Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
1.
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
2.
Cancer Epidemiol Biomarkers Prev ; 29(1): 31-38, 2020 01.
Article in English | MEDLINE | ID: mdl-31666283

ABSTRACT

BACKGROUND: Determination of human papillomavirus (HPV) status has become clinically relevant for patient stratification under UICC TNM8 staging. Within the United Kingdom, a combination of p16 IHC and HPV DNA-ISH is recommended for classifying HPV status. This study will assess a series of clinically applicable second-line molecular tests to run in combination with p16 IHC to optimally determine HPV status. METHODS: The ability of HPV RNA-ISH, HPV DNA-ISH, and HPV DNA-PCR to identify p16-positive/HPV-positive patients was investigated in a population-based oropharyngeal squamous cell carcinoma (OPSCC) cohort of patients diagnosed in Northern Ireland from 2000 to 2011. RESULTS: Only 41% of the Northern Irish OPSCC patient population was associated with HPV-driven carcinogenesis. Both ISH assays were more specific than the DNA-PCR assay (100% and 95% vs. 67%) and were less likely to be affected by preanalytic factors such as increasing block age. A pooled HPV genotype probe for RNA-ISH was found to be the most accurate molecular assay assessed (95% accuracy) when compared with p16 positivity. CONCLUSIONS: Our study demonstrates the advantage of tissue-based molecular assays when determining HPV status in retrospective samples. Specifically, we demonstrate the enhanced sensitivity and specificity of ISH techniques compared with PCR-based methodology when working with formalin-fixed paraffin-embedded tissue, and found HPV RNA-ISH to be the most effective assay for determining HPV status. IMPACT: As p16 IHC is a relatively inexpensive, accessible, and sensitive test for stratifying patients by HPV status, this study finds that more patients would benefit from first-line p16 IHC followed by specific HPV testing using HPV RNA-ISH to confirm HPV status.


Subject(s)
Alphapapillomavirus/isolation & purification , Human Papillomavirus DNA Tests/methods , Oropharyngeal Neoplasms/diagnosis , Papillomavirus Infections/diagnosis , Squamous Cell Carcinoma of Head and Neck/diagnosis , Age Factors , Alphapapillomavirus/genetics , Alphapapillomavirus/immunology , Biomarkers, Tumor/analysis , Biomarkers, Tumor/immunology , Cyclin-Dependent Kinase Inhibitor p16/analysis , Cyclin-Dependent Kinase Inhibitor p16/immunology , DNA, Viral/isolation & purification , Female , Follow-Up Studies , Humans , Immunohistochemistry , In Situ Hybridization , Kaplan-Meier Estimate , Male , Middle Aged , Neoplasm Staging , Northern Ireland/epidemiology , Oropharyngeal Neoplasms/immunology , Oropharyngeal Neoplasms/pathology , Oropharyngeal Neoplasms/virology , Papillomavirus Infections/mortality , Papillomavirus Infections/pathology , Papillomavirus Infections/virology , Prognosis , RNA, Viral/isolation & purification , Retrospective Studies , Sensitivity and Specificity , Squamous Cell Carcinoma of Head and Neck/mortality , Squamous Cell Carcinoma of Head and Neck/pathology , Squamous Cell Carcinoma of Head and Neck/virology
3.
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
4.
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
5.
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
6.
Mutat Res Rev Mutat Res ; 772: 67-77, 2017.
Article in English | MEDLINE | ID: mdl-28528691

ABSTRACT

Human papillomaviruses (HPV) infect and replicate in stratified epithelium at cutaneous and mucosal surfaces. The proliferation and maintenance of keratinocytes, the cells which make up this epithelium, are controlled by a number of growth factor receptors such as the keratinocyte growth factor receptor (KGFR, also called fibroblast growth factor receptor 2b (FGFR2b)), the epithelial growth factor receptor (EGFR) and the insulin-like growth factor receptors 1 and 2 (IGF1R and IGF2R). In this review, we will delineate the mutation, gene transcription, translation and processing of the IGF axis within HPV associated cancers. The IGFs are key for developmental and postnatal growth of almost all tissues; we explore whether this crucial axis has been hijacked by HPV.


Subject(s)
Gene Expression Regulation, Neoplastic , Neoplasms/genetics , Neoplasms/virology , Papillomaviridae/pathogenicity , Somatomedins/genetics , Cell Proliferation , ErbB Receptors/genetics , ErbB Receptors/metabolism , Humans , Keratinocytes/cytology , Keratinocytes/virology , Receptor, Fibroblast Growth Factor, Type 2/genetics , Receptor, Fibroblast Growth Factor, Type 2/metabolism , Receptor, IGF Type 1 , Receptor, IGF Type 2/genetics , Receptor, IGF Type 2/metabolism , Receptors, Somatomedin/genetics , Receptors, Somatomedin/metabolism , Somatomedins/metabolism
7.
Oncotarget ; 8(10): 16202-16219, 2017 Mar 07.
Article in English | MEDLINE | ID: mdl-26001294

ABSTRACT

Using microarray information from oro-pharyngeal data sets and results from primary human foreskin keratinocytes (HFK) expressing Human Papilloma Virus (HPV)-16 E6/E7 proteins, we show that p63 expression regulates signalling molecules which initiate cell migration such as Src and focal adhesion kinase (FAK) and induce invasion in 3D-organotypic rafts; a phenotype that can be reversed by depletion of p63. Knockdown of Src or FAK in the invasive cells restored focal adhesion protein paxillin at cell periphery and impaired the cell migration. In addition, specific inhibition of FAK (PF573228) or Src (dasatinib) activities mitigated invasion and attenuated the expression/activity of matrix metalloproteinase 14 (MMP14), a pivotal MMP in the MMP activation cascade. Expression of constitutively active Src in non-invasive HFK expressing E6/E7 proteins upregulated the activity of c-Jun and MMP14, and induced invasion in rafts. Depletion of Src, FAK or AKT in the invasive cells normalised the expression/activity of c-Jun and MMP14, thus implicating the Src-FAK/AKT/AP-1 signalling in MMP14-mediated extra-cellular matrix remodelling. Up-regulation of Src, AP-1, MMP14 and p63 expression was confirmed in oro-pharyngeal cancer. Since p63 transcriptionally regulated expression of many of the genes in this signalling pathway, it suggests that it has a central role in cancer progression.


Subject(s)
Focal Adhesion Kinase 1/metabolism , Keratinocytes/metabolism , Oncogene Proteins, Viral/metabolism , Papillomavirus E7 Proteins/metabolism , Repressor Proteins/metabolism , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism , src-Family Kinases/metabolism , Blotting, Western , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/virology , Cell Movement/drug effects , Cell Movement/genetics , Dasatinib/pharmacology , Fluorescent Antibody Technique , Focal Adhesion Kinase 1/antagonists & inhibitors , Focal Adhesion Kinase 1/genetics , Foreskin/cytology , Humans , Keratinocytes/cytology , Male , Matrix Metalloproteinase 14/genetics , Matrix Metalloproteinase 14/metabolism , Oncogene Proteins, Viral/genetics , Oropharyngeal Neoplasms/genetics , Oropharyngeal Neoplasms/metabolism , Oropharyngeal Neoplasms/virology , Papillomavirus E7 Proteins/genetics , Papillomavirus Infections/genetics , Papillomavirus Infections/metabolism , Papillomavirus Infections/virology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-jun/genetics , Proto-Oncogene Proteins c-jun/metabolism , Quinolones/pharmacology , RNA Interference , Repressor Proteins/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Signal Transduction/genetics , Sulfones/pharmacology , Transcription Factor AP-1/genetics , Transcription Factor AP-1/metabolism , Transcription Factors/genetics , Tumor Suppressor Proteins/genetics , src-Family Kinases/antagonists & inhibitors , src-Family Kinases/genetics
8.
Cancer Res ; 76(22): 6598-6606, 2016 11 15.
Article in English | MEDLINE | ID: mdl-27569214

ABSTRACT

A rising incidence of oropharyngeal squamous cell carcinoma (OPSCC) incidence has occurred throughout the developed world, where it has been attributed to an increasing impact of human papillomavirus (HPV) on disease etiology. This report presents the findings of a multicenter cross-sectional retrospective study aimed at determining the proportion of HPV-positive and HPV-negative OPSCC within the United Kingdom. Archival tumor tissue blocks from 1,602 patients previously diagnosed with OPSCC (2002-2011) were collated from 11 centers. HPV status was determined with three validated commercial tests to provide valid data for 1,474 cases in total. Corresponding national incidence data from the same decade were obtained from UK Cancer registries. The overall proportion of HPV+ OPSCC between 2002 and 2011 was 51.8% [95% confidence interval (CI), 49.3-54.4], and this remained unchanged throughout the decade [unadjusted RR = 1.00 (95% CI, 0.99-1.02)]. However, over the same period, the incidence of OPSCC in the broader UK population underwent a 2-fold increase [age-standardized rate 2002: 2.1 (95% CI, 1.9-2.2); 2011: 4.1 (95% CI, 4.0-4.3)]. Although the number of OPSCCs diagnosed within the United Kingdom from 2002 to 2011 nearly doubled, the proportion of HPV+ cases remained static at approximately 50%. Our results argue that the rapidly increasing incidence of OPSCC in the United Kingdom cannot be solely attributable to the influence of HPV. The parallel increase in HPV+ and HPV- cases we documented warrants further investigation, so that appropriate future prevention strategies for both types of disease can be implemented. Cancer Res; 76(22); 6598-606. ©2016 AACR.


Subject(s)
Oropharyngeal Neoplasms/etiology , Papillomaviridae/genetics , Papillomavirus Infections/virology , Female , Humans , Male , Oropharyngeal Neoplasms/virology , United Kingdom
9.
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
10.
Article in English | MEDLINE | ID: mdl-25774149

ABSTRACT

The role of insulin-like growth factor binding protein 2 (IGFBP2) in cancer is unclear. In general, IGFBP2 is considered to be oncogenic and its expression is often observed to be elevated in cancer. However, there are a number of conflicting reports in vitro and in vivo where IGFBP2 acts in a tumor suppressor manner. In this mini-review, we discuss the factors influencing the variation in IGFBP2 expression in cancer and our interpretation of these findings.

11.
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
12.
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
13.
Virology ; 448: 210-6, 2014 Jan 05.
Article in English | MEDLINE | ID: mdl-24314651

ABSTRACT

A screen of microRNA (miRNA) expression following differentiation in human foreskin keratinocytes (HFKs) identified changes in several miRNAs, including miR-24 and miR-205. We investigated how expression of Human Papilloma Virus Type-16 (HPV16) onco-proteins E6 and E7 affected expression of miR-24 and miR-205 during proliferation and differentiation of HFKs. We show that the induction of both miR-24 and miR-205 observed during differentiation of HFKs is lost in HFKs expressing E6 and E7. We demonstrate that the effect on miR-205 is due to E7 activity, as miR-205 expression is dependent on pRb expression. Finally, we provide evidence that miR-24 effects in the cell may be due to targeting of cyclin dependent kinase inhibitor p27. In summary, these results indicate that expression of both miR-24 and miR-205 are impacted by E6 and/or E7 expression, which may be one mechanism by which HPV onco-proteins can disrupt the balance between proliferation and differentiation in keratinocytes.


Subject(s)
Human papillomavirus 16/metabolism , Keratinocytes/metabolism , MicroRNAs/genetics , Oncogene Proteins, Viral/metabolism , Papillomavirus E7 Proteins/metabolism , Papillomavirus Infections/genetics , Repressor Proteins/metabolism , Cell Proliferation , Foreskin/cytology , Foreskin/metabolism , Foreskin/virology , Human papillomavirus 16/genetics , Humans , Keratinocytes/cytology , Keratinocytes/virology , Male , MicroRNAs/metabolism , Oncogene Proteins, Viral/genetics , Papillomavirus E7 Proteins/genetics , Papillomavirus Infections/metabolism , Papillomavirus Infections/physiopathology , Papillomavirus Infections/virology , Repressor Proteins/genetics
14.
Oncotarget ; 4(7): 1103-16, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23867201

ABSTRACT

The tumour microenvironment has an important role in cancer progression and recent reports have proposed that stromal AKT is activated and regulates tumourigenesis and invasion. We have shown, by immuno-fluorescent analysis of oro-pharyngeal cancer biopsies, an increase in AKT activity in tumour associated stromal fibroblasts compared to normal stromal fibroblasts. Using organotypic raft co-cultures, we show that activation of stromal AKT can induce the invasion of keratinocytes expressing the HPV type 16 E6 and E7 proteins, in a Keratinocyte Growth Factor (KGF) dependent manner. By depleting stromal fibroblasts of each of the three AKT isoforms independently, or through using isoform specific inhibitors, we determined that stromal AKT2 is an essential regulator of invasion and show in oro-pharyngeal cancers that AKT2 specific phosphorylation events are also identified in stromal fibroblasts. Depletion of stromal AKT2 inhibits epithelial invasion through activating a protective pathway counteracting KGF mediated invasions. AKT2 depletion in fibroblasts stimulates the cleavage and release of IL1B from stromal fibroblasts resulting in down-regulation of the KGF receptor (fibroblast growth factor receptor 2B (FGFR2B)) expression in the epithelium. We also show that high IL1B is associated with increased overall survival in a cohort of patients with oro-pharyngeal cancers. Our findings demonstrate the importance of stromal derived growth factors and cytokines in regulating the process of tumour cell invasion.


Subject(s)
Fibroblasts/enzymology , Fibroblasts/pathology , Oropharyngeal Neoplasms/enzymology , Oropharyngeal Neoplasms/pathology , Proto-Oncogene Proteins c-akt/metabolism , Carcinogenesis/metabolism , Carcinogenesis/pathology , Cells, Cultured , Disease Progression , Epithelial Cells/enzymology , Epithelial Cells/pathology , Humans , Keratinocytes/enzymology , Keratinocytes/pathology , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Stromal Cells/enzymology , Stromal Cells/pathology , Tumor Microenvironment
15.
J Invest Dermatol ; 132(12): 2691-9, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22696061

ABSTRACT

Signaling between the epithelium and stromal cells is crucial for growth, differentiation, and repair of the epithelium. Although the retinoblastoma protein (Rb) is known to regulate the growth of keratinocytes in a cell-autonomous manner, here we describe a function of Rb in the stromal compartment. We find that Rb depletion in fibroblasts leads to inhibition of differentiation and enhanced proliferation of the epithelium. Analysis of conditioned medium identified that keratinocyte growth factor (KGF) levels were elevated following Rb depletion. These findings were also observed with organotypic co-cultures. Treatment of keratinocytes with KGF inhibited differentiation and enhanced keratinocyte proliferation, whereas reduction of KGF levels in Rb-depleted fibroblasts was able to restore expression of differentiation markers. Our findings suggest a crucial role for dermal fibroblasts in regulating the differentiation and proliferation of keratinocytes, and we demonstrate a role for stromal Rb in this cross-talk.


Subject(s)
Epidermal Cells , Keratinocytes/cytology , Retinoblastoma Protein/genetics , Retinoblastoma Protein/metabolism , Stromal Cells/cytology , Cell Differentiation/physiology , Cell Line, Transformed , Cell Proliferation , Cells, Cultured , Epidermis/growth & development , Epidermis/metabolism , Epithelial Cells/cytology , Epithelial Cells/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Foreskin/cytology , Humans , Interleukin-1alpha/metabolism , Interleukin-1alpha/pharmacology , Interleukin-1beta/metabolism , Interleukin-1beta/pharmacology , Keratinocytes/metabolism , Male , Mouth/cytology , Signal Transduction/drug effects , Signal Transduction/physiology , Stromal Cells/metabolism
16.
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
17.
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
18.
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
19.
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
20.
Cancer Res ; 71(5): 1933-44, 2011 Mar 01.
Article in English | MEDLINE | ID: mdl-21363924

ABSTRACT

Little is known about the origin of basal-like breast cancers, an aggressive disease that is highly similar to BRCA1-mutant breast cancers. p63 family proteins that are structurally related to the p53 suppressor protein are known to function in stem cell regulation and stratified epithelia development in multiple tissues, and p63 expression may be a marker of basal-like breast cancers. Here we report that ΔNp63 isoforms of p63 are transcriptional targets for positive regulation by BRCA1. Our analyses of breast cancer tissue microarrays and BRCA1-modulated breast cancer cell lines do not support earlier reports that p63 is a marker of basal-like or BRCA1 mutant cancers. Nevertheless, we found that BRCA1 interacts with the specific p63 isoform ΔNp63γ along with transcription factor isoforms AP-2α and AP-2γ. BRCA1 required ΔNp63γ and AP-2γ to localize to an intronic enhancer region within the p63 gene to upregulate transcription of the ΔNp63 isoforms. In mammary stem/progenitor cells, siRNA-mediated knockdown of ΔNp63 expression resulted in genomic instability, increased cell proliferation, loss of DNA damage checkpoint control, and impaired growth control. Together, our findings establish that transcriptional upregulation of ΔNp63 proteins is critical for BRCA1 suppressor function and that defects in BRCA1-ΔNp63 signaling are key events in the pathogenesis of basal-like breast cancer.


Subject(s)
Breast Neoplasms/genetics , Gene Expression Regulation, Neoplastic/genetics , Trans-Activators/genetics , Tumor Suppressor Proteins/genetics , Ubiquitin-Protein Ligases/genetics , Blotting, Western , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Chromatin Immunoprecipitation , Female , Fluorescent Antibody Technique , Gene Expression , Humans , Protein Isoforms/biosynthesis , Protein Isoforms/genetics , RNA, Small Interfering , Reverse Transcriptase Polymerase Chain Reaction , Tissue Array Analysis , Trans-Activators/biosynthesis , Transcription Factors , Transfection , Tumor Suppressor Proteins/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL