Cisplatin-induced synthetic lethality to arginine-starvation therapy by transcriptional suppression of ASS1 is regulated by DEC1, HIF-1α, and c-Myc transcription network and is independent of ASS1 promoter DNA methylation

Oncotarget. 2016 Dec 13;7(50):82658-82670. doi: 10.18632/oncotarget.12308.

Abstract

Many human tumors require extracellular arginine (Arg) for growth because the key enzyme for de novo biosynthesis of Arg, argininosuccinate synthetase 1 (ASS1), is silenced. These tumors are sensitive to Arg-starvation therapy using pegylated arginine deiminase (ADI-PEG20) which digests extracellular Arg. Many previous studies reported that ASS1 silencing is due to epigenetic inactivation of ASS1 expression by DNA methylation, and that the demethylation agent 5-aza-deoxycytidine (Aza-dC) can induce ASS1 expression. Moreover, it was reported that cisplatin suppresses ASS1 expression through ASS1 promoter methylation, leading to synthetic lethality to ADI-PEG20 treatment. We report here that cisplatin supppresses ASS1 expression is due to upregulation of HIF-1α and downregulation of c-Myc, which function as negative and positive regulators of ASS1 expression, respectively, by reciprocal bindings to the ASS1 promoter. In contrast, we found that Aza-dC induces ASS1 expression by downregulation of HIF-1α but upregulation of c-Myc. We further demonstrated that the clock protein DEC1 is the master regulator of HIF-1α and c-Myc that regulate ASS1. cDDP upregulates DEC1, whereas Aza-dC suppresses its expression. Using two proteasomal inhibitors bortezomib and carfilzomib which induce HIF-1α accumulation, we further demonstrated that HIF-1α is involved in ASS1 silencing for the maintenance of Arg auxotrophy for targeted Arg-starvation therapy.

Keywords: ASS1; DEC1-HIF-1α-c-Myc axis; DNA methylation; arginine-starvation; cisplatin.

MeSH terms

  • Antineoplastic Combined Chemotherapy Protocols / pharmacology*
  • Arginine / deficiency*
  • Argininosuccinate Synthase / genetics
  • Argininosuccinate Synthase / metabolism*
  • Basic Helix-Loop-Helix Transcription Factors / genetics
  • Basic Helix-Loop-Helix Transcription Factors / metabolism*
  • Cell Line, Tumor
  • Cell Survival / drug effects
  • Cisplatin / pharmacology*
  • DNA Methylation* / drug effects
  • Dose-Response Relationship, Drug
  • Down-Regulation
  • Drug Resistance, Neoplasm
  • Gene Expression Regulation, Neoplastic
  • Histone Deacetylase Inhibitors / pharmacology
  • Homeodomain Proteins / genetics
  • Homeodomain Proteins / metabolism*
  • Humans
  • Hydrolases / pharmacology*
  • Hypoxia-Inducible Factor 1, alpha Subunit / genetics
  • Hypoxia-Inducible Factor 1, alpha Subunit / metabolism*
  • Neoplasms / drug therapy*
  • Neoplasms / enzymology
  • Neoplasms / genetics
  • Neoplasms / pathology
  • Polyethylene Glycols / pharmacology*
  • Promoter Regions, Genetic*
  • Proteasome Inhibitors / pharmacology
  • Proto-Oncogene Proteins c-myc / genetics
  • Proto-Oncogene Proteins c-myc / metabolism*
  • RNA Interference
  • Signal Transduction / drug effects
  • Time Factors
  • Transcription, Genetic / drug effects*
  • Transfection

Substances

  • BHLHE40 protein, human
  • Basic Helix-Loop-Helix Transcription Factors
  • HIF1A protein, human
  • Histone Deacetylase Inhibitors
  • Homeodomain Proteins
  • Hypoxia-Inducible Factor 1, alpha Subunit
  • MYC protein, human
  • Proteasome Inhibitors
  • Proto-Oncogene Proteins c-myc
  • Polyethylene Glycols
  • Arginine
  • Hydrolases
  • ADI PEG20
  • Argininosuccinate Synthase
  • Cisplatin