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.
PLoS Pathog ; 14(5): e1007086, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29782554

RESUMO

RNA viruses induce specialized membranous structures for use in genome replication. These structures are often referred to as replication organelles (ROs). ROs exhibit distinct lipid composition relative to other cellular membranes. In many picornaviruses, phosphatidylinositol-4-phosphate (PI4P) is a marker of the RO. Studies to date indicate that the viral 3A protein hijacks a PI4 kinase to induce PI4P by a mechanism unrelated to the cellular pathway, which requires Golgi-specific brefeldin A-resistance guanine nucleotide exchange factor 1, GBF1, and ADP ribosylation factor 1, Arf1. Here we show that a picornaviral 3CD protein is sufficient to induce synthesis of not only PI4P but also phosphatidylinositol-4,5-bisphosphate (PIP2) and phosphatidylcholine (PC). Synthesis of PI4P requires GBF1 and Arf1. We identified 3CD derivatives: 3CDm and 3CmD, that we used to show that distinct domains of 3CD function upstream of GBF1 and downstream of Arf1 activation. These same 3CD derivatives still supported induction of PIP2 and PC, suggesting that pathways and corresponding mechanisms used to induce these phospholipids are distinct. Phospholipid induction by 3CD is localized to the perinuclear region of the cell, the outcome of which is the proliferation of membranes in this area of the cell. We conclude that a single viral protein can serve as a master regulator of cellular phospholipid and membrane biogenesis, likely by commandeering normal cellular pathways.


Assuntos
Peptídeo Hidrolases/metabolismo , Fosfolipídeos/biossíntese , Picornaviridae/enzimologia , Proteínas Virais/metabolismo , Fator 1 de Ribosilação do ADP/metabolismo , Brefeldina A/farmacologia , Membrana Celular/ultraestrutura , Dactinomicina/farmacologia , Complexo de Golgi/efeitos dos fármacos , Complexo de Golgi/metabolismo , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Células HeLa , Humanos , Microscopia Eletrônica de Transmissão , Biogênese de Organelas , Fosfatos de Fosfatidilinositol/metabolismo , Poliovirus/enzimologia , Inibidores da Síntese de Proteínas/farmacologia , Piridinas/farmacologia , Quinolinas/farmacologia
2.
PLoS Pathog ; 14(4): e1007036, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29702686

RESUMO

At the culmination of poliovirus (PV) multiplication, membranes are observed that contain phosphatidylinositol-4-phosphate (PI4P) and appear as vesicular clusters in cross section. Induction and remodeling of PI4P and membranes prior to or concurrent with genome replication has not been well studied. Here, we exploit two PV mutants, termed EG and GG, which exhibit aberrant proteolytic processing of the P3 precursor that substantially delays the onset of genome replication and/or impairs virus assembly, to illuminate the pathway of formation of PV-induced membranous structures. For WT PV, changes to the PI4P pool were observed as early as 30 min post-infection. PI4P remodeling occurred even in the presence of guanidine hydrochloride, a replication inhibitor, and was accompanied by formation of membrane tubules throughout the cytoplasm. Vesicular clusters appeared in the perinuclear region of the cell at 3 h post-infection, a time too slow for these structures to be responsible for genome replication. Delays in the onset of genome replication observed for EG and GG PVs were similar to the delays in virus-induced remodeling of PI4P pools, consistent with PI4P serving as a marker of the genome-replication organelle. GG PV was unable to convert virus-induced tubules into vesicular clusters, perhaps explaining the nearly 5-log reduction in infectious virus produced by this mutant. Our results are consistent with PV inducing temporally distinct membranous structures (organelles) for genome replication (tubules) and virus assembly (vesicular clusters). We suggest that the pace of formation, spatiotemporal dynamics, and the efficiency of the replication-to-assembly-organelle conversion may be set by both the rate of P3 polyprotein processing and the capacity for P3 processing to yield 3AB and/or 3CD proteins.


Assuntos
Membrana Celular/química , Organelas/virologia , Fosfatos de Fosfatidilinositol/metabolismo , Poliomielite/virologia , Poliovirus/patogenicidade , Proteínas Virais/metabolismo , Replicação Viral , Membrana Celular/metabolismo , Genoma Viral , Células HeLa , Humanos , Mutação , Fosfatos de Fosfatidilinositol/química , Poliomielite/genética , Poliomielite/metabolismo , Poliovirus/genética , Análise Espaço-Temporal , Proteínas Virais/genética , Montagem de Vírus
3.
Nucleic Acids Res ; 46(5): 2537-2547, 2018 03 16.
Artigo em Inglês | MEDLINE | ID: mdl-29385522

RESUMO

Numerous mammalian proto-oncogene and other growth-regulatory transcripts are upregulated in malignancy due to abnormal mRNA stabilization. In hepatoma cells expressing a hepatitis C virus (HCV) subgenomic replicon, we found that the viral nonstructural protein 5A (NS5A), a protein known to bind to viral RNA, also bound specifically to human cellular transcripts that encode regulators of cell growth and apoptosis, and this binding correlated with transcript stabilization. An important subset of human NS5A-target transcripts contained GU-rich elements, sequences known to destabilize mRNA. We found that NS5A bound to GU-rich elements in vitro and in cells. Mutation of the NS5A zinc finger abrogated its GU-rich element-binding and mRNA stabilizing activities. Overall, we identified a molecular mechanism whereby HCV manipulates host gene expression by stabilizing host transcripts in a manner that would promote growth and prevent death of virus-infected cells, allowing the virus to establish chronic infection and lead to the development of hepatocellular carcinoma.


Assuntos
Estabilidade de RNA , Proteínas não Estruturais Virais/metabolismo , Linhagem Celular Tumoral , Células HeLa , Hepacivirus/metabolismo , Humanos , Proto-Oncogene Mas , RNA Mensageiro/metabolismo , Proteínas não Estruturais Virais/química
4.
Oncotarget ; 7(45): 72395-72414, 2016 Nov 08.
Artigo em Inglês | MEDLINE | ID: mdl-27590350

RESUMO

Despite ubiquitous activation in human cancer, essential downstream effector pathways of the MYC transcription factor have been difficult to define and target. Using a structure/function-based approach, we identified the mitochondrial RNA polymerase (POLRMT) locus as a critical downstream target of MYC. The multifunctional POLRMT enzyme controls mitochondrial gene expression, a process required both for mitochondrial function and mitochondrial biogenesis. We further demonstrate that inhibition of this newly defined MYC effector pathway causes robust and selective tumor cell apoptosis, via an acute, checkpoint-like mechanism linked to aberrant electron transport chain complex assembly and mitochondrial reactive oxygen species (ROS) production. Fortuitously, MYC-dependent tumor cell death can be induced by inhibiting the mitochondrial gene expression pathway using a variety of strategies, including treatment with FDA-approved antibiotics. In vivo studies using a mouse model of Burkitt's Lymphoma provide pre-clinical evidence that these antibiotics can successfully block progression of MYC-dependent tumors.


Assuntos
Regulação Neoplásica da Expressão Gênica , Genes Mitocondriais , Genes myc , Neoplasias/genética , Animais , Linhagem Celular Tumoral , RNA Polimerases Dirigidas por DNA/genética , RNA Polimerases Dirigidas por DNA/metabolismo , Feminino , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mitocôndrias/genética , Mitocôndrias/metabolismo , Neoplasias/patologia , Proteínas Proto-Oncogênicas c-myc , Espécies Reativas de Oxigênio/metabolismo , Transfecção
6.
PLoS One ; 8(3): e57905, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23483938

RESUMO

Interferon inducible protein kinase PKR is an essential component of innate immunity. It is activated by long stretches of dsRNA and provides the first line of host defense against pathogens by inhibiting translation initiation in the infected cell. Many cellular and viral transcripts contain nucleoside modifications and/or tertiary structure that could affect PKR activation. We have previously demonstrated that a 5'-end triphosphate-a signature of certain viral and bacterial transcripts-confers the ability of relatively unstructured model RNA transcripts to activate PKR to inhibit translation, and that this activation is abrogated by certain modifications present in cellular RNAs. In order to understand the biological implications of native RNA tertiary structure and nucleoside modifications on PKR activation, we study here the heavily modified cellular tRNAs and the unmodified or the lightly modified mitochondrial tRNAs (mt-tRNA). We find that both a T7 transcript of yeast tRNA(Phe) and natively extracted total bovine liver mt-tRNA activate PKR in vitro, whereas native E. coli, bovine liver, yeast, and wheat tRNA(Phe) do not, nor do a variety of base- or sugar-modified T7 transcripts. These results are further supported by activation of PKR by a natively folded T7 transcript of tRNA(Phe)in vivo supporting the importance of tRNA modification in suppressing PKR activation in cells. We also examine PKR activation by a T7 transcript of the A14G pathogenic mutant of mt-tRNA(Leu), which is known to dimerize, and find that the misfolded dimeric form activates PKR in vitro while the monomeric form does not. Overall, the in vitro and in vivo findings herein indicate that tRNAs have an intrinsic ability to activate PKR and that nucleoside modifications and native RNA tertiary folding may function, at least in part, to suppress such activation, thus serving to distinguish self and non-self tRNA in innate immunity.


Assuntos
Imunidade Inata/imunologia , Conformação de Ácido Nucleico , Nucleosídeos/metabolismo , RNA de Transferência/química , RNA de Transferência/metabolismo , eIF-2 Quinase/metabolismo , Animais , Sequência de Bases , Bovinos , Linhagem Celular Tumoral , Dimerização , Ativação Enzimática , Humanos , Dados de Sequência Molecular , Mutação/genética , Ligação Proteica , RNA/química , RNA/genética , RNA Mitocondrial , RNA de Transferência/genética , Saccharomyces cerevisiae/metabolismo
7.
PLoS Pathog ; 8(11): e1003030, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23166498

RESUMO

Ribonucleoside analogues have potential utility as anti-viral, -parasitic, -bacterial and -cancer agents. However, their clinical applications have been limited by off target effects. Development of antiviral ribonucleosides for treatment of hepatitis C virus (HCV) infection has been hampered by appearance of toxicity during clinical trials that evaded detection during preclinical studies. It is well established that the human mitochondrial DNA polymerase is an off target for deoxyribonucleoside reverse transcriptase inhibitors. Here we test the hypothesis that triphosphorylated metabolites of therapeutic ribonucleoside analogues are substrates for cellular RNA polymerases. We have used ribonucleoside analogues with activity against HCV as model compounds for therapeutic ribonucleosides. We have included ribonucleoside analogues containing 2'-C-methyl, 4'-methyl and 4'-azido substituents that are non-obligate chain terminators of the HCV RNA polymerase. We show that all of the anti-HCV ribonucleoside analogues are substrates for human mitochondrial RNA polymerase (POLRMT) and eukaryotic core RNA polymerase II (Pol II) in vitro. Unexpectedly, analogues containing 2'-C-methyl, 4'-methyl and 4'-azido substituents were inhibitors of POLRMT and Pol II. Importantly, the proofreading activity of TFIIS was capable of excising these analogues from Pol II transcripts. Evaluation of transcription in cells confirmed sensitivity of POLRMT to antiviral ribonucleosides, while Pol II remained predominantly refractory. We introduce a parameter termed the mitovir (mitochondrial dysfunction caused by antiviral ribonucleoside) score that can be readily obtained during preclinical studies that quantifies the mitochondrial toxicity potential of compounds. We suggest the possibility that patients exhibiting adverse effects during clinical trials may be more susceptible to damage by nucleoside analogs because of defects in mitochondrial or nuclear transcription. The paradigm reported here should facilitate development of ribonucleosides with a lower potential for toxicity.


Assuntos
Antivirais/farmacologia , Núcleo Celular/metabolismo , RNA Polimerases Dirigidas por DNA/metabolismo , Hepacivirus/metabolismo , Mitocôndrias/metabolismo , RNA Polimerase II/metabolismo , Ribonucleosídeos/farmacologia , Transcrição Gênica/efeitos dos fármacos , Animais , Antivirais/efeitos adversos , Bovinos , Linhagem Celular , Hepatite C/tratamento farmacológico , Hepatite C/enzimologia , RNA Viral/biossíntese , Ribonucleosídeos/efeitos adversos
8.
J Biol Chem ; 287(36): 30861-73, 2012 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-22801423

RESUMO

Hepatitis C virus (HCV) is the main agent of acute and chronic liver diseases leading to cirrhosis and hepatocellular carcinoma. The current standard therapy has limited efficacy and serious side effects. Thus, the development of alternate therapies is of tremendous importance. HCV NS5A (nonstructural 5A protein) is a pleiotropic protein with key roles in HCV replication and cellular signaling pathways. Here we demonstrate that NS5A dimerization occurs through Domain I (amino acids 1-240). This interaction is not mediated by nucleic acids because benzonase, RNase, and DNase treatments do not prevent NS5A-NS5A interactions. Importantly, DTT abrogates NS5A-NS5A interactions but does not affect NS5A-cyclophilin A interactions. Other reducing agents such as tris(2-carboxyethyl)phosphine and 2-mercaptoethanol also abrogate NS5A-NS5A interactions, implying that disulfide bridges may play a role in this interaction. Cyclophilin inhibitors, cyclosporine A, and alisporivir and NS5A inhibitor BMS-790052 do not block NS5A dimerization, suggesting that their antiviral effects do not involve the disruption of NS5A-NS5A interactions. Four cysteines, Cys-39, Cys-57, Cys-59, and Cys-80, are critical for dimerization. Interestingly, the four cysteines have been proposed to form a zinc-binding motif. Supporting this notion, NS5A dimerization is greatly facilitated by Zn(2+) but not by Mg(2+) or Mn(2+). Importantly, the four cysteines are vital not only for viral replication but also critical for NS5A binding to RNA, revealing a correlation between NS5A dimerization, RNA binding, and HCV replication. Altogether our data suggest that NS5A-NS5A dimerization and/or multimerization could represent a novel target for the development of HCV therapies.


Assuntos
Hepacivirus/fisiologia , Multimerização Proteica/fisiologia , RNA Viral/metabolismo , Proteínas não Estruturais Virais/metabolismo , Replicação Viral/fisiologia , Carbamatos , Ciclofilina A/genética , Ciclofilina A/metabolismo , Humanos , Imidazóis/química , Imidazóis/farmacologia , Multimerização Proteica/efeitos dos fármacos , Estrutura Terciária de Proteína , Pirrolidinas , RNA Viral/química , RNA Viral/genética , Valina/análogos & derivados , Proteínas não Estruturais Virais/química , Proteínas não Estruturais Virais/genética , Replicação Viral/efeitos dos fármacos
9.
PLoS One ; 7(3): e32661, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22438882

RESUMO

Retinoic acid inducible gene-I (RIG-I) is a key regulator of antiviral immunity. RIG-I is generally thought to be activated by ssRNA species containing a 5'-triphosphate (PPP) group or by unphosphorylated dsRNA up to ~300 bp in length. However, it is not yet clear how changes in the length, nucleotide sequence, secondary structure, and 5' end modification affect the abilities of these ligands to bind and activate RIG-I. To further investigate these parameters in the context of naturally occurring ligands, we examined RNA sequences derived from the 5' and 3' untranslated regions (UTR) of the influenza virus NS1 gene segment. As expected, RIG-I-dependent interferon-ß (IFN-ß) induction by sequences from the 5' UTR of the influenza cRNA or its complement (26 nt in length) required the presence of a 5'PPP group. In contrast, activation of RIG-I by the 3' UTR cRNA sequence or its complement (172 nt) exhibited only a partial 5'PPP-dependence, as capping the 5' end or treatment with CIP showed a modest reduction in RIG-I activation. Furthermore, induction of IFN-ß by a smaller, U/A-rich region within the 3' UTR was completely 5'PPP-independent. Our findings demonstrated that RNA sequence, length, and secondary structure all contributed to whether or not the 5'PPP moiety is needed for interferon induction by RIG-I.


Assuntos
RNA Helicases DEAD-box/metabolismo , Genoma Viral , Vírus da Influenza A/genética , Vírus da Influenza A/metabolismo , RNA Viral/genética , RNA Viral/metabolismo , Regiões 3' não Traduzidas , Regiões 5' não Traduzidas , Sítios de Ligação/genética , Linhagem Celular , Proteína DEAD-box 58 , RNA Helicases DEAD-box/química , Interações Hospedeiro-Patógeno/genética , Interações Hospedeiro-Patógeno/imunologia , Interações Hospedeiro-Patógeno/fisiologia , Humanos , Vírus da Influenza A/imunologia , Interferon beta/biossíntese , Interferon beta/genética , Cinética , Ligantes , Conformação de Ácido Nucleico , RNA Viral/química , Receptores Imunológicos , Proteínas não Estruturais Virais/genética
10.
J Biol Chem ; 285(30): 22725-31, 2010 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-20457607

RESUMO

Hepatitis C virus non-structural protein 3 contains a serine protease and an RNA helicase. Protease cleaves the genome-encoded polyprotein and inactivates cellular proteins required for innate immunity. Protease has emerged as an important target for the development of antiviral therapeutics, but drug resistance has turned out to be an obstacle in the clinic. Helicase is required for both genome replication and virus assembly. Mechanistic and structural studies of helicase have hurled this enzyme into a prominent position in the field of helicase enzymology. Nevertheless, studies of helicase as an antiviral target remain in their infancy.


Assuntos
Antivirais/metabolismo , Antivirais/farmacologia , Hepacivirus/metabolismo , Proteínas não Estruturais Virais/metabolismo , Imunidade Inata/imunologia , Ácidos Nucleicos/química , Ácidos Nucleicos/metabolismo , Multimerização Proteica , Proteínas não Estruturais Virais/antagonistas & inibidores , Proteínas não Estruturais Virais/química , Proteínas não Estruturais Virais/imunologia
11.
Indian J Med Res ; 131: 17-34, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20167971

RESUMO

Hepatitis C virus (HCV) is a small (approximately 55 to 65 nm), spherical, enveloped, hepatotropic RNA virus that causes acute and chronic hepatitis in humans. Persistent virus infection with HCV often leads to cirrhosis and hepatocellular carcinoma (HCC). At present there is neither a selective antiviral therapy nor a preventive vaccine. The only available treatment option is a long-acting pegylated-interferon-alpha, given in combination with nucleoside analog ribavirin, which is not very effective. Molecular studies of HCV began with the successful cloning of its genome in 1989. For many years, research to develop therapeutics was stalled by the inability to grow virus in tissue culture. A major milestone was achieved with the recent development of a robust cell culture system for HCV propagation. HCV proteins assemble and form replication complexes on modified host membranes, called as membranous webs. Even though HCV is detected and targeted by host immune mechanisms, it establishes and maintains a life-long persistent infection. HCV has evolved multiple strategies to survive and persist in hostile cellular environments; and the viral population is known to rapidly change during the course of a natural infection thereby escaping immune surveillance. Rapid mutations also help virus to survive by selecting for the variants which are resistant to antiviral drugs. Although precise mechanisms regulating HCV entry into hepatic cells via receptors remain unknown, HCV also has the capability of direct cell-to-cell transmission. The extremely complex and incompletely understood nature of the HCV lifecycle has complicated the discovery of new therapies. A complete understanding of the functional roles played by the HCV proteins during HCV lifecycle is vital for developing a successful cure. This review deals with current status of efforts in addressing these daunting tasks and challenges in developing therapeutics against chronic and rapidly changing hepatitis C virus.


Assuntos
Hepacivirus/genética , Hepatite C/tratamento farmacológico , Hepatite C/virologia , Linhagem Celular , Hepacivirus/fisiologia , Humanos , Interferon alfa-2 , Interferon-alfa/uso terapêutico , Polietilenoglicóis/uso terapêutico , Proteínas Recombinantes , Proteínas Virais/genética , Replicação Viral
12.
J Physiol ; 575(Pt 1): 229-39, 2006 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-16777938

RESUMO

Chronic intermittent hypoxia (CIH) augments physiological responses to low partial pressures of O2 in the arterial blood. Adrenal medullae from adult rats, however, are insensitive to direct effects of acute hypoxia. In the present study, we examined whether CIH induces hypoxic sensitivity in the adult rat adrenal medulla and, if so, by what mechanism(s). Experiments were performed on adult male rats exposed to CIH (15 s of 5% O2 followed by 5 min of 21% O2; 9 episodes h(-1); 8 h d(-1); for 3 or 10 days) or to comparable, cumulative durations of continuous hypoxia (CH; 4 h of 7% O2 followed by 20 h of 21% O2 for 1 or 10 days). Noradrenaline (NA) and adrenaline (ADR) effluxes were monitored from ex vivo adrenal medullae. In adrenal medullae of rats exposed to CIH, acute hypoxia evoked robust NA and ADR effluxes, whereas these responses were absent in control rats or in those exposed to CH for 1 or 10 days. Hypercapnia (10% CO2; either acidic, pH 6.8, or isohydric, pH 7.4) was ineffective in eliciting catecholamine (CA) efflux from control, CIH or CH rats. Nicotine (100 microM) evoked NA and ADR effluxes in control rats, and this response was abolished in CIH but not in CH rats. Systemic administration of 2-deoxyglucose depleted ADR content in control rats, and CIH attenuated this response, indicating downregulation of neurally regulated CA secretion. Cytosolic and mitochondrial aconitase enzyme activities decreased in CIH adrenal medullae, suggesting increased generation of superoxide anions. Systemic administration of antioxidants reversed the effect of CIH on the adrenal medulla. Rats exposed to CIH exhibited increased blood pressures and elevated plasma CA, and antioxidants abolished these responses. These observations demonstrate that CIH induces hypoxic sensing in the adult rat adrenal medulla via mechanisms involving increased generation of superoxide anions and suggest that hypoxia-evoked CA efflux from the adrenal medulla contributes, in part, to elevated blood pressure and plasma CA.


Assuntos
Medula Suprarrenal/metabolismo , Catecolaminas/metabolismo , Hipóxia/metabolismo , Estresse Oxidativo , Acetilcisteína/farmacologia , Medula Suprarrenal/efeitos dos fármacos , Animais , Antioxidantes/farmacologia , Pressão Sanguínea/efeitos dos fármacos , Catecolaminas/sangue , Epinefrina/sangue , Epinefrina/metabolismo , Hipóxia/sangue , Hipóxia/fisiopatologia , Masculino , Nicotina/farmacologia , Agonistas Nicotínicos/farmacologia , Norepinefrina/sangue , Norepinefrina/metabolismo , Ratos , Ratos Sprague-Dawley , Superóxidos/metabolismo , Fatores de Tempo
13.
J Biol Chem ; 281(6): 3528-35, 2006 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-16306038

RESUMO

The hepatitis C virus (HCV) nonstructural protein 3 (NS3) is a multifunctional enzyme with serine protease and DEXH/D-box helicase domains. A crystal structure of the NS3 helicase domain (NS3h) was generated in the presence of a single-stranded oligonucleotide long enough to accommodate binding of two molecules of enzyme. Several amino acid residues at the interface of the two NS3h molecules were identified that appear to mediate a protein-protein interaction between domains 2 and 3 of adjacent molecules. Mutations were introduced into domain 3 to disrupt the putative interface and subsequently examined using an HCV subgenomic replicon, resulting in significant reduction in replication capacity. The mutations in domain 3 were then examined using recombinant NS3h in biochemical assays. The mutant enzyme showed RNA binding and RNA-stimulated ATPase activity that mirrored wild type NS3h. In DNA unwinding assays under single turnover conditions, the mutant NS3h exhibited a similar unwinding rate and only approximately 2-fold lower processivity than wild type NS3h. Overall biochemical activities of the mutant NS3h were similar to the wild type enzyme, which was not reflective of the large reduction in HCV replicative capacity observed in the biological experiment. Hence, the biological results suggest that the known biochemical properties associated with the helicase activity of NS3h do not reveal all of the likely biological roles of NS3 during HCV replication. Domain 3 of NS3 is implicated in protein-protein interactions that are necessary for HCV replication.


Assuntos
Hepacivirus/genética , Proteínas não Estruturais Virais/química , Replicação Viral , Adenosina Trifosfatases/química , Western Blotting , Linhagem Celular , Membrana Celular/metabolismo , Cristalografia por Raios X , DNA/química , Dimerização , Relação Dose-Resposta a Droga , Genoma Viral , Humanos , Cinética , Modelos Moleculares , Modelos Estatísticos , Mutação , Oligonucleotídeos/química , Plasmídeos/metabolismo , Ligação Proteica , Conformação Proteica , Mapeamento de Interação de Proteínas , Estrutura Terciária de Proteína , RNA/química , Proteínas Recombinantes/química , Espectrofotometria , Proteínas não Estruturais Virais/metabolismo
14.
J Biol Chem ; 280(43): 36417-28, 2005 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-16126720

RESUMO

Hepatitis C virus (HCV) nonstructural protein 5A (NS5A) has been shown to antagonize numerous cellular pathways, including the antiviral interferon-alpha response. However, the capacity of this protein to interact with the viral polymerase suggests a more direct role for NS5A in genome replication. In this study, we employed two bacterially expressed, soluble derivatives of NS5A to probe for novel functions of this protein. We find that NS5A has the capacity to bind to the 3'-ends of HCV plus and minus strand RNAs. The high affinity binding site for NS5A in the 3'-end of plus strand RNA maps to the polypyrimidine tract, an element known to be essential for genome replication and infectivity. NS5A has a preference for single-stranded RNA containing stretches of uridine or guanosine. Values for the equilibrium dissociation constants for high affinity binding sites were in the 10 nM range. Two-dimensional gel electrophoresis followed by Western blotting revealed the presence of unphosphorylated NS5A in Huh-7 cells stably expressing the subgenomic replicon. Moreover, RNA immunoprecipitation and NS5A pull-down experiments showed the capacity of replicon-derived NS5A to bind to synthetic RNA and the HCV genome, respectively. Deletion of all of the casein kinase II phosphorylation sites in NS5A supported stable replication of a subgenomic replicon in Huh-7. However, this derivative could not be labeled with inorganic phosphate, suggesting that extensive phosphorylation of NS5A is not required for the replication functions of NS5A. The discovery that NS5A is an RNA-binding protein defines a new functional target for development of agents to treat HCV infection and a new structural class of RNA-binding proteins.


Assuntos
Proteínas de Ligação a RNA/química , Proteínas não Estruturais Virais/fisiologia , Sequência de Bases , Sítios de Ligação , Ligação Competitiva , Biotinilação , Western Blotting , Colódio/química , Reagentes de Ligações Cruzadas/farmacologia , Relação Dose-Resposta a Droga , Eletroforese em Gel Bidimensional , Eletroforese em Gel de Poliacrilamida , Deleção de Genes , Genoma Viral , Guanosina Monofosfato/química , Imunoprecipitação , Cinética , Modelos Genéticos , Modelos Estatísticos , Dados de Sequência Molecular , Conformação de Ácido Nucleico , Oligonucleotídeos/química , Fosforilação , Ligação Proteica , Estrutura Terciária de Proteína , Pirimidinas/química , RNA/química , Proteínas Recombinantes/química , Transcrição Gênica , Raios Ultravioleta , Uridina Monofosfato/química , Proteínas não Estruturais Virais/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA