4.7 Article

The STAT3-MYC axis promotes survival of leukemia stem cells by regulating SLC1A5 and oxidative phosphorylation

Journal

BLOOD
Volume 139, Issue 4, Pages 584-596

Publisher

AMER SOC HEMATOLOGY
DOI: 10.1182/blood.2021013201

Keywords

-

Categories

Funding

  1. National Cancer Institute, National Institutes of Health [R01CA200707, R35CA242376, P30CA046934, T32CA190216-4]
  2. National Cancer Institute, National Institutes of Health (Ruth L. Kirschstein National Research Service Award) [T32CA190216]
  3. Colorado Clinical and Translational Sciences Institute [TL1TR002533]
  4. Morgan Adams Foundation
  5. Leukemia and Lymphoma Society's Scholar in Clinical Research
  6. Robert H. Allen Chair in Hematology Research
  7. Leukemia and Lymphoma Society Specialized Center of Research (SCOR) grant
  8. Nancy Carroll Allen Endowed Chair

Ask authors/readers for more resources

This study reveals that acute myeloid leukemia (AML) stem cells rely on oxidative phosphorylation (OXPHOS) for energy metabolism and survival, which is mediated by signal transducer and activator of transcription 3 (STAT3). STAT3 regulates AML-specific expression of MYC and SLC1A5, and inhibition of SLC1A5 acts similarly to STAT3 inhibition by impairing OXPHOS. Furthermore, a novel small molecule STAT3 inhibitor selectively induces cell death in AML stem cells while sparing normal hematopoietic cells.
Acute myeloid leukemia (AML) is characterized by the presence of leukemia stem cells (LSCs), and failure to fully eradicate this population contributes to disease persistence/ relapse. Prior studies have characterized metabolic vulnerabilities of LSCs, which demonstrate preferential reliance on oxidative phosphorylation (OXPHOS) for energy metabolism and survival. In the present study, using both genetic and pharmacologic strategies in primary human AML specimens, we show that signal transducer and activator of transcription 3 (STAT3) mediates OXPHOS in LSCs. STAT3 regulates AML-specific expression of MYC, which in turn controls transcription of the neutral amino acid transporter gene SLC1A5. We show that genetic inhibition of MYC or SLC1A5 acts to phenocopy the impairment of OXPHOS observed with STAT3 inhibition, thereby establishing this axis as a regulatory mechanism linking STAT3 to energy metabolism. Inhibition of SLC1A5 reduces intracellular levels of glutamine, glutathione, and multiple tricarboxylic acid (TCA) cycle metabolites, leading to reduced TCA cycle activity and inhibition of OXPHOS. Based on these findings, we used a novel small molecule STAT3 inhibitor, which binds STAT3 and disrupts STAT3-DNA, to evaluate the biological role of STAT3. We show that STAT3 inhibition selectively leads to cell death in AML stem and progenitor cells derived from newly diagnosed patients and patients who have experienced relapse while sparing normal hematopoietic cells. Together, these findings establish a STAT3-mediated mechanism that controls energy metabolism and survival in primitive AML cells.

Authors

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

Reviews

Primary Rating

4.7
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available