Your browser doesn't support javascript.
loading
Metabolic reprogramming in triple-negative breast cancer through Myc suppression of TXNIP.
Shen, Liangliang; O'Shea, John M; Kaadige, Mohan R; Cunha, Stéphanie; Wilde, Blake R; Cohen, Adam L; Welm, Alana L; Ayer, Donald E.
Affiliation
  • Shen L; The State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, The Fourth Military Medical University, Xi'an, 710032 China; and Departments of Oncological Sciences and Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112-5550.
  • O'Shea JM; Departments of Oncological Sciences and Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112-5550.
  • Kaadige MR; Departments of Oncological Sciences and Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112-5550.
  • Cunha S; Departments of Oncological Sciences and Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112-5550.
  • Wilde BR; Departments of Oncological Sciences and Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112-5550.
  • Cohen AL; Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112-5550 Medicine and.
  • Welm AL; Departments of Oncological Sciences and Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112-5550.
  • Ayer DE; Departments of Oncological Sciences and Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112-5550 don.ayer@hci.utah.edu.
Proc Natl Acad Sci U S A ; 112(17): 5425-30, 2015 Apr 28.
Article in En | MEDLINE | ID: mdl-25870263
ABSTRACT
Triple-negative breast cancers (TNBCs) are aggressive and lack targeted therapies. Understanding how nutrients are used in TNBCs may provide new targets for therapeutic intervention. We demonstrate that the transcription factor c-Myc drives glucose metabolism in TNBC cells but does so by a previously unappreciated mechanism that involves direct repression of thioredoxin-interacting protein (TXNIP). TXNIP is a potent negative regulator of glucose uptake, aerobic glycolysis, and glycolytic gene expression; thus its repression by c-Myc provides an alternate route to c-Myc-driven glucose metabolism. c-Myc reduces TXNIP gene expression by binding to an E-box-containing region in the TXNIP promoter, possibly competing with the related transcription factor MondoA. TXNIP suppression increases glucose uptake and drives a dependence on glycolysis. Ectopic TXNIP expression decreases glucose uptake, reduces cell proliferation, and increases apoptosis. Supporting the biological significance of the reciprocal relationship between c-Myc and TXNIP, a Mychigh/TXNIPlow gene signature correlates with decreased overall survival and decreased metastasis-free survival in breast cancer. The correlation between the Mychigh/TXNIPlow gene signature and poor clinical outcome is evident only in TNBC, not in other breast cancer subclasses. Mutation of TP53, which is a defining molecular feature of TNBC, enhances the correlation between the Mychigh/TXNIPlow gene signature and death from breast cancer. Because Myc drives nutrient utilization and TXNIP restricts glucose availability, we propose that the Mychigh/TXNIPlow gene signature coordinates nutrient utilization with nutrient availability. Further, our data suggest that loss of the p53 tumor suppressor cooperates with Mychigh/TXNIPlow-driven metabolic dysregulation to drive the aggressive clinical behavior of TNBC.
Subject(s)
Key words

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Breast Neoplasms / Carrier Proteins / Proto-Oncogene Proteins c-myc / Cellular Reprogramming Limits: Female / Humans Language: En Journal: Proc Natl Acad Sci U S A Year: 2015 Type: Article

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Breast Neoplasms / Carrier Proteins / Proto-Oncogene Proteins c-myc / Cellular Reprogramming Limits: Female / Humans Language: En Journal: Proc Natl Acad Sci U S A Year: 2015 Type: Article