Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Am J Clin Oncol ; 39(6): 575-580, 2016 12.
Article in English | MEDLINE | ID: mdl-24879476

ABSTRACT

OBJECTIVES: The effect of ethnicity on nasopharyngeal cancer (NPC) outcomes is unclear. This retrospective analysis examines survival and the impact of concurrent chemoradiation (chemoRT) among Asian and non-Asian patients. METHODS: Subjects included 380 consecutive patients with NPC treated at a Canadian institution from 2000 to 2009. Five-year Kaplan-Meier progression-free survival (PFS), disease-specific survival (DSS), and overall survival (OS) were compared between Asian (n=279) and non-Asian (n=101) subjects. Multivariable analysis was performed using Cox regression modeling. Two-variable interaction terms with concurrent chemoRT were used to examine whether concurrent chemoRT conferred different effects among subgroups. RESULTS: Asian subjects presented with earlier stage (P=0.005), were younger, had better performance status, and were less likely smokers (all P<0.001). Survival among Asian versus non-Asian subjects with stage I/II NPC were: PFS 68% versus 59% (P=0.04), DSS 87% versus 77% (P=0.08), and OS 84% versus 74% (P=0.003). Corresponding rates with stage III/IVA/IVB disease were PFS 49% versus 42% (P=0.12), DSS 72% versus 46% (P=0.001), and OS 70% versus 44% (P<0.001). On multivariable analysis, Asian ethnicity, age below 65 years, ECOG performance status 0-1, early stage, staging MRI use, and concurrent chemoRT were associated with improved DSS and OS (P<0.05). On testing interactions with concurrent chemoRT, Asian versus non-Asian ethnicity was significant (hazard ratio 3.9), suggesting that concurrent chemoRT conferred more benefit among non-Asian compared with Asian subjects. CONCLUSIONS: In this population-based study, Asian ethnicity was associated with improved DSS and OS. Concurrent chemoRT conferred more benefit among non-Asian compared with Asian subjects.


Subject(s)
Asian People/statistics & numerical data , Carcinoma/ethnology , Carcinoma/therapy , Nasopharyngeal Neoplasms/ethnology , Nasopharyngeal Neoplasms/therapy , White People/statistics & numerical data , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , British Columbia , Carcinoma/mortality , Carcinoma/pathology , Chemoradiotherapy/methods , Cohort Studies , Combined Modality Therapy , Disease-Free Survival , Female , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Multivariate Analysis , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms/mortality , Nasopharyngeal Neoplasms/pathology , Neoplasm Invasiveness/pathology , Neoplasm Staging , Prognosis , Proportional Hazards Models , Radiotherapy, Conformal/methods , Retrospective Studies , Survival Analysis , Treatment Outcome
2.
Oncotarget ; 6(37): 40247-54, 2015 Nov 24.
Article in English | MEDLINE | ID: mdl-26450901

ABSTRACT

We examined the regulation of NF-κB in prostate cancer by estrogen receptor ß (ERß) based on the inverse correlation between p65 and ERß expression that exists in prostate carcinomas and reports that ERß can inhibit NF-κB activation, although the mechanism is not known. We demonstrate that ERß functions as a gate-keeper for NF-κB p65 signaling by repressing its expression and nuclear translocation. ERß regulation of NF-κB signaling is mediated by HIF-1. Loss of ERß or hypoxia stabilizes HIF-1α, which we found to be a direct driver of IKKß transcription through a hypoxia response element present in the promoter of the IKKß gene. The increase of IKKß expression in ERß-ablated cells correlates with an increase in phospho-IκBα and concomitant p65 nuclear translocation. An inverse correlation between the expression of ERß and IKKß/p65 was also observed in the prostates of ERß knockout (BERKO) mice, Gleason grade 5 prostate tumors and analysis of prostate cancer databases. These findings provide a novel mechanism for how ERß prevents NF-κB activation and raise the exciting possibility that loss of ERß expression is linked to chronic inflammation in the prostate, which contributes to the development of high-grade prostate cancer.


Subject(s)
Estrogen Receptor beta/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , NF-kappa B/metabolism , Signal Transduction , Animals , Cell Hypoxia , Cell Line , Cell Line, Tumor , Estrogen Receptor beta/genetics , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , I-kappa B Kinase/genetics , I-kappa B Kinase/metabolism , I-kappa B Proteins/genetics , I-kappa B Proteins/metabolism , Immunoblotting , Male , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Confocal , NF-KappaB Inhibitor alpha , NF-kappa B/genetics , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factor RelA/genetics , Transcription Factor RelA/metabolism
3.
Cell Rep ; 10(12): 1982-91, 2015 Mar 31.
Article in English | MEDLINE | ID: mdl-25818291

ABSTRACT

The role of ERß in prostate cancer is unclear, although loss of ERß is associated with aggressive disease. Given that mice deficient in ERß do not develop prostate cancer, we hypothesized that ERß loss occurs as a consequence of tumorigenesis caused by other oncogenic mechanisms and that its loss is necessary for tumorigenesis. In support of this hypothesis, we found that ERß is targeted for repression in prostate cancer caused by PTEN deletion and that loss of ERß is important for tumor formation. ERß transcription is repressed by BMI-1, which is induced by PTEN deletion and important for prostate tumorigenesis. This finding provides a mechanism for how ERß expression is regulated in prostate cancer. Repression of ERß contributes to tumorigenesis because it enables HIF-1/VEGF signaling that sustains BMI-1 expression. These data reveal a positive feedback loop that is activated in response to PTEN loss and sustains BMI-1.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Estrogen Receptor beta/metabolism , Gene Expression Regulation, Neoplastic/physiology , PTEN Phosphohydrolase/metabolism , Prostatic Neoplasms/metabolism , Animals , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Humans , Male , Mice , Prostatic Neoplasms/genetics , Signal Transduction/genetics
4.
Proc Natl Acad Sci U S A ; 110(12): 4708-13, 2013 Mar 19.
Article in English | MEDLINE | ID: mdl-23487784

ABSTRACT

Estrogen receptor ß (ERß) promotes the degradation of hypoxia inducible factor 1α (HIF-1α), which contributes to the ability of this hormone receptor to sustain the differentiation of epithelial and carcinoma cells. Although the loss of ERß and consequent HIF-1 activation occur in prostate cancer with profound consequences, the mechanism by which ERß promotes the degradation of HIF-1α is unknown. We report that ERß regulates the ligand (3ß-adiol)-dependent transcription of prolyl hydroxylase 2 (PHD2) also known as Egl nine homolog 1 (EGLN1), a 2-oxoglutarate-dependent dioxygenase that hydroxylates HIF-1α and targets it for recognition by the von Hippel-Lindau tumor suppressor and consequent degradation. ERß promotes PHD2 transcription by interacting with a unique estrogen response element in the 5' UTR of the PHD2 gene that functions as an enhancer. PHD2 itself is critical for maintaining epithelial differentiation. Loss of PHD2 expression or inhibition of its function results in dedifferentiation with characteristics of an epithelial-mesenchymal transition, and exogenous PHD2 expression in dedifferentiated cells can restore an epithelial phenotype. Moreover, expression of HIF-1α in cells that express PHD2 does not induce dedifferentiation but expression of HIF-1α containing mutations in the proline residues that are hydroxylated by PHD2 induces dedifferentiation. These data describe a unique mechanism for the regulation of HIF-1α stability that involves ERß-mediated transcriptional regulation of PHD2 and they highlight an unexpected role for PHD2 in maintaining epithelial differentiation.


Subject(s)
Cell Differentiation/physiology , Epithelial Cells/metabolism , Estrogen Receptor beta/metabolism , Gene Expression Regulation, Enzymologic/physiology , Procollagen-Proline Dioxygenase/biosynthesis , Response Elements/physiology , Transcription, Genetic/physiology , Cell Line, Tumor , Epithelial Cells/cytology , Estrogen Receptor beta/genetics , Humans , Hydroxylation/physiology , Hypoxia-Inducible Factor 1, alpha Subunit/biosynthesis , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor-Proline Dioxygenases , Male , Mutation , Procollagen-Proline Dioxygenase/genetics , Protein Stability , Proteolysis
5.
Stud Health Technol Inform ; 183: 98-103, 2013.
Article in English | MEDLINE | ID: mdl-23388263

ABSTRACT

Understanding the impact of treatment policies on patient outcomes is essential in improving all aspects of patient care. The BC Cancer Agency is a provincial program that provides cancer care on a population basis for 4.5 million residents. The Lung and Head & Neck Tumour Groups planned to create a generic yet comprehensive software infrastructure that could be used by all Tumour Groups: the Outcomes and Surveillance Integration System (OaSIS). The primary goal was the development of an integrated database that will amalgamate existing provincial data warehouses of varying datasets and provide the infrastructure to support additional routes of data entry, including clinicians from multiple-disciplines, quality of life and survivorship data from patients, and three dimensional dosimetric information archived from the radiotherapy planning and delivery systems. The primary goal is to be able to capture any data point related to patient characteristics, disease factors, treatment details and survivorship, from the point of diagnosis onwards. Through existing and novel data-mining techniques, OaSIS will support unique population based research activities by promoting collaborative interactions between the research centre, clinical activities at the cancer treatment centres and other institutions. This will also facilitate initiatives to improve patient outcomes, decision support in achieving operational efficiencies and an environment that supports knowledge generation.


Subject(s)
Data Mining/methods , Database Management Systems , Databases, Factual , Information Storage and Retrieval/methods , Neoplasms/epidemiology , Registries , Software , Canada/epidemiology , Humans , Software Design
6.
Virology ; 405(1): 1-7, 2010 Sep 15.
Article in English | MEDLINE | ID: mdl-20580051

ABSTRACT

Autophagy is an important cellular process by which ATG5 initiates the formation of double membrane vesicles (DMVs). Upon infection, DMVs have been shown to harbor the replicase complex of positive-strand RNA viruses such as MHV, poliovirus, and equine arteritis virus. Recently, it has been shown that autophagy proteins are proviral factors that favor initiation of hepatitis C virus (HCV) infection. Here, we identified ATG5 as an interacting protein for the HCV NS5B. ATG5/NS5B interaction was confirmed by co-IP and metabolic labeling studies. Furthermore, ATG5 protein colocalizes with NS4B, a constituent of the membranous web. Importantly, immunofluorescence staining demonstrated a strong colocalization of ATG5 and NS5B within perinuclear regions of infected cells at 2 days postinfection. However, colocalization was completely lacking at 5DPI, suggesting that HCV utilizes ATG5 as a proviral factor during the onset of viral infection. Finally, inhibition of autophagy through ATG5 silencing blocks HCV replication.


Subject(s)
Hepacivirus/enzymology , Microtubule-Associated Proteins/metabolism , Viral Nonstructural Proteins/metabolism , Autophagy-Related Protein 5 , Cell Line , Gene Expression Regulation/physiology , Gene Silencing , Humans , Microtubule-Associated Proteins/chemistry , Microtubule-Associated Proteins/genetics , Protein Binding , Protein Structure, Tertiary , Two-Hybrid System Techniques , Viral Nonstructural Proteins/genetics , Virus Replication
7.
Cancer Cell ; 17(4): 319-32, 2010 Apr 13.
Article in English | MEDLINE | ID: mdl-20385358

ABSTRACT

High Gleason grade prostate carcinomas are aggressive, poorly differentiated tumors that exhibit diminished estrogen receptor beta (ERbeta) expression. We report that a key function of ERbeta and its specific ligand 5alpha-androstane-3beta,17beta-diol (3beta-adiol) is to maintain an epithelial phenotype and repress mesenchymal characteristics in prostate carcinoma. Stimuli (TGF-beta and hypoxia) that induce an epithelial-mesenchymal transition (EMT) diminish ERbeta expression, and loss of ERbeta is sufficient to promote an EMT. The mechanism involves ERbeta-mediated destabilization of HIF-1alpha and transcriptional repression of VEGF-A. The VEGF-A receptor neuropilin-1 drives the EMT by promoting Snail1 nuclear localization. Importantly, this mechanism is manifested in high Gleason grade cancers, which exhibit significantly more HIF-1alpha and VEGF expression, and Snail1 nuclear localization compared to low Gleason grade cancers.


Subject(s)
Estrogen Receptor beta/therapeutic use , Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Prostatic Neoplasms/prevention & control , Transcription Factors/physiology , Vascular Endothelial Growth Factor A/physiology , Epithelial Cells/cytology , Epithelial Cells/physiology , Estrogen Receptor beta/physiology , Humans , Male , Mesoderm/cytology , Mesoderm/physiology , Prostatic Neoplasms/pathology , Snail Family Transcription Factors , Transforming Growth Factor beta/physiology
8.
Biochem Biophys Res Commun ; 373(4): 618-23, 2008 Sep 05.
Article in English | MEDLINE | ID: mdl-18602367

ABSTRACT

Protein kinase D1 (PKD1), founding member of PKD protein family, is down-regulated in advanced prostate cancer (PCa). We demonstrate that PKD1 and androgen receptor (AR) are present as a protein complex in PCa cells. PKD1 is associated with a transcriptional complex which contains AR and promoter sequence of the Prostate Specific Antigen (PSA) gene. Ectopic expression of wild type PKD1 and the kinase dead mutant PKD1 (K628W) attenuated the ligand-dependent transcriptional activation of AR in prostate cancer cells and yeast cells indicating that PKD1 can affect AR transcription activity, whereas knocking down PKD1 enhanced the ligand-dependent transcriptional activation of AR. Co-expression of kinase dead mutant with AR significantly inhibited androgen-mediated cell proliferation in both LNCaP and DU145 PC cells. Our data demonstrate for the first time that PKD1 can influence AR function in PCa cells.


Subject(s)
Androgen Receptor Antagonists , Gene Expression Regulation, Neoplastic , Prostate-Specific Antigen/genetics , Prostatic Neoplasms/metabolism , Protein Kinase C/metabolism , Receptors, Androgen/metabolism , Cell Line, Tumor , Cell Proliferation , Humans , Male , Promoter Regions, Genetic , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Transcription, Genetic , Transcriptional Activation
9.
J Cell Biochem ; 104(1): 82-95, 2008 May 01.
Article in English | MEDLINE | ID: mdl-17979146

ABSTRACT

We have previously demonstrated that Protein Kinase D1 (PKD1) interacts with E-cadherin and is associated with altered cell aggregation and motility in prostate cancer (PC). Because both PKD1 and E-cadherin are known to be dysregulated in PC, in this study we investigated the functional consequences of combined dysregulation of PKD1 and E-cadherin using a panel of human PC cell lines. Gain and loss of function studies were carried out by either transfecting PC cells with full-length E-cadherin and/or PKD1 cDNA or by protein silencing by siRNAs, respectively. We studied major malignant phenotypic characteristics including cell proliferation, motility, and invasion at the cellular level, which were corroborated with appropriate changes in representative molecular markers. Down regulation or ectopic expression of either E-cadherin or PKD1 significantly increased or decreased cell proliferation, motility, and invasion, respectively, and combined down regulation cumulatively influenced the effects. Loss of PKD1 or E-cadherin expression was associated with increased expression of the pro-survival molecular markers survivin, beta-catenin, cyclin-D, and c-myc, whereas overexpression of PKD1 and/or E-cadherin resulted in an increase of caspases. The inhibitory effect of PKD1 and E-cadherin on cell proliferation was rescued by coexpression with beta-catenin, suggesting that beta-catenin mediates the effect of proliferation by PKD1 and E-cadherin. This study establishes the functional significance of combined dysregulation of PKD1 and E-cadherin in PC and that their effect on cell growth is mediated by beta-catenin.


Subject(s)
Cadherins/genetics , Cell Proliferation , Gene Expression Regulation, Neoplastic , Neoplasm Invasiveness/pathology , Prostatic Neoplasms/pathology , Protein Kinase C/genetics , beta Catenin/physiology , Apoptosis Regulatory Proteins , Cell Line, Tumor , Cell Movement , Humans , Male
10.
Neoplasia ; 8(11): 896-904, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17132221

ABSTRACT

Two flavonoids, genistein and apigenin, have been implicated as chemopreventive agents against prostate and breast cancers. However, the mechanisms behind their respective cancer-protective effects may vary significantly. The goal of this study was to determine whether the antiproliferative action of these flavonoids on prostate (DU-145) and breast (MDA-MB-231) cancer cells expressing only estrogen receptor (ER) beta is mediated by this ER subtype. It was found that both genistein and apigenin, although not 17beta-estradiol, exhibited antiproliferative effects and proapoptotic activities through caspase-3 activation in these two cell lines. In yeast transcription assays, both flavonoids displayed high specificity toward ERbeta transactivation, particularly at lower concentrations. However, in mammalian assay, apigenin was found to be more ERbeta-selective than genistein, which has equal potency in inducing transactivation through ERalpha and ERbeta. Small interfering RNA-mediated downregulation of ERbeta abrogated the antiproliferative effect of apigenin in both cancer cells but did not reverse that of genistein. Our data unveil, for the first time, that the anticancer action of apigenin is mediated, in part, by ERbeta. The differential use of ERalpha and ERbeta signaling for transaction between genistein and apigenin demonstrates the complexity of phytoestrogen action in the context of their anticancer properties.


Subject(s)
Apigenin/pharmacology , Estrogen Receptor beta/metabolism , Gene Expression Regulation, Neoplastic , Anticarcinogenic Agents/pharmacology , Caspase 3/metabolism , Cell Line, Tumor , Cell Survival , Enzyme Activation , Female , Humans , Male , Phytoestrogens/metabolism , RNA, Small Interfering/metabolism , Receptors, Estrogen/metabolism , Signal Transduction
11.
Proc Natl Acad Sci U S A ; 103(35): 13162-7, 2006 Aug 29.
Article in English | MEDLINE | ID: mdl-16938840

ABSTRACT

Estrogen receptor beta (ER-beta) regulates diverse physiological functions in the human body. Current studies are confined to ER-beta1, and the functional roles of isoforms 2, 4, and 5 remain unclear. Full-length ER-beta4 and -beta5 isoforms were obtained from a prostate cell line, and they exhibit differential expression in a wide variety of human tissues/cell lines. Through molecular modeling, we established that only ER-beta1 has a full-length helix 11 and a helix 12 that assumes an agonist-directed position. In ER-beta2, the shortened C terminus results in a disoriented helix 12 and marked shrinkage in the coactivator binding cleft. ER-beta4 and -beta5 completely lack helix 12. We further demonstrated that ER-beta1 is the only fully functional isoform, whereas ER-beta2, -beta4, and -beta5 do not form homodimers and have no innate activities of their own. However, the isoforms can heterodimerize with ER-beta1 and enhance its transactivation in a ligand-dependent manner. ER-beta1 tends to form heterodimers with other isoforms under the stimulation of estrogens but not phytoestrogens. Collectively, these data support the premise that (i) ER-beta1 is the obligatory partner of an ER-beta dimer, whereas the other isoforms function as variable dimer partners with enhancer activity, and (ii) a single functional helix 12 in a dimer is sufficient for gene transactivation. Thus, ER-beta behaves like a noncanonical type-I receptor, and its action may depend on differential amounts of ER-beta1 homo- and heterodimers formed upon stimulation by a specific ligand. Our findings have provided previously unrecognized directions for studying ER-beta signaling and design of ER-beta-based therapies.


Subject(s)
Estrogen Receptor beta/metabolism , Signal Transduction , Amino Acid Sequence , Cells, Cultured , Dimerization , Estrogen Receptor beta/chemistry , Estrogen Receptor beta/genetics , Gene Expression Profiling , Gene Expression Regulation , Humans , Membrane Proteins/genetics , Models, Molecular , Molecular Sequence Data , Presenilin-2 , Protein Binding , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sequence Analysis, Protein , Transcription, Genetic , Transcriptional Activation/genetics , Yeasts
12.
J Vet Med Sci ; 68(6): 543-8, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16820709

ABSTRACT

The surface glycoprotein G is considered as the major neutralizing and protective antigen of bovine ephemeral fever virus (BEFV). Comparison of the deduced amino acid sequence of G protein of BEFV isolates during the period 1984-2004 outbreaks in Taiwan showed amino acid substitutions in the neutralizing epitopes. All the isolates differ markedly in the neutralizing epitope at the same amino acid positions compared to the currently available killed vaccine strain (Tn73). Tn88128 strain isolated in 1999 showed the maximum variability of 12 amino acids, 5 amino acid in the neutralization epitope and 7 apart from, respectively. Combinations of both Tn88128 (1999) and commercially available vaccine strain (Tn73) were developed and its safety was evaluated in mice, guinea pigs, calves, and pregnant cows. None of the animals showed any adverse effect or clinical signs. Calves were immunized with commercial vaccine (Tn73) and, combined vaccine (Tn73 and Tn88128), respectively, with adjuvants such as Al-gel and water-in-oil-in-water (w/o/w) oil and PBS alone and challenged with Tn88128 strains. Except PBS administered animals, all the vaccinated animals showed protective immune response. However, animals immunized with combined vaccine plus w/o/w adjuvant elicited stronger neutralization antibodies and long lasting immunity compared to other vaccines.


Subject(s)
Emulsions/chemistry , Ephemeral Fever Virus, Bovine/genetics , Ephemeral Fever/prevention & control , Viral Proteins/genetics , Viral Proteins/immunology , Viral Vaccines/immunology , Amino Acid Sequence , Animals , Base Sequence , Cattle , DNA, Viral , Ephemeral Fever/immunology , Guinea Pigs , Mice , Molecular Sequence Data , Viral Proteins/chemistry , Viral Vaccines/adverse effects
13.
Virology ; 313(1): 158-69, 2003 Aug 15.
Article in English | MEDLINE | ID: mdl-12951030

ABSTRACT

A class of disulfide constrained peptides containing a core motif FPWG was identified from a screen of phage displayed library using the HCV RNA-dependent RNA polymerase (NS5B) as a bait. Surface plasmon resonance studies showed that three highly purified synthetic constrained peptides bound to immobilized NS5B with estimated K(d) values ranging from 30 to 60 microM. In addition, these peptides inhibited the NS5B activity in vitro with IC(50) ranging from 6 to 48 microM, whereas in contrast they had no inhibitory effect on the enzymatic activities of calf thymus polymerase alpha, human polymerase beta, RSV polymerase, and HIV reverse transcriptase in vitro. Two peptides demonstrated conformation-dependent inhibition since their synthetic linear versions were not inhibitory in the NS5B assay. A constrained peptide with the minimum core motif FPWG retained selective inhibition of NS5B activity with an IC(50) of 50 microM. Alanine scan analyses of a representative constrained peptide, FPWGNTW, indicated that residues F1 and W7 were critical for the inhibitory effect of this peptide, although residues P2 and N5 had some measurable inhibitory effect as well. Further analyses of the mechanism of inhibition indicated that these peptides inhibited the formation of preelongation complexes required for the elongation reaction. However, once the preelongation complex was formed, its activity was refractory to peptide inhibition. Furthermore, the constrained peptide FPWGNTW inhibited de novo initiated RNA synthesis by NS5B from a poly(rC) template. These data indicate that the peptides confer selective inhibition of NS5B activity by binding to the enzyme and perturbing an early step preceding the processive elongation step of RNA synthesis.


Subject(s)
Enzyme Inhibitors/pharmacology , Hepacivirus/enzymology , Peptides/pharmacology , RNA-Dependent RNA Polymerase/antagonists & inhibitors , Viral Nonstructural Proteins/antagonists & inhibitors , Disulfides , Peptide Library , Protein Binding , Protein Conformation , RNA-Dependent RNA Polymerase/metabolism , Viral Nonstructural Proteins/metabolism , Virus Replication
14.
J Biol Chem ; 277(41): 38838-46, 2002 Oct 11.
Article in English | MEDLINE | ID: mdl-12145289

ABSTRACT

The hepatitis C virus (HCV) encodes an RNA-dependent RNA polymerase (NS5B), which is indispensable for the viral genome replication. Although structural comparison among HCV NS5B, poliovirus 3D-pol, and human immunodeficiency virus-reverse transcriptase RNA-dependent polymerase reveals the canonical palm, fingers, and thumb domains, the crystal structure of HCV NS5B highlights the presence of a unique A1-loop, which extends from the fingers to the thumb domain (amino acids 12-46), providing many contact points for the proposed "closed" conformation of the enzyme. The polymerase also possesses a tunnel, which starts at the active site and terminates on the back surface of the enzyme. This tunnel of 19 A contains five basic amino acids, which may be engaged in NTP trafficking. In the present study, we exploited the crystal structure of the enzyme to elucidate the involvement of these two structural motifs in enzyme activity by site-directed mutagenesis. As predicted, the replacement of leucine 30 located in the Lambda 1-loop is detrimental to the NS5B activity. Heparin-Sepharose column chromatography and analytical ultracentrifugation experiments strongly suggest a local alteration in the structure of the Leu-30 mutant. An analysis of amino acid substitutions in Arg-222 and Lys-151 within the putative NTP tunnel indicates that Arg-222 was critical in delivering NTPs to the active site, whereas Lys-151 was dispensable. Interestingly, the substitution of lysine 151 for a glutamic acid resulted in an enzyme that was consistently more active in de novo synthesis as well as by "copy-back" mechanism of a self-primed substrate when compared with the wild type NS5B enzyme. Burst kinetic analyses indicate that the gain in function of K151E enzyme was primarily the result of the formation of more productive pre-initiation complexes that were used for the elongation reaction. In contrast to the recent observations, both the wild type and mutant enzymes were monomeric in solution, whereas molecules of higher order were apparent in the presence of RNA template.


Subject(s)
Protein Structure, Tertiary , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/metabolism , Base Sequence , Humans , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Nucleotides/metabolism , RNA-Dependent RNA Polymerase/chemistry , RNA-Dependent RNA Polymerase/genetics , RNA-Dependent RNA Polymerase/metabolism , Structure-Activity Relationship , Viral Nonstructural Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...