Etoposide-induced DNA damage is increased in p53 mutants: identification of ATR and other genes that influence effects of p53 mutations on Top2-induced cytotoxicity

The functional status of the tumor suppressor p53 is a critical component in determining the sensitivity of cancer cells to many chemotherapeutic agents. DNA topoisomerase II (Top2) plays essential roles in DNA metabolism and is the target of FDA approved chemotherapeutic agents. Topoisomerase targeting drugs convert the enzyme into a DNA damaging agent and p53 influences cellular responses to these agents. We assessed the impact of the loss of p53 function on the formation of DNA damage induced by the Top2 poison etoposide. Using human HCT116 cells, we found resistance to etoposide in cell growth assays upon the functional loss of p53. Nonetheless, cells lacking fully functional p53 were etoposide hypersensitive in clonogenic survival assays. This complex role of p53 led us to directly examine the effects of p53 status on topoisomerase-induced DNA damage. A deficiency in functional p53 resulted in elevated levels of the Top2 covalent complexes (Top2cc) in multiple cell lines. Employing genome-wide siRNA screens, we identified a set of genes for which reduced expression resulted in enhanced synthetic lethality upon etoposide treatment of p53 defective cells. We focused on one hit from this screen, ATR, and showed that decreased expression sensitized the p53-defective cells to etoposide in all assays and generated elevated levels of Top2cc in both p53 proficient and deficient cells. Our findings suggest that a combination of etoposide treatment with functional inactivation of DNA repair in p53 defective cells could be used to enhance the therapeutic efficacy of Top2 targeting agents.

[1]  N. Osheroff,et al.  Topoisomerase II Poisons: Converting Essential Enzymes into Molecular Scissors. , 2021, Biochemistry.

[2]  Y. Pommier,et al.  A conserved SUMO pathway repairs topoisomerase DNA-protein cross-links by engaging ubiquitin-mediated proteasomal degradation , 2020, Science Advances.

[3]  J. Berger,et al.  Trapped topoisomerase II initiates formation of de novo duplications via the nonhomologous end-joining pathway in yeast , 2020, Proceedings of the National Academy of Sciences.

[4]  Y. Pommier,et al.  Excision repair of topoisomerase DNA-protein crosslinks (TOP-DPC). , 2020, DNA repair.

[5]  Y. Pommier,et al.  Suppressing proteasome mediated processing of topoisomerase II DNA-protein complexes preserves genome integrity , 2020, eLife.

[6]  Kejiao Li,et al.  A Genetic Map of the Response to DNA Damage in Human Cells , 2019, Cell.

[7]  J. Bargonetti,et al.  Gain-of-function mutant p53: history and speculation , 2019, Journal of molecular cell biology.

[8]  R. London,et al.  ZATT (ZNF451)–mediated resolution of topoisomerase 2 DNA-protein cross-links , 2017, Science.

[9]  Scott W. Lowe,et al.  Putting p53 in Context , 2017, Cell.

[10]  D. Menendez,et al.  The Cytidine Deaminase APOBEC3 Family Is Subject to Transcriptional Regulation by p53 , 2017, Molecular Cancer Research.

[11]  Y. Pommier,et al.  Roles of eukaryotic topoisomerases in transcription, replication and genomic stability , 2016, Nature Reviews Molecular Cell Biology.

[12]  Dmitriy Sonkin,et al.  TP53 Variations in Human Cancers: New Lessons from the IARC TP53 Database and Genomics Data , 2016, Human mutation.

[13]  M. DePamphilis,et al.  Identification of genes that are essential to restrict genome duplication to once per cell division , 2016, Oncotarget.

[14]  G. Jackson,et al.  Proteasomal inhibition potentiates drugs targeting DNA topoisomerase II , 2016, Biochemical pharmacology.

[15]  Reham Atteya,et al.  Topoisomerase-mediated chromosomal break repair: an emerging player in many games , 2015, Nature Reviews Cancer.

[16]  Davide Heller,et al.  STRING v10: protein–protein interaction networks, integrated over the tree of life , 2014, Nucleic Acids Res..

[17]  R. Muschel,et al.  Targeting ATR in DNA damage response and cancer therapeutics. , 2014, Cancer treatment reviews.

[18]  Johannes M. Freudenberg,et al.  Diverse stresses dramatically alter genome-wide p53 binding and transactivation landscape in human cancer cells , 2013, Nucleic acids research.

[19]  X. Jacq,et al.  Discovery of 4-{4-[(3R)-3-Methylmorpholin-4-yl]-6-[1-(methylsulfonyl)cyclopropyl]pyrimidin-2-yl}-1H-indole (AZ20): a potent and selective inhibitor of ATR protein kinase with monotherapy in vivo antitumor activity. , 2013, Journal of medicinal chemistry.

[20]  I. Kubacka,et al.  Mutant p53 cooperates with ETS2 to promote etoposide resistance. , 2012, Genes & development.

[21]  Oscar Fernandez-Capetillo,et al.  Targeting ATR and Chk1 kinases for cancer treatment: A new model for new (and old) drugs , 2011, Molecular oncology.

[22]  E. Brown,et al.  Tissue regenerative delays and synthetic lethality in adult mice upon combined deletion of ATR and p53 , 2009, Nature Genetics.

[23]  Y. Pommier DNA topoisomerase I inhibitors: chemistry, biology, and interfacial inhibition. , 2009, Chemical reviews.

[24]  J. Nitiss DNA topoisomerase II and its growing repertoire of biological functions , 2009, Nature Reviews Cancer.

[25]  J. Nitiss Targeting DNA topoisomerase II in cancer chemotherapy , 2009, Nature Reviews Cancer.

[26]  D. Cortez,et al.  ATR: an essential regulator of genome integrity , 2008, Nature Reviews Molecular Cell Biology.

[27]  M. Klagsbrun,et al.  Attenuated p53 activation in tumour-associated stromal cells accompanies decreased sensitivity to etoposide and vincristine , 2008, British Journal of Cancer.

[28]  Jesse D. Martinez,et al.  Different mutant/wild-type p53 combinations cause a spectrum of increased invasive potential in nonmalignant immortalized human mammary epithelial cells. , 2008, Neoplasia.

[29]  Marc Ferrer,et al.  Median Absolute Deviation to Improve Hit Selection for Genome-Scale RNAi Screens , 2008, Journal of biomolecular screening.

[30]  H. Koyama,et al.  Absence of p53 enhances growth defects and etoposide sensitivity of human cells lacking the Bloom syndrome helicase BLM. , 2007, DNA and cell biology.

[31]  M. Olivier,et al.  Impact of mutant p53 functional properties on TP53 mutation patterns and tumor phenotype: lessons from recent developments in the IARC TP53 database , 2007, Human mutation.

[32]  F. Bunz,et al.  Loss of ataxia telangiectasia mutated– and Rad3-related function potentiates the effects of chemotherapeutic drugs on cancer cell survival , 2007, Molecular Cancer Therapeutics.

[33]  C. Chiu,et al.  Etoposide (VP-16) sensitizes p53-deficient human non-small cell lung cancer cells to caspase-7-mediated apoptosis , 2005, Apoptosis.

[34]  A. Sartorelli,et al.  Stable Suppression of the R2 Subunit of Ribonucleotide Reductase by R2-targeted Short Interference RNA Sensitizes p53(–/–) HCT-116 Colon Cancer Cells to DNA-damaging Agents and Ribonucleotide Reductase Inhibitors* , 2004, Journal of Biological Chemistry.

[35]  G. Stark,et al.  G2 arrest in response to topoisomerase II inhibitors: the role of p53. , 2003, Cancer research.

[36]  D. Sullivan,et al.  Tumor p53 status and response to topoisomerase II inhibitors. , 2003, Drug resistance updates : reviews and commentaries in antimicrobial and anticancer chemotherapy.

[37]  B. Kaina,et al.  Resistance of p53 knockout cells to doxorubicin is related to reduced formation of DNA strand breaks rather than impaired apoptotic signaling. , 2003, DNA repair.

[38]  R. Paules,et al.  ATR Enforces the Topoisomerase II-dependent G2 Checkpoint through Inhibition of Plk1 Kinase* , 2002, The Journal of Biological Chemistry.

[39]  S. Schreiber,et al.  ATR Is Not Required for p53 Activation but Synergizes with p53 in the Replication Checkpoint* , 2002, The Journal of Biological Chemistry.

[40]  J. Darbon,et al.  Etoposide and adriamycin but not genistein can activate the checkpoint kinase Chk2 independently of ATM/ATR. , 2001, Biochemical and biophysical research communications.

[41]  N. Pellegata,et al.  Effects of topoisomerase II inhibitors on gastric cancer cells characterized by different genetic lesions. , 2001, Anticancer research.

[42]  K. Kinzler,et al.  Disruption of p53 in human cancer cells alters the responses to therapeutic agents. , 1999, The Journal of clinical investigation.

[43]  S. Inoue,et al.  Differential etoposide sensitivity of cells deficient in the Ku and DNA-PKcs components of the DNA-dependent protein kinase. , 1998, Carcinogenesis.

[44]  G. Zambetti,et al.  Inhibition of DNA topoisomerase II alpha gene expression by the p53 tumor suppressor , 1997, Molecular and cellular biology.

[45]  T. Jacks,et al.  DNA damage can induce apoptosis in proliferating lymphoid cells via p53-independent mechanisms inhibitable by Bcl-2 , 1994, Cell.

[46]  C. Purdie,et al.  Thymocyte apoptosis induced by p53-dependent and independent pathways , 1993, Nature.

[47]  M. Fritsche,et al.  Induction of nuclear accumulation of the tumor-suppressor protein p53 by DNA-damaging agents. , 1993, Oncogene.

[48]  J. Wang,et al.  DNA topoisomerase-targeting antitumor drugs can be studied in yeast. , 1988, Proceedings of the National Academy of Sciences of the United States of America.

[49]  Yilun Sun,et al.  Detection of Topoisomerase Covalent Complexes in Eukaryotic Cells. , 2018, Methods in molecular biology.

[50]  D. J. Clarke,et al.  DNA Topoisomerases , 2009, Methods in Molecular Biology™.

[51]  C. Harris,et al.  p53: traffic cop at the crossroads of DNA repair and recombination , 2005, Nature Reviews Molecular Cell Biology.

[52]  M. Boiocchi,et al.  Pharmacokinetic Optimisation of Treatment with Oral Etoposide , 2004, Clinical pharmacokinetics.

[53]  D. Banerjee,et al.  Effects of wild-type p53 expression on the quantity and activity of topoisomerase IIalpha and beta in various human cancer cell lines. , 1999, Journal of cellular biochemistry.

[54]  Mark T. Muller,et al.  Dna Repair : Possible Relationship with in Vivo I-dna Complex Formation Ultraviolet-induced Dna Damage Stimulates Topoisomerase Updated Version Citing Articles E-mail Alerts Ultraviolet-induced Dna Damage Stimulates Topoisomerase I-dna Complex Formation in Vivo: Possible Relationship with Dna Repair , 2022 .