Elsevier

Pharmacological Research

Volume 136, October 2018, Pages 45-55
Pharmacological Research

n-3 Polyunsaturated fatty acids induce acute myeloid leukemia cell death associated with mitochondrial glycolytic switch and Nrf2 pathway activation

https://doi.org/10.1016/j.phrs.2018.08.015Get rights and content

Abstract

Acute Myeloid Leukemia (AML) remains a therapeutic challenge and improvements in chemotherapy are needed. n-3 polyunsaturated fatty acids (PUFAs), present in fish oil (FO) at high concentrations, have antitumoral properties in various cancer models. We investigated the effects of two n-3 PUFAs, docosahexaenoic acid (DHA) and eicosapentaenoic acid (EPA), in AML cell lines and primary AML blasts. EPA and DHA induced a dose-dependent decrease in cell viability in five AML cell lines, which was also observed with FO, but not SO (devoid of n-3 PUFAs) in cell lines and primary leucoblasts. Mitochondrial energy metabolism shifted from oxidative respiration to glycolytic metabolism in the U937, MOLM-13, and HL-60 cell lines. This phenomenon was associated with major disorganization of the mitochondrial network and mitochondrial swelling. Transcriptomic analysis after 6 h and 24 h of exposure to FO revealed a Nrf2 activation signature, which was confirmed by evidence of Nrf2 nuclear translocation in response to oxidative stress, but insufficient to prevent cell death following prolonged exposure. Apoptosis studies showed consistent phosphatidylserine exposition among the AML cell lines tested and a reduced mitochondrial membrane potential. The cell-killing effect of FO was additive with that of cytarabine (AraC), by the Chou and Talalay method, and this combination effect could be reproduced in primary AML blasts. Altogether, our results show deleterious effects of n-3 PUFAs on mitochondrial metabolism of AML cells, associated with oxidative stress and Nrf2 response, leading to cell death. These observations support further investigation of n-3 PUFA addition to standard chemotherapy in AML.

Introduction

Acute Myeloid Leukemia(AML) is a heterogeneous group of hematological malignancies for which improvements in therapy are needed, mostly in the adult setting. Upfront standard care for fit patients younger than 60 years is a combination of anthracyclines with cytarabine (AraC), except for specific molecular subgroups, such as FLT3-ITD, for which targeted therapy may be added [1]. Other treatment combinations with anthracyclines and AraC have shown a survival benefit in adult AML, such as gemtuzumab ozogamicin [2] or norethandrolone [3]. Nevertheless, relapses still occur, even with allogeneic transplantation consolidation. AML thus remains an unmet medical need, particularly in the elderly population.

cis-4,7,10,13,16,19-docosahexaenoic acid (DHA) and all-cis-5,8,11,14,17-eicosapentaenoic acid (EPA) are long-chain n-3 polyunsaturated fatty acids (PUFAs) which are found at high levels in fish oil (FO) [4]. Low n-3 PUFA intake has been associated with an increased risk of breast cancer [5], and higher consumption of n-3 PUFAs with lower pancreatic cancer risk [6]. Furthermore, a high level of whole blood n-3 PUFAs was associated with lower prostate cancer risk in 476 men participating in a Scottish study [7]. These beneficial effects of n-3 PUFAs in cancer prevention suggest a potential role for these compounds in cancer therapy.

Preclinical data show that n-3 PUFAs reduce the IC50 of chemotherapy agents in breast cancer cell lines [8,9], sensitize murine mammary cancers to anthracyclines [10], and trigger cytotoxicity in numerous solid cancer models, such as ovarian cancer [11] and colon cancer [12]. Several mechanisms of action of n-3 PUFAs have been described in solid tumor models. Structurally, EPA and DHA incorporate into cancer cell membranes [13] and modify lipid raft composition [14]. They have been shown to inhibit COX-2 and PGE2 pro-inflammatory activity and to be associated with reduced cancer invasion [15], and n-3 PUFAs can inhibit TLR-induced MHC class II upregulation and cytokine production [16]. DHA can also impede I-κB phosphorylation, thus suppressing NF-κB transcription factor activity [17]. The PI3K/AKT signaling pathway is inhibited by DHA in the MDA-MB-231 cell line engrafted to BALB/c nude mice fed a high DHA diet [18]. Evidence of cell arrest in G1-phase has been observed in breast cancer [19]. An anti-angiogenic effect has also been described through the attenuation of VEGF-induced angiogenesis [20].

In acute leukemia, it has been reported that DHA can induce direct in vitro cytotoxicity in the KG1a cell line [21], and cycle arrest in S-phase of the Jurkat cell line [22]. Hallmarks of apoptosis, such as nuclear fragmentation, have been demonstrated in the U937 cell line exposed to 100 μM DHA [21], as well as upregulation of Bax mRNA in the HL-60 cell line [23]. Exposure of HL-60 cells to EPA induces increased fluorescence of H2-DCFDA, suggesting the involvement of reactive oxygen species (ROS) in triggering cell death [24]. Mechanisms involving cytoplasmic calcium release, associated with cell death, have also been demonstrated in U937 cells [25,26]. A ROS-induced mechanism for the sensitization of HL-60 cells to arsenic trioxide by DHA has been described [27] and lymphoid cell lines may be sensitized to other anticancer agents, with an increase in lipid peroxide formation [28].

Whether n-3 PUFAs can induce mitochondrial impairment leading to cell death in AML, is still unclear, and the potential synergism of n-3 PUFAs with chemotherapy has not been yet evaluated. Here, we show that EPA, DHA and clinical-grade FO can induce in vitro cell death of AML cells, associated with phosphatidylserine exposure and a decrease in mitochondrial membrane potential. We demonstrate a mitochondrial metabolic shift and mitochondrial genomic damage, as well as an oxidative stress-related Nrf2 pathway activation, unable to avoid cell death. Furthermore, these effects of FO were additive with the cytotoxicity of AraC.

Section snippets

Leukemic cells and exposure to PUFAs

AML cell lines (U937, KG1a, THP-1, HL-60, ML-2 and MOLM-13) were purchased from DSMZ (Braunschweig, Germany) and incubated at 37 °C and 5% CO2 in RPMI medium with 10% FCS, 2 mM glutamine, 100U/mL penicillin, and 100 μg/mL streptomycin (Thermo Fisher Scientific, MA, USA) for 6 h, 24 h, or 48 h. Primary leucoblasts were obtained from blood samples from patients with hyperleucocytic AML, collected with informed consent, in the context of FILO (French Innovative Leukemia Organization) clinical

DHA or EPA dose-dependently inhibit AML cell growth

Six AML cell lines (KG1a, ML-2, HL-60, THP-1, U937 and MOLM13) were exposed to various concentrations of DHA (from 0 to 50 μM, Fig. 1A) or EPA (from 0 to 150 μM, Fig. 1B) for 48 h. DHA or EPA significantly inhibited cell growth in a dose dependent manner, whereas OA had no effect (Fig. 1A and B). The cell lines showed heterogeneous sensitivity to PUFAs, with a significant decrease in the number of viable cells at 50 μM DHA or 150 μM EPA for KG1a and HL-60 cells, contrasting with only 30 μM DHA

Discussion

In this study we have demonstrated a role for EPA, DHA, and FO in inhibiting the growth of AML cell lines and primary leucoblasts in vitro, associated with a strong decrease of mitochondrial OCR and a compensatory increase in ECAR, along with activation of the oxidative stress-related Nrf2 pathway. Our results are consistent with other studies which found a negative effect of long-chain n-3 PUFAs on AML cell-line viability, such as the inhibition of U937 proliferation following exposure to EPA [

Conclusions

EPA, DHA, and FO induce a dose-dependent mitochondrial glycolytic switch associated with apoptosis and activation of the Nrf2 pathway, leading to cell death both in AML cell lines and primary leucoblasts. This effect is additive with AraC. A phase II clinical trial conducted by the FILO collaborative group is investigating the addition of intravenous FO to induction chemotherapy in high-risk AML (ClinicalTrials.gov Identifier: NCT01999413).

Conflict of interest

Prof. Emmanuel Gyan is the coordinating investigator of the FAMYLY trial (ClinicalTrials.gov Identifier: NCT01999413) studying the role of the addition of a FO emulsion to chemotherapy in adult acute myeloid leukemia, for which funding was received from FRESENIUS KABI. The other authors declare no competing interests.

Acknowledgements

All authors revised the final version of the manuscript. This work was partly supported by the Ligue Nationale Contre le Cancer, the International Rotary Club of Blois and the Laurette Fugain, Sapins de l’Espoir Contre le Cancer, CANCEN, Tours Autogreffe, and AHB associations.

References (53)

  • S. Sriskanthadevan et al.

    AML cells have low spare reserve capacity in their respiratory chain that renders them susceptible to oxidative metabolic stress

    Blood

    (2015)
  • T.C. Chou et al.

    Quantitative analysis of dose-effect relationships: the combined effects of multiple drugs or enzyme inhibitors

    Adv. Enzyme Regul.

    (1984)
  • J. Abdi et al.

    Omega-3 fatty acids, EPA and DHA induce apoptosis and enhance drug sensitivity in multiple myeloma cells but not in normal peripheral mononuclear cells

    J. Nutr. Biochem.

    (2014)
  • C.H. Tsai et al.

    Docosahexaenoic acid increases the expression of oxidative stress-induced growth inhibitor 1 through the PI3K/Akt/Nrf2 signaling pathway in breast cancer cells

    Food Chem. Toxicol.

    (2017)
  • R.M. Stone et al.

    Midostaurin plus chemotherapy for acute myeloid leukemia with a FLT3 mutation

    N. Engl. J. Med.

    (2017)
  • A. Pigneux et al.

    Addition of androgens improves survival in elderly patients with acute myeloid leukemia: a GOELAMS study

    J. Clin. Oncol.

    (2016)
  • J.K. Bassett et al.

    Plasma phospholipids fatty acids, dietary fatty acids, and breast cancer risk

    Cancer Causes Control

    (2016)
  • J.E. Chavarro et al.

    A prospective study of polyunsaturated fatty acid levels in blood and prostate Cancer risk

    Cancer Epidemiol. Biomarkers Prev.

    (2007)
  • K. Mahe et al.
    (2006)
  • G. Gelsomino et al.

    Omega 3 fatty acids chemosensitize multidrug resistant colon cancer cells by down-regulating cholesterol synthesis and altering detergent resistant membranes composition

    Mol. Cancer

    (2013)
  • P.A. Corsetto et al.

    Effects of n-3 PUFAs on breast cancer cells through their incorporation in plasma membrane

    Lipids Health Dis.

    (2011)
  • P.A. Corsetto et al.

    Chemical-physical changes in cell membrane microdomains of breast Cancer cells after Omega-3 PUFA incorporation

    Cell Biochem. Biophys.

    (2012)
  • N. Merendino et al.

    Dietary ω-3 polyunsaturated fatty acid DHA: a potential adjuvant in the treatment of cancer

    Biomed. Res. Int.

    (2013)
  • A.R. Weatherill et al.

    Modulate dendritic cell functions mediated through TLR4 1

    J. Immunol.

    (2005)
  • Y. Wei et al.

    Toll-like receptor alterations in myelodysplastic syndrome

    Leukemia

    (2013)
  • Z. Chen et al.

    mTORC1/2 targeted by n-3 polyunsaturated fatty acids in the prevention of mammary tumorigenesis and tumor progression

    Oncogene

    (2013)
  • Cited by (24)

    • Eicosapentaenoic acid (EPA) exhibits antioxidant activity via mitochondrial modulation

      2022, Food Chemistry
      Citation Excerpt :

      Bearing unsaturated double bonds, EPA can be easily oxidized, which makes it a pro-oxidant compound that promotes the generation of ROS. Several studies have shown that EPA increased ROS and lipid peroxide levels and promoted cancer cell apoptosis (Picou, Debeissat, Bourgeais, Gallay, Ferrié, Foucault, et al., 2018). However, the beneficial effects of EPA may in part be derived from its antioxidant activity.

    • The central role of mitochondria in the relationship between dietary lipids and cancer progression

      2021, Seminars in Cancer Biology
      Citation Excerpt :

      Dietary lipids can exert modulatory effects in cancer cell bioenergetics. N-3 PUFA have shown deleterious effects on mitochondrial metabolism the human acute myeloid leukemia cell lines U937, MOLM-13 and HL-60 [20]. Extracellular acidification rate (ECAR) and oxygen consumption rate (OCR) values found in treated cells suggested that docosahexaenoic acid (DHA) and eicosapentaenoic acid (EPA) or the n-3 polyunsaturated fatty acids (PUFA)-rich fish oil, but not the n-6 PUFA-rich sunflower oil caused shift in mitochondrial energy metabolism from OXPHOS to glycolytic metabolism in the mentioned cell lines along with dose-dependent decrease in cell viability [20].

    • Potential use of n-3 PUFAs to prevent oxidative stress-derived ototoxicity caused by platinum-based chemotherapy

      2020, Free Radical Biology and Medicine
      Citation Excerpt :

      In this sense, n-3 PUFA supplementation in pediatric patients with lymphoblastic leukemia (n = 32) decreased methotrexate-induced hepatotoxicity compared to placebo (n = 33) (p < 0.01) without side effects [13]. Moreover, DHA can increase Nrf2 activity at least in breast cancer [181] and leukemia [182] cells mediating cancer cell apoptosis, suggesting that PUFAs can effectively protect normal cells and enhance cisplatin ROS-induced cell death. Therefore, n-3 PUFA supplementation would also be a feasible and safe strategy in pediatric patients.

    • EPA and DHA have selective toxicity for PBMCs from multiple myeloma patients in a partly caspase-dependent manner

      2020, Clinical Nutrition
      Citation Excerpt :

      In our studies, we have shown that PBMCs from healthy subjects are refractory to the toxic actions of EPA and DHA [26]. Similarly, Picou et al. showed that normal granulocytic cells derived from CD34+ were not affected by coculturing with EPA or DHA [37]. Differences between tumor cells and healthy cells in metabolism may be an important factor in determining their differential sensitivity to n-3 PUFAs.

    View all citing articles on Scopus
    1

    Contributed equally to this manuscript.

    2

    Contributed equally to this manuscript.

    View full text