Skip to main content

Cystine addiction of triple-negative breast cancer associated with EMT augmented death signaling.

Publication ,  Journal Article
Tang, X; Ding, C-K; Wu, J; Sjol, J; Wardell, S; Spasojevic, I; George, D; McDonnell, DP; Hsu, DS; Chang, JT; Chi, J-T
Published in: Oncogene
July 27, 2017

Despite the advances in the diagnosis and treatment of breast cancer, breast cancers still cause significant mortality. For some patients, especially those with triple-negative breast cancer, current treatments continue to be limited and ineffective. Therefore, there remains an unmet need for a novel therapeutic approach. One potential strategy is to target the altered metabolic state that is rewired by oncogenic transformation. Specifically, this rewiring may render certain outside nutrients indispensable. To identify such a nutrient, we performed a nutrigenetic screen by removing individual amino acids to identify possible addictions across a panel of breast cancer cells. This screen revealed that cystine deprivation triggered rapid programmed necrosis, but not apoptosis, in the basal-type breast cancer cells mostly seen in TNBC tumors. In contrast, luminal-type breast cancer cells are cystine-independent and exhibit little death during cystine deprivation. The cystine addiction phenotype is associated with a higher level of cystine-deprivation signatures noted in the basal type breast cancer cells and tumors. We found that the cystine-addicted breast cancer cells and tumors have strong activation of TNFα and MEKK4-p38-Noxa pathways that render them susceptible to cystine deprivation-induced necrosis. Consistent with this model, silencing of TNFα and MEKK4 dramatically reduces cystine-deprived death. In addition, the cystine addiction phenotype can be abrogated in the cystine-addictive cells by miR-200c, which converts the mesenchymal-like cells to adopt epithelial features. Conversely, the introduction of inducers of epithelial-mesenchymal transition (EMT) in cystine-independent breast cancer cells conferred the cystine-addiction phenotype by modulating the signaling components of cystine addiction. Together, our data reveal that cystine-addiction is associated with EMT in breast cancer during tumor progression. These findings provide the genetic and mechanistic basis to explain how cystine deprivation triggers necrosis by activating pre-existing oncogenic pathways in cystine-addicted TNBC with prominent mesenchymal features.

Duke Scholars

Altmetric Attention Stats
Dimensions Citation Stats

Published In

Oncogene

DOI

EISSN

1476-5594

Publication Date

July 27, 2017

Volume

36

Issue

30

Start / End Page

4235 / 4242

Location

England

Related Subject Headings

  • Triple Negative Breast Neoplasms
  • Signal Transduction
  • Phenotype
  • Oncology & Carcinogenesis
  • Necrosis
  • Humans
  • Female
  • Epithelial-Mesenchymal Transition
  • Cysteine
  • 3211 Oncology and carcinogenesis
 

Citation

APA
Chicago
ICMJE
MLA
NLM
Tang, X., Ding, C.-K., Wu, J., Sjol, J., Wardell, S., Spasojevic, I., … Chi, J.-T. (2017). Cystine addiction of triple-negative breast cancer associated with EMT augmented death signaling. Oncogene, 36(30), 4235–4242. https://doi.org/10.1038/onc.2016.394
Tang, X., C. -. K. Ding, J. Wu, J. Sjol, S. Wardell, I. Spasojevic, D. George, et al. “Cystine addiction of triple-negative breast cancer associated with EMT augmented death signaling.Oncogene 36, no. 30 (July 27, 2017): 4235–42. https://doi.org/10.1038/onc.2016.394.
Tang X, Ding C-K, Wu J, Sjol J, Wardell S, Spasojevic I, et al. Cystine addiction of triple-negative breast cancer associated with EMT augmented death signaling. Oncogene. 2017 Jul 27;36(30):4235–42.
Tang, X., et al. “Cystine addiction of triple-negative breast cancer associated with EMT augmented death signaling.Oncogene, vol. 36, no. 30, July 2017, pp. 4235–42. Pubmed, doi:10.1038/onc.2016.394.
Tang X, Ding C-K, Wu J, Sjol J, Wardell S, Spasojevic I, George D, McDonnell DP, Hsu DS, Chang JT, Chi J-T. Cystine addiction of triple-negative breast cancer associated with EMT augmented death signaling. Oncogene. 2017 Jul 27;36(30):4235–4242.

Published In

Oncogene

DOI

EISSN

1476-5594

Publication Date

July 27, 2017

Volume

36

Issue

30

Start / End Page

4235 / 4242

Location

England

Related Subject Headings

  • Triple Negative Breast Neoplasms
  • Signal Transduction
  • Phenotype
  • Oncology & Carcinogenesis
  • Necrosis
  • Humans
  • Female
  • Epithelial-Mesenchymal Transition
  • Cysteine
  • 3211 Oncology and carcinogenesis