Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Int J Mol Sci ; 20(5)2019 Mar 04.
Article in English | MEDLINE | ID: mdl-30836646

ABSTRACT

Supplementation of micronutrients like folate is a double-edged sword in terms of their ambivalent role in cell metabolism. Although several epidemiological studies support a protective role of folate in carcinogenesis, there are also data arguing for an opposite effect. To address this issue in the context of human papillomavirus (HPV)-induced transformation, the molecular events of different folate availability on human keratinocytes immortalized by HPV16 E6 and E7 oncoproteins were examined. Several sublines were established: Control (4.5 µM folate), folate deficient (0.002 µM folate), and repleted cells (4.5 µM folate). Cells were analyzed in terms of oncogene expression, DNA damage and repair, karyotype changes, whole-genome sequencing, and transcriptomics. Here we show that folate depletion irreversibly induces DNA damage, impairment of DNA repair fidelity, and unique chromosomal alterations. Repleted cells additionally underwent growth advantage and enhanced clonogenicity, while the above mentioned impaired molecular properties became even more pronounced. Overall, it appears that a period of folate deficiency followed by repletion can shape immortalized cells toward an anomalous phenotype, thereby potentially contributing to carcinogenesis. These observations should elicit questions and inquiries for broader additional studies regarding folate fortification programs, especially in developing countries with micronutrient deficiencies and high HPV prevalence.


Subject(s)
Folic Acid Deficiency/genetics , Human papillomavirus 16/genetics , Papillomavirus Infections/genetics , Transcription, Genetic , Carcinogenesis/genetics , DNA Damage/ethics , DNA Repair/genetics , Folic Acid/genetics , Folic Acid Deficiency/pathology , Folic Acid Deficiency/virology , Genomics , Human papillomavirus 16/pathogenicity , Humans , Keratinocytes/virology , Oncogene Proteins, Viral/genetics , Papillomavirus E7 Proteins/genetics , Papillomavirus Infections/virology , Repressor Proteins/genetics
2.
mSphere ; 4(2)2019 03 27.
Article in English | MEDLINE | ID: mdl-30918060

ABSTRACT

Although the role of high-risk human papillomaviruses (hrHPVs) as etiological agents in cancer development has been intensively studied during the last decades, there is still the necessity of understanding the impact of the HPV E6 and E7 oncogenes on host cells, ultimately leading to malignant transformation. Here, we used newly established immortalized human keratinocytes with a well-defined HPV16 E6E7 expression cassette to get a more complete and less biased overview of global changes induced by HPV16 by employing transcriptome sequencing (RNA-Seq) and stable isotope labeling by amino acids in cell culture (SILAC). This is the first study combining transcriptome and proteome data to characterize the impact of HPV oncogenes in human keratinocytes in comparison with their virus-negative counterparts. To enhance the informative value and accuracy of the RNA-Seq data, four different bioinformatic workflows were used. We identified potential novel upstream regulators (e.g., CNOT7, SPDEF, MITF, and PAX5) controlling distinct clusters of genes within the HPV-host cell network as well as distinct factors (e.g., CPPED1, LCP1, and TAGLN) with essential functions in cancer. Validated results in this study were compared to data sets from The Cancer Genome Atlas (TCGA), demonstrating that several identified factors were also differentially expressed in cervical squamous cell carcinoma and endocervical adenocarcinoma (CESC) and HPV-positive head and neck squamous cell carcinomas (HNSCs). This highly integrative approach allows the identification of novel HPV-induced cellular changes that are also reflected in cancer patients, providing a promising omics data set for future studies in both basic and translational research.IMPORTANCE Human papillomavirus (HPV)-associated cancers still remain a big health problem, especially in developing countries, despite the availability of prophylactic vaccines. Although HPV oncogenes have been intensively investigated for decades, a study applying recent advances in RNA-Seq and quantitative proteomic approaches to a precancerous model system with well-defined HPV oncogene expression alongside HPV-negative parental cells has been missing until now. Here, combined omics analyses reveal global changes caused by the viral oncogenes in a less biased way and allow the identification of novel factors and key cellular networks potentially promoting malignant transformation. In addition, this system also provides a basis for mechanistic research on novel key factors regulated by HPV oncogenes, especially those that are confirmed in vivo in cervical cancer as well as in head and neck cancer patient samples from TCGA data sets.


Subject(s)
Carcinogenesis/genetics , Gene Regulatory Networks , Keratinocytes/virology , Oncogene Proteins, Viral/genetics , Proteome/genetics , Transcriptome , Adenocarcinoma/genetics , Adenocarcinoma/virology , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/virology , Cell Transformation, Neoplastic , Computational Biology , Female , Gene Expression Profiling , High-Throughput Nucleotide Sequencing , Human papillomavirus 16/genetics , Humans , Proteomics , Squamous Cell Carcinoma of Head and Neck/genetics , Squamous Cell Carcinoma of Head and Neck/virology , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/virology
3.
Hum Mol Genet ; 21(8): 1693-705, 2012 Apr 15.
Article in English | MEDLINE | ID: mdl-22210627

ABSTRACT

In the present study, we used the human papillomavirus type 16 (HPV16)-positive cervical carcinoma cell line CaSki as a paradigmatic model to understand epigenetic silencing of viral multi-copy genomes. We show that most of the hypermethylated HPV16 copies are kept as 'occluded' chromatin that defines a condition where genes were refractory in their response to trans-acting transcription factors and to external reactivation efforts. This provides the first example that viral genomes are silenced by such a host cell mechanism, hitherto only known for endogenous genes to preserve a stable and robust phenotype. Moreover, considering an adaptive cross-talk between viral proteins and the epigenetic modification machinery, we demonstrate that particularly E2-but also ectopically delivered E6/E7-can induce significant de novo methylation within the enhancer and, to a less extent, within the promoter region. These data suggest that under certain physiological conditions, HPV can down-regulate its own gene expression, regardless of the presence of transcriptional activators. We propose that self-methylation of multi-copy HPV could be the first event prior to heterochromatin formation. These processes favour an 'occluded' chromatin conformation, finally being unresponsive to transcriptional activation. The shift from potentially competent heterochromatin towards an occluded state is basically irreversible, possibly using the same mechanism described for lineage differentiation. Along this line, it is tempting to speculate that virus-cell interaction is able to 'sense' viral copy number and down-regulates excess of gene activity in order to guarantee cell viability.


Subject(s)
Chromatin/metabolism , DNA Methylation , DNA, Viral/genetics , Epigenesis, Genetic , Gene Silencing , Human papillomavirus 16/genetics , Human papillomavirus 16/physiology , Cell Line, Tumor , Chromatin/chemistry , DNA, Viral/metabolism , DNA-Binding Proteins/metabolism , Enhancer Elements, Genetic , Gene Expression Regulation, Viral , Genome, Viral , Histones/metabolism , Host-Pathogen Interactions , Humans , Oncogene Proteins, Viral/metabolism , Papillomavirus E7 Proteins/metabolism , Repressor Proteins/metabolism , Transcription, Genetic , Viral Proteins/metabolism
4.
J Biol Chem ; 282(39): 28520-28529, 2007 Sep 28.
Article in English | MEDLINE | ID: mdl-17686773

ABSTRACT

Retinoic acid receptor beta2 (RAR beta2) is often down-regulated during the multistep process to cervical cancer. In that way, its inhibitory function on the transcription factor AP-1, indispensable to maintain human papillomavirus (HPV) gene expression is relieved. Using HPV-18 positive HeLa cells as a model system, we show that ectopic expression of RAR beta2 is able to down-regulate HPV-18 transcription by selectively abrogating the binding of AP-1 to the viral regulatory region in a ligand-independent manner. This resulted in down-regulation of the viral mRNAs at the level of initiation of transcription. Decreased oncogene expression was accompanied by a re-induction of cell cycle inhibitory proteins such as p53, p21(CIP1), and p27(KIP) as well as by a cessation of cellular growth. Reduced transcriptional activity as a consequence of AP-1 reduction by selective c-Jun degradation apparently targets the HPV-18 regulatory region for epigenetic modification such as de novo methylation and nucleosomal condensation. This mechanism is otherwise counterbalanced by active and abundant viral transcription in malignant cells, because RAR beta2 itself becomes inactivated during cervical carcinogenesis. Hence, our study shows that the temporal co-existence of a potential repressor and viral oncoproteins is mutually exclusive and provides evidence of a cross-talk between a nuclear receptor, AP-1, and the epigenetic machinery.


Subject(s)
Cell Transformation, Viral , DNA Methylation , DNA, Viral/metabolism , Gene Expression Regulation, Viral , Heterochromatin/virology , Human papillomavirus 18/metabolism , Receptors, Retinoic Acid/metabolism , Transcription Factor AP-1/metabolism , Uterine Cervical Neoplasms/virology , Calcium-Binding Proteins/metabolism , Cell Cycle , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Down-Regulation , Female , Gene Silencing , Genes, jun , HeLa Cells , Heterochromatin/metabolism , Humans , Models, Biological , Nucleosomes/metabolism , RNA, Viral/biosynthesis , Response Elements , Transcription, Genetic , Tumor Suppressor Protein p53/metabolism , Uterine Cervical Neoplasms/metabolism , Viral Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...