Purpose: Quiescent leukemia stem cells (LSC) are important resources of resistance and relapse in chronic myelogenous leukemia (CML). Thus, strategies eradicating CML LSCs are required for cure. In this study, we discovered that AXL tyrosine kinase was selectively overexpressed in primary CML CD34+ cells. However, the role of AXL and its ligand Gas6 secreted by stromal cells in the regulation of self-renewal capacity of LSCs has not been well investigated.

Experimental Design: The function of CML CD34+ cells was evaluated by flow cytometer, CFC/replating, long-term culture-initiating cells (LTC-IC), CML mouse model driven by human BCR-ABL gene and NOD-scid-IL2Rg−/− (NSI) mice.

Results: AXL was selectively overexpressed in primary CML CD34+ cells. AXL knockdown reduced the survival and self-renewal capacity of human CML CD34+ cells. Pharmacologic inhibition of AXL reduced the survival and self-renewal capacity of human CML LSCs in vitro and in long-term grafts in NSI mice. Human CML CD34+ cells conscripted bone marrow–derived stromal cells (BMDSC) and primary mesenchymal stem cells (MSC) to secrete Gas6 to form a paracrine loop that promoted self-renewal of LSCs. Suppression of AXL by shRNA and inhibitor prolonged survival of CML mice and reduced the growth of LSCs in mice. Gas6/AXL ligation stabilizes β-catenin in an AKT-dependent fashion in human CML CD34+ cells.

Conclusions: Our findings improve the understanding of LSC regulation and validate Gas6/AXL as a pair of therapeutic targets to eliminate CML LSCs. Clin Cancer Res; 23(11); 2842–55. ©2016 AACR.

This article is featured in Highlights of This Issue, p. 2607

Translational Relevance

Quiescent leukemia stem cells (LSC) that are intrinsically insensitive to BCR-ABL tyrosine kinase inhibitors is a root cause of relapse for chronic myelogenous leukemia (CML) treated with imatinib mesylate. Treatments to eliminate LSCs are urgently needed to cure CML. In this study, we discovered AXL receptor tyrosine kinase was overexpressed in CML, and was an oncogenic protein required for survival and self-renewal of CML LSCs. Human CML CD34+ cells conscripted bone marrow–derived stromal cells (BMDSC) to secrete extra Gas6 to form a paracrine loop that promoted self-renewal of LSCs. AXL knockdown and pharmacologic inhibition of AXL prolonged survival of BCR-ABL–driven CML mice and eliminated LSCs in mice. Gas6/AXL ligation stabilizes β-catenin in an AKT-dependent manner in human CML CD34+ cells. Disruption of Gas6/AXL axis is an attractive approach to selectively eliminate LSCs in CML.

Chronic myelogenous leukemia (CML) is a type of myeloproliferative disorder characterized by uncontrolled clonal expansion of immature granulocytes and their precursors (1). CML is believed to stem from the malignant transformation of bone marrow hematopoietic stem cells (HSC) triggered by a fusion gene BCR-ABL because of a reciprocal chromosomal translocation t(9,22)(q34;q11) (2). Most adult patients with chronic-phase CML respond well to tyrosine kinase inhibitor (TKI) imatinib mesylate (IM; ref. 3). However, acquired resistance to IM conferred by multiple mechanisms [e.g., BCR-ABL mutations (4), leukemia stem cells (LSC; ref. 5)] is an emerging problem. In addition, BCR-ABL–positive pediatric acute lymphoblastic leukemia and adult accelerated phase- or blast-crisis-CML usually respond poor to TKIs (6).

LSCs in CML show common characteristics of cancer stem cells (CSC; refs. 7–9). These traits endow LSCs with resistance to IM. CML patients with complete molecular response, still showed long-term BCR-ABL–negative LSCs (10). Only approaches to effectively eliminate LSCs when combined with IM may help cure CML (7, 8, 11). Therefore, development of rational approaches to eliminate LSCs is imperative.

The self-renewal capacity of LSCs is regulated by intrinsic CSC regulation pathways (e.g., WNT/β-catenin, Hedgehog; refs. 12, 13), metabolism regulators (e.g., ALOX5, SCD; refs. 14, 15), transcription factors (e.g., Foxo3a, HIF1α, γ-catenin; refs. 16–18) and epigenetic regulators (e.g., PRMT5; refs. 19, 20). However, the whole regulation network of LSCs is not fully realized. The complexity is enhanced by the LSC local microenvironment in bone marrow (21). Stromal cells tightly regulate the self-renewal of LSCs via direct contact and by secreting soluble cytokines (22, 23).

AXL is a member of the TAM receptor tyrosine kinases (TAMR), which also includes Mer and Tyro3 (Sky) (24, 25). Two highly homologous ligands for TAMRs are growth-arrest–specific gene 6 (Gas6) and protein S (25). Gas6 has the highest affinity for AXL, whereas protein S predominantly binds to Tyro3 and Mer (26). AXL was originally identified in 1991 from patients with CML (27), then found to be overexpressed in multiple human cancers (24). AXL is involved in acquired resistance to endothelial growth factor receptor inhibitors in lung cancer (28), FLT3-targeted therapy in acute myelogenous leukemia (29) and IM through a bypass mechanism (30).

AXL overexpression was found to induce the epithelial–mesenchymal transition (EMT) and increase tumorigenicity in breast cancer cells (31). The underlying mechanism may involve upregulating β-catenin, snail, slug, and NF-κB triggered by Gas6/AXL activation (31). Recently, AXL was identified as a key regulator for mesenchymal glioblastoma stem-like cells (32). We discovered that among the three TAMR family members, AXL was selectively expressed in primary CML CD34+ cells and hypothesized that overexpression of AXL in LSCs and Gas6 by bone marrow stromal cells might increase the self-renewal capacity of LSCs and confer resistance to IM. In this study, we tested this hypothesis and investigated the impact of forced expression of AXL, silencing it via shRNA or inactivating it by small-molecule inhibitors on survival and self-renewal capacity of LSCs in vitro and in CML mice.

Primary leukemia cells

Peripheral blood or bone marrow samples (Supplementary Table S1) were obtained from patients with CML and from healthy adult donors in Sun Yat-sen Memorial Hospital, The First Affiliated Hospital of Sun Yat-sen University, Guangdong General Hospital, and The First Affiliated Hospital of Jinan University after informed consent according to the institutional guidelines and the Declaration of Helsinki principles. The isolation and culture of human CD34+ cells were performed as described previously (16, 19, 20, 33).

Mesenchymal stromal cells isolation and culture

Peripheral blood samples were obtained from patients with CML and from healthy adult donors. Mononuclear cells isolated by histopaque gradient centrifugation were resuspended in DMEM-low glucose medium (Gibco) supplemented with 10% FBS and 1% penicillin–streptomycin. The cells were kept at 37°C in a humidified incubator with 5% CO2 for 48 hours. After discarding the nonadherent cells, the adherent cells were cultured for 2 to 4 weeks and used as mesenchymal stromal cells (MSC) in subsequent experiments (34).

Western blot analysis

Whole cell lysates were prepared in RIPA buffer (35). The membranes were scanned by using the Odyssey infrared imaging system (LI-COR). Detailed information for antibodies and their sources is described in Supplementary Materials and Methods.

Enzyme-linked immunosorbent assay

Human and mouse Gas6 immunoassay involved use of the Quantikine ELISA Kit (R&D Systems; ref. 36). Cell culture supernatants or plasma from CML patients or healthy individuals were used for detecting the secreted Gas6.

Lentivirus transduction in CML CD34+ cells

Lentivirus were produced by transfection in 293T cells with control shRNA (Scramble) or specific shRNA targeting AXL together with the pCMV-dR8.2 packing construct and the pCMV-VSVG envelope construct. CML or NBM CD34+ cells (1 × 106 cells/mL) were transduced by spinoculation (1,500 g, 90 minutes, 32°C) with virus-containing supernatants two rounds. Cells were harvested 48 hours later for further analysis (16, 20).

Analysis of cell apoptosis in primary CD34+ cells

Apoptosis of primary CD34+ cells in CML patients or NBM was determined as described previously (35).

CFSE assay.

CFSE-labeled cells were cultured with XL880 and R428 for 96 hours, cells were harvested and stained with Annexin V-PE, and quiescent cell apoptosis (CD34+ CFSEmax Annexin V+) was analyzed by using with BD FACS Aria II flow cytometer (8, 16, 20).

Colony-formation cell/replating and long-term culture-initiating cell (LTC-IC) assay

Colony-formation cell (CFC)/replating and LTC-IC assay were performed as reported previously (16, 20, 37). Detailed information is described in Supplementary Materials and Methods.

CML mouse model driven by human BCRABL fusion gene

The retrovirus was produced by transient transfection with the MSCV-BCR-ABL-IRES-EGFP construct in Plat-E cells (9). Bone marrow cells from 6- to 8-week-old donor male C57BL/6 mice primed with 5-fluorouracil (5-FU) were stimulated with cytokines in vitro, then transplanted by tail-vein injection into irradiated (550 cGy) recipient female C57BL/6 mice after transduction two rounds with the MSCV-BCR-ABL-IRES-EGFP retrovirus (16, 20). All the mice developed CML in approximately 2 to 3 weeks.

The detailed information for the treatment of CML mice and detection of LSK, LT-HSC and ST-HSC is described in Supplementary Materials and Methods.

Engraftment of human cells in immunodeficient mice

Primary CD34+ cells from 2 CML patients (1 CP-CML patient and 1 BP-CML patient) were cultured with XL880 (1.0 μmol/L) for 72 hours, then cells (1 × 106 cells/mouse) were transplanted by tail-vein injection into sublethally irradiated (300 cGy) 8-week-old NOD-scid-IL2Rg−/− (NSI) which were generated by TALEN-mediated Il2rg gene targeting in NOD/SCID mice (Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences; refs. 20, 38). Mice were euthanized after 12 weeks, then bone marrow cells and spleen cells were harvested; the engraftment of human cells was analyzed by using BD FACS Aria II flow cytometer.

Statistical analysis

All experiments were performed 3 times, and results are presented as mean ± SD. GraphPad Prism 5.0 (GraphPad Software) was used for statistical analysis. Data for 2 groups were compared by Student t test and for multiple groups by one-way ANOVA, post hoc intergroup comparisons, Tukey test. A P value of <0.05 was considered statistically significant.

Selective overexpression of AXL in primary CML CD34+ cells

We first examined the expression of AXL and other TAMRs members in primary CD34+ cells purified with immunomagnetic columns from CML patients and NBM from healthy donors. The mRNA level of AXL, but not other TAMR members (e.g., Mer and Tyro3), was significantly higher in the primary CD34+ cells from CML patients than from NBM (Fig. 1A). The levels of phospho-AXL and AXL were higher in CML CD34+ cells than NBM CD34+ cells as determined by Western blot analysis (Fig. 1B). Flow cytometry results showed that there was surface expression of AXL in CML CD34+ cells (Fig. 1C). We next examined whether BCR-ABL regulated AXL expression in CML CD34+ cells. The levels of phospho-AXL and AXL were not changed after treatment with IM (Fig. 1D). However, BCR-ABL knockdown by lentiviral shRNA in the primary CML CD34+ cells dramatically decreased the AXL mRNA levels (Fig. 1E). These data suggest that AXL expression is dependent on BCR-ABL protein level rather than its tyrosine kinase activity.

Figure 1.

AXL is overexpressed in CML CD34+ cells and knockdown AXL reduces survival and CFC/replating ability in human CML CD34+ cells. A, Selective overexpression of AXL in primary CD34+ cells from CML patients. The mRNA level of AXL (left), Mer (middle) and Tyro3 (right) in CML patients (n = 6) and normal bone marrow (NBM; n = 4) CD34+ cells analyzed by qRT-PCR. ***, P < 0.0001, Student t test. B, The protein levels of phospho-AXL and AXL were detected by Western blot analysis in CML CD34+ cells (n = 5) and NBM CD34+ cells (n = 3). C, The surface expression of AXL in CML CD34+ cells was examined by flow cytometer. D, CML CD34+ cells were treated with IM for 24 hours, the expression of phospho-AXL and AXL were detected by Western blot analysis (left); the mRNA levels of AXL were detected by qRT-PCR (right). E,BCR-ABL knockdown decreased AXL mRNA levels. Human CML CD34+ (n = 3) were transduced with control shRNA (Scramble), shBCR-ABL #1, or shBCR-ABL #2 lentivirus for 48 hours. The knockdown effect of BCR-ABL was confirmed by Western blot analysis (left). AXL mRNA level was determined by qRT-PCR (right). F and J, Human CML CD34+ (n = 3) were transduced with control shRNA (Scramble), shAXL #1, or shAXL #2 for 48 hours, then treated with IM (2.5 μmol/L) for 24 hours. The knockdown effect of AXL was confirmed by qRT-PCR (F) and Western blot (G) analysis. H, Representative flow cytometry plots for apoptosis in CML CD34+ cells. I, Apoptosis in CD34+ cells in CML analyzed by Annexin V-FITC and anti–CD38-PE labeling. J, BCR-ABL phosphorylation in CML CD34+ cells was inhibited after IM treatment. K, Knockdown of AXL suppressed the serially replating capacity of CML CD34+ cells. The same number of Scramble, shAXL #1 or shAXL #2 cells (5,000 cells/well) were seeded in methylcellulose medium (H4434) for three rounds, and colonies were counted on day 14 after each round of culture. *, P < 0.05; **, P < 0.01; ***, P < 0.0001, one-way ANOVA, post hoc intergroup comparisons, Tukey test.

Figure 1.

AXL is overexpressed in CML CD34+ cells and knockdown AXL reduces survival and CFC/replating ability in human CML CD34+ cells. A, Selective overexpression of AXL in primary CD34+ cells from CML patients. The mRNA level of AXL (left), Mer (middle) and Tyro3 (right) in CML patients (n = 6) and normal bone marrow (NBM; n = 4) CD34+ cells analyzed by qRT-PCR. ***, P < 0.0001, Student t test. B, The protein levels of phospho-AXL and AXL were detected by Western blot analysis in CML CD34+ cells (n = 5) and NBM CD34+ cells (n = 3). C, The surface expression of AXL in CML CD34+ cells was examined by flow cytometer. D, CML CD34+ cells were treated with IM for 24 hours, the expression of phospho-AXL and AXL were detected by Western blot analysis (left); the mRNA levels of AXL were detected by qRT-PCR (right). E,BCR-ABL knockdown decreased AXL mRNA levels. Human CML CD34+ (n = 3) were transduced with control shRNA (Scramble), shBCR-ABL #1, or shBCR-ABL #2 lentivirus for 48 hours. The knockdown effect of BCR-ABL was confirmed by Western blot analysis (left). AXL mRNA level was determined by qRT-PCR (right). F and J, Human CML CD34+ (n = 3) were transduced with control shRNA (Scramble), shAXL #1, or shAXL #2 for 48 hours, then treated with IM (2.5 μmol/L) for 24 hours. The knockdown effect of AXL was confirmed by qRT-PCR (F) and Western blot (G) analysis. H, Representative flow cytometry plots for apoptosis in CML CD34+ cells. I, Apoptosis in CD34+ cells in CML analyzed by Annexin V-FITC and anti–CD38-PE labeling. J, BCR-ABL phosphorylation in CML CD34+ cells was inhibited after IM treatment. K, Knockdown of AXL suppressed the serially replating capacity of CML CD34+ cells. The same number of Scramble, shAXL #1 or shAXL #2 cells (5,000 cells/well) were seeded in methylcellulose medium (H4434) for three rounds, and colonies were counted on day 14 after each round of culture. *, P < 0.05; **, P < 0.01; ***, P < 0.0001, one-way ANOVA, post hoc intergroup comparisons, Tukey test.

Close modal

Forced overexpressing AXL in CML cells increases side population and ALDH+ cells and contributes to IM resistance

Forced overexpression of human AXL in K562 cells reduced sensitivity to IM, suggesting that increased AXL confers resistance to IM (Supplementary Fig. S1A and S1B). The percentage of side population (SP) cells and Aldefluor+ cells were also significantly increased in K562 overexpressing AXL as compared with the control (Supplementary Fig. S1C and S1D). In addition, AXL overexpression upregulated the expression of stemness-associated genes ALDHA1, OCT4, SOX2 and NOTCH1 but not LIN28A and NANOG (Supplementary Fig. S1E).

Silencing AXL reduces survival and self-renewal capacity of human primary CML CD34+ cells

To further define the role of AXL in CML CD34+ cells, we transduced the primary CD34+ cells purified from CML patients or NBM with scramble shRNA or lentivirus AXL shRNA for 48 hours, then treated them with IM for 24 hours. The knockdown effect of AXL in CML CD34+ cells and NBM CD34+ cells was confirmed by qRT-PCR (Fig. 1F and Supplementary Fig. S2A). Given the gene homology of AXL with Mer and Tyro3, we examined the levels of these receptors in cells treated with shRNA against AXL by Western blot analysis. The results showed that the effect of AXL knockdown was of selective (Fig. 1G). AXL silencing significantly increased apoptosis in CML CD34+ cells (Fig. 1H and I) but not in NBM CD34+ cells (Supplementary Fig. S2B and S2C). The effect of AXL knockdown on CML CD34+ cells was augmented with combined IM treatment (Fig. 1I and Supplementary Fig. S2C), in which the BCR-ABL activity as reflected by its phosphorylation (Y245) was completely inhibited by IM (Fig. 1J). Because the self-renewal capacity is an important trait of CSCs, we determined whether silencing AXL by its specific lentivirus shRNA impaired the CFC/replating ability of CML stem cells. The primary CD34+ cells of human CML and NBM were transduced with lentiviral shAXL for 72 hours and underwent three rounds of 14-day serially replating of CFC culture in methylcellulose medium. When AXL was silenced, the CFC/replating ability was significantly decreased in human CML CD34+ but not in NBM CD34+ cells (Fig. 1K and Supplementary Fig. S2D). Thus, AXL knockdown reduced CML CD34+ cell survival and growth, and IM treatment further suppressed CML CD34+ cell growth.

AXL knockdown eliminates LSCs and prolongs survival of BCR-ABL–driven CML mice

We next used a widely used human BCR-ABL gene-driven CML mouse model (Fig. 2A) to examine the in vivo effect of AXL knockdown on CML LSCs (14, 16, 20, 33). We knocked down AXL with shRNA in spleen GFP+ cells from the first generation of CML mice (Fig. 2B), then transplanted these spleen cells into sublethally irradiated recipient C57BL/6 mice. The results showed that AXL knockdown alone or combination with IM potently blocked splenomegaly (Fig. 2C) and markedly decreased the proportion of BCR-ABL–expressing (i.e., GFP+) leukemia cells, myeloid cells (Gr-1+ Mac-1+) in bone marrow cells (Fig. 2D and E) and splenic cells (Supplementary Fig. S3A and S3B) of CML mice.

Figure 2.

AXL knockdown eliminates LSCs and prolongs survival in CML mice. A, A schema to generate retroviral human BCR-ABL–driven CML mouse model. B,AXL knockdown in spleen cells from CML mice was confirmed by Western blot analysis. C, A representative photograph of spleens from each group. D–I,AXL knockdown eliminated LSCs and sensitized CML LSCs to IM in vivo. GFP+ (leukemia) cells (D) and GFP+ myeloid cells (Gr-1+ Mac-1+; E) in bone marrow were analyzed by flow cytometer. F, Representative flow cytometry plots of LSKs, LT-HSCs, and ST-HSCs in bone marrow from CML mice. Results for the GFP+ populations in bone marrow are shown: LSK cells (G), LT-HSCs (H), and ST-HSCs (I). J, Representative flow cytometry plots of progenitor cells in the bone marrow of mice. GMP cells (K) and CMP cells (L) in bone marrow. ***, P < 0.0001, compared with control; #, P < 0.05; ##, P < 0.01, shAXL compared with shAXL+IM; ns: not significant, one-way ANOVA, post hoc intergroup comparisons, Tukey test. M,AXL knockdown significantly prolonged survival of CML mice. Kaplan–Meier survival curves were plotted. Control (n = 10), IM (n = 12), shAXL (n = 10), shAXL+IM (n = 10), Control versus IM: ***, P = 0.0003; Control versus shAXL: ***, P < 0.0001; Control versus shAXL+IM: ***, P < 0.0001. Log-rank test.

Figure 2.

AXL knockdown eliminates LSCs and prolongs survival in CML mice. A, A schema to generate retroviral human BCR-ABL–driven CML mouse model. B,AXL knockdown in spleen cells from CML mice was confirmed by Western blot analysis. C, A representative photograph of spleens from each group. D–I,AXL knockdown eliminated LSCs and sensitized CML LSCs to IM in vivo. GFP+ (leukemia) cells (D) and GFP+ myeloid cells (Gr-1+ Mac-1+; E) in bone marrow were analyzed by flow cytometer. F, Representative flow cytometry plots of LSKs, LT-HSCs, and ST-HSCs in bone marrow from CML mice. Results for the GFP+ populations in bone marrow are shown: LSK cells (G), LT-HSCs (H), and ST-HSCs (I). J, Representative flow cytometry plots of progenitor cells in the bone marrow of mice. GMP cells (K) and CMP cells (L) in bone marrow. ***, P < 0.0001, compared with control; #, P < 0.05; ##, P < 0.01, shAXL compared with shAXL+IM; ns: not significant, one-way ANOVA, post hoc intergroup comparisons, Tukey test. M,AXL knockdown significantly prolonged survival of CML mice. Kaplan–Meier survival curves were plotted. Control (n = 10), IM (n = 12), shAXL (n = 10), shAXL+IM (n = 10), Control versus IM: ***, P = 0.0003; Control versus shAXL: ***, P < 0.0001; Control versus shAXL+IM: ***, P < 0.0001. Log-rank test.

Close modal

AXL knockdown or combination with IM greatly decreased the proportions of LSK cells (LinSca-1+c-Kit+), LT-HSCs (LSK Flt3CD150+CD48) and ST-HSCs (LSK Flt3CD150CD48) in bone marrow cells (Fig. 2F–I) and splenic cells (Supplementary Fig. S3C–S3E) of CML mice. Similarly, the proportions of granulocyte-macrophage progenitors (GMPs, LinSca-1c-Kit+CD34+FcγRII/IIIhig) and common myeloid progenitors (CMPs, LinSca-1c-Kit+CD34+FcγRII/IIIlow) in bone marrow cells (Fig. 2J–L) and splenic cells (Supplementary Fig. S3F and S3G) were significantly reduced in AXL knockdown mice. IM treatment alone did not significantly reduce the proportions of LSKs, LT-HSCs, ST-HSCs, GMPs and CMPs in bone marrow cells and splenic cells of CML mice (Fig. 2F–L and Supplementary Fig. S3C–S3G), which agreed with previous reports (7, 8). AXL knockdown significantly prolonged survival of CML mice (Fig. 2M). In conclusion, silencing AXL eliminated LSCs in vivo and prolonged survival of CML mice.

Pharmacologic inhibition of AXL reduces survival and self-renewal capacity of human CML CD34+ cells

Next, we determined the effect of the small-molecule AXL inhibitors XL880 (39) and R428 (40) on survival of primary CML CD34+ cells. The data showed that XL880 treatment increased the apoptosis in CML CD34+ cells (Fig. 3A, top). Combined treatment with XL880 and IM enhanced the apoptosis in CML CD34+ cells, suggesting that XL880 sensitized CML CD34+ cells to apoptosis in response to IM. XL880 alone or with IM did not induce apoptosis in CD34+ cells from NBM (Supplementary Fig. S4A and S4B). Similar results were obtained when the primary CML CD34+ cells were exposed to R428, a more selective AXL inhibitor (Fig. 3A, bottom). In these settings, both XL880 and R428 treatment blocked the AXL tyrosine phosphorylation in CML CD34+ cells (Fig. 3B).

Figure 3.

Pharmacologic inhibition of AXL activity suppresses survival and self-renewal of human primary CML CD34+ cells. A, Representative flow cytometry plots for apoptosis in human CML CD34+ cells treated with IM and AXL inhibitors XL880 (top) or R428 (bottom). CD34+ cells from CML bone marrow were treated with XL880 (top) or R428 (bottom) with IM (2.5 μmol/L) for 24 hours; apoptotic cells were examined by Annexin V-FITC and anti–CD38-PE labeling. B, XL880 or R428 treatment inhibited AXL phosphorylation in CML CD34+ cells. C, Pharmacologic inhibition of AXL by XL880 selectively induced apoptosis in quiescent CML LSCs. Results for CFSEmax and Annexin V+ cells are shown. **, P < 0.01, Student t test. D, Pharmacologic inhibition of AXL by XL880 and R428 induced apoptosis in quiescent CML LSCs. Results for CFSEmax and Annexin V+ cells are shown. E and F, Pharmacologic inhibition of AXL selectively suppressed the serially replating capacity of CML LSCs. G, Pharmacologic inhibition of AXL selectively lessened the long-term culture-initiating cell (LTC-IC) capacity of CML LSCs. **, P < 0.01; ***, P < 0.0001, one-way ANOVA, post hoc intergroup comparisons, Tukey test.

Figure 3.

Pharmacologic inhibition of AXL activity suppresses survival and self-renewal of human primary CML CD34+ cells. A, Representative flow cytometry plots for apoptosis in human CML CD34+ cells treated with IM and AXL inhibitors XL880 (top) or R428 (bottom). CD34+ cells from CML bone marrow were treated with XL880 (top) or R428 (bottom) with IM (2.5 μmol/L) for 24 hours; apoptotic cells were examined by Annexin V-FITC and anti–CD38-PE labeling. B, XL880 or R428 treatment inhibited AXL phosphorylation in CML CD34+ cells. C, Pharmacologic inhibition of AXL by XL880 selectively induced apoptosis in quiescent CML LSCs. Results for CFSEmax and Annexin V+ cells are shown. **, P < 0.01, Student t test. D, Pharmacologic inhibition of AXL by XL880 and R428 induced apoptosis in quiescent CML LSCs. Results for CFSEmax and Annexin V+ cells are shown. E and F, Pharmacologic inhibition of AXL selectively suppressed the serially replating capacity of CML LSCs. G, Pharmacologic inhibition of AXL selectively lessened the long-term culture-initiating cell (LTC-IC) capacity of CML LSCs. **, P < 0.01; ***, P < 0.0001, one-way ANOVA, post hoc intergroup comparisons, Tukey test.

Close modal

To further identify the effectiveness of AXL inhibition on quiescent LSCs, we labeled primary CD34+ cells of human CML patients or NBM with carboxyfluorescein diacetate succinimidyl ester (CFSE), then incubated with XL880 (1.0 μmol/L) for 96 hours. XL880 treatment markedly increased apoptosis in undivided CML CD34+ cells but not undivided NBM HSCs (Fig. 3C and Supplementary Fig. S4C and S4D). R428 treatment exhibited similar effect compared with XL880 in undivided CML CD34+ cells (Fig. 3D).

We next evaluated whether inhibiting AXL by XL880 and R428 impaired the self-renewal ability of CML stem cells by CFC/replating and LTC-IC assay. In the presence of XL880 and R428, the CFC/replating ability was greatly decreased in the primary CML CD34+ cells but not in NBM CD34+ cells (Fig. 3E and F and Supplementary Fig. S4E). Similarly, XL880 greatly decreased the number of LTC-IC–derived colony-forming units for CML bone marrow cells as compared with NBM cells (Fig. 3G and Supplementary Fig. S4F). Combined treatment with XL880 and IM enhanced the decreased CML LTC-IC frequency as compared with either agent alone (Fig. 3G). Thus, pharmacologic inhibition of AXL may reduce the survival and self-renewal ability of human CML LSCs.

Pharmacologic inhibition of AXL prolongs survival of CML mice and reduces the percentage of CML stem cells in mice

To examine the in vivo effect of pharmacologic inhibition of AXL on CML LSCs, we used the CML mouse model again mentioned before. Mice were treated with XL880, IM or both (Fig. 4A). Splenomegaly was ameliorated in mice that received XL880 or both XL880 and IM (Fig. 4B). XL880-treated CML mice showed significantly prolonged survival, and XL880 with IM extended survival as compared with each alone (Fig. 4C).

Figure 4.

Pharmacologic inhibition of AXL reduces in vivo growth of CML stem cells and prolongs survival of CML mice. A, A schema of drug treatment in CML mice. B, A representative photograph of spleens from each group. C, Treatment with XL880 alone or with IM significantly prolonged the survival of CML mice. Kaplan-Meier survival curves of CML mice treated with IM, XL880 or their combination. Control (n = 9), IM (n = 9), XL880 (n = 9), XL880+IM (n = 9), Control versus IM: *, P = 0.0327; Control versus XL880: *, P = 0.0210; Control versus XL880+IM: ***, P = 0.0001; XL880 versus XL880+IM: *, P = 0.0298. Log-rank test. D–I, XL880 eliminated LSCs and sensitized CML LSCs to IM in vivo. GFP+ (leukemia) cells (D) and GFP+ myeloid cells (Gr-1+ Mac-1+; E) in bone marrow were analyzed by flow cytometry. F, Representative flow cytometry plots of LSKs, LT-HSCs, and ST-HSCs in bone marrow from CML mice. Results for the GFP+ populations in the bone marrow are shown: LSK cells (G), LT-HSCs (H), and ST-HSCs (I). J, Representative flow cytometry plots of progenitor cells in the bone marrow of mice. GMP cells (K) and CMP cells (L) in bone marrow. *, P < 0.05; ***, P < 0.0001, compared with control; #, P < 0.05; ##, P < 0.01; ###, P < 0.0001, XL880 compared with XL880+IM; ns: not significant, one-way ANOVA, post hoc intergroup comparisons, Tukey test.

Figure 4.

Pharmacologic inhibition of AXL reduces in vivo growth of CML stem cells and prolongs survival of CML mice. A, A schema of drug treatment in CML mice. B, A representative photograph of spleens from each group. C, Treatment with XL880 alone or with IM significantly prolonged the survival of CML mice. Kaplan-Meier survival curves of CML mice treated with IM, XL880 or their combination. Control (n = 9), IM (n = 9), XL880 (n = 9), XL880+IM (n = 9), Control versus IM: *, P = 0.0327; Control versus XL880: *, P = 0.0210; Control versus XL880+IM: ***, P = 0.0001; XL880 versus XL880+IM: *, P = 0.0298. Log-rank test. D–I, XL880 eliminated LSCs and sensitized CML LSCs to IM in vivo. GFP+ (leukemia) cells (D) and GFP+ myeloid cells (Gr-1+ Mac-1+; E) in bone marrow were analyzed by flow cytometry. F, Representative flow cytometry plots of LSKs, LT-HSCs, and ST-HSCs in bone marrow from CML mice. Results for the GFP+ populations in the bone marrow are shown: LSK cells (G), LT-HSCs (H), and ST-HSCs (I). J, Representative flow cytometry plots of progenitor cells in the bone marrow of mice. GMP cells (K) and CMP cells (L) in bone marrow. *, P < 0.05; ***, P < 0.0001, compared with control; #, P < 0.05; ##, P < 0.01; ###, P < 0.0001, XL880 compared with XL880+IM; ns: not significant, one-way ANOVA, post hoc intergroup comparisons, Tukey test.

Close modal

XL880 alone or with IM markedly decreased the proportion of BCR-ABL–expressing (GFP+) leukemia cells, myeloid cells (Gr-1+ Mac-1+) in bone marrow cells (Fig. 4D and E) and splenic cells (Supplementary Fig. S5A and S5B) of CML mice. XL880 alone or with IM greatly decreased the proportions of LSK cells (LinSca-1+c-Kit+), LT-HSCs (LSK Flt3CD150+CD48), and ST-HSCs (LSK Flt3CD150CD48) in bone marrow cells (Fig. 4F–I) and splenic cells (Supplementary Fig. S5C–S5E) of CML mice.

The proportions of granulocyte-macrophage progenitors (GMPs, LinSca-1c-Kit+CD34+FcγRII/IIIhig) and common myeloid progenitors (CMPs, LinSca-1c-Kit+CD34+FcγRII/IIIlow) in bone marrow cells (Fig. 4J–L) and splenic cells (Supplementary Fig. S5F and S5G) were significantly reduced in XL880-treated mice. Taken together, pharmacologic inhibition of AXL eliminated LSCs and sensitized CML LSCs to IM in vivo.

AXL inhibition reduces long-term engraftment of human CML CD34+ cells in NSI mice

We investigated the effect of ex vivo treatment with XL880 in human CML CD34+ cells on their ability to be engrafted in NOD-scid-IL2Rg−/− (NSI) mice (Supplementary Fig. S6A). XL880 treatment reduced the engraftment of human CML CD45+ cells in bone marrow (Supplementary Fig. S6B) and spleens (Supplementary Fig. S6C) at 12 weeks after transplantation. The engrafted human CML CD45+ cells sorted by flow cytometry from NSI murine bone marrow cells expressed BCR-ABL, and XL880 treatment markedly decreased BCR-ABL mRNA levels (Supplementary Fig. S6D). The proportions of engrafted human CML CD33+ and CD14+ myeloid cells in NSI murine bone marrow cells were decreased with XL880 treatment (Supplementary Fig. S6E and S6F). XL880 treatment may selectively target human primitive CML cells with in vivo engraftment capacity.

Gas6/AXL paracrine loop confers self-renewal capacity of human CML CD34+ cells

Wild-type AXL executes its function dependently by binding its ligand Gas6. Given that AXL was overexpressed in CML LSCs, we wondered whether CML CD34+ cells produced extra Gas6 to form an autocrine regulation loop. We evaluated Gas6 mRNA level in primary CML and NBM CD34+ cells and found that Gas6 gene transcription was similar in the primary CD34+ cells from CML patients (Fig. 5A). The data did not support a Gas6/AXL autocrine regulation loop in CML LSCs.

Figure 5.

Gas6/AXL paracrine regulation loop elevates self-renewal of human CML CD34+ cells. A, mRNA levels of Gas6 gene in CML (n = 6) and NBM (n = 4) CD34+ cells analyzed by qRT-PCR. B, Plasma levels of human Gas6 in CML patients versus healthy individuals detected by ELISA. *, P < 0.05, Student t test. C, MSCs were isolated from CML patients (n = 2) and healthy adult donors (n = 3). Human Gas6 mRNA levels in the MSCs cells were detected by qRT-PCR. D, Gas6 levels in the MSCs cell culture supernatants were measured by ELISA. E, CD34+ cells from CML patients (n = 2) were co-cultured with MSCs for 10 days, then the cells (10,000/35 mm dish) were seeded in the methylcellulose medium. Colonies were counted on day 14. ***, P < 0.0001, Student t test. F, Human and murine Gas6 level in the culture medium of human CML K562 cells, murine bone marrow–derived stromal cells (BMDSCs) OP9, or co-culture detected by ELISA. ND, not detectable. G, Gas6 stimulation induced AXL activation in CML CD34+ cells. H, Gas6 increased the growth of CML cells. Starved CML cells were stimulated with recombinant human Gas6 (500 ng/mL) for 48 h; cell number was examined by trypan blue exclusion assay. *, P < 0.05; **, P < 0.001, compared with control. I, Total cell numbers were plotted after incubation in the presence or absence of AXL-Fc chimeric proteins (1.0 μg/mL) without or with human BMDSCs HS5. J, Number of K562 cells counted after incubation in the presence or absence of AXL-Fc chimeric proteins (1.0 μg/mL) without or with Gas6 stimulation. K, Gas6 stimulation increased the self-renewal ability of CML CD34+ cells. L–O, Disturbed Gas6/AXL paracrine regulation loop inhibited the self-renewal of human CML stem cells. L and M, CML CD34+ cells were transduced with human AXL shRNA lentivirus for 48 hours, co-cultured with HS5 cells for 72 hours, then underwent CFC/replating assay. N and O, HS5 cells were infected with human Gas6 shRNA lentivirus 48 hours, co-cultured with CML CD34+ cells for 72 hours, then underwent CFC/replating assay. *, P < 0.05; **, P < 0.01; ***, P < 0.0001, one-way ANOVA, post hoc intergroup comparisons, Tukey test.

Figure 5.

Gas6/AXL paracrine regulation loop elevates self-renewal of human CML CD34+ cells. A, mRNA levels of Gas6 gene in CML (n = 6) and NBM (n = 4) CD34+ cells analyzed by qRT-PCR. B, Plasma levels of human Gas6 in CML patients versus healthy individuals detected by ELISA. *, P < 0.05, Student t test. C, MSCs were isolated from CML patients (n = 2) and healthy adult donors (n = 3). Human Gas6 mRNA levels in the MSCs cells were detected by qRT-PCR. D, Gas6 levels in the MSCs cell culture supernatants were measured by ELISA. E, CD34+ cells from CML patients (n = 2) were co-cultured with MSCs for 10 days, then the cells (10,000/35 mm dish) were seeded in the methylcellulose medium. Colonies were counted on day 14. ***, P < 0.0001, Student t test. F, Human and murine Gas6 level in the culture medium of human CML K562 cells, murine bone marrow–derived stromal cells (BMDSCs) OP9, or co-culture detected by ELISA. ND, not detectable. G, Gas6 stimulation induced AXL activation in CML CD34+ cells. H, Gas6 increased the growth of CML cells. Starved CML cells were stimulated with recombinant human Gas6 (500 ng/mL) for 48 h; cell number was examined by trypan blue exclusion assay. *, P < 0.05; **, P < 0.001, compared with control. I, Total cell numbers were plotted after incubation in the presence or absence of AXL-Fc chimeric proteins (1.0 μg/mL) without or with human BMDSCs HS5. J, Number of K562 cells counted after incubation in the presence or absence of AXL-Fc chimeric proteins (1.0 μg/mL) without or with Gas6 stimulation. K, Gas6 stimulation increased the self-renewal ability of CML CD34+ cells. L–O, Disturbed Gas6/AXL paracrine regulation loop inhibited the self-renewal of human CML stem cells. L and M, CML CD34+ cells were transduced with human AXL shRNA lentivirus for 48 hours, co-cultured with HS5 cells for 72 hours, then underwent CFC/replating assay. N and O, HS5 cells were infected with human Gas6 shRNA lentivirus 48 hours, co-cultured with CML CD34+ cells for 72 hours, then underwent CFC/replating assay. *, P < 0.05; **, P < 0.01; ***, P < 0.0001, one-way ANOVA, post hoc intergroup comparisons, Tukey test.

Close modal

The stemness of LSCs is tightly regulated by the components of its surrounding niches (22). We next examined the plasma concentration of Gas6 in CML patients and found increased plasma Gas6 level in CML patients as compared with healthy donors (Fig. 5B). We detected the cellular human Gas6 expression in the primary MSCs from CML patients and healthy individuals by qRT-PCR, the results showed that the Gas6 mRNA levels were higher in CML MSCs than normal MSCs (Fig. 5C). Furthermore, the secreted Gas6 levels as determined by ELISA were also increased in the cell culture supernatants of CML MSCs compared with normal MSCs (Fig. 5D). These data together suggest the cellular and secreted Gas6 levels appears increased in CML MSCs. Co-cultured with CML MSCs for 10 days significantly promoted growth of CML CD34+ cells (Supplementary Fig. S7A). The number of CFCs for CML CD34+ cells co-cultured with MSCs was significantly elevated in comparison with those without co-culture with MSCs (Fig. 5E).

To determine the reason for the increased Gas6 level in CML, we seeded human leukemia K562 cells on the feeder layer of murine BMDSCs OP9 cells, then evaluated murine Gas6 secretion in the culture medium by ELISA. The level of murine Gas6 was higher in co-culture than with OP9 or K562 cells alone. Of note, murine Gas6 but not human soluble Gas6 level was increased in the co-culture system, suggesting that CML cells might educate OP9 cells to secrete Gas6 (Fig. 5F). Indeed, with the number of K562 cells increased in the system of a constant number of OP9 cells, the soluble murine Gas6 level was increased, CML cells indeed educated OP9 cells to dose-dependently secrete Gas6 (Supplementary Fig. S7B). These data suggest that BMDSCs secreted Gas6, which activated the AXL membrane receptor on leukemia cells to form a paracrine regulation loop.

To assess whether Gas6 regulates CML responses in vitro, we found that Gas6 stimulation induced AXL activation (Fig. 5G) and increased growth of CML cells (Fig. 5H). The protective role of stromal cells or Gas6 on CML cells was then investigated in the co-culture system with or without AXL-Fc chimeric protein treatment. AXL-Fc contains the extracellular domain of the AXL protein and the Fc portion of human IgG1. AXL-Fc binds Gas6 and prevents it from interacting with AXL (39). We counted the number of K562 cells when incubated in the presence or absence of AXL-Fc chimeric proteins without or with co-culture with human HS5 BMDSCs or Gas6 stimulation. AXL-Fc treatment counteracted the protective role of BMDSCs (Fig. 5I) or Gas6 (Fig. 5J).

In addition, Gas6 stimulation increased the self-renewal ability of CML CD34+ cells detected by CFC/replating assay (Fig. 5K). Next, we evaluated whether blocking the Gas6/AXL paracrine loop inhibited the self-renewal of human CML CD34+ cells. After transduced with human AXL lentiviral shRNA for 48 hours, the CML CD34+ cells were co-cultured with human HS5 cells for another 72 hours for CFC/replating assay. In parallel, HS5 cells were infected with human Gas6 lentiviral shRNA for 48 hours, then co-cultured with the primary CML CD34+ cells for 72 hours for CFC/replating assay. Silencing AXL in primary CML CD34+ cells or Gas6 in stromal cells markedly inhibited the CFC/replating ability of primary CML CD34+ cells in the co-culture system (Fig. 5L–O). Similarly, blocking the Gas6/AXL paracrine loop diminished the self-renewal of human CML CD34+ cells co-cultured with human CML MSCs (Supplementary Fig. S7C–S7F).

Collectively, the Gas6/AXL paracrine loop positively regulated the self-renewal capacity of human CML LSCs, hinting that targeting the Gas6/AXL paracrine loop may be of benefit for treatment of CML.

Gas6/AXL ligation stabilizes β-catenin in an AKT-dependent manner in CML CD34+ cells

The WNT/β-catenin pathway regulates the self-renewal capacity of CML LSCs (12, 41). We examined whether AXL affected β-catenin level. Levels of β-catenin and its downstream target genes (e.g., c-MYC and LEF1) were greatly increased in AXL- overexpressing K562 cells (Fig. 6A, left and middle). Given that DVL (Dishevelled) is a positive upstream regulator in the WNT/β-catenin pathway, we set to examine DVL. Among the three isoforms of DVL, only DVL2 and DVL3 were detectable in K562 cells. The AXL-overexpressing K562 cells showed an increased DVL3 but not DVL2 (Fig. 6A, left and middle). Similar results were obtained in Gas6-stimulated K562 cells (Fig. 6A, right). Conversely, AXL knockdown in the primary CML CD34+ cells dramatically inhibited WNT/β-catenin pathway (Fig. 6B). However, the level of BCR-ABL was unaltered, suggesting that BCR-ABL was not likely to be regulated by AXL in the primary CML CD34+ cells (Fig. 6B). Similarly, AXL inhibition by XL880 reduced levels of β-catenin and downstream genes in primary human CML CD34+ cells (Supplementary Fig. S8A).

Figure 6.

Gas6/AXL stabilized β-catenin in an AKT-dependent manner in human CML CD34+ cells. A, AXL overexpression or ligation by Gas6 activates WNT/β-catenin signaling in K562 cells. Cells stably expressing AXL (C1 and C6; left), (C2 and B3; middle) or starved K562 cells were stimulated with Gas6 (400 ng/mL; right) for 24 hours; the activation of WNT/β-catenin pathway was examined by Western blot analysis. B, Knockdown of AXL by lentiviral shRNA inhibited β-catenin pathway but not BCR-ABL expression and activity in primary human CML CD34+ cells detected by Western blot analysis. C and D, AXL overexpression or Gas6 stimulation decelerated the turnover rate of β-catenin protein. E, Gas6/AXL signaling increased β-catenin levels in an AKT-dependent fashion. The phosphorylation of AKT and ERK1/2 were examined by Western blot analysis in K562 cells stably overexpressing AXL (C1 and C6; left) or starved K562 cells were stimulated with Gas6 for 24 hours (middle). Pharmacologic inhibition of AXL inhibited phosphorylation of AKT and ERK1/2 in primary human CML CD34+ cells (right). F, Gas6/AXL ligation enables β-catenin accumulation via AKT but not ERK1/2. CML CD34+ cells were pretreated with MEK inhibitor U0126 (10.0 μmol/L) or PI3K inhibitor LY294002 (10.0 μmol/L) for 2 hours, then stimulated with Gas6 (400 ng/mL) for 24 hours; the protein level of β-catenin and phosphorylation of AKT and ERK1/2 were examined by Western blot analysis. *, P < 0.05; **, P < 0.01; ***, P < 0.0001, versus the corresponding control. G, Primary CML CD34+ cells were transduced with lentivirus BCR-ABL shRNA for 48 hours, then the cells were stimulated with Gas6 for 24 hours, BCR-ABL knockdown and AKT activation were confirmed by Western blot analysis. H, Ectopic expression of β-catenin rescued the AXL knockdown-mediated decrease in CFC formation in CML CD34+ cells. CML CD34+ cells were transduced with Scramble (Scr) or AXL lentivirus for 48 hours, then the cells underwent electrotransfection with pcDNA3 (Vector) or pcDNA3-β-catenin (β-catenin) for another 48 hours. AXL knockdown and β-catenin overexpression were confirmed by Western blot analysis (left). The cells were seeded in the methylcellulose medium. Colonies were counted on day 14 (right). *, P < 0.05, Student t test. I, Proposed model by which self-renewal capacity of CML LSCs is positively regulated by Gas6/AXL paracrine loop.

Figure 6.

Gas6/AXL stabilized β-catenin in an AKT-dependent manner in human CML CD34+ cells. A, AXL overexpression or ligation by Gas6 activates WNT/β-catenin signaling in K562 cells. Cells stably expressing AXL (C1 and C6; left), (C2 and B3; middle) or starved K562 cells were stimulated with Gas6 (400 ng/mL; right) for 24 hours; the activation of WNT/β-catenin pathway was examined by Western blot analysis. B, Knockdown of AXL by lentiviral shRNA inhibited β-catenin pathway but not BCR-ABL expression and activity in primary human CML CD34+ cells detected by Western blot analysis. C and D, AXL overexpression or Gas6 stimulation decelerated the turnover rate of β-catenin protein. E, Gas6/AXL signaling increased β-catenin levels in an AKT-dependent fashion. The phosphorylation of AKT and ERK1/2 were examined by Western blot analysis in K562 cells stably overexpressing AXL (C1 and C6; left) or starved K562 cells were stimulated with Gas6 for 24 hours (middle). Pharmacologic inhibition of AXL inhibited phosphorylation of AKT and ERK1/2 in primary human CML CD34+ cells (right). F, Gas6/AXL ligation enables β-catenin accumulation via AKT but not ERK1/2. CML CD34+ cells were pretreated with MEK inhibitor U0126 (10.0 μmol/L) or PI3K inhibitor LY294002 (10.0 μmol/L) for 2 hours, then stimulated with Gas6 (400 ng/mL) for 24 hours; the protein level of β-catenin and phosphorylation of AKT and ERK1/2 were examined by Western blot analysis. *, P < 0.05; **, P < 0.01; ***, P < 0.0001, versus the corresponding control. G, Primary CML CD34+ cells were transduced with lentivirus BCR-ABL shRNA for 48 hours, then the cells were stimulated with Gas6 for 24 hours, BCR-ABL knockdown and AKT activation were confirmed by Western blot analysis. H, Ectopic expression of β-catenin rescued the AXL knockdown-mediated decrease in CFC formation in CML CD34+ cells. CML CD34+ cells were transduced with Scramble (Scr) or AXL lentivirus for 48 hours, then the cells underwent electrotransfection with pcDNA3 (Vector) or pcDNA3-β-catenin (β-catenin) for another 48 hours. AXL knockdown and β-catenin overexpression were confirmed by Western blot analysis (left). The cells were seeded in the methylcellulose medium. Colonies were counted on day 14 (right). *, P < 0.05, Student t test. I, Proposed model by which self-renewal capacity of CML LSCs is positively regulated by Gas6/AXL paracrine loop.

Close modal

Next, we ascertained whether AXL overexpression activates TCF4/LEF1-dependent gene transcription. The data indicated that AXL overexpression increased TCF4/LEF1-dependent luciferase activity (Supplementary Fig. S8B). AXL overexpression also increased the levels of β-catenin target genes (e.g., AXIN2, c-MYC, LEF1 and CCND1; Supplementary Fig. S8C). In a separate approach, the addition of Gas6 increased TCF4/LEF1-dependent luciferase activity (Supplementary Fig. S8D). Conversely, treatment with AXL inhibitor XL880 in K562 cells decreased luciferase activity (Supplementary Fig. S8E), as well as the transcription of these β-catenin target genes in human primary CML CD34+ cells (Supplementary Fig. S8F).

To explore the mechanism by which AXL regulates β-catenin, we performed time–chase experiments. AXL overexpression decelerated the turnover of β-catenin protein (Fig. 6C). Similar results were obtained with Gas6 stimulation (Fig. 6D). K562 cells stably overexpressing AXL and Gas6-stimulated K562 cells showed increased phospho-(S9)-GSK3β, phospho-AKT and phospho-ERK1/2 levels (Fig. 6E, left and middle). Conversely, pharmacologic inhibition of AXL decreased the levels in human primary CML CD34+ cells (Fig. 6E, right). Because phospho-(S9)-GSK3β inhibited the activity of GSK3β and decreased the degradation of β-catenin in primary CML CD34+ cells (Fig. 6E, right), we hypothesized that AXL activation might phosphorylate GSK3β at S9. To identify the regulator responsible for GSK3β S9 phosphorylation, we used specific inhibitors against MEK/ERK1/2 or PI3K/AKT pathway. The PI3K inhibitor LY294002 but not MEK inhibitor U0126 abrogated the Gas6-induced accumulation of β-catenin (Fig. 6F), which suggests that AKT but not ERK1/2 mediated the phosphorylation of GSK3β S9 in primary CML CD34+ cells.

To examine the effect of Gas6/AXL on AKT activation in the absence of BCR/ABL, we knocked down BCR-ABL in primary CD34+ cells with lentivirus BCR-ABL shRNA, and then the cells were stimulated with Gas6, the results showed that AKT was activated with Gas6 stimulation in BCR-ABL knockdown cells (Fig. 6G), which is indicative of the critical role of Gas6/AXL on AKT.

Finally, we determined whether AXL exerted its function through β-catenin. The CML CD34+ cells were transduced with Scramble (Scr) or AXL lentivirus for 48 hours, then the cells underwent transfection with pcDNA3 (Vector) or pcDNA3-β-catenin (β-catenin) for another 48 hours. Ectopic expression of β-catenin rescued the AXL knockdown-mediated decrease in CFC formation in CML CD34+ cells (Fig. 6H).

Taken together, Gas6/AXL ligation leads to AKT-dependent stabilization of β-catenin protein in CML CD34+ cells.

LSCs are important resources of TKI resistance and CML relapse. In the present study, we discovered that among three of the TAMR family members, AXL was selectively overexpressed in primary CML CD34+ cells. Inhibiting AXL by shRNA or small-molecule inhibitors reduced the survival and self-renewal capacity of human CML LSCs. AXL knockdown and pharmacologic inhibition of AXL prolonged the survival of CML mice and eliminated LSCs in mice. AXL inhibition reduced long-term engraftment of human CML CD34+ cells in NSI mice. A Gas6/AXL paracrine loop conferred the self-renewal capacity of human CML CD34+ cells. Gas6/AXL ligation stabilizes β-catenin in an AKT-dependent manner in CML CD34+ cells. Our findings improve the understanding of LSC regulation and validate a pharmacologic target for eliminating LSCs. Our study represents the first description of Gas6/AXL as a critical regulator of the self-renewal capacity of CML LSCs conferring IM resistance.

Among all members of the TAMR family, only AXL has been reported to be involved in the EMT in glioblastoma and breast cancer (31, 32, 42). Consistently, we identified AXL overexpression in CML LSCs. AXL-overexpressing in CML cells increased the proportions of SP and Aldefluor+ cells and stemness-related genes including ALDHA1. In agreement with our findings, erlotinib-resistant non–small cell lung carcinoma cell populations contained a greater proportion of Aldefluor+ cells than parental erlotinib-responsive cells with concurrent increase in AXL expression (43).

Overexpressed AXL may be abundantly accessed by its ligand Gas6. However, identifying any gain-of-function mutations or genomic amplification in AXL in CML LSCs needs further investigation.

Little is known about the underlying mechanism of AXL overexpression in LSCs. Our results showed that AXL mRNA and protein levels were higher in human primary CML CD34+ cells as compared with NBM counterparts. Furthermore, AXL transcriptional expression appears dependent on BCR-ABL protein level rather than its tyrosine kinase activity. It was previously reported that transcription factors (e.g., AP1) and miRNAs (e.g., miR-34a) lead to AXL overexpression (40). TIG1 in breast cancer cells can increase AXL protein stabilization by physical interaction (44). Further study is needed to identify the mechanism of increased AXL expression in LSCs.

Multiple types of cells, including fibroblasts, endothelial cells, osteoblasts, liver cells, and cancer cells can secrete Gas6 (45). In bone marrow, Gas6 is believed to be secreted by fibroblasts and other stromal cells to maintain hematopoietic homeostasis (46). Our results support that Gas6 ligation to AXL to promote survival and self-renewal capacity of LSCs. Conversely, Gas6 knockdown in different stromal cells (e.g., HS5, primary CML MSCs) attenuated the self-renewal of LSCs. Similarly, the addition of soluble AXL-Fc into the co-culture of LSCs and HS5 BMDSCs blocked the pro-survival and pro-self-renewal effect of Gas6 on CML LSCs. Our data did not support that LSCs generated Gas6 for autocrine regulation. This finding disagrees with a previous report that bulk AML cells enhanced Gas6 secretion to form an autocrine loop (45). The difference may be due to variant cell types. Of interest, CML cells educated BMDSCs to secrete Gas6, which can increase the self-renewal ability of CML CD34+ cells.

Overexpression of AXL in CML cells increased the protein level of β-catenin and facilitated its transcriptional activity, as reflected by the expression of its downstream target genes. Stimulation of Gas6 in CML cells provoked similar effects. Conversely, silencing AXL decreased β-catenin protein and its transcriptional activity. In function, the critical role of β-catenin in CML LSCs was revealed as forced expression of β-catenin rescued the AXL knockdown-mediated decrease in CFC formation in CD34+ cells.

BCR-ABL tyrosine kinase can directly phosphorylate β-catenin at Y86 and Y654 residues to increase β-catenin protein stability (47). However, the phosphorylation at Y654 of β-catenin was not altered in AXL-overexpressing CML cells or when starved K562 cells were stimulated with Gas6 (data not shown), which suggests that AXL might not increase β-catenin protein stability in such a direct manner. Therefore, we turned our attention to the canonical cascade of WNT/β-catenin. After inhibiting AXL by shRNA or small-molecule inhibitor XL880 in CML CD34+ cells, phospho-(S9)-GSK3β was markedly decreased, which was helpful to explain the decreased β-catenin levels. Regarding the mediator to render the decreased phospho-(S9)-GSK3β, our results pinpointed its upstream regulators—AKT but not ERK1/2. AKT is able to phosphorylate β-catenin at S552 to facilitate its nucleus accumulation and transcriptional activity (48). Consistently, AKT is required for Gas6/AXL to protect cells from apoptosis in ovarian cancer (49). Because LY294002 is a PI3K inhibitor with toxicity, future studies should define the potential application value of more specific inhibitors of the PI3K/AKT pathway in eliminating LSCs.

In conclusion, overexpression of AXL or ligation of Gas6/AXL increases the self-renewal capacity of CML LSCs in vitro and in vivo. LSCs may educate BMDSCs to secrete Gas6, assumably forming a vicious circle (proposed model, Fig. 6I). Our findings may provide a rationale for a clinical trial of IM combined with AXL inhibitor foretinib, which is tolerant during a phase II clinical trial, in resistant and refractory CML (50).

No potential conflicts of interest were disclosed.

Conception and design: Y. Jin, J. Pan

Development of methodology: Y. Jin, C. Liu, J. Zhou

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): Y. Jin, D. Nie, Y. Lu, Y. Li, C. Liu, J. Zhou, J. Pan

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): Y. Jin, J. Zhou, J. Pan

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

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): Y. Jin, J. Pan

Study supervision: J. Pan

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

This study was supported by grants from National Natural Science funds (grant nos. U1301226, 81373434, 81025021, and 91213304 to J. Pan; 81473247 and 81673451; to Y. Jin), Guangdong Natural Science Funds for Distinguished Young Scholars (Grant no. 2016A030306036 to Y. Jin the Research Foundation of Education Bureau of Guangdong Province, China (grant no. cxzd1103; to J. Pan).

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.
Druker
BJ
. 
Translation of the Philadelphia chromosome into therapy for CML
.
Blood
2008
;
112
:
4808
17
.
2.
Perrotti
D
,
Jamieson
C
,
Goldman
J
,
Skorski
T
. 
Chronic myeloid leukemia: mechanisms of blastic transformation
.
J Clin Invest
2010
;
120
:
2254
64
.
3.
Druker
BJ
,
Talpaz
M
,
Resta
DJ
,
Peng
B
,
Buchdunger
E
,
Ford
JM
, et al
Efficacy and safety of a specific inhibitor of the BCR-ABL tyrosine kinase in chronic myeloid leukemia
.
N Engl J Med
2001
;
344
:
1031
7
.
4.
Shah
NP
,
Nicoll
JM
,
Nagar
B
,
Gorre
ME
,
Paquette
RL
,
Kuriyan
J
, et al
Multiple BCR-ABL kinase domain mutations confer polyclonal resistance to the tyrosine kinase inhibitor imatinib (STI571) in chronic phase and blast crisis chronic myeloid leukemia
.
Cancer Cell
2002
;
2
:
117
25
.
5.
Jiang
X
,
Zhao
Y
,
Smith
C
,
Gasparetto
M
,
Turhan
A
,
Eaves
A
, et al
Chronic myeloid leukemia stem cells possess multiple unique features of resistance to BCR-ABL targeted therapies
.
Leukemia
2007
;
21
:
926
35
.
6.
Quintas-Cardama
A
,
Kantarjian
H
,
Cortes
J
. 
Flying under the radar: the new wave of BCR-ABL inhibitors
.
Nat Rev Drug Discov
2007
;
6
:
834
48
.
7.
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
.
8.
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
.
9.
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
.
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.
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
.
12.
Zhao
C
,
Blum
J
,
Chen
A
,
Kwon
HY
,
Jung
SH
,
Cook
JM
, et al
Loss of β-catenin impairs the renewal of normal and CML stem cells in vivo
.
Cancer Cell
2007
;
12
:
528
41
.
13.
Zhao
C
,
Chen
A
,
Jamieson
CH
,
Fereshteh
M
,
Abrahamsson
A
,
Blum
J
, et al
Hedgehog signalling is essential for maintenance of cancer stem cells in myeloid leukaemia
.
Nature
2009
;
458
:
776
9
.
14.
Chen
Y
,
Hu
Y
,
Zhang
H
,
Peng
C
,
Li
S
. 
Loss of the Alox5 gene impairs leukemia stem cells and prevents chronic myeloid leukemia
.
Nat Genet
2009
;
41
:
783
92
.
15.
Zhang
H
,
Li
H
,
Ho
N
,
Li
D
,
Li
S
. 
Scd1 plays a tumor-suppressive role in survival of leukemia stem cells and the development of chronic myeloid leukemia
.
Mol Cell Biol
2012
;
32
:
1776
87
.
16.
Jin
Y
,
Yao
Y
,
Chen
L
,
Zhu
X
,
Jin
B
,
Shen
Y
, et al
Depletion of γ-catenin by histone deacetylase inhibition confers elimination of CML stem cells in combination with imatinib
.
Theranostics
2016
;
6
:
1947
62
.
17.
Naka
K
,
Hoshii
T
,
Muraguchi
T
,
Tadokoro
Y
,
Ooshio
T
,
Kondo
Y
, et al
TGF-β-FOXO signalling maintains leukaemia-initiating cells in chronic myeloid leukaemia
.
Nature
2010
;
463
:
676
80
.
18.
Ng
KP
,
Manjeri
A
,
Lee
KL
,
Huang
W
,
Tan
SY
,
Chuah
CT
, et al
Physiologic hypoxia promotes maintenance of CML stem cells despite effective BCR-ABL1 inhibition
.
Blood
2014
;
123
:
3316
26
.
19.
Chen
Y
,
Li
S
. 
Molecular signatures of chronic myeloid leukemia stem cells
.
Biomark Res
2013
;
1
:
21
.
20.
Jin
Y
,
Zhou
J
,
Xu
F
,
Jin
B
,
Cui
L
,
Wang
Y
, et al
Targeting methyltransferase PRMT5 eliminates leukemia stem cells in chronic myelogenous leukemia
.
J Clin Invest
2016
;
126
:
3961
80
.
21.
Goff
DJ
,
Court Recart
A
,
Sadarangani
A
,
Chun
HJ
,
Barrett
CL
,
Krajewska
M
, et al
A Pan-BCL2 inhibitor renders bone-marrow-resident human leukemia stem cells sensitive to tyrosine kinase inhibition
.
Cell Stem Cell
2013
;
12
:
316
28
.
22.
Crews
LA
,
Jamieson
CH
. 
Selective elimination of leukemia stem cells: hitting a moving target
.
Cancer Lett
2013
;
338
:
15
22
.
23.
Zhang
B
,
Ho
YW
,
Huang
Q
,
Maeda
T
,
Lin
A
,
Lee
SU
, et al
Altered microenvironmental regulation of leukemic and normal stem cells in chronic myelogenous leukemia
.
Cancer Cell
2012
;
21
:
577
92
.
24.
Graham
DK
,
DeRyckere
D
,
Davies
KD
,
Earp
HS
. 
The TAM family: phosphatidylserine sensing receptor tyrosine kinases gone awry in cancer
.
Nat Rev Cancer
2014
;
14
:
769
85
.
25.
Hafizi
S
,
Dahlback
B
. 
Gas6 and protein S. Vitamin K-dependent ligands for the Axl receptor tyrosine kinase subfamily
.
FEBS J
2006
;
273
:
5231
44
.
26.
Nagata
K
,
Ohashi
K
,
Nakano
T
,
Arita
H
,
Zong
C
,
Hanafusa
H
, et al
Identification of the product of growth arrest-specific gene 6 as a common ligand for Axl, Sky, and Mer receptor tyrosine kinases
.
J Biol Chem
1996
;
271
:
30022
7
.
27.
O'Bryan
JP
,
Frye
RA
,
Cogswell
PC
,
Neubauer
A
,
Kitch
B
,
Prokop
C
, et al
axl, a transforming gene isolated from primary human myeloid leukemia cells, encodes a novel receptor tyrosine kinase
.
Mol Cell Biol
1991
;
11
:
5016
31
.
28.
Zhang
Z
,
Lee
JC
,
Lin
L
,
Olivas
V
,
Au
V
,
LaFramboise
T
, et al
Activation of the AXL kinase causes resistance to EGFR-targeted therapy in lung cancer
.
Nat Genet
2012
;
44
:
852
60
.
29.
Park
IK
,
Mundy-Bosse
B
,
Whitman
SP
,
Zhang
X
,
Warner
SL
,
Bearss
DJ
, et al
Receptor tyrosine kinase Axl is required for resistance of leukemic cells to FLT3-targeted therapy in acute myeloid leukemia
.
Leukemia
2015
;
29
:
2382
9
.
30.
Dufies
M
,
Jacquel
A
,
Belhacene
N
,
Robert
G
,
Cluzeau
T
,
Luciano
F
, et al
Mechanisms of AXL overexpression and function in Imatinib-resistant chronic myeloid leukemia cells
.
Oncotarget
2011
;
2
:
874
85
.
31.
Asiedu
MK
,
Beauchamp-Perez
FD
,
Ingle
JN
,
Behrens
MD
,
Radisky
DC
,
Knutson
KL
. 
AXL induces epithelial-to-mesenchymal transition and regulates the function of breast cancer stem cells
.
Oncogene
2014
;
33
:
1316
24
.
32.
Cheng
P
,
Phillips
E
,
Kim
SH
,
Taylor
D
,
Hielscher
T
,
Puccio
L
, et al
Kinome-wide shRNA screen identifies the receptor tyrosine kinase AXL as a key regulator for mesenchymal glioblastoma stem-like cells
.
Stem Cell Rep
2015
;
4
:
899
913
.
33.
Jin
B
,
Wang
C
,
Li
J
,
Du
X
,
Ding
K
,
Pan
J
. 
Anthelmintic niclosamide disrupts the interplay of p65 and FOXM1/β-catenin and eradicates leukemia stem cells in chronic myelogenous leukemia
.
Clin Cancer Res
2016
Aug 4.
[Epub ahead of print]
.
PMID:27492973.
34.
Schmal
O
,
Seifert
J
,
Schaffer
TE
,
Walter
CB
,
Aicher
WK
,
Klein
G
. 
Hematopoietic stem and progenitor cell expansion in contact with mesenchymal stromal cells in a hanging drop model uncovers disadvantages of 3D culture
.
Stem Cells Int
2016
;
2016
:
4148093
.
35.
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-κB pathway and generation of reactive oxygen species
.
Cancer Res
2010
;
70
:
2516
27
.
36.
Waizenegger
JS
,
Ben-Batalla
I
,
Weinhold
N
,
Meissner
T
,
Wroblewski
M
,
Janning
M
, et al
Role of Growth arrest-specific gene 6-Mer axis in multiple myeloma
.
Leukemia
2015
;
29
:
696
704
.
37.
Neviani
P
,
Harb
JG
,
Oaks
JJ
,
Santhanam
R
,
Walker
CJ
,
Ellis
JJ
, et al
PP2A-activating drugs selectively eradicate TKI-resistant chronic myeloid leukemic stem cells
.
J Clin Invest
2013
;
123
:
4144
57
.
38.
Xiao
Y
,
Jiang
Z
,
Li
Y
,
Ye
W
,
Jia
B
,
Zhang
M
, et al
ANGPTL7 regulates the expansion and repopulation of human hematopoietic stem and progenitor cells
.
Haematologica
2015
;
100
:
585
94
.
39.
Park
IK
,
Mishra
A
,
Chandler
J
,
Whitman
SP
,
Marcucci
G
,
Caligiuri
MA
. 
Inhibition of the receptor tyrosine kinase Axl impedes activation of the FLT3 internal tandem duplication in human acute myeloid leukemia: implications for Axl as a potential therapeutic target
.
Blood
2013
;
121
:
2064
73
.
40.
Scaltriti
M
,
Elkabets
M
,
Baselga
J
. 
Molecular pathways: AXL, a membrane receptor mediator of resistance to therapy
.
Clin Cancer Res
2016
;
22
:
1313
7
.
41.
Heidel
FH
,
Bullinger
L
,
Feng
Z
,
Wang
Z
,
Neff
TA
,
Stein
L
, et al
Genetic and pharmacologic inhibition of β-catenin targets imatinib-resistant leukemia stem cells in CML
.
Cell Stem Cell
2012
;
10
:
412
24
.
42.
Bosurgi
L
,
Bernink
JH
,
Delgado Cuevas
V
,
Gagliani
N
,
Joannas
L
,
Schmid
ET
, et al
Paradoxical role of the proto-oncogene Axl and Mer receptor tyrosine kinases in colon cancer
.
Proc Natl Acad Sci U S A
2013
;
110
:
13091
6
.
43.
Corominas-Faja
B
,
Oliveras-Ferraros
C
,
Cuyas
E
,
Segura-Carretero
A
,
Joven
J
,
Martin-Castillo
B
, et al
Stem cell-like ALDH(bright) cellular states in EGFR-mutant non-small cell lung cancer: a novel mechanism of acquired resistance to erlotinib targetable with the natural polyphenol silibinin
.
Cell Cycle
2013
;
12
:
3390
404
.
44.
Wang
X
,
Saso
H
,
Iwamoto
T
,
Xia
W
,
Gong
Y
,
Pusztai
L
, et al
TIG1 promotes the development and progression of inflammatory breast cancer through activation of Axl kinase
.
Cancer Res
2013
;
73
:
6516
25
.
45.
Ben-Batalla
I
,
Schultze
A
,
Wroblewski
M
,
Erdmann
R
,
Heuser
M
,
Waizenegger
JS
, et al
Axl, a prognostic and therapeutic target in acute myeloid leukemia mediates paracrine crosstalk of leukemia cells with bone marrow stroma
.
Blood
2013
;
122
:
2443
52
.
46.
Dormady
SP
,
Zhang
XM
,
Basch
RS
. 
Hematopoietic progenitor cells grow on 3T3 fibroblast monolayers that overexpress growth arrest-specific gene-6 (GAS6)
.
Proc Natl Acad Sci U S A
2000
;
97
:
12260
5
.
47.
Coluccia
AM
,
Vacca
A
,
Dunach
M
,
Mologni
L
,
Redaelli
S
,
Bustos
VH
, et al
Bcr-Abl stabilizes β-catenin in chronic myeloid leukemia through its tyrosine phosphorylation
.
EMBO J
2007
;
26
:
1456
66
.
48.
Fang
D
,
Hawke
D
,
Zheng
Y
,
Xia
Y
,
Meisenhelder
J
,
Nika
H
, et al
Phosphorylation of β-catenin by AKT promotes β-catenin transcriptional activity
.
J Biol Chem
2007
;
282
:
11221
9
.
49.
Lee
WP
,
Wen
Y
,
Varnum
B
,
Hung
MC
. 
Akt is required for Axl-Gas6 signaling to protect cells from E1A-mediated apoptosis
.
Oncogene
2002
;
21
:
329
36
.
50.
Choueiri
TK
,
Vaishampayan
U
,
Rosenberg
JE
,
Logan
TF
,
Harzstark
AL
,
Bukowski
RM
, et al
Phase II and biomarker study of the dual MET/VEGFR2 inhibitor foretinib in patients with papillary renal cell carcinoma
.
J Clin Oncol
2013
;
31
:
181
6
.