1
|
Trammel J, Amusan O, Hultgren A, Raikhy G, Bodily JM. Epidermal growth factor receptor-dependent stimulation of differentiation by human papillomavirus type 16 E5. Virology 2024; 590:109952. [PMID: 38103269 PMCID: PMC10842332 DOI: 10.1016/j.virol.2023.109952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 11/14/2023] [Accepted: 11/22/2023] [Indexed: 12/19/2023]
Abstract
Human papillomaviruses (HPVs) infect keratinocytes of stratified squamous epithelia, and persistent infection with high-risk HPV types, such as HPV16, may lead to the development of malignancies. HPV evades host immunity in part by linking its gene expression to the host differentiation program, and therefore relies on differentiation to complete its life cycle. Based on previous reports indicating that the HPV16 protein E5 is important in the late stages of the differentiation-dependent life cycle, we found that organotypic cultures harboring HPV16 genomes lacking E5 showed reduced markers of terminal differentiation compared to wild type HPV16-containing cultures. We found that epidermal growth factor receptor (EGFR) levels and activation were increased in an E5-depdendent manner in these tissues, and that EGFR promoted terminal differentiation and expression of the HPV16 L1 gene. These findings suggest a function for E5 in preserving the ability of HPV16 containing keratinocytes to differentiate, thus facilitating the production of new virus progeny.
Collapse
Affiliation(s)
- Jessica Trammel
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center - Shreveport, Shreveport, LA, 71103, USA
| | - Oluwamuyiwa Amusan
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center - Shreveport, Shreveport, LA, 71103, USA
| | - Allison Hultgren
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center - Shreveport, Shreveport, LA, 71103, USA; School of Medicine, Louisiana State University Health Sciences Center - Shreveport, Shreveport, LA, 71103, USA
| | - Gaurav Raikhy
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center - Shreveport, Shreveport, LA, 71103, USA
| | - Jason M Bodily
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center - Shreveport, Shreveport, LA, 71103, USA.
| |
Collapse
|
2
|
Kogure G, Tanaka K, Matsui T, Onuki M, Matsumoto K, Iwata T, Kukimoto I. Intra-Patient Genomic Variations of Human Papillomavirus Type 31 in Cervical Cancer and Precancer. Viruses 2023; 15:2104. [PMID: 37896881 PMCID: PMC10612030 DOI: 10.3390/v15102104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Revised: 10/13/2023] [Accepted: 10/14/2023] [Indexed: 10/29/2023] Open
Abstract
Human papillomavirus type 31 (HPV31) is detected less frequently in cervical cancer than two major causative types, HPV16 and HPV18. Here, we report a comprehensive analysis of HPV31 genome sequences in cervical lesions collected from Japanese women. Of 52 HPV31-positive cervical specimens analyzed by deep sequencing, 43 samples yielded complete genome sequences of around 7900 base pairs and 9 samples yielded partially deleted genome sequences. Phylogenetic analysis showed that HPV31 variant distribution was lineage A in 19 samples (36.5%), lineage B in 28 samples (53.8%), and lineage C in 5 samples (9.6%), indicating that lineage B variants are dominant among HPV31 infections in Japan. Deletions in the viral genome were found in the region from the E1 to L1 genes, but all the deleted genomes retained the E6/E7 genes. Among intra-patient nucleotide variations relative to a consensus genome sequence in each sample, C-to-T substitutions were most frequently detected, followed by T-to-C and C-to-A substitutions. High-frequency, intra-patient mutations (>10%) in cervical cancer samples were found in the E1, E2, and E7 genes, and all of them were nonsynonymous substitutions. The enrichment of high-frequency nonsynonymous substitutions strongly suggests that these intra-patient mutations are positively selected during the development of cervical cancer/precancer.
Collapse
Affiliation(s)
- Gota Kogure
- Department of Obstetrics and Gynecology, Showa University School of Medicine, Tokyo 142-8666, Japan; (G.K.); (M.O.); (K.M.)
| | - Kohsei Tanaka
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo 160-0016, Japan; (K.T.); (T.M.); (T.I.)
| | - Tomoya Matsui
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo 160-0016, Japan; (K.T.); (T.M.); (T.I.)
| | - Mamiko Onuki
- Department of Obstetrics and Gynecology, Showa University School of Medicine, Tokyo 142-8666, Japan; (G.K.); (M.O.); (K.M.)
| | - Koji Matsumoto
- Department of Obstetrics and Gynecology, Showa University School of Medicine, Tokyo 142-8666, Japan; (G.K.); (M.O.); (K.M.)
| | - Takashi Iwata
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo 160-0016, Japan; (K.T.); (T.M.); (T.I.)
| | - Iwao Kukimoto
- Pathogen Genomics Center, National Institute of Infectious Diseases, Tokyo 208-0011, Japan
| |
Collapse
|
3
|
Yu L, Majerciak V, Zheng ZM. HPV16 and HPV18 Genome Structure, Expression, and Post-Transcriptional Regulation. Int J Mol Sci 2022; 23:ijms23094943. [PMID: 35563334 PMCID: PMC9105396 DOI: 10.3390/ijms23094943] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 04/14/2022] [Accepted: 04/15/2022] [Indexed: 12/18/2022] Open
Abstract
Human papillomaviruses (HPV) are a group of small non-enveloped DNA viruses whose infection causes benign tumors or cancers. HPV16 and HPV18, the two most common high-risk HPVs, are responsible for ~70% of all HPV-related cervical cancers and head and neck cancers. The expression of the HPV genome is highly dependent on cell differentiation and is strictly regulated at the transcriptional and post-transcriptional levels. Both HPV early and late transcripts differentially expressed in the infected cells are intron-containing bicistronic or polycistronic RNAs bearing more than one open reading frame (ORF), because of usage of alternative viral promoters and two alternative viral RNA polyadenylation signals. Papillomaviruses proficiently engage alternative RNA splicing to express individual ORFs from the bicistronic or polycistronic RNA transcripts. In this review, we discuss the genome structures and the updated transcription maps of HPV16 and HPV18, and the latest research advances in understanding RNA cis-elements, intron branch point sequences, and RNA-binding proteins in the regulation of viral RNA processing. Moreover, we briefly discuss the epigenetic modifications, including DNA methylation and possible APOBEC-mediated genome editing in HPV infections and carcinogenesis.
Collapse
|
4
|
Carobeli LR, Meirelles LEDF, Damke GMZF, Damke E, de Souza MVF, Mari NL, Mashiba KH, Shinobu-Mesquita CS, Souza RP, da Silva VRS, Gonçalves RS, Caetano W, Consolaro MEL. Phthalocyanine and Its Formulations: A Promising Photosensitizer for Cervical Cancer Phototherapy. Pharmaceutics 2021; 13:pharmaceutics13122057. [PMID: 34959339 PMCID: PMC8705941 DOI: 10.3390/pharmaceutics13122057] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 10/19/2021] [Accepted: 10/21/2021] [Indexed: 12/13/2022] Open
Abstract
Cervical cancer is one of the most common causes of cancer-related deaths in women worldwide. Despite advances in current therapies, women with advanced or recurrent disease present poor prognosis. Photodynamic therapy (PDT) has emerged as an effective therapeutic alternative to treat oncological diseases such as cervical cancer. Phthalocyanines (Pcs) are considered good photosensitizers (PS) for PDT, although most of them present high levels of aggregation and are lipophilic. Despite many investigations and encouraging results, Pcs have not been approved as PS for PDT of invasive cervical cancer yet. This review presents an overview on the pathophysiology of cervical cancer and summarizes the most recent developments on the physicochemical properties of Pcs and biological results obtained both in vitro in tumor-bearing mice and in clinical tests reported in the last five years. Current evidence indicates that Pcs have potential as pharmaceutical agents for anti-cervical cancer therapy. The authors firmly believe that Pc-based formulations could emerge as a privileged scaffold for the establishment of lead compounds for PDT against different types of cervical cancer.
Collapse
Affiliation(s)
- Lucimara R. Carobeli
- Department of Clinical Analysis and Biomedicine, Universidade Estadual de Maringá, Maringá 87020-900, PR, Brazil; (L.R.C.); (L.E.d.F.M.); (G.M.Z.F.D.); (E.D.); (M.V.F.d.S.); (N.L.M.); (K.H.M.); (C.S.S.-M.); (R.P.S.); (V.R.S.d.S.)
| | - Lyvia E. de F. Meirelles
- Department of Clinical Analysis and Biomedicine, Universidade Estadual de Maringá, Maringá 87020-900, PR, Brazil; (L.R.C.); (L.E.d.F.M.); (G.M.Z.F.D.); (E.D.); (M.V.F.d.S.); (N.L.M.); (K.H.M.); (C.S.S.-M.); (R.P.S.); (V.R.S.d.S.)
| | - Gabrielle M. Z. F. Damke
- Department of Clinical Analysis and Biomedicine, Universidade Estadual de Maringá, Maringá 87020-900, PR, Brazil; (L.R.C.); (L.E.d.F.M.); (G.M.Z.F.D.); (E.D.); (M.V.F.d.S.); (N.L.M.); (K.H.M.); (C.S.S.-M.); (R.P.S.); (V.R.S.d.S.)
| | - Edilson Damke
- Department of Clinical Analysis and Biomedicine, Universidade Estadual de Maringá, Maringá 87020-900, PR, Brazil; (L.R.C.); (L.E.d.F.M.); (G.M.Z.F.D.); (E.D.); (M.V.F.d.S.); (N.L.M.); (K.H.M.); (C.S.S.-M.); (R.P.S.); (V.R.S.d.S.)
| | - Maria V. F. de Souza
- Department of Clinical Analysis and Biomedicine, Universidade Estadual de Maringá, Maringá 87020-900, PR, Brazil; (L.R.C.); (L.E.d.F.M.); (G.M.Z.F.D.); (E.D.); (M.V.F.d.S.); (N.L.M.); (K.H.M.); (C.S.S.-M.); (R.P.S.); (V.R.S.d.S.)
| | - Natália L. Mari
- Department of Clinical Analysis and Biomedicine, Universidade Estadual de Maringá, Maringá 87020-900, PR, Brazil; (L.R.C.); (L.E.d.F.M.); (G.M.Z.F.D.); (E.D.); (M.V.F.d.S.); (N.L.M.); (K.H.M.); (C.S.S.-M.); (R.P.S.); (V.R.S.d.S.)
| | - Kayane H. Mashiba
- Department of Clinical Analysis and Biomedicine, Universidade Estadual de Maringá, Maringá 87020-900, PR, Brazil; (L.R.C.); (L.E.d.F.M.); (G.M.Z.F.D.); (E.D.); (M.V.F.d.S.); (N.L.M.); (K.H.M.); (C.S.S.-M.); (R.P.S.); (V.R.S.d.S.)
| | - Cristiane S. Shinobu-Mesquita
- Department of Clinical Analysis and Biomedicine, Universidade Estadual de Maringá, Maringá 87020-900, PR, Brazil; (L.R.C.); (L.E.d.F.M.); (G.M.Z.F.D.); (E.D.); (M.V.F.d.S.); (N.L.M.); (K.H.M.); (C.S.S.-M.); (R.P.S.); (V.R.S.d.S.)
| | - Raquel P. Souza
- Department of Clinical Analysis and Biomedicine, Universidade Estadual de Maringá, Maringá 87020-900, PR, Brazil; (L.R.C.); (L.E.d.F.M.); (G.M.Z.F.D.); (E.D.); (M.V.F.d.S.); (N.L.M.); (K.H.M.); (C.S.S.-M.); (R.P.S.); (V.R.S.d.S.)
| | - Vânia R. S. da Silva
- Department of Clinical Analysis and Biomedicine, Universidade Estadual de Maringá, Maringá 87020-900, PR, Brazil; (L.R.C.); (L.E.d.F.M.); (G.M.Z.F.D.); (E.D.); (M.V.F.d.S.); (N.L.M.); (K.H.M.); (C.S.S.-M.); (R.P.S.); (V.R.S.d.S.)
| | - Renato S. Gonçalves
- Department of Chemistry, Universidade Estadual de Maringá, Maringá 87020-900, PR, Brazil; (R.S.G.); (W.C.)
| | - Wilker Caetano
- Department of Chemistry, Universidade Estadual de Maringá, Maringá 87020-900, PR, Brazil; (R.S.G.); (W.C.)
| | - Márcia E. L. Consolaro
- Department of Clinical Analysis and Biomedicine, Universidade Estadual de Maringá, Maringá 87020-900, PR, Brazil; (L.R.C.); (L.E.d.F.M.); (G.M.Z.F.D.); (E.D.); (M.V.F.d.S.); (N.L.M.); (K.H.M.); (C.S.S.-M.); (R.P.S.); (V.R.S.d.S.)
- Correspondence: ; Tel.: +55-44-3011-5455
| |
Collapse
|
5
|
Ferguson J, Campos-León K, Pentland I, Stockton JD, Günther T, Beggs AD, Grundhoff A, Roberts S, Noyvert B, Parish JL. The chromatin insulator CTCF regulates HPV18 transcript splicing and differentiation-dependent late gene expression. PLoS Pathog 2021; 17:e1010032. [PMID: 34735550 PMCID: PMC8594839 DOI: 10.1371/journal.ppat.1010032] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 11/16/2021] [Accepted: 10/13/2021] [Indexed: 11/18/2022] Open
Abstract
The ubiquitous host protein, CCCTC-binding factor (CTCF), is an essential regulator of cellular transcription and functions to maintain epigenetic boundaries, stabilise chromatin loops and regulate splicing of alternative exons. We have previously demonstrated that CTCF binds to the E2 open reading frame (ORF) of human papillomavirus (HPV) 18 and functions to repress viral oncogene expression in undifferentiated keratinocytes by co-ordinating an epigenetically repressed chromatin loop within HPV episomes. Keratinocyte differentiation disrupts CTCF-dependent chromatin looping of HPV18 episomes promoting induction of enhanced viral oncogene expression. To further characterise CTCF function in HPV transcription control we utilised direct, long-read Nanopore RNA-sequencing which provides information on the structure and abundance of full-length transcripts. Nanopore analysis of primary human keratinocytes containing HPV18 episomes before and after synchronous differentiation allowed quantification of viral transcript species, including the identification of low abundance novel transcripts. Comparison of transcripts produced in wild type HPV18 genome-containing cells to those identified in CTCF-binding deficient genome-containing cells identifies CTCF as a key regulator of differentiation-dependent late promoter activation, required for efficient E1^E4 and L1 protein expression. Furthermore, our data show that CTCF binding at the E2 ORF promotes usage of the downstream weak splice donor (SD) sites SD3165 and SD3284, to the dominant E4 splice acceptor site at nucleotide 3434. These findings demonstrate that in the HPV life cycle both early and late virus transcription programmes are facilitated by recruitment of CTCF to the E2 ORF. Oncogenic human papillomavirus (HPV) infection is the cause of a subset of epithelial cancers of the uterine cervix, other anogenital areas and the oropharynx. HPV infection is established in the basal cells of epithelia where a restricted programme of viral gene expression is required for replication and maintenance of the viral episome. Completion of the HPV life cycle is dependent on the maturation (differentiation) of infected cells which induces enhanced viral gene expression and induction of capsid production. We previously reported that the host cell transcriptional regulator, CTCF, is hijacked by HPV to control viral gene expression. In this study, we use long-read mRNA sequencing to quantitatively map the variety and abundance of HPV transcripts produced in early and late stages of the HPV life cycle and to dissect the function of CTCF in controlling HPV gene expression and transcript processing.
Collapse
Affiliation(s)
- Jack Ferguson
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, West Midlands, United Kingdom
| | - Karen Campos-León
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, West Midlands, United Kingdom
| | - Ieisha Pentland
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, West Midlands, United Kingdom
| | - Joanne D. Stockton
- Genomics Birmingham, University of Birmingham, Birmingham, West Midlands, United Kingdom
| | - Thomas Günther
- Heinrich-Pette Institute, Leibniz Institute for Experimental Virology, Hamburg, Germany
| | - Andrew D. Beggs
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, West Midlands, United Kingdom
- Genomics Birmingham, University of Birmingham, Birmingham, West Midlands, United Kingdom
| | - Adam Grundhoff
- Heinrich-Pette Institute, Leibniz Institute for Experimental Virology, Hamburg, Germany
| | - Sally Roberts
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, West Midlands, United Kingdom
| | - Boris Noyvert
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, West Midlands, United Kingdom
- CRUK Birmingham Centre and Centre for Computational Biology, University of Birmingham, Birmingham, West Midlands, United Kingdom
| | - Joanna L. Parish
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, West Midlands, United Kingdom
- * E-mail:
| |
Collapse
|
6
|
Epigenetic Regulation of the Human Papillomavirus Life Cycle. Pathogens 2020; 9:pathogens9060483. [PMID: 32570816 PMCID: PMC7350343 DOI: 10.3390/pathogens9060483] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 06/16/2020] [Accepted: 06/17/2020] [Indexed: 12/20/2022] Open
Abstract
Persistent infection with certain types of human papillomaviruses (HPVs), termed high risk, presents a public health burden due to their association with multiple human cancers, including cervical cancer and an increasing number of head and neck cancers. Despite the development of prophylactic vaccines, the incidence of HPV-associated cancers remains high. In addition, no vaccine has yet been licensed for therapeutic use against pre-existing HPV infections and HPV-associated diseases. Although persistent HPV infection is the major risk factor for cancer development, additional genetic and epigenetic alterations are required for progression to the malignant phenotype. Unlike genetic mutations, the reversibility of epigenetic modifications makes epigenetic regulators ideal therapeutic targets for cancer therapy. This review article will highlight the recent advances in the understanding of epigenetic modifications associated with HPV infections, with a particular focus on the role of these epigenetic changes during different stages of the HPV life cycle that are closely associated with activation of DNA damage response pathways.
Collapse
|
7
|
Gautam D, Johnson BA, Mac M, Moody CA. SETD2-dependent H3K36me3 plays a critical role in epigenetic regulation of the HPV31 life cycle. PLoS Pathog 2018; 14:e1007367. [PMID: 30312361 PMCID: PMC6200281 DOI: 10.1371/journal.ppat.1007367] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 10/24/2018] [Accepted: 09/28/2018] [Indexed: 12/14/2022] Open
Abstract
The life cycle of HPV is tied to the differentiation status of its host cell, with productive replication, late gene expression and virion production restricted to the uppermost layers of the stratified epithelium. HPV DNA is histone-associated, exhibiting a chromatin structure similar to that of the host chromosome. Although HPV chromatin is subject to histone post-translational modifications, how the viral life cycle is epigenetically regulated is not well understood. SETD2 is a histone methyltransferase that places the trimethyl mark on H3K36 (H3K36me3), a mark of active transcription. Here, we define a role for SETD2 and H3K36me3 in the viral life cycle. We have found that HPV positive cells exhibit increased levels of SETD2, with SETD2 depletion leading to defects in productive viral replication and splicing of late viral RNAs. Reducing H3K36me3 by overexpression of KDM4A, an H3K36me3 demethylase, or an H3.3K36M transgene also blocks productive viral replication, indicating a significant role for this histone modification in facilitating viral processes. H3K36me3 is enriched on the 3' end of the early region of the high-risk HPV31 genome in a SETD2-dependent manner, suggesting that SETD2 may regulate the viral life cycle through the recruitment of H3K36me3 readers to viral DNA. Intriguingly, we have found that activation of the ATM DNA damage kinase, which is required for productive viral replication, is necessary for the maintenance of H3K36me3 on viral chromatin and for processing of late viral RNAs. Additionally, we have found that the HPV31 E7 protein maintains the increased SETD2 levels in infected cells through an extension of protein half-life. Collectively, our findings highlight the importance of epigenetic modifications in driving the viral life cycle and identify a novel role for E7 as well as the DNA damage response in the regulation of viral processes through epigenetic modifications.
Collapse
Affiliation(s)
- Dipendra Gautam
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Bryan A. Johnson
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Michelle Mac
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Cary A. Moody
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- * E-mail:
| |
Collapse
|
8
|
Xue XY, Majerciak V, Uberoi A, Kim BH, Gotte D, Chen X, Cam M, Lambert PF, Zheng ZM. The full transcription map of mouse papillomavirus type 1 (MmuPV1) in mouse wart tissues. PLoS Pathog 2017; 13:e1006715. [PMID: 29176795 PMCID: PMC5720830 DOI: 10.1371/journal.ppat.1006715] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Revised: 12/07/2017] [Accepted: 10/25/2017] [Indexed: 12/17/2022] Open
Abstract
Mouse papillomavirus type 1 (MmuPV1) provides, for the first time, the opportunity to study infection and pathogenesis of papillomaviruses in the context of laboratory mice. In this report, we define the transcriptome of MmuPV1 genome present in papillomas arising in experimentally infected mice using a combination of RNA-seq, PacBio Iso-seq, 5’ RACE, 3’ RACE, primer-walking RT-PCR, RNase protection, Northern blot and in situ hybridization analyses. We demonstrate that the MmuPV1 genome is transcribed unidirectionally from five major promoters (P) or transcription start sites (TSS) and polyadenylates its transcripts at two major polyadenylation (pA) sites. We designate the P7503, P360 and P859 as “early” promoters because they give rise to transcripts mostly utilizing the polyadenylation signal at nt 3844 and therefore can only encode early genes, and P7107 and P533 as “late” promoters because they give rise to transcripts utilizing polyadenylation signals at either nt 3844 or nt 7047, the latter being able to encode late, capsid proteins. MmuPV1 genome contains five splice donor sites and three acceptor sites that produce thirty-six RNA isoforms deduced to express seven predicted early gene products (E6, E7, E1, E1^M1, E1^M2, E2 and E8^E2) and three predicted late gene products (E1^E4, L2 and L1). The majority of the viral early transcripts are spliced once from nt 757 to 3139, while viral late transcripts, which are predicted to encode L1, are spliced twice, first from nt 7243 to either nt 3139 (P7107) or nt 757 to 3139 (P533) and second from nt 3431 to nt 5372. Thirteen of these viral transcripts were detectable by Northern blot analysis, with the P533-derived late E1^E4 transcripts being the most abundant. The late transcripts could be detected in highly differentiated keratinocytes of MmuPV1-infected tissues as early as ten days after MmuPV1 inoculation and correlated with detection of L1 protein and viral DNA amplification. In mature warts, detection of L1 was also found in more poorly differentiated cells, as previously reported. Subclinical infections were also observed. The comprehensive transcription map of MmuPV1 generated in this study provides further evidence that MmuPV1 is similar to high-risk cutaneous beta human papillomaviruses. The knowledge revealed will facilitate the use of MmuPV1 as an animal virus model for understanding of human papillomavirus gene expression, pathogenesis and immunology. Papillomavirus (PV) infections lead to development of both benign warts and cancers. Because PVs are epitheliotropic and species specific, it has been extremely challenging to study PV infection in the context of a naturally occurring infection in a tractable laboratory animal. The recent discovery of the papillomavirus, MmuPV1, that infects laboratory mice, provides an important new animal model system for understanding the pathogenesis of papillomavirus-associated diseases. By using state of the art RNA-seq to provide deep sequencing analysis of what regions of the viral genome are transcribed and PacBio Iso-seq that produces longer reads to define the complete sequences of individual transcripts in combination with several conventional technologies to confirm transcription starts sites, splice sites, and polyadenylation sites, we provide the first detailed description of the MmuPV1 transcript map using RNA from MmuPV1-induced mouse warts. This study reveals the presence of mRNA transcripts capable of coding for ten protein products in the MmuPV1 genome and leads to correctly re-assigning the E1^E4, L2 and L1 coding regions. We were able to detect individual transcripts from the infected wart tissues by RT-PCR, Northern blot and RNA ISH, to define the temporal onset of productive viral infection and to ectopically express a predicted viral protein for functional studies. The constructed MmuPV1 transcript map provides a foundation to advance our understanding of papillomavirus biology and pathogenesis.
Collapse
Affiliation(s)
- Xiang-Yang Xue
- Tumor Virus RNA Biology Section, RNA Biology Laboratory, Center for Cancer Research, NCI/NIH, Frederick, Maryland, United States of America
- Department of Microbiology and Immunology, Wenzhou Medical University, Zhejiang, China
| | - Vladimir Majerciak
- Tumor Virus RNA Biology Section, RNA Biology Laboratory, Center for Cancer Research, NCI/NIH, Frederick, Maryland, United States of America
| | - Aayushi Uberoi
- McArdle Laboratory for Cancer Research, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, United States of America
| | - Bong-Hyun Kim
- Collaborative Bioinformatics Resource, Center for Cancer Research, NCI/NIH, Bethesda, Maryland, United States of America
- Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Frederick, Maryland, United States of America
| | - Deanna Gotte
- Tumor Virus RNA Biology Section, RNA Biology Laboratory, Center for Cancer Research, NCI/NIH, Frederick, Maryland, United States of America
| | - Xiongfong Chen
- Collaborative Bioinformatics Resource, Center for Cancer Research, NCI/NIH, Bethesda, Maryland, United States of America
- Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Frederick, Maryland, United States of America
| | - Maggie Cam
- Collaborative Bioinformatics Resource, Center for Cancer Research, NCI/NIH, Bethesda, Maryland, United States of America
| | - Paul F. Lambert
- McArdle Laboratory for Cancer Research, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, United States of America
| | - Zhi-Ming Zheng
- Tumor Virus RNA Biology Section, RNA Biology Laboratory, Center for Cancer Research, NCI/NIH, Frederick, Maryland, United States of America
- * E-mail:
| |
Collapse
|
9
|
Graham SV. Keratinocyte Differentiation-Dependent Human Papillomavirus Gene Regulation. Viruses 2017; 9:E245. [PMID: 28867768 PMCID: PMC5618011 DOI: 10.3390/v9090245] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Revised: 08/24/2017] [Accepted: 08/25/2017] [Indexed: 12/29/2022] Open
Abstract
Human papillomaviruses (HPVs) cause diseases ranging from benign warts to invasive cancers. HPVs infect epithelial cells and their replication cycle is tightly linked with the differentiation process of the infected keratinocyte. The normal replication cycle involves an early and a late phase. The early phase encompasses viral entry and initial genome replication, stimulation of cell division and inhibition of apoptosis in the infected cell. Late events in the HPV life cycle include viral genome amplification, virion formation, and release into the environment from the surface of the epithelium. The main proteins required at the late stage of infection for viral genome amplification include E1, E2, E4 and E5. The late proteins L1 and L2 are structural proteins that form the viral capsid. Regulation of these late events involves both cellular and viral proteins. The late viral mRNAs are expressed from a specific late promoter but final late mRNA levels in the infected cell are controlled by splicing, polyadenylation, nuclear export and RNA stability. Viral late protein expression is also controlled at the level of translation. This review will discuss current knowledge of how HPV late gene expression is regulated.
Collapse
Affiliation(s)
- Sheila V Graham
- MRC-University of Glasgow Centre for Virus Research, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Garscube Estate, Glasgow G61 1QH, UK.
| |
Collapse
|
10
|
Human Papillomavirus E2 Regulates SRSF3 (SRp20) To Promote Capsid Protein Expression in Infected Differentiated Keratinocytes. J Virol 2016; 90:5047-58. [PMID: 26962216 DOI: 10.1128/jvi.03073-15] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Accepted: 02/29/2016] [Indexed: 12/15/2022] Open
Abstract
UNLABELLED The human papillomavirus (HPV) life cycle is tightly linked to differentiation of the infected epithelial cell, suggesting a sophisticated interplay between host cell metabolism and virus replication. Previously, we demonstrated in differentiated keratinocytes in vitro and in vivo that HPV type 16 (HPV16) infection caused increased levels of the cellular SR splicing factors (SRSFs) SRSF1 (ASF/SF2), SRSF2 (SC35), and SRSF3 (SRp20). Moreover, the viral E2 transcription and replication factor that is expressed at high levels in differentiating keratinocytes could bind and control activity of the SRSF1 gene promoter. Here, we show that the E2 proteins of HPV16 and HPV31 control the expression of SRSFs 1, 2, and 3 in a differentiation-dependent manner. E2 has the greatest transactivation effect on expression of SRSF3. Small interfering RNA depletion experiments in two different models of the HPV16 life cycle (W12E and NIKS16) and one model of the HPV31 life cycle (CIN612-9E) revealed that only SRSF3 contributed significantly to regulation of late events in the virus life cycle. Increased levels of SRSF3 are required for L1 mRNA and capsid protein expression. Capsid protein expression was regulated specifically by SRSF3 and appeared independent of other SRSFs. Taken together, these data suggest a significant role of the HPV E2 protein in regulating late events in the HPV life cycle through transcriptional regulation of SRSF3 expression. IMPORTANCE Human papillomavirus replication is accomplished in concert with differentiation of the infected epithelium. Virus capsid protein expression is confined to the upper epithelial layers so as to avoid immune detection. In this study, we demonstrate that the viral E2 transcription factor activates the promoter of the cellular SRSF3 RNA processing factor. SRSF3 is required for expression of the E4(^)L1 mRNA and so controls expression of the HPV L1 capsid protein. Thus, we reveal a new dimension of virus-host interaction crucial for production of infectious virus. SRSF proteins are known drug targets. Therefore, this study provides an excellent basis for developing strategies to regulate capsid protein production in the infected epithelium and the production of new virions.
Collapse
|
11
|
Li X, Johansson C, Cardoso Palacios C, Mossberg A, Dhanjal S, Bergvall M, Schwartz S. Eight nucleotide substitutions inhibit splicing to HPV-16 3'-splice site SA3358 and reduce the efficiency by which HPV-16 increases the life span of primary human keratinocytes. PLoS One 2013; 8:e72776. [PMID: 24039800 PMCID: PMC3767658 DOI: 10.1371/journal.pone.0072776] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2013] [Accepted: 07/11/2013] [Indexed: 01/05/2023] Open
Abstract
The most commonly used 3'-splice site on the human papillomavirus type 16 (HPV-16) genome named SA3358 is used to produce HPV-16 early mRNAs encoding E4, E5, E6 and E7, and late mRNAs encoding L1 and L2. We have previously shown that SA3358 is suboptimal and is totally dependent on a downstream splicing enhancer containingmultiple potential ASF/SF2 binding sites. Here weshow that only one of the predicted ASF/SF2 sites accounts for the majority of the enhancer activity. We demonstrate that single nucleotide substitutions in this predicted ASF/SF2 site impair enhancer function and that this correlates with less efficient binding to ASF/SF2 in vitro. We provide evidence that HPV-16 mRNAs that arespliced to SA3358 interact with ASF/SF2 in living cells. In addition,mutational inactivation of the ASF/SF2 site weakened the enhancer at SA3358 in episomal forms of the HPV-16 genome, indicating that the enhancer is active in the context of the full HPV-16 genome.This resulted in induction of HPV-16 late gene expression as a result of competition from late splice site SA5639. Furthermore, inactivation of the ASF/SF2 site of the SA3358 splicing enhancer reduced the ability of E6- and E7-encoding HPV-16 plasmids to increase the life span of primary keratinocytes in vitro, demonstrating arequirement for an intact splicing enhancer of SA3358 forefficient production of the E6 and E7 mRNAs. These results link the strength of the HPV-16 SA3358 splicing enhancer to expression of E6 and E7 and to the pathogenic properties of HPV-16.
Collapse
Affiliation(s)
- Xiaoze Li
- Department of Laboratory Medicine, Lund University, Lund, Sweden
| | | | | | - Anki Mossberg
- Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Soniya Dhanjal
- Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Monika Bergvall
- Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Stefan Schwartz
- Department of Laboratory Medicine, Lund University, Lund, Sweden
- * E-mail:
| |
Collapse
|
12
|
Construction of a full transcription map of human papillomavirus type 18 during productive viral infection. J Virol 2011; 85:8080-92. [PMID: 21680515 DOI: 10.1128/jvi.00670-11] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Human papillomavirus type 18 (HPV18) is the second most common oncogenic HPV genotype, responsible for ∼15% of cervical cancers worldwide. In this study, we constructed a full HPV18 transcription map using HPV18-infected raft tissues derived from primary human vaginal or foreskin keratinocytes. By using 5' rapid amplification of cDNA ends (RACE), we mapped two HPV18 transcription start sites (TSS) for early transcripts at nucleotide (nt) 55 and nt 102 and the HPV18 late TSS frequently at nt 811, 765, or 829 within the E7 open reading frame (ORF) of the virus genome. HPV18 polyadenylation cleavage sites for early and late transcripts were mapped to nt 4270 and mainly to nt 7299 or 7307, respectively, by using 3' RACE. Although all early transcripts were cleaved exclusively at a single cleavage site, HPV18 late transcripts displayed the heterogeneity of 3' ends, with multiple minor cleavage sites for late RNA polyadenylation. HPV18 splice sites/splice junctions for both early and late transcripts were identified by 5' RACE and primer walking techniques. Five 5' splice sites (donor sites) and six 3' splice sites (acceptor sites) that are highly conserved in other papillomaviruses were identified in the HPV18 genome. HPV18 L1 mRNA translates a L1 protein of 507 amino acids (aa), smaller than the 568 aa residues previously predicted. Collectively, a full HPV18 transcription map constructed from this report will lead us to further understand HPV18 gene expression and virus oncogenesis.
Collapse
|
13
|
Multiple ASF/SF2 sites in the human papillomavirus type 16 (HPV-16) E4-coding region promote splicing to the most commonly used 3'-splice site on the HPV-16 genome. J Virol 2010; 84:8219-30. [PMID: 20519389 DOI: 10.1128/jvi.00462-10] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Our results presented here demonstrate that the most abundant human papillomavirus type 16 (HPV-16) mRNAs expressing the viral oncogenes E6 and E7 are regulated by cellular ASF/SF2, itself defined as a proto-oncogene and overexpressed in cervical cancer cells. We show that the most frequently used 3'-splice site on the HPV-16 genome, site SA3358, which is used to produce primarily E4, E6, and E7 mRNAs, is regulated by ASF/SF2. Splice site SA3358 is immediately followed by 15 potential binding sites for the splicing factor ASF/SF2. Recombinant ASF/SF2 binds to the cluster of ASF/SF2 sites. Mutational inactivation of all 15 sites abolished splicing to SA3358 and redirected splicing to the downstream-located, late 3'-splice site SA5639. Overexpression of a mutant ASF/SF2 protein that lacks the RS domain, also totally inhibited the usage of SA3358 and redirected splicing to the late 3'-splice site SA5639. The 15 ASF/SF2 binding sites could be replaced by an ASF/SF2-dependent, HIV-1-derived splicing enhancer named GAR. This enhancer was also inhibited by the mutant ASF/SF2 protein that lacks the RS domain. Finally, silencer RNA (siRNA)-mediated knockdown of ASF/SF2 caused a reduction in spliced HPV-16 mRNA levels. Taken together, our results demonstrate that the major HPV-16 3'-splice site SA3358 is dependent on ASF/SF2. SA3358 is used by the most abundantly expressed HPV-16 mRNAs, including those encoding E6 and E7. High levels of ASF/SF2 may therefore be a requirement for progression to cervical cancer. This is supported by our earlier findings that ASF/SF2 is overexpressed in high-grade cervical lesions and cervical cancer.
Collapse
|
14
|
Abstract
Human papillomaviruses complete their life cycle in differentiating epithelial cells that would not normally be competent for either cellular or viral DNA replication. To overcome this, papillomaviruses encode two groups of proteins that work together in the upper epithelial layers to amplify viral genomes. The E6 and E7 proteins play a critical role in driving differentiating epithelial cells that have left the basal layer, back into the cell cycle, in order to produce a replication-competent environment that can be used by the virus for genome amplification. Papillomavirus replication is heavily dependent on cellular replication proteins, but in addition needs the viral E1 and E2 proteins, which act to unwind viral DNA around the origin of replication, and to recruit essential cellular proteins to the replication site. Recent work using mutant viral genomes has suggested that two other viral proteins, E4 and E5, contribute to efficient replication in the upper epithelial layers, although the mechanisms by which they do this have not yet been clearly established. Genome amplification in the upper epithelial layers differs from maintenance replication in the basal layer, where viral genome replication appears coupled to that of the cellular genome. The onset of genome amplification during differentiation is thought to be triggered at least in part by an increase in E1 and E2 levels, and possibly also by a change in the relative levels of the two proteins. The role of E6 and E7 in basal cell replication is, however, uncertain and there is even some question as to the exact requirement for E1. Although similarities in papillomavirus lifecycle organization and protein function suggest a common mechanism by which viral DNA replication is regulated, differences in the site of infection and transmission route appear to manifest themselves as differences in the timing and extent of genome amplification. Understanding the patterns of protein expression seen during natural infection will be important in fully understanding how these differences arise.
Collapse
Affiliation(s)
- John Doorbar
- National Institute for Medical Research, Division of Virology, The Ridgeway, Mill Hill, London, NW7 1AA, UK
| | - Kenneth Raj
- National Institute for Medical Research, Division of Virology, The Ridgeway, Mill Hill, London, NW7 1AA, UK
| |
Collapse
|
15
|
Gu W, Ding J, Wang X, de Kluyver RL, Saunders NA, Frazer IH, Zhao KN. Generalized substitution of isoencoding codons shortens the duration of papillomavirus L1 protein expression in transiently gene-transfected keratinocytes due to cell differentiation. Nucleic Acids Res 2007; 35:4820-32. [PMID: 17621583 PMCID: PMC1950544 DOI: 10.1093/nar/gkm496] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Recently we reported that gene codon composition determines differentiation-dependent expression of the PV L1 genes in mouse primary keratinocytes (KCs) in vitro and in vivo (Zhao et al. 2005, Mol. Cell Biol. 25:8643–8655). Here, we investigated whether generalized substitution of isoencoding codons affects the duration of expression of PV L1 genes in mouse and human KCs in day 1 culture transiently transfected with native (Nat) and codon modified (Mod) L1 genes. Following transient transfection, KC continuously transcribed both Nat and Mod PV L1 genes for at least 12 days, with the levels of L1 mRNAs from the Mod L1 genes significantly higher than those from the Nat L1 genes. However, continuous L1 protein expression at day 9 post-transfection was observed for both mouse and human KCs transfected with the Nat L1 genes only. Further, aa-tRNAs prepared from D8 KC cultures enhanced translation of two PV Nat L1 DNAs in RRL lysate and PV Nat L1 mRNAs in D0 cell-free lysate, whereas aa-tRNAs from D0 KCs enhanced translation of PV Mod L1 mRNAs in D8 cell-free lysate. It appears that aa-tRNAs in less-differentiated and differentiated KCs differentially match the PV Nat and Mod L1 mRNAs to regulate their translations in vitro.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Kong-Nan Zhao
- *To whom correspondence should be addressed.+61 07 3240 5282+61 07 3240 5946
| |
Collapse
|
16
|
Fang NX, Gu W, Ding J, Saunders NA, Frazer IH, Zhao KN. Calcium enhances mouse keratinocyte differentiation in vitro to differentially regulate expression of papillomavirus authentic and codon modified L1 genes. Virology 2007; 365:187-97. [PMID: 17462691 DOI: 10.1016/j.virol.2007.03.038] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2007] [Revised: 02/12/2007] [Accepted: 03/19/2007] [Indexed: 11/27/2022]
Abstract
Here, we first wished to establish for mouse primary keratinocytes (KCs) the Ca(2+) concentrations that were associated with KC differentiation in vitro. Using the range of Ca(2+) concentrations (0-6 mM) to differentiate primary KCs in culture to varying extents for 2 days, we then examined how KC differentiation impacted on expression of papillomavirus (PV) native (Nat) and codon modified (Mod) L1 genes. L1 mRNAs transcribed from either Nat or Mod L1 genes were present in similar amounts in KCs exposed to six Ca(2+) concentrations. However, expression of the L1 proteins from two Mod L1 genes were down-regulated, with no L1 signal detected in KCs exposed to 6 mM Ca(2+). In contrast, L1 proteins expressed from the two Nat L1 genes were not detectable in KCs without Ca(2+), but dramatically up-regulated as the KC cultures exposed to Ca(2+) from 0.5 to 2 mM, then down-regulated in KCs exposed to Ca(2+) from 4 to 6 mM. The different regulatory roles of the Ca(2+) in L1 protein expression from Nat and Mod L1 genes in cultured KCs were confirmed by TGF-beta1 experiments. We observed that aminoacyl-tRNAs (aa-tRNAs) from the 2 mM Ca(2+)-treated KCs only significantly enhanced the Nat L1 mRNAs translation in vitro, suggesting that aa-tRNAs play a differentially regulatory role in translations of the PV Nat and Mod L1 mRNAs. Importantly, the Ca(2+) experimental model provides evidence that mouse primary KCs could be transiently infected by BPV1 virus to express L1 mRNA and protein, which is very useful for future HPV virus infection study.
Collapse
Affiliation(s)
- Ning-Xia Fang
- Diamantina Institute for Cancer, Immunology and Metabolic Medicine, The University of Queensland, Research Extension, Building 1, Princess Alexandra Hospital, Ipswich Road, Woolloongabba, Queensland 4102, Australia
| | | | | | | | | | | |
Collapse
|
17
|
Milligan SG, Veerapraditsin T, Ahamet B, Mole S, Graham SV. Analysis of novel human papillomavirus type 16 late mRNAs in differentiated W12 cervical epithelial cells. Virology 2006; 360:172-81. [PMID: 17098271 PMCID: PMC2151308 DOI: 10.1016/j.virol.2006.10.012] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2006] [Revised: 09/12/2006] [Accepted: 10/05/2006] [Indexed: 11/15/2022]
Abstract
The life cycle of human papillomavirus type 16 (HPV16) is intimately linked to differentiation of the epithelium it infects, and late events in the life cycle are restricted to the suprabasal layers. Here we have used 5′RACE of polyadenylated RNA isolated from differentiated W12 cells (cervical epithelial cells containing episomal copies of the HPV16 genome) that express virus late proteins to map virus late mRNAs. Thirteen different transcripts were identified. Extensive alternative splicing and use of two late polyadenylation sites were noted. A novel promoter located in the long control region was detected as well as P97 and Plate. Promoters in the E4 and E5 open reading frames were active yielding transcripts where L1 or L2 respectively are the first open reading frames. Finally, mRNAs that could encode novel proteins E6*^*E7, E6*^E4, E1^*E4 and E1^E2C (putative repressor E2) were identified, indicating that HPV16 may encode more late proteins than previously accepted.
Collapse
|
18
|
Zheng ZM, Baker CC. Papillomavirus genome structure, expression, and post-transcriptional regulation. FRONT BIOSCI-LANDMRK 2006; 11:2286-302. [PMID: 16720315 PMCID: PMC1472295 DOI: 10.2741/1971] [Citation(s) in RCA: 298] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Papillomaviruses are a group of small non-enveloped DNA tumor viruses whose infection usually causes benign epithelial lesions (warts). Certain types of HPVs, such as HPV-16, HPV-18, and HPV-31, have been recognized as causative agents of cervical cancer and anal cancer and their infections, which arise via sexual transmission, are associated with more than 95% of cervical cancer. Papillomaviruses infect keratinocytes in the basal layer of stratified squamous epithelia and replicate in the nucleus of infected keratinocytes in a differentiation-dependent manner. Viral gene expression in infected cells depends on cell differentiation and is tightly regulated at the transcriptional and post-transcriptional levels. A noteworthy feature of all papillomavirus transcripts is that they are transcribed as a bicistronic or polycistronic form containing two or more ORFs and are polyadenylated at either an early or late poly(A) site. In the past ten years, remarkable progress has been made in understanding how this complex viral gene expression is regulated at the level of transcription (such as via DNA methylation) and particularly post-transcription (including RNA splicing, polyadenylation, and translation). Current knowledge of papillomavirus mRNA structure and RNA processing has provided some clues on how to control viral oncogene expression. However, we still have little knowledge about which mRNAs are used to translate each viral protein. Continuing research on post-transcriptional regulation of papillomavirus infection will remain as a future focus to provide more insights into papillomavirus-host interactions, the virus life-cycle, and viral oncogenesis.
Collapse
Affiliation(s)
- Zhi-Ming Zheng
- HIV and AIDS Malignancy Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| | | |
Collapse
|
19
|
Drubin DA, McLaughlin-Drubin ME, Clawson GA, Meyers C. A Protease Inhibitor Specifically Inhibits Growth of HPV-Infected Keratinocytes. Mol Ther 2006; 13:1142-8. [PMID: 16564747 DOI: 10.1016/j.ymthe.2006.02.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2005] [Revised: 07/07/2005] [Accepted: 08/31/2005] [Indexed: 11/23/2022] Open
Abstract
Protease inhibitors have been known to exhibit anticarcinogenic activity in a variety of model systems, although the biological target(s) and mechanism remain enigmatic. Human papillomavirus (HPV) is the primary etiological agent of cervical cancer. Here we show that a nuclear chymotrypsin-like protease activity (NCLPA), which appears to be involved in transformation in several different experimental models, is significantly elevated in keratinocytes infected with high-risk HPV. Further, we demonstrate a marked growth inhibition of organotypic raft cultures, which is specific for cells infected with high-risk HPV types, using a chloromethyl ketone inhibitor previously shown to be relatively selective for the NCLPA. Surprisingly, this HPV-dependent inhibitory effect is independent of any alterations in the NCLPA. This finding has clear implications for the development of novel therapeutics specifically targeted to cervical dysplasias with HPV-infected cells.
Collapse
Affiliation(s)
- David A Drubin
- The Jake Gittlen Cancer Research Institute, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | | | | | | |
Collapse
|
20
|
Bodily JM, Alam S, Meyers C. Regulation of human papillomavirus type 31 late promoter activation and genome amplification by protein kinase C. Virology 2006; 348:328-40. [PMID: 16500689 DOI: 10.1016/j.virol.2006.01.036] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2005] [Revised: 09/19/2005] [Accepted: 01/26/2006] [Indexed: 10/25/2022]
Abstract
The life cycle of papillomaviruses is tightly linked to differentiation of host keratinocytes, but the mechanisms and cues by which life cycle events are tied to differentiation remain obscure. We have begun a systematic study of the differentiation-dependent life cycle of HPV31. A variety of signaling pathways have been implicated in controlling keratinocyte differentiation, especially the protein kinase C (PKC) pathway. We have used pharmacological inhibitors to determine that genome amplification and late transcription depend on specific PKC isoforms, and that transcription and replication are independently controlled. We found that tyrosine kinases are necessary for viral amplification but not viral transcription. These studies indicate that the PKC pathway is an important regulator of differentiation-dependent HPV31 replication and transcription.
Collapse
Affiliation(s)
- Jason M Bodily
- Department of Microbiology and Immunology, The Pennsylvania State University College of Medicine, Hershey, 17033, USA
| | | | | |
Collapse
|
21
|
Rush M, Zhao X, Schwartz S. A splicing enhancer in the E4 coding region of human papillomavirus type 16 is required for early mRNA splicing and polyadenylation as well as inhibition of premature late gene expression. J Virol 2005; 79:12002-15. [PMID: 16140776 PMCID: PMC1212645 DOI: 10.1128/jvi.79.18.12002-12015.2005] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Successful inhibition of human papillomavirus type 16 (HPV-16) late gene expression early in the life cycle is essential for persistence of infection, the highest risk factor for cervical cancer. Our study aimed to locate regulatory RNA elements in the early region of HPV-16 that influence late gene expression. For this purpose, subgenomic HPV-16 expression plasmids under control of the strong human cytomegalovirus immediate early promoter were used. An exonic splicing enhancer that firmly supported the use of the E4 3' splice site at position 3358 in the early region of the HPV-16 genome was identified. The enhancer was mapped to a 65-nucleotide AC-rich sequence located approximately 100 nucleotides downstream of the position 3358 3' splice site. Deletion of the enhancer caused loss of both splicing at the upstream position 3358 3' splice site and polyadenylation at the early polyadenylation signal, pAE. Direct splicing occurred at the competing L1 3' splice site at position 5639 in the late region. Optimization of the position 3358 3' splice site restored splicing to that site and polyadenylation at pAE. Additionally, a sequence of 40 nucleotides with a negative effect on late mRNA production was located immediately downstream of the enhancer. As the E4 3' splice site is employed by both early and late mRNAs, the enhancer constitutes a key regulator of temporal HPV-16 gene expression, which is required for early mRNA production as well as for the inhibition of premature late gene expression.
Collapse
Affiliation(s)
- Margaret Rush
- Department of Medical Biochemistry and Microbiology, Uppsala University, Biomedical Centre, Husargatan 3, Box 582, 75123 Uppsala, Sweden
| | | | | |
Collapse
|
22
|
Spink KM, Laimins LA. Induction of the human papillomavirus type 31 late promoter requires differentiation but not DNA amplification. J Virol 2005; 79:4918-26. [PMID: 15795277 PMCID: PMC1069532 DOI: 10.1128/jvi.79.8.4918-4926.2005] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The human papillomavirus (HPV) life cycle is linked to the differentiation state of the host cell. In virus-infected undifferentiated basal epithelial cells, HPV genomes are maintained as episomes at low copy number. Upon differentiation, a concomitant increase in viral copy number and an induction of late gene expression from a differentiation-specific promoter is seen. To investigate whether late gene expression was dependent on the amplification of the viral genome, inhibitors of DNA replication and in vitro systems for epithelial differentiation were used in conjunction with cells that stably maintain HPV31 episomes. Treatment of cells induced to differentiate in methylcellulose with the DNA synthesis inhibitor cytosine beta-arabinofuranoside (AraC) blocked viral DNA amplification but did not prevent induction of late transcription. This suggests that late gene expression does not strictly require amplification of the viral genome and that differentiation signals alone are sufficient to activate transcription from the late promoter. However, DNA amplification does appear to be necessary for maximal induction of the late promoter. In order to examine the cis-acting elements that contribute to the activation of the late promoter, a transient reporter assay was developed. In these assays, an induction of late gene expression was seen upon differentiation that was specific to the late promoter. Mapping studies localized important regulatory elements to the E6/E7 region and identified short sequences that could serve as binding sites for transcription factors. Elements within the upstream regulatory region were also found to positively and negatively influence transcription from the late promoter. These results identify mechanisms important for the differentiation-dependent activation of late gene expression of high-risk papillomaviruses.
Collapse
Affiliation(s)
- Kathryn M Spink
- Microbiology-Immunology Department, Northwestern University, 303 E. Chicago Ave, Chicago, IL 60611, USA
| | | |
Collapse
|
23
|
Bodily JM, Meyers C. Genetic analysis of the human papillomavirus type 31 differentiation-dependent late promoter. J Virol 2005; 79:3309-21. [PMID: 15731225 PMCID: PMC1075705 DOI: 10.1128/jvi.79.6.3309-3321.2005] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Human papillomaviruses infect stratifying squamous epithelia, causing benign and malignant lesions. Upon differentiation of the host keratinocyte, the virus undergoes a dramatic increase in both DNA replication and transcription from the late promoter, leading to expression of late genes and virion morphogenesis. In human papillomavirus type 31 (HPV31), the late promoter is designated p742 and includes multiple start sites embedded within the E7 gene. In this report, we mapped viral DNA elements that control transcriptional activity from p742. Enhancer elements in the viral upstream regulatory region positively regulate this promoter. The region containing the transcriptional start sites is dispensable for activity, and at least two separate elements in the E6/E7 region are capable of supporting transcription. Of these, we mapped one to a 150-bp region of the E7 open reading frame and designate it the core p742 promoter. Using GF109203X, an inhibitor of protein kinase C signaling, we show that p742 activation is independent of viral genome amplification. Finally, we mapped elements in the region of p742 that confer responsiveness to differentiation and show that the upstream regulatory region does not contribute to the differentiation response of p742. These studies are an important step toward understanding the functioning and regulation of this multiple-start promoter.
Collapse
Affiliation(s)
- Jason M Bodily
- Department of Microbiology and Immunology H107, P. O. Box 850, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | | |
Collapse
|
24
|
Knight GL, Grainger JR, Gallimore PH, Roberts S. Cooperation between different forms of the human papillomavirus type 1 E4 protein to block cell cycle progression and cellular DNA synthesis. J Virol 2004; 78:13920-33. [PMID: 15564500 PMCID: PMC533915 DOI: 10.1128/jvi.78.24.13920-13933.2004] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Posttranslational modification-oligomerization, phosphorylation, and proteolytic cleavage-of the human papillomavirus (HPV) E4 protein occurs as the infected keratinocytes migrate up through the suprabasal wart layers. It has been postulated that these events modify E4 function during the virus life cycle. In HPV type 1 (HPV1)-induced warts, N-terminal sequences are progressively cleaved from the full-length E4 protein (E1(wedge)E4) of 17 kDa to produce a series of polypeptides of 16, 11 and 10 kDa. Here, we have shown that in human keratinocytes, a truncated protein (E4-16K), equivalent to the 16-kDa species, mediated a G(2) arrest in the cell cycle that was dependent on a threonine amino acid in a proline-rich domain of the protein. Reconstitution of cyclin B1 expression in E4-16K cells reversed the G(2) arrest. Expression of E4-16K also induced chromosomal rereplication, and this was associated with aberrant nuclear morphology. Perturbation of the mitotic cell cycle was a biological activity specific to the truncated protein. However, coexpression of the full-length E1(wedge)E4 protein and the truncated E4-16K protein inhibited normal cellular proliferation and cellular DNA rereplication but did not prevent cells from arresting in G(2). Our findings provide the first evidence to support the hypothesis that proteolytic cleavage of the E1(wedge)E4 protein modifies its function. Also, different forms of the HPV1 E4 protein cooperate to negatively influence keratinocyte proliferation. We predict that these distinct biological activities of E4 act to support efficient amplification of the viral genome in suprabasal keratinocytes.
Collapse
Affiliation(s)
- Gillian L Knight
- Cancer Research UK Institute for Cancer Studies, University of Birmingham, Vincent Dr., Edgbaston, Birmingham B15 2TT, United Kingdom
| | | | | | | |
Collapse
|
25
|
Zhao X, Rush M, Schwartz S. Identification of an hnRNP A1-dependent splicing silencer in the human papillomavirus type 16 L1 coding region that prevents premature expression of the late L1 gene. J Virol 2004; 78:10888-905. [PMID: 15452209 PMCID: PMC521837 DOI: 10.1128/jvi.78.20.10888-10905.2004] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
We have previously identified cis-acting RNA sequences in the human papillomavirus type 16 (HPV-16) L1 coding region which inhibit expression of L1 from eukaryotic expression plasmids. Here we have determined the function of one of these RNA elements, and we provide evidence that this RNA element is a splicing silencer which suppresses the use of the 3' splice site located immediately upstream of the L1 AUG. We also show that this splice site is inefficiently utilized as a result of a suboptimal polypyrimidine tract. Introduction of point mutations in the L1 coding region that altered the RNA sequence without affecting the L1 protein sequence resulted in the inactivation of the splicing silencer and induced splicing to the L1 3' splice site. These mutations also prevented the interaction of the RNA silencer with a 35-kDa cellular protein identified here as hnRNP A1. The splicing silencer in L1 inhibits splicing in vitro, and splicing can be restored by the addition of RNAs containing an hnRNP A1 binding site to the reaction, demonstrating that hnRNP A1 inhibits splicing of the late HPV-16 mRNAs through the splicing silencer sequence. While we show that one role of the splicing silencer is to determine the ratio between partially spliced L2/L1 mRNAs and spliced L1 mRNAs, we also demonstrate that it inhibits splicing from the major 5' splice site in the early region to the L1 3' splice site, thereby playing an essential role in preventing late gene expression at an early stage of the viral life cycle. We speculate that the activity of the splicing silencer and possibly the concentration of hnRNP A1 in the HPV-16-infected cell determines the ability of the virus to establish a persistent infection which remains undetected by the host immune surveillance.
Collapse
Affiliation(s)
- Xiaomin Zhao
- Department of Medical Biochemistry and Microbiology, Biomedical Center, Uppsala University, Husargatan 3, Box 582, 751 23 Uppsala, Sweden
| | | | | |
Collapse
|
26
|
Elementos víricos y celulares que intervienen en el proceso de replicación del virus del papiloma humano. Clin Transl Oncol 2004. [DOI: 10.1007/bf02711833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
27
|
Oh ST, Longworth MS, Laimins LA. Roles of the E6 and E7 proteins in the life cycle of low-risk human papillomavirus type 11. J Virol 2004; 78:2620-6. [PMID: 14963169 PMCID: PMC369251 DOI: 10.1128/jvi.78.5.2620-2626.2004] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Many important functions have been attributed to the high-risk human papillomavirus (HPV) E6 and E7 proteins, including binding and degradation of p53 as well as interacting with Rb proteins. In contrast, the physiological roles of the low-risk E6 and E7 proteins remain unclear. Previous studies demonstrated that the high-risk E6 and E7 proteins also play roles in the productive life cycle by facilitating the maintenance of viral episomes (J. T. Thomas, W. G. Hubert, M. N. Ruesch, and L. A. Laimins, Proc. Natl. Acad. Sci. USA 96:8449-8454, 1999). In order to determine whether low-risk E6 or E7 is similarly necessary for the stable maintenance of episomes, HPV type 11 (HPV-11) genomes that contained translation termination mutations in E6 or E7 were constructed. Upon transfection into normal human keratinocytes, genomes in which E6 function was abolished were unable to be maintained episomally. Transfection of genomes containing substitution mutations in amino acids conserved in high- and low-risk HPV types suggested that multiple protein domains are involved in this process. Examination of cells transfected with HPV-11 genomes in which E7 function was inhibited were found to exhibit a more complex phenotype. At the second passage following transfection, mutant genomes were maintained as episomes but at significantly reduced levels than in cells transfected with the wild-type HPV-11 genome. Upon further passage in culture, however, the episomal forms of these E7 mutant genomes quickly disappeared. These findings identify important new functions for the low-risk E6 and E7 proteins in the episomal maintenance of low-risk HPV-11 genomes and suggest that they may act in a manner similar to that observed for the high-risk proteins.
Collapse
Affiliation(s)
- Stephen T Oh
- Department of Microbiology-Immunology, The Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, USA
| | | | | |
Collapse
|
28
|
Middleton K, Peh W, Southern S, Griffin H, Sotlar K, Nakahara T, El-Sherif A, Morris L, Seth R, Hibma M, Jenkins D, Lambert P, Coleman N, Doorbar J. Organization of human papillomavirus productive cycle during neoplastic progression provides a basis for selection of diagnostic markers. J Virol 2003; 77:10186-201. [PMID: 12970404 PMCID: PMC228472 DOI: 10.1128/jvi.77.19.10186-10201.2003] [Citation(s) in RCA: 165] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The productive cycle of human papillomaviruses (HPVs) can be divided into discrete phases. Cell proliferation and episomal maintenance in the lower epithelial layers are followed by genome amplification and the expression of capsid proteins. These events, which occur in all productive infections, can be distinguished by using antibodies to viral gene products or to surrogate markers of their expression. Here we have compared precancerous lesions caused by HPV type 16 (HPV16) with lesions caused by HPV types that are not generally associated with human cancer. These include HPV2 and HPV11, which are related to HPV16 (supergroup A), as well as HPV1 and HPV65, which are evolutionarily divergent (supergroups E and B). HPV16-induced low-grade squamous intraepithelial lesions (CIN1) are productive infections which resemble those caused by other HPV types. During progression to cancer, however, the activation of late events is delayed, and the thickness of the proliferative compartment is progressively increased. In many HPV16-induced high-grade squamous intraepithelial lesions (CIN3), late events are restricted to small areas close to the epithelial surface. Such heterogeneity in the organization of the productive cycle was seen only in lesions caused by HPV16 and was not apparent when lesions caused by other HPV types were compared. By contrast, the order in which events in the productive cycle were initiated was invariant and did not depend on the infecting HPV type or the severity of disease. The distribution of viral gene products in the infected cervix depends on the extent to which the virus can complete its productive cycle, which in turn reflects the severity of cervical neoplasia. It appears from our work that the presence of such proteins in cells at the epithelial surface allows the severity of the underlying disease to be predicted and that markers of viral gene expression may improve cervical screening.
Collapse
Affiliation(s)
- Kate Middleton
- National Institute for Medical Research, Mill Hill, London
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Bromberg-White JL, Sen E, Alam S, Bodily JM, Meyers C. Induction of the upstream regulatory region of human papillomavirus type 31 by dexamethasone is differentiation dependent. J Virol 2003; 77:10975-83. [PMID: 14512546 PMCID: PMC225009 DOI: 10.1128/jvi.77.20.10975-10983.2003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2002] [Accepted: 07/10/2003] [Indexed: 11/20/2022] Open
Abstract
Glucocorticoids have been shown to play a role in the transforming abilities of human papillomaviruses (HPVs), and glucocorticoid response elements (GREs) have been identified in the upstream regulatory regions (URRs) of various HPV types. These findings have made glucocorticoids potential therapeutic targets for HPV infection. We have previously shown that the URR of HPV type 31 (HPV31) is insensitive to induction by the synthetic glucocorticoid dexamethasone (dex) in monolayer culture, despite the identification of three potential GREs in the 5' region of the URR. Due to the fact that the HPV life cycle is intimately linked to the differentiation of the host tissue, we chose to determine whether the URR of HPV31 was inducible by dex under differentiating conditions. Upon suspension of cells in a semisolid medium of methylcellulose, we found that the URR of HPV31 was inducible by dex. The three GREs appear to play roles as independent repressors of this inducibility. By 5' deletion analysis, the element(s) responsible for this induction was localized to nucleotides (nt) 7238 to 7557. Furthermore, we found that the region between nt 7883 and 7900 appears to act as a repressor of dex inducibility. These findings indicate that epithelial differentiation has a profound effect on the action of dex on the URR of HPV31, suggesting that glucocorticoids play an important role in the differentiation-dependent life cycle of HPV.
Collapse
Affiliation(s)
- Jennifer L Bromberg-White
- Department of Microbiology and Immunology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033, USA.
| | | | | | | | | |
Collapse
|
30
|
Kim KY, Taylor MW. Identification of a novel promoter in the E2 open reading frame of the human papillomavirus type 18 genome. J Med Virol 2003; 69:122-31. [PMID: 12436488 DOI: 10.1002/jmv.10256] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Northern blot and RT-PCR analyses indicated that the human papillomavirus E4 open reading frame is expressed in HeLa cells. Because integration at the E1 or E2 open reading frame would place the viral upstream regulatory region downstream of the viral late genes, the expression of E4 in HeLa cells is most likely regulated by host cellular promoter(s) or unidentified viral promoter(s) in the E2 region. Primer extension analysis and transient transfection experiments with luciferase reporter constructs under the transcriptional control of various subgenomic fragments of HPV-18 were carried out to identify and characterize functional promoters within the E2 region. The in vivo activity of a novel promoter located within the 5'-end region of the E2 open reading frame of human papillomavirus type 18 is demonstrated.
Collapse
Affiliation(s)
- Kelly Y Kim
- Department of Biology, Indiana University, Bloomington, Indiana 47405, USA
| | | |
Collapse
|
31
|
Ozbun MA. Infectious human papillomavirus type 31b: purification and infection of an immortalized human keratinocyte cell line. J Gen Virol 2002; 83:2753-2763. [PMID: 12388811 DOI: 10.1099/0022-1317-83-11-2753] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Human papillomaviruses (HPVs) are aetiological agents of human malignancies, most notably cervical cancers. The life-cycles of HPVs are dependent on epithelial differentiation, and this has impeded many basic studies of HPV biology. The organotypic (raft) culture system supports epithelial differentiation such that infectious virions are synthesized in raft tissues from epithelial cells that replicate extrachromosomal HPV genomes. The CIN-612 9E cell line maintains episomal copies of HPV type 31b (HPV31b), an HPV type associated with cervical cancers. Many previous studies, including our own, have focused on characterizing the later stages of the HPV31b life-cycle in CIN-612 9E raft tissues. In this study, we have used the raft system to generate large numbers of HPV31b viral DNA (vDNA)-containing particles. We found a biologically contained homogenization system to be efficient at virion extraction from raft epithelial tissues. We also determined that vDNA-containing particles could be directly quantified from density-gradient fractions. Using an RT-PCR assay, the presence of newly synthesized, spliced HPV31b transcripts was detected following HPV31b infection of the immortalized HaCaT epithelial cell line. Spliced E6 and E1( wedge )E4 RNAs were detected using a single round of RT-PCR from cells infected with a dose as low as 1.0 vDNA-containing particle per cell. Spliced E1*I,E2 transcripts were found in cells infected with an HPV31b dose as low as 10 vDNA-containing particles per cell. Infectivity was blocked by HPV31 antiserum, but was not affected by DNase I. This work lays a foundation for a detailed analysis of the early events in HPV infection.
Collapse
Affiliation(s)
- Michelle A Ozbun
- Department of Molecular Genetics and Microbiology, The University of New Mexico School of Medicine, Albuquerque, New Mexico 87131, USA1
| |
Collapse
|
32
|
Ozbun MA. Human papillomavirus type 31b infection of human keratinocytes and the onset of early transcription. J Virol 2002; 76:11291-300. [PMID: 12388689 PMCID: PMC136784 DOI: 10.1128/jvi.76.22.11291-11300.2002] [Citation(s) in RCA: 77] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2002] [Accepted: 08/12/2002] [Indexed: 11/20/2022] Open
Abstract
Human papillomaviruses (HPVs) cause a number of human tumors and malignancies, including cervical cancers. Epithelial differentiation is required for the complete HPV life cycle and can be achieved using the organotypic (raft) culture system. The CIN-612 9E cell line maintains episomal copies of HPV type 31b (HPV31b), an HPV type associated with cervical cancers. When grown in the raft system, CIN-612 9E cells form a differentiated epithelium such that infectious virions can be synthesized. Many aspects of the later stages of the HPV31b life cycle have been investigated in CIN-612 9E raft tissues. We used a biologically contained homogenization system for efficient virion extraction from raft epithelial tissues. Purified HPV31b virions were used to infect low-passage-number human foreskin keratinocytes and a variety of epithelial cell lines. Newly synthesized, spliced HPV31b transcripts were detected by reverse transcription and PCR (RT-PCR) following HPV31b infection. HPV31b infection was most efficient and reproducible in HaCaT cells. The onset of viral transcription following infection was also investigated using RT-PCR techniques. Spliced E1(*)I,E2 RNAs were present as early as 4 h postinfection (p.i.), whereas the other major viral transcripts were detected by 8 to 10 h p.i. Furthermore, we characterized the structures and temporal expression of seven novel spliced early transcripts expressed following infection.
Collapse
Affiliation(s)
- Michelle A Ozbun
- Department of Molecular Genetics and Microbiology, The University of New Mexico School of Medicine, Albuquerque, New Mexico 87131, USA.
| |
Collapse
|
33
|
Steele BK, Meyers C, Ozbun MA. Variable expression of some "housekeeping" genes during human keratinocyte differentiation. Anal Biochem 2002; 307:341-7. [PMID: 12202253 DOI: 10.1016/s0003-2697(02)00045-3] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
We investigated the expression levels of four cellular "housekeeping" genes during epithelial differentiation. Differentiation is a dynamic process and various cellular RNAs have been targeted for use as internal controls during differentiation of human keratinocytes, but the consistent expression of such standards has not been previously validated. We used the organotypic (raft) culture system to grow stratified and differentiated epithelium in vitro. We compared cellular RNAs from epithelial tissues of both normal human keratinocytes and keratinocytes whose differentiation scheme is altered by the replication of human papillomavirus. Using ribonuclease protection assays to quantify RNA expression levels, we found that beta-actin and glyceraldehyde-3-phosphate dehydrogenase levels fluctuated during epithelial differentiation, whereas cyclophilin RNA and 28S-ribosomal RNA were the most consistently expressed during epithelial differentiation. These stably expressed cellular RNAs can be targeted as controls to permit quantitative expression analyses of cellular and pathogen RNAs during epithelial differentiation under various experimental conditions.
Collapse
Affiliation(s)
- Brandi K Steele
- Department of Molecular Genetics and Microbiology, The University of New Mexico School of Medicine, Albuquerque, NM 87131, USA
| | | | | |
Collapse
|
34
|
Sen E, Bromberg-White JL, Meyers C. Genetic analysis of cis regulatory elements within the 5' region of the human papillomavirus type 31 upstream regulatory region during different stages of the viral life cycle. J Virol 2002; 76:4798-809. [PMID: 11967297 PMCID: PMC136139 DOI: 10.1128/jvi.76.10.4798-4809.2002] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The function of the 5' region of the upstream regulatory region (URR) in regulating E6/E7 expression in cancer-associated papillomaviruses has been largely uncharacterized. In this study we used linker-scanning mutational analysis to identify potential cis regulatory elements contained within a portion of the 5' region of the URR that are involved in regulating transcription of the E6/E7 promoter at different stages of the viral life cycle. The mutational analysis illustrated differences in the transcriptional utilization of specific regions of the URR depending on the stage of the viral life cycle. This study identified (i) viral cis elements that regulate transcription in the presence and absence of any viral gene products or viral DNA replication, (ii) the role of host tissue differentiation in viral transcriptional regulation, and (iii) cis regulatory regions that are effected by induction of the protein kinase C pathway. Our studies have provided an extensive map of functional elements in the 5' region (nuncleotides 7259 to 7510) of the human papillomavirus type 31 URR that are involved in the regulation of p99 promoter activity at different stages of the viral life cycle.
Collapse
Affiliation(s)
- Ellora Sen
- Department of Microbiology and Immunology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033, USA
| | | | | |
Collapse
|
35
|
Hubert WG, Laimins LA. Human papillomavirus type 31 replication modes during the early phases of the viral life cycle depend on transcriptional and posttranscriptional regulation of E1 and E2 expression. J Virol 2002; 76:2263-73. [PMID: 11836404 PMCID: PMC153800 DOI: 10.1128/jvi.76.5.2263-2273.2002] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The E1 and E2 proteins are both required for papillomavirus DNA replication, and replication efficiency is controlled by the abundance of these factors. In human papillomaviruses (HPVs), the regulation of E1 and E2 expression and its effect on viral replication are not well understood. In particular, it is not known if E1 and E2 modulate their own expression and how posttranscriptional mechanisms may affect the levels of the replication proteins. Previous studies have implicated splicing within the E6 open reading frame (ORF) as being important for modulating replication of HPV type 31 (HPV31) through altered expression of E1 and E2. To analyze the function of the E6 intron in viral replication more specifically, we examined the effects of E6 splicing mutations in the context of entire viral genomes in transient assays. HPV31 genomes which had mutations in the splice donor site (E6SD) or the splice acceptor site (E6SA), a deletion of the intron (E6ID), or substituted heterologous intron sequences (E6IS) were constructed. Compared to wild-type (wt) HPV31, pHPV31-E6SD, -E6SA, and -E6IS replicated inefficiently while pHPV31-E6ID replicated at an intermediate level. Cotransfection of the E6 mutant genomes with an E1 expression vector strongly activated their replication levels, indicating that efficient expression of E1 requires E6 internal splicing. In contrast, replication was activated only moderately with an E2 expression vector. Replacing the wt E6 intron in HPV31 with a heterologous intron from simian virus 40 (E6SR2) resulted in replication levels similar to that of the wt in the absence of expression vectors, suggesting that mRNA splicing upstream of the E1 ORF is important for high-level replication. To examine the effects of E6 intron splicing on E1 and E2 expression directly, we constructed reporter DNAs in which the luciferase coding sequences were fused in frame to the E1 (E1Luc) or E2 (E2Luc) gene. Reporter activities were then analyzed in transient assays with cotransfected E1 or E2 expression vectors. Both reporters were moderately activated by E1 in a dose-dependent manner. In addition, E1Luc was activated by low doses of E2 but was repressed at high doses. In contrast, E2 had little effect on E2Luc activity. These data indicate that E1 expression and that of E2 are interdependent and regulated differentially. When the E6 splicing mutations were analyzed in both reporter backgrounds, only E1Luc activities correlated with splicing competence in the E6 ORF. These findings support the hypothesis that the E6 intron primarily regulates expression of E1. Finally, in long-term replication assays, none of the E6 mutant genomes could be stably maintained. However, cotransfection of the E6 splicing mutant genomes with pHPV31-E7NS, which contains a nonsense mutation in the E7 coding sequence, restored stable replication of some mutants. Our observations indicate that E1 expression and that of E2 are differentially regulated at multiple levels and that efficient expression of E1 is required for transient and stable viral replication. These regulatory mechanisms likely act to control HPV copy number during the various phases of the viral life cycle.
Collapse
Affiliation(s)
- Walter G Hubert
- Department of Dermatology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | | |
Collapse
|
36
|
del Mar Peña LM, Laimins LA. Differentiation-dependent chromatin rearrangement coincides with activation of human papillomavirus type 31 late gene expression. J Virol 2001; 75:10005-13. [PMID: 11559836 PMCID: PMC114575 DOI: 10.1128/jvi.75.20.10005-10013.2001] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The life cycle of human papillomaviruses (HPVs) is tightly linked to the differentiation status of the host cell. While early genes are expressed during the initial stages of viral infection, late gene expression occurs in the suprabasal layers of the cervical epithelium. Late genes encode E1-E4, a cytosolic protein, and capsid proteins L1 and L2. We have mapped over 30 initiation sites for late transcripts and show that the transcripts initiate in a 200-nucleotide region within the E7 open reading frame. The mechanisms regulating the activation of late gene expression, however, are not yet understood. DNase I hypersensitivity analysis of HPV-31 chromatin in cell lines that maintain viral genomes extrachromosomally indicates that a major shift in nuclease digestion occurs upon differentiation. In undifferentiated cells, hypersensitive regions exist in the upstream regulatory region proximal to the E6 open reading frame. Upon differentiation, a region between nucleotides 659 and 811 in the E7 open reading frame becomes accessible to DNase I. These results indicate that the late transcript initiation region becomes accessible to transcription factor binding upon differentiation. Several complexes mediate chromatin rearrangement, and we tested whether histone acetylation was sufficient for late transcript activation. Treatment with the histone deacetylase inhibitor trichostatin A was found to be insufficient to activate late gene expression in undifferentiated cells. However, it did activate expression of early transcripts. These results suggest that chromatin remodeling around the late promoter occurs upon epithelial differentiation and that mechanisms in addition to histone deacetylation contribute to activation of late gene expression.
Collapse
Affiliation(s)
- L M del Mar Peña
- Department of Microbiology-Immunology, Northwestern University Medical School, Chicago, Illinois 60611, USA
| | | |
Collapse
|
37
|
Terhune SS, Hubert WG, Thomas JT, Laimins LA. Early polyadenylation signals of human papillomavirus type 31 negatively regulate capsid gene expression. J Virol 2001; 75:8147-57. [PMID: 11483760 PMCID: PMC115059 DOI: 10.1128/jvi.75.17.8147-8157.2001] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The L1 and L2 capsid genes of human papillomavirus type 31 (HPV-31) are expressed upon keratinocyte differentiation from a promoter located in the E7 open reading frame (ORF) of the early region. Late transcripts must therefore pass through and ignore the early polyadenylation sequences to use the downstream late AAUAAA element located at the end of the L1 ORF. To identify sequences which modulate downstream capsid gene expression, a variety of substitution mutations were introduced into the early polyadenylation signal and studied first in the context of polycistronic luciferase reporter constructs. Removal of the G/U-rich cleavage stimulation factor (CstF) binding sites and the degenerate cleavage and polyadenylation specificity factor binding sites, UAUAUA, had minimal effect on downstream expression as defined by luciferase activities. This is in contrast to the deletion of the HPV-31 early AAUAAA element, which resulted in a dramatic increase in downstream expression. Additional sequences within the first 800 bp of the L2 ORF were also found to negatively regulate capsid expression in luciferase assays. To determine how these mutations influence gene expression in the context of the complete HPV-31 genome, recombinant genomes were constructed that contained a substitution in the AAUAAA sequence, an inserted strong CstF binding site, an inserted simian virus 40 (SV40) late poly(A) signal, or a substitution of the 5'-most 800 nucleotides of the L2 ORF. Reductions in both transient and stable replication were observed with the recombinant genomes containing the strong CstF site or the late SV40 signal, suggesting that alterations in the strength of the upstream poly(A) signal influence expression of viral replication factors. Similarly, disruption of the L2 ORF resulted in a significant reduction in genome replication and an inability to be maintained stably. In contrast, genomes containing a substitution of the AAUAAA sequence had increased levels of transient and stable replication. Quantitation of late transcripts following keratinocyte differentiation in methylcellulose also showed a reduction in downstream capsid gene expression in lines containing genomes with the strong CstF site or the late SV40 signal mutations, while a significant increase in expression was detected in the lines with genomes lacking the AAUAAA sequence. These studies demonstrate that capsid gene expression in HPV-31 requires an inefficient early poly(A) signal which is defined primarily by the AAUAAA element as well as a major negative regulatory element located within the L2 ORF.
Collapse
Affiliation(s)
- S S Terhune
- Department of Microbiology-Immunology, Northwestern University Medical School, Chicago, Illinois 606113, USA
| | | | | | | |
Collapse
|
38
|
Stacey SN, Jordan D, Williamson AJ, Brown M, Coote JH, Arrand JR. Leaky scanning is the predominant mechanism for translation of human papillomavirus type 16 E7 oncoprotein from E6/E7 bicistronic mRNA. J Virol 2000; 74:7284-97. [PMID: 10906182 PMCID: PMC112249 DOI: 10.1128/jvi.74.16.7284-7297.2000] [Citation(s) in RCA: 67] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2000] [Accepted: 05/19/2000] [Indexed: 11/20/2022] Open
Abstract
Human papillomaviruses (HPV) are unique in that they generate mRNAs that apparently can express multiple proteins from tandemly arranged open reading frames. The mechanisms by which this is achieved are uncertain and are at odds with the basic predictions of the scanning model for translation initiation. We investigated the unorthodox mechanism by which the E6 and E7 oncoproteins from human papillomavirus type 16 (HPV-16) can be translated from a single, bicistronic mRNA. The short E6 5' untranslated region (UTR) was shown to promote translation as efficiently as a UTR from Xenopus beta-globin. Insertion of a secondary structural element into the UTR inhibited both E6 and E7 expression, suggesting that E7 expression depends on ribosomal scanning from the 5' end of the mRNA. E7 translation was found to be cap dependent, but E6 was more dependent on capping and eIF4F activity than E7. Insertion of secondary structural elements at various points in the region upstream of E7 profoundly inhibited translation, indicating that scanning was probably continuous. Insertion of the E6 region between Renilla and firefly luciferase genes revealed little or no internal ribosomal entry site activity. However when E6 was located at the 5' end of the mRNA, it permitted over 100-fold-higher levels of downstream cistron translation than did the Renilla open reading frame. Internal AUGs in the E6 region with strong or intermediate Kozak sequence contexts were unable to inhibit E7 translation, but initiation at the E7 AUG was efficient and accurate. These data support a model in which E7 translation is facilitated by an extreme degree of leaky scanning, requiring the negotiation of 13 upstream AUGs. Ribosomal initiation complexes which fail to initiate at the E6 start codon can scan through to the E7 AUG without initiating translation, but competence to initiate is achieved once the E7 AUG is reached. These findings suggest that the E6 region of HPV-16 comprises features that sponsor both translation of the E6 protein and enhancement of translation at a downstream site.
Collapse
MESH Headings
- 5' Untranslated Regions
- Base Sequence
- Codon, Initiator
- Hot Temperature
- Humans
- Models, Molecular
- Molecular Sequence Data
- Oncogene Proteins, Viral/biosynthesis
- Oncogene Proteins, Viral/genetics
- Oncogene Proteins, Viral/metabolism
- Open Reading Frames/genetics
- Papillomaviridae/genetics
- Papillomaviridae/metabolism
- Papillomavirus E7 Proteins
- Potassium Chloride/metabolism
- Protein Biosynthesis
- Protein Structure, Secondary
- RNA Caps/genetics
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- RNA, Viral/genetics
- RNA, Viral/metabolism
- Repressor Proteins
- Ribosomes/genetics
- Ribosomes/metabolism
Collapse
Affiliation(s)
- S N Stacey
- Cancer Research Campaign, Department of Molecular Biology, Paterson Institute for Cancer Research, Christie Hospital, Manchester M20 4BX, United Kingdom.
| | | | | | | | | | | |
Collapse
|
39
|
Ai W, Narahari J, Roman A. Yin yang 1 negatively regulates the differentiation-specific E1 promoter of human papillomavirus type 6. J Virol 2000; 74:5198-205. [PMID: 10799595 PMCID: PMC110873 DOI: 10.1128/jvi.74.11.5198-5205.2000] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Human papillomavirus type 6 (HPV-6) is a low-risk HPV whose replication cycle, like that of all HPVs, is differentiation dependent. We have previously shown that CCAAT displacement protein (CDP) binds the differentiation-induced HPV-6 E1 promoter and negatively regulates its activity in undifferentiated cells (W. Ai, E. Toussaint, and A. Roman, J. Virol. 73:4220-4229, 1999). Using electrophoretic mobility shift assays (EMSAs), we now report that Yin Yang 1 (YY1), a multifunctional protein that can act as a transcriptional activator or repressor and that can also inhibit HPV replication in vitro, binds the HPV-6 E1 promoter. EMSAs, using subfragments of the promoter as competitors, showed that the YY1 binding site is located at the 5' end of the E1 promoter. When a putative YY1 site was mutated, the ability of YY1 to bind was greatly decreased. The activity of the mutated E1 promoter, monitored with the reporter gene luciferase, was threefold greater than that of the wild-type promoter, suggesting that YY1 negatively regulates HPV-6 E1 promoter activity. Nuclear extracts from differentiated keratinocytes showed decreased binding of YY1 to the wild-type promoter. Consistent with this, in differentiated keratinocytes, the activity of the transfected luciferase gene transcribed from the mutated promoter was comparable to that of the wild-type promoter; both promoters were up-regulated in differentiated keratinocytes compared to undifferentiated cells. These data suggest that YY1 functions in undifferentiated keratinocytes but not in differentiated keratinocytes. Both the wild-type and mutated promoters could be negatively regulated by overexpression of a plasmid encoding CDP. Thus, both YY1 and CDP appear to be negative regulators of the differentiation-induced HPV-6 E1 promoter and thereby the HPV life cycle. In contrast, only binding of CDP was detected using the E1 promoter of the high-risk HPV-31.
Collapse
Affiliation(s)
- W Ai
- Department of Microbiology and Immunology, Indiana University School of Medicine, and Walther Cancer Institute, Indianapolis, Indiana 46202-5120, USA
| | | | | |
Collapse
|
40
|
Koffa MD, Graham SV, Takagaki Y, Manley JL, Clements JB. The human papillomavirus type 16 negative regulatory RNA element interacts with three proteins that act at different posttranscriptional levels. Proc Natl Acad Sci U S A 2000; 97:4677-82. [PMID: 10781073 PMCID: PMC18292 DOI: 10.1073/pnas.070049097] [Citation(s) in RCA: 43] [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
In human papillomaviruses, expression of the late genes L1 and L2, encoding the capsid proteins, is restricted to the upper layers of the infected epithelium. A 79-nt GU-rich negative regulatory element (NRE) located at the 3' untranslated region of the human papillomavirus 16 L1 gene was identified previously as key to the posttranscriptional control of late gene expression. Here, we demonstrate that in epithelial cells, the NRE can directly bind the U2 auxiliary splicing factor 65-kDa subunit, the cleavage stimulation factor 64-kDa subunit, and the Elav-like HuR protein. On induction of epithelial cell differentiation, levels of the U2 auxiliary splicing factor 65-kDa subunit decrease, levels of the cleavage stimulation factor 64-kDa subunit increase, and the levels of HuR remain unchanged, although redistribution of the HuR from the nucleus to the cytoplasm is observed. Late gene transcripts, which appear to be fully processed, are detected in undifferentiated W12 cells, but are confined in the nucleus. We propose that repression of late gene expression in basal epithelial cells may be caused by nuclear retention or cytoplasmic instability of NRE-containing late gene transcripts.
Collapse
Affiliation(s)
- M D Koffa
- Division of Virology, Institute of Biomedical and Life Sciences, University of Glasgow, Church Street, Glasgow, G11 5JR, Scotland, United Kingdom
| | | | | | | | | |
Collapse
|
41
|
Abstract
Productive infection by human papillomaviruses (HPV) is dependent upon the differentiation of the host cell. Following entry into basal epithelial cells, HPV genomes are established as autonomous replicating extrachromosomal elements and a low level of HPV expression occurs. Upon differentiation of infected cells, productive replication and expression of capsid genes is induced resulting in the synthesis of progeny virions. Evidence from immunosuppressed patients as well as individuals with recurring laryngeal papillomatosis suggest that certain HPV types can exist in a latent state. In latently infected cells, HPV DNA may be present but no differentiation-dependent synthesis of virions occurs. The presence of a latent state for HPVs can be a determining factor in the effectiveness of therapeutic methods for treatment of infections.
Collapse
Affiliation(s)
- F Stubenrauch
- Universitatsklinikum Tu@a2bingen, Abt. Medizinische Virologie, Sektion Experimentelle Virologie, Calwerstr. 7/6, D72076 Tuebingen, Germany
| | | |
Collapse
|
42
|
Braunstein TH, Madsen BS, Gavnholt B, Rosenstierne MW, Koefoed Johnsen C, Norrild B. Identification of a new promoter in the early region of the human papillomavirus type 16 genome. J Gen Virol 1999; 80 ( Pt 12):3241-3250. [PMID: 10567657 DOI: 10.1099/0022-1317-80-12-3241] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Transcription of the human papillomavirus type 16 (HPV-16) genome is controlled by several promoters; the P(97) promoter is considered to be the main one. An additional promoter has been identified within the E7 ORF as well as an antisense promoter just upstream of the L2 ORF. The significance of these promoters for early and late gene expression and their activity related to cell differentiation is not known in detail. Identification of two new, previously undescribed transcription start sites at nt 542 just upstream of the E7 ORF and at nt 611 within the E7 ORF is reported. The promoter responsible for the start site at nt 542 (P(542)) was active in SiHa, HeLa and C33A cells. Very low promoter activity was found upstream of the nt 611 start site. The E7 protein has previously been shown to be synthesized from a polycistronic mRNA encoding both the E6 and E7 proteins under the control of the P(97) promoter. The data reported in the present paper suggest that promoter P(542) may control synthesis of the E7 oncoprotein from a monocistronic mRNA.
Collapse
Affiliation(s)
- T H Braunstein
- Institute of Molecular Pathology, The Protein Laboratory, University of Copenhagen, Panum Institute, Bldg 6.2, Blegdamsvej 3C, DK-2200 Copenhagen N, Denmark1
| | - B S Madsen
- Institute of Molecular Pathology, The Protein Laboratory, University of Copenhagen, Panum Institute, Bldg 6.2, Blegdamsvej 3C, DK-2200 Copenhagen N, Denmark1
| | - B Gavnholt
- Institute of Molecular Pathology, The Protein Laboratory, University of Copenhagen, Panum Institute, Bldg 6.2, Blegdamsvej 3C, DK-2200 Copenhagen N, Denmark1
| | - M W Rosenstierne
- Institute of Molecular Pathology, The Protein Laboratory, University of Copenhagen, Panum Institute, Bldg 6.2, Blegdamsvej 3C, DK-2200 Copenhagen N, Denmark1
| | - C Koefoed Johnsen
- Institute of Molecular Pathology, The Protein Laboratory, University of Copenhagen, Panum Institute, Bldg 6.2, Blegdamsvej 3C, DK-2200 Copenhagen N, Denmark1
| | - B Norrild
- Institute of Molecular Pathology, The Protein Laboratory, University of Copenhagen, Panum Institute, Bldg 6.2, Blegdamsvej 3C, DK-2200 Copenhagen N, Denmark1
| |
Collapse
|
43
|
Dolei A, Curreli S, Marongiu P, Pierangeli A, Gomes E, Bucci M, Serra C, Degener AM. Human immunodeficiency virus infection in vitro activates naturally integrated human papillomavirus type 18 and induces synthesis of the L1 capsid protein. J Gen Virol 1999; 80 ( Pt 11):2937-2944. [PMID: 10580055 DOI: 10.1099/0022-1317-80-11-2937] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Human papillomavirus (HPV) infections are prevalent in human immunodeficiency virus (HIV)-positive individuals. To highlight the effect of HIV on HPV expression, HPV-18-positive HIV-permissive HeLa-T4 cells were either infected with HIV-1 or treated with Tat or with the cytokines IL-1alpha, IL-1beta, IL-6 and TNF-alpha. The presence of HPV-18 E1 (early) and L1 (late) transcripts was then determined by dot-blot or Northern blot hybridization with E1 and L1 or with genomic HPV-18 DNA probes, respectively. Protein extracts from parallel cultures were challenged by Western blotting with an antiserum raised against an L1-beta-galactosidase hybrid protein. Results indicated that HeLa-T4 cells constitutively express E1 and L1 transcripts. When cells were infected with HIV, the amounts of E1 and L1 RNAs increased with time, followed by the de novo appearance of L1 protein. E1 and L1 transcripts were also increased, in a dose-dependent manner, by treatment of uninfected cultures with Tat or with IL-6, but were not affected by IL-1alpha, IL-1beta and TNF- alpha. Neither Tat nor IL-6 could induce L1 translation. These findings raise the hypothesis that the increase of HPV shedding and of HPV-associated diseases in HIV-infected individuals could be due in part to a direct or cytokine-mediated action of HIV, in addition to the HIV-induced immunodeficiency.
Collapse
Affiliation(s)
- Antonina Dolei
- Department of Biomedical Sciences, Microbiology Section, University of Sassari, Viale S. Pietro 43B, I-07100 Sassari, Italy 1
| | - Sabrina Curreli
- Department of Biomedical Sciences, Microbiology Section, University of Sassari, Viale S. Pietro 43B, I-07100 Sassari, Italy 1
| | - Patrizia Marongiu
- Department of Biomedical Sciences, Microbiology Section, University of Sassari, Viale S. Pietro 43B, I-07100 Sassari, Italy 1
| | - Alessandra Pierangeli
- Department of Cellular and Developmental Biology, La Sapienza University, Rome, Italy2
| | - Eduarda Gomes
- Department of Biomedical Sciences, Microbiology Section, University of Sassari, Viale S. Pietro 43B, I-07100 Sassari, Italy 1
| | - Mauro Bucci
- Department of Cellular and Developmental Biology, La Sapienza University, Rome, Italy2
| | - Caterina Serra
- Department of Biomedical Sciences, Microbiology Section, University of Sassari, Viale S. Pietro 43B, I-07100 Sassari, Italy 1
| | - Anna Marta Degener
- Department of Cellular and Developmental Biology, La Sapienza University, Rome, Italy2
| |
Collapse
|
44
|
Terhune SS, Milcarek C, Laimins LA. Regulation of human papillomavirus type 31 polyadenylation during the differentiation-dependent life cycle. J Virol 1999; 73:7185-92. [PMID: 10438805 PMCID: PMC104242 DOI: 10.1128/jvi.73.9.7185-7192.1999] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The L1 and L2 capsid genes of human papillomavirus type 31 (HPV-31) are expressed late in the differentiation-dependent life cycle from a promoter located in the E7 open reading frame (ORF) of the early region. These late HPV genes are transcribed by RNA polymerase II which reads through the region containing early polyadenylation signals and proceeds to a poly(A) site downstream of L1. In this study, we have investigated the mechanisms regulating differentiation-dependent polyadenylation and read-through in HPV-31. HPV-31 early transcripts were found to utilize a heterogeneous series of polyadenylation sites in undifferentiated cells. The sites for polyadenylation extended over a range of 100 nucleotides from within the E5 ORF to upstream of L2. Upon differentiation, the transcription of early genes increased, but no change in the heterogeneous distribution of 3' ends was detected. The early polyadenylation region was found to contain a single consensus hexanucleotide sequence, AAUAAA, as well as three weak binding sites for the cleavage stimulatory factor, CstF. In contrast to the heterogeneity at the early site, the 3' ends of late transcripts encoding L1 and L2 were localized to a narrow region downstream of the late AAUAAA element. The late polyadenylation signal was found to contain a single high-affinity site for CstF, as well as one consensus hexanucleotide sequence. By using a reporter assay, it was determined that the HPV-31 early polyadenylation sequences allowed significant levels of read-through into the late region in undifferentiated cells. Upon differentiation, this read-through was increased by approximately 50%, indicating that use of the early site decreased. Differentiation was also found to induce a 40% reduction in the levels of CstF subunits, which may contribute to the increased read-through of the early sequence. The insertion of the late high-affinity binding site for CstF into the early polyadenylation region significantly reduced the level of read-through, suggesting that these factors modulate read-through activity. Our studies demonstrate that HPV-31 late gene expression is regulated in a large part by posttranscriptional mechanisms, including the polyadenylation of early transcripts.
Collapse
Affiliation(s)
- S S Terhune
- Department of Microbiology-Immunology, Northwestern University Medical School, Chicago, Illinois 60611, USA
| | | | | |
Collapse
|
45
|
Klumpp DJ, Laimins LA. Differentiation-induced changes in promoter usage for transcripts encoding the human papillomavirus type 31 replication protein E1. Virology 1999; 257:239-46. [PMID: 10208937 DOI: 10.1006/viro.1999.9636] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The life cycle of human papillomaviruses (HPVs) is tied to keratinocyte differentiation. One key event in the viral life cycle is the differentiation-dependent increase in viral replication. This increase in replication activity results in an amplification of the HPV genome from approximately 50 copies per cell in basal keratinocytes to thousands of copies of the viral genome per cell in suprabasal keratinocytes. To characterize the events associated with this differentiation-dependent increase in HPV replication, we have initiated studies of mRNAs encoding the HPV replication protein E1 during the differentiation of cell lines that stably maintain episomal HPV DNA. Differentiation induced the expression of several transcripts that hybridized to an E1-specific probe. One of these messages, a 3.7-kb transcript, did not hybridize to a probe specific for the early promoter upstream of the E6 open reading frame. RNase protection analysis confirmed an induction of unspliced messages derived from the differentiation-dependent promoter at nucleotide 742 in the middle of the E7 open reading frame. These observations demonstrate a differentiation-induced increase in E1 mRNAs derived from the viral late promoter and suggest a role for increased E1 expression during amplification of the HPV genome.
Collapse
Affiliation(s)
- D J Klumpp
- Department of Microbiology-Immunology, Northwestern University Medical School, 303 E. Chicago Avenue, Chicago, Illinois, 60611, USA
| | | |
Collapse
|
46
|
Remm M, Remm A, Ustav M. Human papillomavirus type 18 E1 protein is translated from polycistronic mRNA by a discontinuous scanning mechanism. J Virol 1999; 73:3062-70. [PMID: 10074156 PMCID: PMC104066 DOI: 10.1128/jvi.73.4.3062-3070.1999] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Papillomaviruses are small double-stranded DNA viruses that replicate episomally in the nuclei of infected cells. The full-length E1 protein of papillomaviruses is required for the replication of viral DNA. The viral mRNA from which the human papillomavirus type 18 E1 protein is expressed is not known. We demonstrate that in eukaryotic cells, the E1 protein is expressed from polycistronic mRNA containing E6, E7, and E1 open reading frames (ORFs). The translation of adjacent E7 and E1 ORFs is not associated; it is performed by separate populations of ribosomes. The translation of the downstream E1 gene is preceded by ribosome scanning. Scanning happens at least at the 5' end of the polycistronic mRNA and also approximately 100 bp in front of the E1 gene. Long areas in middle of the mRNA are bypassed by ribosomes, possibly by ribosomal "shunting." Inactivation of short minicistrons in the upstream area of the E1 gene did not change the expression level of the E1 gene.
Collapse
Affiliation(s)
- M Remm
- Department of Microbiology and Virology, University of Tartu, and Estonian Biocentre, Tartu 51010, Estonia.
| | | | | |
Collapse
|
47
|
Ozbun MA, Meyers C. Two novel promoters in the upstream regulatory region of human papillomavirus type 31b are negatively regulated by epithelial differentiation. J Virol 1999; 73:3505-10. [PMID: 10074210 PMCID: PMC104120 DOI: 10.1128/jvi.73.4.3505-3510.1999] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Organotypic cultures support the stratification and differentiation of keratinocytes and the human papillomavirus (HPV) life cycle. We report transcription from four novel promoters in the HPV31b upstream regulatory region during the viral life cycle in organotypic cultures. Promoter initiation was not differentiation dependent; two promoters were down-regulated upon epithelial differentiation.
Collapse
Affiliation(s)
- M A Ozbun
- Department of Molecular Genetics and Microbiology, The University of New Mexico School of Medicine, Albuquerque, New Mexico 87131, USA
| | | |
Collapse
|
48
|
Bodily JM, Hoopes DJ, Roeder BL, Gilbert SG, Pettit GR, Herald CL, Rollins DN, Robison RA. The inhibitory effects of bryostatin 1 administration on the growth of rabbit papillomas. Cancer Lett 1999; 136:67-74. [PMID: 10211941 DOI: 10.1016/s0304-3835(98)00310-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Bryostatin 1 is a protein kinase C modulator that shows antineoplastic activity in a variety of tumor systems. This study examined the effects of bryostatin 1 administration on papilloma growth in rabbits. Investigations of optimal route, dose, and schedule were performed. Several groups of rabbits were inoculated with cottontail rabbit papillomavirus (CRPV) DNA. Bryostatin 1 was administered i.v., both daily and weekly, and intralesionally both weekly and bi-weekly. Intralesionally dosed papillomas were examined histologically for immune cell infiltration. In weekly and daily i.v. trials, 2.5 and 1.0 microg/kg, respectively, showed the greatest overall reduction in tumor size. Bryostatin 1 administered intralesionally also slowed papilloma growth. Treated lesions had significantly higher numbers of heterophils and eosinophils.
Collapse
Affiliation(s)
- J M Bodily
- Department of Microbiology, Brigham Young University, Provo, UT 84602-5133, USA
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Abstract
Genome and pre-genome replication in all animal DNA viruses except poxviruses occurs in the cell nucleus (Table 1). In order to reproduce, an infecting virion enters the cell and traverses through the cytoplasm toward the nucleus. Using the cell's own nuclear import machinery, the viral genome then enters the nucleus through the nuclear pore complex. Targeting of the infecting virion or viral genome to the multiplication site is therefore an essential process in productive viral infection as well as in latent infection and transformation. Yet little is known about how infecting genomes of animal DNA viruses reach the nucleus in order to reproduce. Moreover, this nuclear locus for viral multiplication is remarkable in that the sizes and composition of the infectious particles vary enormously. In this article, we discuss virion structure, life cycle to reproduce infectious particles, viral protein's nuclear import signal, and viral genome nuclear targeting.
Collapse
Affiliation(s)
- H Kasamatsu
- Molecular, Cell and Developmental Biology and Molecular Biology Institute, University of California at Los Angeles 90095, USA
| | | |
Collapse
|
50
|
Phelps WC, Barnes JA, Lobe DC. Molecular targets for human papillomaviruses: prospects for antiviral therapy. Antivir Chem Chemother 1998; 9:359-77. [PMID: 9875390 DOI: 10.1177/095632029800900501] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
A substantial medical need exists for the development of antiviral medicines for the treatment of diseases associated with infection by human papillomaviruses (HPVs). HPVs are associated with various benign and malignant lesions including benign genital condyloma, common skin warts, laryngeal papillomas and anogenital cancer. Since treatment options are limited and typically not very satisfactory, the development of safe and effective antiviral drugs for HPV could have substantial clinical impact. In the last few years, exciting advances have been made in our understanding of papillomavirus replication and the effects that the virus has on growth of the host cell. Although still somewhat rudimentary, techniques have been developed for limited virion production in vitro offering the promise of more rapid advances in the dissection and understanding of the virus life cycle. Of the 8-10 HPV gene products that are made during infection, only one encodes enzymatic activities, the E1 helicase. Successful antiviral therapies have traditionally targeted viral enzymes such as polymerases, kinases and proteases. In contrast, macromolecular interactions which mediate the functions of E6, E7 and E2 are thought to be more difficult targets for small molecule therapy.
Collapse
Affiliation(s)
- W C Phelps
- Department of Virology, Glaxo Wellcome Inc, Research Triangle Park, North Carolina 27709-3398, USA.
| | | | | |
Collapse
|