Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
Virology ; 590: 109952, 2024 02.
Article in English | MEDLINE | ID: mdl-38103269

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.


Subject(s)
Human papillomavirus 16 , Oncogene Proteins, Viral , Papillomavirus Infections , Humans , ErbB Receptors/genetics , ErbB Receptors/metabolism , Human papillomavirus 16/physiology , Keratinocytes , Oncogene Proteins, Viral/genetics , Oncogene Proteins, Viral/metabolism , Cell Differentiation
2.
J Virol ; 97(2): e0103222, 2023 02 28.
Article in English | MEDLINE | ID: mdl-36719239

ABSTRACT

Coinfection of human papillomavirus (HPV) and Epstein-Barr virus (EBV) has been detected in oropharyngeal squamous cell carcinoma. Although HPV and EBV replicate in differentiated epithelial cells, we previously reported that HPV epithelial immortalization reduces EBV replication within organotypic raft culture and that the HPV16 oncoprotein E7 was sufficient to inhibit EBV replication. A well-established function of HPV E7 is the degradation of the retinoblastoma (Rb) family of pocket proteins (pRb, p107, and p130). Here, we show that pRb knockdown in differentiated epithelia and EBV-positive Burkitt lymphoma (BL) reduces EBV lytic replication following de novo infection and reactivation, respectively. In differentiated epithelia, EBV immediate early (IE) transactivators were expressed, but loss of pRb blocked expression of the early gene product, EA-D. Although no alterations were observed in markers of epithelial differentiation, DNA damage, and p16, increased markers of S-phase progression and altered p107 and p130 levels were observed in suprabasal keratinocytes after pRb knockdown. In contrast, pRb interference in Akata BX1 Burkitt lymphoma cells showed a distinct phenotype from differentiated epithelia with no significant effect on EBV IE or EA-D expression. Instead, pRb knockdown reduced the levels of the plasmablast differentiation marker PRDM1/Blimp1 and increased the abundance of c-Myc protein in reactivated Akata BL with pRb knockdown. c-Myc RNA levels also increased following the loss of pRb in epithelial rafts. These results suggest that pRb is required to suppress c-Myc for efficient EBV replication in BL cells and identifies a mechanism for how HPV immortalization, through degradation of the retinoblastoma pocket proteins, interferes with EBV replication in coinfected epithelia. IMPORTANCE Terminally differentiated epithelium is known to support EBV genome amplification and virion morphogenesis following infection. The contribution of the cell cycle in differentiated tissues to efficient EBV replication is not understood. Using organotypic epithelial raft cultures and genetic interference, we can identify factors required for EBV replication in quiescent cells. Here, we phenocopied HPV16 E7 inhibition of EBV replication through knockdown of pRb. Loss of pRb was found to reduce EBV early gene expression and viral replication. Interruption of the viral life cycle was accompanied by increased S-phase gene expression in postmitotic keratinocytes, a process also observed in E7-positive epithelia, and deregulation of other pocket proteins. Together, these findings provide evidence of a global requirement for pRb in EBV lytic replication and provide a mechanistic framework for how HPV E7 may facilitate a latent EBV infection through its mediated degradation of pRb in copositive epithelia.


Subject(s)
Burkitt Lymphoma , Epstein-Barr Virus Infections , Retinoblastoma Protein , Virus Replication , Humans , Burkitt Lymphoma/virology , Cell Differentiation , Epithelium/virology , Epstein-Barr Virus Infections/metabolism , Herpesvirus 4, Human/genetics , Herpesvirus 4, Human/physiology , Papillomavirus Infections , Retinoblastoma Protein/metabolism
4.
Viruses ; 12(3)2020 03 13.
Article in English | MEDLINE | ID: mdl-32183180

ABSTRACT

Persistent infection by human papillomaviruses (HPVs), small, double-stranded DNA viruses that infect keratinocytes of the squamous epithelia, can lead to the development of cervical and other cancers. The viral oncoprotein E7 contributes to viral persistence in part by regulating host gene expression through binding host transcriptional regulators, although mechanisms responsible for E7-mediated transcriptional regulation are incompletely understood. Type I IFN signaling promotes the expression of anti-viral genes, called interferon-stimulated genes (ISGs), through the phosphorylation and activation of STAT1. In this study, we have observed that the CR3 domain of E7 contributes to the episomal maintenance of viral genomes. Transcriptome analysis revealed that E7 transcriptionally suppresses a subset of ISGs but not through regulation of STAT1 activation. Instead, we discovered that E7 associates with Mediator kinase CDK8 and this is correlated with the recruitment of CDK8 to ISG promoters and reduced ISG expression. E7 fails to suppress ISGs in the absence of CDK8, indicating that CDK8 function contributes to the suppression of ISGs by E7. Altogether, E7/CDK8 association may be a novel mechanism by which E7 inhibits innate immune signaling.


Subject(s)
Cyclin-Dependent Kinase 8/genetics , Cyclin-Dependent Kinase 8/metabolism , Human papillomavirus 16/metabolism , Interferons/metabolism , Papillomavirus E7 Proteins/metabolism , Cell Line , Gene Expression , Gene Expression Regulation , Human papillomavirus 16/genetics , Humans , Keratinocytes/virology , Mutation , Oncogene Proteins, Viral/metabolism , Papillomavirus E7 Proteins/genetics , Papillomavirus Infections/virology , Phosphorylation , STAT1 Transcription Factor , Signal Transduction
5.
J Virol ; 94(2)2020 01 06.
Article in English | MEDLINE | ID: mdl-31666385

ABSTRACT

Human papillomaviruses (HPVs) infect keratinocytes of stratified epithelia. Long-term persistence of infection is a critical risk factor for the development of HPV-induced malignancies. Through the actions of its oncogenes, HPV evades host immune responses to facilitate its productive life cycle. In this work, we discovered a previously unknown function of the HPV16 E5 oncoprotein in the suppression of interferon (IFN) responses. This suppression is focused on keratinocyte-specific IFN-κ and is mediated through E5-induced changes in growth factor signaling pathways, as identified through phosphoproteomics analysis. The loss of E5 in keratinocytes maintaining the complete HPV16 genome results in the derepression of IFNK transcription and subsequent JAK/STAT-dependent upregulation of several IFN-stimulated genes (ISGs) at both the mRNA and protein levels. We also established a link between the loss of E5 and the subsequent loss of genome maintenance and stability, resulting in increased genome integration.IMPORTANCE Persistent human papillomavirus infections can cause a variety of significant cancers. The ability of HPV to persist depends on evasion of the host immune system. In this study, we show that the HPV16 E5 protein can suppress an important aspect of the host immune response. In addition, we find that the E5 protein is important for helping the virus avoid integration into the host genome, which is a frequent step along the pathway to cancer development.


Subject(s)
Genome, Viral , Human papillomavirus 16/metabolism , Interferon Type I/metabolism , Keratinocytes , Oncogene Proteins, Viral/metabolism , Papillomavirus Infections , Plasmids/metabolism , Signal Transduction , Cell Line , Genomic Instability , Human papillomavirus 16/genetics , Humans , Interferon Type I/genetics , Keratinocytes/metabolism , Keratinocytes/pathology , Keratinocytes/virology , Oncogene Proteins, Viral/genetics , Papillomavirus Infections/genetics , Papillomavirus Infections/metabolism , Papillomavirus Infections/pathology , Plasmids/genetics
6.
mSphere ; 4(4)2019 07 17.
Article in English | MEDLINE | ID: mdl-31315968

ABSTRACT

Jason Bodily works in the field of tumor virology. In this mSphere of Influence article, he reflects on how "Inactivation of Rb in stromal fibroblasts promotes epithelial cell invasion" by Adam Pickard et al. (EMBO J 31:3092-3103, 2012, https://doi.org/10.1038/emboj.2012.153) has impacted his work by making him think about the role of stromal cells in human papillomavirus infections.


Subject(s)
Epithelial Cells/virology , Oncogenes , Papillomaviridae/genetics , Stromal Cells/virology , Humans , Narration , Papillomaviridae/physiology , Papillomavirus Infections/virology
7.
J Virol ; 93(19)2019 10 01.
Article in English | MEDLINE | ID: mdl-31292244

ABSTRACT

Human papillomaviruses (HPVs) infect squamous epithelia and cause several important cancers. Immune evasion is critical for viral persistence. Fibroblasts in the stromal microenvironment provide growth signals and cytokines that are required for proper epithelial differentiation, maintenance, and immune responses and are critical in the development of many cancers. In this study, we examined the role of epithelial-stromal interactions in the HPV16 life cycle using organotypic (raft) cultures as a model. Rafts were created using uninfected human foreskin keratinocytes (HFKs) and HFKs containing either wild-type HPV16 or HPV16 with a stop mutation to prevent the expression of the viral oncogene E5. Microarray analysis revealed significant changes in gene expression patterns in the stroma in response to HPV16, some of which were E5 dependent. Interferon (IFN)-stimulated genes (ISGs) and extracellular matrix remodeling genes were suppressed, the most prominent pathways affected. STAT1, IFNAR1, IRF3, and IRF7 were knocked down in stromal fibroblasts using lentiviral short hairpin RNA (shRNA) transduction. HPV late gene expression and viral copy number in the epithelium were increased when the stromal IFN pathway was disrupted, indicating that the stroma helps control the late phase of the HPV life cycle in the epithelium. Increased late gene expression correlated with increased late keratinocyte differentiation but not decreased IFN signaling in the epithelium. These studies show HPV16 has a paracrine effect on stromal innate immunity, reveal a new role for E5 as a stromal innate immune suppressor, and suggest that stromal IFN signaling may influence keratinocyte differentiation.IMPORTANCE The persistence of high-risk human papillomavirus (HPV) infections is the key risk factor for developing HPV-associated cancers. The ability of HPV to evade host immunity is a critical component of its ability to persist. The environment surrounding a tumor is increasingly understood to be critical in cancer development, including immune evasion. Our studies show that HPV can suppress the expression of immune-related genes in neighboring fibroblasts in a three-dimensional (3D) model of human epithelium. This finding is significant, because it indicates that HPV can control innate immunity not only in the infected cell but also in the microenvironment. In addition, the ability of HPV to regulate stromal gene expression depends in part on the viral oncogene E5, revealing a new function for this protein as an immune evasion factor.


Subject(s)
Host-Pathogen Interactions , Human papillomavirus 16/growth & development , Human papillomavirus 16/immunology , Immune Evasion , Immunity, Innate , Immunologic Factors/antagonists & inhibitors , Interferons/antagonists & inhibitors , Cells, Cultured , Fibroblasts/immunology , Fibroblasts/virology , Gene Expression Profiling , Humans , Keratinocytes/immunology , Keratinocytes/virology , Models, Biological , Papillomavirus Infections/immunology , Papillomavirus Infections/virology , Signal Transduction
8.
Virology ; 519: 1-11, 2018 06.
Article in English | MEDLINE | ID: mdl-29609071

ABSTRACT

Human papillomaviruses (HPVs) cause benign lesions that can lead to malignancy. How cellular changes induced by viral oncogenes contribute to the progeny virion production is not always clear. Stromally-derived growth factors and their receptors are critical for development of malignancy, but their impact on the pre-malignant HPV life cycle is unknown. We show that HPV16 increases levels of Met, a growth factor receptor critical for tumor cell invasion, motility, and cancer metastasis. The viral oncogene E5 is primarily responsible for Met upregulation, with E6 playing a minor role. Met induction by E5 requires the epidermal growth factor receptor, which is also increased by E5 at the mRNA level. E5-induced Met contributes motility of HPV-containing cells. Finally, Met signaling is necessary for viral gene expression, particularly in the differentiation-dependent phase of the viral life cycle. These studies show a new role for E5 in epithelial-stromal interactions, with implications for cancer development.


Subject(s)
Human papillomavirus 16/metabolism , Keratinocytes/virology , Oncogene Proteins, Viral/metabolism , Proto-Oncogene Proteins c-met/genetics , Proto-Oncogene Proteins c-met/metabolism , Up-Regulation , Cell Differentiation , Cell Movement , Cells, Cultured , Human papillomavirus 16/genetics , Humans , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction , Transcriptional Activation
9.
PLoS Pathog ; 14(3): e1006846, 2018 03.
Article in English | MEDLINE | ID: mdl-29494681

ABSTRACT

Herein, we describe a novel infection model that achieves highly efficient infection of primary keratinocytes with human papillomavirus type 16 (HPV16). This cell culture model does not depend on immortalization and is amenable to extensive genetic analyses. In monolayer cell culture, the early but not late promoter was active and yielded a spliced viral transcript pattern similar to HPV16-immortalized keratinocytes. However, relative levels of the E8^E2 transcript increased over time post infection suggesting the expression of this viral repressor is regulated independently of other early proteins and that it may be important for the shift from the establishment to the maintenance phase of the viral life cycle. Both the early and the late promoter were strongly activated when infected cells were subjected to differentiation by growth in methylcellulose. When grown as organotypic raft cultures, HPV16-infected cells expressed late E1^E4 and L1 proteins and replication foci were detected, suggesting that they supported the completion of the viral life cycle. As a proof of principle that the infection system may be used for genetic dissection of viral factors, we analyzed E1, E6 and E7 translation termination linker mutant virus for establishment of infection and genome maintenance. E1 but not E6 and E7 was essential to establish infection. Furthermore, E6 but not E7 was required for episomal genome maintenance. Primary keratinocytes infected with wild type HPV16 immortalized, whereas keratinocytes infected with E6 and E7 knockout virus began to senesce 25 to 35 days post infection. The novel infection model provides a powerful genetic tool to study the role of viral proteins throughout the viral life cycle but especially for immediate early events and enables us to compare low- and high-risk HPV types in the context of infection.


Subject(s)
Gene Expression Regulation, Viral , Human papillomavirus 16/pathogenicity , Keratinocytes/virology , Oncogene Proteins, Viral/metabolism , Papillomavirus Infections/virology , Virus Replication , Cells, Cultured , Humans , Keratinocytes/metabolism , Keratinocytes/pathology , Oncogene Proteins, Viral/genetics , Papillomavirus Infections/genetics , Papillomavirus Infections/metabolism , Papillomavirus Infections/pathology
10.
J Virol ; 92(8)2018 04 15.
Article in English | MEDLINE | ID: mdl-29437968

ABSTRACT

Persistent high-risk human papillomavirus (HPV) infection is the major causal factor in cervical and other anogenital cancers. Because there are currently no therapeutics capable of preventing neoplastic progression of HPV infections, understanding the mechanisms of HPV-mediated persistence, including immune evasion, is a major research priority. The multifunctional growth factor transforming growth factor beta (TGFß) has been shown to inhibit expression of early viral transcripts from cells harboring integrated HPV genomes or cells infected with retroviruses expressing HPV oncoproteins. However, the mechanism of TGFß-induced inhibition has not been fully defined. In this study, we have observed a previously uncharacterized ability of TGFß to repress the differentiation-induced upregulation of late HPV16 gene expression. In addition, interferon kappa (IFN-κ), a keratinocyte-specific, constitutively expressed cytokine suppressed by differentiation, can be transcriptionally induced by TGFß1. TGFß-mediated IFN-κ transcription only occurs in cells containing HPV16, and this is due to TGFß1-mediated reversal of HPV-induced methylation of the IFN-κ promoter through active DNA demethylation mediated by thymine DNA glycosylase (TDG). This novel interaction between growth factor and innate immune signaling may shed light on the mechanisms of HPV persistence and how the virus manipulates both immune and growth factor signaling to promote its life cycle.IMPORTANCE Persistent infection by high-risk HPVs is the primary risk factor for development of HPV-induced cancers. Persistence involves viral evasion of the immune response, including the IFN response. HPV is also known to suppress TGFß signaling, which inhibits viral gene expression. Here, we show that the TGFß and IFN pathways are interrelated in the context of HPV16 infection through the upregulation of IFN-κ by TGFß. The ability of TGFß to induce IFN-κ promoter demethylation and transcriptional activation provides a new explanation for why HPV has evolved mechanisms to inhibit TGFß in infected cells.


Subject(s)
DNA Methylation , Gene Expression Regulation, Viral , Human papillomavirus 16/metabolism , Interferon Type I/metabolism , Keratinocytes/metabolism , Papillomavirus Infections/metabolism , Promoter Regions, Genetic , Transcriptional Activation , Transforming Growth Factor beta/metabolism , Humans , Keratinocytes/pathology , Keratinocytes/virology , Papillomavirus Infections/pathology
11.
J Virol ; 91(24)2017 12 15.
Article in English | MEDLINE | ID: mdl-28978708

ABSTRACT

High-risk human papillomaviruses (HPVs) link their life cycle to epithelial differentiation and require activation of DNA damage pathways for efficient replication. HPVs modulate the expression of cellular transcription factors, as well as cellular microRNAs (miRNAs) to control these activities. One miRNA that has been reported to be repressed in HPV-positive cancers of the cervix and oropharynx is miR-424. Our studies show that miR-424 levels are suppressed in cell lines that stably maintain HPV 31 or 16 episomes, as well as cervical cancer lines that contain integrated genomes such as SiHa. Introduction of expression vectors for miR-424 reduced both the levels of HPV genomes in undifferentiated cells and amplification upon differentiation. Our studies show that the levels of two putative targets of miR-424 that function in DNA damage repair, CHK1 and Wee1, are suppressed in HPV-positive cells, providing an explanation for why this microRNA is targeted in HPV-positive cells.IMPORTANCE We describe here for the first time a critical role for miR-424 in the regulation of HPV replication. HPV E6 and E7 proteins suppress the levels of miR-424, and this is important for controlling the levels of CHK1, which plays a central role in viral replication.


Subject(s)
Alphapapillomavirus/genetics , Genome, Viral , MicroRNAs/genetics , MicroRNAs/metabolism , Virus Replication , Alphapapillomavirus/physiology , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Differentiation , Cells, Cultured , Checkpoint Kinase 1/genetics , Checkpoint Kinase 1/metabolism , Female , Host-Pathogen Interactions/genetics , Humans , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Oncogene Proteins, Viral/genetics , Papillomavirus E7 Proteins/genetics , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Uterine Cervical Neoplasms/virology
12.
Virology ; 507: 179-191, 2017 07.
Article in English | MEDLINE | ID: mdl-28448849

ABSTRACT

Transcripts from the late promoter of human papillomavirus type 16 (HPV16) are upregulated upon host cell differentiation. Differentiation-dependent transcript regulation is thought to sequester viral antigens in the uppermost epithelial layers, facilitating immune evasion. The mechanisms regulating late promoter upregulation during differentiation are poorly characterized. We show that the late promoter is upregulated at the transcriptional level and that the viral enhancer stimulates promoter activity. Using kinase inhibition and chromatin immunoprecipitation analysis, we show evidence for differentiation-dependent enhancement of transcript elongation. Three factors that promote transcript elongation, cyclin dependent kinase 9 (CDK9), CDK8 (a subunit of the Mediator complex), and bromodomain containing protein 4 (Brd4) are recruited to viral genomes upon differentiation, and each plays a role in promoter activity. These results shed light on the transcriptional processes utilized by HPV16 for proper regulation of gene expression during the viral life cycle.


Subject(s)
Gene Expression Regulation, Viral , Human papillomavirus 16/genetics , Papillomavirus Infections/virology , Promoter Regions, Genetic , Transcription, Genetic , Cell Cycle Proteins , Cyclin-Dependent Kinase 8/genetics , Cyclin-Dependent Kinase 8/metabolism , Cyclin-Dependent Kinase 9/genetics , Cyclin-Dependent Kinase 9/metabolism , Human papillomavirus 16/metabolism , Humans , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Papillomavirus Infections/genetics , Papillomavirus Infections/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
13.
Mol Carcinog ; 56(2): 722-734, 2017 02.
Article in English | MEDLINE | ID: mdl-27434882

ABSTRACT

Tumor cell invasion through the extracellular matrix is facilitated by the secretion of lysosome-associated proteases. As a common mechanism for secretion, lysosomes must first traffic to the cell periphery (anterograde trafficking), consistent with invasive cells often containing lysosomes closer to the plasma membrane compared to non-invasive cells. Epithelial to mesenchymal transition (EMT) is a transcriptionally driven program that promotes an invasive phenotype, and Zeb1 is one transcription factor that activates the mesenchymal gene expression program. The role of lysosome trafficking in EMT-driven invasion has not been previously investigated. We found that cells with increased levels of Zeb1 displayed lysosomes located closer to the cell periphery and demonstrated increased protease secretion and invasion in 3-dimensional (3D) cultures compared to their epithelial counterparts. Additionally, preventing anterograde lysosome trafficking via pharmacological inhibition of Na+/H+ exchanger 1 (NHE1) or shRNA depletion of ADP-ribosylation like protein 8b (Arl8b) reversed the invasive phenotype of mesenchymal cells, thus supporting a role for lysosome positioning in EMT-mediated tumor cell invasion. Immunoblot revealed that expression of Na+/H+ exchanger 1 correlated with Zeb1 expression. Furthermore, we found that the transcription factor Zeb1 binds to the Na+/H+ exchanger 1 promoter, suggesting that Zeb1 directly controls Na+/H+ transcription. Collectively, these results provide insight into a novel mechanism regulating Na+/H+ exchanger 1 expression and support a role for anterograde lysosome trafficking in Zeb1-driven cancer progression. © 2016 Wiley Periodicals, Inc.


Subject(s)
Cation Transport Proteins/genetics , Neoplasm Invasiveness/genetics , Prostatic Neoplasms/genetics , Sodium-Hydrogen Exchangers/genetics , Up-Regulation , Zinc Finger E-box-Binding Homeobox 1/metabolism , Cell Line, Tumor , Cell Movement , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic , Humans , Lysosomes/genetics , Lysosomes/metabolism , Lysosomes/pathology , Male , Neoplasm Invasiveness/pathology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Sodium-Hydrogen Exchanger 1 , Transcriptional Activation , Zinc Finger E-box-Binding Homeobox 1/genetics
14.
Virus Res ; 231: 56-75, 2017 03 02.
Article in English | MEDLINE | ID: mdl-27818212

ABSTRACT

High-risk human papillomaviruses (HPVs) encode oncoproteins which manipulate gene expression patterns in the host keratinocytes to facilitate viral replication, regulate viral transcription, and promote immune evasion and persistence. In some cases, oncoprotein-induced changes in host cell behavior can cause progression to cancer, but a complete picture of the functions of the viral oncoproteins in the productive HPV life cycle remains elusive. E7 is the HPV-encoded factor most responsible for maintaining cell cycle competence in differentiating keratinocytes. Through interactions with dozens of host factors, E7 has an enormous impact on host gene expression patterns. In this review, we will examine the role of E7 specifically as a regulator of transcription. We will discuss mechanisms of regulation of cell cycle-related genes by E7 as well as genes involved in immune regulation, growth factor signaling, DNA damage responses, microRNAs, and others pathways. We will also discuss some unanswered questions about how transcriptional regulation by E7 impacts the biology of HPV in both benign and malignant conditions.


Subject(s)
Immune Evasion , Papillomaviridae/genetics , Papillomavirus E7 Proteins/genetics , Papillomavirus Infections/virology , Skin Neoplasms/virology , Transcription, Genetic , Carcinogenesis/genetics , Carcinogenesis/immunology , Carcinogenesis/pathology , Cell Cycle/genetics , Cell Cycle/immunology , Cell Cycle Proteins/genetics , Cell Cycle Proteins/immunology , Cell Differentiation , Cell Line , Gene Expression Regulation , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/immunology , Keratinocytes/immunology , Keratinocytes/virology , Papillomaviridae/growth & development , Papillomaviridae/pathogenicity , Papillomavirus E7 Proteins/immunology , Papillomavirus Infections/genetics , Papillomavirus Infections/immunology , Papillomavirus Infections/pathology , Signal Transduction , Skin Neoplasms/genetics , Skin Neoplasms/immunology , Skin Neoplasms/pathology , Virus Replication
15.
PLoS Pathog ; 11(9): e1005181, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26405826

ABSTRACT

Human papillomaviruses (HPV) regulate their differentiation-dependent life cycles by activating a number of cellular pathways, such as the DNA damage response, through control of post-translational protein modification. Sirtuin 1 (SIRT1) is a protein deacetylase that modulates the acetylation of a number of cellular substrates, resulting in activation of pathways controlling gene expression and DNA damage repair. Our studies indicate that SIRT1 levels are increased in cells containing episomes of high-risk HPV types through the combined action of the E6 and E7 oncoproteins. Knockdown of SIRT1 in these cells with shRNAs impairs viral activities including genome maintenance, amplification and late gene transcription, with minimal effects on cellular proliferation ability. Abrogation of amplification was also seen following treatment with the SIRT1 deacetylase inhibitor, EX-527. Importantly, SIRT1 binds multiple regions of the HPV genome in undifferentiated cells, but this association is lost upon of differentiation. SIRT1 regulates the acetylation of Histone H1 (Lys26) and H4 (Lys16) bound to HPV genomes and this may contribute to regulation of viral replication and gene expression. The differentiation-dependent replication of high-risk HPVs requires activation of factors in the Ataxia Telangiectasia Mutated (ATM) pathway and SIRT1 regulates the recruitment of both NBS1 and Rad51 to the viral genomes. These observations demonstrate that SIRT1 is a critical regulator of multiple aspects of the high-risk HPV life cycle.


Subject(s)
Cell Cycle Proteins/metabolism , Nuclear Proteins/metabolism , Papillomavirus Infections/metabolism , Rad51 Recombinase/metabolism , Sirtuin 1/metabolism , Virus Replication/physiology , Acetylation , Blotting, Western , Cells, Cultured , Chromatin Immunoprecipitation , Fluorescent Antibody Technique , Histones/metabolism , Humans , Immunoblotting , Life Cycle Stages/physiology , Papillomaviridae/physiology , RNA, Small Interfering , Transduction, Genetic
16.
J Virol ; 89(9): 4980-91, 2015 May.
Article in English | MEDLINE | ID: mdl-25694599

ABSTRACT

UNLABELLED: To replicate the double-stranded human papillomavirus 16 (HPV16) DNA genome, viral proteins E1 and E2 associate with the viral origin of replication, and E2 can also regulate transcription from adjacent promoters. E2 interacts with host proteins in order to regulate both transcription and replication; TopBP1 and Brd4 are cellular proteins that interact with HPV16 E2. Previous work with E2 mutants demonstrated the Brd4 requirement for the transactivation properties of E2, while TopBP1 is required for DNA replication induced by E2 from the viral origin of replication in association with E1. More-recent studies have also implicated Brd4 in the regulation of DNA replication by E2 and E1. Here, we demonstrate that both TopBP1 and Brd4 are present at the viral origin of replication and that interaction with E2 is required for optimal initiation of DNA replication. Both cellular proteins are present in E1-E2-containing nuclear foci, and the viral origin of replication is required for the efficient formation of these foci. Short hairpin RNA (shRNA) against either TopBP1 or Brd4 destroys the E1-E2 nuclear bodies but has no effect on E1-E2-mediated levels of DNA replication. An E2 mutation in the context of the complete HPV16 genome that compromises Brd4 interaction fails to efficiently establish episomes in primary human keratinocytes. Overall, the results suggest that interactions between TopBP1 and E2 and between Brd4 and E2 are required to correctly initiate DNA replication but are not required for continuing DNA replication, which may be mediated by alternative processes such as rolling circle amplification and/or homologous recombination. IMPORTANCE: Human papillomavirus 16 (HPV16) is causative in many human cancers, including cervical and head and neck cancers, and is responsible for the annual deaths of hundreds of thousands of people worldwide. The current vaccine will save lives in future generations, but antivirals targeting HPV16 are required for the alleviation of disease burden on the current, and future, generations. Targeting viral DNA replication that is mediated by two viral proteins, E1 and E2, in association with cellular proteins such as TopBP1 and Brd4 would have therapeutic benefits. This report suggests a role for these cellular proteins in the initiation of viral DNA replication by HPV16 E1-E2 but not for continuing replication. This is important if viral replication is to be effectively targeted; we need to understand the viral and cellular proteins required at each phase of viral DNA replication so that it can be effectively disrupted.


Subject(s)
Carrier Proteins/metabolism , DNA Replication , DNA-Binding Proteins/metabolism , Host-Pathogen Interactions , Human papillomavirus 16/physiology , Nuclear Proteins/metabolism , Oncogene Proteins, Viral/metabolism , Transcription Factors/metabolism , Cell Cycle Proteins , Humans , Virus Replication
17.
Methods Mol Biol ; 1249: 81-91, 2015.
Article in English | MEDLINE | ID: mdl-25348299

ABSTRACT

Human papillomaviruses are the causative agents of several cancers, but only a minority of HPV infections progress to malignancy. In order to better understand HPV biology during the normal, differentiation-dependent life cycle, a cell culture model that maintains the complete episomal genome and permits host cell differentiation is critical. Furthermore, the use of cloned DNA as a starting material is important to facilitate genetic analyses. In this chapter, procedures for isolating human keratinocytes, establishing cell lines maintaining HPV16 genomes, and inducing cellular differentiation, which permits analysis of both early and late stages in the viral life cycle, are described.


Subject(s)
Genes, Viral , Genetic Techniques , Oncogenes , Papillomaviridae/genetics , Cell Differentiation , Cell Line , Foreskin/cytology , Humans , Infant, Newborn , Keratinocytes/cytology , Male , Plasmids/genetics , Transfection
18.
Virology ; 443(1): 11-9, 2013 Aug 15.
Article in English | MEDLINE | ID: mdl-23725693

ABSTRACT

Human papillomaviruses (HPVs) are the causative agents of cervical and other cancers. The oncoprotein E7 activates the cell cycle and makes possible replication of the viral genome in differentiating epithelia. The HPV16 late promoter is activated upon cellular differentiation and regulates late gene expression. We investigated the effect of E7 on the late promoter and found that E7 was able to activate the promoter. In contrast, the other known viral transcriptional regulator, E2, had no effect on the late promoter. Promoter activation by E7 occurred despite inhibition of promoter activity by factors involved in the cell cycle, such as cyclin dependent kinases and E2F transcription factors, and by the ability of E7 to disrupt several aspects of cellular differentiation. These results suggest a new role for E7 in the context of the viral life cycle and shed light on the complex regulation of viral gene expression in infected, differentiating epithelia.


Subject(s)
Cell Cycle , Gene Expression Regulation, Viral , Host-Pathogen Interactions , Human papillomavirus 16/physiology , Papillomavirus E7 Proteins/metabolism , Promoter Regions, Genetic , Virus Replication , Cells, Cultured , DNA-Binding Proteins/metabolism , Human papillomavirus 16/genetics , Humans , Keratinocytes/virology , Oncogene Proteins, Viral/metabolism
19.
J Virol ; 86(23): 12806-15, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22973044

ABSTRACT

In human papillomavirus DNA replication, the viral protein E2 forms homodimers and binds to 12-bp palindromic DNA sequences surrounding the origin of DNA replication. Via a protein-protein interaction, it then recruits the viral helicase E1 to an A/T-rich origin of replication, whereupon a dihexamer forms, resulting in DNA replication initiation. In order to carry out DNA replication, the viral proteins must interact with host factors that are currently not all known. An attractive cellular candidate for regulating viral replication is TopBP1, a known interactor of the E2 protein. In mammalian DNA replication, TopBP1 loads DNA polymerases onto the replicative helicase after the G(1)-to-S transition, and this process is tightly cell cycle controlled. The direct interaction between E2 and TopBP1 would allow E2 to bypass this cell cycle control, resulting in DNA replication more than once per cell cycle, which is a requirement for the viral life cycle. We report here the generation of an HPV16 E2 mutant compromised in TopBP1 interaction in vivo and demonstrate that this mutant retains transcriptional activation and repression functions but has suboptimal DNA replication potential. Introduction of this mutant into a viral life cycle model results in the failure to establish viral episomes. The results present a potential new antiviral target, the E2-TopBP1 interaction, and increase our understanding of the viral life cycle, suggesting that the E2-TopBP1 interaction is essential.


Subject(s)
Carrier Proteins/metabolism , DNA Replication/physiology , DNA-Binding Proteins/metabolism , Human papillomavirus 16/physiology , Nuclear Proteins/metabolism , Oncogene Proteins, Viral/metabolism , Blotting, Southern , Blotting, Western , DNA Primers/genetics , Densitometry , Dimerization , HEK293 Cells , Human papillomavirus 16/metabolism , Humans , Immunoprecipitation , Mutagenesis, Site-Directed , Plasmids/genetics , Replication Origin/physiology , Reverse Transcriptase Polymerase Chain Reaction
20.
J Virol ; 85(17): 8852-62, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21697473

ABSTRACT

Human papillomaviruses (HPVs) are the causative agents of several important genital and other mucosal cancers. The HPV16 E7 gene encodes a viral oncogene that is necessary for the continued growth of cancer cells, but its role in the normal, differentiation-dependent life cycle of the virus is not fully understood. The function of E7 in the viral life cycle was examined using a series of mutations of E7 created in the context of the complete HPV16 genome. The effect of these E7 mutations on key events of the viral life cycle, including immortalization, episomal maintenance, late promoter activation, and infectious virion synthesis, was examined. Our studies show that the pRb binding domain is indispensable for early viral activities, whereas the C-terminal zinc finger domain contributed primarily to very late events. Mutations of the casein kinase II phosphorylation site caused a complex phenotype involving both the function of E7 protein and a cis element necessary for the activation of the late promoter, identifying for the first time a promoter element important for late promoter function in the context of the viral genome. All mutant genomes tested showed reduced viral titers following growth in organotypic raft cultures. These studies clarify the role of E7 as a regulator of late events in the differentiation-dependent HPV life cycle.


Subject(s)
Cell Transformation, Viral , Human papillomavirus 16/pathogenicity , Open Reading Frames , Papillomavirus E7 Proteins/genetics , Virus Replication , Cell Differentiation , Cells, Cultured , DNA Mutational Analysis , Humans , Papillomavirus E7 Proteins/metabolism , Promoter Regions, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...