Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 76
Filter
Add more filters










Publication year range
1.
Proc Natl Acad Sci U S A ; 120(35): e2305907120, 2023 08 29.
Article in English | MEDLINE | ID: mdl-37611058

ABSTRACT

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.


Subject(s)
Carcinoma, Squamous Cell , Fanconi Anemia , Papillomavirus Infections , Humans , R-Loop Structures , Tumor Suppressor Protein p53/genetics , Papillomavirus Infections/genetics
2.
PLoS Pathog ; 18(7): e1010725, 2022 07.
Article in English | MEDLINE | ID: mdl-35877778

ABSTRACT

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.


Subject(s)
Alphapapillomavirus , Papillomavirus Infections , Alphapapillomavirus/metabolism , Apoptosis , DNA Damage , Humans , Immunity, Innate , Nucleotidyltransferases/genetics , Nucleotidyltransferases/metabolism , Papillomaviridae/metabolism
4.
Viruses ; 13(9)2021 09 14.
Article in English | MEDLINE | ID: mdl-34578402

ABSTRACT

Human papillomaviruses (HPV) are the causative agents of cervical and other anogenital cancers as well as those of the oropharynx. HPV proteins activate host DNA damage repair factors to promote their viral life cycle in stratified epithelia. Activation of both the ATR pathway and the ATM pathway are essential for viral replication and differentiation-dependent genome amplification. These pathways are also important for maintaining host genomic integrity and their dysregulation or mutation is often seen in human cancers. The APOBEC3 family of cytidine deaminases are innate immune factors that are increased in HPV positive cells leading to the accumulation of TpC mutations in cellular DNAs that contribute to malignant progression. The activation of DNA damage repair factors may corelate with expression of APOBEC3 in HPV positive cells. These pathways may actively drive tumor development implicating/suggesting DNA damage repair factors and APOBEC3 as possible therapeutic targets.


Subject(s)
Alphapapillomavirus/genetics , DNA Damage , DNA Repair , Genomic Instability , Host-Pathogen Interactions , Metabolic Networks and Pathways/genetics , Cell Differentiation , Humans , Keratinocytes/virology , Mutation , Virus Replication
5.
Viruses ; 13(7)2021 07 17.
Article in English | MEDLINE | ID: mdl-34372596

ABSTRACT

Persistent infection with high-risk human papillomaviruses (HPVs) is the major risk factor associated with development of anogenital and oropharyngeal cancers. Initial infection by HPVs occurs into basal epithelial cells where viral genomes are established as nuclear episomes and persist until cleared by the immune response. Productive replication or amplification occurs upon differentiation and is dependent upon activation of the ataxia-telangiectasia mutated (ATM), ataxia telangiectasia and RAD3-related (ATR) DNA damage repair (DDR) pathways. In addition to activating DDR pathways, HPVs must escape innate immune surveillance mechanisms by antagonizing sensors, adaptors, interferons and antiviral gene expression. Both DDR and innate immune pathways are key host mechanisms that crosstalk with each other to maintain homeostasis of cells persistently infected with HPVs. Interestingly, it is still not fully understood why some HPV infections get cleared while others do not. Targeting of these two processes with antiviral therapies may provide opportunities for treatment of cancers caused by high-risk HPVs.


Subject(s)
Alphapapillomavirus/genetics , Alphapapillomavirus/immunology , DNA Damage , DNA Repair , Host-Pathogen Interactions/immunology , Immunity, Innate , Cell Differentiation , Genome, Viral , Humans , Virus Replication
6.
mBio ; 12(1)2021 02 09.
Article in English | MEDLINE | ID: mdl-33563836

ABSTRACT

Topoisomerases regulate higher-order chromatin structures through the transient breaking and religating of one or both strands of the phosphodiester backbone of duplex DNA. TOP2ß is a type II topoisomerase that induces double-strand DNA breaks at topologically associated domains (TADS) to relieve torsional stress arising during transcription or replication. TADS are anchored by CCCTC-binding factor (CTCF) and SMC1 cohesin proteins in complexes with TOP2ß. Upon DNA cleavage, a covalent intermediate DNA-TOP2ß (TOP2ßcc) is transiently generated to allow for strand passage. The tyrosyl-DNA phosphodiesterase TDP2 can resolve TOP2ßcc, but failure to do so quickly can lead to long-lasting DNA breaks. Given the role of CTCF/SMC1 proteins in the human papillomavirus (HPV) life cycle, we investigated whether TOP2ß proteins contribute to HPV pathogenesis. Our studies demonstrated that levels of both TOP2ß and TDP2 were substantially increased in cells with high-risk HPV genomes, and this correlated with large amounts of DNA breaks. Knockdown of TOP2ß with short hairpin RNAs (shRNAs) reduced DNA breaks by over 50% as determined through COMET assays. Furthermore, this correlated with substantially reduced formation of repair foci such as phosphorylated H2AX (γH2AX), phosphorylated CHK1 (pCHK1), and phosphorylated SMC1 (pSMC1) indicative of impaired activation of DNA damage repair pathways. Importantly, knockdown of TOP2ß also blocked HPV genome replication. Our previous studies demonstrated that CTCF/SMC1 factors associate with HPV genomes at sites in the late regions of HPV31, and these correspond to regions that also bind TOP2ß. This study identifies TOP2ß as responsible for enhanced levels of DNA breaks in HPV-positive cells and as a regulator of viral replication.IMPORTANCE High-risk human papillomaviruses (HPVs) infect epithelial cells and induce viral genome amplification upon differentiation. HPV proteins activate DNA damage repair pathways by inducing high numbers of DNA breaks in both viral and cellular DNAs. This activation is required for HPV genome replication. TOP2ß is a type II topoisomerase that induces double-strand DNA breaks at topologically associated domains (TADS) to relieve torsional stress arising during transcription or replication. Our studies demonstrate that TOP2ß levels are increased in HPV-positive cells and that this is required for HPV replication. Importantly, our studies further show that knockdown of TOP2ß reduces the number of breaks by over 50% in HPV-positive cells and that this correlates with substantially impaired activation of DNA repair pathways. This study identifies a critical mechanism by which HPV replication is regulated by the topoisomerase TOP2ß through DNA break formation.


Subject(s)
DNA Breaks, Double-Stranded , DNA Topoisomerases, Type II/genetics , Gene Expression Regulation , Host-Pathogen Interactions/genetics , Papillomaviridae/physiology , Virus Replication , Cells, Cultured , Foreskin/cytology , Humans , Keratinocytes/virology , Male
7.
Recent Results Cancer Res ; 217: 141-155, 2021.
Article in English | MEDLINE | ID: mdl-33200365

ABSTRACT

Human papillomaviruses (HPVs) are small DNA viruses that infect basal epithelial cells and are the causative agents of cervical, anogenital, as well as oral cancers. High-risk HPVs are responsible for nearly half of all virally induced cancers. Viral replication and amplification are intimately linked to the stratified epithelium differentiation program. The E6 and E7 proteins contribute to the development of cancers in HPV positive individuals by hijacking cellular processes and causing genetic instability. This genetic instability induces a robust DNA damage response and activating both ATM and ATR repair pathways. These pathways are critical for the productive replication of high-risk HPVs, and understanding how they contribute to the viral life cycle can provide important insights into HPV's role in oncogenesis. This review will discuss the role that differentiation and the DNA damage responses play in productive replication of high-risk HPVs as well as in the development of cancer.


Subject(s)
Alphapapillomavirus , DNA Repair , Oncogene Proteins, Viral , Papillomaviridae , Papillomavirus Infections , Humans , Oncogene Proteins, Viral/genetics , Papillomaviridae/genetics , Papillomaviridae/pathogenicity , Virus Replication
8.
mBio ; 11(4)2020 08 12.
Article in English | MEDLINE | ID: mdl-32788179

ABSTRACT

High-risk human papillomaviruses (HPVs) constitutively activate the ataxia telangiectasia and Rad3-related (ATR) DNA damage response pathway, and this is required for viral replication. In fibroblasts, activated ATR regulates transcription of inflammatory genes through its negative effects on the autophagosome cargo protein p62. In addition, suppression of p62 results in increased levels of the transcription factor GATA4, leading to cellular senescence. In contrast, in HPV-positive keratinocytes, we observed that activation of ATR resulted in increased levels of phosphorylated p62, which in turn lead to reduced levels of GATA4. Knockdown of ATR in HPV-positive cells resulted in decreased p62 phosphorylation and increased GATA4 levels. Transcriptome sequencing (RNA-seq) analysis of HPV-positive cells identified inflammatory genes and interferon factors as negative transcriptional targets of ATR. Furthermore, knockdown of p62 or overexpression of GATA4 in HPV-positive cells leads to inhibition of viral replication. These findings identify a novel role of the ATR/p62 signaling pathway in HPV-positive cells.IMPORTANCE High-risk human papillomaviruses (HPVs) infect epithelial cells and induce viral genome amplification upon differentiation. HPV proteins activate the ATR DNA damage repair pathway, and this is required for HPV genome amplification. In the present study, we show that HPV-induced ATR activation also leads to suppression of expression of inflammatory response genes. This suppression results from HPV-induced phosphorylation of the autophagosome cargo protein p62 which regulates the levels of the transcription factor GATA4. Activation of p62 in normal fibroblasts results in senescence, but this is not seen in HPV-positive keratinocytes. Importantly, knockdown of p62 or overexpression of GATA4 in HPV-positive cells abrogates viral replication. This study demonstrates that activation of ATR in HPV-positive cells triggers a p62-directed pathway inducing suppression of inflammatory gene expression independent of DNA repair and facilitating HPV replication.


Subject(s)
Alphapapillomavirus/genetics , Alphapapillomavirus/pathogenicity , Ataxia Telangiectasia Mutated Proteins/genetics , Autophagy/genetics , Host-Pathogen Interactions/genetics , RNA-Binding Proteins/genetics , Cell Differentiation , Cell Line , Cells, Cultured , Fibroblasts/virology , GATA4 Transcription Factor/genetics , Humans , Inflammation/genetics , Inflammation/virology , Keratinocytes/virology , Male , Papillomavirus Infections/virology , Phosphorylation , Signal Transduction , Virus Replication
9.
Viruses ; 12(7)2020 07 10.
Article in English | MEDLINE | ID: mdl-32664381

ABSTRACT

Human papillomaviruses are the causative agents of cervical and other anogenital cancers along with approximately 60% of oropharyngeal cancers. These small double-stranded DNA viruses infect stratified epithelia and link their productive life cycles to differentiation. HPV proteins target cellular factors, such as those involved in DNA damage repair, as well as epigenetic control of host and viral transcription to regulate the productive life cycle. HPVs constitutively activate the ATM and ATR DNA repair pathways and preferentially recruit these proteins to viral genomes to facilitate productive viral replication. In addition, the sirtuin deacetylases along with histone acetyltransferases, including Tip60, are targeted in HPV infections to regulate viral transcription and replication. These pathways provide potential targets for drug therapy to treat HPV-induced disease.


Subject(s)
Alphapapillomavirus/physiology , DNA Damage/genetics , DNA Repair/genetics , Epigenesis, Genetic/genetics , Alphapapillomavirus/genetics , Humans , Papillomavirus Infections/virology , Virus Replication
10.
Virology ; 547: 27-34, 2020 08.
Article in English | MEDLINE | ID: mdl-32560902

ABSTRACT

The mechanisms regulating viral pathogenesis of human papillomavirus (HPV) associated oropharyngeal squamous cell cancers (OPSCC) are not well understood. In the cervix, activation of DNA damage repair pathways is critical for viral replication but little is known about their role in OPSCC. APOBEC factors have been shown to be increased in OPSCC but the significance of this is unclear. We therefore examined activation of DNA damage and APOBEC factors in HPV-induced OPSCC. Our studies show significantly increased levels of pCHK1, FANCD2, BRCA1, RAD51, pSMC1 and γH2AX foci in HPV-positive samples as compared to HPV-negative while the ATM effector kinase, pCHK2, was not increased. Similar differences were observed when the levels of proteins were examined in OPSCC cell lines. In contrast, the levels of APOBEC3B and 3A were found to be similar in both HPV-positive and -negative OPSCC. Our studies suggest members of ATR pathway and FANCD2 may be important in HPV-induced OPSCC.


Subject(s)
Neoplasms, Squamous Cell/metabolism , Oropharyngeal Neoplasms/metabolism , Papillomaviridae/physiology , Papillomavirus Infections/metabolism , ATPases Associated with Diverse Cellular Activities/genetics , ATPases Associated with Diverse Cellular Activities/metabolism , Acid Anhydride Hydrolases/genetics , Acid Anhydride Hydrolases/metabolism , Checkpoint Kinase 1/genetics , Checkpoint Kinase 1/metabolism , Cytidine Deaminase/genetics , Cytidine Deaminase/metabolism , DNA Repair , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Fanconi Anemia Complementation Group D2 Protein/genetics , Fanconi Anemia Complementation Group D2 Protein/metabolism , Humans , Minor Histocompatibility Antigens/genetics , Minor Histocompatibility Antigens/metabolism , Neoplasms, Squamous Cell/genetics , Neoplasms, Squamous Cell/virology , Oropharyngeal Neoplasms/genetics , Oropharyngeal Neoplasms/virology , Papillomaviridae/genetics , Papillomavirus Infections/genetics , Papillomavirus Infections/virology
11.
Nat Commun ; 10(1): 2300, 2019 05 24.
Article in English | MEDLINE | ID: mdl-31127091

ABSTRACT

Single-stranded circular RNAs (circRNAs), generated through 'backsplicing', occur more extensively than initially anticipated. The possible functions of the vast majority of circRNAs remain unknown. Virus-derived circRNAs have recently been described in gamma-herpesviruses. We report that oncogenic human papillomaviruses (HPVs) generate circRNAs, some of which encompass the E7 oncogene (circE7). HPV16 circE7 is detectable by both inverse RT-PCR and northern blotting of HPV16-transformed cells. CircE7 is N6-methyladenosine (m6A) modified, preferentially localized to the cytoplasm, associated with polysomes, and translated to produce E7 oncoprotein. Specific disruption of circE7 in CaSki cervical carcinoma cells reduces E7 protein levels and inhibits cancer cell growth both in vitro and in tumor xenografts. CircE7 is present in TCGA RNA-Seq data from HPV-positive cancers and in cell lines with only episomal HPVs. These results provide evidence that virus-derived, protein-encoding circular RNAs are biologically functional and linked to the transforming properties of some HPV.


Subject(s)
Cell Transformation, Neoplastic/pathology , Host-Pathogen Interactions/genetics , RNA, Viral/metabolism , RNA/metabolism , Uterine Cervical Neoplasms/virology , Animals , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Datasets as Topic , Female , Gene Knockdown Techniques , Human papillomavirus 16/genetics , Human papillomavirus 16/pathogenicity , Humans , Mice , Mice, Inbred NOD , Papillomavirus E7 Proteins/genetics , Polyribosomes/genetics , Polyribosomes/metabolism , RNA/genetics , RNA/isolation & purification , RNA, Circular , RNA, Small Interfering/metabolism , RNA, Viral/genetics , RNA, Viral/isolation & purification , Sequence Analysis, RNA , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/pathology , Xenograft Model Antitumor Assays
12.
Oncogene ; 38(17): 3274-3287, 2019 04.
Article in English | MEDLINE | ID: mdl-30631149

ABSTRACT

High-risk human papillomaviruses (HPVs) constitutively activate ataxia telangiectasia mutated (ATM) and ataxia telangiectasia- and Rad3-related (ATR) DNA damage repair pathways for viral genome amplification. HPVs activate these pathways through the immune regulator STAT-5. For the ATR pathway, STAT-5 increases expression of the topoisomerase IIß-binding protein 1 (TopBP1), a scaffold protein that binds ATR and recruits it to sites of DNA damage. TopBP1 also acts as a transcriptional regulator, and we investigated how this activity influenced the HPV life cycle. We determined that TopBP1 levels are increased in cervical intraepithelial neoplasias as well as cervical carcinomas, consistent with studies in HPV-positive cell lines. Suppression of TopBP1 by shRNAs impairs HPV genome amplification and activation of the ATR pathway but does not affect the total levels of ATR and CHK1. In contrast, knockdown reduces the expression of other DNA damage factors such as RAD51 and Mre11 but not BRCA2 or NBS1. Interestingly, TopBP1 positively regulates the expression of E2F1, a TopBP1-binding partner, and p73 in HPV-positive cells in contrast to its effects in other cell types. TopBP1 transcriptional activity is regulated by AKT, and treatment with AKT inhibitors suppresses expression of E2F1 and p73 without interfering with ATR signaling. Importantly, the levels of p73 are elevated in HPV-positive cells and its knockdown impairs HPV genome amplification. This demonstrates that p73, like p63 and p53, is an important regulator of the HPV life cycle that is controlled by the transcriptional activating properties of the multifunctional TopBP1 protein.


Subject(s)
Carrier Proteins/genetics , Cell Differentiation/genetics , DNA-Binding Proteins/genetics , E2F1 Transcription Factor/genetics , Epithelial Cells/pathology , Gene Amplification/genetics , Nuclear Proteins/genetics , Papillomavirus Infections/genetics , Tumor Protein p73/genetics , Animals , Ataxia Telangiectasia Mutated Proteins/genetics , Cell Line , Checkpoint Kinase 1/genetics , DNA Damage/genetics , Female , Gene Expression Regulation/genetics , Humans , MRE11 Homologue Protein/genetics , Mice , NIH 3T3 Cells , Papillomaviridae/pathogenicity , Rad51 Recombinase/genetics , STAT5 Transcription Factor/genetics , Signal Transduction/genetics , Transcription, Genetic/genetics , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/virology , Uterine Cervical Dysplasia/genetics , Uterine Cervical Dysplasia/virology
13.
Int J Gynecol Pathol ; 38(1): 1-10, 2019 Jan.
Article in English | MEDLINE | ID: mdl-29995652

ABSTRACT

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.


Subject(s)
Papillomaviridae/isolation & purification , Papillomavirus Infections/diagnosis , Uterine Cervical Dysplasia/diagnosis , Uterine Cervical Neoplasms/diagnosis , Cell Differentiation , Cervix Uteri/metabolism , Cervix Uteri/pathology , Cervix Uteri/virology , DNA Damage , DNA Repair , Disease Progression , Female , Genotype , Humans , Immunohistochemistry , Papillomaviridae/genetics , Papillomaviridae/physiology , Papillomavirus Infections/metabolism , Papillomavirus Infections/pathology , Papillomavirus Infections/virology , Sensitivity and Specificity , Uterine Cervical Neoplasms/metabolism , Uterine Cervical Neoplasms/pathology , Uterine Cervical Neoplasms/virology , Virus Replication , Uterine Cervical Dysplasia/metabolism , Uterine Cervical Dysplasia/pathology , Uterine Cervical Dysplasia/virology
14.
mBio ; 9(1)2018 02 13.
Article in English | MEDLINE | ID: mdl-29440569

ABSTRACT

High-risk human papillomaviruses (HPVs) activate the ataxia telangiectasia mutated-dependent (ATM) DNA damage response as well as the ataxia telangiectasia mutated-dependent DNA-related (ATR) pathway in the absence of external DNA damaging agents for differentiation-dependent genome amplification. Through the use of comet assays and pulsed-field gel electrophoresis, our studies showed that these pathways are activated in response to DNA breaks induced by the viral proteins E6 and E7 alone and independently of viral replication. The majority of these virally induced DNA breaks are present in cellular DNAs and only minimally in HPV episomes. Treatment of HPV-positive cells with inhibitors of both ATM and ATR leads to the generation of DNA breaks and the fragmentation of viral episomes, indicating that DNA breaks are introduced into HPV genomes. These breaks, however, are rapidly repaired through the preferential recruitment of homologous recombination repair enzymes, such as RAD51 and BRCA1, to viral genomes at the expense of cellular DNAs. When HPV-positive cells are treated with hydroxyurea, this recruitment of RAD51 and BRCA1 to viral genomes is greatly enhanced with little recruitment to damaged cellular DNAs and with retention of the ability of viral genomes to amplify. Overall, our studies demonstrated that human papillomaviruses induce breaks into cellular and viral DNAs and that the preferential repair of these lesions in viral episomes leads to genome amplification.IMPORTANCE High-risk human papillomaviruses (HPVs) are the etiologic agents of cervical cancer and are linked to the development of many other anogenital and oropharyngeal cancers. Replication of high-risk HPVs requires the activation of the ataxia telangiectasia-mutated (ATM) and ATM- and Rad3-related (ATR) DNA repair pathways. Our studies have shown that HPVs activate these pathways by inducing double-strand breaks primarily in cellular DNAs and minimally in viral genomes. Breaks are induced in viral genomes but are rapidly repaired through the preferential recruitment of homologous repair factors such as RAD51 and BRCA1 to HPV episomes. The preferential repair of breaks in viral genomes leads to amplification. Our study identified a novel mechanism by which human papillomaviruses manipulate DNA repair pathways to productively replicate viral genomes. The induction of genetic instability in cellular DNAs likely contributes to the generation of mutations that lead to the development of cancers.


Subject(s)
DNA Breaks , DNA Repair Enzymes/metabolism , DNA Repair , DNA, Viral/metabolism , Host-Pathogen Interactions , Papillomaviridae/physiology , Virus Replication , Cells, Cultured , Comet Assay , Electrophoresis, Gel, Pulsed-Field , Humans , Keratinocytes/virology , Oncogene Proteins, Viral/metabolism , Protein Binding , Time Factors
15.
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
16.
Viruses ; 9(8)2017 08 18.
Article in English | MEDLINE | ID: mdl-28820495

ABSTRACT

High-risk human papillomaviruses (HPVs) are the causative agents of cervical and other genital cancers. In addition, HPV infections are associated with the development of many oropharyngeal cancers. HPVs activate and repress a number of host cellular pathways to promote their viral life cycles, including those of the DNA damage response. High-risk HPVs activate the ataxia telangiectasia-mutated (ATM) and ATM and Rad3-related (ATR) DNA damage repair pathways, which are essential for viral replication (particularly differentiation-dependent genome amplification). These DNA repair pathways are critical in maintaining host genomic integrity and stability and are often dysregulated or mutated in human cancers. Understanding how these pathways contribute to HPV replication and transformation may lead to the identification of new therapeutic targets for the treatment of existing HPV infections.


Subject(s)
DNA Damage , Papillomaviridae/physiology , Papillomavirus Infections/genetics , Papillomavirus Infections/virology , Virus Replication , Animals , DNA Repair , Host-Pathogen Interactions , Humans , Papillomaviridae/genetics
17.
mBio ; 8(1)2017 02 14.
Article in English | MEDLINE | ID: mdl-28196964

ABSTRACT

The life cycle of human papillomavirus (HPV) is dependent on the differentiation state of its host cell. HPV genomes are maintained as low-copy episomes in basal epithelial cells and amplified to thousands of copies per cell in differentiated layers. Replication of high-risk HPVs requires the activation of the ataxia telangiectasia-mutated (ATM) and ATM and Rad3-related (ATR) DNA repair pathways. The Fanconi anemia (FA) pathway is a part of the DNA damage response and mediates cross talk between the ATM and ATR pathways. Our studies show that HPV activates the FA pathway, leading to the accumulation of a key regulatory protein, FANCD2, in large nuclear foci. These HPV-dependent foci colocalize with a distinct population of DNA repair proteins, including ATM components γH2AX and BRCA1, but infrequently with p-SMC1, which is required for viral genome amplification in differentiated cells. Furthermore, FANCD2 is found at viral replication foci, where it is preferentially recruited to viral genomes compared to cellular chromosomes and is required for maintenance of HPV episomes in undifferentiated cells. These findings identify FANCD2 as an important regulator of HPV replication and provide insight into the role of the DNA damage response in the differentiation-dependent life cycle of HPV.IMPORTANCE High-risk human papillomaviruses (HPVs) are the etiological agents of cervical cancer and are linked to the development of many other anogenital and oropharyngeal cancers. Identification of host cellular pathways involved in regulating the viral life cycle may be helpful in identifying treatments for HPV lesions. Mutations in genes of the Fanconi anemia (FA) DNA repair pathway lead to genomic instability in patients and a predisposition to HPV-associated malignancies. Our studies demonstrate that FA pathway component FANCD2 is recruited to HPV DNA, associates with members of the ATM DNA repair pathway, and is essential for the maintenance of viral episomes in basal epithelial cells. Disruption of the FA pathway may result in increased integration events and a higher incidence of HPV-related cancer. Our study identifies new links between HPV and the FA pathway that may help to identify new therapeutic targets for the treatment of existing HPV infections and cancers.


Subject(s)
Fanconi Anemia Complementation Group D2 Protein/metabolism , Gene Expression Regulation , Genome, Viral , Papillomaviridae/physiology , Papillomavirus Infections/genetics , Virus Replication , ATPases Associated with Diverse Cellular Activities , Adenosine Triphosphatases/metabolism , BRCA1 Protein/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Differentiation , Cell Line , DNA Repair , Fanconi Anemia/metabolism , Histones/metabolism , Host-Pathogen Interactions/genetics , Humans , Papillomaviridae/genetics , Papillomavirus E7 Proteins/genetics , Papillomavirus Infections/virology , Virus Replication/physiology
18.
Virus Res ; 231: 34-40, 2017 03 02.
Article in English | MEDLINE | ID: mdl-27826042

ABSTRACT

The innate immune response constitutes the first line of defense against infections by pathogens. Successful pathogens such as human papillomaviruses (HPVs) have evolved mechanisms that target several points in these pathways including sensing of viral genomes, blocking the synthesis of interferons and inhibiting the action of JAK/STAT transcription factors. Disruption of these inhibitory mechanisms contributes to the ability of HPVs to establish persistent infections, which is the major etiological factor in the development of anogenital cancers. Interestingly, HPVs also positively activate several members of these pathways such as STAT-5 that are important for their differentiation-dependent life cycle. STAT-5 activation induces the ATM and ATR DNA damage response pathways that play critical roles in HPV genome amplification. Targeting of these pathways by pharmaceuticals can provide novel opportunities to inhibit infections by these important human pathogens.


Subject(s)
Genome, Viral , Immune Evasion , Keratinocytes/immunology , NF-kappa B/genetics , Oncogene Proteins, Viral/genetics , Papillomaviridae/genetics , Papillomavirus Infections/virology , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/immunology , Gene Expression Regulation , Humans , Immunity, Innate , Interferons/genetics , Interferons/immunology , Janus Kinases/genetics , Janus Kinases/immunology , Keratinocytes/virology , NF-kappa B/immunology , Oncogene Proteins, Viral/immunology , Papillomaviridae/growth & development , Papillomaviridae/pathogenicity , Papillomavirus Infections/genetics , Papillomavirus Infections/immunology , Papillomavirus Infections/pathology , STAT5 Transcription Factor/genetics , STAT5 Transcription Factor/immunology , Signal Transduction , Virus Replication
19.
Trends Cancer ; 2(5): 234-240, 2016 May.
Article in English | MEDLINE | ID: mdl-27617309

ABSTRACT

Human papillomaviruses are the causative agents of cervical, anal as well as many oropharyngeal cancers. While prophylactic vaccines have been developed, uptake is low in the US and other Western countries, and access is limited in less developed countries. A number of areas are emerging as critical for future study. These include investigation of the mechanisms regulating infection and progression to cancer at both cervical and oropharyngeal sites as these appear to be distinct. HPV-induced cancers also may be susceptible to immune therapy, revealing opportunities for treating advanced cervical disease and reducing the morbidity of treatments for oropharyngeal cancers. We believe these areas are critical focal points for HPV cancer research in the next decade.

20.
PLoS Pathog ; 12(7): e1005747, 2016 07.
Article in English | MEDLINE | ID: mdl-27386862

ABSTRACT

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.


Subject(s)
Gene Expression Regulation/physiology , Keratinocytes/virology , Kruppel-Like Transcription Factors/metabolism , Papillomaviridae/physiology , Virus Replication/physiology , Cell Differentiation , Cells, Cultured , Chromatin Immunoprecipitation , Fluorescent Antibody Technique , Host-Pathogen Interactions/physiology , Humans , Immunoblotting , Kruppel-Like Factor 4 , Life Cycle Stages , Mutagenesis, Site-Directed , Oligonucleotide Array Sequence Analysis , Papillomavirus Infections , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...