Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
Methods Mol Biol ; 1805: 233-250, 2018.
Article in English | MEDLINE | ID: mdl-29971721

ABSTRACT

Over the recent years single-molecule fluorescence resonance energy transfer (smFRET) technique has proven to be one of the most powerful tools for revealing mechanistic insights into helicase activities. Here we describe details of single-molecule FRET assays for probing DNA unwinding activities as well as functional dynamics by replicative helicases in real time. The ability of smFRET to measure the behavior of biomolecules at a nanometer scale enabled us to address how the leading and lagging strand synthesis are coordinated during DNA replication, to resolve DNA unwinding steps of Bacteriophage T7 helicase, and to observe heterogeneous unwinding patterns modulated by the DNA binding domain of E1 helicase. These single-molecule FRET assays are generally applicable to other replicative and nonreplicative hexameric helicases.


Subject(s)
DNA Helicases/metabolism , DNA Replication , Fluorescence Resonance Energy Transfer/methods , Single Molecule Imaging , Bacteriophage T7/enzymology , DNA/chemistry , DNA-Directed DNA Polymerase/metabolism , Multienzyme Complexes/metabolism , Papillomaviridae/enzymology , Polyethylene Glycols/chemistry
2.
Int J Mol Sci ; 17(7)2016 Jul 14.
Article in English | MEDLINE | ID: mdl-27428950

ABSTRACT

Bladder cancer is the second most common urological malignancy in the world. In 70% of cases it is initially diagnosed as non-muscle-invasive bladder cancer (NMIBC) and it is amenable to local treatments, with intravesical (IVES) Bacillus-Calmette-Guerin (BCG) immunotherapy being routinely used after transurethral resection of the lesion. However, this treatment is associated with significant side-effects and treatment failures, highlighting the necessity of novel strategies. One potent approach is the suicide-gene mediated therapy/prodrug combination, provided tumor-specificity can be ensured and anti-tumor immune responses induced. Using the mouse syngeneic orthotopic MB49-bladder tumor model, here we show that IVES human papillomavirus non-replicative pseudovirions (PsV) can pseudoinfect tumors with a ten-fold higher efficacy than normal bladders. In addition, PsV carrying the suicide-gene herpes-simplex virus thymidine kinase (PsV-TK) combined to Ganciclovir (GCV) led to immunogenic cell-death of tumor cells in vitro and to MB49-specific CD8 T-cells in vivo. This was associated with reduction in bladder-tumor growth and increased mice survival. Altogether, our data show that IVES PsV-TK/GCV may be a promising alternative or combinatory treatment for NMIBC.


Subject(s)
Ganciclovir/therapeutic use , Genetic Therapy , Papillomaviridae/genetics , Papillomaviridae/immunology , Thymidine Kinase/metabolism , Urinary Bladder Neoplasms/therapy , Animals , Antiviral Agents/therapeutic use , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Combined Modality Therapy , Female , Genetic Vectors , Humans , Mice , Mice, Inbred C57BL , Papillomaviridae/enzymology , Thymidine Kinase/genetics , Urinary Bladder Neoplasms/enzymology , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/immunology
3.
Nucleic Acids Res ; 43(17): 8551-63, 2015 Sep 30.
Article in English | MEDLINE | ID: mdl-26240379

ABSTRACT

Hexameric helicases are processive DNA unwinding machines but how they engage with a replication fork during unwinding is unknown. Using electron microscopy and single particle analysis we determined structures of the intact hexameric helicase E1 from papillomavirus and two complexes of E1 bound to a DNA replication fork end-labelled with protein tags. By labelling a DNA replication fork with streptavidin (dsDNA end) and Fab (5' ssDNA) we located the positions of these labels on the helicase surface, showing that at least 10 bp of dsDNA enter the E1 helicase via a side tunnel. In the currently accepted 'steric exclusion' model for dsDNA unwinding, the active 3' ssDNA strand is pulled through a central tunnel of the helicase motor domain as the dsDNA strands are wedged apart outside the protein assembly. Our structural observations together with nuclease footprinting assays indicate otherwise: strand separation is taking place inside E1 in a chamber above the helicase domain and the 5' passive ssDNA strands exits the assembly through a separate tunnel opposite to the dsDNA entry point. Our data therefore suggest an alternative to the current general model for DNA unwinding by hexameric helicases.


Subject(s)
DNA Helicases/chemistry , DNA/chemistry , DNA/metabolism , DNA Helicases/metabolism , DNA Helicases/ultrastructure , DNA Replication , DNA, Single-Stranded/chemistry , DNA, Single-Stranded/metabolism , DNA, Single-Stranded/ultrastructure , Models, Molecular , Papillomaviridae/enzymology , Protein Structure, Tertiary
4.
Methods Mol Biol ; 1249: 67-80, 2015.
Article in English | MEDLINE | ID: mdl-25348298

ABSTRACT

Replication of the human papillomavirus (HPV) double-stranded DNA genome in the nucleus of infected cells relies on the viral proteins E1 and E2 in conjunction with the host DNA replication machinery. This process is tightly linked to the replication of cellular DNA, in part through the cyclin-dependent phosphorylation of E1, which inhibits its export out of the nucleus to promote its accumulation in this compartment during S-phase. It has been recently shown that accumulation of E1 in the nucleus, while a prerequisite for viral DNA replication, leads to the inhibition of cellular proliferation and the activation of a DNA damage response (DDR). Here we describe methods to monitor the subcellular localization of E1 and to assess the deleterious effects of its nuclear accumulation on cellular proliferation, cell cycle progression and the induction of a DDR, using a combination of colony formation assays, immunofluorescence microcopy, and flow cytometry approaches.


Subject(s)
Cell Nucleus/metabolism , DNA Damage , DNA Helicases/metabolism , Molecular Biology/methods , Papillomaviridae/enzymology , Active Transport, Cell Nucleus , Amino Acid Sequence , Amino Acid Substitution , Bromodeoxyuridine/metabolism , Cell Line, Tumor , Cell Proliferation , DNA Helicases/chemistry , Histones/metabolism , Humans , Molecular Sequence Data , Phosphorylation , Protein Transport , Subcellular Fractions/enzymology
5.
J Virol ; 89(2): 1129-42, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25378487

ABSTRACT

UNLABELLED: Viruses frequently combine multiple activities into one polypeptide to conserve coding capacity. This strategy creates regulatory challenges to ascertain that the combined activities are compatible and do not interfere with each other. The papillomavirus E1 protein, as many other helicases, has the intrinsic ability to form hexamers and double hexamers (DH) that serve as the replicative DNA helicase. However, E1 also has the more unusual ability to generate local melting by forming a double trimer (DT) complex that can untwist the double-stranded origin of DNA replication (ori) DNA in preparation for DH formation. Here we describe a switching mechanism that allows the papillomavirus E1 protein to form these two different kinds of oligomers and to transition between them. We show that a conserved regulatory module attached to the E1 helicase domain blocks hexamer and DH formation and promotes DT formation. In the presence of the appropriate trigger, the inhibitory effect of the regulatory module is relieved and the transition to DH formation can occur. IMPORTANCE: This study provides a mechanistic understanding into how a multifunctional viral polypeptide can provide different, seemingly incompatible activities. A conserved regulatory sequence module attached to the AAA+ helicase domain in the papillomavirus E1 protein allows the formation of different oligomers with different biochemical activities.


Subject(s)
DNA Helicases/genetics , DNA Helicases/metabolism , Gene Expression Regulation, Viral , Papillomaviridae/enzymology , Papillomaviridae/genetics , Protein Multimerization , Conserved Sequence , Protein Structure, Tertiary
6.
Virology ; 445(1-2): 35-56, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24029589

ABSTRACT

E1, an ATP-dependent DNA helicase, is the only enzyme encoded by papillomaviruses (PVs). It is essential for replication and amplification of the viral episome in the nucleus of infected cells. To do so, E1 assembles into a double-hexamer at the viral origin, unwinds DNA at the origin and ahead of the replication fork and interacts with cellular DNA replication factors. Biochemical and structural studies have revealed the assembly pathway of E1 at the origin and how the enzyme unwinds DNA using a spiral escalator mechanism. E1 is tightly regulated in vivo, in particular by post-translational modifications that restrict its accumulation in the nucleus. Here we review how different functional domains of E1 orchestrate viral DNA replication, with an emphasis on their interactions with substrate DNA, host DNA replication factors and modifying enzymes. These studies have made E1 one of the best characterized helicases and provided unique insights on how PVs usurp different host-cell machineries to replicate and amplify their genome in a tightly controlled manner.


Subject(s)
DNA Helicases/metabolism , DNA Replication , Genome, Viral , Oncogene Proteins, Viral/metabolism , Papillomaviridae/enzymology , Virus Replication , Cell Nucleus/virology , DNA Helicases/genetics , DNA, Viral/genetics , Enzyme Activation , Humans , Oncogene Proteins, Viral/genetics , Papillomaviridae/genetics , Papillomaviridae/physiology , Protein Multimerization , Protein Processing, Post-Translational , Protein Structure, Tertiary
7.
J Otolaryngol Head Neck Surg ; 41 Suppl 1: S65-70, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22569052

ABSTRACT

BACKGROUND: Epigenetic modifications are defined as heritable changes in gene expression that are not encoded in deoxyribonucleic acid (DNA). Despite the importance of epigenetics in tumorigenesis, there is a paucity of information regarding the epigenetic profiles of oropharyngeal squamous cell carcinoma (OPSCC). OBJECTIVE: The objective of this study was to identify epigenetic signatures associated with human papillomavirus (HPV)-positive and -negative OPSCC. METHODS: We collected demographic, pathologic, and survival data from 44 patients with advanced-stage OPSCC treated with surgery and chemoradiation at the University of Alberta between January 2006 and December 2008. Tumour specimen from these patients were retreived and sectioned for immunohistochemical analysis. Double immunofluorescence staining was performed with p16 (HPV surrogate) and a panel of epigenetic markers, namely, histone methyl-lysines 4, 9, and 27 and H4 methyl-lysine 20. Correlation between p16 and epigenetic markers was measured using Metamorph and Image J software. RESULTS: Forty-one percent of patients were p16 positive. No statistically significant differences were found between p16-positive and -negative patients in terms of age at diagnosis, tumour subsite, or smoking history. We found significant differences in histone methylation between p16-positive and -negative tumours. OPSCC tumours positive for p16 had global elevations of histone H4 monomethylated lysine 20 (H4K20me1) and H3 trimethylated lysine 27 (H3K27me3) with depletions of H4 trimethylated lysine 20 (H4K20me3). In contrast, p16-negative tumours had depleted levels of H4K20me1 and H3K27me3 with high levels of H4K20me3. CONCLUSIONS: HPV-positive and -negative OPSCCs have distinct epigenetic profiles representing broad gene expression differences between these tumours.


Subject(s)
Carcinoma, Squamous Cell/virology , Cyclin-Dependent Kinase Inhibitor p16/genetics , DNA, Viral/analysis , Epigenomics/methods , Oropharyngeal Neoplasms/virology , Papillomaviridae/genetics , Papillomavirus Infections/virology , Biomarkers, Tumor/analysis , Biomarkers, Tumor/genetics , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Cyclin-Dependent Kinase Inhibitor p16/biosynthesis , Female , Genetic Markers , Humans , Immunohistochemistry , Male , Middle Aged , Neoplasm Staging , Oropharyngeal Neoplasms/genetics , Oropharyngeal Neoplasms/pathology , Papillomaviridae/enzymology , Papillomavirus Infections/genetics , Papillomavirus Infections/pathology , Prognosis , Retrospective Studies
8.
Mol Cell ; 43(5): 776-87, 2011 Sep 02.
Article in English | MEDLINE | ID: mdl-21884978

ABSTRACT

Preparation of DNA templates for replication requires opening of the duplex to expose single-stranded (ss) DNA. The locally melted DNA is required for replicative DNA helicases to initiate unwinding. How local melting is generated in eukaryotic replicons is unknown, but initiator proteins from a handful of eukaryotic viruses can perform this function. Here we dissect the local melting process carried out by the papillomavirus E1 protein. We characterize the melting process kinetically and identify mutations in the E1 helicase and in the ori that arrest the local melting process. We show that a subset of these mutants have specific defects for melting of the center of the ori containing the binding sites for E1 and demonstrate that these mutants fail to untwist the ori DNA. This understanding of how E1 generates local melting suggests possible mechanisms for local melting in other replicons.


Subject(s)
DNA Helicases/chemistry , DNA, Viral/chemistry , DNA-Binding Proteins/chemistry , Papillomaviridae/enzymology , Replication Origin , Viral Proteins/chemistry , Binding Sites , DNA Helicases/genetics , DNA Replication , DNA-Binding Proteins/genetics , Models, Molecular , Mutation , Protein Conformation , Viral Proteins/genetics
10.
Oncogene ; 27(12): 1800-4, 2008 Mar 13.
Article in English | MEDLINE | ID: mdl-17934525

ABSTRACT

Human papillomavirus (HPV) type 16 and 18 E6 proteins target many of their cellular substrates for proteasome-mediated degradation. In the case of p53, this is mediated by the E6AP ubiquitin ligase. However it is still unclear whether other E6 substrates, in particular those containing PDZ domains, are also degraded in a similar manner. To investigate this, we established an epithelial cell line from E6AP-null mice and used these cells as a background to perform E6-mediated in vivo degradation assays. We show that the PDZ domain-containing substrates of E6, including Scribble, MAGI-1 and MAGI-3, are all subject to E6-mediated degradation in these cells. Strikingly, we also found that p53 could be degraded by E6 within these cells in a proteasome-dependent manner. These results demonstrate that HPV-16 and -18 E6 can target substrates for degradation in a manner independent of the E6AP ubiquitin ligase.


Subject(s)
DNA-Binding Proteins/physiology , Oncogene Proteins, Viral/physiology , PDZ Domains/physiology , Papillomaviridae/physiology , Repressor Proteins/physiology , Tumor Suppressor Protein p53/metabolism , Ubiquitin-Protein Ligases/deficiency , Ubiquitin-Protein Ligases/genetics , Cell Line , DNA-Binding Proteins/genetics , Humans , Oncogene Proteins, Viral/genetics , PDZ Domains/genetics , Papillomaviridae/enzymology , Repressor Proteins/genetics , Substrate Specificity/genetics
11.
J Virol ; 81(1): 384-94, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17035309

ABSTRACT

The papillomavirus E1 protein is essential for the initiation of viral replication. We previously showed that the bovine papillomavirus E1 protein is unstable and becomes resistant to ubiquitin-mediated degradation when tightly bound to cyclin E-cyclin-dependent kinase 2 (Cdk2) before the start of DNA synthesis. However, neither the protection nor the targeted degradation of E1 appears to depend on its phosphorylation by Cdk. Here, we report that Cdk phosphorylation of E1 is also not a prerequisite for the initiation of viral DNA replication either in vitro or in vivo. Nevertheless, we found that phosphorylation of one Cdk site, Ser283, abrogates E1 replicative activity only in a cellular context. We show that this site-specific phosphorylation of E1 drives its export from the nucleus and promotes its continuous nucleocytoplasmic shuttling. In addition, we find that E1 shuttling occurs in S phase, when cyclin A-Cdk2 is activated. E1 interacts with the active cyclin A-Cdk2 complex and is phosphorylated on Ser283 by this kinase. These data suggest that the phosphorylation of E1 on Ser283 is a negative regulatory event that is involved in preventing the amplification of viral DNA during S phase. This finding reveals a novel facet of E1 regulation that could account for the variations of the viral replication capacity during different cell cycle phases, as well as in different stages of the viral cycle.


Subject(s)
Cell Nucleus/virology , DNA Helicases/metabolism , DNA Replication/physiology , DNA, Viral/biosynthesis , DNA-Binding Proteins/metabolism , Down-Regulation , Papillomaviridae/enzymology , S Phase , Viral Proteins/metabolism , Virus Replication/physiology , Active Transport, Cell Nucleus , Animals , Cyclin-Dependent Kinases/metabolism , Cytoplasm/virology , DNA Helicases/chemistry , DNA-Binding Proteins/chemistry , Papillomaviridae/genetics , Papillomaviridae/physiology , Phosphorylation , Protein Transport , Viral Proteins/chemistry , Xenopus
13.
Curr Pharm Des ; 12(11): 1315-38, 2006.
Article in English | MEDLINE | ID: mdl-16611118

ABSTRACT

Helicases are promising antiviral drug targets because their enzymatic activities are essential for viral genome replication, transcription, and translation. Numerous potent inhibitors of helicases encoded by herpes simplex virus, severe acute respiratory syndrome coronavirus, hepatitis C virus, Japanese encephalitis virus, West Nile virus, and human papillomavirus have been recently reported in the scientific literature. Some inhibitors have also been shown to decrease viral replication in cell culture and animal models. This review discusses recent progress in understanding the structure and function of viral helicases to help clarify how these potential antiviral compounds function and to facilitate the design of better inhibitors. The above helicases and all related viral proteins are classified here based on their evolutionary and functional similarities, and the key mechanistic features of each group are noted. All helicases share a common motor function fueled by ATP hydrolysis, but differ in exactly how the motor moves the protein and its cargo on a nucleic acid chain. The helicase inhibitors discussed here influence rates of helicase-catalyzed DNA (or RNA) unwinding by preventing ATP hydrolysis, nucleic acid binding, nucleic acid release, or by disrupting the interaction of a helicase with a required cofactor.


Subject(s)
Antiviral Agents/pharmacology , DNA Helicases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , RNA Helicases/antagonists & inhibitors , DNA Helicases/chemistry , DNA Helicases/physiology , DNA Primase/antagonists & inhibitors , Papillomaviridae/drug effects , Papillomaviridae/enzymology , RNA Helicases/chemistry , RNA Helicases/physiology , Severe acute respiratory syndrome-related coronavirus/drug effects , Severe acute respiratory syndrome-related coronavirus/enzymology , Serine Endopeptidases , Simplexvirus/drug effects , Simplexvirus/enzymology , Viral Nonstructural Proteins/antagonists & inhibitors , Viral Proteins/antagonists & inhibitors
14.
Biochemistry ; 45(3): 657-67, 2006 Jan 24.
Article in English | MEDLINE | ID: mdl-16411741

ABSTRACT

E7 oncoprotein is the major transforming activity in human papillomavirus and shares sequence and functional properties with adenovirus E1A and SV40 T-antigen, in particular by targeting the pRb tumor suppressor. HPV 16 E7 forms spherical oligomers that display chaperone activity in thermal denaturation and chemical refolding assays of two model polypeptide substrates, citrate synthase and luciferase, and it does so at substoichiometric concentrations. We show that the E7 chaperone can stably bind model polypeptides and hold them in a state with significant tertiary structure, but does not bind the fully native proteins. The E7 oligomers bind native in vitro translated pRb without the requirement of it being unfolded, since the N-terminal domain of E7 containing the LXCXE binding motif is exposed. The N-terminal domain of E7 can interfere with pRb binding but not with the chaperone activity, which requires the C-terminal domain, as in most reported E7 activities. The ability to bind up to approximately 72 molecules of pRb by the oligomeric E7 form could be important either for sequestering pRb from Rb-E2F complexes or for targeting it for proteasome degradation. Thus, both the dimeric and oligomeric chaperone forms of E7 can bind Rb and various potential targets. We do not know at present if the chaperone activity of E7 plays an essential role in the viral life cycle; however, a chaperone activity may explain the large number of cellular targets reported for this oncoprotein.


Subject(s)
Molecular Chaperones/metabolism , Oncogene Proteins, Viral/metabolism , Papillomaviridae/enzymology , Kinetics , Models, Molecular , Molecular Chaperones/genetics , Oncogene Proteins, Viral/genetics , Papillomavirus E7 Proteins , Protein Conformation , Protein Denaturation
15.
Antimicrob Agents Chemother ; 49(12): 4834-42, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16304143

ABSTRACT

Human papillomaviruses (HPVs) are the causative agents of benign and malignant lesions of the epithelium. Despite their high prevalence, there is currently no antiviral drug for the treatment of HPV-induced lesions. The ATPase and helicase activities of the highly conserved E1 protein of HPV are essential for viral DNA replication and pathogenesis and hence are considered valid antiviral targets. We recently described novel biphenylsulfonacetic acid inhibitors of the ATPase activity of E1 from HPV type 6 (HPV6). Based on kinetics and mutagenesis studies, we now report that these compounds act by an allosteric mechanism. They are hyperbolic competitive inhibitors of the ATPase activity of HPV6 E1 and also inhibit its helicase activity. Compounds in this series can also inhibit the ATPase activity of the closely related enzyme from HPV11; however, the most potent inhibitors of HPV6 E1 are significantly less active against the type 11 protein. We identified a single critical residue in HPV6 E1, Tyr-486, substituted by a cysteine in HPV11, which is primarily responsible for this difference in inhibitor potency. Interestingly, HPV18 E1, which also has a tyrosine at this position, could be inhibited by biphenylsulfonacetic acid derivatives, thereby raising the possibility that this class of inhibitors could be optimized as antiviral agents against multiple HPV types. These studies implicate Tyr-486 as a key residue for inhibitor binding and define an allosteric pocket on HPV E1 that can be exploited for future drug discovery efforts.


Subject(s)
Acetates/pharmacology , Adenosine Triphosphate/metabolism , Biphenyl Compounds/pharmacology , Enzyme Inhibitors/pharmacology , Oncogene Proteins, Viral/antagonists & inhibitors , Sulfones/pharmacology , Tyrosine/metabolism , Allosteric Regulation , Biphenyl Compounds/chemistry , Humans , Hydrolysis , Oncogene Proteins, Viral/metabolism , Papillomaviridae/enzymology , Structure-Activity Relationship
16.
EMBO J ; 24(10): 1821-30, 2005 May 18.
Article in English | MEDLINE | ID: mdl-15861133

ABSTRACT

The E7 proteins of human papillomaviruses (HPVs) contribute to oncogenesis by associating with Rb family members as well class I histone deacetylases (HDACs). The binding of HDACs is also important for the maintenance of viral episomes during the differentiation-dependent productive life cycle. The effects of E7 and other viral proteins on E2F family members were examined in differentiating keratinocytes. E7 was found to specifically activate E2F2 transcription in suprabasal keratinocytes through its ability to bind HDACs. Chromatin immunoprecipitation assays demonstrated that, in differentiating cells, E7 acts to inhibit HDAC binding to the E2F2 promoter resulting in activation of expression. Reduction of E2F2 levels through the use of siRNA confirmed that E2F2 expression facilitated HPV replication but its loss did not affect cell proliferation. Our study demonstrates a mechanism by which binding of HDACs to E7 directly modulates viral replication and identifies E2F2 as a possible target for antiviral therapies.


Subject(s)
Histone Deacetylases/metabolism , Oncogene Proteins, Viral/metabolism , Transcription Factors/genetics , E2F2 Transcription Factor , Epithelial Cells/enzymology , Epithelial Cells/virology , Humans , Keratinocytes/enzymology , Keratinocytes/virology , Papillomaviridae/enzymology , Papillomaviridae/genetics , Papillomavirus E7 Proteins , Promoter Regions, Genetic , Transcription Factors/biosynthesis , Transcription, Genetic/physiology , Up-Regulation
17.
J Laryngol Otol ; 118(10): 750-6, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15550179

ABSTRACT

OBJECTIVE: Determination of early prognostic factors in patients with recurrent respiratory papillomatosis is extremely important, so the major goal of our prospective, multicentre study was to evaluate (1) the feasibility of various factors to determine prognosis of the clinical course, as well as (2) the response to interferon-alpha therapy in recurrent respiratory papillomatosis. METHODS: Forty-two patients with recurrent respiratory papillomatosis were treated with interferon-alpha (3 MU/m(2) three times per week; mean therapy duration was 2.7 +/- 1.8 years) in 1983-1994 and followed-up until 2003. Human papilloma virus (HPV) type, recurrent respiratory papillomatosis severity and 2',5'-oligoadenylate synthetase activity were determined by standard methods and analysed for correlation with the results of long-term clinical outcome. RESULTS AND CONCLUSION: Patients with HPV type 11, a severity score >4, a high number of surgical procedures prior to interferon-alpha therapy and a high basal 2',5'-oligoadenylate synthetase activity should be considered at high risk of an aggressive clinical course, often with spread to lower airway passages, malignant transformation and death. Human papilloma virus type, score for recurrent respiratory papillomatosis severity, number of surgical procedures and 2',5'-oligoadenylate synthetase activity showed significant association with response to interferon-alpha therapy and the long-term clinical course, so these factors have value in predicting prognosis in recurrent respiratory papillomatosis.


Subject(s)
2',5'-Oligoadenylate Synthetase/analysis , Papilloma/enzymology , Papillomaviridae/enzymology , Respiratory Tract Neoplasms/enzymology , Adolescent , Adult , Antineoplastic Agents/therapeutic use , Child , Child, Preschool , DNA, Viral/analysis , Female , Humans , Interferon-alpha/therapeutic use , Leukocytes, Mononuclear/enzymology , Male , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/enzymology , Papilloma/drug therapy , Papillomaviridae/classification , Prognosis , Prospective Studies , Respiratory Tract Neoplasms/drug therapy , Severity of Illness Index , Treatment Outcome
18.
Genes Dev ; 18(16): 1981-96, 2004 Aug 15.
Article in English | MEDLINE | ID: mdl-15289463

ABSTRACT

DNA replication of the papillomaviruses is specified by cooperative binding of two proteins to the ori site: the enhancer E2 and the viral initiator E1, a distant member of the AAA+ family of proteins. Formation of this prereplication complex is an essential step toward the construction of a functional, multimeric E1 helicase and DNA melting. To understand how E2 interacts with E1 to regulate this process, we have solved the X-ray structure of a complex containing the HPV18 E2 activation domain bound to the helicase domain of E1. Modeling the monomers of E1 to a hexameric helicase shows that E2 blocks hexamerization of E1 by shielding a region of the E1 oligomerization surface and stabilizing a conformation of E1 that is incompatible with ATP binding. Further biochemical experiments and structural analysis show that ATP is an allosteric effector of the dissociation of E2 from E1. Our data provide the first molecular insights into how a protein can regulate the assembly of an oligomeric AAA+ complex and explain at a structural level why E2, after playing a matchmaker role by guiding E1 to the DNA, must dissociate for subsequent steps of initiation to occur. Building on previously proposed ideas, we discuss how our data advance current models for the conversion of E1 in the prereplication complex to a hexameric helicase assembly.


Subject(s)
DNA Helicases/chemistry , Papillomaviridae/enzymology , Amino Acid Sequence , Crystallography, X-Ray , Models, Molecular , Molecular Sequence Data , Protein Conformation , Sequence Homology, Amino Acid , Viral Proteins/chemistry
19.
Oncogene ; 23(17): 2988-95, 2004 Apr 15.
Article in English | MEDLINE | ID: mdl-14968115

ABSTRACT

Human papillomavirus (HPV) 16 is involved in causing cervical cancer. The E6 and E7 proteins of HPV 16 immortalize human keratinocytes and this is due, at least in part, to inactivation of the tumor suppressor proteins p53 and pRB. These tumor suppressor proteins also regulate the expression of pro- and antiangiogenic factors by cells. For this reason, experiments were conducted to determine whether the expression of E6 and E7 in primary keratinocytes alters the phenotype of these cells such that they express diminished levels of antiangiogenic factors and/or increased levels of proangiogenic factors. To avoid variances in experimental observations, pools of human foreskin keratinocytes from multiple sources were infected with recombinant retrovirus expressing HPV 16 E6 and E7 or control retrovirus. Gene array analysis, RT-PCR, ELISAs and Western blotting showed that in cells expressing HPV 16 E6 and E7, expression levels of two potent angiogenesis inhibitors, thrombospondin-1 and maspin, were lower compared to controls. Additionally, major angiogenesis inducers, interleukin-8 and vascular endothelial growth factor (VEGF), were increased relative to controls. VEGF can be produced as multiple splice variants, all of which are required for the formation of patent blood vessels. The expression of HPV 16 E6 and E7 in keratinocytes augmented expression of VEGF 121, 145, 165 and 189. These observations show that HPV 16 E6 and E7 alter the phenotype of primary keratinocytes, diminishing expression of inhibitors and increasing expression of inducers of angiogenesis. This altered phenotype may permit keratinocytes infected by HPV 16 to play a role in the progression of cancer by permitting tumors to acquire a blood supply permissive of growth and spread.


Subject(s)
Keratinocytes/virology , Oncogene Proteins, Viral/genetics , Protein-Tyrosine Kinases/genetics , Repressor Proteins , Cells, Cultured , Humans , Infant, Newborn , Interleukin-8/metabolism , Male , Oligonucleotide Array Sequence Analysis , Oncogene Proteins, Viral/metabolism , Papillomaviridae/enzymology , Papillomaviridae/genetics , Papillomavirus E7 Proteins , Protein-Tyrosine Kinases/metabolism , Recombinant Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Skin/virology , Transfection
20.
J Med Chem ; 47(1): 18-21, 2004 Jan 01.
Article in English | MEDLINE | ID: mdl-14695816

ABSTRACT

The Boehringer Ingelheim compound collection was screened for inhibitors of the ATPase activity of human papillomavirus E1 helicase to develop antiviral agents that inhibit human papillomavirus (HPV) DNA replication. This screen led to the discovery of (biphenyl-4-sulfonyl)acetic acid 1, which inhibits the ATPase activity of HPV type 6 E1 helicase with a low micromolar IC(50) value. A hit-to-lead exercise rapidly converted 1 into a low nanomolar lead series.


Subject(s)
Acetates/chemical synthesis , Adenosine Triphosphatases/antagonists & inhibitors , Biphenyl Compounds/chemical synthesis , Oncogene Proteins, Viral/antagonists & inhibitors , Papillomaviridae/enzymology , Sulfones/chemical synthesis , Acetates/chemistry , Adenosine Triphosphatases/chemistry , Antiviral Agents/chemical synthesis , Antiviral Agents/chemistry , Biphenyl Compounds/chemistry , Humans , Oncogene Proteins, Viral/chemistry , Structure-Activity Relationship , Sulfones/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...