, also expressed as a GST fusion protein. Upon cleavage with N0, a single 25-mer peptide is released from GST-56, which is soluble in trichloroacetic acid. Using this assay, the protease displayed a pH optimum between 7 and 9 but most importantly had an absolute requirement for high concentrations of an antichaeotrophic agent. Strong salting out salts such as Na2SO4 and KPO4 (> or = 1 M) stimulated activity, whereas NaCl and KCl had no effect. The degree of stimulation by 1.25 M Na2SO4 and KPO4 were 100-150- and 200-300-fold, respectively. Using the fluorescent probe 1-anilino-8-naphthalene sulfonate, the protease was shown to bind the dye in the presence of 1.25 M Na2SO4 or KPO4, but not at low ionic strength or in the presence of 1.25 or 2.2 M NaCl. This binding was most likely at the protease active site because a high affinity cleavage site peptide, but not a control peptide, could displace the dye. In addition to cleaving GST-56, the herpes simplex virus type I protease also cleaved the purified 56-mer peptide. Circular dichroism and NMR spectroscopy showed the peptide to be primarily random coil under physiological conditions, suggesting that antichaeotrophic agents affect the conformation of the substrate as well as the protease.


Assuntos
Sais/farmacologia , Serina Endopeptidases/metabolismo , Proteínas Virais , Acetatos/farmacologia , Sequência de Aminoácidos , Cloretos/farmacologia , Ativação Enzimática , Genes Virais , Glutationa Transferase/biossíntese , Glutationa Transferase/metabolismo , Herpesvirus Humano 1/enzimologia , Herpesvirus Humano 1/genética , Cinética , Dados de Sequência Molecular , Peso Molecular , Fases de Leitura Aberta , Peptídeos/síntese química , Peptídeos/química , Peptídeos/metabolismo , Fosfatos/farmacologia , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/isolamento & purificação , Proteínas Recombinantes de Fusão/metabolismo , Serina Endopeptidases/biossíntese , Serina Endopeptidases/isolamento & purificação , Espectrometria de Fluorescência , Especificidade por Substrato , Sulfatos/farmacologia
10.
J Virol ; 69(7): 4347-56, 1995 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-7769696

RESUMO

The herpes simplex virus type 1 protease and its substrate, ICP35, are involved in the assembly of viral capsids. Both proteins are encoded by a single open reading frame from overlapping mRNAs. The protease is autoproteolytically processed at two sites. The protease cleaves itself at the C-terminal site (maturation site) and also cleaves ICP35 at an identical site, releasing a 25-amino-acid (aa) peptide from each protein. To determine whether these 25 aa play a role in capsid assembly, we constructed a mutant virus expressing only Prb, the protease without the C-terminal 25 aa. Phenotypic analysis of the Prb virus in the presence and absence of ICP35 shows the following: (i) Prb retains the functional activity of the wild-type protease which supports virus growth in the presence of ICP35; (ii) in contrast to the ICP35 null mutant delta ICP35 virus, the Prb virus fails to grow in the absence of ICP35; and (iii) trans-complementation experiments indicated that full-length ICP35 (ICP35 c,d), but not the cleaved form (ICP35 e,f), complements the growth of the Prb virus. The most striking phenotype of the Prb virus is that only unsealed aberrant capsid structures are observed by electron microscopy in mutant-infected Vero cells. Our results demonstrate that the growth of herpes simplex virus type 1 requires the C-terminal 25 aa of either the protease or its substrate, ICP35, and that the C-terminal 25 aa are involved in the formation of sealed capsids.


Assuntos
Capsídeo/química , Serina Endopeptidases/fisiologia , Proteínas Virais/fisiologia , Animais , Sequência de Bases , Chlorocebus aethiops , Microscopia Eletrônica , Dados de Sequência Molecular , Mutação , Fragmentos de Peptídeos/fisiologia , Células Vero
11.
J Biol Chem ; 270(9): 4753-8, 1995 Mar 03.
Artigo em Inglês | MEDLINE | ID: mdl-7876248

RESUMO

The 28-kilodalton (kDa) catalytic domain of the human cytomegalovirus (HCMV) protease undergoes autoproteolytic cleavage at an internal site (I site), yielding amino-terminal 15-kDa (N15) and carboxyl-terminal 13-kDa (C13) fragments. I site autocleavage has been postulated to inactivate the protease and provide a mechanism for the negative regulation of enzyme activity during viral infection. We purified recombinant enzymes to demonstrate I site autocleavage in vitro and used site-directed mutagenesis of the I site to stabilize the protease. No difference in the kinetic properties of wild type and stabilized mutant proteases were observed in an in vitro peptide cleavage assay. The consequences of I site cleavage on enzyme activity were investigated two ways. First, autodigestion of the wild type enzyme converted the intact protease to N15 and C13 autocleavage products without a corresponding loss in enzyme activity. Second, genetic constructs encoding the N15 and C13 autocleavage products were generated and expressed separately in Escherichia coli, and each fragment was purified. An active enzyme was reconstituted by refolding a mixture of the purified fragments in vitro to form a noncovalent complex. The kinetic properties of this complex were very similar to the wild type and stabilized enzymes under optimal reaction conditions. We concluded from these studies that I site cleavage does not inactivate the HCMV protease, in the absence of other virally induced factors, and that limited potential exists for the regulation of catalytic activity by I site cleavage.


Assuntos
Citomegalovirus/enzimologia , Endopeptidases/metabolismo , Sequência de Bases , Sítios de Ligação , Catálise , Linhagem Celular Transformada , Endopeptidases/genética , Estabilidade Enzimática , Hidrólise , Cinética , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Oligodesoxirribonucleotídeos
12.
Eur J Biochem ; 226(2): 361-7, 1994 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-8001553

RESUMO

Human cytomegalovirus (HCMV) encodes a protease that cleaves itself and the HCMV assembly protein. Two proteolytic processing sites within the protease were identified at Ala 256-Ser 257 (release site) and Ala 643-Ser 644 (maturation site). Identification of rP5-P4' and mP4-P6' as the minimal peptide substrates spanning the release and maturation cleavage sites, respectively, demonstrated a requirement for residues flanking the conserved core in substrate recognition and hydrolysis, which are unique to HCMV. Kinetic parameters determined for release-site-derived and maturation-site-derived peptides revealed a 10-fold increase in kcat/Km for a maturational peptide (mP4-P8') over release-site peptide (rP5-P5'). Experimental results with a panel of class-specific protease inhibitors were consistent with the protease being a member of the serine or cysteine family of proteases. Further investigation revealed that the HCMV protease activity decreased with incorporation of [14C]iodoacetic acid, but when approximately 4.5 mol 14C were incorporated/mol enzyme, the enzyme retained approximately 20% of its original activity, indicating that hydrolysis does not require a cysteine nucleophile. Analysis of diisopropyl-fluorophosphate-inactivated protease by mass spectrometry indicated a stoichiometry of 1 diisopropyl phosphate/protease molecule, suggesting that hydrolysis requires a single serine nucleophile. The residue modified by diisopropyl fluorophosphate was identified as Ser132 by modification with 3H-labeled diisopropyl fluorophosphate, peptide mapping and Edman degradation. This residue and the region in which it is found is highly conserved among the herpes virus proteases. These data demonstrates that HCMV protease is a serine protease and that Ser132 is the active-site nucleophile.


Assuntos
Citomegalovirus/enzimologia , Endopeptidases/química , Endopeptidases/metabolismo , Serina Endopeptidases/química , Serina Endopeptidases/metabolismo , Sequência de Aminoácidos , Sítios de Ligação , Escherichia coli , Iodoacetatos/metabolismo , Ácido Iodoacético , Isoflurofato/farmacologia , Cinética , Dados de Sequência Molecular , Mapeamento de Peptídeos , Peptídeos/química , Peptídeos/metabolismo , Inibidores de Proteases/farmacologia , Proteínas Recombinantes de Fusão/metabolismo , Homologia de Sequência , Relação Estrutura-Atividade , Especificidade por Substrato , Tripsina
13.
J Virol ; 68(9): 5384-94, 1994 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-8057422

RESUMO

The herpes simplex virus type 1 ICP35 assembly protein is involved in the formation of viral capsids. ICP35 is encoded by the UL26.5 gene and is specifically processed by the herpes simplex virus type 1 protease encoded by the UL26 gene. To better understand the functions of ICP35 in infected cells, we have isolated and characterized an ICP35 mutant virus, delta ICP35. The mutant virus was propagated in complementing 35J cells, which express wild-type ICP35. Phenotypic analysis of delta ICP35 shows that (i) mutant virus growth in Vero cells was severely restricted, although small amounts of progeny virus was produced; (ii) full-length ICP35 protein was not produced, although autoproteolysis of the protease still occurred in mutant-infected nonpermissive cells; (iii) viral DNA replication of the mutant proceeded at wild-type levels, but only a very small portion of the replicated DNA was processed to unit length and encapsidated; (iv) capsid structures were observed in delta ICP35-infected Vero cells by electron microscopy and by sucrose sedimentation analysis; (v) assembly of VP5 into hexons of the capsids was conformationally altered; and (vi) ICP35 has a novel function which is involved in the nuclear transport of VP5.


Assuntos
Herpesvirus Humano 1/ultraestrutura , Proteínas Virais/fisiologia , Animais , Capsídeo/metabolismo , DNA Viral/genética , Genes Virais , Teste de Complementação Genética , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/crescimento & desenvolvimento , Técnicas In Vitro , Mapeamento por Restrição , Deleção de Sequência , Células Vero , Proteínas Estruturais Virais/genética , Replicação Viral
14.
J Biol Chem ; 269(17): 12672-6, 1994 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-8175677

RESUMO

Herpes simplex virus type 1 (HSV-1) encodes a protease that is essential for proteolytic processing of itself and of the nucleocapsid-associated protein, ICP35 (infected cell protein 35) (Liu, F., and Roizman, B. (1991) J. Virol. 65, 5149-5156). Inhibitor studies indicated that the HSV-1 protease is sensitive to the serine protease inactivator diisopropyl fluorophosphate (DFP). Inactivation is irreversible and dependent on time and concentration of DFP. Loss of activity correlates linearly with the incorporation of [3H]DFP. Analysis of completely inactivated protease by mass spectrometry indicated a stoichiometry of 1 DFP/protease. In order to identify the specific residue modified by DFP, the protease was labeled with [3H]DFP and subsequently digested with trypsin or chymotrypsin. The peptides resulting from each digestion were separated by reverse phase HPLC, and the radioactivity was recovered in a single peak. Mass spectrometric studies and sequencing analysis by Edman degradation identified Ser-129 as the residue modified by DFP. This residue and the region in which it is found is highly conserved among the herpes viral proteases. These data demonstrate that HSV-1 protease is a serine protease and that Ser-129 is the active site nucleophile.


Assuntos
Endopeptidases/metabolismo , Serina Endopeptidases/metabolismo , Simplexvirus/enzimologia , Proteínas Virais , Sequência de Aminoácidos , Animais , Baculoviridae/genética , Sítios de Ligação , Células Cultivadas , Cromatografia Líquida de Alta Pressão , Endopeptidases/química , Endopeptidases/genética , Isoflurofato/farmacologia , Cinética , Espectrometria de Massas , Dados de Sequência Molecular , Mariposas , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/isolamento & purificação , Mapeamento de Peptídeos , Inibidores de Proteases/farmacologia , Serina/análise , Serina Endopeptidases/química , Serina Endopeptidases/genética
15.
J Biol Chem ; 268(34): 25449-54, 1993 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-8244978

RESUMO

Herpes simplex virus type-1 (HSV-1) protease is responsible for proteolytic processing of itself and the virus assembly protein ICP35 (infected cell protein 35). Two proteolytic processing sites within the protease have recently been identified between Ala247 and Ser248 and between Ala610 and Ser611. In this report we demonstrate that peptides corresponding to each of these cleavage sites are substrates for recombinant HSV protease-glutathione S-transferase fusion protein in vitro by high performance liquid chromatography analysis of cleavage reactions. Analysis of the products by fast atom bombardment-mass spectrometry confirmed that cleavage occurred at the expected position between the Ala and Ser residues of the substrate. Peptide cleavage was linear with respect to time and enzyme concentration and proceeded optimally at pH 8.0. A peptide that spans Ala99/Ser100 of the protease but does not correspond to a naturally occurring cleavage site was not a substrate for the protease in vitro confirming that sequence elements outside the conserved dipeptide sequence are required for substrate recognition and cleavage. Identification of P5-P8' as the minimal substrate peptide for the Ala610/Ser611 cleavage site revealed a requirement for residues flanking the conserved core P4-LVNA/S-P1' in substrate recognition and hydrolysis. Kinetic analysis with peptide P5-P8' yielded a Km of 190 microM and kcat of 0.2 min-1. Experiments with a panel of class-specific protease inhibitors were consistent with the protease being a member of the general class of serine proteases.


Assuntos
Endopeptidases/metabolismo , Herpesvirus Humano 1/enzimologia , Sequência de Aminoácidos , Sítios de Ligação , Cromatografia Líquida de Alta Pressão , Proteínas de Ligação a DNA/biossíntese , Endopeptidases/biossíntese , Cinética , Dados de Sequência Molecular , Peptídeos/síntese química , Peptídeos/isolamento & purificação , Peptídeos/metabolismo , Inibidores de Proteases/farmacologia , Processamento de Proteína Pós-Traducional , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/metabolismo , Especificidade por Substrato , Proteínas Virais/biossíntese
16.
J Virol ; 67(10): 5813-22, 1993 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-8396657

RESUMO

The UL26 gene of herpes simplex virus type 1 (HSV-1) encodes a 635-amino-acid protease that cleaves itself and the HSV-1 assembly protein ICP35cd (F. Liu and B. Roizman, J. Virol. 65:5149-5156, 1991). We previously examined the HSV protease by using an Escherichia coli expression system (I. C. Deckman, M. Hagen, and P. J. McCann III, J. Virol. 66:7362-7367, 1992) and identified two autoproteolytic cleavage sites between residues 247 and 248 and residues 610 and 611 of UL26 (C. L. DiIanni, D. A. Drier, I. C. Deckman, P. J. McCann III, F. Liu, B. Roizman, R. J. Colonno, and M. G. Cordingley, J. Biol. Chem. 268:2048-2051, 1993). In this study, a series of C-terminal truncations of the UL26 open reading frame was tested for cleavage activity in E. coli. Our results delimit the catalytic domain of the protease to the N-terminal 247 amino acids of UL26 corresponding to No, the amino-terminal product of protease autoprocessing. Autoprocessing of the full-length protease was found to be unnecessary for catalysis, since elimination of either or both cleavage sites by site-directed mutagenesis fails to prevent cleavage of ICP35cd or an unaltered protease autoprocessing site. Catalytic activity of the 247-amino-acid protease domain was confirmed in vitro by using a glutathione-S-transferase fusion protein. The fusion protease was induced to high levels of expression, affinity purified, and used to cleave purified ICP35cd in vitro, indicating that no other proteins are required. By using a set of domain-specific antisera, all of the HSV-1 protease cleavage products predicted from studies in E. coli were identified in HSV-1-infected cells. At least two protease autoprocessing products, in addition to fully processed ICP35cd (ICP35ef), were associated with intermediate B capsids in the nucleus of infected cells, suggesting a key role for proteolytic maturation of the protease and ICP35cd in HSV-1 capsid assembly.


Assuntos
Capsídeo/metabolismo , Endopeptidases/metabolismo , Simplexvirus/enzimologia , Proteínas Virais , Alanina , Sequência de Aminoácidos , Sítios de Ligação , Western Blotting , Centrifugação com Gradiente de Concentração , Cromatografia de Afinidade , Clonagem Molecular , Endopeptidases/biossíntese , Endopeptidases/isolamento & purificação , Escherichia coli/genética , Genes Virais , Mutagênese Sítio-Dirigida , Plasmídeos , Mutação Puntual , Reação em Cadeia da Polimerase , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/isolamento & purificação , Proteínas Recombinantes de Fusão/metabolismo , Mapeamento por Restrição , Serina , Simplexvirus/genética , Trombina/metabolismo
17.
J Virol ; 66(1): 258-69, 1992 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-1309245

RESUMO

VP16 (also called Vmw65 and alpha TIF) is a structural protein of herpes simplex virus type 1 (HSV-1) that trans-induces HSV-1 immediate-early gene transcription. This report describes an HSV-1 VP16 deletion mutant that was constructed and propagated in a cell line transformed with a VP16 expression vector. The VP16 deletion mutant replicated like wild-type HSV-1 during infection of the VP16-expressing cell line. Deletion mutant virions propagated in this cell line contained wild-type, cell-derived VP16 protein that was recruited during virion assembly and was functional for immediate-early gene trans-induction. The mutant failed to replicate during subsequent infection of cells that do not express VP16, as determined in plaque assays and single-step replication assays. The deletion mutant induced nearly normal levels of viral DNA synthesis and capsid production during these infections, but it induced slightly lower levels of viral DNA encapsidation and appeared by transmission electron microscopy to be defective in further steps of virion maturation. A genetic revertant of the deletion mutant that was restored for VP16-coding sequences exhibited fully wild-type replication properties in both VP16-expressing and nonexpressing cells. The absence of VP16 protein synthesis at late times of HSV-1 infection prevents the production of infectious progeny virus and correlates with a profound defect in HSV-1 particle assembly.


Assuntos
Fases de Leitura Aberta , Simplexvirus/genética , Transativadores/genética , Proteínas Virais/genética , Alelos , Sequência de Aminoácidos , Animais , Southern Blotting , Linhagem Celular Transformada , Replicação do DNA , DNA Viral/biossíntese , Immunoblotting , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Recombinação Genética , Simplexvirus/fisiologia , Simplexvirus/ultraestrutura , Células Vero , Replicação Viral/genética
18.
J Mol Biol ; 195(4): 819-33, 1987 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-2821283

RESUMO

Gene expression by herpes simplex virus type 1 (HSV-1) results in the synthesis of three temporal classes of viral proteins. The three classes of viral proteins are expressed in a cascade manner of sequential dependency. The molecular mechanisms that account for the HSV-1 protein synthesis cascade are poorly understood. In order to provide a detailed description of the metabolic levels at which HSV-1 protein synthesis is regulated, we have measured transcription rates and mRNA accumulation levels for 11 HSV-1 genes. These measurements were made over a time-course of infection in the presence or absence of metabolic inhibitors of either viral protein synthesis or viral DNA synthesis. Our observations show that the protein synthesis cascade of HSV-1 is established as a consequence of mechanisms that regulate both the transcription and accumulation of viral messenger RNA.


Assuntos
Regulação da Expressão Gênica , Processamento de Proteína Pós-Traducional , Simplexvirus/genética , Transcrição Gênica , Proteínas Virais/biossíntese , Cicloeximida/farmacologia , DNA Viral/antagonistas & inibidores , Genes Virais/efeitos dos fármacos , Modelos Genéticos , Ácido Fosfonoacéticos/farmacologia , Regiões Promotoras Genéticas , RNA Mensageiro/biossíntese , RNA Viral/biossíntese
19.
Science ; 234(4772): 53-9, 1986 Oct 03.
Artigo em Inglês | MEDLINE | ID: mdl-3018926

RESUMO

Viral infection of mammalian cells entails the regulated induction of viral gene expression. The induction of many viral genes, including the herpes simplex virus gene encoding thymidine kinase (tk), depends on viral regulatory proteins that act in trans. Because recognition of the tk promoter by cellular transcription factors is well understood, its trans induction by viral regulatory proteins may serve as a useful model for the regulation of eukaryotic gene expression. A comprehensive set of mutations was therefore introduced into the chromosome of herpes simplex virus at the tk promoter to directly analyze the effects of promoter mutations on tk transcription. The promoter domains required for efficient tk expression under conditions of trans induction corresponded to those important for recognition by cellular transcription factors. Thus, trans induction of tk expression may be catalyzed initially by the interaction of viral regulatory proteins with cellular transcription factors.


Assuntos
Regulação da Expressão Gênica , Regiões Promotoras Genéticas , Ativação Viral , Animais , Genes Virais , Mutação , RNA Mensageiro/genética , Simplexvirus/genética , Simplexvirus/crescimento & desenvolvimento , Transcrição Gênica , Xenopus
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...