Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
J Med Virol ; 96(8): e29815, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39073137

ABSTRACT

Human papillomaviruses (HPVs) are non-enveloped double-stranded DNA viruses. When HPV infection persists, infected tissues can develop many HPV-related diseases such as cervical cancer and head and neck squamous cell carcinoma. To establish their persistent infection, HPVs have evolved mechanisms to manipulate the host cellular processes such as DNA damage response (DDR), which includes homologous recombination, nonhomologous end joining, and microhomology-mediated end joining. Additionally, HPVs utilize host inflammatory processes to facilitate their life cycles. Here, we bridge the concepts of DDR and inflammatory response, and discuss how HPV proteins orchestrate a sophisticated manipulation of DDR and inflammation to promote their viral replication, ultimately fostering the progression of infected cells towards oncogenic transformation to malignancy.


Subject(s)
DNA Damage , Inflammation , Papillomaviridae , Papillomavirus Infections , Humans , Papillomavirus Infections/virology , Inflammation/virology , Papillomaviridae/genetics , Papillomaviridae/pathogenicity , Host-Pathogen Interactions , DNA Repair , Virus Replication , Cell Transformation, Neoplastic
2.
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
3.
J Virol ; 89(8): 4668-75, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25673709

ABSTRACT

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.


Subject(s)
Alphapapillomavirus/physiology , Cell Differentiation/physiology , Epithelial Cells/virology , Glycogen Synthase Kinase 3/metabolism , Histone Acetyltransferases/metabolism , STAT5 Transcription Factor/metabolism , Acetylation , Alphapapillomavirus/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , Blotting, Northern , Blotting, Southern , Blotting, Western , Cells, Cultured , DNA Primers/genetics , Epithelial Cells/physiology , Gene Knockdown Techniques , Glycogen Synthase Kinase 3 beta , Histone Acetyltransferases/genetics , Humans , Lysine Acetyltransferase 5 , Phosphorylation , Plasmids/genetics , RNA, Small Interfering/genetics , Reverse Transcriptase Polymerase Chain Reaction
4.
PLoS Pathog ; 9(4): e1003295, 2013.
Article in English | MEDLINE | ID: mdl-23593005

ABSTRACT

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.


Subject(s)
Alphapapillomavirus/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , DNA Repair/genetics , Papillomavirus E7 Proteins/metabolism , STAT5 Transcription Factor/metabolism , Tumor Suppressor Proteins/metabolism , Alphapapillomavirus/physiology , Cell Differentiation , Cells, Cultured , DNA Damage/genetics , Epithelial Cells/metabolism , Humans , Keratinocytes/metabolism , PPAR gamma/metabolism , Papillomavirus E7 Proteins/genetics , Phosphorylation , Pimozide/pharmacology , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA Interference , STAT5 Transcription Factor/genetics , Tumor Suppressor Proteins/genetics , Virus Replication
5.
Front Pharmacol ; 15: 1450875, 2024.
Article in English | MEDLINE | ID: mdl-39156107

ABSTRACT

Background: Cervical cancer (CC) stands as a significant health threat to women globally, with high-risk human papillomaviruses as major etiologic agents. The DNA damage repair (DDR) protein topoisomerase I (TOP1) has been linked to various cancers, yet its distinct roles and mechanisms in CC are not fully elucidated. Methods: We investigated TOP1 expression in cervical intraepithelial neoplasia (CIN) and CC tissues utilizing qRT-PCR and IHC, correlating findings with patient prognosis. Subsequent knockdown studies were performed in vitro and in vivo to evaluate the influence of TOP1 on tumor growth, DNA repair, and inflammatory responses. Results: TOP1 was highly expressed in CIN and CC, negatively correlating with patient prognosis. Inhibition of TOP1 impeded CC cell growth and disrupted DNA repair. TOP1 was shown to regulate tumor-promoting inflammation and programmed death-ligand 1 (PD-L1) production in a cGAS-dependent manner. HPV oncoproteins E6 and E7 upregulated TOP1 and activated the cGAS-PD-L1 pathway. Conclusions: TOP1 acts as a DNA repair mediator, promoting CC development and immune evasion. Targeting the TOP1-cGAS-PD-L1 axis could be a potential therapeutic strategy for CC.

6.
J Virol ; 85(18): 9486-94, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21734056

ABSTRACT

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.


Subject(s)
Host-Pathogen Interactions , Keratinocytes/virology , Oncogene Proteins, Viral/metabolism , Papillomaviridae/pathogenicity , Papillomavirus E7 Proteins/metabolism , Repressor Proteins/metabolism , STAT1 Transcription Factor/antagonists & inhibitors , Virus Replication , Cells, Cultured , Humans , Immune Evasion , Keratinocytes/immunology , Papillomaviridae/immunology , Plasmids/metabolism , STAT1 Transcription Factor/immunology , Transcription, Genetic , Ubiquitin-Protein Ligases/metabolism
7.
Cell Death Dis ; 13(4): 342, 2022 04 13.
Article in English | MEDLINE | ID: mdl-35418185

ABSTRACT

High grade serous ovarian cancer (HGSOC) is the most aggressive subtype of ovarian cancer and HGSOC patients often appear with metastasis, leading to the poor prognosis. Up to date, the extrachromosomal circular DNAs (eccDNAs) have been shown to be involved in cancer genome remodeling but the roles of eccDNAs in metastatic HGSOC are still not clear. Here we explored eccDNA profiles in HGSOC by Circle-Sequencing analysis using four pairs of primary and metastatic tissues of HGSOC patients. Within the differentially expressed eccDNAs screened out by our analysis, eight candidates were validated by outward PCR and qRT-PCR analysis. Among them, DNMT1circle10302690-10302961 was further confirmed by FISH assay and BaseScope assay, as the most significantly down-regulated eccDNA in metastatic tumors of HGSOC. Lower expression of DNMT1circle10302690-10302961 in both primary and metastatic tumors was associated with worse prognosis of HGSOC. Taken together, our finding firstly demonstrated the eccDNAs landscape of primary and metastatic tissues of HGSOC. The eccDNA DNMT1circle10302690-10302961 can be considered as a potential biomarker or a therapeutically clinical target of HGSOC metastasis and prognosis.


Subject(s)
Ovarian Neoplasms , Carcinoma, Ovarian Epithelial , DNA , DNA, Circular/genetics , Female , Humans , Ovarian Neoplasms/pathology
8.
J Immunol ; 183(1): 578-92, 2009 Jul 01.
Article in English | MEDLINE | ID: mdl-19542469

ABSTRACT

Extracellular NAD induces the ATP-independent activation of the ionotropic P2X(7) purinergic receptor (P2X(7)R) in murine T lymphocytes via a novel covalent pathway involving ADP-ribosylation of arginine residues on the P2X(7)R ectodomain. This modification is catalyzed by ART2.2, a GPI-anchored ADP-ribosyltransferase (ART) that is constitutively expressed in murine T cells. We previously reported that ART2.1, a related ecto-ART, is up-regulated in inflammatory murine macrophages that constitutively express P2X(7)R. Thus, we tested the hypothesis that extracellular NAD acts via ART2.1 to regulate P2X(7)R function in murine macrophages. Coexpression of the cloned murine P2X(7)R with ART2.1 or ART2.2 in HEK293 cells verified that P2X(7)R is an equivalent substrate for ADP-ribosylation by either ART2.1 or ART2.2. However, in contrast with T cells, the stimulation of macrophages or HEK293 cells with NAD alone did not activate the P2X(7)R. Rather, NAD potentiated ATP-dependent P2X(7)R activation as indicated by a left shift in the ATP dose-response relationship. Thus, extracellular NAD regulates the P2X(7)R in both macrophages and T cells but via distinct mechanisms. Although ADP-ribosylation is sufficient to gate a P2X(7)R channel opening in T cells, this P2X(7)R modification in macrophages does not gate the channel but decreases the threshold for gating in response to ATP binding. These findings indicate that extracellular NAD and ATP can act synergistically to regulate P2X(7)R signaling in murine macrophages and also suggest that the cellular context in which P2X(7)R signaling occurs differs between myeloid and lymphoid leukocytes.


Subject(s)
ADP Ribose Transferases/physiology , Macrophages/immunology , NAD/physiology , Receptors, Purinergic P2/metabolism , T-Lymphocytes/immunology , ADP Ribose Transferases/biosynthesis , ADP Ribose Transferases/genetics , Animals , Cell Line , Cells, Cultured , Dose-Response Relationship, Immunologic , Extracellular Space/enzymology , Extracellular Space/immunology , Extracellular Space/metabolism , Humans , Inflammation Mediators/physiology , Macrophages/enzymology , Macrophages/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred NZB , Mice, Knockout , Protein Structure, Tertiary , Receptors, Purinergic P2/biosynthesis , Receptors, Purinergic P2/deficiency , Receptors, Purinergic P2/physiology , Receptors, Purinergic P2X7 , Signal Transduction/genetics , Signal Transduction/immunology , Substrate Specificity/genetics , Substrate Specificity/immunology , T-Lymphocytes/enzymology , T-Lymphocytes/metabolism
9.
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
10.
Cell Death Discov ; 7(1): 259, 2021 Sep 22.
Article in English | MEDLINE | ID: mdl-34552062

ABSTRACT

The poly(adenosine diphosphate-ribose) polymerase (PARP) inhibitors show survival benefits in ovarian cancer patients with BRCA1/2 mutation or homologous recombination (HR) deficiency, but only limited efficacy in HR-proficient ones. Another drug, arsenic trioxide (ATO) or arsenic drug (RIF), exerts antitumor effects via inducing DNA damage. Here, we investigated the combined therapeutic effects of the PARP inhibitors and the arsenic compound in HR-proficient ovarian cancer. The combined treatment of niraparib, olaparib, or fluazolepali with ATO showed a significant suppression in tumor cell viability and colony formation. The drug treatment also induced synergistic inhibition of cell proliferation and DNA damage, and acceleration of cell apoptosis in two HR-proficient ovarian cancer cell lines SKOV3 and CAOV3, either by simultaneous or sequential administration. The mechanism underlying these synergistic effects were reflected by the significantly increased ratio of cleaved-PARP/total PARP and decreased ratio of p-AKT/total AKT. Consistently, the combination of olaparib with RIF synergistically reduced the tumor growth in mouse xenograft models. In conclusion, the arsenic compound greatly sensitizes HR-proficient ovarian cancer cells to the PARP inhibitors, and our findings provide an evidence for the clinical treatment development of this combination in HR-proficient ovarian cancer patients.

11.
Pathogens ; 9(6)2020 Jun 23.
Article in English | MEDLINE | ID: mdl-32585979

ABSTRACT

Human papillomavirus (HPV) infection leads to a variety of benign lesions and malignant tumors such as cervical cancer and head and neck squamous cell carcinoma. Several HPV vaccines have been developed that can help to prevent cervical carcinoma, but these vaccines are only effective in individuals with no prior HPV infection. Thus, it is still important to understand the HPV life cycle and in particular the association of HPV with human pathogenesis. HPV production requires activation of the DNA damage response (DDR), which is a complex signaling network composed of multiple sensors, mediators, transducers, and effectors that safeguard cellular DNAs to maintain the host genome integrity. In this review, we focus on the roles of the ataxia telangiectasia mutant and Rad3-related (ATR) DNA damage response in HPV DNA replication. HPV can induce ATR expression and activate the ATR pathway. Inhibition of the ATR pathway results in suppression of HPV genome maintenance and amplification. The mechanisms underlying this could be through various molecular pathways such as checkpoint signaling and transcriptional regulation. In light of these findings, other downstream mechanisms of the ATR pathway need to be further investigated for better understanding HPV pathogenesis and developing novel ATR DDR-related inhibitors against HPV infection.

12.
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
13.
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
14.
J Zhejiang Univ Sci B ; 18(3): 215-232, 2017 03.
Article in English | MEDLINE | ID: mdl-28271657

ABSTRACT

The DNA damage response (DDR) is activated when DNA is altered by intrinsic or extrinsic agents. This pathway is a complex signaling network and plays important roles in genome stability, tumor transformation, and cell cycle regulation. Human papillomaviruses (HPVs) are the main etiological agents of cervical cancer. Cervical cancer ranks as the fourth most common cancer among women and the second most frequent cause of cancer-related death worldwide. Over 200 types of HPVs have been identified and about one third of these infect the genital tract. The HPV life cycle is associated with epithelial differentiation. Recent studies have shown that HPVs deregulate the DDR to achieve a productive life cycle. In this review, I summarize current findings about how HPVs mediate the ataxia-telangiectasia mutated kinase (ATM) and the ATM-and RAD3-related kinase (ATR) DDRs, and focus on the roles that ATM and ATR signalings play in HPV viral replication. In addition, I demonstrate that the signal transducer and activator of transcription-5 (STAT)-5, an important immune regulator, can promote ATM and ATR activations through different mechanisms. These findings may provide novel opportunities for development of new therapeutic targets for HPV-related cancers.


Subject(s)
DNA Damage , Papillomaviridae/physiology , Papillomavirus Infections/virology , Animals , Ataxia Telangiectasia Mutated Proteins/genetics , Cell Cycle , Cell Cycle Proteins/metabolism , Cell Differentiation , DNA-Binding Proteins/metabolism , Female , Gene Expression Regulation, Viral , Genome, Viral , Humans , Mice , MicroRNAs/metabolism , Papillomaviridae/pathogenicity , Papillomavirus Infections/genetics , STAT5 Transcription Factor/genetics , Signal Transduction , Uterine Cervical Neoplasms/virology , Viral Proteins/metabolism , Virus Replication
15.
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
16.
mBio ; 6(6): e02006-15, 2015 Dec 22.
Article in English | MEDLINE | ID: mdl-26695634

ABSTRACT

UNLABELLED: The life cycle of high-risk human papillomaviruses (HPVs) is dependent upon epithelial differentiation. Following infection of basal cells, HPV genomes are stably maintained at low copy numbers, and productive replication or amplification is restricted to highly differentiated suprabasal cells. In high-risk HPV infections, the ATM pathway is constitutively activated in the absence of external DNA-damaging agents and is required for productive viral replication. The ataxia telangiectasia (ATM) pathway repairs double-strand breaks in DNA, while the ataxia telangiectasia and Rad3-related (ATR) pathway targets single-strand breaks. Our studies show that the ATR pathway, like the ATM pathway, is activated in HPV-positive cells and that inhibitors of ATR or CHK1 phosphorylation block both amplification and late viral gene expression in differentiated cells while moderately reducing stable copy numbers in undifferentiated cells. TopBP1 is a critical upstream activator of the ATR pathway and is expressed at elevated levels in HPV-positive cells. This increased expression of TopBP1 is necessary for ATR/CHK1 activation in HPV-positive cells, and knockdown blocks amplification. Furthermore, TopBP1 activation is shown to be regulated at the level of transcription initiation by the innate immune regulator STAT-5, which is activated by HPV proteins. STAT-5 has also been shown to be a regulator of the ATM response, demonstrating that these two pathways are coordinately regulated in HPV-positive cells. These findings identify a novel link between the innate immune response and activation of the ATR DNA damage response in regulating the life cycle of high-risk HPVs. IMPORTANCE: High-risk human papillomaviruses (HPVs) are the causative agents of cervical and other anogenital cancers, as well as many oral cancers. HPVs infect epithelial cells and restrict productive viral replication or amplification and virion production to differentiated cells. Our studies demonstrate that HPVs activate the ATR single-strand DNA repair pathway and this activation is necessary for HPV genome amplification. The innate immune regulator STAT-5 is shown to regulate transcription of the ATR binding factor TopBP1, and this is critical for the induction of the ATR pathway. Our study identifies important links between innate immune signaling, the ATR DNA damage pathway, and productive HPV replication that may lead to the characterization of new targets for the development of therapeutics to treat HPV-induced infections.


Subject(s)
Carrier Proteins/metabolism , DNA-Binding Proteins/metabolism , Host-Pathogen Interactions , Nuclear Proteins/metabolism , Papillomaviridae/physiology , STAT5 Transcription Factor/metabolism , Viral Proteins/metabolism , Virus Replication , Ataxia Telangiectasia Mutated Proteins/metabolism , Cells, Cultured , Gene Expression Regulation , Gene Regulatory Networks , Humans , Keratinocytes/virology , Transcription, Genetic
17.
Cancer Res ; 74(22): 6682-92, 2014 Nov 15.
Article in English | MEDLINE | ID: mdl-25267065

ABSTRACT

Cervical cancers, a malignancy associated with oncogenic papilloma viruses, remain a major disease burden in the absence of effective implementation of preventive strategies. CD66(+) cells have previously been identified as a tumor-propagating subset in cervical cancers. We investigated the existence, differentiation state, and neoplastic potential of CD66(+) cells in a precancer cell line harboring HPV31b episomes. The gene expression profile of CD66(high) cells overlaps with differentiated keratinocytes, neoplastic mesenchymal transition, cells of the squamocolumnar junction, and cervical cancer cell line-derived spheroids. There is elevated expression of DNMT1, Notch1, and the viral gene product E1⁁E4 in CD66(high) cells. Thus, CD66(high) cells, in the absence of differentiating signals, express higher levels of key regulators of keratinocytes stemness, differentiation, and the viral life cycle, respectively. We also find a striking association of neoplastic traits, including migration, invasion, and colony formation, in soft agar with CD66(high) cells. These properties and a distinct G2-M-enriched cell-cycle profile are conserved in cells from cervical cancers. Principally, using a precancerous cell line, we propose that CD66(high) cells have an intermediate differentiation state, with a cellular milieu connected with both viral replication and neoplastic potential, and validate some key features in precancer lesions. Such pathophysiologically relevant systems for defining cellular changes in the early phases of the disease process provide both mechanistic insight and potential therapeutic strategies. Collectively, our data provide a rationale for exploring novel therapeutic targets in CD66(+) subsets during cancer progression.


Subject(s)
Antigens, CD/analysis , Cell Adhesion Molecules/analysis , Neoplastic Stem Cells/cytology , Precancerous Conditions/pathology , Uterine Cervical Neoplasms/pathology , Cell Differentiation , Cell Line, Tumor , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/analysis , Female , Humans , Membrane Proteins/analysis , Neoplasm Invasiveness , Papillomaviridae/genetics , Precancerous Conditions/virology , Receptor, Notch1/analysis , Uterine Cervical Neoplasms/virology
18.
Future Microbiol ; 8(12): 1547-57, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24266355

ABSTRACT

HPVs are the causative agents of cervical and other anogenital cancers. HPVs infect stratified epithelia and link their productive life cycles to cellular differentiation. Low levels of viral genomes are stably maintained in undifferentiated cells and productive replication or amplification is restricted to differentiated suprabasal cells. Amplification is dependent on the activation of the ATM DNA damage factors that are recruited to viral replication centers and inhibition of this pathway blocks productive replication. The STAT-5 protein appears to play a critical role in mediating activation of the ATM pathway in HPV-positive cells. While HPVs need to activate the DNA damage pathway for replication, cervical cancers contain many genomic alterations suggesting that this pathway is circumvented during progression to malignancy.


Subject(s)
Alphapapillomavirus/physiology , Cell Differentiation , DNA Damage , Papillomavirus Infections/virology , Alphapapillomavirus/genetics , Animals , Humans , Papillomavirus Infections/genetics , Papillomavirus Infections/physiopathology , Virus Replication
19.
Purinergic Signal ; 5(3): 369-83, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19404775

ABSTRACT

ADP-ribosylation of cell surface proteins in mammalian cells is a post-translational modification by which ecto-ADP-ribosyltransferases (ARTs) transfer ADP-ribose from extracellular NAD to protein targets. The ART2 locus at murine chromosome 7 encompasses the tandem Art2a and Art2b genes that encode the distinct ART2.1 and ART2.2 proteins. Although both ecto-enzymes share 80% sequence identity, ART2.1 activity is uniquely regulated by an allosteric disulfide bond that is reducible in the presence of extracellular thiols, such as cysteine and glutathione, that accumulate in hypoxic and ischemic tissues. Previous studies have characterized the expression of ART2.1 and ART2.2 in murine T lymphocytes but not in other major classes of lymphoid and myeloid leukocytes. Here, we describe the expression of ART2.1 activity in a wide range of freshly isolated or tissue-cultured murine myeloid and lymphoid leukocytes. Spleen-derived macrophages, dendritic cells (DC), and B cells constitutively express ART2.1 as their predominant ART while spleen T cells express both ART2.1 and the thiol-independent ART2.2 isoform. Although bone-marrow-derived macrophages (BMDM) and dendritic cells (BMDC) constitutively express ART2.1 at low levels, it is markedly up-regulated when these cells are stimulated in vitro with IFNbeta or IFNgamma. ART2.1 expression and activity in splenic B cells is modestly up-regulated during incubation in vitro for 24 h, a condition that promotes B cell apoptosis. This increase in ART2.1 is attenuated by IL-4 (a B cell survival factor), but is not affected by IFNbeta/gamma, suggesting a possible induction of ART2.1 as an ancillary response to B cell apoptosis. In contrast, ART2.1 and ART2.2, which are highly expressed in freshly isolated splenic T cells, are markedly down-regulated when purified T cells are incubated in vitro for 12-24 h. Studies with the BW5147 mouse thymocyte line verified basal expression of ART2.1 and ART2.2, as in primary spleen T cells, and demonstrated that both isoforms can be up-regulated when T cells are maintained in the presence of IFNs. Comparison of the surface proteins which are ADP-ribosylated by ART2.1 in the different leukocyte subtypes indicated both shared and cell-specific proteins as ART2.1 substrates. The LFA-1 integrin, a major target for ART2.2 in T cells, is also ADP-ribosylated by the ART2.1 expressed in macrophages. Thus, ART2.1, in contrast to ART2.2, is expressed in a broad range of myeloid and lymphoid leukocytes. The thiol redox-sensitive nature of this ecto-enzyme suggests an involvement in purinergic signaling that occurs in the combined context of inflammation and hypoxia/ischemia.

20.
J Immunol ; 179(9): 6215-27, 2007 Nov 01.
Article in English | MEDLINE | ID: mdl-17947697

ABSTRACT

Nicotinamide adenosine dinucleotide (NAD) can act as a modulator of multiple immune and inflammatory responses when released into extracellular compartments. These actions of extracellular NAD are largely mediated by a family of mammalian ecto-ADP-ribosyltransferases (ARTs) that covalently modify target extracellular or cell surface proteins by transferring ADP-ribose to arginine or cysteine residues. In this study, we report that bone marrow-derived macrophages (BMDM) from BALB/c mice lack constitutive expression of any of the six murine ecto-ART subtypes, but selectively up-regulate ART2.1 in response to multiple proinflammatory mediators including agonists for TLR and type I and type II IFN. Stimulation of BMDM with LPS, IFN-beta, or IFN-gamma induced high expression of ART2.1, but not ART2.2, as a GPI-anchored cell surface ectoenzyme. ART2.1 expression in response to LPS was potentiated by inhibition of ERK1/2 signaling, but inhibited by blockade of the NF-kappaB, PI3K, and JAK-STAT pathways or the presence of neutralizing anti-IFN-beta. The catalytic function of the induced cell surface ART2.1 was strictly dependent on the presence of extracellular thiol-reducing cofactors, suggesting that in vivo activity of ART2.1-expressing macrophages may be potentiated in hypoxic or ischemic compartments. Consistent with the mutated art2a gene in C57BL/6 mice, LPS- or IFN-stimulated BMDM from this strain lacked expression of cell surface ART2 activity in the presence or absence of extracellular thiol reductants. Collectively, these studies identify ART2.1 as a new candidate for linking autocrine/paracrine activation of inflammatory macrophages to the release of NAD, a critical intracellular metabolite.


Subject(s)
ADP Ribose Transferases/metabolism , Interferon-beta/pharmacology , Interferon-gamma/pharmacology , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Macrophages/enzymology , Sulfhydryl Compounds/metabolism , ADP Ribose Transferases/chemistry , ADP Ribose Transferases/genetics , Animals , Bone Marrow Cells/cytology , Cell Differentiation , Cell Membrane/drug effects , Cell Membrane/enzymology , Cells, Cultured , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Regulation, Enzymologic , Inflammation/enzymology , Inflammation/genetics , Isoenzymes/genetics , Isoenzymes/metabolism , Macrophages/cytology , Mice , Models, Molecular , NF-kappa B/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein Structure, Tertiary , Rats , Sensitivity and Specificity , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL