Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Mol Cell ; 54(3): 445-59, 2014 May 08.
Article in English | MEDLINE | ID: mdl-24746700

ABSTRACT

Mutations within BRCA1 predispose carriers to a high risk of breast and ovarian cancers. BRCA1 functions to maintain genomic stability through the assembly of multiple protein complexes involved in DNA repair, cell-cycle arrest, and transcriptional regulation. Here, we report the identification of a DNA damage-induced BRCA1 protein complex containing BCLAF1 and other key components of the mRNA-splicing machinery. In response to DNA damage, this complex regulates pre-mRNA splicing of a number of genes involved in DNA damage signaling and repair, thereby promoting the stability of these transcripts/proteins. Further, we show that abrogation of this complex results in sensitivity to DNA damage, defective DNA repair, and genomic instability. Interestingly, mutations in a number of proteins found within this complex have been identified in numerous cancer types. These data suggest that regulation of splicing by the BRCA1-mRNA splicing complex plays an important role in the cellular response to DNA damage.


Subject(s)
BRCA1 Protein/metabolism , DNA Repair , Genomic Instability , RNA, Messenger/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Basic-Leucine Zipper Transcription Factors/genetics , Basic-Leucine Zipper Transcription Factors/metabolism , Cell Survival/radiation effects , DNA Damage , DNA Repair Enzymes/genetics , DNA Repair Enzymes/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Exodeoxyribonucleases/genetics , Exodeoxyribonucleases/metabolism , Fanconi Anemia Complementation Group Proteins/genetics , Fanconi Anemia Complementation Group Proteins/metabolism , Genome, Human , HEK293 Cells , Humans , Phosphorylation , Protein Processing, Post-Translational , RNA Splicing , Radiation Tolerance , Repressor Proteins/metabolism , Tumor Suppressor Proteins/metabolism
2.
Br J Cancer ; 120(8): 827-833, 2019 04.
Article in English | MEDLINE | ID: mdl-30890775

ABSTRACT

BACKGROUND: TNM8 staging for oropharyngeal squamous cell carcinomas (OPSCC) surrogates p16 immunohistochemistry for HPV testing. Patients with p16+ OPSCC may lack HPV aetiology. Here, we evaluate the suitability of TNM8 staging for guiding prognosis in such patients. METHODS: HPV status was ascertained using p16 immunohistochemistry and high-risk HPV RNA and DNA in situ hybridisation. Survival by stage in a cohort of OPSCC patients was evaluated using TNM7/TNM8 staging. Survival of p16+/HPV- patients was compared to p16 status. RESULTS: TNM8 staging was found to improve on TNM7 (log rank p = 0·0190 for TNM8 compared with p = 0·0530 for TNM7) in p16+ patients. Patients who tested p16+ but were HPV- (n = 20) had significantly reduced five-year survival (33%) compared to p16+ patients (77%) but not p16- patients (35%). Cancer stage was reduced in 95% of p16+/HPV- patients despite having a mortality rate twice (HR 2.66 [95% CI: 1.37-5.15]) that of p16+/HPV+ patients under new TNM8 staging criteria. CONCLUSION: Given the significantly poorer survival of p16+/HPV- OPSCCs, these data provide compelling evidence for use of an HPV-specific test for staging classification. This has particular relevance in light of potential treatment de-escalation that could expose these patients to inappropriately reduced treatment intensity as treatment algorithms evolve.


Subject(s)
Oropharyngeal Neoplasms/genetics , Papillomavirus Infections/genetics , Viral Proteins/genetics , Adolescent , Adult , Child , Child, Preschool , Disease-Free Survival , Female , Gene Expression Regulation, Neoplastic/genetics , Humans , Infant , Infant, Newborn , Male , Middle Aged , Neoplasm Staging , Oropharyngeal Neoplasms/mortality , Oropharyngeal Neoplasms/pathology , Oropharyngeal Neoplasms/virology , Papillomavirus Infections/pathology , Papillomavirus Infections/virology , Young Adult
3.
PLoS Pathog ; 11(6): e1004988, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26107517

ABSTRACT

Cervical cancer is a multi-stage disease caused by human papillomaviruses (HPV) infection of cervical epithelial cells, but the mechanisms regulating disease progression are not clearly defined. Using 3-dimensional organotypic cultures, we demonstrate that HPV16 E6 and E7 proteins alter the secretome of primary human keratinocytes resulting in local epithelial invasion. Mechanistically, absence of the IGF-binding protein 2 (IGFBP2) caused increases in IGFI/II signalling and through crosstalk with KGF/FGFR2b/AKT, cell invasion. Repression of IGFBP2 is mediated by histone deacetylation at the IGFBP2 promoter and was reversed by treatment with histone deacetylase (HDAC) inhibitors. Our in vitro findings were confirmed in 50 invasive cancers and 79 cervical intra-epithelial neoplastic lesions caused by HPV16 infection, where IGFBP2 levels were reduced with increasing disease severity. In summary, the loss of IGFBP2 is associated with progression of premalignant disease, and sensitises cells to pro-invasive IGF signalling, and together with stromal derived factors promotes epithelial invasion.


Subject(s)
Epithelial Cells/metabolism , Human papillomavirus 16 , Insulin-Like Growth Factor Binding Protein 2/metabolism , Cells, Cultured , Down-Regulation , Female , Human papillomavirus 16/genetics , Humans , Oncogene Proteins, Viral/metabolism , Papillomavirus E7 Proteins/genetics , Repressor Proteins/metabolism , Transcription, Genetic/genetics , Uterine Cervical Neoplasms/metabolism , Uterine Cervical Neoplasms/pathology , Uterine Cervical Neoplasms/virology
4.
EMBO J ; 31(14): 3092-103, 2012 May 29.
Article in English | MEDLINE | ID: mdl-22643222

ABSTRACT

Stromal-derived growth factors are required for normal epithelial growth but are also implicated in tumour progression. We have observed inactivation of the retinoblastoma protein (Rb), through phosphorylation, in cancer-associated fibroblasts in oro-pharyngeal cancer specimens. Rb is well known for its cell-autonomous effects on cancer initiation and progression; however, cell non-autonomous functions of Rb are not well described. We have identified a cell non-autonomous role of Rb, using three-dimensional cultures, where depletion of Rb in stromal fibroblasts enhances invasive potential of transformed epithelia. In part, this is mediated by upregulation of keratinocyte growth factor (KGF), which is produced by the depleted fibroblasts. KGF drives invasion of epithelial cells through induction of MMP1 expression in an AKT- and Ets2-dependent manner. Our data identify that stromal fibroblasts can alter the invasive behaviour of the epithelium, and we show that altered expression of KGF can mediate these functions.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Epithelial Cells/metabolism , Fibroblasts/metabolism , Retinoblastoma Protein/metabolism , Cell Line, Transformed , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Epithelial Cells/pathology , Fibroblast Growth Factor 7/genetics , Fibroblast Growth Factor 7/metabolism , Fibroblasts/pathology , Humans , Matrix Metalloproteinase 1/genetics , Matrix Metalloproteinase 1/metabolism , Proto-Oncogene Protein c-ets-2/genetics , Proto-Oncogene Protein c-ets-2/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Retinoblastoma Protein/genetics
5.
Nucleic Acids Res ; 42(10): 6270-85, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24823795

ABSTRACT

In response to genotoxic stress the TP53 tumour suppressor activates target gene expression to induce cell cycle arrest or apoptosis depending on the extent of DNA damage. These canonical activities can be repressed by TP63 in normal stratifying epithelia to maintain proliferative capacity or drive proliferation of squamous cell carcinomas, where TP63 is frequently overexpressed/amplified. Here we use ChIP-sequencing, integrated with microarray analysis, to define the genome-wide interplay between TP53 and TP63 in response to genotoxic stress in normal cells. We reveal that TP53 and TP63 bind to overlapping, but distinct cistromes of sites through utilization of distinctive consensus motifs and that TP53 is constitutively bound to a number of sites. We demonstrate that cisplatin and adriamycin elicit distinct effects on TP53 and TP63 binding events, through which TP53 can induce or repress transcription of an extensive network of genes by direct binding and/or modulation of TP63 activity. Collectively, this results in a global TP53-dependent repression of cell cycle progression, mitosis and DNA damage repair concomitant with activation of anti-proliferative and pro-apoptotic canonical target genes. Further analyses reveal that in the absence of genotoxic stress TP63 plays an important role in maintaining expression of DNA repair genes, loss of which results in defective repair.


Subject(s)
Mutagens/toxicity , Stress, Physiological/genetics , Transcription Factors/metabolism , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/metabolism , Binding Sites , Cells, Cultured , Cisplatin/toxicity , DNA Breaks, Double-Stranded , DNA Repair , Doxorubicin/toxicity , Genome, Human , Humans , Keratinocytes/drug effects , Keratinocytes/metabolism , Transcription, Genetic
6.
Nucleic Acids Res ; 40(15): 7190-206, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22573176

ABSTRACT

The p63 transcription factor (TP63) is critical in development, growth and differentiation of stratifying epithelia. This is highlighted by the severity of congenital abnormalities caused by TP63 mutations in humans, the dramatic phenotypes in knockout mice and de-regulation of TP63 expression in neoplasia altering the tumour suppressive roles of the TP53 family. In order to define the normal role played by TP63 and provide the basis for better understanding how this network is perturbed in disease, we used chromatin immunoprecipitation combined with massively parallel sequencing (ChIP-seq) to identify >7500 high-confidence TP63-binding regions across the entire genome, in primary human neonatal foreskin keratinocytes (HFKs). Using integrative strategies, we demonstrate that only a subset of these sites are bound by TP53 in response to DNA damage. We identify a role for TP63 in transcriptional regulation of multiple genes genetically linked to cleft palate and identify AP-2alpha (TFAP2A) as a co-regulator of a subset of these genes. We further demonstrate that AP-2gamma (TFAP2C) can bind a subset of these regions and that acute depletion of either TFAP2A or TFAP2C alone is sufficient to reduce terminal differentiation of organotypic epidermal skin equivalents, indicating overlapping physiological functions with TP63.


Subject(s)
Epidermal Cells , Keratinocytes/metabolism , Transcription Factor AP-2/metabolism , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism , Binding Sites , Cell Differentiation , Cells, Cultured , Cleft Palate/genetics , Gene Expression Regulation , Genome, Human , Humans , Keratinocytes/cytology , Molecular Sequence Annotation , Regulatory Elements, Transcriptional , Transcription Factor AP-2/antagonists & inhibitors , Tumor Suppressor Protein p53/metabolism
7.
J Cell Sci ; 124(Pt 10): 1635-43, 2011 May 15.
Article in English | MEDLINE | ID: mdl-21511729

ABSTRACT

p63 is a master regulator of proliferation and differentiation in stratifying epithelia, and its expression is frequently altered in carcinogenesis. However, its role in maintaining proliferative capacity remains unclear. Here, we demonstrate that hypoproliferation and loss of differentiation in organotypic raft cultures of primary neonatal human foreskin keratinocytes (HFKs) depleted of the α and ß isoforms of p63 result from p53-p21-mediated accumulation of retinoblastoma (Rb) family member p130. Hypoproliferation in p63-depleted HFKs can be rescued by depletion of p53, p21(CIP1) or p130. Furthermore, we identified the gene encoding S-phase kinase-associated protein 2 (Skp2), the recognition component of the SCF(Skp2) E3 ubiquitin ligase, as a novel target of p63, potentially influencing p130 levels. Expression of Skp2 is maintained by p63 binding to a site in intron 2 and mRNA levels are downregulated in p63-depleted cells. Hypoproliferation in p63-depleted cells can be restored by re-expression of Skp2. Taken together, these results indicate that p63 plays a multifaceted role in maintaining proliferation in the mature regenerating epidermis, in addition to being required for differentiation.


Subject(s)
Keratinocytes/cytology , Keratinocytes/metabolism , Retinoblastoma-Like Protein p130/metabolism , S-Phase Kinase-Associated Proteins/metabolism , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism , Cell Cycle/physiology , Cell Differentiation/physiology , Cell Growth Processes/physiology , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p21/biosynthesis , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Gene Expression Regulation , Humans , Protein Isoforms , S-Phase Kinase-Associated Proteins/biosynthesis , S-Phase Kinase-Associated Proteins/genetics , Transcription Factors/genetics , Transcription, Genetic , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/genetics
8.
Elife ; 122023 01 16.
Article in English | MEDLINE | ID: mdl-36645410

ABSTRACT

Background: We have previously shown that the long non-coding (lnc)RNA prostate cancer associated 3 (PCA3; formerly prostate cancer antigen 3) functions as a trans-dominant negative oncogene by targeting the previously unrecognized prostate cancer suppressor gene PRUNE2 (a homolog of the Drosophila prune gene), thereby forming a functional unit within a unique allelic locus in human cells. Here, we investigated the PCA3/PRUNE2 regulatory axis from early (tumorigenic) to late (biochemical recurrence) genetic events during human prostate cancer progression. Methods: The reciprocal PCA3 and PRUNE2 gene expression relationship in paired prostate cancer and adjacent normal prostate was analyzed in two independent retrospective cohorts of clinically annotated cases post-radical prostatectomy: a single-institutional discovery cohort (n=107) and a multi-institutional validation cohort (n=497). We compared the tumor gene expression of PCA3 and PRUNE2 to their corresponding expression in the normal prostate. We also serially examined clinical/pathological variables including time to disease recurrence. Results: We consistently observed increased expression of PCA3 and decreased expression of PRUNE2 in prostate cancer compared with the adjacent normal prostate across all tumor grades and stages. However, there was no association between the relative gene expression levels of PCA3 or PRUNE2 and time to disease recurrence, independent of tumor grades and stages. Conclusions: We concluded that upregulation of the lncRNA PCA3 and targeted downregulation of the protein-coding PRUNE2 gene in prostate cancer could be early (rather than late) molecular events in the progression of human prostate tumorigenesis but are not associated with biochemical recurrence. Further studies of PCA3/PRUNE2 dysregulation are warranted. Funding: We received support from the Human Tissue Repository and Tissue Analysis Shared Resource from the Department of Pathology of the University of New Mexico School of Medicine and a pilot award from the University of New Mexico Comprehensive Cancer Center. RP and WA were supported by awards from the Levy-Longenbaugh Donor-Advised Fund and the Prostate Cancer Foundation. EDN reports research fellowship support from the Brazilian National Council for Scientific and Technological Development (CNPq), Brazil, and the Associação Beneficente Alzira Denise Hertzog Silva (ABADHS), Brazil. This work has been funded in part by the NCI Cancer Center Support Grants (CCSG; P30) to the University of New Mexico Comprehensive Cancer Center (CA118100) and the Rutgers Cancer Institute of New Jersey (CA072720).


Subject(s)
Prostatic Neoplasms , RNA, Long Noncoding , Humans , Male , Antigens, Neoplasm/genetics , Biomarkers, Tumor/genetics , Neoplasm Recurrence, Local/genetics , Prostate/metabolism , Prostatic Neoplasms/metabolism , Retrospective Studies , RNA, Long Noncoding/genetics
9.
J Biol Chem ; 286(5): 3915-24, 2011 Feb 04.
Article in English | MEDLINE | ID: mdl-21127042

ABSTRACT

Although members of the p63 family of transcription factors are known for their role in the development and differentiation of epithelial surfaces, their function in cancer is less clear. Here, we show that depletion of the ΔNp63α and ß isoforms, leaving only ΔNp63γ, results in epithelial to mesenchymal transition (EMT) in the normal breast cell line MCF10A. EMT can be rescued by the expression of the ΔNp63α isoform. We also show that ΔNp63γ expressed in a background where all the other ΔNp63 are knocked down causes EMT with an increase in TGFß-1, -2, and -3 and downstream effectors Smads2/3/4. In addition, a p63 binding site in intron 1 of TGFß was identified. Inhibition of the TGFß response with a specific inhibitor results in reversion of EMT in ΔNp63α- and ß-depleted cells. In summary, we show that p63 is involved in inhibiting EMT and reduction of certain p63 isoforms may be important in the development of epithelial cancers.


Subject(s)
Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic , Smad Proteins/genetics , Trans-Activators/physiology , Transforming Growth Factor beta/genetics , Tumor Suppressor Proteins/physiology , Binding Sites , Breast Neoplasms/etiology , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Humans , Neoplasms, Glandular and Epithelial/etiology , Protein Isoforms , Trans-Activators/metabolism , Transcription Factors , Tumor Suppressor Proteins/metabolism , Up-Regulation
10.
J Cell Sci ; 123(Pt 21): 3718-26, 2010 Nov 01.
Article in English | MEDLINE | ID: mdl-20940255

ABSTRACT

Although the retinoblastoma protein (Rb) functions as a checkpoint in the cell cycle, it also regulates differentiation. It has recently been shown that Rb is acetylated during differentiation; however, the role of this modification has not been identified. Depletion of Rb levels with short hairpin RNA resulted in inhibition of human keratinocyte differentiation, delayed cell cycle exit and allowed cell cycle re-entry. Restoration of Rb levels rescued defects in differentiation and cell cycle exit and re-entry; however, re-expression of Rb with the major acetylation sites mutated did not. During keratinocyte differentiation, acetylation of Rb is mediated by PCAF and it is further shown that PCAF acetyltransferase activity is also required for normal differentiation. The major acetylation sites in Rb are located within the nuclear localization sequence and, although mutation did not alter Rb localization in cycling cells, the mutant is mislocalized to the cytoplasm during differentiation. Studies indicate that acetylation is a mechanism for controlling Rb localization in human keratinocytes, with either reduction of the PCAF or exogenous expression of the deacetylase SIRT1, resulting in mislocalization of Rb. These findings identify PCAF-mediated acetylation of Rb as an event required to retain Rb within the nucleus during keratinocyte differentiation.


Subject(s)
Cell Nucleus/metabolism , Keratinocytes/metabolism , Retinoblastoma Protein/metabolism , Sirtuin 1/metabolism , p300-CBP Transcription Factors/metabolism , Acetylation , Active Transport, Cell Nucleus/genetics , Cell Differentiation/genetics , Cloning, Molecular , Enzyme Activation/genetics , Humans , Keratinocytes/pathology , Mutagenesis, Site-Directed , Protein Sorting Signals/genetics , RNA, Small Interfering/genetics , Retinoblastoma Protein/genetics , Sirtuin 1/genetics , Transgenes/genetics , p300-CBP Transcription Factors/genetics
11.
J Virol ; 85(21): 10999-1006, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21865393

ABSTRACT

A number of epigenetic alterations occur in both the virus and host cellular genomes during human papillomavirus (HPV)-associated carcinogenesis, and investigations of such alterations, including changes in chromatin proteins and histone modifications, have the potential to lead to therapeutic epigenetic reversion. We report here that transformed HPV16 E6/E7-expressing primary human foreskin keratinocytes (HFKs) (E6/E7 cells) demonstrate increased expression of the PRC2 methyltransferase EZH2 at both the mRNA and protein levels but do not exhibit the expected increase in trimethylated H3K27 (H3K27me3) compared to normal keratinocytes. In contrast, these cells show a reduction in global H3K27me3 levels in vitro, as well as upregulation of the KDM6A demethylase. We further show for the first time that transformation with the HPV16 E6 and E7 oncogenes also results in an increase in phosphorylated EZH2 serine 21 (P-EZH2-Ser21), mediated by active Akt, and in a downregulation of the PRC1 protein BMI1 in these cells. High-grade squamous cervical intraepithelial lesions also showed a loss of H3K27me3 in the presence of increased expression of EZH2. Correlating with the loss of H3K27me3, E6/E7 cells exhibited derepression of specific EZH2-, KMD6A-, and BMI1-targeted HOX genes. These results suggest that the observed reduction in H3K27me3 may be due to a combination of reduced activities/levels of specific polycomb proteins and increases in demethylases. The dysregulation of multiple chromatin proteins resulting in the loss of global H3K27me3 and the transcriptional reprogramming in HPV16 E6/E7-infected cells could provide an epigenetic signature associated with risk and/or progression of HPV16-associated cancers, as well as the potential for epigenetic reversion in the future.


Subject(s)
DNA-Binding Proteins/metabolism , Epigenesis, Genetic , Histone Demethylases/metabolism , Human papillomavirus 16/pathogenicity , Nuclear Proteins/metabolism , Oncogene Proteins, Viral/metabolism , Papillomavirus E7 Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Repressor Proteins/metabolism , Transcription Factors/metabolism , Cells, Cultured , Enhancer of Zeste Homolog 2 Protein , Gene Expression Profiling , Host-Pathogen Interactions , Humans , Keratinocytes/virology , Polycomb Repressive Complex 1 , Polycomb Repressive Complex 2
12.
J Virol ; 84(21): 10956-64, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20739533

ABSTRACT

Cells expressing human papillomavirus type 16 (HPV-16) E6 and E7 proteins exhibit deregulation of G2/M genes, allowing bypass of DNA damage arrest signals. Normally, cells with DNA damage that override the G2 damage checkpoint would precociously enter mitosis and ultimately face mitotic catastrophe and apoptotic cell death. However, E6/E7-expressing cells (E6/E7 cells) have the ability to enter and exit mitosis in the presence of DNA damage and continue with the next round of the cell cycle. Little is known about the mechanism that allows these cells to gain entry into and exit from mitosis. Here, we show that in the presence of DNA damage, E6/E7 cells have elevated levels of cyclin B, which would allow entry into mitosis. Also, as required for exit from mitosis, cyclin B is degraded in these cells, permitting initiation of the next round of DNA synthesis and cell cycle progression. Proteasomal degradation of cyclin B by anaphase-promoting complex/cyclosome (APC/C) is, in part, due to elevated levels of the E2-conjugating enzyme, Ubch10, and the substrate recognition protein, Cdc20, of APC/C. Also, in E6/E7 cells with DNA damage, while Cdc20 is complexed with BubR1, indicating an active checkpoint, it is also present in complexes free of BubR1, presumably allowing APC/C activity and slippage through the checkpoint.


Subject(s)
Cell Cycle Proteins/genetics , Gene Expression Regulation , Human papillomavirus 16/physiology , Keratinocytes/virology , Oncogene Proteins, Viral/genetics , Papillomavirus E7 Proteins/genetics , Repressor Proteins/genetics , Spindle Apparatus/virology , Ubiquitin-Conjugating Enzymes/genetics , Cdc20 Proteins , Cell Cycle , Cyclin B/metabolism , DNA Damage , Humans , Keratinocytes/metabolism , Mitosis , Spindle Apparatus/pathology
13.
J Virol ; 84(20): 10644-52, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20702634

ABSTRACT

A screen of microRNA (miRNA) expression following differentiation in human foreskin keratinocytes (HFKs) identified changes in several miRNAs, including miRNA 203 (miR-203), which has previously been shown to play an important role in epithelial cell biology by regulating p63 levels. We investigated how expression of human papillomavirus type 16 (HPV16) oncoproteins E6 and E7 affected miR-203 expression during proliferation and differentiation of HFKs. We demonstrated that miR-203 expression is reduced in HFKs where p53 function is compromised, either by the viral oncoprotein E6 or by knockout of p53 using short hairpin RNAs (p53i). We show that the induction of miR-203 observed during calcium-induced differentiation of HFKs is significantly reduced in HFKs expressing E6 and in p53i HFKs. Induction of miR-203 in response to DNA damage is also reduced in the absence of p53. We report that proliferation of HFKs is dependent on the level of miR-203 expression and that overexpression of miR-203 can reduce overproliferation in E6/E7-expressing and p53i HFKs. In summary, these results indicate that expression of miR-203 is dependent on p53, which may explain how expression of HPV16 E6 can disrupt the balance between proliferation and differentiation, as well as the response to DNA damage, in keratinocytes.


Subject(s)
Human papillomavirus 16/physiology , Human papillomavirus 16/pathogenicity , Keratinocytes/metabolism , Keratinocytes/virology , MicroRNAs/genetics , Oncogene Proteins, Viral/metabolism , Repressor Proteins/metabolism , Tumor Suppressor Protein p53/metabolism , Base Sequence , Cell Differentiation , Cell Proliferation , Cells, Cultured , DNA Damage , DNA Primers/genetics , Gene Expression , Gene Knockdown Techniques , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/physiology , Human papillomavirus 16/genetics , Humans , Keratinocytes/pathology , MicroRNAs/antagonists & inhibitors , MicroRNAs/metabolism , Oncogene Proteins, Viral/genetics , Papillomavirus E7 Proteins/genetics , Papillomavirus E7 Proteins/metabolism , Repressor Proteins/genetics , Tumor Suppressor Protein p53/antagonists & inhibitors , Tumor Suppressor Protein p53/genetics
14.
J Virol ; 84(10): 5131-9, 2010 May.
Article in English | MEDLINE | ID: mdl-20237089

ABSTRACT

The E6 and E7 oncoproteins of high-risk human papillomaviruses (HPVs) are together sufficient to cause cellular transformation. Nucleophosmin (NPM) was identified as a protein with increased levels in two-dimensional (2-D) gel analysis of human foreskin keratinocytes (HFKs) expressing E7 following methylcellulose-induced differentiation. Analysis of NPM expression in E7-expressing cells and E6- and E7-expressing cells in culture and in organotypic rafts confirmed the increased levels observed in 2-D gel analysis. The elevated expression of NPM was determined to be posttranscriptional and was attributed to increased v-akt murine thymoma viral oncogene (AKT) activity in the E6- and E7-expressing cells. Depletion of NPM caused a reduction in the replicative capacity of E7- and E6/E7-expressing HFKs and an increase in markers of differentiation. Also, the p53 and pRb tumor suppressor levels are increased with the knockdown of NPM in E6/E7-expressing cells, and, interestingly, p14(ARF) is relocalized from the nucleolus to the nucleoplasm and cytoplasm in these cells. The results show for the first time that NPM is required for the proliferation and inhibition of differentiation observed in HPV E6- and E7-expressing primary cells.


Subject(s)
Human papillomavirus 16/pathogenicity , Nuclear Proteins/biosynthesis , Oncogene Proteins, Viral/physiology , Papillomavirus E7 Proteins/physiology , Repressor Proteins/physiology , Virulence Factors/physiology , Cell Differentiation , Cell Proliferation , Cell Transformation, Viral , Cells, Cultured , Electrophoresis, Gel, Two-Dimensional , Humans , Keratinocytes/virology , Nucleophosmin , Up-Regulation
15.
Curr Biol ; 17(24): 2162-8, 2007 Dec 18.
Article in English | MEDLINE | ID: mdl-18060783

ABSTRACT

When human cells are stressed during G2, they are delayed from entering mitosis via a checkpoint mediated by the p38 kinase, and this delay can be modeled by the selective activation of p38 with anisomycin. Here, we report, on the basis of live-cell studies, that 75 nM anisomycin transiently (1 hr) activates p38 which, in turn, rapidly and completely blocks entry into mitosis for at least 4 hr in all primary, telomerase- or spontaneously immortalized (p53+ and pRB+) human cells. However, the same treatment does not delay entry into mitosis in cancer cells, or the delay in entering mitosis is shortened, even though it induces a similar transient and comparable (or stronger) activation of p38. Because the primary substrate of p38, the MK2 kinase, is also transiently (1-2 hr) activated by anisomycin in both normal and cancer cells, checkpoint disruption in transformed cells occurs downstream of MK2. Finally, observations on isogenic lines reveal that the duration of the stress checkpoint is shortened in cells lacking both p53 and pRb and that the constitutive expression of an active H-Ras oncogene in these cells further attenuates the checkpoint via an ERK1/2-dependent manner. Thus, transformation leads to attenuation of the p38-mediated stress checkpoint. This outcome is likely selected for during transformation because it confers the ability to outgrow normal cells under stressful in vitro (culture) or in vivo (tumor) environments. Our data caution against using cancer cells to study how p38 produces a G2 arrest.


Subject(s)
Cell Transformation, Neoplastic/metabolism , G2 Phase/physiology , Mitosis/physiology , p38 Mitogen-Activated Protein Kinases/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , HeLa Cells , Humans , Retinoblastoma Protein/metabolism , Stress, Physiological/metabolism , ras Proteins/metabolism
16.
Biochem Soc Trans ; 38(Pt 1): 223-8, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20074064

ABSTRACT

The p53 family of transcription factors is made up of p53, p63 and p73, which share significant structural homology. In particular, transcriptional complexity and the expression of multiple protein isoforms are an emergent trait of all family members. p63 is the evolutionarily eldest member of the p53 family and the various isoforms have critical roles in the development of stratifying epithelia. Recent results have uncovered additional splice variants, adding to the complexity of the transcriptional architecture of p63. These observations and the emerging extensive interplay between p63 and p53 in development, proliferation and differentiation underline the importance of considering all isoforms and family members in studies of the function of p53 family members.


Subject(s)
Epidermis , Morphogenesis , Protein Isoforms/metabolism , Skin Neoplasms/metabolism , Trans-Activators/metabolism , Tumor Suppressor Proteins/metabolism , Alternative Splicing , Animals , Epidermis/anatomy & histology , Epidermis/embryology , Epidermis/growth & development , Humans , Mice , Protein Isoforms/genetics , Trans-Activators/genetics , Transcription Factors , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/genetics
17.
Curr Opin Genet Dev ; 15(5): 515-9, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16099158

ABSTRACT

Human papillomaviruses are the causative agent of cancers in stratified epithelial surfaces. They replicate in the upper parts of the epithelium, where cells would normally be dying to produce a cornified layer. Therefore, they need to inhibit or delay differentiation and stimulate cell cycle progression to create an environment conducive for replication of the viral genome. The alterations both in differentiation and in the cell cycle are achieved by the viral proteins E6 and E7, which modulate cellular transcription mainly through their effects on p53 and the retinoblastoma family.


Subject(s)
Papillomaviridae/physiology , Transcription, Genetic/physiology , Cell Differentiation/physiology , Epithelial Cells/virology , Humans , Papillomaviridae/genetics , Viral Proteins/genetics , Viral Proteins/physiology
18.
Oncogene ; 38(48): 7329-7341, 2019 11.
Article in English | MEDLINE | ID: mdl-31417185

ABSTRACT

Radiation therapy (RT) is a cornerstone of treatment in the management of head and neck squamous cell carcinomas (HNSCC), yet treatment failure and disease recurrence are common. The p38/MK2 pathway is activated in response to cellular stressors, including radiation, and promotes tumor inflammation in a variety of cancers. We investigated MK2 pathway activation in HNSCC and the interaction of MK2 and RT in vitro and in vivo. We used a combination of an oropharyngeal SCC tissue microarray, HNSCC cell lines, and patient-derived xenograft (PDX) tumor models to study the effect of RT on MK2 pathway activation and to determine how inhibition of MK2 by pharmacologic (PF-3644022) and genetic (siRNA) methods impacts tumor growth. We show that high phosphorylated MK2 (p-MK2) levels are associated with worsened disease-specific survival in p16-negative HNSCC patients. RT increased p-MK2 in both p16-positive, HPV-positive and p16-negative, HPV-negative HNSCC cell lines. Pharmacologic inhibition or gene silencing of MK2 in vitro abrogated RT-induced increases in p-MK2; inflammatory cytokine expression and expression of the downstream MK2 target, heat shock protein 27 (HSP27); and markers of epithelial-to-mesenchymal transition. Mouse PDX models treated with a combination of RT and MK2 inhibitor experienced decreased tumor growth and increased survival. Our results suggest that MK2 is a potential prognostic biomarker for head and neck cancer and that MK2 pathway activation can mediate radiation resistance in HNSCC.


Subject(s)
Cytokines/metabolism , Head and Neck Neoplasms/pathology , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Papillomavirus Infections/complications , Protein Serine-Threonine Kinases/antagonists & inhibitors , Radiotherapy/methods , Squamous Cell Carcinoma of Head and Neck/pathology , Animals , Apoptosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Proliferation , Epithelial-Mesenchymal Transition , Female , Follow-Up Studies , Gene Expression Regulation, Neoplastic , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/radiotherapy , Head and Neck Neoplasms/virology , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Mice, Nude , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Papillomaviridae/isolation & purification , Papillomavirus Infections/virology , Phosphorylation , Prognosis , Protein Serine-Threonine Kinases/metabolism , Squamous Cell Carcinoma of Head and Neck/metabolism , Squamous Cell Carcinoma of Head and Neck/radiotherapy , Squamous Cell Carcinoma of Head and Neck/virology , Survival Rate , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
19.
Cancer Res ; 66(11): 5555-9, 2006 Jun 01.
Article in English | MEDLINE | ID: mdl-16740689

ABSTRACT

Human papillomaviruses (HPV) are small DNA tumor viruses causally associated with cervical cancer. The early gene product E7 from high-risk HPV is considered the major transforming protein expressed by the virus. Although many functions have been described for E7 in disrupting normal cellular processes, we describe in this study a new cellular target in primary human foreskin keratinocytes (HFK), the serine/threonine kinase AKT. Expression of HPV type 16 E7 in HFK caused inhibition of differentiation, hyperproliferation, and up-regulation of AKT activity in organotypic raft cultures. The ability of E7 to up-regulate AKT activity is dependent on its ability to bind to and inactivate the retinoblastoma (Rb) gene product family of proteins. Furthermore, we show that knocking down Rb alone, with short hairpin RNAs, was sufficient to up-regulate AKT activity in differentiated keratinocytes. Up-regulation of AKT activity and loss of Rb was also observed in HPV-positive cervical high-grade squamous intraepithelial lesions when compared with normal cervical tissue. Together, these data provide evidence linking inactivation of Rb by E7 in the up-regulation of AKT activity during cervical cancer progression.


Subject(s)
Human papillomavirus 16/physiology , Papillomavirus Infections/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Retinoblastoma Protein/metabolism , Cells, Cultured , Enzyme Activation , Female , Human papillomavirus 16/metabolism , Humans , Keratinocytes/metabolism , Keratinocytes/virology , Papillomavirus Infections/enzymology , Papillomavirus Infections/genetics , Papillomavirus Infections/virology , Phosphorylation , RNA, Small Interfering/genetics , Retinoblastoma Protein/antagonists & inhibitors , Retinoblastoma Protein/genetics , Up-Regulation , Uterine Cervical Neoplasms/enzymology , Uterine Cervical Neoplasms/metabolism , Uterine Cervical Neoplasms/pathology , Uterine Cervical Neoplasms/virology , Uterine Cervical Dysplasia/enzymology , Uterine Cervical Dysplasia/metabolism , Uterine Cervical Dysplasia/pathology , Uterine Cervical Dysplasia/virology
20.
Clin Cancer Res ; 24(16): 3917-3927, 2018 08 15.
Article in English | MEDLINE | ID: mdl-29739791

ABSTRACT

Purpose: To investigate the regulation of epithelial-to-mesenchymal transition (EMT) in head and neck squamous cell carcinoma (HNSCC) and its importance in tumor invasion.Experimental Design: We use a three-dimensional invasive organotypic raft culture model of human foreskin keratinocytes expressing the E6/E7 genes of the human papilloma virus-16, coupled with bioinformatic and IHC analysis of patient samples to investigate the role played by EMT in invasion and identify effectors and upstream regulatory pathways.Results: We identify SNAI2 (Slug) as a critical effector of EMT-activated downstream of TP63 overexpression in HNSCC. Splice-form-specific depletion and rescue experiments further identify the ΔNp63γ isoform as both necessary and sufficient to activate the SRC signaling axis and SNAI2-mediated EMT and invasion. Moreover, elevated SRC levels are associated with poor outcome in patients with HNSCC in The Cancer Genome Atlas dataset. Importantly, the effects on EMT and invasions and SNAI2 expression can be reversed by genetic or pharmacologic inhibition of SRC.Conclusions: Overexpression of ΔNp63γ modulates cell invasion by inducing targetable SRC-Slug-evoked EMT in HNSCC, which can be reversed by inhibitors of the SRC signaling. Clin Cancer Res; 24(16); 3917-27. ©2018 AACR.


Subject(s)
Snail Family Transcription Factors/genetics , Squamous Cell Carcinoma of Head and Neck/genetics , Transcription Factors/genetics , Tumor Suppressor Proteins/genetics , src-Family Kinases/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic/genetics , Human papillomavirus 16/genetics , Human papillomavirus 16/pathogenicity , Humans , Keratinocytes/virology , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Oncogene Proteins, Viral/genetics , Protein Isoforms/genetics , Repressor Proteins/genetics , Signal Transduction/genetics , Squamous Cell Carcinoma of Head and Neck/pathology , Squamous Cell Carcinoma of Head and Neck/virology
SELECTION OF CITATIONS
SEARCH DETAIL