Your browser doesn't support javascript.
loading
TXNIP loss expands Myc-dependent transcriptional programs by increasing Myc genomic binding.
Lim, Tian-Yeh; Wilde, Blake R; Thomas, Mallory L; Murphy, Kristin E; Vahrenkamp, Jeffery M; Conway, Megan E; Varley, Katherine E; Gertz, Jason; Ayer, Donald E.
Afiliación
  • Lim TY; Department of Oncological Sciences, Huntsman Cancer Institute, Salt Lake City, Utah, United States of America.
  • Wilde BR; Department of Oncological Sciences, Huntsman Cancer Institute, Salt Lake City, Utah, United States of America.
  • Thomas ML; Department of Oncological Sciences, Huntsman Cancer Institute, Salt Lake City, Utah, United States of America.
  • Murphy KE; Department of Oncological Sciences, Huntsman Cancer Institute, Salt Lake City, Utah, United States of America.
  • Vahrenkamp JM; Department of Oncological Sciences, Huntsman Cancer Institute, Salt Lake City, Utah, United States of America.
  • Conway ME; Department of Oncological Sciences, Huntsman Cancer Institute, Salt Lake City, Utah, United States of America.
  • Varley KE; Department of Oncological Sciences, Huntsman Cancer Institute, Salt Lake City, Utah, United States of America.
  • Gertz J; Department of Oncological Sciences, Huntsman Cancer Institute, Salt Lake City, Utah, United States of America.
  • Ayer DE; Department of Oncological Sciences, Huntsman Cancer Institute, Salt Lake City, Utah, United States of America.
PLoS Biol ; 21(3): e3001778, 2023 03.
Article en En | MEDLINE | ID: mdl-36930677
ABSTRACT
The c-Myc protooncogene places a demand on glucose uptake to drive glucose-dependent biosynthetic pathways. To meet this demand, c-Myc protein (Myc henceforth) drives the expression of glucose transporters, glycolytic enzymes, and represses the expression of thioredoxin interacting protein (TXNIP), which is a potent negative regulator of glucose uptake. A Mychigh/TXNIPlow gene signature is clinically significant as it correlates with poor clinical prognosis in triple-negative breast cancer (TNBC) but not in other subtypes of breast cancer, suggesting a functional relationship between Myc and TXNIP. To better understand how TXNIP contributes to the aggressive behavior of TNBC, we generated TXNIP null MDA-MB-231 (231TKO) cells for our study. We show that TXNIP loss drives a transcriptional program that resembles those driven by Myc and increases global Myc genome occupancy. TXNIP loss allows Myc to invade the promoters and enhancers of target genes that are potentially relevant to cell transformation. Together, these findings suggest that TXNIP is a broad repressor of Myc genomic binding. The increase in Myc genomic binding in the 231TKO cells expands the Myc-dependent transcriptome we identified in parental MDA-MB-231 cells. This expansion of Myc-dependent transcription following TXNIP loss occurs without an apparent increase in Myc's intrinsic capacity to activate transcription and without increasing Myc levels. Together, our findings suggest that TXNIP loss mimics Myc overexpression, connecting Myc genomic binding and transcriptional programs to the nutrient and progrowth signals that control TXNIP expression.
Asunto(s)

Texto completo: 1 Colección: 01-internacional Banco de datos: MEDLINE Asunto principal: Neoplasias de la Mama Triple Negativas Tipo de estudio: Prognostic_studies Límite: Humans Idioma: En Revista: PLoS Biol Asunto de la revista: BIOLOGIA Año: 2023 Tipo del documento: Article País de afiliación: Estados Unidos

Texto completo: 1 Colección: 01-internacional Banco de datos: MEDLINE Asunto principal: Neoplasias de la Mama Triple Negativas Tipo de estudio: Prognostic_studies Límite: Humans Idioma: En Revista: PLoS Biol Asunto de la revista: BIOLOGIA Año: 2023 Tipo del documento: Article País de afiliación: Estados Unidos