Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
1.
Proc Natl Acad Sci U S A ; 120(35): e2305907120, 2023 08 29.
Artigo em Inglês | MEDLINE | ID: mdl-37611058

RESUMO

R-loops are trimeric RNA: DNA hybrids that are important physiological regulators of transcription; however, their aberrant formation or turnover leads to genomic instability and DNA breaks. High-risk human papillomaviruses (HPV) are the causative agents of genital as well as oropharyngeal cancers and exhibit enhanced amounts of DNA breaks. The levels of R-loops were found to be increased up to 50-fold in cells that maintain high-risk HPV genomes and were readily detected in squamous cell cervical carcinomas in vivo but not in normal cells. The high levels of R-loops in HPV-positive cells were present on both viral and cellular sites together with RNase H1, an enzyme that controls their resolution. Depletion of RNase H1 in HPV-positive cells further increased R-loop levels, resulting in impaired viral transcription and replication along with reduced expression of the DNA repair genes such as FANCD2 and ATR, both of which are necessary for viral functions. Overexpression of RNase H1 decreased total R-loop levels, resulting in a reduction of DNA breaks by over 50%. Furthermore, increased RNase H1 expression blocked viral transcription and replication while enhancing the expression of factors in the innate immune regulatory pathway. This suggests that maintaining elevated R-loop levels is important for the HPV life cycle. The E6 viral oncoprotein was found to be responsible for inducing high levels of R-loops by inhibiting p53's transcriptional activity. Our studies indicate that high R-loop levels are critical for HPV pathogenesis and that this depends on suppressing the p53 pathway.


Assuntos
Carcinoma de Células Escamosas , Anemia de Fanconi , Infecções por Papillomavirus , Humanos , Estruturas R-Loop , Proteína Supressora de Tumor p53/genética , Infecções por Papillomavirus/genética
2.
PLoS Pathog ; 18(7): e1010725, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35877778

RESUMO

The cyclic GMP-AMP synthase (cGAS) is a critical regulator of the innate immune response acting as a sensor of double-strand DNAs from pathogens or damaged host DNA. Upon activation, cGAS signals through the STING/TBK1/IRF3 pathway to induce interferon expression. Double stranded DNA viruses target the cGAS pathway to facilitate infection. In HPV positive cells that stably maintain viral episomes, the levels of cGAS were found to be significantly increased over those seen in normal human keratinocytes. Furthermore the downstream effectors of the cGAS pathway, STING and IRF3, were fully active in response to signaling from the secondary messenger cGAMP or poly (dA:dT). In HPV positive cells cGAS was detected in both cytoplasmic puncta as well as in DNA damage induced micronuclei. E6 was responsible for increased levels of cGAS that was dependent on inhibition of p53. CRISPR-Cas9 mediated knockout of cGAS prevented activation of STING and IRF3 but had a minimal effect on viral replication. A primary function of cGAS in HPV positive cells was in response to treatment with etoposide or cisplatin which lead to increased levels of H2AX phosphorylation and activation of caspase 3/7 cleavage while having only a minimal effect on activation of homologous recombination repair factors ATM, ATR or CHK2. In HPV positive cells cGAS was found to regulate the levels of the phosphorylated non-homologous end-joining kinase, DNA-PK, which may contribute to H2AX phosphorylation along with other factors. Importantly cGAS was also responsible for increased levels of DNA breaks along with enhanced apoptosis in HPV positive cells but not in HFKs. This study identifies an important and novel role for cGAS in mediating the response of HPV positive cells to chemotherapeutic drugs.


Assuntos
Alphapapillomavirus , Infecções por Papillomavirus , Alphapapillomavirus/metabolismo , Apoptose , Dano ao DNA , Humanos , Imunidade Inata , Nucleotidiltransferases/genética , Nucleotidiltransferases/metabolismo , Papillomaviridae/metabolismo
3.
Int J Gynecol Pathol ; 38(1): 1-10, 2019 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-29995652

RESUMO

Human papillomaviruses (HPVs) are DNA viruses with epithelial tropism. High-risk types of HPV are the causative agents of the majority of cervical cancers and are responsible for a number of other anogenital as well as oropharyngeal cancers. The life cycle of HPV is closely linked to the differentiation state of its host cell and is dependent on the activation of specific pathways of the DNA damage response. Several proteins from the ataxia telangiectasia mutated and the ataxia telangiectasia mutated and Rad3-related DNA repair pathways, which are essential for maintaining genomic stability in cells, are upregulated in HPV-positive cells and are required for viral replication. Our studies examine the expression of 5 such DNA repair factors-pCHK2, pCHK1, FANCD2, BRCA1, and H2AX-in cervical specimens from patients diagnosed with low-grade, intermediate-grade, or high-grade lesions. The percentage of cells expressing pCHK2, pCHK1, FANCD2, and BRCA1 is significantly higher in high-grade squamous intraepithelial lesions compared with that of either low-grade squamous intraepithelial lesions or normal tissue, particularly in differentiated cell layers. In addition, the distribution of this staining throughout the epithelium is altered with increasing lesion grade. This study characterizes the expression of pCHK2, pCHK1, FANCD2, H2AX and BRCA1 during cervical cancer progression and provides additional insight into the role of these DNA damage response proteins in viral transformation.


Assuntos
Papillomaviridae/isolamento & purificação , Infecções por Papillomavirus/diagnóstico , Displasia do Colo do Útero/diagnóstico , Neoplasias do Colo do Útero/diagnóstico , Diferenciação Celular , Colo do Útero/metabolismo , Colo do Útero/patologia , Colo do Útero/virologia , Dano ao DNA , Reparo do DNA , Progressão da Doença , Feminino , Genótipo , Humanos , Imuno-Histoquímica , Papillomaviridae/genética , Papillomaviridae/fisiologia , Infecções por Papillomavirus/metabolismo , Infecções por Papillomavirus/patologia , Infecções por Papillomavirus/virologia , Sensibilidade e Especificidade , Neoplasias do Colo do Útero/metabolismo , Neoplasias do Colo do Útero/patologia , Neoplasias do Colo do Útero/virologia , Replicação Viral , Displasia do Colo do Útero/metabolismo , Displasia do Colo do Útero/patologia , Displasia do Colo do Útero/virologia
4.
J Virol ; 91(24)2017 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-28978708

RESUMO

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.


Assuntos
Alphapapillomavirus/genética , Genoma Viral , MicroRNAs/genética , MicroRNAs/metabolismo , Replicação Viral , Alphapapillomavirus/fisiologia , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Diferenciação Celular , Células Cultivadas , Quinase 1 do Ponto de Checagem/genética , Quinase 1 do Ponto de Checagem/metabolismo , Feminino , Interações Hospedeiro-Patógeno/genética , Humanos , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas Oncogênicas Virais/genética , Proteínas E7 de Papillomavirus/genética , Proteínas Tirosina Quinases/genética , Proteínas Tirosina Quinases/metabolismo , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Neoplasias do Colo do Útero/virologia
5.
PLoS Pathog ; 12(7): e1005747, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27386862

RESUMO

Human papillomaviruses (HPVs) are epithelial tropic viruses that link their productive life cycles to the differentiation of infected host keratinocytes. A subset of the over 200 HPV types, referred to as high-risk, are the causative agents of most anogenital malignancies. HPVs infect cells in the basal layer, but restrict viral genome amplification, late gene expression, and capsid assembly to highly differentiated cells that are active in the cell cycle. In this study, we demonstrate that HPV proteins regulate the expression and activities of a critical cellular transcription factor, KLF4, through post-transcriptional and post-translational mechanisms. Our studies show that KLF4 regulates differentiation as well as cell cycle progression, and binds to sequences in the upstream regulatory region (URR) to regulate viral transcription in cooperation with Blimp1. KLF4 levels are increased in HPV-positive cells through a post-transcriptional mechanism involving E7-mediated suppression of cellular miR-145, as well as at the post-translational level by E6-directed inhibition of its sumoylation and phosphorylation. The alterations in KLF4 levels and functions results in activation and suppression of a subset of KLF4 target genes, including TCHHL1, VIM, ACTN1, and POT1, that is distinct from that seen in normal keratinocytes. Knockdown of KLF4 with shRNAs in cells that maintain HPV episomes blocked genome amplification and abolished late gene expression upon differentiation. While KLF4 is indispensable for the proliferation and differentiation of normal keratinocytes, it is necessary only for differentiation-associated functions of HPV-positive keratinocytes. Increases in KLF4 levels alone do not appear to be sufficient to explain the effects on proliferation and differentiation of HPV-positive cells indicating that additional modifications are important. KLF4 has also been shown to be a critical regulator of lytic Epstein Barr virus (EBV) replication underscoring the importance of this cellular transcription factor in the life cycles of multiple human cancer viruses.


Assuntos
Regulação da Expressão Gênica/fisiologia , Queratinócitos/virologia , Fatores de Transcrição Kruppel-Like/metabolismo , Papillomaviridae/fisiologia , Replicação Viral/fisiologia , Diferenciação Celular , Células Cultivadas , Imunoprecipitação da Cromatina , Imunofluorescência , Interações Hospedeiro-Patógeno/fisiologia , Humanos , Immunoblotting , Fator 4 Semelhante a Kruppel , Estágios do Ciclo de Vida , Mutagênese Sítio-Dirigida , Análise de Sequência com Séries de Oligonucleotídeos , Infecções por Papillomavirus , Transcrição Gênica
6.
PLoS Pathog ; 11(9): e1005181, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26405826

RESUMO

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.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Proteínas Nucleares/metabolismo , Infecções por Papillomavirus/metabolismo , Rad51 Recombinase/metabolismo , Sirtuína 1/metabolismo , Replicação Viral/fisiologia , Acetilação , Western Blotting , Células Cultivadas , Imunoprecipitação da Cromatina , Imunofluorescência , Histonas/metabolismo , Humanos , Immunoblotting , Estágios do Ciclo de Vida/fisiologia , Papillomaviridae/fisiologia , RNA Interferente Pequeno , Transdução Genética
7.
PLoS Pathog ; 11(4): e1004763, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25875106

RESUMO

Human papillomaviruses infect stratified epithelia and link their productive life cycle to the differentiation state of the host cell. Productive viral replication or amplification is restricted to highly differentiated suprabasal cells and is dependent on the activation of the ATM DNA damage pathway. The ATM pathway has three arms that can act independently of one another. One arm is centered on p53, another on CHK2 and a third on SMC1/NBS1 proteins. A role for CHK2 in HPV genome amplification has been demonstrated but it was unclear what other factors provided important activities. The cohesin protein, SMC1, is necessary for sister chromatid association prior to mitosis. In addition the phosphorylated form of SMC1 plays a critical role together with NBS1 in the ATM DNA damage response. In normal cells, SMC1 becomes phosphorylated in response to radiation, however, in HPV positive cells our studies demonstrate that it is constitutively activated. Furthermore, pSMC1 is found localized in distinct nuclear foci in complexes with γ-H2AX, and CHK2 and bound to HPV DNA. Importantly, knockdown of SMC1 blocks differentiation-dependent genome amplification. pSMC1 forms complexes with the insulator transcription factor CTCF and our studies show that these factors bind to conserved sequence motifs in the L2 late region of HPV 31. Similar motifs are found in most HPV types. Knockdown of CTCF with shRNAs blocks genome amplification and mutation of the CTCF binding motifs in the L2 open reading frame inhibits stable maintenance of viral episomes in undifferentiated cells as well as amplification of genomes upon differentiation. These findings suggest a model in which SMC1 factors are constitutively activated in HPV positive cells and recruited to viral genomes through complex formation with CTCF to facilitate genome amplification. Our findings identify both SMC1 and CTCF as critical regulators of the differentiation-dependent life cycle of high-risk human papillomaviruses.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Proteínas Cromossômicas não Histona/metabolismo , Queratinócitos/virologia , Papillomaviridae/fisiologia , Infecções por Papillomavirus/metabolismo , Proteínas Repressoras/metabolismo , Replicação Viral/fisiologia , Fator de Ligação a CCCTC , Diferenciação Celular , Células Cultivadas , Imunoprecipitação da Cromatina , Imunofluorescência , Humanos , Immunoblotting , Queratinócitos/metabolismo , Mutagênese Sítio-Dirigida , Transfecção , Ativação Viral
8.
J Virol ; 89(8): 4668-75, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25673709

RESUMO

UNLABELLED: The life cycle of human papillomaviruses (HPVs) is dependent upon differentiation of the infected host epithelial cell as well as activation of the ataxia telangiectasia mutated (ATM) DNA repair pathway that in normal cells acts to repair double-strand DNA breaks. In normal cells, following DNA damage the acetyltransferase Tip60 must acetylate ATM proteins prior to their full activation by autophosphorylation. E6 proteins have been shown to induce the degradation of Tip60, suggesting that Tip60 action may not be required for activation of the ATM pathway in HPV-positive cells. We investigated what role, if any, Tip60 plays in regulating the differentiation-dependent HPV life cycle. Our study indicates that Tip60 levels and activity are increased in cells that stably maintain complete HPV genomes as episomes, while low levels are seen in cells that express only HPV E6 and E7 proteins. Knockdown of Tip60 with short hairpin RNAs in cells that maintain HPV episomes blocked ATM induction and differentiation-dependent genome amplification, demonstrating the critical role of Tip60 in the viral life cycle. The JAK/STAT transcription factor STAT-5 has previously been shown to regulate the phosphorylation of ATM. Our studies demonstrate that STAT-5 regulates Tip60 activation and this occurs in part by targeting glycogen synthase kinase 3ß (GSK3ß). Inhibition of either STAT-5, Tip60, or GSK3ß blocked differentiation-dependent genome amplification. Taken together, our findings identify Tip60 to be an important regulator of HPV genome amplification whose activity during the viral life cycle is controlled by STAT-5 and the kinase GSK3ß. IMPORTANCE: Human papillomaviruses (HPVs) are the etiological agents of cervical and other anogenital cancers. HPVs regulate their differentiation-dependent life cycle by activation of DNA damage pathways. This study demonstrates that HPVs regulate the ATM DNA damage pathway through the action of the acetyltransferase Tip60. Furthermore, the innate immune regulator STAT-5 and the kinase GSK3ß mediate the activation of Tip60 in HPV-positive cells. This study identifies critical regulators of the HPV life cycle.


Assuntos
Alphapapillomavirus/fisiologia , Diferenciação Celular/fisiologia , Células Epiteliais/virologia , Quinase 3 da Glicogênio Sintase/metabolismo , Histona Acetiltransferases/metabolismo , Fator de Transcrição STAT5/metabolismo , Acetilação , Alphapapillomavirus/genética , Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Northern Blotting , Southern Blotting , Western Blotting , Células Cultivadas , Primers do DNA/genética , Células Epiteliais/fisiologia , Técnicas de Silenciamento de Genes , Glicogênio Sintase Quinase 3 beta , Histona Acetiltransferases/genética , Humanos , Lisina Acetiltransferase 5 , Fosforilação , Plasmídeos/genética , RNA Interferente Pequeno/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
9.
PLoS Pathog ; 9(4): e1003295, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23593005

RESUMO

High-risk human papillomavirus (HPV) must evade innate immune surveillance to establish persistent infections and to amplify viral genomes upon differentiation. Members of the JAK-STAT family are important regulators of the innate immune response and HPV proteins downregulate expression of STAT-1 to allow for stable maintenance of viral episomes. STAT-5 is another member of this pathway that modulates the inflammatory response and plays an important role in controlling cell cycle progression in response to cytokines and growth factors. Our studies show that HPV E7 activates STAT-5 phosphorylation without altering total protein levels. Inhibition of STAT-5 phosphorylation by the drug pimozide abolishes viral genome amplification and late gene expression in differentiating keratinocytes. In contrast, treatment of undifferentiated cells that stably maintain episomes has no effect on viral replication. Knockdown studies show that the STAT-5ß isoform is mainly responsible for this activity and that this is mediated through the ATM DNA damage response. A downstream target of STAT-5, the peroxisome proliferator-activated receptor γ (PPARγ) contributes to the effects on members of the ATM pathway. Overall, these findings identify an important new regulatory mechanism by which the innate immune regulator, STAT-5, promotes HPV viral replication through activation of the ATM DNA damage response.


Assuntos
Alphapapillomavirus/genética , Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Reparo do DNA/genética , Proteínas E7 de Papillomavirus/metabolismo , Fator de Transcrição STAT5/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Alphapapillomavirus/fisiologia , Diferenciação Celular , Células Cultivadas , Dano ao DNA/genética , Células Epiteliais/metabolismo , Humanos , Queratinócitos/metabolismo , PPAR gama/metabolismo , Proteínas E7 de Papillomavirus/genética , Fosforilação , Pimozida/farmacologia , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Interferência de RNA , Fator de Transcrição STAT5/genética , Proteínas Supressoras de Tumor/genética , Replicação Viral
10.
J Virol ; 87(10): 6037-43, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23468503

RESUMO

Human papillomaviruses (HPVs) modulate expression of host microRNAs. Our deep-sequencing analysis of organotypic raft cultures identified microRNA 145 (miR-145) as a differentiation-dependent microRNA that has functionally active target sequences in the HPV-31 E1 and E2 open reading frames. Overexpression of miR-145 in HPV-positive cells resulted in reduced genome amplification and late gene expression, along with decreased levels of cellular transcription factor KLF-4. Our studies show that HPV modulates miR-145 expression to control its own life cycle.


Assuntos
Regulação da Expressão Gênica , Interações Hospedeiro-Patógeno , MicroRNAs/biossíntese , Papillomaviridae/fisiologia , Replicação Viral , Humanos
11.
Recent Results Cancer Res ; 193: 135-48, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24008297

RESUMO

Human papillomaviruses (HPVs) are the causative agents of cervical and other anogenital as well as oral cancers. Approximately fifty percent of virally induced cancers in the USA are associated with HPV infections. HPVs infect stratified epithelia and link productive replication with differentiation. The viral oncoproteins, E6, E7, and E5, play important roles in regulating viral functions during the viral life cycle and also contribute to the development of cancers. p53 and Rb are two major targets of the E6 and E7 oncoproteins, but additional cellular proteins also play important roles. E5 plays an auxiliary role in contributing to the development of cancers. This review will discuss the various targets of these viral proteins and what roles they play in viral pathogenesis.


Assuntos
Transformação Celular Neoplásica/patologia , Papillomaviridae/patogenicidade , Infecções por Papillomavirus/virologia , Neoplasias do Colo do Útero/virologia , Feminino , Humanos , Infecções por Papillomavirus/patologia , Neoplasias do Colo do Útero/patologia
12.
J Virol ; 86(17): 9520-6, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22740399

RESUMO

Human papillomaviruses (HPV) activate the ataxia telangiectasia mutated (ATM)-dependent DNA damage response to induce viral genome amplification upon epithelial differentiation. Our studies show that along with members of the ATM pathway, HPV proteins also localize factors involved in homologous DNA recombination to distinct nuclear foci that contain HPV genomes and cellular replication factors. These studies indicate that HPV activates the ATM pathway to recruit repair factors to viral genomes and allow for efficient replication.


Assuntos
Alphapapillomavirus/fisiologia , Reparo do DNA , Recombinação Homóloga , Infecções por Papillomavirus/genética , Replicação Viral , Alphapapillomavirus/genética , Proteínas Mutadas de Ataxia Telangiectasia , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular , Núcleo Celular/genética , Núcleo Celular/metabolismo , Quinase do Ponto de Checagem 2 , Dano ao DNA , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Humanos , Infecções por Papillomavirus/metabolismo , Infecções por Papillomavirus/virologia , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Transporte Proteico , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo
13.
J Virol ; 86(23): 12806-15, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22973044

RESUMO

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.


Assuntos
Proteínas de Transporte/metabolismo , Replicação do DNA/fisiologia , Proteínas de Ligação a DNA/metabolismo , Papillomavirus Humano 16/fisiologia , Proteínas Nucleares/metabolismo , Proteínas Oncogênicas Virais/metabolismo , Southern Blotting , Western Blotting , Primers do DNA/genética , Densitometria , Dimerização , Células HEK293 , Papillomavirus Humano 16/metabolismo , Humanos , Imunoprecipitação , Mutagênese Sítio-Dirigida , Plasmídeos/genética , Origem de Replicação/fisiologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa
14.
J Virol ; 86(15): 8131-8, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22623785

RESUMO

High-risk human papillomaviruses (HPVs) deregulate epidermal differentiation and cause anogenital and head and neck squamous cell carcinomas (SCCs). The E7 gene is considered the predominant viral oncogene and drives proliferation and genome instability. While the implementation of routine screens has greatly reduced the incidence of cervical cancers which are almost exclusively HPV positive, the proportion of HPV-positive head and neck SCCs is on the rise. High levels of HPV oncogene expression and genome load are linked to disease progression, but genetic risk factors that regulate oncogene abundance and/or genome amplification remain poorly understood. Fanconi anemia (FA) is a genome instability syndrome characterized at least in part by extreme susceptibility to SCCs. FA results from mutations in one of 15 genes in the FA pathway, whose protein products assemble in the nucleus and play important roles in DNA damage repair. We report here that loss of FA pathway components FANCA and FANCD2 stimulates E7 protein accumulation in human keratinocytes and causes increased epithelial proliferation and basal cell layer expansion in the HPV-positive epidermis. Additionally, FANCD2 loss stimulates HPV genome amplification in differentiating cells, demonstrating that the intact FA pathway functions to restrict the HPV life cycle. These findings raise the possibility that FA genes suppress HPV infection and disease and suggest possible mechanism(s) for reported associations of HPV with an FA cohort in Brazil and for allelic variation of FA genes with HPV persistence in the general population.


Assuntos
Proteína do Grupo de Complementação A da Anemia de Fanconi/metabolismo , Proteína do Grupo de Complementação D2 da Anemia de Fanconi/metabolismo , Genoma Viral/fisiologia , Papillomavirus Humano 16/fisiologia , Proteínas E7 de Papillomavirus/metabolismo , Infecções por Papillomavirus/metabolismo , Replicação Viral/fisiologia , Brasil/epidemiologia , Carcinoma de Células Escamosas/epidemiologia , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patologia , Carcinoma de Células Escamosas/virologia , Linhagem Celular Transformada , Anemia de Fanconi/epidemiologia , Anemia de Fanconi/genética , Anemia de Fanconi/metabolismo , Anemia de Fanconi/patologia , Anemia de Fanconi/virologia , Proteína do Grupo de Complementação A da Anemia de Fanconi/genética , Proteína do Grupo de Complementação D2 da Anemia de Fanconi/genética , Feminino , Neoplasias de Cabeça e Pescoço/epidemiologia , Neoplasias de Cabeça e Pescoço/genética , Neoplasias de Cabeça e Pescoço/metabolismo , Neoplasias de Cabeça e Pescoço/patologia , Neoplasias de Cabeça e Pescoço/virologia , Humanos , Queratinócitos/metabolismo , Queratinócitos/patologia , Queratinócitos/virologia , Masculino , Proteínas E7 de Papillomavirus/genética , Infecções por Papillomavirus/epidemiologia , Infecções por Papillomavirus/genética , Infecções por Papillomavirus/patologia
15.
J Virol ; 85(17): 8863-9, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21715473

RESUMO

The late phase of the human papillomavirus (HPV) life cycle is linked to epithelial differentiation, and we investigated the factors that regulate this process. One potential regulator is p63, a member of the p53 family of proteins, which modulates epithelial development, as well as proliferation capability, in stem cells. In this study, we examined the role of p63 in the HPV life cycle using a lentiviral knockdown system for p63. In epithelial cells, the ΔN truncated isoforms of p63 predominate, while the full-length TA isoforms are present at very low levels. Upon the differentiation of normal keratinocytes, p63 levels rapidly decreased while higher levels were retained in HPV-positive cells. Our studies indicate that reducing p63 levels in differentiated HPV-positive cells resulted in the loss of viral genome amplification and late gene expression. p63 regulates the expression of cell cycle regulators, and we determined that cyclin A, cyclin B1, cdk1, and cdc25c were reduced in p63-deficient, HPV-positive keratinocytes, which suggests a possible mechanism of action. In addition, activation of the DNA repair pathway is necessary for genome amplification, and the expression of two members, BRCA2 and RAD51, was altered in the absence of p63 in HPV-positive cells. Our studies indicate that p63 is necessary for the activation of differentiation-dependent HPV late viral functions and provide insights into relevant cellular targets.


Assuntos
Diferenciação Celular , Regulação Viral da Expressão Gênica , Interações Hospedeiro-Patógeno , Queratinócitos/fisiologia , Queratinócitos/virologia , Proteínas de Membrana/metabolismo , Papillomaviridae/patogenicidade , Proteínas de Ciclo Celular/biossíntese , Células Cultivadas , Humanos , Proteínas Virais/biossíntese
16.
J Virol ; 85(18): 9486-94, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21734056

RESUMO

High-risk human papillomaviruses (HPVs) infect stratified epithelia to establish persistent infections that maintain low-copy-number episomes in infected basal cells. Amplification of viral genomes occurs upon keratinocyte differentiation, followed by virion synthesis. During persistent HPV infections, viral proteins act to evade surveillance by both innate and adaptive immune responses. One of the primary pathways regulating the innate immune response is the JAK/STAT pathway. Our studies indicate that the expression of STAT-1, but not other members of interferon (IFN)-stimulated gene factor 3 (ISGF-3) complex such as STAT-2 and IFN regulatory factor 9 (IRF9), is selectively suppressed by HPV proteins at the level of transcription. Both E6 and E7 oncoproteins independently suppress the expression of STAT-1, and mutational analyses indicate that the E6 targeting E6-associated protein (E6AP) is responsible for suppression. The levels of STAT-1 proteins increase upon differentiation of both normal and HPV-positive cells but are still significantly reduced in the latter cells. Transient restoration of STAT-1 levels in HPV-positive cells using recombinant retroviruses significantly impaired viral amplification upon differentiation while long-term increases abrogated maintenance of episomes. Similarly, increased levels of STAT-1 induced by gamma interferon treatment inhibited HPV genome amplification upon differentiation. Overall, our findings demonstrate that suppression of STAT-1 expression by HPV proteins is necessary for genome amplification and maintenance of episomes, suggesting an important role for this activity in viral pathogenesis.


Assuntos
Interações Hospedeiro-Patógeno , Queratinócitos/virologia , Proteínas Oncogênicas Virais/metabolismo , Papillomaviridae/patogenicidade , Proteínas E7 de Papillomavirus/metabolismo , Proteínas Repressoras/metabolismo , Fator de Transcrição STAT1/antagonistas & inibidores , Replicação Viral , Células Cultivadas , Humanos , Evasão da Resposta Imune , Queratinócitos/imunologia , Papillomaviridae/imunologia , Plasmídeos/metabolismo , Fator de Transcrição STAT1/imunologia , Transcrição Gênica , Ubiquitina-Proteína Ligases/metabolismo
17.
J Virol ; 85(17): 8996-9012, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21734051

RESUMO

Replication of the papillomavirus genome is initiated by the assembly of a complex between the viral E1 and E2 proteins at the origin. The E1 helicase is comprised of a C-terminal ATPase/helicase domain, a central domain that binds to the origin, and an N-terminal regulatory region that contains nuclear import and export signals mediating its nucleocytoplasmic shuttling. We previously reported that nuclear accumulation of E1 has a deleterious effect on cellular proliferation which can be prevented by its nuclear export. Here we have shown that nuclear accumulation of E1 from different papillomavirus types blocks cell cycle progression in early S phase and triggers the activation of a DNA damage response (DDR) and of the ATM pathway in a manner that requires both the origin-binding and ATPase activities of E1. Complex formation with E2 reduces the ability of E1 to induce a DDR but does not prevent cell cycle arrest. Transient viral DNA replication still occurs in S-phase-arrested cells but surprisingly is neither affected by nor dependent on induction of a DDR and of the ATM kinase. Finally, we provide evidence that a DDR is also induced in human papillomavirus type 31 (HPV31)-immortalized keratinocytes expressing a mutant E1 protein defective for nuclear export. We propose that nuclear export of E1 prevents cell cycle arrest and the induction of a DDR during the episomal maintenance phase of the viral life cycle and that complex formation with E2 further safeguards undifferentiated cells from undergoing a DDR when E1 is in the nucleus.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Ciclo Celular , DNA Helicases/metabolismo , Reparo do DNA , Proteínas de Ligação a DNA/metabolismo , Interações Hospedeiro-Patógeno , Proteínas Oncogênicas Virais/metabolismo , Papillomaviridae/patogenicidade , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Proteínas Mutadas de Ataxia Telangiectasia , Linhagem Celular , Humanos , Papillomaviridae/crescimento & desenvolvimento , Replicação Viral
18.
J Virol ; 85(17): 8852-62, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21697473

RESUMO

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.


Assuntos
Transformação Celular Viral , Papillomavirus Humano 16/patogenicidade , Fases de Leitura Aberta , Proteínas E7 de Papillomavirus/genética , Replicação Viral , Diferenciação Celular , Células Cultivadas , Análise Mutacional de DNA , Humanos , Proteínas E7 de Papillomavirus/metabolismo , Regiões Promotoras Genéticas
19.
J Virol ; 84(10): 5212-21, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20219920

RESUMO

Human papillomaviruses (HPV) link their life cycles to epithelial differentiation and induce productive replication of viral DNA in suprabasal cells. Viral-DNA amplification requires cells to remain active in the cell cycle upon differentiation. This is in contrast to normal cells, which lose proliferative capability upon differentiation. One factor that negatively regulates proliferative capability upon differentiation is microRNA 203 (miR-203), which is expressed primarily in suprabasal epithelial cells. Although HPVs do not encode their own microRNAs (miRNAs), they modulate expression of cellular miRNAs to regulate the activities of cellular proteins. We show that the HPV E7 protein downregulates miR-203 expression upon differentiation, which may occur through the mitogen-activated protein (MAP) kinase/protein kinase C (PKC) pathway. One target of miR-203 is the p63 family of transcription factors, and we demonstrate that HPV-positive cells maintain significantly higher levels of these factors upon differentiation than do normal keratinocytes. Several downstream targets of p63, CARM-1, p21, and Bax, were also increased in E7-expressing cells, and their levels were inversely correlated with amounts of miR-203. Introduction of expression vectors for miR-203 into keratinocytes that stably maintain HPV episomes resulted in short-term elevation of HPV genome copy numbers, but these were rapidly lost upon subsequent passage. When HPV-positive cells expressing high levels of miR-203 were induced to differentiate in methylcellulose, impaired genome amplification was observed. We conclude that high levels of miR-203 are inhibitory to HPV amplification and that HPV proteins act to suppress expression of this microRNA to allow productive replication in differentiating cells.


Assuntos
Interações Hospedeiro-Patógeno , Queratinócitos/virologia , MicroRNAs/antagonistas & inibidores , Papillomaviridae/patogenicidade , Proteínas E7 de Papillomavirus/metabolismo , Transativadores/biossíntese , Proteínas Supressoras de Tumor/biossíntese , Células Cultivadas , Inibidor de Quinase Dependente de Ciclina p21/biossíntese , Humanos , Proteína-Arginina N-Metiltransferases/biossíntese , Fatores de Transcrição , Proteína X Associada a bcl-2/biossíntese
20.
J Virol ; 84(22): 11747-60, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20844047

RESUMO

The initiator protein E1 from human papillomavirus (HPV) is a helicase essential for replication of the viral genome. E1 contains three functional domains: a C-terminal enzymatic domain that has ATPase/helicase activity, a central DNA-binding domain that recognizes specific sequences in the origin of replication, and a N-terminal region necessary for viral DNA replication in vivo but dispensable in vitro. This N-terminal portion of E1 contains a conserved nuclear export signal (NES) whose function in the viral life cycle remains unclear. In this study, we provide evidence that nuclear export of HPV31 E1 is inhibited by cyclin E/A-Cdk2 phosphorylation of two serines residues, S92 and S106, located near and within the E1 NES, respectively. Using E1 mutant proteins that are confined to the nucleus, we determined that nuclear export of E1 is not essential for transient viral DNA replication but is important for the long-term maintenance of the HPV episome in undifferentiated keratinocytes. The findings that E1 nuclear export is not required for viral DNA replication but needed for genome maintenance over multiple cell divisions raised the possibility that continuous nuclear accumulation of E1 is detrimental to cellular growth. In support of this possibility, we observed that nuclear accumulation of E1 dramatically reduces cellular proliferation by delaying cell cycle progression in S phase. On the basis of these results, we propose that nuclear export of E1 is required, at least in part, to limit accumulation of this viral helicase in the nucleus in order to prevent its detrimental effect on cellular proliferation.


Assuntos
Alphapapillomavirus/fisiologia , Núcleo Celular/metabolismo , Quinase 2 Dependente de Ciclina/metabolismo , Genoma Viral , Infecções por Papillomavirus/enzimologia , Proteínas Virais/metabolismo , Replicação Viral , Transporte Ativo do Núcleo Celular , Alphapapillomavirus/química , Alphapapillomavirus/genética , Motivos de Aminoácidos , Sequência de Aminoácidos , Linhagem Celular Tumoral , Núcleo Celular/química , Núcleo Celular/genética , Quinase 2 Dependente de Ciclina/genética , Humanos , Dados de Sequência Molecular , Infecções por Papillomavirus/metabolismo , Infecções por Papillomavirus/virologia , Fosforilação , Alinhamento de Sequência , Proteínas Virais/química , Proteínas Virais/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA