PMID- 33292639 OWN - NLM STAT- PubMed-not-MEDLINE LR - 20210428 IS - 2049-3002 (Print) IS - 2049-3002 (Electronic) IS - 2049-3002 (Linking) VI - 8 IP - 1 DP - 2020 Dec 4 TI - Protein synthesis inhibitors stimulate MondoA transcriptional activity by driving an accumulation of glucose 6-phosphate. PG - 27 LID - 10.1186/s40170-020-00233-6 [doi] LID - 27 AB - BACKGROUND: Protein synthesis is regulated by the availability of amino acids, the engagement of growth factor signaling pathways, and adenosine triphosphate (ATP) levels sufficient to support translation. Crosstalk between these inputs is extensive, yet other regulatory mechanisms remain to be characterized. For example, the translation initiation inhibitor rocaglamide A (RocA) induces thioredoxin-interacting protein (TXNIP). TXNIP is a negative regulator of glucose uptake; thus, its induction by RocA links translation to the availability of glucose. MondoA is the principal regulator of glucose-induced transcription, and its activity is triggered by the glycolytic intermediate, glucose 6-phosphate (G6P). MondoA responds to G6P generated by cytoplasmic glucose and mitochondrial ATP (mtATP), suggesting a critical role in the cellular response to these energy sources. TXNIP expression is entirely dependent on MondoA; therefore, we investigated how protein synthesis inhibitors impact its transcriptional activity. METHODS: We investigated how translation regulates MondoA activity using cell line models and loss-of-function approaches. We examined how protein synthesis inhibitors effect gene expression and metabolism using RNA-sequencing and metabolomics, respectively. The biological impact of RocA was evaluated using cell lines and patient-derived xenograft organoid (PDxO) models. RESULTS: We discovered that multiple protein synthesis inhibitors, including RocA, increase TXNIP expression in a manner that depends on MondoA, a functional electron transport chain and mtATP synthesis. Furthermore, RocA and cycloheximide increase mtATP and G6P levels, respectively, and TXNIP induction depends on interactions between the voltage-dependent anion channel (VDAC) and hexokinase (HK), which generates G6P. RocA treatment impacts the regulation of ~ 1200 genes, and ~ 250 of those genes are MondoA-dependent. RocA treatment is cytotoxic to triple negative breast cancer (TNBC) cell lines and shows preferential cytotoxicity against estrogen receptor negative (ER-) PDxO breast cancer models. Finally, RocA-driven cytotoxicity is partially dependent on MondoA or TXNIP. CONCLUSIONS: Our data suggest that protein synthesis inhibitors rewire metabolism, resulting in an increase in mtATP and G6P, the latter driving MondoA-dependent transcriptional activity. Further, MondoA is a critical component of the cellular transcriptional response to RocA. Our functional assays suggest that RocA or similar translation inhibitors may show efficacy against ER- breast tumors and that the levels of MondoA and TXNIP should be considered when exploring these potential treatment options. FAU - Wilde, Blake R AU - Wilde BR AD - Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, 84112, USA. AD - Present Address: Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA. FAU - Kaadige, Mohan R AU - Kaadige MR AD - Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, 84112, USA. AD - Present Address: Translational Genomics Research Institute, Phoenix, AZ, 85004, USA. FAU - Guillen, Katrin P AU - Guillen KP AD - Department of Surgery, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, 84112, USA. FAU - Butterfield, Andrew AU - Butterfield A AD - Department of Surgery, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, 84112, USA. FAU - Welm, Bryan E AU - Welm BE AD - Department of Surgery, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, 84112, USA. FAU - Ayer, Donald E AU - Ayer DE AD - Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, 84112, USA. don.ayer@hci.utah.edu. LA - eng GR - R01 CA222650/CA/NCI NIH HHS/United States GR - R01CA222650/CA/NCI NIH HHS/United States GR - U54 CA224076/CA/NCI NIH HHS/United States GR - P30CA042014-31/CA/NCI NIH HHS/United States PT - Journal Article DEP - 20201204 PL - England TA - Cancer Metab JT - Cancer & metabolism JID - 101607582 PMC - PMC7718662 COIS- The authors declare that they have no competing interests. EDAT- 2020/12/10 06:00 MHDA- 2020/12/10 06:01 PMCR- 2020/12/04 CRDT- 2020/12/09 05:45 PHST- 2020/07/01 00:00 [received] PHST- 2020/11/25 00:00 [accepted] PHST- 2020/12/09 05:45 [entrez] PHST- 2020/12/10 06:00 [pubmed] PHST- 2020/12/10 06:01 [medline] PHST- 2020/12/04 00:00 [pmc-release] AID - 10.1186/s40170-020-00233-6 [pii] AID - 233 [pii] AID - 10.1186/s40170-020-00233-6 [doi] PST - epublish SO - Cancer Metab. 2020 Dec 4;8(1):27. doi: 10.1186/s40170-020-00233-6.