Background: Wnt/β-catenin signaling plays a strong role in maintaining homeostasis of hematopoietic progenitor cells (HPCs).
Results: Activated β-catenin deregulates the survival of HPCs by induction of the mitochondrial apoptotic pathway.
Conclusion: Wnt/β-catenin signaling is critical for HPC pool maintenance.
Significance: This study clarifies the role of the Wnt/β-catenin signaling in HPCs by showing that it has negative impact on the function and survival of the cells.
Keywords: Bcl-2, Beta-Catenin, Hematopoiesis, Stem Cells, Wnt Pathway, Progenitor Cells
Abstract
The canonical Wnt/β-catenin signaling is activated during development, tumorigenesis, and in adult homeostasis, yet its role in maintenance of hematopoietic stem/progenitor cells is not firmly established. Here, we demonstrate that conditional expression of an active form of β-catenin in vivo induces a marked increase in the frequency of apoptosis in hematopoietic stem/progenitor cells (HSCs/HPCs). Activation of Wnt/β-catenin signaling in HPCs in vitro elevates the activity of caspases 3 and 9 and leads to a loss of mitochondrial membrane potential (ΔΨm), indicating that it induces the intrinsic mitochondrial apoptotic pathway. In vivo, expression of activated β-catenin in HPCs is associated with down-regulation of Bcl2 and expression of Casp3. Bone marrow transplantation assays reveal that enhanced cell survival by a Bcl2 transgene re-establishes the reconstitution capacity of HSCs/HPCs that express activated β-catenin. In addition, a Bcl2 transgene prevents exhaustion of these HSCs/HPCs in vivo. Our data suggest that activation of the Wnt/β-catenin pathway contributes to the defective function of HPCs in part by deregulating their survival.
Introduction
The canonical Wnt/β-catenin pathway plays an important regulatory role in hematopoiesis; however, its involvement in regulation of HSC3 function is controversial. Mice with deletion of β-catenin (1) or β-catenin and γ-catenin (2) had no obvious HSC defects. In contrast, other investigators showed the existence of hematopoietic abnormalities in mice with deletion of β-catenin from fetal life (3). Retroviral expression of activated β-catenin enhances the self-renewal of Bcl2 transgenic HSCs in vitro, and the Wnt signaling inhibitor Axin represses HSC proliferation and decreases reconstitution capacity of HSCs (4). However, others reported that purified Wnt protein in vitro (5) or inhibition of GSK3β in vivo (6) promotes the self-renewal of HSCs. Baba et al. (7, 8) showed that constitutive activation of β-catenin impaired multilineage differentiation, whereas others found that expression of constitutively active β-catenin in hematopoietic cells in vivo leads to loss of HSC repopulation and multilineage differentiation block (9, 10).
The role of β-catenin in the regulation of cell proliferation and apoptosis is cell context-dependent. β-Catenin is an enhancer of proliferation and survival in tumor cells (11, 12) but can also induce apoptosis in a variety of cells. Forced expression of an activated of β-catenin lacking the NH2 terminus promoted proliferation and apoptosis in mouse intestinal epithelial cells (13). However, in fibroblasts, the overexpression of β-catenin induced apoptosis independently of its transactivation function with LEF1 (14). In enzastaurin-treated multiple myeloma cells, it resulted in cell death-dependent up-regulation of JUN and TP73 (15).
Taken together, these contradictory reports emphasize the difficulty of understanding the role of the Wnt/β-catenin pathway in HSCs/HPCs. Here, we have revisited this controversy and have examined the effects of activated β-catenin in hematopoietic progenitor cells in vitro and in vivo. In vitro our studies reveal that activation of the Wnt/β-catenin signaling induces a mitochondria-dependent apoptotic pathway. In vivo we find that it promotes apoptosis of HPCs by suppressing Bcl2 and inducing Casp3. In addition, we show that an enhanced survival signal is required for maintenance and function of HSC/HPCs with activated β-catenin in vivo. Our studies suggest that the negative effect of Wnt/β-catenin signaling in HPCs results from the deregulation of their survival.
MATERIALS AND METHODS
Mice
C57BL/6 and B6.SJL mice were purchased from Taconic (Hudson, NY) for breeding in the mouse facility at the University of Illinois at Chicago. The mice (6–12 weeks) were used for bone marrow (BM) collection. All animal protocols were approved by the Animal Care and Use Committee of the University of Illinois at Chicago. Vav-Bcl2 transgenic mice were kindly provided by Dr. Jerry Adams (The Walter and Eliza Hall Institute of Medical Research, Bundoora, Victoria Australia). β-Cateninflox/flox mice were purchased from The Jackson Laboratory (Bar Harbor, ME). Ctnnb1(Ex3)flox/flox mice have been described (16) and were obtained from Dr. Kathleen Goss (University of Chicago). Effective deletion of floxed exons was monitored by PCR. The sequence of the primers is listed in supplemental Table S1.
Quantitative Real-time PCR
To evaluate the expression of candidate genes deregulated in mutant β-catenin mice, HPCs were sorted from Mx1-cre Ctnnb1(Ex3)Δ/+ and control mice (n = 3) 4 days after three doses of pI-pC injection. RNA was isolated with TRIzol reagent (Invitrogen) and quantified. Relative abundance of specific candidate gene transcripts normalized to β-actin was determined by quantitative RT-PCR by SYBR Green incorporation. The primer sequence for each gene is listed in supplemental Table S1.
Retroviral Vectors and BM Infection
The pBabe-β-cat-S33Y vector encoding an activated form of β-catenin was kindly provided by Dr. Eric Fearon (University of Michigan School of Medicine, Ann Arbor, MI). The BamHI-EcoRI DNA fragment encoding β-cat-S33Y was cloned into the MSCV-IRES-EGFP (MIGR1) vector. The Bcl2 cDNA was subcloned into MSCV-EYFP vector. Using Effectene transfection reagent (Qiagen), high titer retrovirus was produced by transient transfection of 293T cells with the pCL ecotropic packaging plasmid and the appropriate retroviral vectors (17). The MSCV-Cre-IRES-EYFP and MSCV-puro-Cre vectors were provided by Dr. Jiwang Zhang (Loyola University, Maywood, IL). The BM cells isolated from mice treated with 5-fluorouracil (150 mg/Kg) for 4 days, the lineage-negative BM cells, and the day 14.5 fetal liver cells were infected with the appropriate retrovirus by spinoculation as described previously (18).
Cell Culture
BM cells were collected by flushing murine femurs and tibias with IMDM containing 2% fetal bovine serum (FBS). To isolate lineage-negative BM cells, the cells were stained with biotinylated mouse antibodies specific for lineage markers (CD3, B220, IgM, Ter119, Gr-1, and CD19). The Lin+ cells were depleted with streptavidin microbeads and LS columns (Miltenyi Biotec, Auburn, CA) following the manufacturer's instructions. The BM cells were cultured in IMDM, supplemented with 10% FBS, penicillin/streptomycin (10,000 units/ml), β-mercaptoethanol (50 μm), 10 ng/ml murine IL-3 and IL-6, and 100 ng/ml murine stem cell factor (eBioscience, San Diego CA). Fetal liver cells were isolated at embryonic day 14.5 and grown in 50% IMDM and DMEM containing 20% FBS, 10 ng/ml murine IL-3 and IL-6, and 100 ng/ml murine stem cell factor.
Colony-forming Unit Assays
Isolated BM progenitor cells were infected with MIGR1 or β-catenin retrovirus. The EGFP-positive cells, isolated by flow cytometry, were plated in duplicate in methylcellulose medium (MethoCult M3434, StemCell Technologies) supplemented with IL-3, IL-6, erythropoietin and stem cell factor. The number of colonies was scored 10 days later.
Flow Cytometric Analysis
We used single-cell suspensions of BM and PB. Flow cytometric analysis of HSCs/HPCs has been described previously (19). Briefly, the infected BM cells were stained with biotinylated antibodies specific for lineage markers (CD3, B220, IgM, Ter119, Gr-1, Thy1.1, Il-7R, and CD19) followed by PE-Cy5-conjugated streptavidin, Kit-APC-eFluo780, Sca-PE, CD48-PE-Cy7, and CD150-APC staining. Flow cytometry was performed at the University of Illinois at Chicago facility using FACSCalibur or CyAn flow cytometers. Cell cycle analysis with DAPI staining was performed as described previously (20). Cell cycle analysis with Hoechst 3342 was performed as described (4). For detection of apoptosis, BM cells were cultured overnight at a density of 0.2 × 106/ml. The next day, the cells were stained with annexin V (BD Biosciences) and 7-AAD or DAPI following the manufacturer's instructions. Caspase 3 and caspase 9 staining was performed using kits from BD Biosciences and BioVision according to the manufacturer's instructions. All data were analyzed by the FlowJo software (TreeStar).
Transplantation Assays
BM cells and day 14.5 fetal liver cells from C57BL/6 or BM cells from Vav-Bcl2-transgenic mice were infected with MIGR1 or β-catenin retrovirus and were transplanted into lethally irradiated (960 radians) B6.SJL mice by retro-orbital injection. After transplantation, the mice were closely monitored. FACS analysis of EGFP-positive cells in PB was performed every month after transplantation.
Mitochondrial Membrane Potential (ΔΨm) Measurement
Tetramethylrhodamine methyl ester perchlorate (TMRM) (Sigma) was used as an indicator of ΔΨm. Live BM cells were washed once with PBS and stained with freshly prepared 10 μm TMRM for 30 min at 37 °C. The cells were then washed twice with PBS and suspended in PBS for flow cytometric analysis.
Western Blot Analysis
Cell extracts were prepared using lysis buffer containing 1% Triton X-100, 2 mm EDTA, 100 mm NaCl, 10% glycerol, 50 mm Tris-HCL, and a protease inhibitor mixture (Sigma). Mouse anti-β-catenin (BD Transduction Laboratories) and mouse anti-tubulin (Millipore, Billerica, MA) were used for Western blot analysis, which was performed as described previously (21).
Luciferase Reporter Assay
The luciferase assay was performed with the Dual-GloTM luciferase assay system (Promega).
Statistical Analysis
Statistical significance was calculated using the two-tailed Student's t test (Excel, Microsoft, Redmond, WA).
RESULTS
Activation of β-Catenin Induces Apoptosis in Vivo
To determine whether in vivo activation of β-catenin induces apoptosis of HSCs/HPCs, we used the Cre-loxP system to activate β-catenin in hematopoietic stem/progenitor cells in vivo. We generated Mx1-cre+Ctnnb1(Ex3)fl/+ mice by crossing Ctnnb1(Ex3)fl/fl mice with transgenic mice expressing cre recombinase under control of the type I interferon-inducible Mx1 promoter. In Mx1-cre+Ctnnb1(Ex3)fl/+ mice, the exon 3 of Ctnnb1 is flanked by loxP sites. Induction of exon 3 deletion by injection of interferon-α inducer pI-pC leads to expression of activated β-catenin. By PCR analysis, Mx1-cre+Ctnnb1(Ex3)fl/+ mice treated with pI-pC had Cre-mediated deletion of exon 3 of Ctnnb1 in the majority of BM cells (Fig. 1A). After 4 days of treatment with three doses of pI-pC every other day, we characterized the HPCs and Lin−c-Kit+Sca+ cells (LSKs, stem-enriched population) in these mice. The frequency (Fig. 1B, left panel) as well as the total number (Fig. 1B, right panel) of HPCs and LSKs were decreased 3–10-fold in BM cells from Mx1-cre Ctnnb1(Ex3)Δ/+ when compared with control mice. The LSKs and HPCs expressing activated β-catenin displayed a 3–4-fold increase in apoptosis when compared with control cells (Fig. 1C). Our data suggest that in vivo activation of Wnt/β-catenin by stabilized β-catenin induces apoptosis in HPCs and LSKs, leading to the depletion of these cells.
FIGURE 1.
Expression of activated β-catenin in vivo leads to a significant increase in the frequency of apoptosis of HPCs and LSKs. A, semiquantitative PCR analysis of genomic DNA confirms excision of exon 3 (right lane) in BM cells isolated from Mx1-cre+Ctnnb1(Ex3)fl/+ and Mx1-cre−Ctnnb1(Ex3)fl/+mice after pI-pC induction. B, marked decrease in the number of HPCs and LSKs in cells expressing activated β-catenin when compared with the control BM cells. The left panel shows the histograms of the flow cytometric analysis of HPCs and LSKs. The right panel shows the total number of HPCs and LSKs in BM from the pI-pC-treated mice (mean ± S.D.; n = 6; **, p < 0.01). C, activated β-catenin promotes apoptosis of HPCs and LSKs. The frequency of apoptosis in gated LSKs and HPCs stained with annexin V and DAPI from Mx1-cre+Ctnnb1(Ex3)fl/+ (left panels) and Mx1-cre−Ctnnb1(Ex3)fl/+ (right panels) mice 4 days after Cre induction (mean ± S.D.; n = 3; **, p < 0.01) was determined. D, gene expression by quantitative RT-PCR of RNA isolated from the sorted HPCs from Mx1-cre+Ctnnb1(Ex3)fl/+ and Mx1-cre−Ctnnb1(Ex3)fl/+mice 4 days after induction. Gene expression was normalized to β-actin as internal control. Values represent the -fold changes in gene expression relative to that in HPCs from control mice (mean ± S.D.; n = 3; *, p < 0.05). All mice used for studies were 5–8 weeks old.
To gain insight into the molecular mechanism underlying the increased apoptosis of HPCs that express the mutant β-catenin, we examined several genes regulating the apoptotic and survival pathways. HPC cells were isolated from the Mx1-cre Ctnnb1(Ex3)Δ/+ and control mice 4 days after three doses of pI-pC injection. In sorted Ctnnb1-mutant HPCs, Bcl2 is down-regulated (>3-fold, Fig. 1D, black bars) whereas Casp3 is up-regulated (>3-fold, Fig. 1D, black bars) when compared with HPCs of control mice (Fig. 1D, gray bars). However, the expression of other genes involved in cell apoptosis and survival was not significantly altered.
Activation of Wnt/β-Catenin Induces Apoptosis in Hematopoietic Progenitors in Vitro
To determine whether these effects are also seen in primary BM cells expressing the active form of β-catenin (β-cat-S33Y) in vitro, we cloned β-cat-S33Y in the retroviral vector MIGR1 and expressed the protein in primary BM cells (supplemental Fig. S1A). As a control for endogenous expression of the protein at a physiological level, we used Apc−/− BM cells. Apc is an essential component of β-catenin destruction complex, and its loss leads to retention of the protein in the cytoplasm (11). Western blot analyses showed no significant difference between β-cat-S33Y-transduced Lin− cells and Apc−/− Lin− cells, indicating that the retroviral expression of β-cat is within a normal range (supplemental Fig. S1B). To confirm that β-cat-S33Y activates the Wnt/β-catenin signaling, we performed luciferase reporter assays. The Wnt signaling was stimulated by this mutant β-catenin (supplemental Fig. S1C). In addition, we found that in β-cat-S33Y-EGFP+ HPCs (Lin−Kit+) or Lin− cells, the frequency of early apoptotic cells (Fig. 2A) and the level of caspase 3 (Fig. 2B) were increased significantly when compared with MIGR1-transduced HPCs and Lin− cells.
FIGURE 2.
Expression of β-cat-S33Y in BM cells induces apoptosis in vitro. A, the fraction of apoptotic Lin−c-Kit+ cells expressing MIGR1 (left panel) or MIGR1-β-cat-S33Y (right panel) was determined by FACS. The frequency of early apoptotic cells (annexin V+DAPI− or annexin V+7-AAD−) is shown (three independent experiments; mean ± S.D.; *, p < 0.05; **, p < 0.01). B, the activity of caspase 3 in primary progenitor-enriched cells (Lin−) expressing MIGR1 (left panel) or β-cat-S33Y (right panel). The percentage of cells with active caspase 3 is shown (three independent experiments, mean ± S.D.; *, p < 0.05). All analyses were performed on a gated EGFP+ cell population.
The Wnt/β-catenin pathway can be activated by the addition of LiCl, which inhibits GSK-3β, a component of the degradation complex, thereby stabilizing β-catenin. In methylcellulose-based medium, treatment of BM cells with LiCl, but not with equimolar amounts of NaCl, repressed colony formation (supplemental Fig. S2A), and the number of early apoptotic BM cells was significantly increased (supplemental Fig. S2B). Similarly, in liquid culture, the early apoptotic Lin−Kit+ progenitor cells (Fig. 3A) and progenitor cells with an elevated caspase 3 activity (Fig. 3B) were increased significantly after LiCl treatment when compared with NaCl treatment. The frequency of apoptosis was increased more significantly at high concentration of LiCl than at low concentration of LiCl, suggesting that LiCl-induced apoptosis is dosage-dependent (Fig. 3A).
FIGURE 3.
β-Catenin mediates the induction of HPC apoptosis after LiCl treatment. A, early apoptotic cells (annexin V+DAPI−) of Lin−c-Kit+ BM cells treated with increasing concentration of NaCl or LiCl (three independent experiments; mean ± S.D.; *, p < 0.05) for 24 h. Black bars indicate cells treated with NaCl and used as controls; gray bars indicate cells treated with LiCl. B, Lin− BM cells were treated with 15 mm NaCl or 15 mm LiCl for 24 h. The histogram depicts the mean percentage of cells with active caspase 3 (three independent experiments, mean ± S.D.) **, p < 0.01. C, the frequency of early apoptotic Lin− Kit+ BM cells isolated from β-cateninfl/fl mice with expression of MIGR1 (gray bars) or EYFP-Cre to excise β-catenin (black bars). The BM cells were treated with 10 mm LiCl for 24 h (three independent experiments; mean ± S.D.; *, p < 0.05).
Indomethacin is a Cox inhibitor that blocks the Wnt signaling pathway by repressing Ctnnb (22). To confirm that the effects of LiCl are dependent on β-catenin, we treated HPCs with LiCl alone or with indomethacin. The results showed that LiCl-induced apoptosis is reduced by indomethacin (supplemental Fig. S3). Next, we isolated BM cells from β-cateninfl/fl mice in which exons 2–6 can be excised by Cre expression (23) and confirmed the deletion by PCR (supplemental Fig. S4). After treatment with LiCl, apoptosis in β-cateninfl/fl HPCs was comparable with that of control HPCs (Fig. 3C), suggesting that LiCl specifically affects β-catenin and that the activation of Wnt/β-catenin either by expression of active β-catenin or by LiCl leads to apoptosis in HPCs.
Wnt/β-Catenin Signaling Induces Apoptosis in Vitro through the Intrinsic Mitochondrial Apoptotic Pathway, and Apoptosis Is Inhibited by Bcl2
There are two major apoptotic pathways, the mitochondrial “intrinsic” and transmembrane “extrinsic” pathway. To determine whether the mitochondrial apoptotic pathway is activated by Wnt signaling, we examined the activity of caspase 9 in HPCs that expressed β-cat-S33Y or that were treated with LiCl. We found that caspase 9 was activated in β-catenin-positive HPCs (Fig. 4A, black bar) and that LiCl treatment resulted in a higher activity of caspase 9 than what was observed in the control HPCs (supplemental Fig. S5). The proper function of the mitochondria was tested with TMRM, which detects the mitochondrial membrane potential (ΔΨm). HPCs treated with LiCl and TMRM showed a significant drop in fluorescence with increasing concentration of LiCl (Fig. 4B, black bars), indicating that a loss of ΔΨm occurred in the LiCl-treated HPCs. To determine whether expression of Bcl2 protects HPCs from apoptosis, Lin− BM cells isolated from Mx1-cre−Ctnnb1(Ex3)fl/fl mice were infected with MSCV-puro and MSCV-EYFP, MSCV-puro-Cre and MSCV-EYFP, or MSCV-puro-Cre and MSCV-EYFP-Bcl2. The excision of exon 3 of Ctnnb1 in Mx1-cre−Ctnnb1(Ex3)fl/fl BM cells was induced by Cre recombinase expressed in a retroviral vector. As we expected, Lin− cells expressing activated β-catenin (Fig. 4C, middle bar) displayed a frequency of apoptosis higher than the control cells in vitro. However, Bcl2 could repress apoptosis (Fig. 4C, right bar). To confirm the role of Bcl2 in blocking apoptosis induced by activated β-catenin, Lin− BM cells, isolated from Vav-Bcl2 mice, were infected with the β-cat-S33Y or the MIGR1 retrovirus. Flow cytometric analysis of the cells revealed that the frequency of annexin V+DAPI− cells and the activity of caspase 3 were slightly lower in β-cat-S33Y-positive Lin−Kit+ BM cells than in MIGR1-transduced Lin−Kit+ BM cells (Fig. 4, D and E) and that LiCl treatment did not affect apoptosis in HPCs that expressed the Bcl2 transgene (Fig. 4F). Together, our data suggest that Bcl2 can reduce apoptosis induced by activated β-catenin.
FIGURE 4.
Activation of the Wnt/β-catenin pathway initiates the mitochondria-dependent apoptotic program, which can be inhibited by Bcl2. A, the histogram shows the quantification of active caspase 9 in control cells (gray bar) and in cells expressing β-cat-S33Y (black bar). The Lin− kit+ BM cells were infected with MIGR1 or β-cat-S33Y retrovirus. The analysis was performed on gated EGFP+ cells. The results are from three independent experiments (mean ± S.D.; **, p < 0.01). B, the histogram depicts the mean percentage of TMRM-positive cells in three independent experiments (mean ± S.D.; **, p < 0.01). The gated Lin−Il-7R−-kit+ BM cells treated with NaCl and LiCl for 18 h and stained with TMRM were analyzed by flow cytometry. C, flow cytometric analysis of the early apoptotic cells (annexin V+7-AAD−) in Mx1-cre−Ctnnb1(Ex3)fl/fl Lin− BM cells, co-infected with MSCV-puro and MSCV-EYFP (left bar), with MSCV-puro-Cre and MSCV-EYFP (middle bar), or with MSCV-puro-Cre and MSCV-EYFP-Bcl2 (right bar). The EGFP+ cells were analyzed after 7 days of selection in medium containing 1 μg/ml puromycin (mean ± S.D.; *, p < 0.05). D and E, flow cytometric analysis of early apoptotic cells (D) and of cells with active caspase 3 (E) isolated from Bcl2 transgenic BM cells and infected with MIGR1 or β-cat-S33Y retrovirus. Gated Lin− Kit+ GFP+ cells were analyzed, and the mean of three independent experiments is shown. *, p < 0.05. F, flow cytometric analysis of early apoptotic cells (annexin V+7-AAD−) in Bcl2 transgenic BM cells treated with NaCl (left panel) or LiCl (right panel). Gated Lin− Kit+ GFP+ cells were analyzed. The experiments were performed in triplicate and repeated three times.
A Bcl2 Transgene Rescues the Defects of HSCs/HPCs Expressing β-cat-S33Y
We hypothesized that apoptosis induced by activated β-catenin contributes to the defective function of HSCs/HPCs with active Wnt/β-catenin signaling. If this is the case, then Bcl2 expression should rescue the function of these HSCs/HPCs. Therefore, we isolated CD45.2+ BM cells from C57BL/6 donors 4 days after treatment with 5-fluorouracil; cells were infected with β-cat-S33Y or the empty MIGR1 retrovirus and transplanted into lethally irradiated syngeneic mice (CD45.1+). More than 90% of the PB cells from the recipients were donor-derived (CD45.2+), indicating a successful reconstitution (data not shown). Five weeks after transplantation, the percentage of EGFP+ cells in total BM cells was determined by flow cytometry. Before transplantation (in vitro) and 5 weeks after transplantation, in the control mice, the percentage of MIGR1-transduced BM cells was comparable (Fig. 5A, gray bars). In contrast, before transplantation, the percentage of β-cat-S33Y-EGFP-positive cells was about 14%, but the cells were hardly detected in the recipient mice at 5 weeks after transplantation (Fig. 5A, black bars). To confirm these results, we infected day 14.5 fetal liver cells with β-cat-S33Y and MIGR1 retroviruses followed by transplantation. The percentage of MIGR1-transduced fetal liver cells was comparable before or after transplantation, but again, although the percentage of β-cat-S33Y-EGFP-transduced fetal liver cells was about 27% before transplantation, no EGFP+ cells were detected in the PB of the recipient mice at 5 and 14 weeks after transplantation (Fig. 5B, black bars). These data suggest that retroviral expression of the active form of β-catenin disrupts the reconstitution capacity of HSCs/HPCs. To determine whether Bcl2 rescues the defects in the function of HSCs/HPCs expressing activated β-catenin, we isolated BM cells from Vav-Bcl2 transgenic mice (24), infected them with the MIGR1 vector expressing either β-cat-S33Y-EGFP or EGFP, and transplanted the cells into lethally irradiated syngeneic recipients. As expected, the percentage of control cells at time of transplantation and at 7–8 weeks after transplantation did not decrease (Fig. 5C, gray bars). However, in contrast to the previous results, the percentage of β-cat-S33Y-Bcl2-EGFP+ cells was also comparable in vitro before transplantation and in total PB and BM cells at 7–8 weeks after transplantation (Fig. 5C, black bars). We characterized the GFP-positive HSC and HPC compartments in both groups of recipients. The number of LSK cells and CD48−CD150+ LSK cells was slightly lower in the GFP+ BM cells of Bcl2 transgenic/β-cat-S33Y mice than in the control animals (Fig. 6A), suggesting that the expression of Bcl2 partially rescued the reconstitution capacity of short-term HSCs and HPCs expressing activated β-catenin. Because others reported that in vivo expression of β-cat-S33Y leads to BM failure within 2–3 weeks (9), we monitored a cohort of chimeric mice expressing both activated β-catenin and Bcl2 transgene. The EGFP+ cells in PB were stabilized at 6 months and were almost doubled at 10 months after transplantation (Fig. 6B), indicating that the expression of Bcl2 rescues and may enhance the repopulating capacity of long-term HSCs expressing activated β-catenin. Of note, the apoptosis of Bcl2 transgenic LSKs expressing activated β-catenin was slightly higher than the LSKs expressing control vector, whereas the Bcl2 transgenic HPCs expressing activated β-catenin or control vector displayed a comparable frequency of apoptosis (Fig. 6C). These data suggest that Bcl2 can repair the hematopoietic defects of HSCs/HPCs that express activated β-catenin.
FIGURE 5.
Expression of β-cat-S33Y disrupts the reconstitution capacity of HSCs/HPCs, which can be rescued by Bcl2 transgene. A, before transplantation (day 0), the fraction of β-cat-S33Y-EGFP+ BM cells (black bar) was ∼14%. Five weeks later, β-cat-S33Y-EGFP+-positive cells were virtually undetectable (mean ± S.D.; n = 5). Control cells (gray bars) did not change significantly. B, the percentage of β-cat-S33Y-EGFP+ (black bars) and of control fetal liver (FL) cells (gray bars) before transplantation (Day 0) and at 5 and 14 weeks after transplantation indicates the inability of the cells to be maintained in vivo (mean ± S.D.; n = 5). C, the fraction of EGFP+, Bcl2-transgenic/β-cat-S33Y (black bars), or control cells (gray bars) detected by FACS at time of transplantation (day 0) or at 7 weeks (PB) or 8 weeks (BM) after transplantation (mean ± S.D.; n = 5). Similar results were obtained in two independent experiments.
FIGURE 6.
Bcl2 transgene rescues the defects induced by β-catenin in HSCs/HPCs in vivo. A, the top and bottom panels show representative flow cytometric analyses of LSK and long-term HSCs (LT-HSCs) isolated from BM of the chimeric mice reconstituted with Bcl2 transgenic BM cells expressing EGFP (top) or β-cat-S33Y/EGFP (bottom). On the right side, the bars compare the total number of LSKs (top) or long-term HSCs (bottom) in 105 EGFP+ BM cells (mean ± S.D.; n = 5). *, p < 0.05; ** p < 0.01. LSK are defined as Lin−Sca-1+Kit+; long-term HSCs are defined as LSK CD150+CD48−. B, the histogram shows the percentage of EGFP+ cells in the PB of chimeric mice reconstituted with Bcl2 transgenic BM cells expressing β-cat-S33Y/EGFP at 2–6 and 10 months (mean ± S.D.; n = 3–5). *, p < 0.05. C, the histogram indicates the frequency of early apoptotic cells in LSK (Lin−Kit+Sca+) cells and in HPCs (Lin−Kit+Sca−) in BM of the chimeric mice reconstituted with Bcl2 transgenic BM cells expressing EGFP or β-cat-S33Y/EGFP. The analysis was performed on gated EGFP+ cells. The mice were analyzed 2 months after transplantation (mean ± S.D., n = 4). *, p < 0.05.
DISCUSSION
Whether β-catenin acts as a proapoptotic factor in hematopoietic cells has not been conclusively defined. Previous studies revealed that expression of activated β-catenin did not alter the frequency of apoptosis in HSC population (9, 10). However, Li and colleagues (25) showed that activated β-catenin enhances apoptosis of HSCs in vitro. The goal of this study was to help to solve, if possible, the existing discrepancies on β-catenin in HSCs/HPCs. Our results include strong data, obtained in vitro and in vivo, which support the apoptotic effect that this protein exerts on HPCs. In addition, our results show the molecular mechanisms by which this occurs and indicate that regulation of survival by β-catenin is critical for the maintenance of functional HSCs/HPCs.
By using an Mx1-cre/loxP system to induce the deletion of exon 3 of Ctnnb1 gene, leading to activated β-catenin in vivo, we showed that a significant decrease in the number of HPCs and LSKs and a markedly increased frequency of apoptosis occur in these cells. Consistently, Cre-dependent activation of β-catenin in Ctnnb1(Ex3)fl/fl HPCs enhances apoptosis in vitro. The result was confirmed by retroviral expression of activated β-cat-S33Y in HPCs. However, the frequency of apoptosis is induced by activated β-catenin more significantly in vivo than it is in vitro. In our opinion, it is likely that the in vivo microenvironment could contribute to the survival of HPCs. Bcl2 is a survival factor, and ectopic expression of Bcl2 prevents apoptosis in HSCs (26). Caspase 3 plays a central role in the execution of cell apoptosis (27). Down-regulation of Bcl2 and induced expression of Casp3 occur in Ctnnb1 mutant HPCs in vivo. The expression of p53 is not altered in the mutant HPCs (Fig. 1D). Thus, increased apoptosis in Ctnnb1 mutant HPCs may be a consequence of the combined deregulation of Bcl2, Casp3, and perhaps other genes involved in apoptosis and survival of HPCs, which are still unidentified and which could be regulated directly or indirectly by β-catenin. Our in vitro study suggests that activation of Wnt/β-catenin signaling activates caspase 3 and caspase 9 and reduces the mitochondrial membrane potential, leading to a mitochondria-mediated apoptosis. We previously showed that loss of Apc induces apoptosis in HSCs/HPCs (19). Apc is a multifunction, cell context-dependent protein (28), and it is still unclear whether β-catenin is a critical function mediator of Apc. Our data suggest that β-catenin is likely to mediate, at least in part, the function of Apc in regulating the survival of HPCs.
Although previous studies suggested that the retroviral expression of active β-catenin in Bcl2 transgenic HSCs enhanced their self-renewal, there was no indication on the potential pathways utilized by β-cat-S33Y (4). It was therefore unclear whether the effects of β-catenin on self-renewal depended on Bcl2 expression, and the mechanism by which Bcl2 cooperates with β-catenin in maintenance of HSCs/HPCs remained not well defined. In this study, we show that retroviral expression of active β-catenin disrupts the reconstitution capacity of HSCs/HPCs but that Bcl2 transgenic HSCs/HPCs expressing activated β-catenin maintain their reconstitution capacity. We further show that a Bcl2 transgene inhibits the apoptosis induced by activated β-catenin in vitro and prevents the exhaustion of HSCs/HPCs in vivo. We believe that taken together, our studies clarify the existing contradictory data on the effects of activated β-catenin in HSCs/HPCs not only by supporting the notion that activation of Wnt/β-catenin negatively impacts the function of HSCs/HPCs, but more importantly, by strongly indicating that an enhanced survival signal is required for the survival of these cells. It is very likely that regulation of the function of HSCs by Wnt/β-catenin is dosage-dependent (29). Our study suggests that the negative impact of Wnt/β-catenin in HSCs/HPCs is due, at least in part, to an increased apoptosis induced by high dosage of Wnt signaling.
Finally, the activation of Wnt/β-catenin signaling and the overexpression of Bcl2 have been frequently associated with acute myeloid leukemia (30–34). Our data suggest that Wnt/β-catenin may cooperate with survival signaling in promoting the self-renewal or proliferation of leukemia stem/progenitor cells in some acute myelogenous leukemia patients.
Supplementary Material
This work was supported, in whole or in part, by National Institutes of Health Grant CA140979 (to Z. Q.). This work was also supported in part by the American Cancer Society Illinois Division (to Z. Q.).

This article contains supplemental Table 1 and Figs. S1–S5.
- HSC
- hematopoietic stem cell
- HPC
- hematopoietic progenitor cell
- BM
- bone marrow
- 7-AAD
- 7-aminoactinomycin D
- IMDM
- Iscove's modified Dulbecco's medium
- β-cat
- β-catenin
- TMRM
- tetramethylrhodamine methyl ester perchlorate
- pI-pC
- polyinosinic-polycytidylic acid
- PE
- phycoerythrin
- EGFP
- enhanced green fluorescent protein
- LSK
- Lin−c-Kit+Sca+
- PB
- peripheral blood
- Apc
- adenomatous polyposis coli.
REFERENCES
- 1. Cobas M., Wilson A., Ernst B., Mancini S. J., MacDonald H. R., Kemler R., Radtke F. (2004) β-Catenin is dispensable for hematopoiesis and lymphopoiesis. J. Exp. Med. 199, 221–229 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2. Jeannet G., Scheller M., Scarpellino L., Duboux S., Gardiol N., Back J., Kuttler F., Malanchi I., Birchmeier W., Leutz A., Huelsken J., Held W. (2008) Long-term, multilineage hematopoiesis occurs in the combined absence of β-catenin and γ-catenin. Blood 111, 142–149 [DOI] [PubMed] [Google Scholar]
- 3. Zhao C., Blum J., Chen A., Kwon H. Y., Jung S. H., Cook J. M., Lagoo A., Reya T. (2007) Loss of β-catenin impairs the renewal of normal and CML stem cells in vivo. Cancer Cell 12, 528–541 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Reya T., Duncan A. W., Ailles L., Domen J., Scherer D. C., Willert K., Hintz L., Nusse R., Weissman I. L. (2003) A role for Wnt signaling in self-renewal of hematopoietic stem cells. Nature 423, 409–414 [DOI] [PubMed] [Google Scholar]
- 5. Willert K., Brown J. D., Danenberg E., Duncan A. W., Weissman I. L., Reya T., Yates J. R., 3rd, Nusse R. (2003) Wnt proteins are lipid-modified and can act as stem cell growth factors. Nature 423, 448–452 [DOI] [PubMed] [Google Scholar]
- 6. Trowbridge J. J., Xenocostas A., Moon R. T., Bhatia M. (2006) Glycogen synthase kinase-3 is an in vivo regulator of hematopoietic stem cell repopulation. Nat. Med. 12, 89–98 [DOI] [PubMed] [Google Scholar]
- 7. Baba Y., Garrett K. P., Kincade P. W. (2005) Constitutively active β-catenin confers multilineage differentiation potential on lymphoid and myeloid progenitors. Immunity 23, 599–609 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Baba Y., Yokota T., Spits H., Garrett K. P., Hayashi S., Kincade P. W. (2006) Constitutively active β-catenin promotes expansion of multipotent hematopoietic progenitors in culture. J. Immunol. 177, 2294–2303 [DOI] [PubMed] [Google Scholar]
- 9. Kirstetter P., Anderson K., Porse B. T., Jacobsen S. E., Nerlov C. (2006) Activation of the canonical Wnt pathway leads to loss of hematopoietic stem cell repopulation and multilineage differentiation block. Nat. Immunol. 7, 1048–1056 [DOI] [PubMed] [Google Scholar]
- 10. Scheller M., Huelsken J., Rosenbauer F., Taketo M. M., Birchmeier W., Tenen D. G., Leutz A. (2006) Hematopoietic stem cell and multilineage defects generated by constitutive β-catenin activation. Nat. Immunol. 7, 1037–1047 [DOI] [PubMed] [Google Scholar]
- 11. He T. C., Sparks A. B., Rago C., Hermeking H., Zawel L., da Costa L. T., Morin P. J., Vogelstein B., Kinzler K. W. (1998) Identification of c-MYC as a target of the APC pathway. Science 281, 1509–1512 [DOI] [PubMed] [Google Scholar]
- 12. Tetsu O., McCormick F. (1999) β-Catenin regulates expression of cyclin D1 in colon carcinoma cells. Nature 398, 422–426 [DOI] [PubMed] [Google Scholar]
- 13. Wong M. H., Rubinfeld B., Gordon J. I. (1998) Effects of forced expression of an NH2-terminal truncated β-catenin on mouse intestinal epithelial homeostasis. J. Cell Biol. 141, 765–777 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Kim K., Pang K. M., Evans M., Hay E. D. (2000) Overexpression of β-catenin induces apoptosis independent of its transactivation function with LEF-1 or the involvement of major G1 cell cycle regulators. Mol. Biol. Cell 11, 3509–3523 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Raab M. S., Breitkreutz I., Tonon G., Zhang J., Hayden P. J., Nguyen T., Fruehauf J. H., Lin B. K., Chauhan D., Hideshima T., Munshi N. C., Anderson K. C., Podar K. (2009) Targeting PKC: a novel role for β-catenin in ER stress and apoptotic signaling. Blood 113, 1513–1521 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Harada N., Tamai Y., Ishikawa T., Sauer B., Takaku K., Oshima M., Taketo M. M. (1999) Intestinal polyposis in mice with a dominant stable mutation of the β-catenin gene. EMBO J. 18, 5931–5942 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Qian Z., Mao L., Fernald A. A., Yu H., Luo R., Jiang Y., Anastasi J., Valk P. J., Delwel R., Le Beau M. M. (2009) Enhanced expression of FHL2 leads to abnormal myelopoiesis in vivo. Leukemia 23, 1650–1657 [DOI] [PubMed] [Google Scholar]
- 18. Lavau C., Szilvassy S. J., Slany R., Cleary M. L. (1997) Immortalization and leukemic transformation of a myelomonocytic precursor by retrovirally transduced HRX-ENL. EMBO J. 16, 4226–4237 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Qian Z., Chen L., Fernald A. A., Williams B. O., Le Beau M. M. (2008) A critical role for Apc in hematopoietic stem and progenitor cell survival. J. Exp. Med. 205, 2163–2175 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Zhang J., Grindley J. C., Yin T., Jayasinghe S., He X. C., Ross J. T., Haug J. S., Rupp D., Porter-Westpfahl K. S., Wiedemann L. M., Wu H., Li L. (2006) PTEN maintains hematopoietic stem cells and acts in lineage choice and leukemia prevention. Nature 441, 518–522 [DOI] [PubMed] [Google Scholar]
- 21. Qian Z., Okuhara D., Abe M. K., Rosner M. R. (1999) Molecular cloning and characterization of a mitogen-activated protein kinase-associated intracellular chloride channel. J. Biol. Chem. 274, 1621–1627 [DOI] [PubMed] [Google Scholar]
- 22. Goessling W., North T. E., Loewer S., Lord A. M., Lee S., Stoick-Cooper C. L., Weidinger G., Puder M., Daley G. Q., Moon R. T., Zon L. I. (2009) Genetic interaction of PGE2 and Wnt signaling regulates developmental specification of stem cells and regeneration. Cell 136, 1136–1147 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Brault V., Moore R., Kutsch S., Ishibashi M., Rowitch D. H., McMahon A. P., Sommer L., Boussadia O., Kemler R. (2001) Inactivation of the β-catenin gene by Wnt1-Cre-mediated deletion results in dramatic brain malformation and failure of craniofacial development. Development 128, 1253–1264 [DOI] [PubMed] [Google Scholar]
- 24. Ogilvy S., Metcalf D., Print C. G., Bath M. L., Harris A. W., Adams J. M. (1999) Constitutive Bcl-2 expression throughout the hematopoietic compartment affects multiple lineages and enhances progenitor cell survival. Proc. Natl. Acad. Sci. U.S.A. 96, 14943–14948 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25. Perry J. M., He X. C., Sugimura R., Grindley J. C., Haug J. S., Ding S., Li L. (2011) Cooperation between both Wnt/β-catenin and PTEN/PI3K/Akt signaling promotes primitive hematopoietic stem cell self-renewal and expansion. Genes Dev. 25, 1928–1942 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26. Domen J., Weissman I. L. (2000) Hematopoietic stem cells need two signals to prevent apoptosis; BCL-2 can provide one of these, Kitl/c-Kit signaling the other. J. Exp. Med. 192, 1707–1718 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Porter A. G., Jänicke R. U. (1999) Emerging roles of caspase-3 in apoptosis. Cell Death Differ. 6, 99–104 [DOI] [PubMed] [Google Scholar]
- 28. Fodde R., Smits R., Clevers H. (2001) APC, signal transduction, and genetic instability in colorectal cancer. Nat. Rev. Cancer 1, 55–67 [DOI] [PubMed] [Google Scholar]
- 29. Luis T. C., Naber B. A., Roozen P. P., Brugman M. H., de Haas E. F., Ghazvini M., Fibbe W. E., van Dongen J. J., Fodde R., Staal F. J. (2011) Canonical Wnt signaling regulates hematopoiesis in a dosage-dependent fashion. Cell Stem Cell 9, 345–356 [DOI] [PubMed] [Google Scholar]
- 30. Chung E. J., Hwang S. G., Nguyen P., Lee S., Kim J. S., Kim J. W., Henkart P. A., Bottaro D. P., Soon L., Bonvini P., Lee S. J., Karp J. E., Oh H. J., Rubin J. S., Trepel J. B. (2002) Regulation of leukemic cell adhesion, proliferation, and survival by β-catenin. Blood 100, 982–990 [DOI] [PubMed] [Google Scholar]
- 31. Simon M., Grandage V. L., Linch D. C., Khwaja A. (2005) Constitutive activation of the Wnt/β-catenin signaling pathway in acute myeloid leukemia. Oncogene 24, 2410–2420 [DOI] [PubMed] [Google Scholar]
- 32. Tickenbrock L., Schwäble J., Wiedehage M., Steffen B., Sargin B., Choudhary C., Brandts C., Berdel W. E., Müller-Tidow C., Serve H. (2005) Flt3 tandem duplication mutations cooperate with Wnt signaling in leukemic signal transduction. Blood 105, 3699–3706 [DOI] [PubMed] [Google Scholar]
- 33. Ysebaert L., Chicanne G., Demur C., De Toni F., Prade-Houdellier N., Ruidavets J. B., Mansat-De Mas V., Rigal-Huguet F., Laurent G., Payrastre B., Manenti S., Racaud-Sultan C. (2006) Expression of β-catenin by acute myeloid leukemia cells predicts enhanced clonogenic capacities and poor prognosis. Leukemia 20, 1211–1216 [DOI] [PubMed] [Google Scholar]
- 34. Del Principe M. I., Del Poeta G., Venditti A., Buccisano F., Maurillo L., Mazzone C., Bruno A., Neri B., Irno Consalvo M., Lo Coco F., Amadori S. (2005) Apoptosis and immaturity in acute myeloid leukemia. Hematology 10, 25–34 [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.






