Cancer stem cells (CSC) maintain both undifferentiated self-renewing CSCs and differentiated, non-self-renewing non-CSCs through cellular division. However, molecular mechanisms that maintain self-renewal in CSCs versus non-CSCs are not yet clear. Here, we report that in a transgenic mouse model of MYC-induced T-cell leukemia, MYC, maintains self-renewal in Sca1+ CSCs versus Sca-1 non-CSCs. MYC preferentially bound to the promoter and activated hypoxia-inducible factor-2α (HIF2α) in Sca-1+ cells only. Furthermore, the reprogramming factors, Nanog and Sox2, facilitated MYC regulation of HIF2α in Sca-1+ versus Sca-1 cells. Reduced expression of HIF2α inhibited the self-renewal of Sca-1+ cells; this effect was blocked through suppression of ROS by N-acetyl cysteine or the knockdown of p53, Nanog, or Sox2. Similar results were seen in ABCG2+ CSCs versus ABCG2 non-CSCs from primary human T-cell lymphoma. Thus, MYC maintains self-renewal exclusively in CSCs by selectively binding to the promoter and activating the HIF2α stemness pathway. Identification of this stemness pathway as a unique CSC determinant may have significant therapeutic implications.

Significance:

These findings show that the HIF2α stemness pathway maintains leukemic stem cells downstream of MYC in human and mouse T-cell leukemias.

A hallmark of many tumors is the capacity to maintain a stable population of cancer stem cells (CSC) during multiple generations (1). This is attributed to CSC's ability to undergo asymmetric cellular division, where one daughter cell retains self-renewal ability, whereas the other daughter cell differentiates into non-CSCs, composing the bulk of the tumor (2). Numerous studies demonstrate that CSCs retain this ability of selective or exclusive self-renewal through asymmetric cellular division even after numerous serial transplantations and maintain a stable proportion of CSCs (3, 4). Hence, this maintenance of a stable proportion of CSCs via asymmetric division suggests a revision in the notion of the clonal evolution in cancer (2, 3, 5, 6).

Various mechanisms have been proposed by which CSCs maintain asymmetric self-renewal, including cell polarity, fate determinants, microenvironment modulation (4, 7, 8), phenotypic equilibrium (9), and activation of developmental pathways such as Notch and Wnt (1, 3, 4, 10). In addition, gene products that can confer self-renewal in cancer have been identified including the iPS gene products MYC, Nanog, Sox2, Oct-4, as well as hypoxia-inducible factors (HIF; refs. 11–23). However, it is not clear how MYC and other iPS genes cooperate with HIFs to maintain self-renewal in CSCs versus non-CSCs.

The MYC oncogene plays an important role in the self-renewal of normal stem cells and CSCs (22, 24, 25). MYC is a transcription factor that regulates gene expression. When overexpressed, MYC commonly contributes to human cancer (11, 14). MYC induces an embryonic stem cell signature in CSCs (26). While in cooperation with other iPS genes such as Sox2, Nanog, and Oct-4, MYC elicits reprograming of differentiated cells enabling self-renewal (27) and thereby modulating the iPS genes (19, 28). MYC cooperate with HIF2α (29, 30), a stemness-associated transcription factor that increases self-renewal of embryonic stem cells through coordinated upregulation of Oct-4 and Nanog (31, 32), and the negative regulation of p53 (33). Hence, MYC through interaction with HIF2α and iPS genes could regulate exclusive self-renewal of CSCs.

We investigated self-renewal of CSCs in a transgenic mouse model of MYC-induced T-cell acute lymphocytic lymphoma (T-ALL; refs. 34, 35) and human lymphoma. In MYC-induced T-ALL, we identified Sca-1+ CSCs that exhibit dependency on HIF2α for self-renewal. In CSCs but not non-CSCs, MYC preferentially binds to the promoter and activates transcription of HIF2α that is facilitated by Nanog and Sox-2. Finally, MYC mediated activation of HIF2α in ABCG2+ but not ABCG2 human lymphoma CSCs. Our observations thereby suggest that MYC maintains exclusive self-renewal of CSCs by preferential activation of HIF2α in CSC versus non-CSCs.

Details of Materials and Methods are provided in the Supplementary Method section.

Sca-1 cell sorting of MYC-induced transgenic lymphoma

All the necessary experimental procedures were approved and undertaken in accordance with guidelines of Stanford University (Stanford, CA), Forsyth Institute (Cambridge, MA), Gauhati University (Guwahati, Assam, India), and KaviKrishna Laboratory (Guwahati, Assam, India) institutional animal ethics committee. Seven such transgenic mice were selected for the study and genotype confirmed (Supplementary Table S1). The generation and genotyping of Eu-tTA/tetO-MYC system transgenic lines for conditional MYC-driven lymphoma has been used as described previously (34). The thymus obtained from moribund animals was dissociated to flow cytometry or immunomagnetic sort Sca-1+ cells (36), and these cells were expanded in serum-free media containing IL7 and stem cell factors, and then subjected to phenotypic analysis. Multi-color flow cytometry for HIF2α (#NB100-132, Novus Biologicals) and Nanog (#ab184609, Abcam) was done as described previously (33).

Measurement of intracellular glutathione, reactive oxygen species, apoptosis, and proliferation

Glutathione (GSH) and reactive oxygen species (ROS) levels were measured as described previously (37), whereas apoptosis was measured by calorimetric assay as per the manufacturer's instructions. Analysis of the relative cell number was performed by using Alamar blue assay as described previously (37).

Measurement of senescence-associated β-galactosidase activity

The fluorescence method using 5-dodecanoylaminofluorescein di-β-d-galactopyranoside (C12FDG; Thermo Fisher Scientific, #D2893) was used as described previously (38). MYC-inactivated lymphoma cells served as positive control for senescence (39). Details are given in Supplementary Materials and Methods.

Clonogenic assay

It was performed in methylcellulose medium (Methocult M3134, Stem Cell Technologies) as described previously (15, 37).

IHC and Western blot analysis

IHC was done using a Vector Mouse on Mouse Elite Peroxidase Immunodetection Kit (Vector Laboratories, Inc.) according to the manufacturer's instructions (40). Mouse HIF2α (Novus Biologicals) antibody was used in 1:500 dilution. Western blot analysis was done as previously described by using mouse HIF2α (#NB-132; Novus Biologicals) and mouse β-actin (Cell Signaling Technology; ref. 40).

Real-time PCR and specific inhibition of genes

The real-time PCR (qPCR) was performed using TaqMan gene expression assay as described previously (33). The in vitro inhibition of mouse HIF2α, HIF1α, p53, Sox2, and Nanog were achieved by Accell siRNA obtained from GE Healthcare Dharmacon Inc. as described previously (33). The in vivo inhibitions of HIFs and p53 were achieved by FM19G11 (Millipore; 5 mg/kg i.p.) and pifithrin α (Sigma-Aldrich; 4 mg/kg i.p.) after dissolving in 1.5 % DMSO, and given three times weekly for 2 weeks (41). Inhibition of ROS was accomplished by N-acetylcysteine (NAC; 100 mg/kg i.p.) 30 minutes before the injection of FM19G11.

Chromatin immunoprecipitation and re-ChIP assays

Chromatin immunoprecipitation (ChIP) was performed using the Magnify chromatin IP Kit (#1626969, Invitrogen) according to the manufacturer's instructions. Briefly, Sca-1+ cells were fixed with 1% formaldehyde, sonicated to produce DNA fragments of approximately 100–600 bp and then subjected to immunoprecipitation (19) with c-MYC (2 μg, N-262, #sc-764, Santa Cruz Biotechnology), as well as isotype-matched control for rabbit IgG (NB810-56910; Novus Biologicals) as described previously (42). The mouse and human DNA products were subjected to SYBR green PCR using EpiTect ChIP qPCR Primers (SABiosciences, Qiagen). Results were analyzed using percent input method, where 1% of starting chromatin was used as input (42). For re-ChIP analysis, chromatin products from first immunoprecipitation were treated with 10 mmol/L DTT for 30 minutes at 37°C to prevent the majority of the first antibody from participating in the second immunoprecipitation reaction. Eluates were diluted in dilution buffer and then used for second immunoprecipitation by Nanog (#5232, Cell Signaling Technology) rabbit antibody, as described previously (42).

Collection of primary T-cell lymphoblastic lymphoma leukemia cells, ABCG2+ cell sorting, and xenotransplantation in NOD/SCID mice

Peripheral blood and lymph node samples from patients with leukemia (Supplementary Table S2) were obtained after written informed consent and with the approval from the institutional research ethics committee of respective institutions and Declaration of Helsinki. The approved institutions are Stanford University School of Medicine (Stanford, CA), KaviKrishna Telemedicine Care, a branch of Kavi Krishna Laboratory, Dr. B. Borooah Cancer Institute, and Gauhati University (Guwahati, Assam, India). The collection and expansion of T-cell lymphoblastic lymphoma (T-LBL) samples (n = 6, Supplementary Table S2), immunomagnetic sorting of ABCG2+, and in vivo transplantation in NOD/SCID mice were performed as described previously (20, 43, 44). Mice were prior treated with 22 mg/kg i.p. busulfan (6 mg/mL injection, Taj Pharma) daily over 3 days (20). The human cell engraftment was confirmed by flow cytometry staining with a human CD45 antibody (BioLegend).

Mutant HIF2α transfection assay

ABCG2+ cells were transfected using the JetPEI Reagent (Polyplus transfection) with plasmids encoding constitutively active HIF2α mutant, the HA-HIF-2α-P405A/P531A (gift from Professor William Kaelin, Harvard Medical School, Cambridge, MA; Addgene plasmid #18956; ref. 45) and confirmed by Western blot analysis. The control circular plasmid encoding HA with no HIFα insert was generated as described previously (46).

Statistical analysis

The statistical calculations were performed with GraphPad Prism 4.0 (Hearne Scientific Software) using Student t test and One-Way ANOVA with Dunnett post-hoc test. The data on the in vivo limiting dilution assay was analyzed by the extreme limiting dilution analysis software, available online (http://bioinf.wehi.edu.au/software/elda/). Statistical comparison of Kaplan–Meier curves was based on the log-rank test.

MYC-induced lymphoma contains rare Sca-1+ CSCs

In a transgenic mouse model of Tet system regulated MYC-induced T-ALL, we have examined whether there was an identifiable CSC population (34) that activates HIF2α pathway (Fig. 1A). We used multiple transplanted T-ALL tumors (D9476, D9482, E0366, E2824, E2825, E6550, and D9479). We confirmed that all tumors exhibited clonal expression of the T-cell receptor Vβ 2 or 4 (Supplementary Table S1), as demonstrated previously (34).

Figure 1.

Sca-1+ versus Sca-1 cells of MYC-driven T-cell lymphoma exhibit self-renewal. A, Hypothesis: HIF2α pathway is differentially activated in CSC versus non-CSCs in MYC-driven T-ALL. B, Representative flow cytometry panels of Sca-1+/CD4+/CD8+/CD19 cells in MYC-induced thymic tumor (D9476 parental monoclonal tumor; Supplementary Table S1). C, qPCR analysis of stemness-related gene expression; five independent samples from each parental tumor. *, P < 0.05; **, P < 0.001 comparing with control (Student t test). D, Demarcated tumor images from monoclonal serial transplantation assay. Number of mice with tumor/total number of injection is given below each image (additional images in Supplementary Fig. S2). E, Flow cytometry quantification of Sca-1+ cell frequency in parental, primary (1°), secondary (2°), and tertiary (3°) tumors (five independent samples; details in Supplementary Table S3A). F, Ratio of CSC frequency in serially transplanted tumors. ***, P < 0.0001 (analyzed by ELDA; Supplementary Table S4). Data in C and E represents mean ± SEM.

Figure 1.

Sca-1+ versus Sca-1 cells of MYC-driven T-cell lymphoma exhibit self-renewal. A, Hypothesis: HIF2α pathway is differentially activated in CSC versus non-CSCs in MYC-driven T-ALL. B, Representative flow cytometry panels of Sca-1+/CD4+/CD8+/CD19 cells in MYC-induced thymic tumor (D9476 parental monoclonal tumor; Supplementary Table S1). C, qPCR analysis of stemness-related gene expression; five independent samples from each parental tumor. *, P < 0.05; **, P < 0.001 comparing with control (Student t test). D, Demarcated tumor images from monoclonal serial transplantation assay. Number of mice with tumor/total number of injection is given below each image (additional images in Supplementary Fig. S2). E, Flow cytometry quantification of Sca-1+ cell frequency in parental, primary (1°), secondary (2°), and tertiary (3°) tumors (five independent samples; details in Supplementary Table S3A). F, Ratio of CSC frequency in serially transplanted tumors. ***, P < 0.0001 (analyzed by ELDA; Supplementary Table S4). Data in C and E represents mean ± SEM.

Close modal

To identify potential CSC markers, we first examined the expression of multiple thymic lymphocyte lineage markers. T-cell development in the thymus involves migration of bone marrow cells that differentiate into early thymocyte progenitors (ETP) and express Sca-1, c-kit, and CD44 (36). We found that a small number of tumor cells expressed Sca-1 (Fig. 1B, left), c-kit, and CD44 (Supplementary Fig. S1A; Supplementary Table S3A and S3B). The Sca-1+ cells were positive for both CD4 and CD8, but did not express B-cell maker, CD19 (Fig. 1B, right; Supplementary Table S3A; ref. 34). Hence, a small number of tumor cells expressing Sca-1 could be identified as potential CSC population.

Next, we examined whether Sca-1+ versus Sca-1 cells differentially expressed other stem cell genes including: HIF2α, and the pluripotency factors Nanog, Sox2, and Oct4 (33). Sca-1+ cells exhibited 6- to 10-fold higher expression of all of these genes except Oct4, which was not significantly expressed (Fig. 1C; Supplementary Fig. S1A and S1B; Supplementary Table S3B). HIF1α and p53 were downregulated by 4- to 6-fold, whereas the level of endogenous MYC remained consistent in Sca-1+ versus Sca-1 cells (Fig. 1C; Supplementary Fig. S1C). Flow cytometry confirmed the qPCR results (Supplementary Fig. S1A and S1B; Supplementary Table S3A and S3B). Thus, Sca-1+ versus Sca-1 cells exhibited differential expression of stemness associated gene products.

Next, we evaluated the self-renewal capacity of Sca-1+ cells by performing in vivo limiting dilution assay. Flow cytometry–sorted Sca-1+ and Sca-1 cell population from parental tumors were transplanted into immunocompetent syngeneic mice. Intravenous injection of 10 Sca-1+ cells but 1 × 106 Sca-1 cells resulted in engraftment of lymphoma in recipient syngeneic mice (Fig. 1D, left; Supplementary Fig. S2A–S2C; Supplementary Table S4). The maximum likelihood analysis revealed that Sca-1+ cells exhibit 5,724-fold higher tumorigenic capacity in comparison with Sca-1 cells (P < 0.00001; Supplementary Table S4). These Sca-1+ cells–derived primary tumors reestablished parental heterogeneity. The expression of ETPs and stemness markers CD44, C-kit, HIF-2α, Nanog, and Sox2 were consistently high in Sca-1+ versus Sca-1 cells. Whereas the expression of p53 and HIF1α remained low, similar to parental tumors (Supplementary Table S3B). Thus, we have identified Sca-1+ cell population enriched with CSCs in MYC-induced lymphoma.

Sca-1+ cells maintain exclusive self-renewal during serial transplantation

To evaluate whether Sca-1+ cells maintain stable frequency and self-renewal state during serial transplantation, a monoclonal transplantation assay was performed where tumors were derived from a single CSC (47). For this purpose, Sca-1+ cells were isolated from multiple primary tumors: D9476, E6550, D9479, and E0366 (Fig. 1D left; Supplementary Fig. S2A–S2C). A single Sca-1+ cell from these primary tumors was then injected to syngeneic mice to obtain secondary tumors or further reinjected to obtain tertiary tumors. We found secondary and tertiary tumors derived from each of the four parental tumors maintained stable proportion of Sca-1+ cells, and their phenotype including the high HIF2α protein expression in comparison with Sca-1 cells (Fig. 1D and E; Supplementary Fig. S2D; Supplementary Table S3A and S3B). In addition, Sca-1 cells from these tumors expressed TCR V β expression as parental Sca-1+ cells (Supplementary Tables S1 and S3C). Then, Sca-1+ and Sca-1 cells from these primary, secondary, and tertiary tumors were subjected to in vivo limiting dilution assay to obtain CSCs proportions. We found that the ratio of CSC frequency between Sca-1+ versus Sca-1 cells remained stable. (Fig. 1F; Supplementary Table S4). Our results suggest that Sca-1+ CSCs maintain stable proportion during serial transplantation.

A stable proportion of CSC may be maintained either via asymmetric self-renewal or by conversion of non-CSC to CSCs by the process of phenotypic equilibrium (7, 9). To address this possibility, we subjected Sca-1 cells from primary, secondary, and tertiary tumors (Fig. 1D–F) to in vivo limiting dilution assay. Interestingly, CSC frequency did not become elevated during serial transplantation (Supplementary Table S4), suggesting the unlikely contribution of Sca-1 cells to CSC frequency.

HIF2α pathway is essential to maintain the stemness of Sca-1+ cells

Inactivation of MYC by doxycycline treatment downregulates HIF2α without significantly changing HIF1α in Sca-1+ cells (Supplementary Fig. S3A), suggesting that HIF2α pathway may be required to maintain the stemness state of Sca-1+ cells. By Flow cytometry, the expression of HIF2α, Nanog, and Sox2 was found to be consistently higher in Sca-1+ cells of serially transplanted tumors, while the expression of HIF1α and p53 remained low in the Sca-1+ cells (Fig. 2A; Supplementary Table S3B). Thus, Sca-1+ cells maintained high HIF2α during serial transplantation. In contrast, the Sca-1 cells maintained low HIF2α expression (Supplementary Fig. S2D). Thus, serially transplanted Sca-1+ cells but not Sca-1 cells maintain high HIF2α, suggesting the potential role of HIF2α in the exclusive self-renewal of Sca-1+ versus Sca-1 cells.

Figure 2.

Fate of Sca-1+ cells following HIF2α gene silencing. HIF2α silencing was performed by treating Sca-1+ cells with 1 μmol/L of Accell HIF2α siRNA for 3 days and various assays (B–H) were performed on day 4. A, Flow cytometry analysis reports expression of the indicated transcription factors in primary (1°) and secondary (2°) single Sca-1+ cell–derived tumors (details in Supplementary Table S3B). B, qPCR analysis of indicated genes in HIF2α-silenced Sca-1+ cells. NT, nontargeted siRNA. C, Alamar blue proliferation assay result. D, Sca-1+ cell phenotype following HIF2α inactivation. Values were compared with nontargeted group. E, Representative flow cytometry panels of GSH (monochlorobimane, mBCl), DCFH-DA (ROS), and p53-stained Sca-1+ cells. T, targeted (siRNA HIF2α treated); gray dots, isotype control. Quantification of the results is given in Supplementary Table S5A. F, Levels of p53 and Sca-1 (ELISA), and GSH and ROS (calorimetric methods) following HIF2α silencing. G, Western blot to quantitate p53 levels in siRNA HIF2α and p53-silenced cells. H, siRNA HIF2α-treated cells were cotreated with NAC (1 mmol/L) or siRNA p53 to perform rescue experiment (additional data in Supplementary Fig. S3). Data in A–D and F–H are presented as mean ± SEM of at least four independent samples. *, P < 0.05; **, P < 0.001; ***, P < 0.0001; ANOVA and Dunnett post hoc test.

Figure 2.

Fate of Sca-1+ cells following HIF2α gene silencing. HIF2α silencing was performed by treating Sca-1+ cells with 1 μmol/L of Accell HIF2α siRNA for 3 days and various assays (B–H) were performed on day 4. A, Flow cytometry analysis reports expression of the indicated transcription factors in primary (1°) and secondary (2°) single Sca-1+ cell–derived tumors (details in Supplementary Table S3B). B, qPCR analysis of indicated genes in HIF2α-silenced Sca-1+ cells. NT, nontargeted siRNA. C, Alamar blue proliferation assay result. D, Sca-1+ cell phenotype following HIF2α inactivation. Values were compared with nontargeted group. E, Representative flow cytometry panels of GSH (monochlorobimane, mBCl), DCFH-DA (ROS), and p53-stained Sca-1+ cells. T, targeted (siRNA HIF2α treated); gray dots, isotype control. Quantification of the results is given in Supplementary Table S5A. F, Levels of p53 and Sca-1 (ELISA), and GSH and ROS (calorimetric methods) following HIF2α silencing. G, Western blot to quantitate p53 levels in siRNA HIF2α and p53-silenced cells. H, siRNA HIF2α-treated cells were cotreated with NAC (1 mmol/L) or siRNA p53 to perform rescue experiment (additional data in Supplementary Fig. S3). Data in A–D and F–H are presented as mean ± SEM of at least four independent samples. *, P < 0.05; **, P < 0.001; ***, P < 0.0001; ANOVA and Dunnett post hoc test.

Close modal

Hence, we evaluated the role of HIF2α in the self-renewal of Sca-1+ cells. Suppression of HIF2α but not HIF1α expression by siRNA resulted in decreased Nanog and Sox2, but increased p53 expression (Fig. 2B; Supplementary Fig. S3A and S3B). These changes were associated with decreased HIF2α protein levels, in vitro cell growth, but no change in cell death (Fig. 2B and C; Supplementary Fig. S3C and S3D). Suppression of HIF2α but not HIF1α was associated with changes in senescence and differentiation markers including reduced BrdU incorporation and increased β-galactosidase staining (Fig. 2D) p21, and p16 (39) and decreased expression of CD44 and C-kit (Supplementary Fig. S3E). Apoptosis measured by caspase-3 activity was not altered (Fig. 2D). Therefore, HIF2α is required to maintain the stemness state of Sca-1+ cells.

HIF2α maintains the self-renewal of Sca-1+ cells by suppressing p53 and ROS

In embryonic stem cells, HIF2α negatively regulated p53 and ROS to maintain an undifferentiated state of high GSH redox state (33). We explored whether HIF2α employs similar mechanisms of maintaining low p53 and ROS levels in Sca-1+ cells. Flow cytometry–based evaluation of p53, ROS, and GSH were performed in Sca-1+ cells subjected to HIF2α silencing. Interestingly, suppression of HIF2α indicated decreased GSH, whereas increased p53 and ROS levels in Sca-1+ cells. Furthermore, these changes were accompanied by loss of Sca-1 expression (Fig. 2E and F; Supplementary Table S5A). Therefore, HIF2α appears to negatively regulate p53 and ROS for maintaining the stemness state of Sca-1+ cells. Indeed, siRNA p53 gene silencing blocked HIF2α silencing from inducing proliferative arrest or senescence in Sca-1+ cells (Fig. 2G and H; Supplementary Fig. S3F). Similarly, inhibition of ROS by cotreatment with NAC blocked p53 induction following HIF2α silencing (Supplementary Fig. S3F) and proliferative arrest or senescence (Fig. 2H). Hence, HIF2α suppresses both p53 and ROS to maintain proliferation of Sca-1+ cells.

Next, we examined whether HIF2α was required for the self-renewal of Sca-1+ cells by injecting these cells to immunocompetent mice. We measured in vivo tumorigenic growth and self-renewal of HIF2α-silenced Sca-1+ cells with or without p53 silencing or NAC treatment. HIF2α silencing in Sca1+ cells decreased tumorigenic growth by 115-fold (Fig. 3A and B; Supplementary Table S5B) and self-renewal by 11-fold (Fig. 3C–E; Supplementary Table S5C and S5D). However, concomitant NAC treatment or p53 silencing restored both tumorigenic growth and self-renewal (Fig. 3B–E; Supplementary Table S5C and S5D). Importantly, HIF2α silencing did not prevent Sca-1+ cells from entering the thymus (Supplementary Fig. S4A–S4C). Thus, HIF2α appears to maintain the self-renewal of Sca-1+ cells by negative regulation of ROS and p53.

Figure 3.

HIF2α maintains self-renewal of Sca-1+ cells by negative regulation of p53 and ROS. A, Sca-1+ cells were treated with siRNA HIF2α with or without NAC (1 mmol/L) or siRNA p53 for 3 days and on day 4 cells were injected into syngeneic mice to perform limiting dilution analysis (see details in Supplementary Table S5). B, NAC or siRNA p53 silencing rescued CSC frequency of HIF2α-silenced Sca-1+ cells (Supplementary Table S5B). C–E, Self-renewal capacity was evaluated by performing serial transplantation assay. A total of 5 × 103 cells of each treatment group were injected intravenously, and mice were sacrificed after 10 weeks to find thymic tumors. Thymic tumors were dissociated and Sca-1+ cells were flow cytometry sorted and subjected to limiting dilution assay to estimate CSC frequency. Representative images of recovered thymic tumors are presented with demarcated circle. Inset shows number of mice with tumors per total number of injection. D, Representative flow cytometry panels indicate percent of Sca-1+ cells in recovered tumors. E, CSC frequency of tumor cells recovered from the tumors shown in C (Supplementary Table S5C and S5D). ***, P < 0.0001; ELDA; error bar, 95% confidence intervals.

Figure 3.

HIF2α maintains self-renewal of Sca-1+ cells by negative regulation of p53 and ROS. A, Sca-1+ cells were treated with siRNA HIF2α with or without NAC (1 mmol/L) or siRNA p53 for 3 days and on day 4 cells were injected into syngeneic mice to perform limiting dilution analysis (see details in Supplementary Table S5). B, NAC or siRNA p53 silencing rescued CSC frequency of HIF2α-silenced Sca-1+ cells (Supplementary Table S5B). C–E, Self-renewal capacity was evaluated by performing serial transplantation assay. A total of 5 × 103 cells of each treatment group were injected intravenously, and mice were sacrificed after 10 weeks to find thymic tumors. Thymic tumors were dissociated and Sca-1+ cells were flow cytometry sorted and subjected to limiting dilution assay to estimate CSC frequency. Representative images of recovered thymic tumors are presented with demarcated circle. Inset shows number of mice with tumors per total number of injection. D, Representative flow cytometry panels indicate percent of Sca-1+ cells in recovered tumors. E, CSC frequency of tumor cells recovered from the tumors shown in C (Supplementary Table S5C and S5D). ***, P < 0.0001; ELDA; error bar, 95% confidence intervals.

Close modal

We examined whether the in vivo inactivation of HIF2α through the small-molecule FM19G11 influenced tumor growth. Tumors in mice treated with FM19G11 showed increased survival comparable with vehicle-treated group (Fig. 4A). Importantly, Sca-1+ cells from FM19G11-treated tumors indicated increased p53 but decreased HIF2α, as well as self-renewal capacity of Sca-1+ cells (Fig. 4B and C; Supplementary Table S6Aand S6B). FM19G11 combined with NAC reduced the p53 and ROS levels, increased GSH and rescued the self-renewal capacity of Sca-1+ cells (Fig. 4 D–F; Supplementary Table S6A and S6B). This further supports the conclusion that HIF2α is required for self-renewal of Sca-1+ cells.

Figure 4.

In vivo inhibition of HIF2α in Sca-1+ cells by FM19G11 small-molecular inhibitor. Three weeks after the injection of 5,000 Sca-1+ cells intravenously, syngeneic mice were treated with FM19G11 (5 mg/kg; three times weekly) or vehicle for 2 weeks. Animals were either observed for long-term (A) or sacrificed to recover Sca-1+ cells from the treated tumors (B–G). A, Kaplan–Meier survival estimation; n = 10 animals in each group. MYC dependency was confirmed by doxycycline treatment. Difference between vehicle and FM19G11 treatment was calculated using log-rank test; P < 0.001. B, Sca-1+ cells frequency as measured by flow cytometry (five independent samples). C, Western blot analysis indicates sustained decrease of intracellular HIF2α and gradual increase of p53 levels. D, CSC frequency following FM19G11 treatment (Supplementary Table S6A and S6B). E, Flow cytometry analysis of FM19G11 ± NAC (100 mg/kg)-treated tumor cells. Anti-cleaved caspase-3 (Asp175, Cell Signaling Technology) was used to stain apoptotic cells. F, Sca-1+ cells in E were evaluated for shown values and compared with untreated Sca-1+ cells. G, Phenotypes (right) and limiting dilution assay (left) of tumors that escaped FM19G11 sensitivity (as shown in A). F and G, Phenotype data are presented as mean ± SEM, four independent samples. Protein levels were measured by ELISA. Raw data of limiting dilution is given in Supplementary Table S6C. *, P < 0.05; **, P < 0.001; ***, P < 0.0001 (Student t test); error bar, 95% confidence intervals.

Figure 4.

In vivo inhibition of HIF2α in Sca-1+ cells by FM19G11 small-molecular inhibitor. Three weeks after the injection of 5,000 Sca-1+ cells intravenously, syngeneic mice were treated with FM19G11 (5 mg/kg; three times weekly) or vehicle for 2 weeks. Animals were either observed for long-term (A) or sacrificed to recover Sca-1+ cells from the treated tumors (B–G). A, Kaplan–Meier survival estimation; n = 10 animals in each group. MYC dependency was confirmed by doxycycline treatment. Difference between vehicle and FM19G11 treatment was calculated using log-rank test; P < 0.001. B, Sca-1+ cells frequency as measured by flow cytometry (five independent samples). C, Western blot analysis indicates sustained decrease of intracellular HIF2α and gradual increase of p53 levels. D, CSC frequency following FM19G11 treatment (Supplementary Table S6A and S6B). E, Flow cytometry analysis of FM19G11 ± NAC (100 mg/kg)-treated tumor cells. Anti-cleaved caspase-3 (Asp175, Cell Signaling Technology) was used to stain apoptotic cells. F, Sca-1+ cells in E were evaluated for shown values and compared with untreated Sca-1+ cells. G, Phenotypes (right) and limiting dilution assay (left) of tumors that escaped FM19G11 sensitivity (as shown in A). F and G, Phenotype data are presented as mean ± SEM, four independent samples. Protein levels were measured by ELISA. Raw data of limiting dilution is given in Supplementary Table S6C. *, P < 0.05; **, P < 0.001; ***, P < 0.0001 (Student t test); error bar, 95% confidence intervals.

Close modal

However, we noted that 4 of 10 tumor-bearing mice treated with FM19G11 exhibited morbidity within 20–30 weeks (Fig. 4A), suggesting continuing growth of thymic tumors despite treatment. Sca-1+ versus Sca-1 cells recovered from these tumors maintained the expression of high HIF2α and low p53, whereas MYC level remained equal (Fig. 4G). Furthermore, Sca-1+ versus Sca-1 cells of these post-FM19G11–treated tumors maintained high tumorigenic capacity (Supplementary Table S6C). This suggests that Sca-1+ cells that escape sensitivity to FM19G11 did not evolve to a different phenotype but maintained the high HIF2α phenotype.

MYC through Nanog and Sox2 regulates HIF2α in Sca-1+ versus Sca-1 cells

We examined MYC's role in the exclusive regulation of HIF2α in Sca-1+ versus Sca-1 cells. MYC was found to be equally expressed in Sca-1+ and Sca-1 cells, obtained from a single Sca-1+ cell–derived tumor #D9476 (Figs. 1D and 5A) and expressing TCR V β 4 (Supplementary Table S3C). We infer that other transcription factors may be required for differential regulation of HIF2α in Sca-1+ versus Sca-1 cells. One possibility was that the iPS genes Nanog and Sox2 might be involved in this MYC-mediated regulation of HIF2α, because they both were found to be highly expressed in Sca-1+ but not Sca-1 cells (Fig. 1C; Supplementary Fig. S1A–S1C; Supplementary Table S3B). Indeed, we found that MYC extensively binds to the promoters of HIF2α, Nanog, and Sox2 but not Oct4 in Sca-1+ versus Sca-1 cells (Fig. 5B). Notably, MYC binds preferentially to the HIF1α promoters in Sca-1 versus Sca-1+ cells (Fig. 5B). Suppression of MYC expression decreased HIF2α, Nanog, and Sox-2, but upregulated HIF1α expressions in Sca-1+ versus Sca-1 cells (Fig. 5C). Thus, MYC preferentially bound and regulated the expression of HIF2α, Sox-2, and Nanog in Sca-1+ versus Sca-1 cells.

Figure 5.

MYC interacts with Nanog and Sox2 to induce HIF2α in Sca-1+ versus Sca-1 cells. A, ELISA based analysis of Sca-1 and MYC proteins in Sca-1+ and Sca-1 cells derived from a monoclonal tumor (#D9476, Fig. 1D). B, qPCR analysis of MYC ChIP data in Sca-1+ and Sca-1 cell samples at the indicated promoters. 1GX1A served as negative control, whereas ODC1, a MYC target gene, served as positive control. The IgG panel represented matched isotype. Data was quantified as percentage of input DNA (N = 3 independent samples; Student t test). C, qPCR data of indicated genes in Sca-1+ and Sca-1 cells following MYC inactivation upon doxycycline treatment (N = 4 independent experiments; Student t test). D, qPCR analysis of MYC ChIP in Sca-1+ cells at the HIF2α promoter following siRNA silencing of Nanog and Sox2. E,HIF2α protein (ELISA) following cosilencing of Nanog and Sox2 (N = 4 independent experiments; Student t test). F, Clonogenic capacity following cosilencing of Nanog and Sox2 (Supplementary Fig. S5). G, ELISA for Nanog and Sox2 cosilenced Sca-1+ cells. E–G,N = 4 independent samples; ANOVA and Dunnett post hoc test. Data are presented as mean ± SEM. *, P < 0.05; **, P < 0.001.

Figure 5.

MYC interacts with Nanog and Sox2 to induce HIF2α in Sca-1+ versus Sca-1 cells. A, ELISA based analysis of Sca-1 and MYC proteins in Sca-1+ and Sca-1 cells derived from a monoclonal tumor (#D9476, Fig. 1D). B, qPCR analysis of MYC ChIP data in Sca-1+ and Sca-1 cell samples at the indicated promoters. 1GX1A served as negative control, whereas ODC1, a MYC target gene, served as positive control. The IgG panel represented matched isotype. Data was quantified as percentage of input DNA (N = 3 independent samples; Student t test). C, qPCR data of indicated genes in Sca-1+ and Sca-1 cells following MYC inactivation upon doxycycline treatment (N = 4 independent experiments; Student t test). D, qPCR analysis of MYC ChIP in Sca-1+ cells at the HIF2α promoter following siRNA silencing of Nanog and Sox2. E,HIF2α protein (ELISA) following cosilencing of Nanog and Sox2 (N = 4 independent experiments; Student t test). F, Clonogenic capacity following cosilencing of Nanog and Sox2 (Supplementary Fig. S5). G, ELISA for Nanog and Sox2 cosilenced Sca-1+ cells. E–G,N = 4 independent samples; ANOVA and Dunnett post hoc test. Data are presented as mean ± SEM. *, P < 0.05; **, P < 0.001.

Close modal

Next, we examined whether MYC regulation of HIF2α expression was mediated by Nanog and/or Sox-2. We found that co-siRNA silencing of Nanog and Sox2 decreased MYC binding to HIF2α, reduced HIF2α protein levels by 4-fold, and decreased the self-renewal capacity without altering MYC protein level (Fig. 5D–G; Supplementary Fig. S5A–S5C). Thus, MYC cooperates with Nanog and Sox2 to increase HIF2α expression and regulate self-renewal in Sca-1+ versus Sca-1 cells.

MYC through HIF2α maintains self-renewal in human lymphoma

To confirm our findings from transgenic mouse model system in a human equivalent tumor, we first identified the equivalent ABCG2+/HIF2α CSC subpopulation in relapsed cases of human adult acute lymphoblastic lymphoma (T-LBL). ABCG2, a stem cell marker regulated by HIF2α (33), was highly expressed in patients with T-LBL treated with chemotherapy/radiation (48). Our results indicated that T cells recovered from cervical lymph nodes and peripheral blood of the subjects (n = 6; Supplementary Table S2) were highly enriched in ABCG2+/HIF2α+ cells (Supplementary Fig. S6A–S6C). These ABCG2+ cells were CD7-positive but CD8- and CD1a-negative (Fig. 6A; Supplementary Fig. S6D), a phenotype associated with T-ALL CSCs (49).

Figure 6.

ABCG2+ cells in the human T-LBL subjects are HIF2α dependent. A, Representative flow cytometry panel indicates enrichment and purity analysis of ABCG2+/CD7+ cells from the cervical lymph nodes of T-LBL subject #5 (90% ± 4.5%; n = 4; Supplementary Fig. S6D). B, Engraftment of human CD45 cells in the bone marrow of NOD/SCID mice injected with 5 × 103 ABCG2+ or ABCG2 cells of subject #5 (quantification data is in Supplementary Fig. S7A and S7B). C, Serial transplantation of ABCG2+ cells in NOD/SCID mice (details in Supplementary Fig. S7C and S7D). D, qPCR analysis of indicated gene expression in cells obtained during serial transplantation (five independent samples from each patient; Student t test). E, Effect of NAC or siRNA p53 cotreatment in HIF2α-silenced ABCG2+ cells (four independent experiments, patient #5 and 6; ANOVA with Dunnett test). F and G, Cosilenced ABCG2+ cells (1 × 104) were subjected to serial engraftment (details in Supplementary Fig. S8B). At least three independent samples, Student t test. Data in D–G are presented as mean ± SEM. *, P < 0.05; **, P < 0.001.

Figure 6.

ABCG2+ cells in the human T-LBL subjects are HIF2α dependent. A, Representative flow cytometry panel indicates enrichment and purity analysis of ABCG2+/CD7+ cells from the cervical lymph nodes of T-LBL subject #5 (90% ± 4.5%; n = 4; Supplementary Fig. S6D). B, Engraftment of human CD45 cells in the bone marrow of NOD/SCID mice injected with 5 × 103 ABCG2+ or ABCG2 cells of subject #5 (quantification data is in Supplementary Fig. S7A and S7B). C, Serial transplantation of ABCG2+ cells in NOD/SCID mice (details in Supplementary Fig. S7C and S7D). D, qPCR analysis of indicated gene expression in cells obtained during serial transplantation (five independent samples from each patient; Student t test). E, Effect of NAC or siRNA p53 cotreatment in HIF2α-silenced ABCG2+ cells (four independent experiments, patient #5 and 6; ANOVA with Dunnett test). F and G, Cosilenced ABCG2+ cells (1 × 104) were subjected to serial engraftment (details in Supplementary Fig. S8B). At least three independent samples, Student t test. Data in D–G are presented as mean ± SEM. *, P < 0.05; **, P < 0.001.

Close modal

Serial transplantation assay in NOD/SCID mice showed a 2,000-fold higher self-renewal capacity of ABCG2+ versus ABCG2 cells (Fig. 6B and C; Supplementary Fig. S7A–S7D). Phenotypically, ABCG2+ cells represented undifferentiated lymphoma gene signature with high expression of MYC, HIF2α, Nanog, Sox2, and CD44. p53 expression was lower in ABCG2+ versus ABCG2 cells (Fig. 6D), thereby indicating similar CSC property and stemness phenotype to Sca-1+ cells. Importantly, silencing of HIF2α but not HIF1α in ABCG2+ cells exhibited similar results that we observed in Sca-1+ CSCs (Fig. 6E–G; Supplementary Fig. S8A and S8B). The results suggest that HIF2α maintains the self-renewal of ABCG2+ cells in human equivalent cancer.

We furthered our findings and revealed that MYC differentially binds to promoter of HIF2α in ABCG2+ versus ABCG2 cells while directly regulates the Nanog and Sox2 binding. Inhibition of MYC led to marked decrease of HIF2α, Nanog, and Sox2 expression in ABCG2+ cells (Fig. 7A and B; Supplementary Fig. S8C), indicating that MYC and HIF2α may mediate self-renewal of ABCG2+ cells.

Figure 7.

MYC interacts with Nanog to induce HIF2α in ABCG2+ versus ABCG2 cells. A, qPCR analysis of ChIP-MYC in ABCG2+ and ABCG2 cell samples at the indicated promoters. ODC1, a MYC target gene, served as positive control. The IgG panel represented matched isotype. Data was quantified as percentage of input DNA (N = 4 independent samples; Student t test). B, qPCR data of indicated genes in MYC-silenced ABCG2+ cells. C, qPCR analysis of MYC ChIP in ABCG2+ cells at the HIF2α promoter following siRNA silencing of Nanog and Sox2. (N = 4 independent samples; Student t test). D, Chromatin materials pulled down with MYC primary immunoprecipitates from A was used for re-ChIP with Nanog antibody or IgG. Re-ChIP qPCR signal is shown as fold enrichment relative to the IgG control. E–G, siRNA Nanog-silenced ABCG2+ cells were evaluated for the reversal of GSH, p53, and self-renewal following introduction of a constitutively active mutant HIF2α transgene (Supplementary Fig. S8E). ELISA and calorimetric assay measured proteins and GSH, respectively. For in vivo engraftment, 1 × 104 ABCG2+ cells, patient #5 and #6 were used. Data in D–F are presented as mean ± SEM of four independent experiments. H, A model figure summarizing the results on the identification of the MYC-mediated HIF2α stemness pathway. *, P < 0.05; **, P < 0.001 (Student t test).

Figure 7.

MYC interacts with Nanog to induce HIF2α in ABCG2+ versus ABCG2 cells. A, qPCR analysis of ChIP-MYC in ABCG2+ and ABCG2 cell samples at the indicated promoters. ODC1, a MYC target gene, served as positive control. The IgG panel represented matched isotype. Data was quantified as percentage of input DNA (N = 4 independent samples; Student t test). B, qPCR data of indicated genes in MYC-silenced ABCG2+ cells. C, qPCR analysis of MYC ChIP in ABCG2+ cells at the HIF2α promoter following siRNA silencing of Nanog and Sox2. (N = 4 independent samples; Student t test). D, Chromatin materials pulled down with MYC primary immunoprecipitates from A was used for re-ChIP with Nanog antibody or IgG. Re-ChIP qPCR signal is shown as fold enrichment relative to the IgG control. E–G, siRNA Nanog-silenced ABCG2+ cells were evaluated for the reversal of GSH, p53, and self-renewal following introduction of a constitutively active mutant HIF2α transgene (Supplementary Fig. S8E). ELISA and calorimetric assay measured proteins and GSH, respectively. For in vivo engraftment, 1 × 104 ABCG2+ cells, patient #5 and #6 were used. Data in D–F are presented as mean ± SEM of four independent experiments. H, A model figure summarizing the results on the identification of the MYC-mediated HIF2α stemness pathway. *, P < 0.05; **, P < 0.001 (Student t test).

Close modal

To confirm the role of Nanog and Sox2 in the MYC-mediated regulation of HIF2α in ABCG2+ versus ABCG2 cells, we performed ChIP assay. Inhibition of Nanog but not Sox2 led to marked decrease of MYC binding to HIF2α promoter (Fig. 7C; Supplementary Fig. S8C). Importantly, Nanog binding to the HIF2α promoter was observed in re-ChIP samples of MYC (Fig. 7D), suggesting that MYC and Nanog may cooperate to regulate HIF2α expression. Furthermore, in Nanog-silenced ABCG2+ cells, while MYC protein levels remained unaltered, the HIF2α protein and GSH levels were markedly reduced, p53 and its target genes p21, MDM2, and BAX were induced, and the self-renewal was negatively affected as measured by serial transplantation assay (Fig. 7E–G; Supplementary Fig. S8D). Interestingly, Nanog's role in ABCG2+ cell self-renewal was not independent of HIF2α, because Nanog knockdown ABCG2+ cells could be rescued by the constitutive expression of a HIF2α transgene (Fig. 7F–G; Supplementary Fig. S8E; ref. 45). These results therefore suggest that Nanog participated in MYC regulation of HIF2α in ABCG2+ versus ABCG2 cells to confer exclusive self-renewal capacity and tumor stemness.

CSCs contribute to chemotherapy failure and tumor relapse (2, 3). How CSCs maintain exclusive self-renewal is not clear. Here, we report that Sca-1+ CSCs in transgenic mouse model of MYC-driven lymphoma and ABCG2+ CSCs from lymphoma subjects demonstrate exclusive self-renewal via specific MYC binding to HIF2α promoter regions specifically in CSCs versus non-CSCs. Furthermore, stemness factors Nanog and Sox2 cooperates with MYC to regulate HIF2α that in turn decreases p53 expression and reduces ROS levels in CSCs. Thus, MYC-HIF2α stemness pathway (Fig. 7H) may contribute to the exclusive self-renewal mechanism of CSCs and regulate their frequency during multiple generations.

HIFs signaling have a complex role in cancer self-renewal, where both HIF1α and HIF2α either promote or inhibit the self-renewal of leukemia (10, 20, 21, 50). MYC is a key regulator of pluripotency and differentiation that acts downstream of NOTCH (51), a developmental pathway that cooperates with HIFs to maintain self-renewal of T-ALL (43, 49, 50, 52, 53) Also, MYC maintains self-renewal in stem cells and CSCs through effects on cellular metabolism (14, 24, 29). How MYC cooperates with HIFs to regulate CSC self-renewal, as well as frequency is less understood.

To investigate the role of HIF2α in the self-renewal of MYC-dependent CSCs, we characterized Sca-1+ CSCs in mouse, and human T-lymphoblastic lymphoma patient–derived ABCG2+ CSCs. In these CSCs, HIF2α a transcription factor known to maintain stem cell robustness (15), negatively regulates p53 expression and ROS levels to confer self- renewal capacity. Importantly, our findings reveal that high HIF2α expression in Sca-1+ CSCs is enabled by distinct MYC binding to HIF2α promoter regions. Pluripotency factors such as Nanog and Sox2 together facilitated this binding, thereby conferring selective or exclusive self-renewal capacity to CSCs versus non-CSCs. Nanog and Sox2 are broadly expressed in human cancers (15, 54, 55). Nanog is known to maintain self-renewing T-cell leukemia cells by suppressing p53 (13), however the molecular mechanism was not clear. Now, our findings in human ABCG2+ CSCs suggest that Nanog-mediated p53 suppression was HIF2α dependent. Furthermore, in Sca-1+ cells, we found that Nanog cooperates with Sox2 to facilitate MYC-mediated selective upregulation of HIF2α in CSCs versus non-CSCs. Selective or exclusive MYC binding to HIF2α promoters in human ABCG2+ CSCs versus non-CSCs was facilitated by Nanog alone. Further experiments are required to understand the independent role of Nanog in self-renewal of human CSCs.

Involvement of the iPS factors Oct4, Nanog, and Sox2 in CSC self-renewal may be tumor type specific (6, 15). Only Nanog was required to regulate human ABCG2+ CSCs self-renewal, whereas both Nanog and Sox2 regulated the mouse CSCs self-renewal. Also, MYC protein levels were higher in human ABCG2+ CSCs as compared with mouse CSCs. Interestingly, Oct4, a downstream mediator of HIF2α pathway (31, 33) was neither involved in human or mouse T-ALL–derived CSCs. These observations indicate that the role of these stemness effectors might be tumor specific (6, 11, 12, 15, 17).

Our study is suggestive, but does not confirm the possibility of CSCs asymmetric self-renewal at single-cell level. Indirect evidences indicate that single Sca-1+ cell may give rise to tumors, representing a clonal population of both Sca-1+ and Sca-1 cells with distinct phenotype. Stemness factors, Nanog and Sox2, were involved in regulating HIF2α expression in Sca-1+ versus Sca-1 cells, suggesting the possibility of asymmetric self-renewal and maintenance of CSC frequency. Furthermore, previous observations illustrate that tumors maintain a stable population of CSCs (1) that may be attributed to asymmetric cellular division (2). Similarly, in a MYC-induced T-ALL model in zebrafish and in human CD7+ lymphoma stem cells, CSCs maintained their frequency during serial transplantation (47, 49). Therefore, CSCs may maintain their frequency by undergoing asymmetric self-renewal. Our T-ALL CSCs model of Sca-1+ cell self-renewal may serve as a model to understand the underlying mechanisms that maintain asymmetric self-renewal in cancer.

We identified that MYC and HIF2α negatively regulate p53 and ROS in CSCs and this appears to be required to maintain stemness. Nanog and Sox2 interacted with MYC and HIF2α to maintain balance between stemness and differentiation (Fig. 7H). Thus, targeting this MYC-HIF2α stemness pathway could be a targeted therapy against CSCs.

No potential conflicts of interest were disclosed.

Conception and design: B. Das, B. Pal, D.W. Felsher

Development of methodology: B. Das, B. Pal, D.W. Felsher

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): B. Das, B. Pal, R. Bhuyan, H. Li, A. Sarma, S. Gayan, J. Talukdar, S. Sandhya, S. Bhuyan, G. Gogoi, D. Baishya, J.R. Gotlib

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): B. Das, B. Pal, S. Gayan, A.M. Gouw, A.C. Kataki, D.W. Felsher

Writing, review, and/or revision of the manuscript: B. Das, B. Pal, R. Bhuyan, H. Li, S. Gayan, A.M. Gouw, A.C. Kataki, D.W. Felsher

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): B. Das, A. Sarma, J. Talukdar, S. Sandhya, S. Bhuyan, G. Gogoi, D. Baishya, J.R. Gotlib, D.W. Felsher

Study supervision: B. Das, D.W. Felsher

We thank members of Felsher laboratory, the Stanford flow cytometry facility, Animal Facility, and Laboratory of Immunology and Vascular Biology at the Palo Alto VA Health Care System, CA, Forsyth Institute, Cambridge, MA, and KaviKrishna laboratory, Guwahati Biotech Park, Indian Institute of Technology, Guwahati, India. This research project was funded by grants from the Canadian Cancer Society (to B. Das), Laurel Foundation (to B. Das), and KaviKrishna Foundation, Assam, India (to B. Das). Additional funding was obtained from the Bill & Melinda Gates Foundation through the “Grand Challenges Exploration Initiatives” (to B. Das), Stem Cell Altruism Fund, Thoreau Laboratory for Global Health, University of Massachusetts, Lowell (to B. Das), NIH grants R01CA105102, CA89305-0351, and CA112973 (to D.W. Felsher), Department of Defense grant PR080163 (to D.W. Felsher), Emerson Collective Foundation, KaviKrishna Foundation Fellowship award (to J. Talukdar and S. Bhuyan), KaviKrishna USA award (to R. Bhuyan and B. Pal), and Department of Biotechnology-India grant (to A. Sarma, D. Baishya, and A.C. Kataki).

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.
Kreso
A
,
Dick
JE
. 
Evolution of the cancer stem cell model
.
Cell Stem Cell
2014
;
14
:
275
91
.
2.
Pattabiraman
DR
,
Weinberg
RA
. 
Tackling the cancer stem cells - what challenges do they pose?
Nat Rev Drug Discov
2014
;
13
:
497
512
.
3.
Rosen
JM
,
Jordan
CT
. 
The increasing complexity of the cancer stem cell paradigm
.
Science
2009
;
324
:
1670
3
.
4.
Yoo
YD
,
Kwon
YT
. 
Molecular mechanisms controlling asymmetric and symmetric self-renewal of cancer stem cells
.
J Anal Sci Technol
2015
;
6
:
28
.
5.
Maenhaut
C
,
Dumont
JE
,
Roger
PP
,
van Staveren
WC
. 
Cancer stem cells: a reality, a myth, a fuzzy concept or a misnomer? An analysis
.
Carcinogenesis
2010
;
31
:
149
58
.
6.
Das,
B
. 
Altruistic stem cells and cancer stem cells
. In:
V.K. Rajasekhar, editors. Cancer Stem Cells
.
Hoboken, NJ: John Wiley & Sons
; 
2014
,
89
106
.
7.
Iliopoulos
D
,
Hirsch
HA
,
Wang
G
,
Struhl
K
. 
Inducible formation of breast cancer stem cells and their dynamic equilibrium with non-stem cancer cells via IL6 secretion
.
Proc Natl Acad Sci U S A
2011
;
108
:
1397
402
.
8.
Pardal
R
,
Clarke
MF
,
Morrison
SJ
. 
Applying the principles of stem-cell biology to cancer
.
Nat Rev Cancer
2003
;
3
:
895
902
.
9.
Gupta
PB
,
Fillmore
CM
,
Jiang
G
,
Shapira
SD
,
Tao
K
,
Kuperwasser
C
, et al
Stochastic state transitions give rise to phenotypic equilibrium in populations of cancer cells
.
Cell
2011
;
146
:
633
44
.
10.
Belmonte
M
,
Hoofd
C
,
Weng
AP
,
Giambra
V
. 
Targeting leukemia stem cells: which pathways drive self-renewal activity in T-cell acute lymphoblastic leukemia?
Curr Oncol
2016
;
23
:
34
41
.
11.
Bellovin
DI
,
Das
B
,
Felsher
DW
. 
Tumor dormancy, oncogene addiction, cellular senescence, and self-renewal programs
.
Adv Exp Med Biol
2013
;
734
:
91
107
.
12.
Ben-Porath
I
,
Thomson
MW
,
Carey
VJ
,
Ge
R
,
Bell
GW
,
Regev
A
, et al
An embryonic stem cell-like gene expression signature in poorly differentiated aggressive human tumors
.
Nat Genet
2008
;
40
:
499
507
.
13.
Cao
J
,
Li
L
,
Chen
C
,
Lv
C
,
Meng
F
,
Zeng
L
, et al
RNA interference-mediated silencing of NANOG leads to reduced proliferation and self-renewal, cell cycle arrest and apoptosis in T-cell acute lymphoblastic leukemia cells via the p53 signaling pathway
.
Leuk Res
2013
;
37
:
1170
7
.
14.
Dang
CV
. 
MYC on the path to cancer
.
Cell
2012
;
149
:
22
35
.
15.
Das
B
,
Tsuchida
R
,
Malkin
D
,
Koren
G
,
Baruchel
S
,
Yeger
H
. 
Hypoxia enhances tumor stemness by increasing the invasive and tumorigenic side population fraction
.
Stem Cells
2008
;
26
:
1818
30
.
16.
Deynoux
M
,
Sunter
N
,
Herault
O
,
Mazurier
F
. 
Hypoxia and hypoxia-inducible factors in leukemias
.
Front Oncol
2016
;
6
:
41
.
17.
Jeter
CR
,
Yang
T
,
Wang
J
,
Chao
HP
,
Tang
DG
. 
Concise review: NANOG in cancer stem cells and tumor development: an update and outstanding questions
.
Stem Cells
2015
;
33
:
2381
90
.
18.
Kim
JW
,
Gao
P
,
Dang
CV
. 
Effects of hypoxia on tumor metabolism
.
Cancer Metastasis Rev
2007
;
26
:
291
8
.
19.
Marzi
I
,
Cipolleschi
MG
,
D'Amico
M
,
Stivarou
T
,
Rovida
E
,
Vinci
MC
, et al
The involvement of a Nanog, Klf4 and c-Myc transcriptional circuitry in the intertwining between neoplastic progression and reprogramming
.
Cell Cycle
2013
;
12
:
353
64
.
20.
Rouault-Pierre
K
,
Lopez-Onieva
L
,
Foster
K
,
Anjos-Afonso
F
,
Lamrissi-Garcia
I
,
Serrano-Sanchez
M
, et al
HIF-2alpha protects human hematopoietic stem/progenitors and acute myeloid leukemic cells from apoptosis induced by endoplasmic reticulum stress
.
Cell Stem Cell
2013
;
13
:
549
63
.
21.
Vukovic
M
,
Guitart
AV
,
Sepulveda
C
,
Villacreces
A
,
O'Duibhir
E
,
Panagopoulou
TI
, et al
Hif-1α and Hif-2α synergize to suppress AML development but are dispensable for disease maintenance
.
J Exp Med
2015
;
212
:
2223
34
.
22.
Wang
J
,
Wang
H
,
Li
Z
,
Wu
Q
,
Lathia
JD
,
McLendon
RE
, et al
c-Myc is required for maintenance of glioma cancer stem cells
.
PLoS One
2008
;
3
:
e3769
.
23.
Wang
Y
,
Liu
Y
,
Malek
SN
,
Zheng
P
,
Liu
Y
. 
Targeting HIF1α eliminates cancer stem cells in hematological malignancies
.
Cell Stem Cell
2011
;
8
:
399
411
.
24.
Roderick
JE
,
Tesell
J
,
Shultz
LD
,
Brehm
MA
,
Greiner
DL
,
Harris
MH
, et al
c-Myc inhibition prevents leukemia initiation in mice and impairs the growth of relapsed and induction failure pediatric T-ALL cells
.
Blood
2014
;
123
:
1040
50
.
25.
Wilson
A
,
Murphy
MJ
,
Oskarsson
T
,
Kaloulis
K
,
Bettess
MD
,
Oser
GM
, et al
c-Myc controls the balance between hematopoietic stem cell self-renewal and differentiation
.
Genes Dev
2004
;
18
:
2747
63
.
26.
Wong
DJ
,
Liu
H
,
Ridky
TW
,
Cassarino
D
,
Segal
E
,
Chang
HY
. 
Module map of stem cell genes guides creation of epithelial cancer stem cells
.
Cell Stem Cell
2008
;
2
:
333
44
.
27.
Takahashi
K
,
Tanabe
K
,
Ohnuki
M
,
Narita
M
,
Ichisaka
T
,
Tomoda
K
, et al
Induction of pluripotent stem cells from adult human fibroblasts by defined factors
.
Cell
2007
;
131
:
861
72
.
28.
Kwan
KY
,
Shen
J
,
Corey
DP
. 
C-MYC transcriptionally amplifies SOX2 target genes to regulate self-renewal in multipotent otic progenitor cells
.
Stem Cell Rep
2015
;
4
:
47
60
.
29.
Dang
CV
,
Kim
JW
,
Gao
P
,
Yustein
J
. 
The interplay between MYC and HIF in cancer
.
Nat Rev Cancer
2008
;
8
:
51
6
.
30.
Gordan
JD
,
Bertout
JA
,
Hu
CJ
,
Diehl
JA
,
Simon
MC
. 
HIF-2alpha promotes hypoxic cell proliferation by enhancing c-myc transcriptional activity
.
Cancer Cell
2007
;
11
:
335
47
.
31.
Covello
KL
,
Kehler
J
,
Yu
H
,
Gordan
JD
,
Arsham
AM
,
Hu
CJ
, et al
HIF-2alpha regulates Oct-4: effects of hypoxia on stem cell function, embryonic development, and tumor growth
.
Genes Dev
2006
;
20
:
557
70
.
32.
Petruzzelli
R
,
Christensen
DR
,
Parry
KL
,
Sanchez-Elsner
T
,
Houghton
FD
. 
HIF-2α regulates NANOG expression in human embryonic stem cells following hypoxia and reoxygenation through the interaction with an Oct-Sox cis regulatory element
.
PLoS One
2014
;
9
:
e108309
.
33.
Das
B
,
Bayat-Mokhtari
R
,
Tsui
M
,
Lotfi
S
,
Tsuchida
R
,
Felsher
DW
, et al
HIF-2α suppresses p53 to enhance the stemness and regenerative potential of human embryonic stem cells
.
Stem Cells
2012
;
30
:
1685
95
.
34.
Felsher
DW
,
Bishop
JM
. 
Reversible tumorigenesis by MYC in hematopoietic lineages
.
Mol Cell
1999
;
4
:
199
207
.
35.
Giuriato
S
,
Ryeom
S
,
Fan
AC
,
Bachireddy
P
,
Lynch
RC
,
Rioth
MJ
, et al
Sustained regression of tumors upon MYC inactivation requires p53 or thrombospondin-1 to reverse the angiogenic switch
.
Proc Natl Acad Sci U S A
2006
;
103
:
16266
71
.
36.
Spangrude
GJ
,
Aihara
Y
,
Weissman
IL
,
Klein
J
. 
The stem cell antigens Sca-1 and Sca-2 subdivide thymic and peripheral T lymphocytes into unique subsets
.
J Immunol
1988
;
141
:
3697
707
.
37.
Das
B
,
Antoon
R
,
Tsuchida
R
,
Lotfi
S
,
Morozova
O
,
Farhat
W
, et al
Squalene selectively protects mouse bone marrow progenitors against cisplatin and carboplatin-induced cytotoxicity in vivo without protecting tumor growth
.
Neoplasia
2008
;
10
:
1105
19
.
38.
Debacq-Chainiaux
F
,
Erusalimsky
JD
,
Campisi
J
,
Toussaint
O
. 
Protocols to detect senescence-associated beta-galactosidase (SA-betagal) activity, a biomarker of senescent cells in culture and in vivo
.
Nat Protoc
2009
;
4
:
1798
806
.
39.
Wu
CH
,
van Riggelen
J
,
Yetil
A
,
Fan
AC
,
Bachireddy
P
,
Felsher
DW
. 
Cellular senescence is an important mechanism of tumor regression upon c-Myc inactivation
.
Proc Natl Acad Sci U S A
2007
;
104
:
13028
33
.
40.
Das
B
,
Yeger
H
,
Tsuchida
R
,
Torkin
R
,
Gee
MF
,
Thorner
PS
, et al
A hypoxia-driven vascular endothelial growth factor/Flt1 autocrine loop interacts with hypoxia-inducible factor-1alpha through mitogen-activated protein kinase/extracellular signal-regulated kinase 1/2 pathway in neuroblastoma
.
Cancer Res
2005
;
65
:
7267
75
.
41.
Liu
X
,
Chua
CC
,
Gao
J
,
Chen
Z
,
Landy
CL
,
Hamdy
R
, et al
Pifithrin-alpha protects against doxorubicin-induced apoptosis and acute cardiotoxicity in mice
.
Am J Physiol Heart Circ Physiol
2004
;
286
:
H933
9
.
42.
Lau
PN
,
Cheung
P
. 
Histone code pathway involving H3 S28 phosphorylation and K27 acetylation activates transcription and antagonizes polycomb silencing
.
Proc Natl Acad Sci U S A
2011
;
108
:
2801
6
.
43.
Cox
CV
,
Martin
HM
,
Kearns
PR
,
Virgo
P
,
Evely
RS
,
Blair
A
. 
Characterization of a progenitor cell population in childhood T-cell acute lymphoblastic leukemia
.
Blood
2007
;
109
:
674
82
.
44.
Garhyan
J
,
Bhuyan
S
,
Pulu
I
,
Kalita
D
,
Das
B
,
Bhatnagar
R
. 
Preclinical and clinical evidence of mycobacterium tuberculosis persistence in the hypoxic niche of bone marrow mesenchymal stem cells after therapy
.
Am J Pathol
2015
;
185
:
1924
34
.
45.
Yan
Q
,
Bartz
S
,
Mao
M
,
Li
L
,
Jr
Kaelin
WG
. 
The hypoxia-inducible factor 2alpha N-terminal and C-terminal transactivation domains cooperate to promote renal tumorigenesis in vivo
.
Mol Cell Biol
2007
;
27
:
2092
102
.
46.
Rider
P
,
Kaplanov
I
,
Romzova
M
,
Bernardis
L
,
Braiman
A
,
Voronov
E
, et al
The transcription of the alarmin cytokine interleukin-1 alpha is controlled by hypoxia inducible factors 1 and 2 alpha in hypoxic cells
.
Front Immunol
2012
;
3
:
290
.
47.
Blackburn
JS
,
Liu
S
,
Wilder
JL
,
Dobrinski
KP
,
Lobbardi
R
,
Moore
FE
, et al
Clonal evolution enhances leukemia-propagating cell frequency in T cell acute lymphoblastic leukemia through Akt/mTORC1 pathway activation
.
Cancer Cell
2014
;
25
:
366
78
.
48.
Plasschaert
SL
,
van der Kolk
DM
,
de Bont
ES
,
Kamps
WA
,
Morisaki
K
,
Bates
SE
, et al
The role of breast cancer resistance protein in acute lymphoblastic leukemia
.
Clin Cancer Res
2003
;
9
:
5171
7
.
49.
Chiu
PP
,
Jiang
H
,
Dick
JE
. 
Leukemia-initiating cells in human T-lymphoblastic leukemia exhibit glucocorticoid resistance
.
Blood
2010
;
116
:
5268
79
.
50.
Giambra
V
,
Jenkins
CE
,
Lam
SH
,
Hoofd
C
,
Belmonte
M
,
Wang
X
, et al
Leukemia stem cells in T-ALL require active Hif1α and Wnt signaling
.
Blood
2015
;
125
:
3917
27
.
51.
Palomero
T
,
Lim
WK
,
Odom
DT
,
Sulis
ML
,
Real
PJ
,
Margolin
A
, et al
NOTCH1 directly regulates c-MYC and activates a feed-forward-loop transcriptional network promoting leukemic cell growth
.
Proc Natl Acad Sci U S A
2006
;
103
:
18261
6
.
52.
Armstrong
F
,
Brunet de la Grange
P
,
Gerby
B
,
Rouyez
MC
,
Calvo
J
,
Fontenay
M
, et al
NOTCH is a key regulator of human T-cell acute leukemia initiating cell activity
.
Blood
2009
;
113
:
1730
40
.
53.
Chumsri
S
,
Matsui
W
,
Burger
AM
. 
Therapeutic implications of leukemic stem cell pathways
.
Clin Cancer Res
2007
;
13
:
6549
54
.
54.
Lahad
JP
,
Mills
GB
,
Coombes
KR
. 
Stem cell-ness: a "magic marker" for cancer
.
J Clin Invest
2005
;
115
:
1463
7
.
55.
Liu
K
,
Lin
B
,
Zhao
M
,
Yang
X
,
Chen
M
,
Gao
A
, et al
The multiple roles for Sox2 in stem cell maintenance and tumorigenesis
.
Cell Signal
2013
;
25
:
1264
71
.