The STAT3-MYC Axis Promotes Survival of Leukemia Stem Cells by Regulating SLC1A5 and Oxidative Phosphorylation

Blood ◽  
2021 ◽  
Author(s):  
Maria Amaya ◽  
Anagha Inguva ◽  
Shanshan Pei ◽  
Courtney L Jones ◽  
Anna Krug ◽  
...  

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 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 TCA metabolites, leading to reduced TCA cycle activity and inhibition of OXPHOS. Based on these findings, we used a novel small molecule STAT3 inhibitor, that 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 and relapsed patients, while sparing normal hematopoietic cells. Together, these findings establish a STAT3-mediated mechanism that controls energy metabolism and survival in primitive AML cells.

Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 429-429
Author(s):  
Courtney L Jones ◽  
Brett M Stevens ◽  
Rachel Culp-Hill ◽  
Travis Nemkov ◽  
Angelo D'Alessandro ◽  
...  

Abstract Most AML patients who receive intensive chemotherapy achieve a significant clinical response; however, the majority will relapse and succumb to their disease, indicating that leukemia stem cells (LSCs) are not effectively targeted. Further, it has recently been shown that LSC frequency and phenotypic diversity are increased at relapse (Ho et al. Blood, 2016), thereby creating an even more challenging clinical scenario. Thus, novel therapies specifically designed to target LSCs in relapsed AML patients are urgently needed. Previously, we have shown that LSCs can be targeted by perturbing energy metabolism (Lagadinou et al. Cell Stem Cell, 2013). Therefore, the goal of the current study was to identify and target metabolic dependencies of relapsed LSCs, with the hope that this would allow improved efficacy for AML patients with relapsed disease. To achieve this objective we first measured metabolic differences in LSCs isolated from de novo and relapsed patients. This analysis revealed that relapsed LSCs have significantly increased levels of nicotinamide compared to de novo LSCs (Figure 1A). Nicotinamide is a precursor of NAD+, an essential coenzyme in energy metabolism. We hypothesized that relapsed LSCs are dependent on nicotinamide metabolism to maintain energy metabolism. To test this hypothesis, we targeted nicotinamide metabolism with the small molecule APO866, an inhibitor of Nampt, the rate-limiting enzyme for conversion of nicotinamide to NAD+. This resulted in a significant decrease in NAD+ in LSCs isolated from both de novo and relapsed AML specimens (data not shown). However, strikingly, inhibition of nicotinamide metabolism only decreased viability and colony-forming ability of LSCs isolated from relapsed AML patients, not LSCs from untreated patients (Figure 1B). To verify that inhibition of Nampt was targeting functional LSCs, we treated a relapsed AML patient specimen with APO866 for 24 hours and measured the ability of the leukemia cells to engraft into immune deficient mice. We observed a significant reduction in leukemia engraftment upon APO866 treatment (data not shown). Importantly, inhibition of nicotinamide metabolism did not affect normal hematopoietic stem cell frequency or colony forming ability (data not shown). Altogether, these data suggest that inhibition of nicotinamide metabolism specifically targets relapsed LSCs. We next sought to understand the mechanism by which inhibiting nicotinamide metabolism targets relapsed LSCs. To this end we measured changes in the major energy metabolism pathways (oxidative phosphorylation [OXPHOS] and glycolysis) in LSCs isolated de novo and relapsed AML patient specimens. Upon APO866 treatment, we observed a significant decrease in OXPHOS and OXPHOS capacity in relapsed LSCs but not de novo LSCs (Figure 1C). Furthermore, no change in glycolysis was observed (data not shown). These data demonstrate that inhibition of nicotinamide metabolism targets OXPHOS specifically in relapsed LSCs. To determine how APO866 reduced OXPHOS, we measured stable isotope metabolic flux of amino acids, the fatty acid palmitate, and glucose into the TCA cycle after APO866 treatment. We observed an increased accumulation of citrate, malate, and α-ketoglutarate from amino acids and palmitate, consistent with decreased activity of the NAD+ dependent enzymes isocitrate dehydrogenase, α-ketoglutarate dehydrogenase and malate dehydrogenase (data not shown). Through direct measurement of enzyme activity, we confirmed that isocitrate dehydrogenase, α-ketoglutarate dehydrogenase and malate dehydrogenase activity were each significantly decreased upon APO866 treatment (Figure 1D). Consistent with our previous findings we did not observe any changes in glycolysis or glucose contribution to the TCA cycle (data not shown). Overall, these data suggest that inhibition of nicotinamide metabolism through Nampt inhibition results in decreased OXPHOS through decreased TCA cycle activity. In conclusion, we have shown that relapsed LSCs have distinct metabolic properties including increased levels of nicotinamide, which can be selectively targeted to eradicate relapsed LSCs. We propose that therapeutic strategies designed to target nicotinamide metabolism may be useful for relapsed AML patients and may allow for broad efficacy such as that observed when LSCs are targeted in the up-front treatment setting. Disclosures Nemkov: Omix Technologies inc: Equity Ownership. Pollyea:Curis: Membership on an entity's Board of Directors or advisory committees; Agios: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding; AbbVie: Consultancy, Research Funding; Celgene: Membership on an entity's Board of Directors or advisory committees; Celyad: Consultancy, Membership on an entity's Board of Directors or advisory committees; Pfizer: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding; Argenx: Consultancy, Membership on an entity's Board of Directors or advisory committees; Gilead: Consultancy; Karyopharm: Membership on an entity's Board of Directors or advisory committees.


Blood ◽  
2019 ◽  
Vol 134 (4) ◽  
pp. 389-394 ◽  
Author(s):  
Courtney L. Jones ◽  
Brett M. Stevens ◽  
Angelo D’Alessandro ◽  
Rachel Culp-Hill ◽  
Julie A. Reisz ◽  
...  

Abstract We have previously demonstrated that oxidative phosphorylation is required for the survival of human leukemia stem cells (LSCs) from patients with acute myeloid leukemia (AML). More recently, we demonstrated that LSCs in patients with de novo AML rely on amino acid metabolism to drive oxidative phosphorylation. Notably, although overall levels of amino acids contribute to LSC energy metabolism, our current findings suggest that cysteine may be of particular importance for LSC survival. We demonstrate that exogenous cysteine is metabolized exclusively to glutathione. Upon cysteine depletion, glutathione synthesis is impaired, leading to reduced glutathionylation of succinate dehydrogenase A (SDHA), a key component of electron transport chain complex (ETC) II. Loss of SDHA glutathionylation impairs ETC II activity, thereby inhibiting oxidative phosphorylation, reducing production of ATP, and leading to LSC death. Given the role of cysteine in driving LSC energy production, we tested cysteine depletion as a potential therapeutic strategy. Using a novel cysteine-degrading enzyme, we demonstrate selective eradication of LSCs, with no detectable effect on normal hematopoietic stem/progenitor cells. Together, these findings indicate that LSCs are aberrantly reliant on cysteine to sustain energy metabolism, and that targeting this axis may represent a useful therapeutic strategy.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 845-845 ◽  
Author(s):  
Haobin Ye ◽  
Biniam Adane ◽  
Nabilah Khan ◽  
John M. Ashton ◽  
Marlene Balys ◽  
...  

Abstract Adipose tissue (AT) serves as a storage site for lipids as well as an endocrine organ. In the context of cancer, AT can function to facilitate the progression of tumors. Interestingly, recent studies have shown that AT acts as an extramedullary reservoir of hematopoietic stem cells (HSCs), suggesting the presence of a HSC niche in AT. The fact that leukemia stem cells (LSCs) co-opt the HSC marrow and the supportive effect of AT on cancer cells led us to hypothesize that AT functions as a reservoir for LSCs. To test this hypothesis, we first examined whether there were LSCs residing in AT. Using a murine model of primary blast crisis CML, we found enrichment of phenotypically primitive leukemia cells (PLCs) in the gonadal adipose tissue (GAT) relative to bone marrow, spleen and peripheral blood (Figure 1). The high percentage of PLCs in GAT led us to postulate that PLCs are preferentially reliant on fatty acids as an energy source since GAT was found to be lipolytic in leukemic mice. Indeed, we observed that PLCs had a higher rate of fatty acid oxidation (FAO) compared to lineage+ leukemia cells and their non-leukemic counterparts. Additionally, conditioned medium from adipocytes selectively increased the FAO rate in PLCs suggesting a FAO regulatory role of AT. Further, we showed that CD36, a fatty acid transporter, was highly expressed by a subset of PLCs and selectively regulated FAO in PLCs. Interestingly, CD36+ PLCs were strikingly enriched in GAT. Together, these results suggested that GAT functions as a reservoir for CD36+ PLCs and facilitates use of FAO for energy metabolism. Next we examined the characteristics of CD36+ PLCs. CD36+ PLCs differed metabolically from CD36- PLCs with regard to fatty acid metabolism. Specifically, we found CD36+ PLCs had a higher FAO rate and were more dependent on the transportation function of CD36 for FAO (Figure 2). We also compared the cell cycle status between these two populations and found CD36+ PLCs were more quiescent. Interestingly, both CD36+ and CD36- PLCs contained LSCs and were able to reconstitute the whole leukemic BM when transplanted into recipients. Collectively, these findings indicated that at least two metabolically distinct types of leukemia-initiating cells exist in our blast crisis model, and that the major forms of energy metabolism can differ as a function of anatomical location and expression of CD36. Since front line agents commonly used in cancer generally target actively cycling cells, we speculated that CD36+ PLCs might be more drug resistant due to their increased quiescence. Indeed, we found enrichment of CD36+ PLCs in BM after applying a 5-day chemotherapy regimen to leukemic mice, while CD36- PLCs were not protected. More importantly, we observed that CD36+ PLCs were highly enriched in GAT after chemotherapy suggesting GAT conferred chemo-resistance to resident CD36+ PLCs (Figure 3). Taken together, our observations imply that the heterogeneity found in PLCs is translated into drug sensitivities in different leukemic sub-fractions. Specifically, CD36+ PLCs represent a relatively drug resistant subpopulation and GAT serves as a reservoir for resident CD36+ PLCs. Lastly, we investigated whether these findings in the murine model could be recapitulated in primary human bcCML cells. We found that within the CD34+ subpopulation of primary human bcCML cells, CD36+ subset had a high FAO rate compared to CD36- subset. Furthermore, this CD36+ subset was more quiescent and drug resistant. These data suggest that a similar heterogeneity can be found in primary human bcCML cells based on the expression of CD36. Collectively, our findings suggested that GAT in leukemia mice functions as a reservoir for LSCs and confers chemo-resistance to resident leukemia cells, implying a potential role of GAT in the pathogenesis of leukemia and relative efficacy of therapeutic challenge. Furthermore, our data indicate metabolic heterogeneity within LSC populations, where pathways controlling energy consumption can differ. We propose that metabolic heterogeneity in LSCs may contribute to the challenge in effectively eradicating such cells. Disclosures No relevant conflicts of interest to declare.


2020 ◽  
Vol 21 (15) ◽  
pp. 5276 ◽  
Author(s):  
Ge Liu ◽  
Qing Luo ◽  
Hong Li ◽  
Qiuping Liu ◽  
Yang Ju ◽  
...  

Cancer stem cells (CSCs) are considered to be the main cause of tumor recurrence, metastasis, and an unfavorable prognosis. Energy metabolism is closely associated with cell stemness. However, how the stemness of liver cancer stem cells (LCSCs) is regulated by metabolic/oxidative stress remains poorly understood. In this study, we compare the metabolic differences between LCSCs and the hepatocellular carcinoma cell line HCCLM3, and explore the relationship between metabolism and LCSC stemness. We found that LCSCs from the hepatocellular carcinoma cell HCCLM3 exhibited more robust glucose metabolism than HCCLM3, including glycolysis, oxidative phosphorylation (OXPHOS), and pyruvate produced by glycolysis entering mitochondria for OXPHOS. Moreover, 2-deoxy-D-glucose (2-DG) enhanced the LCSC stemness by upregulating OXPHOS. In contrast, Mdivi-1 reduced the levels of OXPHOS and weakened the stemness by inhibiting mitochondrial fission. Together, our findings clarify the relationship between energy metabolism and LCSC stemness and may provide theoretical guidance and potential therapeutic approaches for liver cancer.


Cancers ◽  
2020 ◽  
Vol 12 (7) ◽  
pp. 1731 ◽  
Author(s):  
Carina Neitzel ◽  
Philipp Demuth ◽  
Simon Wittmann ◽  
Jörg Fahrer

Colorectal cancer (CRC) is among the most frequent cancer entities worldwide. Multiple factors are causally associated with CRC development, such as genetic and epigenetic alterations, inflammatory bowel disease, lifestyle and dietary factors. During malignant transformation, the cellular energy metabolism is reprogrammed in order to promote cancer cell growth and proliferation. In this review, we first describe the main alterations of the energy metabolism found in CRC, revealing the critical impact of oncogenic signaling and driver mutations in key metabolic enzymes. Then, the central role of mitochondria and the tricarboxylic acid (TCA) cycle in this process is highlighted, also considering the metabolic crosstalk between tumor and stromal cells in the tumor microenvironment. The identified cancer-specific metabolic transformations provided new therapeutic targets for the development of small molecule inhibitors. Promising agents are in clinical trials and are directed against enzymes of the TCA cycle, including isocitrate dehydrogenase, pyruvate dehydrogenase kinase, pyruvate dehydrogenase complex (PDC) and α-ketoglutarate dehydrogenase (KGDH). Finally, we focus on the α-lipoic acid derivative CPI-613, an inhibitor of both PDC and KGDH, and delineate its anti-tumor effects for targeted therapy.


2018 ◽  
Vol 24 (12) ◽  
pp. 1859-1866 ◽  
Author(s):  
Daniel A. Pollyea ◽  
Brett M. Stevens ◽  
Courtney L. Jones ◽  
Amanda Winters ◽  
Shanshan Pei ◽  
...  

Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 484-484 ◽  
Author(s):  
Carolina Schinke ◽  
Orsolya Giricz ◽  
Shanisha A. K. Gordon ◽  
Laura Barreyro ◽  
Tushar D. Bhagat ◽  
...  

Abstract Acute Myeloid Leukemia (AML) and Myelodysplastic syndrome (MDS) arise from accumulation of multiple stepwise genetic and epigenetic changes in hematopoietic stem cells (HSC) and/or committed progenitors. A series of transforming events can initially give rise to pre-leukemia stem cells (pre-LSC) as well as fully transformed leukemia stem cells (LSC), both of which need to be targeted in strategies aimed at curing these diseases. We conducted parallel transcriptional analysis of multiple, highly fractionated stem and progenitor populations in individual patients of MDS and AML (N=16) and identified candidate genes that are consistently dysregulated at multiple immature stem and progenitor cell stages. Interleukin 8 (IL8), was one of the most consistently overexpressed genes in MDS/AML Hematolpoetic Stem Cells (HSCs) and progenitors when compared to healthy control HSCs and progenitors. IL8 is a pro-inflammatory chemokine, which is able to activate multiple intracellular signaling pathways after binding to its surface receptor CXCR2. Even though increased IL8-CXCR2 signaling has been shown to promote angiogenesis, metastasis and chemotherapy resistance in many solid tumors, its role in AML and MDS is not well elucidated. We further analyzed gene expression profiles of CD34+ cells from 183 MDS patients and found significant increased expression of CXCR2 in MDS when compared to healthy controls (FDR<0.1). Most importantly, analysis of The Cancer Genome Atlas (TCGA) AML (n=200) dataset showed that CXCR2 expression was predictive of significantly adverse prognosis (log rank P value=0.0182; median survival of 245 days in cxcr2 high vs 607 days in cxcr2 low) in patients, further pointing to a critical role of IL8-CXCR2 signaling in AML/MDS. Next, we studied the functional role of IL8 and CXCR2 in AML. A panel of leukemic cell lines (THP-1, U937, KG-1, MOLM13, HL-60, K532) were screened for CXCR2 expression and revealed significantly higher expression when compared to healthy CD34+ control cells. SB-332235, a specific inhibitor of CXCR2 was used for functional studies. CXCR2 inhibition led to significant, (p<0.05) reduction in proliferation in all 6 cell lines tested and an effect was seen as early as 24 hrs of exposure. CXCR2 inhibition was found to lead to G0/G1 cell cycle arrest and trigged apoptosis in THP-1 and U937 cells (p-value 0.004 and 0.02 respectively). Incubation of primary AML/MDS bone marrow samples with SB-332235 similarly lead to significantly reduced proliferation at 24hrs, when compared to healthy CD34+ cells. Selective, and highly significant inhibition of leukemic cell growth was also seen in colony assays from primary MDS/AML samples (mean leukemic colonies in AML/MDS= 73 vs 313 in controls, P < 0.001). Interestingly, inhibition of CXCR2 in primary AML marrow samples led to induction of apoptosis in immature CD34+/CD38- cells when compared to healthy controls. Lastly, xenografting studies with THP-1 leukemic cells revealed that CXCR2 inhibitor treatment led to decreased leukemic burden and organ infiltration when compared to placebo controls in vivo. In summary we have found significantly increased expression of IL8 and its receptor CXCR2 in sorted HSCs and progenitors from AML and MDS patients. High CXCR2 expression was a marker of adverse prognosis in a large cohort of AML patients. Most importantly, in vitro and in vivo functional studies showed that CXCR2 is a potential therapeutic target in AML/MDS and is able to selectively target immature, LSC-enriched cell fractions in AML. Disclosures: No relevant conflicts of interest to declare.


Oncotarget ◽  
2010 ◽  
Vol 1 (2) ◽  
pp. 156-160 ◽  
Author(s):  
Cong Peng ◽  
Yaoyu Chen ◽  
Dongguang Li ◽  
Shaoguang Li

Sign in / Sign up

Export Citation Format

Share Document