Purpose: Leukemia stem cells (LSC), which are insensitive to tyrosine kinase inhibitors (TKI), are an important source of TKI resistance and disease relapse in chronic myelogenous leukemia (CML). Obstacles to eradicating LSCs include limited understanding of the regulation network of LSCs. The current study aimed to examine the interplay between NF-κB and FOXM1/β-catenin, and the effect of its chemical intervention on CML LSCs.

Experimental Design: The interplay between NF-κB and FOXM1/β-catenin was analyzed by reciprocal coimmunoprecipitation (co-IP) and chromatin immunoprecipitation (ChIP) assay in CML cells. The effect of disturbing NF-κB and FOXM1/β-catenin by niclosamide on the self-renewal capacity and survival of LSCs was evaluated in vitro in human primary CML CD34+ cells and in vivo in CML mice.

Results: Reciprocal co-IP experiments showed physical interaction of p65 and FOXM1. p65 promoted transcription of FOXM1 gene. ChIP assay revealed recruitment of p65 on the promoter of FOXM1 gene. Conversely, FOXM1 and β-catenin positively regulated the nuclear translocation and transcriptional activity of NF-κB in CML cells. Niclosamide disrupted the positive feedback loop between NF-κB and FOXM1/β-catenin, thereby impairing the self-renewal capacity and survival of CML LSCs. Niclosamide decreased the long-term engraftment of human CML LSCs in NOD-SCID IL2Rγ chain-deficient (NOG) mice, and prolonged the survival of CML mice.

Conclusions: Interaction of p65 with FOXM1/β-catenin is critical in CML and its disruption by niclosamide eradicates LSCs. These findings may improve the understanding of a self-renewal regulatory mechanism of LSCs and offer a rationale-based approach to eliminate LSCs in CML. Clin Cancer Res; 23(3); 789–803. ©2016 AACR.

Translational Relevance

Leukemia stem cells (LSC) are important source of tyrosine kinase inhibitor (TKI) resistance and disease relapse in chronic myelogenous leukemia (CML). Obstacles to eradicating LSCs include a lack of understanding of the molecular regulation network of LSC survival and self-renewal. Human CML primitive CD34+ cells aberrantly overexpress cellular NF-κB, forkhead box M1 (FOXM1), and β-catenin. We examined the interaction and feedback loop between NF-κB and FOXM1/β-catenin in CML LSCs. Niclosamide disrupted the positive feedback loop, thereby impairing LSC self-renewal capacity and eliminating LSCs. Niclosamide decreased the long-term engraftment of human CML LSCs in immunodeficient mice and prolonged the survival of human BCR-ABL gene–driven CML mice. These findings between p65 and FOXM1/β-catenin interplay may improve understanding the signaling network of CML LSCs.

Chronic myelogenous leukemia (CML) arises from hematopoietic stem cells (HSC) malignantly transformed by the BCR-ABL oncogene. CML generally progresses from a chronic phase (CP) to an accelerated phase (AP), then a stage of blast crisis (BC; refs. 1, 2). A 10-year clinical follow-up demonstrated that treatment with a tyrosine kinase inhibitor (TKI) imatinib mesylate achieved remission in more than 80% of patients with CP CML and significantly prolonged the event-free survival of CML patients harboring the wild-type BCR-ABL (3). However, acquired resistance to imatinib is a challenge in CML treatment. Point mutations (e.g., T315I, G250E, and E255K/V) in BCR-ABL are major causes of imatinib-resistance (4). Nilotinib and dasatinib, the second-generation TKIs, can achieve good clinical response in most CML patients harboring most mutant isoforms of BCR-ABL except T315I (5). Ponatinib, a third-generation TKI, has been approved for treatment in imatinib-resistant CML patients harboring T315I BCR-ABL despite its potential cardiotoxicity (6, 7). Thus, acquired mutation resistance to imatinib is becoming a manageable clinical issue with these novel TKIs.

The other mechanisms of resistance to imatinib may include leukemia stem cells (LSC) or leukemia-initiating cells (8), and BCR-ABL–independent clones (9). LSCs, characterized by their capacity for self-renewal, and insensitivity to TKIs, may confer clinical resistance to imatinib and lead to CML relapse. Current evidence supports that LSCs are retained in CML patients with remission induced by TKI treatment (10), for a potential source of CML recurrence (11). Identification of novel agents capable of eradicating LSCs may be a critical strategy to cure CML.

The regulation of self-renewal in LSCs has not been fully understood. The intrinsic regulators include developmental signaling pathways (e.g., WNT/β-catenin, Hedgehog) and transcription factors [e.g., forkhead box M1 (FOXM1), NF-κB; ref. 12]. Evidence supports that loss of β-catenin impairs LSCs renewal in vivo, and genetic deletion and pharmacologic inhibition of β-catenin targets LSCs in CML (13, 14). Deregulated inflammatory cytokines in leukemic bone marrow may also affect intrinsic regulators of LSCs; for instance, high levels of TNFα in bone marrow promotes CML LSC survival by activating the NF-κB pathway (15). However, whether β-catenin and NF-κB, two common intrinsic regulators of LSCs, have interplay, remains unclear.

The current study aimed to examine the interplay between β-catenin (with its partner FOXM1; ref. 16) and NF-κB, and the effect of its chemical intervention on CML LSCs. We discovered a positive-feedback loop regulation between NF-κB p65 and FOXM1/β-catenin in CML LSCs. Disrupting this loop with niclosamide inhibited survival and self-renewal of CML LSCs in vitro and in vivo.

Chemicals, antibodies, and plasmids

Niclosamide, Annexin V-FITC, cycloheximide, DMSO, anti-Flag, anti-HA, anti-actin, and anti-tubulin were from Sigma-Aldrich. p-Niclosamide, a water-soluble derivative of niclosamide, was designed by adding a phosphate group to niclosamide with diethyl phosphate (17). Imatinib was from Novartis Pharmaceuticals. MG-132 was from Calbiochem. Recombinant human TNFα was a product of Peprotech. Recombinant human WNT3A was from R&D Systems. Protein A/G agarose beads and antibodies against FOXM1 (C-19, K-20), p65 (c-20, F-6), proliferating cell nuclear antigen (PCNA), and IκBα were from Santa Cruz Biotechnology. Anti-β-catenin and FITC conjunct IgG1κ isotype control were from BD Biosciences. Phospho-IκBα (S32) was from Cell Signaling Technology. Anti-active-β-catenin (clone 8E7) was from Upstate Tech. Anti-mouse immunoglobulin G and anti-rabbit immunoglobulin G horseradish peroxidase–conjugated antibodies were from Pierce Biotechnology.

Plasmid encoding FOXM1 gene promoter–driven luciferase (Luc) reporter was described previously (18). The pNF-κB-Luc plasmid was from Stratagene. The TCF/LEF reporter plasmid, pTOPflash, and its mutant control, pFOPflash, were from EMD Millipore. The Renilla luciferase reporter construct, pEFRenilla-Luc, was from Promega. pcDNA3-β-catenin was from Addgene. FOXM1-flag (16), p65-HA, p65 1–286, and p65 286–551 (19), HA-β-catenin (20), and (His)6-ubiquitin (21) were described previously.

siRNA duplexes against p65 (#1, sc-29410; #2, sc-44212) and FOXM1 (#1, sc-270048; #2, sc-43769) were from Santa Cruz Biotechnology. siRNA duplexes against β-catenin (#1, 6225; #2, 6238) were from Cell Signaling Technology; ON-TARGET plus Non-Targeting Pool siRNA control was from Dhamacon RNA Tech (22).

Cell culture

K562 and KBM5-T315I cells were grown as reported previously (23). 293T and Plat-E cells were cultured in DMEM supplemented with 10% FBS. MEF p65+/+ and p65−/− cells were grown in IMDM (Invitrogen) supplemented with 10% FBS (24). All the cell lines were tested and authenticated by using short tandem repeat (STR) matching analysis of cells last month. No cross-contamination of other human cells was found in all six lines of cells.

Transfection of plasmids and siRNA duplexes

For K562 and human primary CD34+ cells, siRNA duplexes were transduced into cells with the Cell Line Nucleofector Kit T (Amaxa) and program O-17 (22). For 293T cells, transfection involved use of Lipofectamine 2000 (Invitrogen).

Preparation of whole-cell lysates and cytoplasmic and nuclear fractions

Whole-cell lysates were prepared in RIPA buffer. Cytoplasmic, mitochondrial, and nuclear extracts were prepared as described previously (17, 22).

Immunoprecipitation and immunoblotting

Immunoprecipitation (IP) and immunoblotting (IB) were performed as reported previously (16).

Chromatin immunoprecipitation assay

An amount of 1 × 107 K562 cells was prepared with a ChIP kit (EMD Millipore). The precipitated DNA complexes were analyzed by real-time quantitative PCR (22). The primers were listed in Supplementary Table S1.

Dual luciferase assay

Dual luciferase assay was followed as reported previously (22). Briefly, cells were transfected with plasmids encoding FOXM1-Luc (0.5 μg), NF-κB-Luc (0.5 μg), pTOPflash (0.5 μg), pFOPflash (0.5 μg), and pEFRenilla-Luc (10 ng) with Lipofectamine 2000. Luciferase activity was measured with Dual Luciferase Assay Kit (Promega) as described previously (22). The luciferase activity was normalized to Renilla luciferase activity.

Immunofluorescence staining

Cells treated with or without niclosamide for 24 hours followed by stimulation of TNFα or WNT3A were collected and cytospun to slides. Immunofluorescence staining was performed as described previously (22).

In vivo ubiquitination assay

KBM5-T315I cells were transfected with the indicated constructs in the presence of DMSO or 2.0 μmol/L niclosamide for 24 hours and then treated with MG-132 (20 μmol/L) for the last 6 hours. In vivo ubiquitination assay was performed as reported previously (21).

Primary cells

Normal bone marrow samples (n = 4) were obtained from healthy donors at the first affiliated hospital of Sun Yat-sen University (Guangzhou, China). CML samples (n = 9) were obtained from the First Affiliated Hospital of Sun Yat-sen University and Guangdong General Hospital. Nucleated cells were isolated by Ficoll separation. CD34+ cells were fractionated by use of a positive magnetic bead selection protocol (Miltenyi Biotec). All patients and healthy donors gave their signed informed consent. The study approved by our institute followed the Declaration of Helsinki principles. The clinical information of CML patients is described in Supplementary Table S2.

Flow cytometry analysis of intracellular active β-catenin, FOXM1, and p65

The staining procedure was described previously (22). Samples were analyzed by Accuri C6 (BD Biosciences) flow cytometry.

Apoptosis analysis of quiescent cells

The apoptosis of quiescent cells was analyzed according to the previous report (25). Briefly, carboxyfluorescein succinimidyl amino ester (CFSE)-stained CD34+ cells (CellTrace CFSE Cell Proliferation Kit, Invitrogen) were incubated with different treatments for 96 hours and then stained with Annexin V–PE and analyzed by Accuri C6 flow cytometry. CFSEmaxCD34+Annexin V+ cells were determined as apoptotic quiescent cells (25).

Long-term culture-initiating cell assay and limiting dilution LTC-IC assay

Long-term culture-initiating cell (LTC-IC) assay and limiting dilution LTC-IC assay were performed following the manufacturer's instructions (9). CML-nucleated cells (2 × 106) were cultured with irradiated (80 Gy) M2-10B4 murine fibroblasts in MyeloCult H5100 (StemCell Technologies) supplemented with 10−6 mol/L hydrocortisone (long-term culture medium) in the presence of drugs for the first week of culture. Medium was replaced by half drug-free medium change weekly. After 6 weeks, all cells were harvested and plated into MethoCult H4435. LTC-IC–derived colonies were counted after 14 days. For limiting dilution assay, pretreated mononuclear CML cells were cultured with irradiated (80 Gy) M2-10B4 murine fibroblasts in MyeloCult H5100 supplemented with 10−6 mol/L hydrocortisone at serial dilutions (104, 3 × 103, 103, 300, 100). Half of the medium was refreshed weekly. After 5 weeks, cells were harvested and seeded in MethoCult H4435, and then the colonies were counted. LT-HSC frequency was analyzed by Poisson statistics online by using the Bioinformatics facility of The Walter & Eliza Hall Institute of Medical Research (Melbourne, Australia; ref. 26).

CFC/replating assay

CD34+ cells (5,000/well) were seeded in the H4434 MethoCult with niclosamide for the first round. Colonies were counted 7–10 days after culture, and then 5,000 cells from the colonies were sequentially replated in the H4434 MethoCult for another two rounds, respectively (9).

Engraftment of human cells in immunodeficient mice

CML CD34+ cells were cultured with or without 2.5 μmol/L niclosamide for 48 hours. Cells (1–2 × 106 cells/mouse) were then collected, washed, and transplanted into 8-week-old NOD.Cg-PrkdcscidII2rgtm1Sug/JicCrl mice (NOG mice, CIEA) via the tail vein (27). Cells were allowed to grow for 10 weeks. Mice were sacrificed, and fractionated mononuclear cells from bone marrow and spleen were labeled with antibodies for flow cytometry analysis (BD LSRFortessa). Antibodies were from BD Biosciences: CD45-APC CyTM7, CD34-FITC, CD33-PE CyTM7, CD14-PerCP-Cy5.5, CD11B-PE, CD19-APC, and CD3-Alexa Fluor 700 (27, 28).

Retroviral BCR-ABL–driven CML mouse model and treatment

The retroviral construct MSCR-IRES-BCR-ABL-WT-EGFP was used to generate high-titer helper-free retrovirus by transient transfection of Plat-E cells as reported previously (27). Bone marrow cells from 5-fluorouracil–treated (200 mg/kg) 6- to 8-week-old C57BL/6 male donor mice were transduced twice with BCR-ABL retrovirus by centrifugation in the presence of IL3, IL6, and stem cell factor (SCF). Cells (0.5 × 106) were then transplanted by tail vein into irradiated (5.50 Gy) receipt female mice. Mice were treated with placebo, imatinib, p-niclosamide, and imatinib combined with p-niclosamide for 2 weeks.

Flow cytometry analysis of bone marrow and splenic cells in CML mice and isolation of LSK cells

Cells were obtained from bone marrow (both femurs and tibias) or spleen. Antibodies were as follows: Lin-APC, Sca-1-PE-CF594, c-Kit-PE, Flt3-PE-Cyanine 5, CD150-PE-Cyanine 7, and CD48-APC-Cyanine7. Analysis and LSK cell sorting were conducted using flow cytometer (BD FACSAria II, BD Biosciences).

Real-time quantitative PCR

The qPCR experiments were carried out as described previously (22). The primers were listed in Supplementary Table S1.

Analysis of leukemia stem cell frequency

Bone marrow and splenic cells from 3–5 CML mice with different treatments were harvested and injected into secondary receipt mice (irradiated at 5.50 Gy) at serial concentrations of cells (2 × 106, 1 × 106, 5 × 105). GFP+ cells in peripheral blood were monitored by cytometry every week. GFP+ cells >0.5% were considered as positive transplantation. LSC frequency was determined 16 weeks after the secondary transplantation using Poisson statistics online by using the Bioinformatics facility of The Walter & Eliza Hall Institute of Medical Research (Parkville, Victoria, Australia; ref. 26).

Statistical analysis

GraphPad Prism 5.0 (GraphPad Prism Software) was used for statistical analysis. All experiments were carried out at least three times, and results were presented as mean ± SEM unless otherwise stated. Comparison between two groups was analyzed by t test and between more than two groups by one-way ANOVA with post hoc comparison by Tukey test. P < 0.05 was considered statistically significant.

NF-κB physically interacts with FOXM1 and promotes transcription of FOXM1 gene

To examine the potential interaction between NF-κB and FOXM1, 293T cells were cotransfected with HA-tagged p65 and Flag-tagged FOXM1 and subjected to IP analysis. IB analysis revealed FOXM1 presented in anti-p65 IP pellets (Fig. 1A, left) and p65 in anti-FOXM1 IP pellets (Fig. 1A, right). Moreover, reciprocal co-IP experiments revealed physical interaction of endogenous p65 and FOXM1 proteins in CML cells (Fig. 1B). Our results suggest that NF-κB protein physically interacts with FOXM1 protein.

Figure 1.

NF-κB positively regulates the expression of FOXM1 and β-catenin. A, p65 physically interacts with FOXM1. After 293T cells were cotransfected with plasmids encoding HA-p65 and flag-FOXM1, the whole-cell lysates were subjected to immunoprecipitation using HA antibody, followed by immunoblotting (IB) with anti-FOXM1 (left). Reciprocal IP was performed using anti-flag antibody, followed by IB with anti-p65 antibody (right). B, Physical interaction of endogenous p65 and endogenous FOXM1 was detected in CML cells. Whole-cell lysates of K562 cells were subjected to immunoprecipitation with anti-p65 antibody, followed by IB with anti-FOXM1 antibody (left). Reciprocal IP was performed using anti-FOXM1 antibody, followed by IB with anti-p65 antibody (right). C–E, p65 promoted expression of FOXM1. 293T cells were cotransfected with the human FOXM1 promoter-luciferase construct along with empty vector or increasing amount of HA-tagged constructs encoding full-length p65 (C) or truncate p65 fragments (D). FOXM1 protein level was measured by IB, the integrated density (IntDen) was analyzed by Image J, and normalized to relevant β-actin and control (D). The FOXM1 promoter activity was determined by luciferase activity assay (E). RHD, Rel homology domain; NLS, nuclear localization signal; TA, transactivation domain. F, p65 directly bound to the promoter of FOXM1 gene. Purified DNA of K562 cells was immunoprecipitated with anti-p65, and then amplified with specific primers of FOXM1 gene promoter, coding sequence (CDS) and intron. Promoter of GAPDH gene served as an irrelevant gene control in the ChIP assay. G, Purity of the fractions of nuclear and cytosol was analyzed by IB. H, NF-κB activation promoted nuclear translocation of FOXM1. Nucleus and cytosolic fractionations of TNFα (20 ng/mL)-treated MEF cells harboring p65+/+ or p65-null (p65−/−) cells were extracted for IB. I, p65 deficiency abolished coupled nuclear translocation of FOXM1 and p65 upon stimulation with TNFα. Immunofluorescence observation of MEF p65+/+ and p65−/− cells treated with TNFα (20 ng/mL) for 30 minutes. Images were recorded with microscopy (Zeiss, LSM710, 63× oil immersion objective). J, Pharmacologic inactivation of NF-κB by niclosamide abrogated nuclear translocation of FOXM1, p65, and β-catenin. K562 cells were pretreated with or without 2.5 μmol/L niclosamide for 24 hours, and exposed to TNFα (20 ng/mL), the samples were fractionated and analyzed by IB. Columns and bars are mean ± SEM. *, P < 0.05; **, P < 0.01; ***, P < 0.0001 compared with control, one-way ANOVA, post hoc intergroup comparisons. Same are hereafter.

Figure 1.

NF-κB positively regulates the expression of FOXM1 and β-catenin. A, p65 physically interacts with FOXM1. After 293T cells were cotransfected with plasmids encoding HA-p65 and flag-FOXM1, the whole-cell lysates were subjected to immunoprecipitation using HA antibody, followed by immunoblotting (IB) with anti-FOXM1 (left). Reciprocal IP was performed using anti-flag antibody, followed by IB with anti-p65 antibody (right). B, Physical interaction of endogenous p65 and endogenous FOXM1 was detected in CML cells. Whole-cell lysates of K562 cells were subjected to immunoprecipitation with anti-p65 antibody, followed by IB with anti-FOXM1 antibody (left). Reciprocal IP was performed using anti-FOXM1 antibody, followed by IB with anti-p65 antibody (right). C–E, p65 promoted expression of FOXM1. 293T cells were cotransfected with the human FOXM1 promoter-luciferase construct along with empty vector or increasing amount of HA-tagged constructs encoding full-length p65 (C) or truncate p65 fragments (D). FOXM1 protein level was measured by IB, the integrated density (IntDen) was analyzed by Image J, and normalized to relevant β-actin and control (D). The FOXM1 promoter activity was determined by luciferase activity assay (E). RHD, Rel homology domain; NLS, nuclear localization signal; TA, transactivation domain. F, p65 directly bound to the promoter of FOXM1 gene. Purified DNA of K562 cells was immunoprecipitated with anti-p65, and then amplified with specific primers of FOXM1 gene promoter, coding sequence (CDS) and intron. Promoter of GAPDH gene served as an irrelevant gene control in the ChIP assay. G, Purity of the fractions of nuclear and cytosol was analyzed by IB. H, NF-κB activation promoted nuclear translocation of FOXM1. Nucleus and cytosolic fractionations of TNFα (20 ng/mL)-treated MEF cells harboring p65+/+ or p65-null (p65−/−) cells were extracted for IB. I, p65 deficiency abolished coupled nuclear translocation of FOXM1 and p65 upon stimulation with TNFα. Immunofluorescence observation of MEF p65+/+ and p65−/− cells treated with TNFα (20 ng/mL) for 30 minutes. Images were recorded with microscopy (Zeiss, LSM710, 63× oil immersion objective). J, Pharmacologic inactivation of NF-κB by niclosamide abrogated nuclear translocation of FOXM1, p65, and β-catenin. K562 cells were pretreated with or without 2.5 μmol/L niclosamide for 24 hours, and exposed to TNFα (20 ng/mL), the samples were fractionated and analyzed by IB. Columns and bars are mean ± SEM. *, P < 0.05; **, P < 0.01; ***, P < 0.0001 compared with control, one-way ANOVA, post hoc intergroup comparisons. Same are hereafter.

Close modal

We next explored whether transcription factor p65 promotes FOXM1 expression. IB analysis indicated increased level of endogenous FOXM1 protein with increasing amount of plasmids encoding full-length p65 (Fig. 1C, top). In a parallel set of experiments cotransfecting a construct containing FOXM1 promoter-driven luciferase reporter (18), p65 dose dependently promoted FOXM1 promoter–driven transcription activity (Fig. 1C, bottom). In contrast, transfection of constructs of truncate p65 (a.a. 1–286) lacking the TAD domain (19), and truncate p65 (a.a. 286–551) containing NLS and C-terminal domains did not elicit an increase in endogenous FOXM1 protein (Fig. 1D), as well as in transcriptional activity of the Luc-FOXM1 promoter (Fig. 1E). These data suggest that the C-terminal portion including the NLS appears to be required for the promotion of FOXM1 transcription.

NF-κB directly binds FOXM1 gene promoter

We next examined whether cellular endogenous p65 directly bound to the promoter sequence of FOXM1 gene by using chromatin immunoprecipitation (ChIP) assay. Fragmented chromatin of K562 cell lysates was immunoprecipitated with anti-p65 antibody, and specific primers were used to amplify p65-binding sites in the FOXM1 promoter region. The results showed recruitment of endogenous p65 to the FOXM1 gene promoter but not CDS and intron regions of FOXM1 or irrelevant gene GAPDH promoter in CML cells (Fig. 1F).

Silencing p65 attenuates FOXM1 and β-catenin expression

We next investigated whether NF-κB was required for FOXM1 expression. K562 cells were transfected with siRNA duplexes against RELA gene, and cell lysates were then subjected to IB and luciferase activity assay. Silencing p65 in K562 cells by transfection of siRNA duplexes against RELA gene led to downregulation of FOXM1 expression as detected by IB (Supplementary Fig. S1A, left). Parallelly, p65 knockdown decreased the transcriptional activity of Luc-FOXM1 promoter which was cotransfected in K562 cells (Supplementary Fig. S1A, middle). These results suggest that NF-κB is required for FOXM1 expression. In addition, parallel experiments showed that p65 knockdown in K562 cells reduced the levels of β-catenin and its downstream TCF/LEF–dependent transcription activity (Supplementary Fig. S1A, right), which further indicates the regulation of β-catenin function by NF-κB.

NF-κB activation is required for nuclear translocation of FOXM1 and β-catenin

To further delineate the regulation of FOXM1 by canonical activation of NF-κB, K562 cells with p65 silenced by siRNA were exposed to TNFα for different durations. Control siRNA–treated cells showed TNFα-upregulated FOXM1 expression, which was abrogated in p65 siRNA–treated cells (Supplementary Fig. S1B). These data suggest that TNFα upregulates FOXM1 in a p65-dependent manner. Furthermore, p65-deficient MEF cells showed higher turnover rate of FOXM1 as compared with MEF p65+/+ cells (Supplementary Fig. S1C).

To better clarify the relationship of p65 and FOXM1, MEF cells harboring p65+/+ or p65−/− were stimulated by TNFα for various durations, nuclear and cytosolic fractions were examined by IB. The purity of nuclear and cytosol fractions was first verified (Fig. 1G). In MEF p65+/+ cells, TNFα stimulation triggered FOXM1 nuclear translocation, coupled with p65 nuclear translocation (Fig. 1H). In stark contrast, TNFα stimulation did not trigger nuclear translocation of FOXM1 in MEF p65−/− cells (Fig. 1H). A similar effect of TNFα treatment on nuclear relocation of FOXM1 and p65 was observed by confocal fluorescence microscopy (Fig. 1I). Moreover, WNT3A stimulation triggered concomitant nuclear translocation of β-catenin and FOXM1 protein in MEF p65+/+ cells but not MEF p65−/− cells (Supplementary Fig. S1D). These results suggest that p65 is required for FOXM1 nuclear translocation.

Because niclosamide is capable of blocking the canonical activation of NF-κB pathway (17), K562 cells pretreated with or without niclosamide were exposed to TNFα. IB examination showed that in the absence of niclosamide, IκBα was phosphorylated shortly after TNFα stimulation (Fig. 1J, left). Accordingly, p65 level was decreased in the cytosolic fraction and increased in the nuclear fraction (Fig. 1J, left). Of note, FOXM1 and β-catenin levels were concomitantly decreased in the cytosolic fraction and increased in the nuclear fraction with the change in p65 level (Fig. 1J, left). Niclosamide completely abolished TNFα-induced nuclear translocation of p65, FOXM1, and β-catenin (Fig. 1J, right).

Similarly, immunofluorescence staining revealed coupled nuclear translocation of p65 and FOXM1, and p65 and β-catenin in K562 cells with TNFα treatment (Supplementary Fig. S1E and S1F), which suggests that pharmacologic inactivation of NF-κB by niclosamide abrogates nuclear translocation of FOXM1 and β-catenin in CML cells.

FOXM1 and β-catenin positively regulate NF-κB

We next examined whether FOXM1 and β-catenin affected NF-κB level. 293T cells were transfected with plasmids encoding human FOXM1 or β-catenin, and p65 level was assessed by IB. Ectopic expression of FOXM1 and β-catenin increased endogenous p65 level (Fig. 2A).

Figure 2.

FOXM1 and its partner β-catenin positively regulate NF-κB. A, Ectopic expression of FOXM1 and β-catenin elevated the level of p65. 293T cells transfected with plasmids encoding FOXM1 or β-catenin were subjected to IB with anti-p65. B, Canonical activation of WNT/β-catenin pathway promoted nuclear translocation of p65. K562 cells were exposed to recombinant human WNT3A (20 ng/mL) for durations as indicated and nuclear and cytosolic fractionations were extracted for IB. C and D, Silencing FOXM1 or β-catenin decreased the level and transcription activity of p65. K562 cells were cotransfected with siRNA duplexes against two independent portions of FOXM1 (C) or CTNNB1 (D) gene in combination with a plasmid encoding FOXM1 gene promoter luciferase; cell lysates were subjected to IB and luciferase reporter assay, respectively. E, Niclosamide decreased the levels of β-catenin. KBM5-T315I cells treated with increasing concentrations of niclosamide for 48 hours were analyzed by IB and Image J, and normalized to relevant β-actin and control. F, Niclosamide abrogated nuclear translocation of p65 and β-catenin in CML cells. K562 cells were pretreated with or without 2.5 μmol/L niclosamide for 24 hours and exposed to recombinant human WNT3A (20 ng/mL); cells were analyzed by fluorescence confocal microscopy after staining with the indicated antibodies (Zeiss, LSM710, 63× oil immersion objective). Columns and bars are mean ± SEM. *, P < 0.05; **, P < 0.01; ***, P < 0.0001 compared with control, one-way ANOVA, post hoc intergroup comparisons.

Figure 2.

FOXM1 and its partner β-catenin positively regulate NF-κB. A, Ectopic expression of FOXM1 and β-catenin elevated the level of p65. 293T cells transfected with plasmids encoding FOXM1 or β-catenin were subjected to IB with anti-p65. B, Canonical activation of WNT/β-catenin pathway promoted nuclear translocation of p65. K562 cells were exposed to recombinant human WNT3A (20 ng/mL) for durations as indicated and nuclear and cytosolic fractionations were extracted for IB. C and D, Silencing FOXM1 or β-catenin decreased the level and transcription activity of p65. K562 cells were cotransfected with siRNA duplexes against two independent portions of FOXM1 (C) or CTNNB1 (D) gene in combination with a plasmid encoding FOXM1 gene promoter luciferase; cell lysates were subjected to IB and luciferase reporter assay, respectively. E, Niclosamide decreased the levels of β-catenin. KBM5-T315I cells treated with increasing concentrations of niclosamide for 48 hours were analyzed by IB and Image J, and normalized to relevant β-actin and control. F, Niclosamide abrogated nuclear translocation of p65 and β-catenin in CML cells. K562 cells were pretreated with or without 2.5 μmol/L niclosamide for 24 hours and exposed to recombinant human WNT3A (20 ng/mL); cells were analyzed by fluorescence confocal microscopy after staining with the indicated antibodies (Zeiss, LSM710, 63× oil immersion objective). Columns and bars are mean ± SEM. *, P < 0.05; **, P < 0.01; ***, P < 0.0001 compared with control, one-way ANOVA, post hoc intergroup comparisons.

Close modal

WNT/β-catenin activation promotes nuclear translocation of p65

We determined the effect of WNT/β-catenin activation by WNT3A on p65 level in K562 cells. IB analysis of K562 cells exposed to WNT3A revealed that β-catenin and FOXM1 levels were progressively decreased over time in the cytosolic fraction and progressively increased in the nuclear fraction (Fig. 2B), which supports nuclear translocation of β-catenin during canonical activation of WNT/β-catenin pathway. IB results showed a coupled and concurrent change of p65 with β-catenin in the cytosolic and nuclear fractions in WNT3A-treated cells (Fig. 2B). WNT/β-catenin activation promoted the nuclear translocation of p65.

Knockdown of β-catenin and FOXM1 downregulates p65 protein and NF-κB–dependent reporter activity

In K562 cells transfected by two independent siRNA duplexes against human FOXM1, the protein levels of p65 and β-catenin were reduced along with FOXM1 knockdown (Fig. 2C). Accordingly, NF-κB– and β-catenin–dependent reporter gene transcription was reduced (Fig. 2C). Similarly, silencing β-catenin by siRNA in K562 cells reduced p65 protein level and NF-κB–dependent reporter gene transcription (Fig. 2D), lowered the FOXM1 protein level and the transcription activity of FOXM1, as evaluated by IB and FOXM1 gene promoter–dependent luciferase activity, respectively (Fig. 2D). Collectively, these data suggest a positive-feedback regulation in expression between NF-κB and FOXM1/β-catenin.

Niclosamide induces ubiquitin-mediated degradation of β-catenin protein

Because niclosamide is capable of blocking canonical activation pathways of NF-κB as well as WNT/β-catenin (17), we determined whether niclosamide disturbed the regulatory loop between NF-κB and FOXM1/β-catenin. Niclosamide dose dependently reduced β-catenin protein level in CML cells (Fig. 2E). Furthermore, in vivo ubiquitination assay showed that the ubiquitination of β-catenin was increased with niclosamide treatment (Supplementary Fig. S2A), which suggests that niclosamide induces ubiquitin–proteosome–dependent degradation of β-catenin.

Furthermore, immunofluorescence staining experiments revealed that niclosamide completely abrogated β-catenin nuclear translocation in K562 cells stimulated with WNT3A (Fig. 2F and Supplementary Fig. S2B). Concomitantly, the nuclear translocation of p65 was abolished in K562 cells treated with WNT3A combined with niclosamide. Collectively, niclosamide blocked NF-κB and FOXM1/β-catenin and disrupted the regulatory loop.

Increased expression of p65, FOXM1, and β-catenin and their niclosamide-sensitive interplay in human CML stem cells

Because TNFα supports the survival of CML stem/progenitor cells by promoting NF-κB pathway activity (15), and β-catenin is essential for stemness maintenance of CML LSCs (14), we examined whether the regulatory loop between NF-κB and FOXM1/β-catenin existed in the primary CD34+ cell populations from CML patients. Purified CD34+ cells from CML patients were labeled with antibodies against p65, FOXM1, and active-β-catenin, then underwent flow cytometry analysis. The results showed that the levels of intracellular p65, FOXM1, and active-β-catenin were significantly higher in CML CD34+ cells than those in normal bone marrow CD34+ cells (Fig. 3A). Immunofluorescence detection revealed that p65, FOXM1, and β-catenin appeared predominantly distributed in nucleus of the CML CD34+ cells versus NBM counterparts (Supplementary Fig. S3). In addition, overlay between p65 and β-catenin under immunofluorescence observation implied interplay of NF-κB and FOXM1/β-catenin pathways in the primary CML CD34+ cells (Supplementary Fig. S3).

Figure 3.

Niclosamide disrupts interplay of p65 and FOXM1/β-catenin in human primary chronic myelogenous leukemia (CML) CD34+ cells. A, Concomitant overexpression of intracellular p65, FOXM1, and active β-catenin was detected in CML leukemia stem cells (LSC) relative to NBM CD34+ cells. Flow cytometry analysis of p65, FOXM1, and active β-catenin in CD34+ cells purified with immunomagnetic beads from specimens of healthy individuals (NBM, n = 3) and CML patients (n = 3). Representative flow cytometry histograms (left) and the bar chart of median fluorescence intensity (MFI; right) were shown. B–E, NF-κB, FOXM1, and β-catenin presented a positive regulation loop of expression in human primary CML CD34+ cells. CD34+ cells from CML patients were cotransfected with plasmids of wt p65, β-catenin, or FOXM1 (B), siRNA duplexes against p65 (C), β-catenin (D), or FOXM1 (E) with plasmids encoding FOXM1 promoter-luciferase reporter, NF-κB, or TOP/FOP luciferase reporter and Renilla luciferase reporter and underwent luciferase activity assay 24 hours later. F and G, Niclosamide disturbed the expression and nuclear translocation of NF-κB, FOXM1, and β-catenin in primary CML CD34+ cells. CD34+ cells from CML patients were treated with niclosamide (2.5 μmol/L) ± imatinib (5 μmol/L) for 24 hours and the levels of p65, FOXM1, and β-catenin were analyzed by IB (F). CD34+ cells from CML patients were treated with niclosamide (2.5 μmol/L) for 24 hours, and then stained with the indicated antibodies and observed and recorded with fluorescence confocal microscopy (G). Zeiss, LSM710, 63× oil immersion objective. Columns and bars are mean ± SEM. *, P < 0.05; **, P < 0.01; ***, P < 0.0001 compared with control, one-way ANOVA, post hoc intergroup comparisons.

Figure 3.

Niclosamide disrupts interplay of p65 and FOXM1/β-catenin in human primary chronic myelogenous leukemia (CML) CD34+ cells. A, Concomitant overexpression of intracellular p65, FOXM1, and active β-catenin was detected in CML leukemia stem cells (LSC) relative to NBM CD34+ cells. Flow cytometry analysis of p65, FOXM1, and active β-catenin in CD34+ cells purified with immunomagnetic beads from specimens of healthy individuals (NBM, n = 3) and CML patients (n = 3). Representative flow cytometry histograms (left) and the bar chart of median fluorescence intensity (MFI; right) were shown. B–E, NF-κB, FOXM1, and β-catenin presented a positive regulation loop of expression in human primary CML CD34+ cells. CD34+ cells from CML patients were cotransfected with plasmids of wt p65, β-catenin, or FOXM1 (B), siRNA duplexes against p65 (C), β-catenin (D), or FOXM1 (E) with plasmids encoding FOXM1 promoter-luciferase reporter, NF-κB, or TOP/FOP luciferase reporter and Renilla luciferase reporter and underwent luciferase activity assay 24 hours later. F and G, Niclosamide disturbed the expression and nuclear translocation of NF-κB, FOXM1, and β-catenin in primary CML CD34+ cells. CD34+ cells from CML patients were treated with niclosamide (2.5 μmol/L) ± imatinib (5 μmol/L) for 24 hours and the levels of p65, FOXM1, and β-catenin were analyzed by IB (F). CD34+ cells from CML patients were treated with niclosamide (2.5 μmol/L) for 24 hours, and then stained with the indicated antibodies and observed and recorded with fluorescence confocal microscopy (G). Zeiss, LSM710, 63× oil immersion objective. Columns and bars are mean ± SEM. *, P < 0.05; **, P < 0.01; ***, P < 0.0001 compared with control, one-way ANOVA, post hoc intergroup comparisons.

Close modal

To validate the above implications, primary CD34+ cells from CML patients were cotransfected with constructs of p65 or β-catenin with constructs of Luc-FOXM1 promoter and pEFRenilla-Luc. The results showed that either p65 or β-catenin significantly increased the transcription of FOXM1 in primary CML CD34+ cells (Fig. 3B, left). Moreover, cotransfection of constructs of pTOPflash (or pFOPflash) and pEFRenilla-Luc in combination with p65 or FOXM1 demonstrated that either p65 or β-catenin remarkably increased the TCF/LEF–dependent transcription in primary CML CD34+ cells (Fig. 3B, middle). In separate cotransfection experiments, either β-catenin or FOXM1 significantly increased NF-κB–dependent reporter gene transcription in primary CML CD34+ cells (Fig. 3B, right).

In contrast, cotransfection experiments with p65 siRNA duplexes in combination with constructs of Luc-FOXM1 promoter and pEFRenilla-Luc showed that p65 knockdown significantly decreased the transcription of FOXM1 in primary CML CD34+ cells (Fig. 3C, left). Cotransfection of constructs of pTOPflash (or pFOPflash) and pEFRenilla-Luc in combination with p65 siRNA duplexes revealed that p65 remarkably decreased the TCF/LEF–dependent transcription in primary CML CD34+ cells (Fig. 3C, right). In addition, FOXM1 knockdown significantly reduced NF-κB– and TCF/LEF–dependent reporter gene transcription in primary CML CD34+ cells (Fig. 3D). Similarly, β-catenin knockdown significantly decreased NF-κB–dependent reporter gene transcription and Luc-FOXM1 promoter transcriptional activity in primary CML CD34+ cells (Fig. 3E).

Next, we evaluated the effect of niclosamide on NF-κB and β-catenin in primary CML CD34+ cells. The purified CML CD34+ cells were incubated with control or niclosamide ± imatinib for 24 hours, levels of p65, FOXM1, and β-catenin were detected by IB. The results showed that niclosamide alone or combined with imatinib elicited a robust suppression in FOXM1 and p65 levels (Fig. 3F). Furthermore, niclosamide decreased levels of p65, FOXM1, and β-catenin by immunofluorescence staining examination (Fig. 3G).

Taken together, these results suggest that the positive feedback loop formed by p65 and FOXM1/β-catenin is active in human CML LSCs, and that niclosamide may abrogate the interplay.

Niclosamide reduces survival and self-renewal capacity in primary CML CD34+ cells

We next ascertained the effect of niclosamide on survival and growth of CML primitive stem/progenitor cells. Purified CML CD34+ cells were labeled with CFSE (25), cultured with niclosamide, then stained with Annexin V–PE for flow cytometry analysis. Niclosamide greatly induced apoptosis in the CFSEmax CML CD34+ cell population but not in the CFSEmax NBM CD34+ cell population (Fig. 4A). Niclosamide may preferentially kill quiescent CML stem/progenitor cells, sparing quiescent NBM HSCs.

Figure 4.

Niclosamide reduces survival and self-renewal of primary CML CD34+ cells from CML patients. A, Niclosamide induced apoptosis in quiescent CML CD34+ cells but not NBM CD34+ cells. CML (n = 3) or NBM (n = 3) CD34+ cells were labeled with CFSE, and then cultured with niclosamide for 96 hours; cells were analyzed by flow cytometry after staining with Annexin V–PE; results for CFSEmax and Annexin V+ cells. A set of representative plots (left) and bar chart (right) are shown. ns, no significance; **, P < 0.01; Student t test. B, Niclosamide impaired CFC/replating capacity of CML stem cells. Results showed the replating efficiency of CFC colonies from NBM and CML specimen treated with niclosamide as indicated. ns, no significance; *, P < 0.05; **, P < 0.01; ***, P < 0.0001. Student t test. C, The quantity of CML hematopoietic stem cells and progenitors were decreased by combination treatment with niclosamide and imatinib. LTC-IC assay showed the colony numbers of NBM and CML cells exposed to the indicated concentrations of niclosamide during the initial week, and another 5-week subsequent culture in drug-free long-term culture medium, with colony scored in methylcellulose at week 6. D, Combination treatment of niclosamide and imatinib decreased the frequency of stem/progenitor cell in CML samples. Columns and bars are mean ± SEM. *, P < 0.05; **, P < 0.01; ***, P < 0.0001 compared with control, one-way ANOVA, post hoc intergroup comparisons.

Figure 4.

Niclosamide reduces survival and self-renewal of primary CML CD34+ cells from CML patients. A, Niclosamide induced apoptosis in quiescent CML CD34+ cells but not NBM CD34+ cells. CML (n = 3) or NBM (n = 3) CD34+ cells were labeled with CFSE, and then cultured with niclosamide for 96 hours; cells were analyzed by flow cytometry after staining with Annexin V–PE; results for CFSEmax and Annexin V+ cells. A set of representative plots (left) and bar chart (right) are shown. ns, no significance; **, P < 0.01; Student t test. B, Niclosamide impaired CFC/replating capacity of CML stem cells. Results showed the replating efficiency of CFC colonies from NBM and CML specimen treated with niclosamide as indicated. ns, no significance; *, P < 0.05; **, P < 0.01; ***, P < 0.0001. Student t test. C, The quantity of CML hematopoietic stem cells and progenitors were decreased by combination treatment with niclosamide and imatinib. LTC-IC assay showed the colony numbers of NBM and CML cells exposed to the indicated concentrations of niclosamide during the initial week, and another 5-week subsequent culture in drug-free long-term culture medium, with colony scored in methylcellulose at week 6. D, Combination treatment of niclosamide and imatinib decreased the frequency of stem/progenitor cell in CML samples. Columns and bars are mean ± SEM. *, P < 0.05; **, P < 0.01; ***, P < 0.0001 compared with control, one-way ANOVA, post hoc intergroup comparisons.

Close modal

We employed CFC/replating and LTC-IC assay to assess whether niclosamide affected the self-renewal capacity of CML LSCs. The results showed that niclosamide alone or combined with imatinib treatment decreased CFC/replating ability and the number of LTC-IC–derived colonies in CML CD34+ cells but not NBM CD34+ cells (Fig. 4B and C).

We further quantify the frequency of CML LSCs with limiting dilution LTC-IC assay. Niclosamide reduced the frequency from 1/104 (control) to 1/457 (niclosamide). Combination treatment of niclosamide and imatinib substantially reduced the LT-HSC frequency to a greater degree (Fig. 4D; Supplementary Table S3). Therefore, niclosamide alone or combinational treatments of niclosamide and imatinib inhibited the self-renewal capacity of CML LSCs and reduced the CML LT-HSC frequency.

Niclosamide abolishes the long-term engraftment of CML CD34+ cells in NOG mice

To evaluate the long-term ex vivo effect of niclosamide on survival of CML CD34+ cells, purified human CML CD34+ cells were exposed to niclosamide, then intravenously injected into NOG mice (Fig. 5A). The proportion of human CD45+ cells in the bone marrow and spleens of NOG mice was analyzed by flow cytometry 10 weeks after transplantation. Niclosamide significantly reduced the engraftment of human CML CD45+ cells in murine bone marrow (Fig. 5B) and spleens (Fig. 5C). In addition, niclosamide lowered the human diverse myeloid lineages in the NOG murine bone marrow (Fig. 5D) and spleens (Fig. 5E) as detected by flow cytometry after staining with antibodies of CD34, CD33, CD11b, CD14, CD19, and CD3, respectively. These results suggest that ex vivo niclosamide treatment inhibits the long-term engraftment of primary human CML CD34+ cells.

Figure 5.

Niclosamide inhibits the long-term engraftment of human CML stem cells in NOG mice. A, The schema of long-term engraftment of human CML stem cells in NOG mice. Human CML CD34+ cells were treated with 2.5 μmol/L niclosamide for 48 hours, and then injected into NOD-SCID IL2R γ chain-deficient (NOG) mice via the tail vein, and allowed to grow for 10 weeks. Human cell engraftment was analyzed by flow cytometry with assessment of human CD45+ cells. B and C, The amount of human CD45+ cells was decreased by niclosamide. Long-term engraftment in nucleated cells of bone marrow (BM; B) and spleens (C) in NOG mice were analyzed by flow cytometry after staining with human CD45 antibody. Each data point represents one sample. D and E, Niclosamide inhibited diverse human CML myeloid cell lineages in NOG bone marrow and spleens. The nucleated cells in bone marrow (D) and spleen (E) of NOG mice engrafted with human CD34+ cells were analyzed by flow cytometry after staining for human CD34, CD33, CD11b, CD14, CD19, and CD3. Bar charts of various engrafted CD45+ cells were shown. ***, P < 0.0001, Student t test.

Figure 5.

Niclosamide inhibits the long-term engraftment of human CML stem cells in NOG mice. A, The schema of long-term engraftment of human CML stem cells in NOG mice. Human CML CD34+ cells were treated with 2.5 μmol/L niclosamide for 48 hours, and then injected into NOD-SCID IL2R γ chain-deficient (NOG) mice via the tail vein, and allowed to grow for 10 weeks. Human cell engraftment was analyzed by flow cytometry with assessment of human CD45+ cells. B and C, The amount of human CD45+ cells was decreased by niclosamide. Long-term engraftment in nucleated cells of bone marrow (BM; B) and spleens (C) in NOG mice were analyzed by flow cytometry after staining with human CD45 antibody. Each data point represents one sample. D and E, Niclosamide inhibited diverse human CML myeloid cell lineages in NOG bone marrow and spleens. The nucleated cells in bone marrow (D) and spleen (E) of NOG mice engrafted with human CD34+ cells were analyzed by flow cytometry after staining for human CD34, CD33, CD11b, CD14, CD19, and CD3. Bar charts of various engrafted CD45+ cells were shown. ***, P < 0.0001, Student t test.

Close modal

p-Niclosamide significantly prolongs the survival of CML mice and reduces the frequency of LSCs in vivo

To investigate the in vivo effect of niclosamide on CML LSCs, we used a human BCR-ABL gene–driven CML mouse model (27). The CML mice were treated with placebo, parenteral p-niclosamide, imatinib, and the combination for 2 weeks (Fig. 6A). p-Niclosamide alone or in combination with imatinib significantly prolonged the overall survival of CML mice as compared with the placebo group (Fig. 6B). The populations of leukemic BCR-ABL-GFP+ WBC and bone marrow myeloid cells (Gr-1+Mac1+) were greatly reduced in the mice receiving p-niclosamide or imatinib, and was further reduced in the mice receiving combinational treatment (Fig. 6C). CML mice receiving p-niclosamide, imatinib, or the combination showed reduction in size and nodule in spleen than those in placebo group (Supplementary Fig. S4A). Pathology of lungs revealed decreased infiltration of leukemic cells in the CML mice receiving p-niclosamide, imatinib, or the combination (Supplementary Fig. S4B).

Figure 6.

p-Niclosamide reduces survival and self-renewal of leukemia stem cells in CML mice. A, The schematic procedure of the CML model induced by retroviralBCR-ABL transduction/transplantation. B, Effect of p-niclosamide on survival of CML mice. Kaplan–Meier survival curve of CML mouse after treatment of p-niclosamide (i.p., 30 mg/kg/day) ± imatinib (oral gavage, 100 mg/kg/day). ***, P = 0.005; **, P = 0.01, log-rank test. C,p-Niclosamide administration decreased the amount of leukemia myeloid cells in bone marrow of CML mice. The percentages of GFP+ WBC (left) and GFP+ myeloid (Gr-1+Mac1+; right) cells in the bone marrow were analyzed by flow cytometry after CML mice were administrated with p-niclosamide ± imatinib for 2 weeks. Each data point represents one patient specimen. *, P < 0.05; ***, P < 0.0001, one-way ANOVA, post hoc intergroup comparisons. D and E, Two weeks of administration of p-niclosamide alone or in combination with imatinib eradicated LSCs in the bone marrow of CML mice. The percentages of LSK, ST-HSC, LT-HSC, in bone marrow were analyzed by flow cytometry. Data are mean ± SEM. ns, no significance; ***, P < 0.0001 (D). Bar charts of LSK, LT-HSC, and ST-HSC for each treatment (E). F,p-Niclosamide suppressed the transcription of Rela, Foxm1, and Ctnnb1 in the LSK cells sorted by flow cytometer from the bone marrow of the CML mice treated with p-niclosamide ± imatinib for 2 weeks; the results of qRT-PCR analysis were shown. ns, no significance; **, P < 0.01; ***, P < 0.0001, one-way ANOVA, post hoc intergroup comparisons. G–I,p-Niclosamide lowered the in vivo CML reconstitution capacity and LSC frequency. Bone marrow cells from CML mice received 2 weeks of treatments were serially diluted with bone marrow cells from normal mice and then transplanted into the secondary irradiated recipient mice. The secondary recipient mice were examined at 8 and 16 weeks after transplantation, respectively (procedure schematic, G). Percentage of GFP+ cells at 8 and 16 weeks after transplantation (H) and the frequency of LSCs after treatment were shown (I). *, P < 0.05; **, P < 0.01; ***, P < 0.0001, one-way ANOVA, post hoc intergroup comparisons. J, A proposed model of interplay of p65/FOXM1/β-catenin and its intervention by niclosamide.

Figure 6.

p-Niclosamide reduces survival and self-renewal of leukemia stem cells in CML mice. A, The schematic procedure of the CML model induced by retroviralBCR-ABL transduction/transplantation. B, Effect of p-niclosamide on survival of CML mice. Kaplan–Meier survival curve of CML mouse after treatment of p-niclosamide (i.p., 30 mg/kg/day) ± imatinib (oral gavage, 100 mg/kg/day). ***, P = 0.005; **, P = 0.01, log-rank test. C,p-Niclosamide administration decreased the amount of leukemia myeloid cells in bone marrow of CML mice. The percentages of GFP+ WBC (left) and GFP+ myeloid (Gr-1+Mac1+; right) cells in the bone marrow were analyzed by flow cytometry after CML mice were administrated with p-niclosamide ± imatinib for 2 weeks. Each data point represents one patient specimen. *, P < 0.05; ***, P < 0.0001, one-way ANOVA, post hoc intergroup comparisons. D and E, Two weeks of administration of p-niclosamide alone or in combination with imatinib eradicated LSCs in the bone marrow of CML mice. The percentages of LSK, ST-HSC, LT-HSC, in bone marrow were analyzed by flow cytometry. Data are mean ± SEM. ns, no significance; ***, P < 0.0001 (D). Bar charts of LSK, LT-HSC, and ST-HSC for each treatment (E). F,p-Niclosamide suppressed the transcription of Rela, Foxm1, and Ctnnb1 in the LSK cells sorted by flow cytometer from the bone marrow of the CML mice treated with p-niclosamide ± imatinib for 2 weeks; the results of qRT-PCR analysis were shown. ns, no significance; **, P < 0.01; ***, P < 0.0001, one-way ANOVA, post hoc intergroup comparisons. G–I,p-Niclosamide lowered the in vivo CML reconstitution capacity and LSC frequency. Bone marrow cells from CML mice received 2 weeks of treatments were serially diluted with bone marrow cells from normal mice and then transplanted into the secondary irradiated recipient mice. The secondary recipient mice were examined at 8 and 16 weeks after transplantation, respectively (procedure schematic, G). Percentage of GFP+ cells at 8 and 16 weeks after transplantation (H) and the frequency of LSCs after treatment were shown (I). *, P < 0.05; **, P < 0.01; ***, P < 0.0001, one-way ANOVA, post hoc intergroup comparisons. J, A proposed model of interplay of p65/FOXM1/β-catenin and its intervention by niclosamide.

Close modal

Flow cytometry analysis indicated that p-niclosamide alone or with imatinib significantly lowered the number of primitive LinSca1+Kit+ (LSK) cells, LT-HSC (Flt3CD150+CD48), ST-HSC (Flt3CD150CD48) in the bone marrow (Fig. 6D and E) and spleens (Supplementary Fig. S4C and S4D) in CML mice.

Sorted LSK cells from bone marrow displayed significant reduction in mRNA levels of Rela, Foxm1, and Ctnnb1 in the mice receiving p-niclosamide alone or with imatinib (Fig. 6F). In vivo limiting dilution analysis of LSCs further showed that p-niclosamide alone prevented the engraftment of GFP+ leukemia cells in the secondary recipient mice at 16 weeks (Fig. 6G and H). p-Niclosamide alone or combined with imatinib significantly decreased CML LSC frequencies (Fig. 6I; Supplementary Table S4).

Elucidation of survival and the self-renewal regulation mechanism of CSCs is of importance for targeting CSCs. In this study, we documented that NF-κB physically interacted with FOXM1, and was recruited to the FOXM1 gene promoter to increase the transcription of FOXM1. Reciprocally, FOXM1 and β-catenin positively regulated NF-κB expression and its transcriptional activity in CML LSCs. Disrupting the interplay of NF-κB and FOXM1/β-catenin by niclosamide treatment significantly induced apoptosis and reduced in vitro LSC self-renewal capacity in human CML CD34+ cells. Niclosamide prolonged survival in a BCR-ABL–driven CML mouse model and decreased the in vivo burden of CML LSCs.

Aberrant overexpression of NF-κB, FOXM1, and β-catenin in LSCs

Oncogene activation in LSCs may cause NF-κB pathway activation (15, 29). LSCs may also produce autocrine TNFα to support their own survival via the NF-κB pathway because increased TNFα was detected in the serum of CML patients (15). We discovered aberrant overexpression of intracellular FOXM1, β-catenin, and NF-κB in human CML primitive CD34+ cells as compared with NBM CD34+ cells. The differential expression may offer a rationale to target LSCs.

Regulatory loop and its significance for LSCs

FOXM1 is widely overexpressed in human tumors, and plays a critical role in cell cycle, DNA replication, mitosis, and genomic stability (16). The association of FOXM1 and β-catenin hints that FOXM1 may be involved in the regulation of self-renewal of CSCs. FOXM1 is essential for the maintenance of HSCs (30). Mechanistically, loss of FOXM1 downregulates cyclin-dependent kinase inhibitors (e.g., p21, p27) by directly suppressing the expression of the gene encoding NURR1 (30). Given that LSCs share properties with normal HSCs, FOXM1 might control LSCs in CML.

Why FOXM1 is overexpressed in malignant hematologic cells is not fully understood. Our results revealed that NF-κB activation might execute three layers of regulation of FOXM1: increasing FOXM1 nuclear translocation, direct binding to the FOXM1 gene promoter to increase transcription, and facilitating the stability of FOXM1.

Although the interplay of p65 and β-catenin is controversial, more studies have supported a cooperative contribution of NF-κB and β-catenin in tumorigenesis and metastasis (31, 32). Simultaneous activation of both β-catenin and NF-κB signaling pathways but neither alone is required for the enhanced CSC phenotypes (33). These findings support a connection between NF-κB and β-catenin. Concordantly, reports have shown the complex crosstalk between NF-κB and WNT/β-catenin in a mouse model of smoke-induced inflammation with lung cancer growth (32). Mice lacking myeloid RelA/p65 displayed tumor growth delay on inhibition of the WNT/β-catenin pathway (34). Our findings provide a mechanistic explanation for such a connection. We found that NF-κB also positively regulates β-catenin by increasing its protein level. NF-κB may increase β-catenin protein stability because we found that pharmacologic inhibition of NF-κB by niclosamide increased ubiquitin-mediated degradation of β-catenin.

Conversely, activation of a canonical WNT/β-catenin pathway resulted in the simultaneous translocation of p65 and β-catenin. Ectopic expression of β-catenin increased p65 protein level, but silencing β-catenin decreased p65 protein level and transcriptional activity. Crosstalk between β-catenin and NF-κB may involve other molecules besides FOXM1. For instance, Schön and colleagues (35) demonstrated that β-catenin regulates NF-κB probably via TNFRSF19, a β-catenin target gene in colorectal cancer cells. Likewise, in our study, forced expression of β-catenin increased p65 protein level, whereas silencing β-catenin decreased p65 protein level and transcriptional activity.

The interplay of FOXM1 and β-catenin in glioma cells was demonstrated by Zhang and colleagues (16). Consistent with that report (16), we confirmed the interdependence of FOXM1 and β-catenin in CML cells. WNT3A stimulation triggered coupled nuclear translocation of β-catenin and FOXM1. Silencing β-catenin lowered FOXM1 expression, while forced expression of β-catenin increased FOXM1 level. Reciprocally, manipulation of FOXM1 positively affected β-catenin levels.

Taken together, we discovered the positive feedback loop of p65/FOXM1/β-catenin in CML LSCs. Therefore, targeting this p65/FOXM1/β-catenin regulatory loop may be a promising approach to eliminate CML LSCs (a proposed model, Fig. 6J).

Pharmacologic disruption of the p65/FOXM1/β-catenin regulatory loop

Niclosamide has been an oral anthelmintic drug used to treat tapeworms for approximately 50 years (17). Recent studies from different groups including us have independently shown that niclosamide is effective against CSCs in acute myeloid leukemia, ovarian carcinoma, breast cancer, glioblastoma, and colon cancer (36–39). The underlying mechanisms may involve inactivation of pathways of NF-κB and WNT/β-catenin and increased ROS production in CSCs (17, 37, 40, 41). Niclosamide lowers the levels of Dishevelled-2 (DVL2) and LRP6, components of the WNT pathway (40, 41). Inhibition of NF-κB niclosamide involves Tak1 and IKK in the NF-κB pathway (17). In the current study, we discovered that niclosamide and its water-soluble derivative downregulated, and thereby disrupted the regulatory loop of p65/FOXM1/β-catenin in human CML CD34+ cells and in LSCs of CML mice, respectively. Niclosamide treatment decreased the survival and self-renewal capacity of human CML LSCs in vitro and in long-term engraftment in vivo. In addition, parenteral p-niclosamide administration significantly prolonged the survival of human BCR-ABL gene–driven CML mice and decreased the in vivo LSC frequency in the bone marrow and spleen of CML mice. Our findings in CML are consistent with previous reports in diverse types of cancer such as ovarian cancer and acute myeloid leukemia (17, 42), and shed new light on its antineoplastic mechanism.

In short, our findings provide new insight into the self-renewal regulatory network of CML LSCs and offer a rational approach to eliminate LSCs in CML. These results warrant further clinical study of parenteral p-niclosamide in CML.

No potential conflicts of interest were disclosed.

Conception and design: B. Jin, J. Pan

Development of methodology: B. Jin, J. Pan

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): B. Jin, C. Wang, J. Pan

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): B. Jin, C. Wang, J. Pan

Writing, review, and/or revision of the manuscript: B. Jin, J. Pan

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): K. Ding

Study supervision: J. Pan

Other (provide specimens of CML patients): J. Li, X. Du

The authors thank Dr. Hans Clever (Department of Immunology, University Hospital Utrecht, the Netherlands) for generously providing plasmid encoding FOXM1 gene promoter–driven luciferase (LUC) reporter. The authors also thank Dr. Sai-Ching J. Yeung (The University of Texas MD Anderson Cancer Center, Houston, TX) for critical reading of the manuscript.

This study was supported by grants from National Natural Science Funds (81025021, U1301226, 81373434, and 91213304; to J. Pan), the National Basic Research Program of China (973 program grant 2009CB825506; to J. Pan), the Research Foundation of Education Bureau of Guangdong Province, China (grant cxzd1103; to J. Pan), the Research Foundation of Guangzhou Bureau of Science and Technology, the Fundamental Research Funds for the Central Universities (to J. Pan), the Natural Science Foundation of Guangdong province (grant 2015A030312014; to J. Pan), and the Fundamental Research Funds of the State Key Laboratory of Ophthalmology (grant 2015QN07; to B. Jin).

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.
Prost
S
,
Relouzat
F
,
Spentchian
M
,
Ouzegdouh
Y
,
Saliba
J
,
Massonnet
G
, et al
Erosion of the chronic myeloid leukaemia stem cell pool by PPARγ agonists
.
Nature
2015
;
525
:
380
3
.
2.
O'Hare
T
,
Zabriskie
MS
,
Eiring
AM
,
Deininger
MW
. 
Pushing the limits of targeted therapy in chronic myeloid leukaemia
.
Nat Rev Cancer
2012
;
12
:
513
26
.
3.
Kalmanti
L
,
Saussele
S
,
Lauseker
M
,
Muller
MC
,
Dietz
CT
,
Heinrich
L
, et al
Safety and efficacy of imatinib in CML over a period of 10 years: data from the randomized CML-study IV
.
Leukemia
2015
;
29
:
1123
32
.
4.
Druker
BJ
. 
Translation of the Philadelphia chromosome into therapy for CML
.
Blood
2008
;
112
:
4808
17
.
5.
Nguyen
T
,
Dai
Y
,
Attkisson
E
,
Kramer
L
,
Jordan
N
,
Nguyen
N
, et al
HDAC inhibitors potentiate the activity of the BCR/ABL kinase inhibitor KW-2449 in imatinib-sensitive or -resistant BCR/ABL+ leukemia cells in vitro and in vivo
.
Clin Cancer Res
2011
;
17
:
3219
32
.
6.
Cortes
JE
,
Kim
DW
,
Pinilla-Ibarz
J
,
le Coutre
P
,
Paquette
R
,
Chuah
C
, et al
A phase 2 trial of ponatinib in Philadelphia chromosome-positive leukemias
.
N Engl J Med
2013
;
369
:
1783
96
.
7.
Jain
P
,
Kantarjian
H
,
Jabbour
E
,
Gonzalez
GN
,
Borthakur
G
,
Pemmaraju
N
, et al
Ponatinib as first-line treatment for patients with chronic myeloid leukaemia in chronic phase: a phase 2 study
.
Lancet Haematol
2015
;
2
:
e376
e83
.
8.
Chen
Y
,
Li
S
. 
Molecular signatures of chronic myeloid leukemia stem cells
.
Biomark Res
2013
;
1
:
21
.
9.
Neviani
P
,
Santhanam
R
,
Oaks
JJ
,
Eiring
AM
,
Notari
M
,
Blaser
BW
, et al
FTY720, a new alternative for treating blast crisis chronic myelogenous leukemia and Philadelphia chromosome-positive acute lymphocytic leukemia
.
J Clin Invest
2007
;
117
:
2408
21
.
10.
Chu
S
,
McDonald
T
,
Lin
A
,
Chakraborty
S
,
Huang
Q
,
Snyder
DS
, et al
Persistence of leukemia stem cells in chronic myelogenous leukemia patients in prolonged remission with imatinib treatment
.
Blood
2011
;
118
:
5565
72
.
11.
Li
L
,
Wang
L
,
Wang
Z
,
Ho
Y
,
McDonald
T
,
Holyoake
TL
, et al
Activation of p53 by SIRT1 inhibition enhances elimination of CML leukemia stem cells in combination with imatinib
.
Cancer Cell
2012
;
21
:
266
81
.
12.
Lien
WH
,
Fuchs
E
. 
Wnt some lose some: transcriptional governance of stem cells by Wnt/beta-catenin signaling
.
Genes Dev
2014
;
28
:
1517
32
.
13.
Heidel
FH
,
Bullinger
L
,
Feng
Z
,
Wang
Z
,
Neff
TA
,
Stein
L
, et al
Genetic and pharmacologic inhibition of beta-catenin targets imatinib-resistant leukemia stem cells in CML
.
Cell Stem Cell
2012
;
10
:
412
24
.
14.
Zhao
C
,
Blum
J
,
Chen
A
,
Kwon
HY
,
Jung
SH
,
Cook
JM
, et al
Loss of beta-catenin impairs the renewal of normal and CML stem cells in vivo
.
Cancer Cell
2007
;
12
:
528
41
.
15.
Gallipoli
P
,
Pellicano
F
,
Morrison
H
,
Laidlaw
K
,
Allan
EK
,
Bhatia
R
, et al
Autocrine TNF-alpha production supports CML stem and progenitor cell survival and enhances their proliferation
.
Blood
2013
;
122
:
3335
9
.
16.
Zhang
N
,
Wei
P
,
Gong
A
,
Chiu
WT
,
Lee
HT
,
Colman
H
, et al
FoxM1 promotes beta-catenin nuclear localization and controls Wnt target-gene expression and glioma tumorigenesis
.
Cancer Cell
2011
;
20
:
427
42
.
17.
Jin
Y
,
Lu
Z
,
Ding
K
,
Li
J
,
Du
X
,
Chen
C
, et al
Antineoplastic mechanisms of niclosamide in acute myelogenous leukemia stem cells: inactivation of the NF-kappaB pathway and generation of reactive oxygen species
.
Cancer Res
2010
;
70
:
2516
27
.
18.
Korver
W
,
Roose
J
,
Heinen
K
,
Weghuis
DO
,
de Bruijn
D
,
van Kessel
AG
, et al
The human TRIDENT/HFH-11/FKHL16 gene: structure, localization, and promoter characterization
.
Genomics
1997
;
46
:
435
42
.
19.
Uranishi
H
,
Tetsuka
T
,
Yamashita
M
,
Asamitsu
K
,
Shimizu
M
,
Itoh
M
, et al
Involvement of the pro-oncoprotein TLS (translocated in liposarcoma) in nuclear factor-kappa B p65-mediated transcription as a coactivator
.
J Biol Chem
2001
;
276
:
13395
401
.
20.
Chen
YH
,
Yang
CK
,
Xia
M
,
Ou
CY
,
Stallcup
MR
. 
Role of GAC63 in transcriptional activation mediated by beta-catenin
.
Nucleic Acids Res
2007
;
35
:
2084
92
.
21.
Yang
WL
,
Wang
J
,
Chan
CH
,
Lee
SW
,
Campos
AD
,
Lamothe
B
, et al
The E3 ligase TRAF6 regulates Akt ubiquitination and activation
.
Science
2009
;
325
:
1134
8
.
22.
Jin
B
,
Ding
K
,
Pan
J
. 
Ponatinib induces apoptosis in imatinib-resistant human mast cells by dephosphorylating mutant D816V KIT and silencing beta-catenin signaling
.
Mol Cancer Ther
2014
;
13
:
1217
30
.
23.
Lu
Z
,
Jin
Y
,
Qiu
L
,
Lai
Y
,
Pan
J
. 
Celastrol, a novel HSP90 inhibitor, depletes Bcr-Abl and induces apoptosis in imatinib-resistant chronic myelogenous leukemia cells harboring T315I mutation
.
Cancer Lett
2010
;
290
:
182
91
.
24.
Song
L
,
Li
J
,
Zhang
D
,
Liu
ZG
,
Ye
J
,
Zhan
Q
, et al
IKKbeta programs to turn on the GADD45alpha-MKK4-JNK apoptotic cascade specifically via p50 NF-kappaB in arsenite response
.
J Cell Biol
2006
;
175
:
607
17
.
25.
Copland
M
,
Hamilton
A
,
Elrick
LJ
,
Baird
JW
,
Allan
EK
,
Jordanides
N
, et al
Dasatinib (BMS-354825) targets an earlier progenitor population than imatinib in primary CML but does not eliminate the quiescent fraction
.
Blood
2006
;
107
:
4532
9
.
26.
Hu
Y
,
Smyth
GK
. 
ELDA: extreme limiting dilution analysis for comparing depleted and enriched populations in stem cell and other assays
.
J Immunol Methods
2009
;
347
:
70
8
.
27.
Zhang
H
,
Peng
C
,
Hu
Y
,
Li
H
,
Sheng
Z
,
Chen
Y
, et al
The Blk pathway functions as a tumor suppressor in chronic myeloid leukemia stem cells
.
Nat Genet
2012
;
44
:
861
71
.
28.
Zhang
B
,
Strauss
AC
,
Chu
S
,
Li
M
,
Ho
Y
,
Shiang
KD
, et al
Effective targeting of quiescent chronic myelogenous leukemia stem cells by histone deacetylase inhibitors in combination with imatinib mesylate
.
Cancer Cell
2010
;
17
:
427
42
.
29.
Guzman
ML
,
Neering
SJ
,
Upchurch
D
,
Grimes
B
,
Howard
DS
,
Rizzieri
DA
, et al
Nuclear factor-kappaB is constitutively activated in primitive human acute myelogenous leukemia cells
.
Blood
2001
;
98
:
2301
7
.
30.
Hou
Y
,
Li
W
,
Sheng
Y
,
Li
L
,
Huang
Y
,
Zhang
Z
, et al
The transcription factor Foxm1 is essential for the quiescence and maintenance of hematopoietic stem cells
.
Nat Immunol
2015
;
16
:
810
8
.
31.
Hwang
I
,
Choi
YS
,
Jeon
MY
,
Jeong
S
. 
NF-kappaB p65 represses beta-catenin-activated transcription of cyclin D1
.
Biochem Biophys Res Commun
2010
;
403
:
79
84
.
32.
Du
Q
,
Zhang
X
,
Cardinal
J
,
Cao
Z
,
Guo
Z
,
Shao
L
, et al
Wnt/beta-catenin signaling regulates cytokine-induced human inducible nitric oxide synthase expression by inhibiting nuclear factor-kappaB activation in cancer cells
.
Cancer Res
2009
;
69
:
3764
71
.
33.
Jia
D
,
Yang
W
,
Li
L
,
Liu
H
,
Tan
Y
,
Ooi
S
, et al
beta-Catenin and NF-kappaB co-activation triggered by TLR3 stimulation facilitates stem cell-like phenotypes in breast cancer
.
Cell Death Differ
2015
;
22
:
298
310
.
34.
Li
D
,
Beisswenger
C
,
Herr
C
,
Hellberg
J
,
Han
G
,
Zakharkina
T
, et al
Myeloid cell RelA/p65 promotes lung cancer proliferation through Wnt/beta-catenin signaling in murine and human tumor cells
.
Oncogene
2014
;
33
:
1239
48
.
35.
Schon
S
,
Flierman
I
,
Ofner
A
,
Stahringer
A
,
Holdt
LM
,
Kolligs
FT
, et al
beta-catenin regulates NF-kappaB activity via TNFRSF19 in colorectal cancer cells
.
Int J Cancer
2014
;
135
:
1800
11
.
36.
Sack
U
,
Walther
W
,
Scudiero
D
,
Selby
M
,
Kobelt
D
,
Lemm
M
, et al
Novel effect of antihelminthic Niclosamide on S100A4-mediated metastatic progression in colon cancer
.
J Natl Cancer Inst
2011
;
103
:
1018
36
.
37.
Pan
JX
,
Ding
K
,
Wang
CY
. 
Niclosamide, an old antihelminthic agent, demonstrates antitumor activity by blocking multiple signaling pathways of cancer stem cells
.
Chin J Cancer
2012
;
31
:
178
84
.
38.
Yo
YT
,
Lin
YW
,
Wang
YC
,
Balch
C
,
Huang
RL
,
Chan
MW
, et al
Growth inhibition of ovarian tumor-initiating cells by niclosamide
.
Mol Cancer Ther
2012
;
11
:
1703
12
.
39.
Wieland
A
,
Trageser
D
,
Gogolok
S
,
Reinartz
R
,
Hofer
H
,
Keller
M
, et al
Anticancer effects of niclosamide in human glioblastoma
.
Clin Cancer Res
2013
;
19
:
4124
36
.
40.
Osada
T
,
Chen
M
,
Yang
XY
,
Spasojevic
I
,
Vandeusen
JB
,
Hsu
D
, et al
Antihelminth compound niclosamide downregulates Wnt signaling and elicits antitumor responses in tumors with activating APC mutations
.
Cancer Res
2011
;
71
:
4172
82
.
41.
Lu
W
,
Lin
C
,
Roberts
MJ
,
Waud
WR
,
Piazza
GA
,
Li
Y
. 
Niclosamide suppresses cancer cell growth by inducing Wnt co-receptor LRP6 degradation and inhibiting the Wnt/beta-catenin pathway
.
PLoS One
2011
;
6
:
e29290
.
42.
Arend
RC
,
Londono-Joshi
AI
,
Samant
RS
,
Li
Y
,
Conner
M
,
Hidalgo
B
, et al
Inhibition of Wnt/beta-catenin pathway by niclosamide: a therapeutic target for ovarian cancer
.
Gynecol Oncol
2014
;
134
:
112
20
.