101
|
Chi CN, Bach A, Engström Å, Strømgaard K, Lundström P, Ferguson N, Jemth P. Biophysical characterization of the complex between human papillomavirus E6 protein and synapse-associated protein 97. J Biol Chem 2010; 286:3597-606. [PMID: 21113079 DOI: 10.1074/jbc.m110.190264] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The E6 protein of human papillomavirus (HPV) exhibits complex interaction patterns with several host proteins, and their roles in HPV-mediated oncogenesis have proved challenging to study. Here we use several biophysical techniques to explore the binding of E6 to the three PDZ domains of the tumor suppressor protein synapse-associated protein 97 (SAP97). All of the potential binding sites in SAP97 bind E6 with micromolar affinity. The dissociation rate constants govern the different affinities of HPV16 and HPV18 E6 for SAP97. Unexpectedly, binding is not mutually exclusive, and all three PDZ domains can simultaneously bind E6. Intriguingly, this quaternary complex has the same apparent hydrodynamic volume as the unliganded PDZ region, suggesting that a conformational change occurs in the PDZ region upon binding, a conclusion supported by kinetic experiments. Using NMR, we discovered a new mode of interaction between E6 and PDZ: a subset of residues distal to the canonical binding pocket in the PDZ(2) domain exhibited noncanonical interactions with the E6 protein. This is consistent with a larger proportion of the protein surface defining binding specificity, as compared with that reported previously.
Collapse
Affiliation(s)
- Celestine N Chi
- Department of Medical Biochemistry and Microbiology, Uppsala University, SE-75123 Uppsala, Sweden
| | | | | | | | | | | | | |
Collapse
|
102
|
Fu L, Van Doorslaer K, Chen Z, Ristriani T, Masson M, Travé G, Burk RD. Degradation of p53 by human Alphapapillomavirus E6 proteins shows a stronger correlation with phylogeny than oncogenicity. PLoS One 2010; 5. [PMID: 20862247 PMCID: PMC2941455 DOI: 10.1371/journal.pone.0012816] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2010] [Accepted: 08/23/2010] [Indexed: 11/25/2022] Open
Abstract
Background Human Papillomavirus (HPV) E6 induced p53 degradation is thought to be an essential activity by which high-risk human Alphapapillomaviruses (alpha-HPVs) contribute to cervical cancer development. However, most of our understanding is derived from the comparison of HPV16 and HPV11. These two viruses are relatively distinct viruses, making the extrapolation of these results difficult. In the present study, we expand the tested strains (types) to include members of all known HPV species groups within the Alphapapillomavirus genus. Principal Findings We report the biochemical activity of E6 proteins from 27 HPV types representing all alpha-HPV species groups to degrade p53 in human cells. Expression of E6 from all HPV types epidemiologically classified as group 1 carcinogens significantly reduced p53 levels. However, several types not associated with cancer (e.g., HPV53, HPV70 and HPV71) were equally active in degrading p53. HPV types within species groups alpha 5, 6, 7, 9 and 11 share a most recent common ancestor (MRCA) and all contain E6 ORFs that degrade p53. A unique exception, HPV71 E6 ORF that degraded p53 was outside this clade and is one of the most prevalent HPV types infecting the cervix in a population-based study of 10,000 women. Alignment of E6 ORFs identified an amino acid site that was highly correlated with the biochemical ability to degrade p53. Alteration of this amino acid in HPV71 E6 abrogated its ability to degrade p53, while alteration of this site in HPV71-related HPV90 and HPV106 E6s enhanced their capacity to degrade p53. Conclusions These data suggest that the alpha-HPV E6 proteins' ability to degrade p53 is an evolved phenotype inherited from a most recent common ancestor of the high-risk species that does not always segregate with carcinogenicity. In addition, we identified an amino-acid residue strongly correlated with viral p53 degrading potential.
Collapse
Affiliation(s)
- Leiping Fu
- Department of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Koenraad Van Doorslaer
- Department of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Zigui Chen
- Department of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Tutik Ristriani
- Équipe Oncoprotéine, UMR CNRS 7100, École Supérieure de Biotechnologie de Strasbourg, Illkirch, France
| | - Murielle Masson
- Équipe Oncoprotéine, UMR CNRS 7100, École Supérieure de Biotechnologie de Strasbourg, Illkirch, France
| | - Gilles Travé
- Équipe Oncoprotéine, UMR CNRS 7100, École Supérieure de Biotechnologie de Strasbourg, Illkirch, France
| | - Robert D. Burk
- Department of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, New York, United States of America
- Departments of Pediatrics, Epidemiology & Population Health and Obstetrics, Gynecology & Women's Health, Albert Einstein College of Medicine, Bronx, New York, United States of America
- * E-mail:
| |
Collapse
|
103
|
Fournane S, Charbonnier S, Chapelle A, Kieffer B, Orfanoudakis G, Travé G, Masson M, Nominé Y. Surface plasmon resonance analysis of the binding of high-risk mucosal HPV E6 oncoproteins to the PDZ1 domain of the tight junction protein MAGI-1. J Mol Recognit 2010; 24:511-23. [DOI: 10.1002/jmr.1056] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2010] [Revised: 04/22/2010] [Accepted: 04/25/2010] [Indexed: 11/10/2022]
|
104
|
Civit L, Fragoso A, O'Sullivan CK. Electrochemical biosensor for the multiplexed detection of human papillomavirus genes. Biosens Bioelectron 2010; 26:1684-7. [PMID: 20667709 DOI: 10.1016/j.bios.2010.06.072] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2010] [Revised: 06/28/2010] [Accepted: 06/30/2010] [Indexed: 02/06/2023]
Abstract
A proof-of-concept of an electrochemical genosensor array for the individual and simultaneous detection of two high-risk human papillomavirus DNA sequences, HPV16E7p and HPV45E6 that exhibits high sensitivity and selectivity is reported. The optimum conditions for surface chemistry preparation and detection of hybridised target were investigated. The LOD obtained are in the pM range, which are sufficient for most real RNA/DNA samples obtained from PCR amplification, usually in the nanomolar range. In a multiplexed detection format, high selectivity was observed over the non-specific sequence, opening the way for the development of an electrochemical high throughput screening assay for multiple high-risk DNA sequences.
Collapse
Affiliation(s)
- L Civit
- Nanobiotechnology and Bioanalysis Group, Departament d'Enginyeria Química, Universitat Rovira i Virgili, Tarragona, Spain
| | | | | |
Collapse
|
105
|
The immortalizing and transforming ability of two common human papillomavirus 16 E6 variants with different prevalences in cervical cancer. Oncogene 2010; 29:3435-45. [PMID: 20383192 DOI: 10.1038/onc.2010.93] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Persistent infection with high-risk human papillomaviruses (HPVs), especially type 16 has been undeniably linked to cervical cancer. The Asian-American (AA) variant of HPV16 is more common in the Americas than the prototype in cervical cancer. The different prevalence is based on three amino acid changes within the E6 protein denoted Q14H/H78Y/L83V. To investigate the mechanism(s) behind this observation, both E6 proteins, in the presence of E7, were evaluated for their ability to extend the life span of and transform primary human foreskin keratinocytes (PHFKs). Long-term cell culture studies resulted in death at passage 9 of vector-transduced PHFKs (negative control), but survival of both E6 PHFKs to passage 65 (and beyond). Compared with E6/E7 PHFKs, AA/E7 PHFKs were significantly faster dividing, developed larger cells in monolayer cultures, showed double the epithelial thickness and expressed cytokeratin 10 when grown as organotypic raft cultures. Telomerase activation and p53 inactivation, two hallmarks of immortalization, were not significantly different between the two populations. Both were resistant to anoikis at later passages, but only AA/E7 PHFKs acquired the capacity for in vitro transformation. Proteomic analysis revealed markedly different protein patterns between E6/E7 and AA/E7, particularly with respect to key cellular metabolic enzymes. Our results provide new insights into the reasons underlying the greater prevalence of the AA variant in cervical cancer as evidenced by characteristics associated with higher oncogenic potential.
Collapse
|
106
|
de Souza RF, Iyer LM, Aravind L. Diversity and evolution of chromatin proteins encoded by DNA viruses. BIOCHIMICA ET BIOPHYSICA ACTA 2010; 1799:302-18. [PMID: 19878744 PMCID: PMC3243496 DOI: 10.1016/j.bbagrm.2009.10.006] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2009] [Revised: 10/21/2009] [Accepted: 10/22/2009] [Indexed: 11/23/2022]
Abstract
Double-stranded DNA viruses display a great variety of proteins that interact with host chromatin. Using the wealth of available genomic and functional information, we have systematically surveyed chromatin-related proteins encoded by dsDNA viruses. The distribution of viral chromatin-related proteins is primarily influenced by viral genome size and the superkingdom to which the host of the virus belongs. Smaller viruses usually encode multifunctional proteins that mediate several distinct interactions with host chromatin proteins and viral or host DNA. Larger viruses additionally encode several enzymes, which catalyze manipulations of chromosome structure, chromatin remodeling and covalent modifications of proteins and DNA. Among these viruses, it is also common to encounter transcription factors and DNA-packaging proteins such as histones and IHF/HU derived from cellular genomes, which might play a role in constituting virus-specific chromatin states. Through all size ranges a subset of domains in viral chromatin proteins appears to have been derived from those found in host proteins. Examples include the Zn-finger domains of the E6 and E7 proteins of papillomaviruses, SET domain methyltransferases and Jumonji-related demethylases in certain nucleocytoplasmic large DNA viruses and BEN domains in poxviruses and polydnaviruses. In other cases, chromatin-interacting modules, such as the LXCXE motif, appear to have been widely disseminated across distinct viral lineages, resulting in similar retinoblastoma targeting strategies. Viruses, especially those with large linear genomes, have evolved a number of mechanisms to manipulate viral chromosomes in the process of replication-associated recombination. These include topoisomerases, Rad50/SbcC-like ABC ATPases and a novel recombinase system in bacteriophages utilizing RecA and Rad52 homologs. Larger DNA viruses also encode SWI2/SNF2 and A18-like ATPases which appear to play specialized roles in transcription and recombination. Finally, it also appears that certain domains of viral provenance have given rise to key functions in eukaryotic chromatin such as a HEH domain of chromosome tethering proteins and the TET/JBP-like cytosine and thymine hydroxylases.
Collapse
Affiliation(s)
- Robson F. de Souza
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD 20894, United States of America
| | - Lakshminarayan M. Iyer
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD 20894, United States of America
| | - L. Aravind
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD 20894, United States of America
| |
Collapse
|
107
|
Construction and identification of a yeast two-hybrid bait vector and its effect on the growth of yeast cells and the self-activating function of reporter genes for screening of HPV18 E6-interacting protein. JOURNAL OF HUAZHONG UNIVERSITY OF SCIENCE AND TECHNOLOGY. MEDICAL SCIENCES = HUA ZHONG KE JI DA XUE XUE BAO. YI XUE YING DE WEN BAN = HUAZHONG KEJI DAXUE XUEBAO. YIXUE YINGDEWEN BAN 2010; 30:8-12. [PMID: 20155448 DOI: 10.1007/s11596-010-0102-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2009] [Indexed: 10/19/2022]
Abstract
By using a yeast two-hybrid system, a yeast two-hybrid bait vector was constructed and identified for screening of the HPV18 E6-interacting proteins, and its effects on the growth of yeast cells and the activation of reporter genes were investigated. Total mRNA extracted from Hela cells was reversely transcribed into cDNA. Fragment of HPV18 E6 cDNA was amplified using RT-PCR and directly ligated to the pGBKT7 vector. The recombinant plasmid was confirmed by restriction endonuclease analysis and DNA sequencing. The recombinant pGBKT7-HPV18 E6 plasmid and empty pGBKT7 vector were transformed into the yeast cell AH109, respectively. After they were cultured respectively in YPDA liquid medium and nutrition-deficient culture medium, their toxicity and transcriptional activation were tested by both the phenotype assay and the color assay. The bait plasmid HPV18 E6 was successfully obtained. After being cultured in YPDA liquid medium for 16h, the A (600 nm) values of two yeast fluids were 0.98+/-0.03 and 0.99+/-0.02, respectively. The recombinant pGBKT7-HPV18 E6 plasmid and empty pGBKT7 vector could grow to white colonies on SD/-Trp/X-alpha-gal plates, while no colony could survive on SD/-His/-Trp/X-alpha-gal, SD/-Ade/-Trp/X-alpha-gal plates, indicating that the bait plasmid pGBKT7-HPV18 E6 was constructed successfully and expressed correctly, and could not activate the transcription of reporter gene alone. The yeast two-hybrid GAL4 system 3 can be utilized to find HPV18 E6 interacting proteins.
Collapse
|
108
|
Poláková I, Pokorná D, Dušková M, Šmahel M. DNA vaccine against human papillomavirus type 16: Modifications of the E6 oncogene. Vaccine 2010; 28:1506-13. [DOI: 10.1016/j.vaccine.2009.11.069] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2009] [Revised: 11/16/2009] [Accepted: 11/21/2009] [Indexed: 11/29/2022]
|
109
|
Köhler A, Gottschling M, Förster J, Röwert-Huber J, Stockfleth E, Nindl I. Genomic characterization of a novel human papillomavirus (HPV-117) with a high viral load in a persisting wart. Virology 2010; 399:129-133. [PMID: 20096912 DOI: 10.1016/j.virol.2009.12.023] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2009] [Revised: 11/24/2009] [Accepted: 12/17/2009] [Indexed: 11/27/2022]
Abstract
Warts from immunosuppressed organ transplant recipients (OTR) persist over years and may progress into non-melanoma skin cancer. Human papillomaviruses (HPV) are considered the causal agents for the development of such warts. We isolated the novel type HPV-117 from a persisting wart by rolling circle amplification. One hundred eighteen warts from immunocompetent patients (IC) and 49 warts from OTR were analyzed by HPV-117 E6 type-specific PCR. As inferred from a phylogenetic analysis, the new type HPV-117 belonged to alpha-PV species 2, including the most similar types HPV-10 and HPV-94. The general prevalence of HPV-117 in warts was 2% in IC (2/118), and 12% in OTR (6/49). The high viral load in dysplastic cells of a Verruca vulgaris was shown by in situ hybridization. Our results suggest an active role of the novel type in the development of cutaneous warts of OTR.
Collapse
Affiliation(s)
- Anja Köhler
- Charité, Department of Dermatology, Venereology and Allergy, Skin Cancer Center Charité, University Hospital of Berlin, Berlin, Germany
| | - Marc Gottschling
- Department of Biology, Systematic Botany and Mycology, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Jana Förster
- Charité, Department of Dermatology, Venereology and Allergy, Skin Cancer Center Charité, University Hospital of Berlin, Berlin, Germany
| | - Joachim Röwert-Huber
- Charité, Department of Dermatology, Venereology and Allergy, Skin Cancer Center Charité, University Hospital of Berlin, Berlin, Germany
| | - Eggert Stockfleth
- Charité, Department of Dermatology, Venereology and Allergy, Skin Cancer Center Charité, University Hospital of Berlin, Berlin, Germany
| | - Ingo Nindl
- Charité, Department of Dermatology, Venereology and Allergy, Skin Cancer Center Charité, University Hospital of Berlin, Berlin, Germany.
| |
Collapse
|
110
|
Abstract
Members of the Alphapapillomavirus genus are the causative agent for virtually all cases of cervical cancer. However, strains (commonly referred to as types) within this genus span the entire range of pathogenicity from highly carcinogenic (e.g., HPV16, odds ratio = 281.9, responsible for 50% of all cervical cancers), moderately carcinogenic (e.g., HPV31) to not carcinogenic (e.g., HPV71). The persistent expression of the viral oncoproteins (E6 and E7) from HPV16 has been shown to be necessary and sufficient to transform primary human keratinocytes in vitro. A plethora of functions have been described for both oncoproteins, and through functional comparisons between HPV16 and HPV6, a subset of these functions have been suggested to be oncogenic. However, extrapolating functional differences from these comparisons is unlikely to tease apart the fine details. In this review, we argue that a thorough understanding of the molecular mechanisms differentiating oncogenic from nononcogenic types should be obtained by performing functional assays in an evolutionary and epidemiological framework. We continue by interpreting some recent results using this paradigm and end by suggesting directions for future inquiries.
Collapse
Affiliation(s)
- Koenraad Van Doorslaer
- Department of Microbiology and Immunology, Albert Einstein Cancer Center, Albert Einstein College of Medicine, New York, USA
| | | |
Collapse
|
111
|
Zanier K, Ruhlmann C, Melin F, Masson M, Ould M'hamed Ould Sidi A, Bernard X, Fischer B, Brino L, Ristriani T, Rybin V, Baltzinger M, Vande Pol S, Hellwig P, Schultz P, Travé G. E6 proteins from diverse papillomaviruses self-associate both in vitro and in vivo. J Mol Biol 2009; 396:90-104. [PMID: 19917295 DOI: 10.1016/j.jmb.2009.11.022] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2009] [Revised: 11/08/2009] [Accepted: 11/09/2009] [Indexed: 11/18/2022]
Abstract
Papillomavirus E6 oncoproteins bind and often provoke the degradation of many cellular proteins important for the control of cell proliferation and/or cell death. Structural studies on E6 proteins have long been hindered by the difficulties of obtaining highly concentrated samples of recombinant E6. Here, we show that recombinant E6 proteins from eight human papillomavirus strains and one bovine papillomavirus strain exist as oligomeric and multimeric species. These species were characterized using a variety of biochemical and biophysical techniques, including analytical gel filtration, activity assays, surface plasmon resonance, electron microscopy and Fourier transform infrared spectroscopy. The characterization of E6 oligomers is facilitated by the fusion to the maltose binding protein, which slows the formation of higher-order multimeric species. The proportion of each oligomeric form varies depending on the viral strain considered. Oligomers appear to consist of folded units, which, in the case of high-risk mucosal human papillomavirus E6, retain binding to the ubiquitin ligase E6-associated protein and the capacity to degrade the proapoptotic protein p53. In addition to the small-size oligomers, E6 proteins spontaneously assemble into large organized multimeric structures, a process that is accompanied by a significant increase in the beta-sheet secondary structure content. Finally, co-localisation experiments using E6 equipped with different tags further demonstrate the occurrence of E6 self-association in eukaryotic cells. The ensemble of these data suggests that self-association is a general property of E6 proteins that occurs both in vitro and in vivo and might therefore be functionally relevant.
Collapse
Affiliation(s)
- Katia Zanier
- Ecole Supérieure de Biotechnologie de Strasbourg (IREBS, FRE 3211), Boulevard Sébastien Brant, BP 10413, 67412 Illkirch Cedex, France.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
112
|
The full-length isoform of human papillomavirus 16 E6 and its splice variant E6* bind to different sites on the procaspase 8 death effector domain. J Virol 2009; 84:1453-63. [PMID: 19906919 DOI: 10.1128/jvi.01331-09] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Human papillomavirus 16 is a causative agent of most cases of cervical cancer and has also been implicated in the development of some head and neck cancers. The early viral E6 gene codes for two alternatively spliced isoforms, E6(large) and E6*. We have previously demonstrated the differential effects of E6(large) and E6* binding on the expression and stability of procaspase 8, a key mediator of the apoptotic pathway. Additionally, we have reported that E6 binds to the FADD death effector domain (DED) at a novel E6 binding domain. Sequence similarities between the FADD and procaspase 8 DEDs suggested a specific region for E6(large)/procaspase 8 binding, which was subsequently confirmed by mutational analysis as well as by the ability of peptides capable of blocking E6/FADD binding to also block E6(large)/caspase 8 binding. However, the binding of the smaller isoform, E6*, to procaspase 8 occurs at a different region, as deletion and point mutations that disrupt E6(large)/caspase 8 DED binding do not disrupt E6*/caspase 8 DED binding. In addition, peptide inhibitors that can block E6(large)/procaspase 8 binding do not affect the binding of E6* to procaspase 8. These results demonstrate that the residues that mediate E6*/procaspase 8 DED binding localize to a different region on the protein and employ a separate binding motif. This provides a molecular explanation for our initial findings that the two E6 isoforms affect procaspase 8 stability in an opposing manner.
Collapse
|
113
|
Abstract
Over the last 30 years, the increasing use of organ and stem cell transplantation and the AIDS epidemic have led to the realization that some, but not all, human cancers occur more frequently in immunosuppressed individuals. With the notable exception of non-melanoma skin cancer (NMSC), most tumors that show strongly increased incidence rates in both transplant recipients and AIDS patients have been found to have a viral etiology. Among these are Kaposi sarcoma, diffuse large cell B-cell lymphoma, cervical cancer, liver cancer, Merkel cell carcinoma and a subset of Hodgkin's disease. A viral etiology for NMSC, i.e., beta- and gamma-subtypes of human papillomavirus, has been suggested and investigated for many years, but remains controversial. In addition, the moderately increased incidence rates of several other cancers in immunosuppressed individuals (e.g., Vajdic and van Leeuwen, Int J Cancer, in press) could indicate that additional infectious causes for at least some human cancers remain to be discovered. The controversy surrounding the role of cutaneous papillomavirus subtypes in the pathogenesis of NMSC illustrates the difficulties encountered when weighing the epidemiological and molecular biology evidence arguing for an involvement of highly prevalent viruses in certain types of cancer.
Collapse
Affiliation(s)
- Thomas F Schulz
- Institute of Virology, Hannover Medical School, Hannover 60325, Germany.
| |
Collapse
|
114
|
Van Doorslaer K, Sidi AOMO, Zanier K, Rybin V, Deryckère F, Rector A, Burk RD, Lienau EK, van Ranst M, Travé G. Identification of unusual E6 and E7 proteins within avian papillomaviruses: cellular localization, biophysical characterization, and phylogenetic analysis. J Virol 2009; 83:8759-70. [PMID: 19553340 PMCID: PMC2738182 DOI: 10.1128/jvi.01777-08] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2008] [Accepted: 05/12/2009] [Indexed: 11/20/2022] Open
Abstract
Papillomaviruses (PVs) are a large family of small DNA viruses infecting mammals, reptiles, and birds. PV infection induces cell proliferation that may lead to the formation of orogenital or skin tumors. PV-induced cell proliferation has been related mainly to the expression of two small oncoproteins, E6 and E7. In mammalian PVs, E6 contains two 70-residue zinc-binding repeats, whereas E7 consists of a natively unfolded N-terminal region followed by a zinc-binding domain which folds as an obligate homodimer. Here, we show that both the novel francolin bird PV Francolinus leucoscepus PV type 1 (FlPV-1) and the chaffinch bird PV Fringilla coelebs PV contain unusual E6 and E7 proteins. The avian E7 proteins contain an extended unfolded N terminus and a zinc-binding domain of reduced size, whereas the avian E6 proteins consist of a single zinc-binding domain. A comparable single-domain E6 protein may have existed in a common ancestor of mammalian and avian PVs. Mammalian E6 C-terminal domains are phylogenetically related to those of single-domain avian E6, whereas mammalian E6 N-terminal domains seem to have emerged by duplication and subsequently diverged from the original ancestral domain. In avian and mammalian cells, both FlPV-1 E6 and FlPV-1 E7 were evenly expressed in the cytoplasm and the nucleus. Finally, samples of full-length FlPV-1 E6 and the FlPV-1 E7 C-terminal zinc-binding domain were prepared for biophysical analysis. Both constructs were highly soluble and well folded, according to nuclear magnetic resonance spectroscopy measurements.
Collapse
Affiliation(s)
- Koenraad Van Doorslaer
- Laboratory of Clinical Virology, Rega Institute for Medical Research, University of Leuven, Minderbroedersstraat 10, 3000 Leuven, Belgium
| | | | | | | | | | | | | | | | | | | |
Collapse
|
115
|
Liu Y, Cherry JJ, Dineen JV, Androphy EJ, Baleja JD. Determinants of stability for the E6 protein of papillomavirus type 16. J Mol Biol 2009; 386:1123-37. [PMID: 19244625 PMCID: PMC2768073 DOI: 10.1016/j.jmb.2009.01.018] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
E6 is an oncoprotein produced by human papillomavirus (HPV). The E6 protein from high-risk HPV type 16 contains two zinc-binding domains with two C-x-x-C motifs each. E6 exerts its transforming functions through formation of a complex with E6AP, which binds p53 and stimulates its degradation. There have been few biophysical and structural studies due to difficulty in preparation of soluble protein; here we describe the preparation of soluble E6 constructs including the two separated zinc-binding domains of E6. These proteins are used to examine the extent to which the two domains cooperate to mediate E6 function, how zinc influences the behavior of E6 protein, and which domains mediate aggregation. We demonstrate, using p53 degradation, E6AP binding, and hDlg (human homolog of the Drosophila discs large tumor suppressor protein) PDZ (postsynaptic density/disc large/zonula occludens) protein binding assays, that these soluble proteins are active, and, using NMR, circular dichroism, and fluorescence spectroscopies, we show that they are folded and stable. We show that the separated N-terminal and C-terminal domains interact, but nonproductively, for E6 function. The two domains bind zinc differently with higher affinity associated with the C-terminal domain. Analyses using surface plasmon resonance and circular dichroism and fluorescence spectroscopies show that aggregation is mediated more through the N-terminal domain than through the C-terminal domain. These studies allow a model in which the C-terminal zinc-binding domain of E6 recruits a target protein such as hDlg and the N-terminal domain is mostly responsible for recruiting a ubiquitin ligase to mediate target protein degradation.
Collapse
Affiliation(s)
- Yuqi Liu
- Department of Biochemistry, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA 02111, U.S.A
| | - Jonathan J. Cherry
- Department of Medicine, University of Massachusetts Medical School, 55 Lake Avenue North, Worcester, MA 01605 U.S.A
| | - Joseph V. Dineen
- Department of Biochemistry, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA 02111, U.S.A
| | - Elliot J. Androphy
- Department of Medicine, University of Massachusetts Medical School, 55 Lake Avenue North, Worcester, MA 01605 U.S.A
| | - James D. Baleja
- Department of Biochemistry, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA 02111, U.S.A
| |
Collapse
|
116
|
Abstract
The papillomaviruses are small DNA viruses that encode approximately eight genes, and require the host cell DNA replication machinery for their viral DNA replication. Thus papillomaviruses have evolved strategies to induce host cell DNA synthesis balanced with strategies to protect the cell from unscheduled replication. While the papillomavirus E1 and E2 genes are directly involved in viral replication by binding to and unwinding the origin of replication, the E6 and E7 proteins have auxillary functions that promote proliferation. As a consequence of disrupting the normal checkpoints that regulate cell cycle entry and progression, the E6 and E7 proteins play a key role in the oncogenic properties of human papillomaviruses with a high risk of causing anogenital cancers (HR HPVs). As a consequence, E6 and E7 of HR HPVs are invariably expressed in cervical cancers. This article will focus on the E6 protein and its numerous activities including inactivating p53, blocking apoptosis, activating telomerase, disrupting cell adhesion, polarity and epithelial differentiation, altering transcription and reducing immune recognition.
Collapse
Affiliation(s)
- Heather L Howie
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Rachel A Katzenellenbogen
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, Washington
- Department of Pediatrics, University of Washington, Seattle, Washington
| | - Denise A Galloway
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, Washington
| |
Collapse
|
117
|
A novel peptide motif binding to and blocking the intracellular activity of the human papillomavirus E6 oncoprotein. J Mol Med (Berl) 2008; 87:321-31. [DOI: 10.1007/s00109-008-0432-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2008] [Revised: 11/25/2008] [Accepted: 11/29/2008] [Indexed: 11/27/2022]
|
118
|
A single-codon mutation converts HPV16 E6 oncoprotein into a potential tumor suppressor, which induces p53-dependent senescence of HPV-positive HeLa cervical cancer cells. Oncogene 2008; 28:762-72. [PMID: 19015633 DOI: 10.1038/onc.2008.422] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
High-risk mucosal human papillomaviruses (HPV), mainly HPV16 and HPV18, are implicated in cervical carcinogenesis. HPV16 E6 oncoprotein binds and often targets for degradation numerous cell proteins, including the tumor suppressor p53 and several PDZ domain proteins. Here, we show that a single-point mutation, F47R, is sufficient to convert the HPV16 E6 oncoprotein into a suppressor of HPV-positive HeLa cervical cancer cells proliferation. The E6 F47R mutant is defective for polyubiquitination and subsequent degradation of p53. When expressed in HPV-positive cervical cancer cells, E6 F47R acts as a dominant negative mutant by counteracting the p53 degradation activity of endogenous E6 and restoring high p53 protein levels. Moreover, the prolonged expression of E6 F47R leads to suppression of HeLa cells proliferation through the induction of premature senescence. This phenotype is independent on the PDZ-binding activity of E6. F47R-senescent HeLa cells exhibit a sustained expression of p53, hMDM2 and p21(CIP) proteins and a reduced expression of endogenous HPV18 E6 protein. Finally, small interfering RNAs directed against p53 counteract the effect of E6 F47R expression, indicating that E6 F47R-induced cellular senescence is strongly dependent on p53 signaling pathway.
Collapse
|
119
|
Simmonds M, Storey A. Identification of the regions of the HPV 5 E6 protein involved in Bak degradation and inhibition of apoptosis. Int J Cancer 2008; 123:2260-6. [PMID: 18752251 PMCID: PMC2677715 DOI: 10.1002/ijc.23815] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
UVB induced DNA damage is the major aetiological agent in NMSC development, but mounting evidence suggests a role for human papillomaviruses (HPV) from genus beta, including HPV 5 and HPV 8, in the development of NMSC on sun exposed body sites. We have previously shown that UVB activates Bak, an apoptogenic mitochondrial factor that, following an apoptotic stimulus, undergoes a conformational change that leads to pore formation in the mitochondrial membrane that releases apoptotic factors. The HPV E6 protein effectively inhibits UVB-induced apoptosis and targets Bak for proteolytic degradation. We have now identified the regions of the HPV5 E6 that are required to mediate Bak proteolysis and contribute toward the antiapoptotic activity of the protein. Interestingly, while wild-type HPV5 E6 does not bind or target p53 for proteolysis, we have isolated specific HPV5 E6 mutants that switch target specificity from Bak to p53 in a p53 codon 72 isoform-dependent manner. Furthermore, we demonstrate that the ability of wild-type HPV5 E6 to target Bak or specific E6 mutants to target p53 for proteolysis is not dependent on the E6-AP ubiquitin ligase.
Collapse
Affiliation(s)
- Mark Simmonds
- Cancer Research UK, Skin Tumour Laboratory, ICMS, 4 Newark Street, London, United Kingdom
| | - Alan Storey
- Cancer Research UK, Skin Tumour Laboratory, ICMS, 4 Newark Street, London, United Kingdom
| |
Collapse
|
120
|
Abstract
Every year, approximately 470,000 new cases of cervical cancer are diagnosed and approximately 230,000 women worldwide die of the disease, with the majority (approximately 80%) of these cases and deaths occurring in developing countries. Human papillomaviruses (HPVs) are the etiological agents in nearly all cases (99.7%) of cervical cancer, and the HPV E6 protein is one of two viral oncoproteins that is expressed in virtually all HPV-positive cancers. E6 hijacks a cellular ubiquitin ligase, E6AP, resulting in the ubiquitylation and degradation of the p53 tumor suppressor, as well as several other cellular proteins. While the recent introduction of prophylactic vaccines against specific HPV types offers great promise for prevention of cervical cancer, there remains a need for therapeutics. Biochemical characterization of E6 and E6AP has suggested approaches for interfering with the activities of these proteins that could be useful for this purpose. PUBLICATION HISTORY : Republished from Current BioData's Targeted Proteins database (TPdb; http://www.targetedproteinsdb.com).
Collapse
Affiliation(s)
| | - Jon M Huibregtse
- Molecular Genetics and Microbiology, Institute for Cellular and Molecular Biology, University of Texas at Austin, Austin, TX 78712, USA
| |
Collapse
|
121
|
New variants of human papillomavirus type 18 identified in central Brazil. Virus Genes 2008; 37:282-7. [DOI: 10.1007/s11262-008-0263-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2008] [Accepted: 07/11/2008] [Indexed: 10/21/2022]
|
122
|
The interaction between human papillomavirus type 16 and FADD is mediated by a novel E6 binding domain. J Virol 2008; 82:9600-14. [PMID: 18632871 DOI: 10.1128/jvi.00538-08] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
High-risk strains of human papillomavirus, such as types 16 and 18, have been etiologically linked to cervical cancer. Most cervical cancer tissues are positive for both the E6 and E7 oncoproteins, since it is their cooperation that results in successful transformation and immortalization of infected cells. We have reported that E6 binds to tumor necrosis factor receptor 1 and to Fas-associated death domain (FADD) and, in doing so, prevents E6-expressing cells from responding to apoptotic stimuli. The binding site of E6 to FADD localizes to the first 23 amino acids of FADD and has now been further characterized by the use of deletion and site-directed mutants of FADD in pull-down and functional assays. The results from these experiments revealed that mutations of serine 16, serine 18, and leucine 20 obstruct FADD binding to E6, suggesting that these residues are part of the E6 binding domain on FADD. Because FADD does not contain the two previously identified E6 binding motifs, the LxxphiLsh motif, and the PDZ motif, a novel binding domain for E6 has been identified on FADD. Furthermore, peptides that correspond to this region can block E6/FADD binding in vitro and can resensitize E6-expressing cells to apoptotic stimuli in vivo. These results demonstrate the existence of a novel E6 binding domain.
Collapse
|
123
|
Charbonnier S, Stier G, Orfanoudakis G, Kieffer B, Atkinson RA, Travé G. Defining the minimal interacting regions of the tight junction protein MAGI-1 and HPV16 E6 oncoprotein for solution structure studies. Protein Expr Purif 2008; 60:64-73. [DOI: 10.1016/j.pep.2008.03.022] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2008] [Revised: 03/17/2008] [Accepted: 03/19/2008] [Indexed: 11/30/2022]
|
124
|
Liu R, Pu D, Liu Y, Cheng Y, Yin L, Li T, Zhao L. Induction of SiHa cells apoptosis by nanometer realgar suspension and its mechanism. ACTA ACUST UNITED AC 2008; 28:317-21. [PMID: 18563332 DOI: 10.1007/s11596-008-0320-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2008] [Indexed: 10/19/2022]
Abstract
The effects of nanometer realgar suspension on proliferation and apoptosis of human uterine cervix cancer cell line SiHa cells and oncogenic genes HPV16E6/E7 were investigated. A "micro-jet efflux" strategy was used for the preparation of nanometer realgar suspension. SiHa cells were treated with nanometer Realgar suspension in various concentrations (6.25, 12.5, 25 and 50 mg/L) for different durations (12, 24, 48 and 72 h). The inhibitive effect of nanometer realgar suspension on growth of SiHa cells was detected by MTT method. Special morphological changes of apoptosis were observed by transmission electron microscopy (TEM) and DNA fragments electrophoresis. The apoptotic rate was quantified by flow cytometry (FCM). The expression of HPV16E6/E7 mRNA and protein was assayed by RT-PCR and Western blot respectively. The results showed after being treated with 25 50 mg/L nanometer realgar suspension for 48 h, the survival rate of SiHa cells was decreased, and apoptotic rate markedly increased in a time-and concentration-dependent manner. TEM and DNA electrophoresis revealed the special morphological changes of apoptosis. The apoptotic rate of SiHa cells treated with nanometer realgar suspension was significantly higher than in the control group (P<0.01), and G(0)/G(1) phase arrest appeared following treatment with nanometer realgar suspension in 25 and 50 mg/L for 48 h. RT-PCR and Western blot assay indicated that nanometer realgar suspension reduced the HPV16E6/E7 gene expression. Nanometer realgar suspension could inhibit the proliferation and induce apoptosis of SiHa cells. The mechanism may be related to the down-regulation of the HPV16E6/E7 gene expression.
Collapse
Affiliation(s)
- Rong Liu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | | | | | | | | | | | | |
Collapse
|
125
|
A novel adenovirus vector for easy cloning in the E3 region downstream of the CMV promoter. Virol J 2008; 5:73. [PMID: 18538014 PMCID: PMC2427025 DOI: 10.1186/1743-422x-5-73] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2008] [Accepted: 06/06/2008] [Indexed: 11/11/2022] Open
Abstract
The construction of expression vectors derived from the human adenovirus type 5 (Ad5), usually based on homologous recombination, is time consuming as a shuttle plasmid has to be selected before recombination with the viral genome. Here, we describe a method allowing direct cloning of a transgene in the E3 region of the Ad5 genome already containing the immediate early CMV promoter upstream of three unique restriction sites. This allowed the construction of recombinant adenoviral genomes in just one step, reducing considerably the time of selection and, of course, production of the corresponding vectors. Using this vector, we produced recombinant adenoviruses, each giving high-level expression of the transgene in the transduced cells.
Collapse
|
126
|
Thomas M, Dasgupta J, Zhang Y, Chen X, Banks L. Analysis of specificity determinants in the interactions of different HPV E6 proteins with their PDZ domain-containing substrates. Virology 2008; 376:371-8. [PMID: 18452965 DOI: 10.1016/j.virol.2008.03.021] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2008] [Revised: 02/14/2008] [Accepted: 03/10/2008] [Indexed: 11/30/2022]
Abstract
The E6 oncoproteins of the cancer-associated human papillomaviruses (high-risk HPV types) characteristically have a PDZ-binding motif at their extreme carboxy-termini. However, they interact with only some of the PDZ domain-containing proteins in the human proteome and, despite many of these proteins having multiple PDZ domains, they interact specifically through only one of those domains. Previous work has shown that the exact sequence of the C-terminal PDZ-binding motif of E6 affects substrate selection, and recently we have shown that an E6 residue peripheral to the binding motif also contributes to the specificity of binding. Here we show that substrate specificity of the HPV E6 PDZ binding is modulated both by the amino acid residues upstream of the binding domain and by the non-canonical residues within it. Using this data we have begun to construct a scheme of substrate preferences for E6 proteins from different HPV types.
Collapse
Affiliation(s)
- Miranda Thomas
- Tumour Virology Laboratory, I.C.G.E.B., Padriciano 99, 34012 Trieste, Italy.
| | | | | | | | | |
Collapse
|
127
|
The E6 oncoproteins from human betapapillomaviruses differentially activate telomerase through an E6AP-dependent mechanism and prolong the lifespan of primary keratinocytes. J Virol 2008; 82:3894-902. [PMID: 18256157 DOI: 10.1128/jvi.01818-07] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Human papillomaviruses (HPVs) belonging to the Betapapillomavirus genus have recently been implicated in squamous cell carcinomas of the skin, though the mechanisms by which they initiate carcinogenesis are unclear. We show that human foreskin keratinocytes (HFKs) expressing several betapapillomavirus E6 (beta-E6) proteins display life span extension, but not to the extent seen in HFKs expressing HPV type 16 E6 (16E6). Additionally, we demonstrate that beta-E6 proteins can differentially activate telomerase. HFKs expressing 38E6 exhibit significant telomerase activity but to a lesser degree than that observed with 16E6; however, other beta-E6 proteins, including 5E6, 8E6, 20E6, and 22E6, exhibit low or background levels of telomerase activity. Utilizing glutathione S-transferase pull-down and coimmunoprecipitation experiments, the beta-E6 proteins were shown to interact with the cellular proteins E6-associated protein (E6AP) and NFX1-91, two proteins known to be important for telomerase activation by 16E6. Interestingly, the relative strength of the interaction between E6 and E6AP or NFX1-91 was proportionate to the activation of telomerase by each beta-E6 protein. To address the requirement for E6AP in telomerase activation by beta-E6 proteins, we utilized a shRNA to knock down endogenous levels of E6AP. Lysates with decreased levels of E6AP showed a reduced ability to activate telomerase, suggesting that E6AP is a necessary component. These data suggest that complex formation between E6, E6AP, and NFX1-91 is a critical step in mediating telomerase activation, which may be one contributing factor to cellular life span extension during human betapapillomavirus infection.
Collapse
|
128
|
Tungteakkhun SS, Duerksen-Hughes PJ. Cellular binding partners of the human papillomavirus E6 protein. Arch Virol 2008; 153:397-408. [PMID: 18172569 PMCID: PMC2249614 DOI: 10.1007/s00705-007-0022-5] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2007] [Accepted: 12/13/2007] [Indexed: 11/25/2022]
Abstract
The high-risk strains of human papillomavirus (HR-HPV) are known to be causative agents of cervical cancer and have recently also been implicated in cancers of the oropharynx. E6 is a potent oncogene of HR-HPVs, and its role in the progression to malignancy has been and continues to be explored. E6 is known to interact with and subsequently inactivate numerous cellular proteins pivotal in the mediation of apoptosis, transcription of tumor suppressor genes, maintenance of epithelial organization, and control of cell proliferation. Binding of E6 to these proteins cumulatively contributes to the oncogenic potential of HPV. This paper provides an overview of these cellular protein partners of HR-E6, the motifs known to mediate oncoprotein binding, and the agents that have the potential to interfere with E6 expression and activity and thus prevent the subsequent progression to oncogenesis.
Collapse
Affiliation(s)
- Sandy S. Tungteakkhun
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92354 USA
| | | |
Collapse
|
129
|
Sibler AP, Baltzinger M, Choulier L, Desplancq D, Altschuh D. SPR identification of mild elution conditions for affinity purification of E6 oncoprotein, using a multivariate experimental design. J Mol Recognit 2008; 21:46-54. [DOI: 10.1002/jmr.865] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
130
|
Töpffer S, Müller-Schiffmann A, Matentzoglu K, Scheffner M, Steger G. Protein tyrosine phosphatase H1 is a target of the E6 oncoprotein of high-risk genital human papillomaviruses. J Gen Virol 2007; 88:2956-2965. [DOI: 10.1099/vir.0.83123-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
The E6 proteins of high-risk genital human papillomaviruses (HPV), such as HPV types 16 and 18, possess a conserved C-terminal PDZ-binding motif, which mediates interaction with some cellular PDZ domain proteins. The binding of E6 usually results in their ubiquitin-mediated degradation. The ability of E6 to bind to PDZ domain proteins correlates with the oncogenic potential. Using a yeast two-hybrid system, GST pull-down experiments and coimmunoprecipitations, we identified the protein tyrosine phosphatase H1 (PTPH1/PTPN3) as a novel target of the PDZ-binding motif of E6 of HPV16 and 18. PTPH1 has been suggested to function as tumour suppressor protein, since mutational analysis revealed somatic mutations in PTPH1 in a minor fraction of various human tumours. We show here that HPV16 E6 accelerated the proteasome-mediated degradation of PTPH1, which required the binding of E6 to the cellular ubiquitin ligase E6-AP and to PTPH1. The endogenous levels of PTPH1 were particularly low in HPV-positive cervical carcinoma cell lines. The reintroduction of the E2 protein into the HPV16-positive cervical carcinoma cell line SiHa, known to lead to a sharp repression of E6 expression and to induce growth suppression, resulted in an increase of the amount of PTPH1. Our data suggest that reducing the level of PTPH1 may contribute to the oncogenic activity of high-risk genital E6 proteins.
Collapse
Affiliation(s)
- Stephanie Töpffer
- Institute of Virology, University of Cologne, Fürst-Pückler-Strasse 56, 50935 Cologne, Germany
| | | | | | - Martin Scheffner
- Department of Biology, University of Konstanz, Konstanz, Germany
| | - Gertrud Steger
- Institute of Virology, University of Cologne, Fürst-Pückler-Strasse 56, 50935 Cologne, Germany
| |
Collapse
|
131
|
Binding of human papillomavirus type 16 E6 to E6AP is not required for activation of hTERT. J Virol 2007; 82:71-6. [PMID: 17942561 DOI: 10.1128/jvi.01776-07] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The human papillomavirus (HPV) type 16 (HPV16) E6 protein stimulates transcription of the catalytic subunit of telomerase, hTERT, in epithelial cells. It has been reported that binding to the ubiquitin ligase E6AP is required for this E6 activity, with E6 directing E6AP to the hTERT promoter. We previously reported two E6AP binding-defective HPV16 E6 mutations that induced immortalization of human mammary epithelial cells. Because activation of hTERT is proposed to be necessary for epithelial cell immortalization, we sought to further characterize the relationship between E6/E6AP association and telomerase induction. We demonstrate that while these E6 mutants do not bind E6AP, they retain the capability to stimulate the expression of hTERT. Chromatin immunoprecipitation assays confirmed the presence of Myc, wild-type E6, and the E6AP binding-defective E6 mutants, but not E6AP itself, at the endogenous hTERT promoter. Interestingly, an immortalization-defective E6 mutant localized to the hTERT promoter but failed to increase transcription. We conclude that binding to E6AP is not necessary for E6 localization to or activation of the hTERT promoter and that another activity of E6 is involved in hTERT activation.
Collapse
|
132
|
Van Bressem MF, Cassonnet P, Rector A, Desaintes C, Van Waerebeek K, Alfaro-Shigueto J, Van Ranst M, Orth G. Genital warts in Burmeister's porpoises: characterization of Phocoena spinipinnis papillomavirus type 1 (PsPV-1) and evidence for a second, distantly related PsPV. J Gen Virol 2007; 88:1928-1933. [PMID: 17554024 DOI: 10.1099/vir.0.82694-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
We identified sequences from two distantly related papillomaviruses in genital warts from two Burmeister's porpoises, including a PV antigen-positive specimen, and characterized Phocoena spinipinnis papillomavirus type 1 (PsPV-1). The PsPV-1 genome comprises 7879 nt and presents unusual features. It lacks an E7, an E8 and a bona fide E5 open reading frame (ORF) and has a large E6 ORF. PsPV-1 L1 ORF showed the highest percentage of nucleotide identity (54-55 %) with human papillomavirus type 5, bovine papillomavirus type 3 (BPV-3) and Tursiops truncatus papillomavirus type 2 (TtPV-2). This warrants the classification of PsPV-1 as the prototype of the genus Omikronpapillomavirus. PsPV-1 clustered with TtPV-2 in the E6 and E1E2 phylogenetic trees and with TtPV-2 and BPV-3 in the L2L1 tree. This supports the hypothesis that PV evolution may not be monophyletic across all genes.
Collapse
Affiliation(s)
- Marie-Françoise Van Bressem
- Cetacean Conservation Medicine Group (CMED), CEPEC/Museo de Delfines, Waldspielplatz 11, 82319 Starnberg, Germany
| | - Patricia Cassonnet
- Département de Virologie, Institut Pasteur, 25 rue du Dr Roux, 75724 Paris Cedex 15, France
| | - Annabel Rector
- Laboratory of Clinical and Epidemiological Virology, Rega Institute for Medical Research, University of Leuven, Minderbroedersstraat 10, B-3000 Leuven, Belgium
| | | | - Koen Van Waerebeek
- Cetacean Conservation Medicine Group (CMED), CEPEC/Museo de Delfines, Waldspielplatz 11, 82319 Starnberg, Germany
| | | | - Marc Van Ranst
- Laboratory of Clinical and Epidemiological Virology, Rega Institute for Medical Research, University of Leuven, Minderbroedersstraat 10, B-3000 Leuven, Belgium
| | - Gérard Orth
- Département de Virologie, Institut Pasteur, 25 rue du Dr Roux, 75724 Paris Cedex 15, France
| |
Collapse
|
133
|
Lagrange M, Boulade-Ladame C, Mailly L, Weiss E, Orfanoudakis G, Deryckere F. Intracellular scFvs against the viral E6 oncoprotein provoke apoptosis in human papillomavirus-positive cancer cells. Biochem Biophys Res Commun 2007; 361:487-92. [PMID: 17658466 DOI: 10.1016/j.bbrc.2007.07.040] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2007] [Accepted: 07/11/2007] [Indexed: 12/01/2022]
Abstract
The E6 protein of human papillomavirus type 16 (16E6) is involved in the tumorigenesis of human cervical cells by targeting numerous cellular proteins. We have designed a strategy for neutralizing 16E6 based on the intracellular expression of single-chain Fv antibodies (scFvs) specific to 16E6. Recombinant adenovirus vectors were constructed to allow expression of two 16E6-binding scFvs and one 16E6-non-binding scFv in HPV16-positive and -negative cells. Expression of the scFvs provoked two types of effects: (i) inhibition of proliferation of all cell lines tested, this aspecific toxicity being likely due to the aggregation of unfolded scFvs; and (ii) apoptosis observed only in HPV16-positive cervical cancer cell lines after expression of 16E6-binding scFvs, this specific effect being proportional to the intracellular solubility of the scFvs. These data demonstrate the feasibility of intracellular immunization with anti-16E6 scFvs and highlight the importance of the solubility of the intracellular antibodies.
Collapse
Affiliation(s)
- Magali Lagrange
- UMR 7175-LC1, CNRS, Université Louis Pasteur (Strasbourg I), ESBS, Boulevard Sébastien Brant, BP 10413, 67412 Illkirch Cedex, France
| | | | | | | | | | | |
Collapse
|
134
|
Camus S, Menéndez S, Cheok CF, Stevenson LF, Laín S, Lane DP. Ubiquitin-independent degradation of p53 mediated by high-risk human papillomavirus protein E6. Oncogene 2007; 26:4059-70. [PMID: 17224909 PMCID: PMC2742713 DOI: 10.1038/sj.onc.1210188] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2006] [Revised: 11/02/2006] [Accepted: 11/06/2006] [Indexed: 11/09/2022]
Abstract
In vitro, high-risk human papillomavirus E6 proteins have been shown, in conjunction with E6-associated protein (E6AP), to mediate ubiquitination of p53 and its degradation by the 26S proteasome by a pathway that is thought to be analogous to Mdm2-mediated p53 degradation. However, differences in the requirements of E6/E6AP and Mdm2 to promote the degradation of p53, both in vivo and in vitro, suggest that these two E3 ligases may promote p53 degradation by distinct pathways. Using tools that disrupt ubiquitination and degradation, clear differences between E6- and Mdm2-mediated p53 degradation are presented. The consistent failure to fully protect p53 protein from E6-mediated degradation by disrupting the ubiquitin-degradation pathway provides the first evidence of an E6-dependent, ubiquitin-independent, p53 degradation pathway in vivo.
Collapse
Affiliation(s)
- S Camus
- Department of Cell Cycle Control, Institute of Molecular and Cell Biology, Proteos, Singapore.
| | | | | | | | | | | |
Collapse
|
135
|
Shai A, Nguyen ML, Wagstaff J, Jiang YH, Lambert PF. HPV16 E6 confers p53-dependent and p53-independent phenotypes in the epidermis of mice deficient for E6AP. Oncogene 2007; 26:3321-8. [PMID: 17130828 PMCID: PMC2858288 DOI: 10.1038/sj.onc.1210130] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2006] [Revised: 10/10/2006] [Accepted: 10/13/2006] [Indexed: 02/01/2023]
Abstract
High-risk human papillomaviruses are the causative agents of cervical and other anogenital cancers. In these cancers, two viral oncogenes, E6 and E7, are expressed. E6 is best known for its ability to inactivate the tumor suppressor p53, which is thought to arise through ubiquitin-mediated degradation of p53 and involve a ternary complex between E6, p53 and the E3 ligase, E6AP. In mice transgenic for wild-type HPV16 E6, its expression leads to epithelial hyperplasia and an abrogation of normal cellular responses to DNA damage. Whereas only the latter phenotype is dependent upon E6's inactivation of p53, both are reduced in transgenic mice expressing an E6 mutant severely reduced in its binding to E6AP and other cellular proteins that bind E6 through a shared alpha-helix motif. Here, we investigated whether E6AP is required for the induction of the above phenotypes through the use of both E6AP-mutant and E6AP-null mice. E6, in the absence of E6AP retains an ability to induce epithelial hyperplasia, abrogate DNA damage responses and inhibit the induction of p53 protein following exposure to ionizing radiation. We conclude that E6 is able to induce both p53-dependent and p53-independent phenotypes through E6AP-independent pathways in the mouse.
Collapse
Affiliation(s)
- A Shai
- Department of Cancer Biology, McArdle Laboratory for Cancer Research, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - ML Nguyen
- Department of Cancer Biology, McArdle Laboratory for Cancer Research, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - J Wagstaff
- Department of Pediatrics, Carolinas Medical Center, Charlotte, NC, USA
| | - Y-h Jiang
- Department of Molecular and Human Genetics, Baylor College of Medicine, Baylor University, Houston, TX, USA
| | - PF Lambert
- Department of Cancer Biology, McArdle Laboratory for Cancer Research, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| |
Collapse
|
136
|
Fradet-Turcotte A, Archambault J. Recent Advances in the Search for Antiviral Agents against Human Papillomaviruses. Antivir Ther 2007. [DOI: 10.1177/135965350701200417] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Infection by human papillomavirus (HPV) is extremely common and associated with the development of benign warts or malignant lesions of the skin and mucosa. Infection by a high-risk (oncogenic) anogenital HPV type, most often through sexual contacts, is the starting point of virtually all cases of cervical cancers and the majority of anal cancers. The same viral types are also increasingly being linked with a subset of head-and-neck and non-melanoma skin cancers. Although prophylactic vaccines are now available to protect against the four types most commonly found in cervical and anal cancers (HPV16 and HPV18) and anogenital warts (HPV6 and HPV11), these neither protect against all genital HPVs nor are of therapeutic utility for already infected patients. Thus, the need for antiviral agents to treat HPV-associated diseases remains great, but none currently exist. This article reviews the recent progress made towards the development of antiviral agents to treat HPV infections, from target identification and validation to the discovery of lead compounds with therapeutic potential. Emphasis has been placed on novel low-molecular-weight compounds that antagonize HPV proteins or, alternatively, inhibit cellular proteins which have been usurped by papillomaviruses and are mediating their pathogenic effects.
Collapse
Affiliation(s)
- Amélie Fradet-Turcotte
- Laboratory of Molecular Virology, Institut de Recherches Cliniques de Montréal, Montreal, Quebec, Canada
- Department of Biochemistry, University of Montreal, Montreal, Quebec, Canada
| | - Jacques Archambault
- Laboratory of Molecular Virology, Institut de Recherches Cliniques de Montréal, Montreal, Quebec, Canada
- Department of Biochemistry, University of Montreal, Montreal, Quebec, Canada
| |
Collapse
|
137
|
Kanda T, Kukimoto I. [Human papillomavirus and cervical cancer]. Uirusu 2007; 56:219-30. [PMID: 17446671 DOI: 10.2222/jsv.56.219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Human papillomavirus (HPV) is a small non-enveloped icosahedral virus with a circular double-stranded DNA genome of 8 kilo base pairs. HPV particles reach and infect the basal cells of the stratified epithelia through small epithelial lesions. In the basal cells the viral DNA is maintained as episomes, which start to replicate when the host cells initiate terminal differentiation. In these differentiating cells the degradation of p53 by the E6 protein and the abrogation of the pRb functions by the E7 protein lead to the reactivation of the DNA synthesis machinery. After virus propagation the host cells usually die. On the other hand, in some of the infected cells, the E6 and E7 genes are integrated on rare occasion into cell DNA. The cell continuously expressing the E6 and E7 proteins from the integrated genes is immortalized and sometimes acquires malignant phenotype induced by the accumulated damages to DNA. Of more than 100 HPV genotypes recorded to date, 13 including types 16 and 18 are associated with cervical cancer. Expression of HPV major capsid protein L1 in some cultured cells results in production of virus-like particles (VLPs). The VLPs of types 6, 11, 16, and 18 were used as a prophylactic vaccine in recent clinical trials and shown to successfully induce type-specific neutralizing antibodies in the recipients.
Collapse
Affiliation(s)
- Tadahito Kanda
- Center for Pathogen Genomics, National Institute of Infectious Diseases.
| | | |
Collapse
|
138
|
Storrs CH, Silverstein SJ. PATJ, a tight junction-associated PDZ protein, is a novel degradation target of high-risk human papillomavirus E6 and the alternatively spliced isoform 18 E6. J Virol 2007; 81:4080-90. [PMID: 17287269 PMCID: PMC1866151 DOI: 10.1128/jvi.02545-06] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2006] [Accepted: 01/30/2007] [Indexed: 12/25/2022] Open
Abstract
The E6 protein from high-risk human papillomavirus types interacts with and degrades several PDZ domain-containing proteins that localize to adherens junctions or tight junctions in polarized epithelial cells. We have identified the tight junction-associated multi-PDZ protein PATJ (PALS1-associated TJ protein) as a novel binding partner and degradation target of high-risk types 16 and 18 E6. PATJ functions in the assembly of the evolutionarily conserved CRB-PALS1-PATJ and Par6-aPKC-Par3 complexes and is critical for the formation of tight junctions in polarized cells. The ability of type 18 E6 full-length to bind to, and the subsequent degradation of, PATJ is dependent on its C-terminal PDZ binding motif. We demonstrate that the spliced 18 E6* protein, which lacks a C-terminal PDZ binding motif, associates with and degrades PATJ independently of full-length 18 E6. Thus, PATJ is the first binding partner that is degraded in response to both isoforms of 18 E6. The ability of E6 to utilize a non-E6AP ubiquitin ligase for the degradation of several PDZ binding partners has been suggested. We also demonstrate that 18 E6-mediated degradation of PATJ is not inhibited in cells where E6AP is silenced by shRNA. This suggests that the E6-E6AP complex is not required for the degradation of this protein target.
Collapse
Affiliation(s)
- Carina H Storrs
- Department of Microbiology, College of Physicians and Surgeons, Columbia University, 701 W. 168th St., New York, NY 10032, USA
| | | |
Collapse
|
139
|
Brimer N, Lyons C, Vande Pol SB. Association of E6AP (UBE3A) with human papillomavirus type 11 E6 protein. Virology 2007; 358:303-10. [PMID: 17023019 PMCID: PMC1892534 DOI: 10.1016/j.virol.2006.08.038] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2006] [Revised: 06/19/2006] [Accepted: 08/23/2006] [Indexed: 11/28/2022]
Abstract
The cellular E3 ubiquitin ligase E6AP (UBE3A) interacts with the cancer-associated HPV E6 oncoproteins, where together with the viral E6 oncoprotein it binds and targets the degradation of the p53 tumor suppressor. We find that the HPV-11E6 protein also associates with E6AP in vivo, and thereby can target the degradation of an E6-associated protein. Mutation of an E6-binding LXXLL peptide motif on E6AP eliminated the association, revealing a common mode of interaction between high- and low-risk E6 proteins and E6AP. E6AP was required for the in vivo degradation of DLG1 by both HVP-18 E6 and a chimeric HPV-11E6. The common functional interaction of both cancer-associated and non-cancer-associated E6 proteins with E6AP establishes a common mechanism for E6 proteins trophic to mucosal squamous epithelium.
Collapse
Affiliation(s)
- Nicole Brimer
- Department of Pathology, University of Virginia School of Medicine
| | - Charles Lyons
- Department of Pathology, University of Virginia School of Medicine
| | | |
Collapse
|
140
|
Uversky VN, Roman A, Oldfield CJ, Dunker AK. Protein intrinsic disorder and human papillomaviruses: increased amount of disorder in E6 and E7 oncoproteins from high risk HPVs. J Proteome Res 2007; 5:1829-42. [PMID: 16889404 DOI: 10.1021/pr0602388] [Citation(s) in RCA: 110] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
It is recognized now that many functional proteins or their long segments are devoid of stable secondary and/or tertiary structure and exist instead as very dynamic ensembles of conformations. They are known by different names including natively unfolded, intrinsically disordered, intrinsically unstructured, rheomorphic, pliable, and different combinations thereof. Many important functions and activities have been associated with these intrinsically disordered proteins (IDPs), including molecular recognition, signaling, and regulation. It is also believed that disorder of these proteins allows function to be readily modified through phosphorylation, acetylation, ubiquitination, hydroxylation, and proteolysis. Bioinformatics analysis revealed that IDPs comprise a large fraction of different proteomes. Furthermore, it is established that the intrinsic disorder is relatively abundant among cancer-related and other disease-related proteins and IDPs play a number of key roles in oncogenesis. There are more than 100 different types of human papillomaviruses (HPVs), which are the causative agents of benign papillomas/warts, and cofactors in the development of carcinomas of the genital tract, head and neck, and epidermis. With respect to their association with cancer, HPVs are grouped into two classes, known as low (e.g., HPV-6 and HPV-11) and high-risk (e.g., HPV-16 and HPV-18) types. The entire proteome of HPV includes six nonstructural proteins [E1, E2, E4, E5, E6, and E7 (the latter two are known to function as oncoproteins in the high-risk HPVs)] and two structural proteins (L1 and L2). To understand whether intrinsic disorder plays a role in the oncogenic potential of different HPV types, we have performed a detailed bioinformatics analysis of proteomes of high-risk and low-risk HPVs with the major focus on E6 and E7 oncoproteins. The results of this analysis are consistent with the conclusion that high-risk HPVs are characterized by the increased amount of intrinsic disorder in transforming proteins E6 and E7.
Collapse
Affiliation(s)
- Vladimir N Uversky
- Department of Biochemistry and Molecular Biology, Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, 46202, USA.
| | | | | | | |
Collapse
|
141
|
Asadurian Y, Kurilin H, Lichtig H, Jackman A, Gonen P, Tommasino M, Zehbe I, Sherman L. Activities of human papillomavirus 16 E6 natural variants in human keratinocytes. J Med Virol 2007; 79:1751-60. [PMID: 17854024 DOI: 10.1002/jmv.20978] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Genetic variations in the E6 oncogene have been associated with different risk for cancer progression. In the present study, the functional significance of human papillomavirus (HPV) polymorphism in the E6 oncogene was investigated. Ten HPV16 E6 variants containing amino acid substitutions in the N-terminal region of E6 were evaluated for different biological and biochemical activities in human keratinocytes, the target cells for HPV infection. Western blot analyses of primary foreskin human keratinocytes or immortalized human keratinocytes, stably transduced with the E6 variants, revealed reduced p53 and Bax levels in all E6 expressing cultures. The reduction induced by most E6 proteins was at similar levels and comparable to the reduction induced by the E6 prototype. The ability of the proteins to induce serum/calcium-differentiation resistant colonies in primary keratinocytes was more variable. Overall activities of the variants ranged between 0.24- and 2.18-fold of the E6 prototype activity. The I27R/L83V variant showed the lowest activity whereas the R8Q variant showed the highest activity. The L83V polymorphism previously associated with risk for cancer progression in some populations, showed significant activity, comparable to that of the E6 prototype, in reducing p53 and Bax levels. Furthermore, this variant showed enhancement in the ability to induce colonies resistant to serum/calcium-triggered differentiation, however, the difference from the prototype was not statistically significant. This, and augmentation of other described functions might result in differences in L83V pathogenicity.
Collapse
Affiliation(s)
- Yulia Asadurian
- Department of Human Microbiology, Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | | | | | | | | | | | | | | |
Collapse
|
142
|
Paul I, Cui J, Maynard EL. Zinc binding to the HCCH motif of HIV-1 virion infectivity factor induces a conformational change that mediates protein-protein interactions. Proc Natl Acad Sci U S A 2006; 103:18475-80. [PMID: 17132731 PMCID: PMC1693687 DOI: 10.1073/pnas.0604150103] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Virion infectivity factor (Vif) is an accessory protein encoded by HIV-1 and is critical for viral infection of the host CD4(+) T cell population. Vif induces ubiquitination and subsequent degradation of Apo3G, a cytosolic cytidine deaminase that otherwise targets the retroviral genome. Interaction of Vif with the cellular Cullin5-based E3 ubiquitin ligase requires a conserved BC box and upstream residues that are part of the conserved H-(Xaa)(5)-C-(Xaa)(17-18)-C-(Xaa)(3-5)-H (HCCH) motif. The HCCH motif is involved in stabilizing the Vif-Cullin 5 interaction, but the exact role of the conserved His and Cys residues remains elusive. In this report, we find that full-length HIV-1 Vif, as well as a HCCH peptide, is capable of binding to zinc with high specificity. Zinc binding induces a conformational change that leads to the formation of large protein aggregates. EDTA reversed aggregation and regenerated the apoprotein conformation. Cysteine modification studies with the HCCH peptide suggest that C114 is critical for stabilizing the fold of the apopeptide, and that C133 is located in a solvent-exposed region with no definite secondary structure. Selective alkylation of C133 reduced metal-binding specificity of the HCCH peptide, allowing cobalt to bind with rates comparable to that with zinc. This study demonstrates that the HCCH motif of HIV-1 Vif is a unique metal-binding domain capable of mediating protein-protein interactions in the presence of zinc and adds to a growing list of examples in which metal ion binding induces protein misfolding and/or aggregation.
Collapse
Affiliation(s)
- Indrani Paul
- Department of Biochemistry and Molecular Biology, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814
| | - Jian Cui
- Department of Biochemistry and Molecular Biology, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814
| | - Ernest L. Maynard
- Department of Biochemistry and Molecular Biology, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814
- *To whom correspondence should be addressed. E-mail:
| |
Collapse
|
143
|
Hegde RS. Papillomavirus proteins and their potential as drug design targets. Future Virol 2006. [DOI: 10.2217/17460794.1.6.795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The papillomaviruses are a family of small, double-stranded DNA viruses that infect the basal cells of cutaneous and mucosal epithelium. While a large percentage of the population is benignly infected with various strains of human papillomavirus (HPV), long-term infection by a subset of HPV strains is associated with malignant transformation. The prospects for prophylaxis against HPV infection have recently received an enormous boost with the approval by the US FDA of a vaccine targeted against the most common cancer-associated HPV strains. However, the large number of people already infected, the high cost of the vaccination regimen (particularly in poorer countries) and the HPV infections that these vaccines do not protect against underscore the need for therapeutic strategies. The elucidation of molecular details underlying fundamental processes in the viral life cycle, such as virus replication, transcription and HPV-induced carcinogenesis, is required to meet this aim. This article provides an overview of high-resolution structures of papillomavirus proteins and their functional complexes, with particular reference to mechanistic and structural features that could be exploited in the rational design of antiviral agents.
Collapse
Affiliation(s)
- Rashmi S Hegde
- Cincinnati Children’s Hospital Medical Center, Division of Developmental Biology, University of Cincinnati School of Medicine, Department of Pediatrics, 3333 Burnet Avenue, Cincinnati, OH 45229, USA
| |
Collapse
|
144
|
Baleja JD, Cherry JJ, Liu Z, Gao H, Nicklaus MC, Voigt JH, Chen JJ, Androphy EJ. Identification of inhibitors to papillomavirus type 16 E6 protein based on three-dimensional structures of interacting proteins. Antiviral Res 2006; 72:49-59. [PMID: 16690141 PMCID: PMC2776632 DOI: 10.1016/j.antiviral.2006.03.014] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2005] [Revised: 03/22/2006] [Accepted: 03/24/2006] [Indexed: 11/25/2022]
Abstract
Human papillomaviruses (HPV) cause cutaneous and genital warts. A subset of HPV types is associated with a high-risk for progression to malignancy. The E6 protein from the high-risk HPV types represents an attractive target for intervention because of its roles in viral propagation and cellular transformation. E6 functions in part by interaction with human cellular proteins, several of which possess a helical E6-binding motif. The role for each amino acid in this motif for binding E6 has been tested through structure determination and site-directed mutagenesis. These structural and molecular biological approaches defined the spatial geometry of functional groups necessary for binding to E6. This E6-binding information (the E6-binding pharmacophore) was transferred into a three-dimensional query format suitable for computational screening of large chemical databases. Compounds were identified and tested using in vitro and cell culture-based assays. Several compounds selectively inhibited E6 interaction with the E6-binding protein E6AP and interfered with the ability of E6 to promote p53 degradation. Such compounds provide leads for the development of new pharmacologic agents to treat papillomavirus infections and their associated cancers.
Collapse
Affiliation(s)
- James D. Baleja
- Dept. of Biochemistry, Tufts University School of Medicine, 136 Harrison Ave., Boston, MA 02111 U.S.A
| | - Jonathan J. Cherry
- Dept. of Medicine, University of Massachusetts Medical School, 55 Lake Avenue North, Worcester, MA 01605, U.S.A
| | - Zhiguo Liu
- Dept. of Medicine, University of Massachusetts Medical School, 55 Lake Avenue North, Worcester, MA 01605, U.S.A
| | - Hua Gao
- Dept. of Medicine, University of Massachusetts Medical School, 55 Lake Avenue North, Worcester, MA 01605, U.S.A
| | - Marc C. Nicklaus
- Laboratory of Medicinal Chemistry, Center for Cancer Research, National Cancer Institute, 376 Boyles Street, Frederick, MD 21702 U.S.A
| | - Johannes H. Voigt
- Laboratory of Medicinal Chemistry, Center for Cancer Research, National Cancer Institute, 376 Boyles Street, Frederick, MD 21702 U.S.A
| | - Jason J. Chen
- Dept. of Medicine, University of Massachusetts Medical School, 55 Lake Avenue North, Worcester, MA 01605, U.S.A
| | - Elliot J. Androphy
- Dept. of Medicine, University of Massachusetts Medical School, 55 Lake Avenue North, Worcester, MA 01605, U.S.A
| |
Collapse
|
145
|
Zanier K, Nominé Y, Charbonnier S, Ruhlmann C, Schultz P, Schweizer J, Travé G. Formation of well-defined soluble aggregates upon fusion to MBP is a generic property of E6 proteins from various human papillomavirus species. Protein Expr Purif 2006; 51:59-70. [PMID: 17055740 DOI: 10.1016/j.pep.2006.07.029] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2006] [Revised: 06/27/2006] [Accepted: 07/03/2006] [Indexed: 10/24/2022]
Abstract
Protein aggregation is a main barrier hindering structural and functional studies of a number of interesting biological targets. The E6 oncoprotein of Human Papillomavirus strain 16 (E6(16)) is difficult to express under a native soluble form in bacteria. Produced as an unfused sequence, it forms inclusion bodies. Fused to the C-terminus of MBP, it is mainly produced in the form of soluble high molecular weight aggregates. Here, we produced as MBP-fusions seven E6 proteins from other HPV strains (5, 11, 18, 33, 45, 52, and 58) belonging to four different species, and we compared their aggregation state to that of MBP-E6(16). Using a fast mutagenesis method, we changed most non-conserved cysteines to the isosteric residue serine to minimize disulfide bridge-mediated aggregation during purification. Static and dynamic light scattering measurements, ultracentrifugation and electron microscopy demonstrated the presence in all MBP-E6 preparations of soluble high-molecular weight aggregates with a well-defined spherical shape. These aggregated particles are relatively monodisperse but their amount and their size vary depending on the conditions of expression and the strain considered. For all strains, minimal aggregate formation occurs when the expression is performed at 15 degrees C. Such observations suggest that the assembly of MBP-E6 aggregates takes place in vivo during protein biosynthesis, rather than occurring during purification. Finally, we show that all MBP-E6 preparations contain two zinc ions per protein monomer, suggesting that E6 domains within the high molecular weight aggregates possess a native-like fold, which enables correct coordination to the metal center.
Collapse
Affiliation(s)
- Katia Zanier
- Ecole Supérieure de Biotechnologie de Strasbourg, Institut Gilbert Laustriat (UMR 7175 LC1), Boulevard Sébastien Brant, BP 10413, 67412 Illkirch, France
| | | | | | | | | | | | | |
Collapse
|
146
|
Affiliation(s)
- Xin Liu
- Wistar Institute and Department of Chemistry, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | | |
Collapse
|