Genomic profiling of the transcription factor Zfp148 and its impact on the p53 pathway

Autor: Ella M. Eklund, Jozefina J. Dzanan, Markus Lindberg, Anna Staffas, Ben O. Titmuss, Angana A. H. Patel, Erik Larsson, Per Lindahl, Zhiyuan V. Zou, Eva Forssell-Aronsson, Kristell Le Gal, Inger Johansson, Nadia Gul, Åsa Tivesten, Abdulmalik A. Bokhari, Volkan I. Sayin, Viktor Garellick, Martin O. Bergo
Rok vydání: 2019
Předmět:
0301 basic medicine
Chromatin Immunoprecipitation
Cell division
lcsh:Medicine
Down-Regulation
Cell Cycle Proteins
Biology
Senescence
Article
Cell Line
03 medical and health sciences
Mice
0302 clinical medicine
Transcription (biology)
RNA interference
Animals
RNA
Small Interfering

lcsh:Science
Transcription factor
Cyclin-Dependent Kinase Inhibitor p16
Etoposide
Multidisciplinary
Cell growth
Activator (genetics)
lcsh:R
Promoter
Cell Cycle Checkpoints
Fibroblasts
Cell Cycle Gene
Cell biology
E2F Transcription Factors
Computational biology and bioinformatics
DNA-Binding Proteins
030104 developmental biology
Gene Ontology
Gene Expression Regulation
lcsh:Q
RNA Interference
CRISPR-Cas Systems
Cisplatin
Tumor Suppressor Protein p53
Chromatin immunoprecipitation
030217 neurology & neurosurgery
DNA Damage
Signal Transduction
Transcription Factors
Zdroj: Scientific Reports
Scientific Reports, Vol 10, Iss 1, Pp 1-13 (2020)
ISSN: 2045-2322
Popis: Recent data suggest that the transcription factor Zfp148 represses activation of the tumor suppressor p53 in mice and that therapeutic targeting of the human orthologue ZNF148 could activate the p53 pathway without causing detrimental side effects. We have previously shown that Zfp148 deficiency promotes p53-dependent proliferation arrest of mouse embryonic fibroblasts (MEFs), but the underlying mechanism is not clear. Here, we showed that Zfp148 deficiency downregulated cell cycle genes in MEFs in a p53-dependent manner. Proliferation arrest of Zfp148-deficient cells required increased expression of ARF, a potent activator of the p53 pathway. Chromatin immunoprecipitation showed that Zfp148 bound to the ARF promoter, suggesting that Zfp148 represses ARF transcription. However, Zfp148 preferentially bound to promoters of other transcription factors, indicating that deletion of Zfp148 may have pleiotropic effects that activate ARF and p53 indirectly. In line with this, we found no evidence of genetic interaction between TP53 and ZNF148 in CRISPR and siRNA screen data from hundreds of human cancer cell lines. We conclude that Zfp148 deficiency, by increasing ARF transcription, downregulates cell cycle genes and cell proliferation in a p53-dependent manner. However, the lack of genetic interaction between ZNF148 and TP53 in human cancer cells suggests that therapeutic targeting of ZNF148 may not increase p53 activity in humans.
Databáze: OpenAIRE