Purpose: The intracellular redox environment of acute myeloid leukemia (AML) cells is often highly oxidized compared to healthy hematopoietic progenitors and this is purported to contribute to disease pathogenesis. However, the redox regulators that allow AML cell survival in this oxidized environment remain largely unknown.

Experimental Design: Utilizing several chemical and genetically-encoded redox sensing probes across multiple human and mouse models of AML, we evaluated the role of the serine/threonine kinase PKC-epsilon (PKCϵ) in intracellular redox biology, cell survival and disease progression.

Results: We show that RNA interference-mediated inhibition of PKCϵ significantly reduces patient-derived AML cell survival as well as disease onset in a genetically engineered mouse model (GEMM) of AML driven by MLL-AF9. We also show that PKCϵ inhibition induces multiple reactive oxygen species (ROS) and that neutralization of mitochondrial ROS with chemical antioxidants or co-expression of the mitochondrial ROS-buffering enzymes SOD2 and CAT, mitigates the anti-leukemia effects of PKCϵ inhibition. Moreover, direct inhibition of SOD2 increases mitochondrial ROS and significantly impedes AML progression in vivo. Furthermore, we report that PKCϵ over-expression protects AML cells from otherwise-lethal doses of mitochondrial ROS-inducing agents. Proteomic analysis reveals that PKCϵ may control mitochondrial ROS by controlling the expression of regulatory proteins of redox homeostasis, electron transport chain flux, as well as outer mitochondrial membrane potential and transport.

Conclusions: This study uncovers a previously unrecognized role for PKCϵ in supporting AML cell survival and disease progression by regulating mitochondrial ROS biology and positions mitochondrial redox regulators as potential therapeutic targets in AML. Clin Cancer Res; 24(3); 608–18. ©2017 AACR.

Translational Relevance

Patient-derived AML cells often display significantly higher levels of intracellular ROS compared with their normal counterparts. Elevated ROS levels are often associated with increased DNA damage, dysfunctional organelle biology, oncogenic signaling, and altered cellular metabolism and thus are purported to contribute to disease pathogenesis and therapeutic responses in a variety of human tumor settings. However, excess ROS can also promote tumor cell death and therefore intracellular ROS are tightly maintained by various regulatory mechanisms. Therefore, regulators of redox biology may provide opportunities for therapeutic intervention. We have revealed that PKCϵ suppresses mitochondrial ROS to support AML and that blocking PKCϵ or enzymes that directly neutralize mitochondrial ROS, such as SOD2, diminishes AML cell survival by inducing lethal ROS levels.

The intracellular redox environment is largely influenced by the production and clearance of reactive species such as reactive oxygen species (ROS). ROS encompass a heterogeneous class of small oxygen-containing reactive species that are produced by several intracellular processes such as aerobic glucose metabolism and enzymatic reactions (1) and contribute to physiologic functions such as innate and acquired immune defense. However, aberrantly elevated ROS levels, referred to as “oxidative stress,” can cause DNA lesions, organelle dysfunction, and metabolic alterations that contribute to tumorigenesis (2). Furthermore, extremely high ROS levels can promote cell death (2). Therefore, intracellular ROS levels are tightly regulated by an array of enzymatic and nonenzymatic systems (1).

Several human cancers, including AML, display perturbations in genes that encode regulators of intracellular ROS biology (3). For example, primary AML patient samples display decreased expression of the mitochondrial superoxide-neutralizing gene SOD2 and decreased glutathione metabolism (4). The activities of the ROS-generating enzymes NADPH-oxidases (NOX) are also elevated in primary human AML samples compared with healthy controls and this is associated with increased steady-state levels of intracellular superoxides (5). Moreover, the total antioxidant capacity of leukemic cells from AML patients at initial diagnosis and relapse is decreased compared with healthy controls (6). Several of the oncogenic signaling molecules that are either mutated (e.g., FLT3ITD, KRASG12D or BCR-ABL1; refs. 7–12) or dysregulated (e.g., c-MYC; ref. 13) in myeloid malignancies are known to drive ROS production. Furthermore, small nucleotide polymorphisms (SNP) of several redox-regulatory enzymes have been identified to be associated with myeloid neoplasia susceptibility and prognosis (14). Despite these studies as well as the emerging evidence that elevated ROS levels promote the proliferation and survival of solid cancers (15, 16), the underlying molecular mechanisms that govern ROS biology in AML remain unresolved.

PKCϵ is a serine/threonine kinase that is expressed in numerous tissues and supports tumorigenesis in many solid cancers (17). In normal hematopoietic development, PKCϵ influences erythrocyte and megakaryocyte lineage commitments (18); however, in primary myelofibrosis, elevated PKCϵ expression antagonizes megakaryocytic differentiation (19). Furthermore, downmodulation of PKCϵ expression is a key event during phorbol ester–induced differentiation of primary human AML samples and increased PKCϵ expression protects AML cells from TRAIL-induced apoptosis (20). However, the molecular mechanism(s) by which PKCϵ influences AML cell fate has yet to be resolved.

Here, we show that PKCϵ inhibition obstructs disease progression in a GEMM of AML driven by MLL-AF9 and impairs cell survival in multiple patient-derived AML samples. Furthermore, we find that PKCϵ inhibition results in increased steady-state levels of multiple mitochondrial ROS and that chemical or genetic neutralization of mitochondrial ROS counteracts the antileukemia effects of PKCϵ. Furthermore, we report that overexpression of PKCϵ protects AML cells from otherwise lethal doses of mitochondrial ROS–inducing agents. Finally, direct inhibition of the mitochondrial ROS–neutralizing enzyme SOD2 phenocopies PKCϵ inhibition indicating that maintenance of mitochondrial ROS homeostasis is crucial to the maintenance of AML pathogenesis.

Cell culture

Human AML cell lines were obtained from the ATCC and the German Collection of Microorganism and Cell Cultures (DMSZ). Cells were cultured in the recommended media conditions. Murine AML cells were cultured in cytokine-enriched media (CEM): RPMI1640 supplemented with 10% FBS and penicillin/streptomycin, 10 ng/mL mSCF (Peprotech), 6 ng/mL mIL3 (Peprotech), and 5 ng/ml mIL6 (Peprotech).

Patient-derived AML samples

Patient-derived AML samples were obtained from The Ohio State Comprehensive Cancer Center and used according to the approved Institutional Review Board protocol 16-9037. Cells were plated on irradiated monolayers of HS27 cells (21) and cultured in Stemspan (Stem Cell Technology) supplemented with 10% FBS, 100 ng/mL hSCF (Peprotech), 100ng/ml hFLT3 ligand (Peprotech), 20ng/ml hIL-3 (Peprotech), 20ng/mL hIL6 (Peprotech), 20 ng/mL G-CSF (Peprotech). Cells (5 × 105) were transduced with pLKO.1 GFP lentiviral shRNA vectors and evaluated for GFP expression every 3 days for 12 days after staining with human CD45 APC-Cy7 (BD Biosciences) and propidium iodide (PI) by flow cytometry.

Lentiviral transduction

A total of 5 × 105 human AML cells were transduced for 30 hours with recombinant pLKO.1 lentiviruses coexpressing either GFP or a puromycin-resistant cassette with shRNAs from the TRC shRNA library (shRNA Pkcϵ: TRCN0000022759; shRNA PKCϵ_1: TRCN0000000848 and shRNA PKCϵ_2: TRCN0000000846: shRNA SOD2: TRCN0000350349; shRNA Sod2_1: TRCN0000123392; shRNA Sod2_2: TRCN0000123390). Cells transduced with recombinant lentiviruses expressing the puromycin-resistant cassette were initially selected with 2 μg/mL puromycin for 48 or 72 hours (MOI = 0.5–0.9) and thereafter maintained in 0.5 μg/mL puromycin. To generate pLKO.1 GFP vectors, the puromycin-resistant cassette was replaced with eGFP. Cells transduced with lentiviruses expressing GFP (MOI = 0.2–0.5) were purified by FACS at the indicated times following transduction.

Retroviral transduction

A total of 5 × 105 human or murine AML cells were subjected to spin transduction with recombinant retroviruses (MOI = 0.2–0.3). The MSCV.PKCϵ.GFP construct was obtained by cloning murine Pkcϵ into the BglII and XhoI sites of the MSCV.IRES.GFP vector. Cells transduced with vectors expressing GFP were purified by FACS 96 hours after transduction. SF91-IRES-eGFP and SF91-SOD2/Catalase-IRES-eGFP vectors, were described previously (22) and provided by M. Milsom. SF91-Grx1-roGFP2, SF91-Mito-Grx1-roGFP2, SF91-roGFP2-Orp1, and SF91-Mito-roGFP2-Orp1 were described previously (23) and provided by Dr. T. Dick (German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, Heidelberg, Germany).

Western blot analysis

Cell pellets were lysed and resolved on 4%–12% Bis Tris gels (Thermo Fisher Scientific; Life Technologies). Following protein transfer and blocking with 5% nonfat milk, blots were incubated with primary antibody overnight at 4°C. Secondary antibody was incubated at room temperature for 1 hour and blots were developed with ECL prime (GE Healthcare). The following antibodies were used: anti-PKCϵ (catalog no.: 2683S: lot no.: 4) and anti-Catalase (catalog no.: 12980S, lot no.: 1) from Cell Signaling Technology with the dilution 1:1,000, anti-SOD2 (catalog no.: ADI-SOD-111, lot no.: 08021202) from Enzo Life Sciences with the dilution 1:1,000, anti-α-tubulin (catalog no.: T9026, lot no.: 083M4847V) from Sigma-Aldrich with the dilution 1:5,000. Secondary antibodies anti-rabbit-HRP (catalog no.: 7074S, lot no.: 26) and anti-mouse-HRP (catalog no.: 7076S, lot no.: 31) were from Cell Signaling Technology with the dilution 1:4,000.

MTS assay

Cells were plated at the density of 1–2 × 105 cells/mL at either 72 or 96 hours posttransduction (day 0). At days 0, 2, and 4, 100 μL of cell suspension was incubated with 20 μL of CellTiter Aqueous One solution (Promega) for 90 minutes. Plates were read at the wavelength of 495 nm. Culture media was used as blank.

Apoptosis assay

Cells were washed in PBS and stained with Annexin V and Propidium Iodide (PI) or 7AAD according to the manufacturer's instruction (BD Biosciences). Cells were acquired and analyzed using an LSRII flow cytometer (Beckton Dickinson). All the data were analyzed using FlowJo software.

CellROX and MitoSOX Staining

Human AML cell lines were washed and incubated with PBS + 5 μmol/L of CellROX DeepRed reagent (Life Technologies; catalog no.: C10422) at 37°C for 30 minutes, or with PBS + 5 μmol/L of MitoSOX Red reagent (Life Technologies; catalog no.: M36008) at 37°C for 10 minutes. After incubation, cells were washed two times in PBS and stained with Annexin V-APC according to the manufacturer's instructions (BD Biosciences). Cells were analyzed using an LSRII flow cytometer (Beckton Dickinson).

roGFP analysis

Human AML cell lines stably expressing the roGFP2 probes (27) were transduced with recombinant lentiviruses for 30 hours, then selected with 2 μg/μL puromycin for 72 hours and subsequently analyzed by flow cytometry. RoGFP2 is excited at 400 nm (oxidized state) and 475–490 nm (reduced state) when fluorescence emission is monitored at 510 nm. Diamide (DIA; 1 mmol/L) or dithiothreitol (DTT; 1 mmol/L) were used as controls to induce a complete oxidized or reduced state of the probes, respectively. Propidium iodide staining was performed to exclude dead cells from the analysis.

Pro-oxidant treatment

Human AML cells stably expressing PKCϵ, PKCϵ-targeting shRNAs, or corresponding controls were seeded at the concentration of 2 × 105 cells/mL in 24-well plates and treated with the indicated concentrations of Antimycin A (AA; Sigma Aldrich A8674) and Thenoyltrifluoroacetone (TTFA, Sigma Aldrich T27006) for 24 hours and then analyzed by flow cytometry after Annexin V and 7AAD staining.

Bone marrow transplant leukemia model

All animal studies conducted were approved by the IUCAC of the Fox Chase Cancer Center. For recombinant viral transduction, bone marrow cells recovered from leukemia mice were cultured in CEM media overnight. The next day, cells were counted and then 5 × 105 cells were subjected to spin transduction with recombinant lentiviruses (MOI = 0.4–0.6) or retroviruses (MOI = 0.2–0.3) supplemented with polybrene (5 μg/mL) in 12-well nonadherent plates. Plates were centrifuged at 2,400 rpm at 30 °C for 90 minutes. Transduced cells were then incubated overnight. The following day, viral supernatants were removed from cells and replenished with fresh CEM. For in vivo survival assays, sorted GFP+ cells (1 × 106 cells/mouse) were transplanted into sublethally irradiated (450 rad) syngenic recipient mice 48 hours post-transduction. For Western blot analysis and colony formation assays, transduced cells were subjected to FACS to isolate GFP+ cells 48 hours post-transduction. For colony formation assays, 500 purified GFP+ leukemia cells were cultured in 1 mL of methylcellulose supplemented with cytokines (M3434, StemCell Technologies) for 5–7 days.

PKCϵ inhibition impairs in vitro and in vivo AML cell expansion and survival

To evaluate the functional role of PKCϵ expression in AML biology, we employed an shRNA approach in a panel of genetically distinct AML cell lines in vitro (OCI-AML3, THP-1, NOMO1, and U937). We identified two shRNA constructs, PKCϵ shRNA_1 and PKCϵ shRNA_2, which target distinct regions of the PKCϵ mRNA and effectively deplete PKCϵ protein levels (Supplementary Fig. S1A). Each of these PKCϵ-targeting shRNAs significantly reduced the expansion of the four AML cell lines compared with nontargeting shRNA controls (CTRL shRNA; Supplementary Fig. S1B). This reduction in cell growth was accompanied by a significant increase in the percentage of Annexin V+ cells (Supplementary Fig. S1C), and CD11b expression (Supplementary Fig. S1D and S1E) indicating that PKCϵ inhibition is detrimental to survival and growth and may induce differentiation of these AML cell lines.

To assess the impact of PKCϵ inhibition in vivo, we utilized a GEMM of AML that is driven by the human leukemogenic fusion protein MLL-AF9 (24). Mouse MLL-AF9 leukemia cells were transduced with recombinant lentiviruses that coexpress GFP in combination with either control (CTRL shRNA) or murine Pkcϵ-targeting shRNAs (Pkcϵ shRNA). The transduced cells were subsequently purified by FACS to assess protein expression, cell growth in vitro and leukemia induction in vivo (Supplementary Figs. S2A and S2B). Depletion of Pkcϵ protein significantly reduced the growth of mouse MLL-AF9 leukemia cells in cytokine-enriched liquid culture (Fig. 1A and B; Supplementary Fig. S2C–S2E). Furthermore, mice transplanted with FACS-purified mouse MLL-AF9 leukemia cells expressing Pkcϵ shRNA exhibited a significantly longer onset of disease compared with CTRL shRNA–expressing cells (Fig. 1C). Furthermore, depletion of Pkcϵ protein significantly reduced the colony-forming capacity (CFC) of mouse MLL-AF9 leukemia cells in cytokine-enriched methylcellulose (Fig. 1D).

Figure 1.

PKCϵ inhibition impairs AML cell expansion and survival in vitro and in vivo. A, Western blot analysis of mouse MLL-AF9 leukemia cells transduced with CTRL or Pkcϵ shRNA. B,In vitro competitive growth curve of mouse MLL-AF9 cells transduced with either CTRL or Pkcϵ shRNA GFP lentiviruses. %GFP+ cells were evaluated every two days by flow cytometry and normalized to fold change in %GFP+ at day 3 post-transduction, which represents day 0 in the figure (day 6 = day 9 post-transduction). C, Kaplan–Meier survival curve analysis of mice transplanted with mouse MLL-AF9 leukemia cells coexpressing GFP and either CTRL or Pkcϵ shRNAs (P = 0.0014; n = 7). MLL-AF9 (D) and MLL-AF9;Flt3ITD knock in (KI; E) cells transduced with lentiviruses coexpressing GFP with either CTRL or Pkcϵ shRNA were FACS-purified and plated in M3434. Dnmt3a−/−;Tet2−/− (F) and Dnmt3a−/−;Tet2−/−;FLT3ITD cells (G) transduced with lentiviruses coexpressing RFP with either CTRL or Pkcϵ shRNA were FACS-purified and plated in M3434. D–G, Bar graph representing the number of colonies formed by mouse MLL-AF9 leukemia cells expressing CTRL or Pkcϵ shRNAs in methylcellulose culture. Data are represented as the mean ± SD of three technical replicates for B and D–G (***, P ≤ 0.001; ***, P ≤ 0.0001).

Figure 1.

PKCϵ inhibition impairs AML cell expansion and survival in vitro and in vivo. A, Western blot analysis of mouse MLL-AF9 leukemia cells transduced with CTRL or Pkcϵ shRNA. B,In vitro competitive growth curve of mouse MLL-AF9 cells transduced with either CTRL or Pkcϵ shRNA GFP lentiviruses. %GFP+ cells were evaluated every two days by flow cytometry and normalized to fold change in %GFP+ at day 3 post-transduction, which represents day 0 in the figure (day 6 = day 9 post-transduction). C, Kaplan–Meier survival curve analysis of mice transplanted with mouse MLL-AF9 leukemia cells coexpressing GFP and either CTRL or Pkcϵ shRNAs (P = 0.0014; n = 7). MLL-AF9 (D) and MLL-AF9;Flt3ITD knock in (KI; E) cells transduced with lentiviruses coexpressing GFP with either CTRL or Pkcϵ shRNA were FACS-purified and plated in M3434. Dnmt3a−/−;Tet2−/− (F) and Dnmt3a−/−;Tet2−/−;FLT3ITD cells (G) transduced with lentiviruses coexpressing RFP with either CTRL or Pkcϵ shRNA were FACS-purified and plated in M3434. D–G, Bar graph representing the number of colonies formed by mouse MLL-AF9 leukemia cells expressing CTRL or Pkcϵ shRNAs in methylcellulose culture. Data are represented as the mean ± SD of three technical replicates for B and D–G (***, P ≤ 0.001; ***, P ≤ 0.0001).

Close modal

In addition to MLL-AF9, we observed that PKCϵ supports the growth of mouse hematopoietic stem and progenitor cells (HSPC) by expressing alterations in genes commonly mutated in AML. Briefly, leukemia cells coexpressing MLL-AF9 and an internal tandem duplication (ITD) of the murine Flt3 gene (MLL-AF9;Flt3ITD) were transduced with lentiviruses expressing GFP in combination with either CTRL or Pkcϵ shRNAs. HSPCs null for the combination of Dnmt3a and Tet2 deletion (Dnmt3a−/−;Tet2−/−) as well as Dnmt3a−/−;Tet2−/− HSPCs coexpressing human FLT3-ITD and GFP (Dnmt3a−/−;Tet2−/−;FLT3ITD) were transduced with lentiviruses expressing RFP in combination with either CTRL or Pkcϵ shRNAs. Following stable transduction, cells from each condition were purified by FACS and plated separately in cytokine-enriched methycellulose. In all of the models analyzed, shRNA-mediated inhibition of PKCϵ expression significantly hindered the CFC of leukemic cells compared with CTRL shRNA–expressing cells (Fig. 1E–G).

PKCϵ supports the survival of patient-derived AML samples

To investigate the functional role of PKCϵ in patient-derived AML samples, we employed an shRNA approach to inhibit the expression of PKCϵ in 10 subtype diverse patient-derived AML samples (Supplementary Table S1; Supplementary Fig. S3A). Briefly, cryopreserved patient-derived samples were thawed and plated on the HS-27 supportive stroma in cytokine-enriched media. After a short recovery period, cells were transduced with lentiviruses coexpressing GFP and either CTRL shRNA or PKCϵ shRNA_1 and subsequently replated on fresh HS-27 stroma cells. Three days after transduction, cells were assessed by flow cytometry to determine the percentage of live hCD45+, GFP+ cells (represented as time point day 0). Cells were cocultured for an additional 9 days and then assessed for the percentage of live hCD45+, GFP+ cells (time point day 9). We then determined the fold change in the percentage of hCD45+, GFP+ cells by dividing the day 9 percentages of hCD45+, GFP+ divided by the percentages of hCD45+, GFP+ at day 0. From this analysis, we observed that 8 of the 10 patient samples expressing PKCϵ-targeting shRNAs displayed a significantly lower fold change in the percentage of hCD45+, GFP+ cells compared with those samples expressing CTRL shRNAs (Fig. 2; Supplementary Fig. S3B). These data indicate that PKCϵ supports leukemia growth in a genetically diverse subset of patient-derived AML samples.

Figure 2.

PKCϵ inhibition impedes patient-derived AML growth. Ex vivo growth curve analysis of 10 patient-derived AML samples transduced with lentiviruses coexpressing GFP and CTRL, PKCϵ shRNA_1 and then analyzed by flow cytometry for the GFP+, hCD45+, PI cells every 3 days for a total of 12 days, post-transduction (P.T.). AML samples displayed maximal GFP mean fluorescence intensity (MFI) at three days post-transduction and thus represents analysis point, day 0. The data presented are expressed as the fold change of GFP+ cells at day 9 (day 12 post-transduction) versus day 0, relative to the mean of the CTRL shRNA (*, P ≤ 0.05; **, P ≤ 0.01; ***, P ≤ 0.001; ****, P < 0.0001). Data shown are the endpoint analyses (day 12 post-transduction) and are represented as mean ± SD of three technical replicates.

Figure 2.

PKCϵ inhibition impedes patient-derived AML growth. Ex vivo growth curve analysis of 10 patient-derived AML samples transduced with lentiviruses coexpressing GFP and CTRL, PKCϵ shRNA_1 and then analyzed by flow cytometry for the GFP+, hCD45+, PI cells every 3 days for a total of 12 days, post-transduction (P.T.). AML samples displayed maximal GFP mean fluorescence intensity (MFI) at three days post-transduction and thus represents analysis point, day 0. The data presented are expressed as the fold change of GFP+ cells at day 9 (day 12 post-transduction) versus day 0, relative to the mean of the CTRL shRNA (*, P ≤ 0.05; **, P ≤ 0.01; ***, P ≤ 0.001; ****, P < 0.0001). Data shown are the endpoint analyses (day 12 post-transduction) and are represented as mean ± SD of three technical replicates.

Close modal

PKCϵ regulates the intracellular redox environment of AML cells

Given the emerging recognition that ROS influences both normal and malignant HSPCs (4–6) and the previous connections of PKCϵ to redox biology (25–28), we investigated the relationship between PKCϵ and ROS biology in AML cells. As an initial assessment of whether shRNA-mediated inhibition of PKCϵ impacted intracellular ROS levels, we stained CTRL and PKCϵ-shRNA expressing cells with the fluorogenic probe CellROX, which detects multiple types of ROS. From this analysis, we found that inhibition of PKCϵ resulted in an increase of CellROX staining in OCI-AML3, NOMO1, and THP-1 cells (Fig. 3A; Supplementary Figs. S3C and S3D).

Figure 3.

PKCϵ regulates intracellular ROS biology in AML. A, OCI-AML3 cells stably expressing CTRL shRNA or PKCϵ shRNAs were stained with CellROX, 5 days post-transduction. Left, representative histogram plot of a single experiment; right, the average MFI of CellROX. Data are represented as the mean ± SD of three technical replicates. B, OCI-AML3 cells were stably transduced with the indicated roGFP2 probes to evaluate glutathione or H2O2 redox potential in cytoplasm or mitochondria. Cells were transduced with lentiviruses expressing CTRL shRNA or PKCϵ shRNA_1 or PKCϵ shRNA_2 and analyzed by flow cytometry 5 days later. Bar graph represents the percentage of live (PI) oxidized cells in the cytoplasm (Cyto) or mitochondria (Mito). Data represents the mean ± SD of three independent experiments. C, OCI-AML3 cells were transduced with CTRL shRNA or PKCϵ shRNA and stained after 4 days with MitoSOX and Annexin V (to exclude dead cells). Left, representative histogram plot of a single experiment; right, shows the average MitoSOX MFI of live cells (Annexin V) expressed as the mean ± SD of three independent experiments. D, MLL-AF9 leukemia cells coexpressing GFP and either CTRL or PKCϵ shRNAs were stained and evaluated for MitoSOX levels by flow cytometry, 5 days after transduction. Bar graph represents the MitoSOX MFI of live cells (Annexin V) expressed as the mean ± SD of three independent experiments (*, P ≤ 0.05; **, P ≤ 0.01; ***, P ≤ 0.001; ****, P < 0.0001).

Figure 3.

PKCϵ regulates intracellular ROS biology in AML. A, OCI-AML3 cells stably expressing CTRL shRNA or PKCϵ shRNAs were stained with CellROX, 5 days post-transduction. Left, representative histogram plot of a single experiment; right, the average MFI of CellROX. Data are represented as the mean ± SD of three technical replicates. B, OCI-AML3 cells were stably transduced with the indicated roGFP2 probes to evaluate glutathione or H2O2 redox potential in cytoplasm or mitochondria. Cells were transduced with lentiviruses expressing CTRL shRNA or PKCϵ shRNA_1 or PKCϵ shRNA_2 and analyzed by flow cytometry 5 days later. Bar graph represents the percentage of live (PI) oxidized cells in the cytoplasm (Cyto) or mitochondria (Mito). Data represents the mean ± SD of three independent experiments. C, OCI-AML3 cells were transduced with CTRL shRNA or PKCϵ shRNA and stained after 4 days with MitoSOX and Annexin V (to exclude dead cells). Left, representative histogram plot of a single experiment; right, shows the average MitoSOX MFI of live cells (Annexin V) expressed as the mean ± SD of three independent experiments. D, MLL-AF9 leukemia cells coexpressing GFP and either CTRL or PKCϵ shRNAs were stained and evaluated for MitoSOX levels by flow cytometry, 5 days after transduction. Bar graph represents the MitoSOX MFI of live cells (Annexin V) expressed as the mean ± SD of three independent experiments (*, P ≤ 0.05; **, P ≤ 0.01; ***, P ≤ 0.001; ****, P < 0.0001).

Close modal

To determine the localization and further define the specific types of ROS regulated by PKCϵ, OCI-AML3 cells were engineered to express the redox-sensitive roGFP2 protein genetically fused with glutaredoxin (Grx1) or the hydrogen peroxide (H2O2)-neutralizing yeast peroxidase Orp1. The Grx1-roGFP2 probe measures the redox potential of GSH:GSSG redox couples, where roGFP2-Orp1 probe measures changes in H2O2 levels. Both probes are expressed in the cytoplasm, however, we also engineered cells to express versions of each probe that are tagged with mitochondrial localization signals (Grx1-roGFP2–mito and roGFP2-Orp1–mito) to evaluate changes in mitochondria ROS biology (Supplementary Figs. S4A and S4B). roGFP2 has an emission of 510 nm as well as excitation peaks at 400 and 490 nm. Upon oxidation, the intensity of the 400-nm roGFP2 peak increases while the amplitude of the 490-nm peak decreases resulting in the ratio of peak intensities shifting toward 400 nm (23). OCI-AML3 cells were treated with either the oxidizing agent diamide (DIA) or the reducing agent dithiothreitol (DTT) to establish the oxidized and reduced gates (data not shown). Using these probes, we observed that the expression of PKCϵ shRNA_1 or shRNA_2 resulted in a significantly higher percentage of oxidized OCI-AML3 cells expressing either cytoplasmic or mitochondrial Grx1-roGFP2, compared with control shRNAs. We also observed that PKCϵ inhibition led to a significant increase in the percentage of cells expressing oxidized roGFP2-Orp1 in the mitochondria (Fig. 3B; Supplementary Fig. S4A and S4B).

The most well-defined ROS generated by mitochondria are superoxides, which are either neutralized by glutathione or converted to H2O2 by superoxide-dimutase (SOD) enzymes. Given that PKCϵ inhibition consistently altered mitochondrial levels of glutathione and H2O2, we next examined how PKCϵ inhibition impacted mitochondrial superoxide levels using the fluorogenic probe MitoSOX. From this analysis, we observed that shRNA-mediated inhibition of PKCϵ led to a significant increase in MitoSOX staining in both human [OCI-AML3, Fig. 3C; Supplementary Figs. S5A (THP-1) and S5B] and mouse (MLL-AF9, Fig. 3D) AML cells. We also observed that shRNA-mediated inhibition of PKCϵ in patient-derived AML cells expressing MLL-AF9 also display elevated MitoSOX levels (Supplementary Fig. S5C). Collectively, these findings reveal that PKCϵ regulates steady-state levels of mitochondrial ROS and possibly certain cytoplasmic ROS in human and mouse AML cells.

Reducing mitochondrial ROS partially reverses the antileukemia effects of PKCϵ inhibition

To determine whether changes in the intracellular redox biology contribute to the antileukemia effects of PKCϵ inhibition, we assessed how various chemical antioxidants impacted the growth and survival of AML cells expressing CTRL and PKCϵ-targeting shRNAs. Twice daily treatment with either N-acetyl-l-cysteine (NAC) or glutathione was unable to reverse the antileukemia effects of PKCϵ inhibition in both human and mouse cells despite being able to effectively block the cytotoxicity of the glutathione-depleting pro-oxidant, Menadione (Supplementary Fig. S6A and S6B and data not shown). However, administration of either butylated hydroxyanisole (BHA) or MitoTEMPO, both compounds that neutralize mitochondrial ROS, was able to significantly reduce the cell death of human and mouse AML cells mediated by PKCϵ inhibition (Fig. 4A and B; Supplementary Fig. S6C).

Figure 4.

Neutralization of ROS partially reverses the antileukemia effects of PKCϵ inhibition. A and B, OCI-AML3 cells were transduced with CTRL shRNA, PKCϵ shRNA_1, or PKCϵ shRNA_2 and then administered 25 μmol/L BHA (A) or 100 nmol/L MitoTEMPO (B). Ninety-six hours post-transduction, cells from each condition were assessed for Annexin V staining by flow cytometry. Data are represented as the mean ± SD of two independent experiments. C, OCI-AML3 cells stably transduced with retroviruses coexpressing SOD2-IRES-Catalase (SOD2-Catalase) and GFP or control vector (CTRL) were transduced with CTRL shRNA, PKCϵ shRNA_1 or PKCϵ shRNA_2. Four days after transduction, cells were analyzed for cell death evaluated as percent of Annexin V+ cells by flow cytometry. Data are represented as the mean ± SD of three independent experiments for each panel (*, P ≤ 0.05; **, P ≤ 0.01). D, SOD2-Catalase or CTRL-expressing mouse MLL-AF9 were transduced with CTRL shRNA or Pkcϵ shRNA, selected with puromycin and grown in methylcellulose. After 5 days of culture, colonies were enumerated and data represents the mean ± SD of two independent experiments (*, P ≤ 0.05; **, P ≤ 0.01; ***, P ≤ 0.001; ****, P < 0.0001).

Figure 4.

Neutralization of ROS partially reverses the antileukemia effects of PKCϵ inhibition. A and B, OCI-AML3 cells were transduced with CTRL shRNA, PKCϵ shRNA_1, or PKCϵ shRNA_2 and then administered 25 μmol/L BHA (A) or 100 nmol/L MitoTEMPO (B). Ninety-six hours post-transduction, cells from each condition were assessed for Annexin V staining by flow cytometry. Data are represented as the mean ± SD of two independent experiments. C, OCI-AML3 cells stably transduced with retroviruses coexpressing SOD2-IRES-Catalase (SOD2-Catalase) and GFP or control vector (CTRL) were transduced with CTRL shRNA, PKCϵ shRNA_1 or PKCϵ shRNA_2. Four days after transduction, cells were analyzed for cell death evaluated as percent of Annexin V+ cells by flow cytometry. Data are represented as the mean ± SD of three independent experiments for each panel (*, P ≤ 0.05; **, P ≤ 0.01). D, SOD2-Catalase or CTRL-expressing mouse MLL-AF9 were transduced with CTRL shRNA or Pkcϵ shRNA, selected with puromycin and grown in methylcellulose. After 5 days of culture, colonies were enumerated and data represents the mean ± SD of two independent experiments (*, P ≤ 0.05; **, P ≤ 0.01; ***, P ≤ 0.001; ****, P < 0.0001).

Close modal

To examine how specific neutralization of mitochondrial ROS impacts the antileukemia effects of PKCϵ inhibition, we engineered OCI-AML3 cells to constitutively coexpress SOD2 and Catalase (SOD2-Catalase), which neutralize mitochondrial superoxides and H2O2, respectively. Control and SOD2-Catalase–expressing OCI-AML3 cells were then transduced with CTRL or PKCϵ shRNAs and subsequently analyzed for MitoSOX levels and cell death. SOD2-Catalase expression significantly blocked the induction of MitoSOX staining mediated by PKCϵ inhibition (Supplementary Fig. S6D) and significantly reduced cell death induced by PKCϵ-targeting shRNAs (Fig. 4C). We also observed that SOD2-Catalase expression restored the CFC of mouse MLL-AF9 leukemia cells expressing Pkcϵ shRNA (Fig. 4D) further indicating that AML cells rely on PKCϵ to maintain mitochondrial ROS levels and survival.

The mitochondrial ROS–neutralizing enzyme, SOD2, supports AML in vitro and in vivo

To examine how a specific induction of mitochondrial superoxides impacts AML cell growth, we evaluated how shRNAs directly targeting SOD2 impacted human AML cell biology (Supplementary Fig. S7A). Similar to PKCϵ inhibition, shRNA-mediated depletion of SOD2 led to increased steady-state levels of MitoSOX and reduced AML cell growth and survival (Supplementary Fig. S7B–S7D). Furthermore, direct inhibition of SOD2 in mouse MLL-AF9 leukemia cells (Fig. 5A) led to increased levels of MitoSOX staining (Fig. 5B), reduced CFC (Supplementary Fig. S7E) and significantly delayed the time of disease onset in vivo (Fig. 5C). These results show that similar to the antileukemia effects of PKCϵ inhibition, SOD2 inhibition leads to an accumulation of mitochondrial ROS and diminishes AML cell growth and survival.

Figure 5.

SOD2 inhibition phenocopies the antileukemic effects of PKCϵ inhibition. A, Mouse MLL-AF9 leukemia cells were transduced with CTRL shRNA or SOD2 shRNA, sorted 48 hours later and analyzed for SOD2 expression by western blot. B, MitoSOX staining of mouse MLL-AF9 leukemia cells evaluated by flow cytometry 5 days after transduction with CRTL, Sod2 shRNA_1, or Sod2 shRNA_2 (the bar graph shows the MitoSOX MFI of live cells (Annexin V; Ctrl shRNA vs. shRNA sod2_1 and _2; *, P ≤ 0.05). Data represents the mean ± SD of three technical replicates. C, Kaplan–Meier survival curve analysis of mice transplanted with mouse MLL-AF9 leukemia cells expressing either CTRL or Sod2 shRNA_2 (P = 0.0042; n = 7).

Figure 5.

SOD2 inhibition phenocopies the antileukemic effects of PKCϵ inhibition. A, Mouse MLL-AF9 leukemia cells were transduced with CTRL shRNA or SOD2 shRNA, sorted 48 hours later and analyzed for SOD2 expression by western blot. B, MitoSOX staining of mouse MLL-AF9 leukemia cells evaluated by flow cytometry 5 days after transduction with CRTL, Sod2 shRNA_1, or Sod2 shRNA_2 (the bar graph shows the MitoSOX MFI of live cells (Annexin V; Ctrl shRNA vs. shRNA sod2_1 and _2; *, P ≤ 0.05). Data represents the mean ± SD of three technical replicates. C, Kaplan–Meier survival curve analysis of mice transplanted with mouse MLL-AF9 leukemia cells expressing either CTRL or Sod2 shRNA_2 (P = 0.0042; n = 7).

Close modal

PKCϵ regulates the expression of proteins that regulate mitochondrial biology

To identify potential downstream effectors of PKCϵ in AML, we performed nanoscale liquid chromatography coupled to tandem mass spectrometry (nanoLC/MS-MS) on OCI-AML3 cells expressing CTRL shRNA, PKCϵ shRNA_1, or PKCϵ shRNA_2. From this analysis, 3,192 peptides were captured and using a statistical cutoff of P < 0.05, we found that 707 peptides were differently expressed between CTRL shRNA and PKCϵ shRNA_1–expressing cells and 641 peptides were differently expressed between CTRL shRNA and PKCϵ shRNA_2–expressing cells. Furthermore, we observed that 226 peptides were similarly differentially expressed between OCI-AML3 cells expressing CTRL shRNA versus PKCϵ shRNA_1 and CTRL shRNA versus PKCϵ shRNA_2. Integrated pathway analysis did not reveal any significant enrichment of particular molecular pathways or processes however, we did observe that approximately 15% (34 proteins) of these differentially expressed proteins between CTRL shRNA and either PKCϵ shRNA_1 or PKCϵ shRNA_2 were related to mitochondrial biology. Aberrant mitochondrial ROS arise from multiple disruptions in mitochondrial biology such as reduced expression of antioxidant systems, perturbations in the activities of the complexes that regulate electron flux through the electron transport chain (ETC), as well as alterations in outer mitochondrial membrane (OMM) potential and transport. Many of the proteins whose expression is altered by PKCϵ inhibition are components of ETC complexes involved in the regulation of OMM potential (UQCR10, ATPC1, ATP5H, COX6C, SDHAF2, and NDUFB10) or proteins involved in mitochondrial membrane transport (VDAC1, VDAC3, TOMM22, SLC25A1, SLC25A11, and SLC25A12; Fig. 6A). We also observed that two antioxidant proteins, GSS and TXN, were significantly reduced by PKCϵ inhibition as well as a variety of other antioxidant proteins that were reduced but not statistically significant (Supplementary Fig. S7F).

Figure 6.

PKCϵ protects AML cells against agents that induce mitochondrial dysfunction and mitochondrial ROS-oxidative stress. A, Heatmap analysis displaying the expression of mitochondrial-regulatory proteins that were significantly differentially expressed between CTRL shRNA versus PKCϵ shRNA_1 or versus PKCϵ shRNA_2 transduced OCI-AML3 cells. B and C, OCI-AML3 cells were transduced with retroviral vectors that constitutively express PKCϵ and GFP (PKCϵ) or just GFP-expressing control (Ctrl) viruses. Four days after transduction, GFP+ cells were isolated by FACS and treated with 100 μmol/L of AA (B) or TTFA (C). The percentage of Annexin V+ cells was evaluated 24 hours after treatment by flow cytometry. Data are represented as the mean ± SD of three independent experiments. D, Ctrl and PKCϵ-expressing mouse MLL-AF9 leukemia cells were generated as described in B and C. GFP+ cells were isolated by FACS and seeded in methylcellulose with vehicle, 100 μmol/L AA or 200 μmol/L TTFA. The bar graph shows the number of colonies formed in methylcellulose after 5 days of culture. The data represent the mean ± SD of three technical replicates. (**, P ≤ 0.01; ***, P ≤ 0.001).

Figure 6.

PKCϵ protects AML cells against agents that induce mitochondrial dysfunction and mitochondrial ROS-oxidative stress. A, Heatmap analysis displaying the expression of mitochondrial-regulatory proteins that were significantly differentially expressed between CTRL shRNA versus PKCϵ shRNA_1 or versus PKCϵ shRNA_2 transduced OCI-AML3 cells. B and C, OCI-AML3 cells were transduced with retroviral vectors that constitutively express PKCϵ and GFP (PKCϵ) or just GFP-expressing control (Ctrl) viruses. Four days after transduction, GFP+ cells were isolated by FACS and treated with 100 μmol/L of AA (B) or TTFA (C). The percentage of Annexin V+ cells was evaluated 24 hours after treatment by flow cytometry. Data are represented as the mean ± SD of three independent experiments. D, Ctrl and PKCϵ-expressing mouse MLL-AF9 leukemia cells were generated as described in B and C. GFP+ cells were isolated by FACS and seeded in methylcellulose with vehicle, 100 μmol/L AA or 200 μmol/L TTFA. The bar graph shows the number of colonies formed in methylcellulose after 5 days of culture. The data represent the mean ± SD of three technical replicates. (**, P ≤ 0.01; ***, P ≤ 0.001).

Close modal

PKCϵ protects AML cells against agents that promote mitochondrial dysfunction and induce superoxides

To assess the role PKCϵ in superoxide-induced oxidative stress, we examined how modulating PKCϵ expression impacted the survival of AML cells challenged with either of the mitochondrial superoxide inducing agents, 2-thenoyltrifluoroacetone (TTFA) or antimycin A (AA). To examine whether increased expression of PKCϵ is able to protect AML cells from the cytotoxic effects of TTFA or AA, we generated OCI-AML3, THP-1, and mouse MLL-AF9–expressing AML cells that constitutively express PKCϵ (Supplementary Fig. S8A and data not shown). Constitutive PKCϵ expression did not impact OCI-AML3 growth or the CFC of mouse MLL-AF9 leukemia cells (Supplementary Fig. S8B and S*C). However, PKCϵ-expressing OCI-AML3 or THP-1 cells treated with TTFA or AA displayed significantly lower percentages of cell death compared with similarly treated control cells (Fig. 6B and C; Supplementary Fig. S8E and S8F). In addition, constitutive PKCϵ expression significantly improved the CFC of mouse MLL-AF9 leukemia cells challenged with TTFA or AA compared with similarly treated control cells (Fig. 6D). Of all the human AML cell lines we tested, U937 cells were the least impacted by PKCϵ inhibition, cell viability–wise (Supplementary Fig. S1B, bottom). Therefore, we evaluated whether PKCϵ inhibition rendered U937 cells more sensitive to TTFA and/or AA treatment. From this analysis, we found that shRNA-mediated inhibition of PKCϵ exacerbated the cytotoxic effects of TTFA and AA in comparison with PKCϵ inhibition or pro-oxidant treatment alone (Supplementary Fig. S8G and S8H). Collectively, these results suggest that PKCϵ protects AML cells from agents that perturb mitochondrial function and induce oxidative stress.

PKCϵ has been implicated as an oncogenic kinase in several human cancers including prostate, breast, colon, lung, and certain forms of squamous cell carcinoma (29–34). In AML, downmodulation of PKCϵ is necessary for the prodifferentiating actions of phorbol esters (20, 35), insinuating that PKCϵ supports leukemia growth and expansion. However, the impact of blocking PKCϵ expression on AML growth and survival has not been comprehensively investigated.

Here, we demonstrate that shRNA-mediated reduction of PKCϵ significantly reduces the survival of human AML cells in vitro and significantly impedes disease progression in a GEMM of AML driven by MLL-AF9. In addition, inhibition of PKCϵ reduced the in vitro growth properties of multiple, genetically diverse patient-derived AML samples confirming that the proleukemia roles of PKCϵ is not restricted to a particular genetic subtype of AML.

At the molecular level, we have discovered that PKCϵ is a key regulator of intracellular redox homeostasis in several mouse and human AML models. Using multiple ROS detection strategies we have found that shRNA-mediated inhibition of PKCϵ increases the steady-state levels of multiple ROS, including several mitochondrial ROS. On the basis of these observations, we postulated that increased/excess production of mitochondrial ROS antagonizes leukemia cell viability and that management of mitochondrial ROS levels is a key proleukemia function of PKCϵ. We have made three central observations that support these hypotheses. First, chemical antioxidants that specifically neutralize mitochondrial ROS, such as BHA (36) and MitoTEMPO (37) are able to significantly and consistently blunt cell death mediated by PKCϵ inhibition, whereas indiscriminant antioxidants such as NAC and glutathione are not. Moreover, reconstitution of mitochondrial ROS-neutralizing enzymes SOD2 and Catalase, partially reverses mitochondrial superoxide induction and cell death mediated by PKCϵ inhibition. Second, similar to PKCϵ inhibition, shRNA-mediated inhibition of SOD2 increased mitochondrial ROS, limited disease progression in vivo and suppressed the growth of patient-derived AML samples. Third, elevated expression of PKCϵ is able to largely protect AML cells from otherwise toxic doses of agents that drive mitochondrial ROS production. Moreover, impeding PKCϵ expression rendered leukemia cells more sensitive to these pro-oxidants. Collectively, these results support that certain sub-types of AML rely on PKCϵ for proper management of mitochondrial ROS biology and survival.

Our observations that the anti-leukemia effects of PKCϵ inhibition could not by fully rescued by SOD2-Catalase expression, MitoTEMPO or BHA suggest that PKCϵ may regulate additional molecular processes to support AML cell survival. Our proteomic analysis shows that, in addition to multiple key ROS-buffering enzymes, such as TXN and GSS, several proteins related to mitochondrial biology and function are impacted by PKCϵ inhibition. Therefore, it remains possible that the increase in mitochondrial ROS mediated by PKCϵ inhibition is due to mitochondrial dysfunction either in addition to or in place of the observed decrease in ROS-regulating enzymes. Consistent with this idea, several studies have shown that the mitochondrial redox state of AML cells is directly related to their metabolic needs. Specifically, compared to bulk AML cells, leukemia-initiating cells from multiple AML patients display a lower oxidized redox environment that is associated with lower rates of oxidative phosphorylation (OXPHOS; ref. 38). Furthermore, cytarabine-treated AML cells display high levels of mitochondrial ROS and OXPHOS and this altered mitochondrial state may contribute to cytarabine resistance (39). Also, multiple AML patients display increased mitochondrial mass compared with healthy HSPCs and as a result are more sensitive to OXPHOS-induced oxidative stress (40).

Although our results indicate that a key proleukemia function of PKCϵ is to manage mitochondrial ROS biology, they do not exclude the possibility that PKCϵ regulates additional redox-related (that are mitochondrial-independent) and/or redox-independent mechanisms to support AML cell survival. For example, our proteomic analysis shows that PKCϵ inhibition, although not statistically significant, impacts multiple redox-regulatory systems. For example, protein levels of both PRDX2 and PRDX4, which have been implicated as growth suppressors in AML and acute promyelocytic leukemia (APL), respectively, increase upon PKCϵ inhibition (41, 42). PKCϵ inhibition also altered the expression of various glutathione-regulatory components and Pei and colleagues (4) have shown that pharmacologic inhibition of glutathione metabolic enzymes, such as GPX1 and GCLC, antagonize primitive human leukemia cell survival. In fact, the relationship of PKCϵ and redox biology varies among distinct biological settings. PKCϵ activation in neuronal and cardiac tissues correlates with the induction of ROS mediated by ischemia, hypoxia or pro-oxidants such as buthionine sulfoximine (BSO) or AA. However, other studies have shown that PKCϵ activation promotes ROS generation in smooth muscle and immortalized epithelial cells and that hepatocytes void of PKCϵ display enhanced stress-induced ROS formation (28, 43). These divergent observations may result from distinct tissue- or ROS-specific roles of PKCϵ; however, in the context of AML, our results establish that PKCϵ works to suppress mitochondrial ROS and possibly other types of ROS.

Several studies suggest that tumor cells, including leukemia cells, maintain high levels of ROS to drive cell growth and survival and therefore targeting redox regulators may be a viable anticancer therapeutic strategy (2, 34, 44). In our models, reducing steady-state levels of mitochondrial superoxides through the combined over-expression of SOD2 and Catalase does not impede colony formation suggesting that elevated mitochondrial ROS are not a central driver of AML cell growth in this model. However, increasing mitochondrial ROS, by inhibiting PKCϵ or SOD2 or by administering chemical ROS-inducing agents diminishes AML cell survival. Collectively, these results suggest that strategies for increasing, rather than decreasing, mitochondrial ROS may carry a significant therapeutic potential. Consistent with this concept, the efficacy of arsenic trioxide, which is commonly used to treat APL, works primarily by inducing ROS (45). Moreover, high doses of vitamin C, which have been previously shown to induce toxic ROS levels in certain types of cancer cells (46), were recently reported to selectively eliminate AML cells carrying TET2 (47, 48) or IDH (49) mutations. However, it should be noted that the proposed mechanism of action of Vitamin C in AML is to activate other TET family members (47, 48).

The intracellular redox environment of AML cells is often distinct from their normal counterparts. Therefore, identifying and defining the molecular regulators of redox biology, such as PKCϵ and SOD2, may provide key insights into the etiology and pathogenesis of AML as well as possibly contribute to the design of more effective antileukemia therapies. However, AML encompasses a wide variety of genetic subtypes and individual tumors often display complex clonal heterogeneity (50) and it remains unclear which genetic subtypes are susceptible to redox imbalances. Our loss-of-function studies in patient-derived AML cells show that not all AML samples rely on PKCϵ for growth and survival. Thus, it is possible that certain AML cells utilize PKCϵ- and/or SOD2-independent mechanisms to regulate mitochondrial superoxide biology or that certain genetic subtypes or clones are insensitive to increases in mitochondrial superoxide levels. Therefore, future studies defining the genetic subtypes that are sensitive to changes in redox homeostasis or PKCϵ/SOD2 inhibition as well as the role of PKCϵ/SOD2 in healthy HSPC biology will be needed to fully gauge the therapeutic potential of targeting these pathways in AML.

No potential conflicts of interest were disclosed.

Conception and design: D.Di Marcantonio, E. Masselli, M.D. Milsom, G. Gobbi, S.M. Sykes

Development of methodology: J. Michael Meadows, G. Gobbi, S.M. Sykes

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): D.Di Marcantonio, E. Martinez, J. Vadaketh, A. Gupta, J. Michael Meadows, G.A. Challen, B. Garcia, R. Garzon, S.M. Sykes

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): D.Di Marcantonio, J. Vadaketh, A. Gupta, J. Michael Meadows, F. Ferraro, M.D. Milsom, S. Froehling, P. Mirandola, G. Gobbi, S.M. Sykes

Writing, review, and/or revision of the manuscript: D.Di Marcantonio, E. Masselli, M.D. Milsom, C. Scholl, S. Froehling, S. Balachandran, T. Skorski, R. Garzon, M. Vitale, S.M. Sykes

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): M. Nieborowska-Skorska, T. Skorski, S.M. Sykes

Study supervision: P. Mirandola, M. Vitale, S.M. Sykes

Other (responsible for the design, execution and interpretation of mass spectrometry studies): S. Sidoli

Other (advised in research design): C. Scholl

This work was supported by the NIH Grant R00 CA158461, the ASH Junior Scholar Award, W.W. Smith and Bob and Jeanne Brennan (to S.M. Sykes); the Rotary Foundation, Grant GG1414529 and the Board of Directors of Fox Chase Cancer Center Fellowship (to D. Di Marcantonio); CURE supplement (CA06927; to J. Vadaketh and A. Gupta); Jeanne E. and Robert F. Ozols Undergraduate Summer Research Fellowship, Fox Chase Cancer Center (to J. Michael Meadows); NIH grant P01CA196539, DOD grant W81XWH-113-1-0426 and the Leukemia and Lymphoma Society Dr. Robert Arceci Scholar Award (to B. Garcia), The Dietmar Hopp Stiftung (to M.D. Milsom), the NIH Grant 1R01DK102428 (to G.A. Challen).

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Holmström
KM
,
Finkel
T
. 
Cellular mechanisms and physiological consequences of redox-dependent signalling
.
Nat Rev Mol Cell Biol
2014
;
15
:
411
21
.
2.
Sabharwal
SS
,
Schumacker
PT
. 
Mitochondrial ROS in cancer: initiators, amplifiers or an Achilles' heel?
Nat Rev Cancer
2014
;
14
:
709
21
.
3.
Irwin
ME
,
Rivera-Del Valle
N
,
Chandra
J
. 
Redox control of leukemia: from molecular mechanisms to therapeutic opportunities
.
Antioxid Redox Signal
2013
;
18
:
1349
83
4.
Pei
S
,
Minhajuddin
M
,
Callahan
KP
,
Balys
M
,
Ashton
JM
,
Neering
SJ
, et al
Targeting aberrant glutathione metabolism to eradicate human acute myelogenous leukemia cells
.
J Biol Chem
2013
;
288
:
33542
58
.
5.
Hole
PS
,
Zabkiewicz
J
,
Munje
C
,
Newton
Z
,
Pearn
L
,
White
P
, et al
Overproduction of NOX-derived ROS in AML promotes proliferation and is associated with defective oxidative stress signaling
.
Blood
2013
;
122
:
3322
30
.
6.
Zhou
FL
,
Zhang
WG
,
Wei
YC
,
Meng
S
,
Bai
GG
,
Wang
BY
, et al
Involvement of oxidative stress in the relapse of acute myeloid leukemia
.
J Biol Chem
2010
;
285
:
15010
5
.
7.
Sallmyr
A
,
Fan
J
,
Datta
K
,
Kim
KT
,
Grosu
D
,
Shapiro
P
, et al
Internal tandem duplication of FLT3 (FLT3/ITD) induces increased ROS production, DNA damage, and misrepair: implications for poor prognosis in AML
.
Blood
2008
;
111
:
3173
82
.
8.
Stanicka
J
,
Russell
EG
,
Woolley
JF
,
Cotter
TG
. 
NADPH oxidase-generated hydrogen peroxide induces DNA damage in mutant FLT3-expressing leukemia cells
.
J Biol Chem
2015
;
290
:
9348
61
.
9.
Weinberg
F
,
Hamanaka
R
,
Wheaton
WW
,
Weinberg
S
,
Joseph
J
,
Lopez
M
, et al
Mitochondrial metabolism and ROS generation are essential for Kras-mediated tumorigenicity
.
Proc Natl Acad Sci U S A
2010
;
107
:
8788
93
.
10.
Hole
PS
,
Pearn
L
,
Tonks
AJ
,
James
PE
,
Burnett
AK
,
Darley
RL
, et al
Ras-induced reactive oxygen species promote growth factor-independent proliferation in human CD34+hematopoietic progenitor cells
.
Blood
2010
;
115
:
1238
46
.
11.
Koptyra
M
,
Falinski
R
,
Nowicki
MO
,
Stoklosa
T
,
Majsterek
I
,
Nieborowska-Skorska
M
, et al
BCR/ABL kinase induces self-mutagenesis via reactive oxygen species to encode imatinib resistance
.
Blood
2006
;
108
:
319
27
.
12.
Naughton
R
,
Quiney
C
,
Turner
SD
,
Cotter
TG
. 
Bcr-Abl-mediated redox regulation of the PI3K/AKT pathway
.
Leukemia
2009
;
23
:
1432
40
.
13.
Tanaka
H
,
Matsumura
I
,
Ezoe
S
,
Satoh
Y
,
Sakamaki
T
,
Albanese
C
, et al
E2F1 and c-Myc potentiate apoptosis through inhibition of NF-kappaB activity that facilitates MnSOD-mediated ROS elimination
.
Mol Cell
2002
;
9
:
1017
29
.
14.
Gonçalves
AC
,
Alves
R
,
Baldeiras
I
,
Cortesão
E
,
Carda
JP
,
Branco
CC
, et al
Genetic variants involved in oxidative stress, base excision repair, DNA methylation, and folate metabolism pathways influence myeloid neoplasias susceptibility and prognosis
.
Mol Carcinog
2017
;
56
:
130
148
.
15.
Jeong
SM
,
Hwang
S
,
Seong
RH
. 
Transferrin receptor regulates pancreatic cancer growth by modulating mitochondrial respiration and ROS generation
.
Biochem Biophys Res Commun
2016
;
471
:
373
9
.
16.
Hambright
HG
,
Meng
P
,
Kumar
AP
,
Ghosh
R
. 
Inhibition of PI3K/AKT/mTOR axis disrupts oxidative stress-mediated survival of melanoma cells
.
Oncotarget
2015
;
6
:
7195
208
.
17.
Totoń
E
,
Ignatowicz
E
,
Skrzeczkowska
K
,
Rybczyńska
M
. 
Protein kinase Cϵ as a cancer marker and target for anticancer therapy
.
Pharmacol Rep
2011
;
63
:
19
29
.
18.
Gobbi
G
,
Mirandola
P
,
Carubbi
C
,
Galli
D
,
Vitale
M
. 
Protein kinase C ϵ in hematopoiesis: conductor or selector?
Semin Thromb Hemost
2013
;
39
:
59
65
.
19.
Masselli
E
,
Carubbi
C
,
Gobbi
G
,
Mirandola
P
,
Galli
D
,
Martini
S
, et al
Protein kinase Cϵ inhibition restores megakaryocytic differentiation of hematopoietic progenitors from primary myelofibrosis patients
.
Leukemia
2015
;
29
:
2192
201
.
20.
Gobbi
G
,
Mirandola
P
,
Carubbi
C
,
Micheloni
C
,
Malinverno
C
,
Lunghi
P
, et al
Phorbol ester-induced PKCepsilon down-modulation sensitizes AML cells to TRAIL-induced apoptosis and cell differentiation
.
Blood
2009
;
113
:
3080
7
.
21.
Klco
JM
,
Spencer
DH
,
Lamprecht
TL
,
Sarkaria
SM
,
Wylie
T
,
Magrini
V
, et al
Genomic impact of transient low-dose decitabine treatment on primary AML cells
.
Blood
2013
;
121
:
1633
43
.
22.
Walter
D
,
Lier
A
,
Geiselhart
A
,
Thalheimer
FB
,
Huntscha
S
,
Sobotta
MC
, et al
Exit from dormancy provokes DNA-damage-induced attrition in haematopoietic stem cells
.
Nature
2015
;
520
:
549
52
.
23.
Morgan
B
,
Sobotta
MC
,
Dick
TP
. 
Measuring E(GSH) and H2O2 with roGFP2-based redox probes
.
Free Radic Biol Med
2011
;
51
:
1943
51
.
24.
Krivtsov
AV
,
Twomey
D
,
Feng
Z
,
Stubbs
MC
,
Wang
Y
,
Faber
J
, et al
Transformation from committed progenitor to leukaemia stem cell initiated by MLL-AF9
.
Nature
2006
;
442
:
818
22
.
25.
Barnett
ME
,
Madgwick
DK
,
Takemoto
DJ
. 
Protein kinase C as a stress sensor
.
Cell Signal
2007
;
19
:
1820
9
.
26.
Jung
YS
,
Ryu
BR
,
Lee
BK
,
Mook-Jung
I
,
Kim
SU
,
Lee
SH
, et al
Role for PKC-epsilon in neuronal death induced by oxidative stress
.
Biochem Biophys Res Commun
2004
;
320
:
789
94
.
27.
Kabir
AM
,
Clark
JE
,
Tanno
M
,
Cao
X
,
Hothersall
JS
,
Dashnyam
S
, et al
Cardioprotection initiated by reactive oxygen species is dependent on activation of PKCepsilon
.
Am J Physiol Heart Circ Physiol
2006
;
291
:
H1893
9
.
28.
Rathore
R
,
Zheng
YM
,
Niu
CF
,
Liu
QH
,
Korde
A
,
Ho
YS
, et al
Hypoxia activates NADPH oxidase to increase [ROS]i and [Ca2+]i through the mitochondrial ROS-PKCepsilon signaling axis in pulmonary artery smooth muscle cells
.
Free Radic Biol Med
2008
;
45
:
1223
31
.
29.
Cornford
P
,
Evans
J
,
Dodson
A
,
Parsons
K
,
Woolfenden
A
,
Neoptolemos
J
, et al
Protein kinase C isoenzyme patterns characteristically modulated in early prostate cancer
.
Am J Pathol
1999
;
154
:
137
44
.
30.
Pan
Q
,
Bao
LW
,
Kleer
CG
,
Sabel
MS
,
Griffith
KA
,
Teknos
TN
, et al
Protein kinase C epsilon is a predictive biomarker of aggressive breast cancer and a validated target for RNA interference anticancer therapy
.
Cancer Res
2005
;
65
:
8366
71
.
31.
Davidson
LA
,
Jiang
YH
,
Derr
JN
,
Aukema
HM
,
Lupton
JR
,
Chapkin
RS
. 
Protein kinase C isoforms in human and rat colonic mucosa
.
Arch Biochem Biophys
1994
;
312
:
547
53
.
32.
Bae
KM
,
Wang
H
,
Jiang
G
,
Chen
MG
,
Lu
L
,
Xiao
L
. 
Protein kinase C epsilon is overexpressed in primary human non-small cell lung cancers and functionally required for proliferation of non-small cell lung cancer cells in a p21/Cip1-dependent manner
.
Cancer Res
2007
;
67
:
6053
63
.
33.
Martínez-Gimeno
C
,
Díaz-Meco
MT
,
Domínguez
I
,
Moscat
J
. 
Alterations in levels of different protein kinase C isotypes and their influence on behavior of squamous cell carcinoma of the oral cavity: epsilon PKC, a novel prognostic factor for relapse and survival
.
Head Neck
1995
;
17
:
516
25
.
34.
Gobbi
G
,
Masselli
E
,
Micheloni
C
,
Nouvenne
A
,
Russo
D
,
Santi
P
, et al
Hypoxia-induced down-modulation of PKCepsilon promotes trail-mediated apoptosis of tumor cells
.
Int J Oncol
2010
;
37
:
719
29
.
35.
Wu
SF
,
Huang
Y
,
Hou
JK
,
Yuan
TT
,
Zhou
CX
,
Zhang
J
, et al
The downregulation of onzin expression by PKCepsilon-ERK2 signaling and its potential role in AML cell differentiation
.
Leukemia
2010
;
24
:
544
51
.
36.
Thapa
RJ
,
Nogusa
S
,
Chen
P
,
Maki
JL
,
Lerro
A
,
Andrake
M
, et al
Interferon-induced RIP1/RIP3-mediated necrosis requires PKR and is licensed by FADD and caspases
.
Proc Natl Acad Sci U S A
2013
;
110
:
E3109
18
.
37.
Dikalov
S
. 
Cross talk between mitochondria and NADPH oxidases
.
Free Radic Biol Med
2011
;
51
:
1289
301
.
38.
Lagadinou
ED
,
Sach
A
,
Callahan
K
,
Rossi
RM
,
Neering
SJ
,
Minhajuddin
M
, et al
BCL-2 inhibition targets oxidative phosphorylation and selectively eradicates quiescent human leukemia stem cells
.
Cell Stem Cell
2013
;
12
:
329
41
.
39.
Farge
T
,
Saland
E
,
de Toni
F
,
Aroua
N
,
Hosseini
M
,
Perry
R
, et al
Chemotherapy-resistant human acute myeloid leukemia cells are not enriched for leukemic stem cells but require oxidative metabolism
.
Cancer Discov
2017
;
7
:
716
735
.
40.
Sriskanthadevan
S
,
Jeyaraju
DV
,
Chung
TE
,
Prabha
S
,
Xu
W
,
Skrtic
M
, et al
AML cells have low spare reserve capacity in their respiratory chain that renders them susceptible to oxidative metabolic stress
.
Blood
2015
;
125
:
2120
30
.
41.
Agrawal-Singh
S
,
Isken
F
,
Agelopoulos
K
,
Klein
HU
,
Thoennissen
NH
,
Koehler
G
, et al
Genome-wide analysis of histone H3 acetylation patterns in AML identifies PRDX2 as an epigenetically silenced tumor suppressor gene
.
Blood
2012
;
119
:
2346
57
.
42.
Palande
KK
,
Beekman
R
,
van der Meeren
LE
,
Beverloo
HB
,
Valk
PJ
,
Touw
IP
. 
The antioxidant protein peroxiredoxin 4 is epigenetically down regulated in acute promyelocytic leukemia
.
PLoS One
2011
;
6
:
e16340
.
43.
Raddatz
K
,
Turner
N
,
Frangioudakis
G
,
Liao
BM
,
Pedersen
DJ
,
Cantley
J
, et al
Time-dependent effects of Prkce deletion on glucose homeostasis and hepatic lipid metabolism on dietary lipid oversupply in mice
.
Diabetologia
2011
;
54
:
1447
56
.
44.
Kalota
A
,
Selak
MA
,
Garcia-Cid
LA
,
Carroll
M
. 
Eltrombopag modulates reactive oxygen species and decreases acute myeloid leukemia cell survival
.
PLoS One
2015
;
10
:
e0126691
.
45.
Verma
A
,
Mohindru
M
,
Deb
DK
,
Sassano
A
,
Kambhampati
S
,
Ravandi
F
, et al
Activation of Rac1 and the p38 mitogen-activated protein kinase pathway in response to arsenic trioxide
.
J Biol Chem
2002
;
277
:
44988
95
.
46.
Ma
Y
,
Chapman
J
,
Levine
M
,
Polireddy
K
,
Drisko
J
,
Chen
Q
. 
High-dose parenteral ascorbate enhanced chemosensitivity of ovarian cancer and reduced toxicity of chemotherapy
.
Sci Transl Med
2014
;
6
:
222ra18
.
47.
Cimmino
L
,
Dolgalev
I
,
Wang
Y
,
Yoshimi
A
,
Martin
GH
,
Wang
J
, et al
Restoration of TET2 function blocks aberrant self-renewal and leukemia progression
.
Cell
2017
;
170
:
1079
1095.e20
.
48.
Agathocleous
M
,
Meacham
CE
,
Burgess
RJ
,
Piskounova
E
,
Zhao
Z
,
Crane
GM
, et al
Ascorbate regulates haematopoietic stem cell function and leukaemogenesis
.
Nature
2017
;
549
:
476
481
.
49.
Mingay
M
,
Chaturvedi
A
,
Bilenky
M
,
Cao
Q
,
Jackson
L
,
Hui
T
, et al
Vitamin C-induced epigenomic remodelling in IDH1 mutant acute myeloid leukaemia
.
Leukemia
2017
.
doi:10.1038/leu2017.171
50.
Jan
M
,
Snyder
TM
,
Corces-Zimmerman
MR
,
Vyas
P
,
Weissman
IL
,
Quake
SR
, et al
Clonal evolution of preleukemic hematopoietic stem cells precedes human acute myeloid leukemia
.
Sci Transl Med
2012
;
4
:
149ra118
.

Supplementary data