4.5 Article

Integration of Downstream Signals of Insulin-like Growth Factor-1 Receptor by Endoplasmic Reticulum Stress for Estrogen-Induced Growth or Apoptosis in Breast Cancer Cells

Journal

MOLECULAR CANCER RESEARCH
Volume 13, Issue 10, Pages 1367-1376

Publisher

AMER ASSOC CANCER RESEARCH
DOI: 10.1158/1541-7786.MCR-14-0494

Keywords

-

Funding

  1. Department of Defense Breast Program Center of Excellence [W81XWH-06-1-0590]
  2. Stand Up To Cancer Dream Team Translational Research Grant [SU2C-AACR-DT0409]
  3. Susan G Komen for the Cure Foundation [SAC100009]
  4. GHUCCTS CTSA [UL1RR031975]
  5. Lombardi Comprehensive Cancer Center Support Grant (CCSG) [NIH P30 CA051008]
  6. NIH MD Anderson's Cancer Center Support Grant [CA016672]

Ask authors/readers for more resources

Estrogen (E2) exerts a dual function on E2-deprived breast cancer cells, with both initial proliferation and subsequent induction of stress responses to cause apoptosis. However, the mechanism by which E2 integrally regulates cell growth or apoptosis-associated pathways remains to be elucidated. Here, E2 deprivation results in many alterations in stress-responsive pathways. For instance, E2-deprived breast cancer cells had higher basal levels of stress-activated protein kinase, c-Jun N-terminal kinase (JNK), compared with wild-type MCF-7 cells. E2 treatment further constitutively activated JNK after 24 hours. However, inhibition of JNK (SP600125) was unable to abolish E2-induced apoptosis, whereas SP600125 alone arrested cells at the G(2) phase of the cell cycle and increased apoptosis. Further examination showed that inhibition of JNK increased gene expression of TNF alpha and did not effectively attenuate expression of apoptosis-related genes induced by E2. A notable finding was that E2 regulated both JNK and Akt as the downstream signals of insulin-like growth factor-1 receptor (IGFIR)/PI3K, but with distinctive modulation patterns: JNK was constitutively activated, whereas Akt and Akt-associated proteins, such as PTEN and mTOR, were selectively degraded. Endoplasmic reticulum-associated degradation (ERAD) was involved in the selective protein degradation. These findings highlight a novel IGFIR/PI3K/JNK axis that plays a proliferative role during the prelude to E2-induced apoptosis and that the endoplasmic reticulum is a key regulatory site to decide cell fate after E2 treatment. (C) 2015 AACR.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.5
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available