Acute BCR-ABL expression during in vitro hematopoietic development of embryonic stem (ES) cells causes expansion of multipotent and myeloid progenitors with a concomitant reduction in differentiation toward erythroblasts. Progenitor cell expansion is due to a rapid, cell autonomous, suppression of programmed cell death with an increase in expression of the antiapoptotic molecule BCL-XL. Other antiapoptotic effectors, including AKT, STAT5, and BCL-2 are not up-regulated by BCR-ABL in this system. In addition, the proapoptotic p38 mitogen–activated protein kinase (MAPK) pathway is suppressed by BCR-ABL expression in ES-derived hematopoietic progenitors. Inhibition of p38 MAPK by the small molecule inhibitor SB203580 expanded ES-derived hematopoietic progenitors by an antiapoptotic mechanism and is sufficient to expand ES-derived hematopoietic progenitors to levels approaching 80% of that seen following BCR-ABL expression. In the cellular context of ES-derived hematopoietic progenitors, BCR-ABL expression expands cells by suppressing programmed cell death with a set of antiapoptotic pathways distinct from those previously reported in continuous cell line studies.

Cancer develops from an accumulation of genetic aberrations.1 Particular genetic lesions may be essential for initiation and maintenance of particular cancers. Inducible transgenic models demonstrate that sustained activation of C-MYC is essential both for the establishment and continual growth of T-cell lymphomas, myeloid leukemias, and osteogenic sarcomas.2,3In human cancer cells at the time of diagnosis, the presence of multiple genetic defects usually prevents the identification of signaling pathways and cellular changes directly altered by the primary genetic lesion.

In more than 90% of chronic myelogenous leukemia (CML) cases, the initial genetic aberration is the Philadelphia chromosome (Ph+), which results from a reciprocal translocation between chromosome 9 within the tyrosine kinase gene ABL and chromosome 22 within the BCR gene.4,5 This creates a fusion gene product BCR-ABL.6-11 The deregulated tyrosine kinase activity of BCR-ABL is required to transform cells in vitro12 and to generate a CML-like disease in vivo.13,14 Inhibition of BCR-ABL's tyrosine kinase activity with the drug imatinib mesylate (STI571; Novartis, Basel, Switzerland) is sufficient to suppress CML in patients15-17 and relapses from therapy are due to reactivation of BCR-ABL via mutation or amplification of the gene.18,19 These results demonstrate that theBCR-ABL oncogene is required both for establishment and maintenance of CML.

CML begins with a long 3- to 5-year chronic phase characterized by an expansion of progenitor and mature myeloid cells, mild anemia, and splenomegaly. This chronic phase transforms to an aggressive stage (blast crisis) associated with numerous genetic abnormalities such as trisomy 8, trisomy 19, i(17q), and loss or mutation of p53as well as epigenetic effects leading to an expansion of immature progenitors.20,21 

The cellular origin of CML begins with formation of the Ph+in the hematopoietic stem cell (HSC) from where it is subsequently transmitted to all hematopoietic lineages.22,23 Studies of purified Ph+ HSCs suggest that either BCR-ABL is not expressed in this population24 or it is insufficient to affect the quiescent status of HSCs.25 Multipotent hematopoietic progenitors are receptive to BCR-ABL expression and generate an abnormal expansion of mature myeloid cells, a clinical feature of CML in chronic phase.26,27 These results demonstrate that although the Ph+ originates in HSCs, multipotent hematopoietic progenitors and not HSCs are the cell population responding to BCR-ABL in CML.

Numerous signaling pathways regulated by BCR-ABL expression have been identified over the past decade; however, their relevance toward the direct effect of BCR-ABL expression in multipotent hematopoietic progenitors remains unclear for 2 reasons. First, mostBCR-ABL studies have been conducted in fibroblast, myeloid, and pro-B cell lines, which do not represent multipotent hematopoietic progenitors.28 Second, BCR-ABL studies conducted in the cellular context of hematopoietic progenitors have largely relied on the availability of materials from patients with CML. These studies could not distinguish direct BCR-ABLconsequences versus other genetic and epigenetic effects that occurred during the evolution of these leukemic cells.

Alternative in vitro cellular systems have been developed that can be used to study BCR-ABL in the cellular context of hematopoietic progenitors. The pluripotent FDCP-Mix cell line has been used to demonstrate that growth factor independence of myeloid cells requires long-term BCR-ABL expression.29 Chronic BCR-ABL expression in human CD34+ multipotent progenitors via retroviral transduction delays apoptosis induced by serum and cytokine withdrawal.26 Embryonic stem (ES) cell–derived multipotent hematopoietic progenitors chronically expressing BCR-ABL via retroviral transduction generate a CML-like disease in mice.30 However, such studies could not evaluate the initial effect of BCR-ABL expression in multipotent hematopoietic progenitors.

We have developed an ES cell in vitro differentiation system with tetracycline-regulated BCR-ABL expression to study its effects during defined stages of hematopoietic differentiation.31 BCR-ABL expression throughout in vitro hematopoiesis leads to an expansion of mature myeloid cells with a concomitant reduction in erythropoiesis mimicking some of the bone marrow features of CML in chronic phase.31 

In this study we determined which specific hematopoietic progenitor populations are responsive to initial BCR-ABL expression. Based on these results we propose a mechanism by which BCR-ABL expands such progenitors and the distinctive combination of signaling pathways that may be involved.

General maintenance of feeder-independent BCR-ABL–inducible ES clones

ES clones 1-8 and 2-4, inducible for BCR-ABL expression via a tetracycline (TET)–suppressible promoter were maintained as previously described.31,32 

Maintenance of OP9 stromal cell line

Maintenance of OP9 cells was as previously described.31,32 

ES in vitro differentiation and induction of BCR-ABL expression during hematopoietic progenitor development

ES cell in vitro differentiation and induction of BCR-ABL expression was as previously described.31,32 Briefly, 104 undifferentiated ES cells, not induced for BCR-ABL expression, were cocultured on a confluent OP9 stroma per well of a 6-well plate by adding 100 ng/mL (TET). On day 5 of differentiation, both differentiated ES cells and the OP9 stroma were harvested in fresh medium in the absence of TET. Cells were replated on new dishes for 20 to 30 minutes to separate OP9 cells from differentiated ES cells. OP9 cells quickly adhered to the dish and differentiated ES cells were simply harvested by pipetting. Then, 106 differentiated ES cells were plated per 10-cm dish on a confluent OP9 cell layer. Cultures induced for BCR-ABL expression were kept without TET and control cultures had TET added. On day 7 of differentiation, hematopoietic progenitors were harvested by gentle pipetting and adherent endothelial cells were not harvested (Figure1A). BCR-ABL–expressing hematopoietic cells were identified by expression of enhanced green fluorescent protein (EGFP) and hematopoietic cells not induced for BCR-ABL expression remained EGFP.

Fig. 1.

Strategy to study the initial effects of BCR-ABL expression during hematopoietic progenitor development.

(A) BCR-ABL expression was turned on at day 5 of ES cell in vitro differentiation when the majority of the cells were mesodermal cells and hematopoietic precursors as defined by FLK-1 and VE-cadherin expression. Effects were analyzed on day 7 ES in vitro–differentiated hematopoietic progenitors by harvesting nonadherent hematopoietic progenitors with gentle pipetting and not harvesting adherent endothelial cells. In this system, removal of TET on day 5 of differentiation induced BCR-ABL IRES EGFP expression in 40% to 60% of day 7 ES-derived hematopoietic progenitors as measured by flow cytometry. BCR-ABL effects were analyzed in EGFP+ cells and EGFP cells both in BCR-ABL–induced and uninduced cultures, used as controls. (B) Exclusive BCR-ABL activation in EGFP-expressing ES-derived day 7 hematopoietic progenitor cultures without TET. EGFP+ and EGFPES-derived hematopoietic progenitors from BCR-ABL–induced cultures without TET were sorted to more than 95% purity using the FACSVantage along with EGFP cells from a control BCR-ABL uninduced culture with TET. Purified cell extracts were prepared and immunoblotted with anti-ABL and antiphosphotyrosine antibodies to determine BCR-ABL expression and activity, respectively. (C) SCF is expressed in OP9 cells and ES-derived hematopoietic progenitors. RNA was extracted from OP9 cells and day 7 ES-derived hematopoietic progenitors expressing BCR-ABL and control day 7 ES-derived hematopoietic progenitors. cDNA was generated from these samples and PCR using SCF and HPRT-specific primers were conducted. SCF primers would generate a 592-bp cDNA fragment and an about 1500-bp genomic band; HPRT primers would generate a 249-bp cDNA fragment and an approximately 1100-bp genomic band.

Fig. 1.

Strategy to study the initial effects of BCR-ABL expression during hematopoietic progenitor development.

(A) BCR-ABL expression was turned on at day 5 of ES cell in vitro differentiation when the majority of the cells were mesodermal cells and hematopoietic precursors as defined by FLK-1 and VE-cadherin expression. Effects were analyzed on day 7 ES in vitro–differentiated hematopoietic progenitors by harvesting nonadherent hematopoietic progenitors with gentle pipetting and not harvesting adherent endothelial cells. In this system, removal of TET on day 5 of differentiation induced BCR-ABL IRES EGFP expression in 40% to 60% of day 7 ES-derived hematopoietic progenitors as measured by flow cytometry. BCR-ABL effects were analyzed in EGFP+ cells and EGFP cells both in BCR-ABL–induced and uninduced cultures, used as controls. (B) Exclusive BCR-ABL activation in EGFP-expressing ES-derived day 7 hematopoietic progenitor cultures without TET. EGFP+ and EGFPES-derived hematopoietic progenitors from BCR-ABL–induced cultures without TET were sorted to more than 95% purity using the FACSVantage along with EGFP cells from a control BCR-ABL uninduced culture with TET. Purified cell extracts were prepared and immunoblotted with anti-ABL and antiphosphotyrosine antibodies to determine BCR-ABL expression and activity, respectively. (C) SCF is expressed in OP9 cells and ES-derived hematopoietic progenitors. RNA was extracted from OP9 cells and day 7 ES-derived hematopoietic progenitors expressing BCR-ABL and control day 7 ES-derived hematopoietic progenitors. cDNA was generated from these samples and PCR using SCF and HPRT-specific primers were conducted. SCF primers would generate a 592-bp cDNA fragment and an about 1500-bp genomic band; HPRT primers would generate a 249-bp cDNA fragment and an approximately 1100-bp genomic band.

Close modal

Phenotyping ES-derived hematopoietic progenitors

ES day 7 hematopoietic cells were stained with the following combination of antibodies from BD Pharmingen (San Diego, CA) except where stated: anti–TER-119–PE, anti-CD117–APC (c-KIT), anti-CD34–biotin with streptavidin-APC-CY7 (Caltag, Burlingame, CA), anti-CD11b–TC (Caltag); lineage panel: phycoerythrin (PE)–conjugated TER-119, B220, CD11b, CD4, CD8, and Gr-1. ES day 0 and day 5 differentiated cells were stained with anti–FLK-1–PE, E-cadherin (Zymed, South San Francisco, CA), or VE-cadherin (CD144) with antirat-TC antibody (Caltag). Data were acquired on a FACSVantage (Becton Dickinson, Franklin Lakes, NJ) and analyzed using WinMDI version 2.8 software (Scripps Research Institute, La Jolla, CA). Dead cells were excluded from analysis based on forward and side-scatter properties.

Morphologic analysis of ES-derived hematopoietic progenitors

ES-differentiated day 7 endothelial and hematopoietic cells were visualized with an inverted microscope at × 200. Cytospin preparations of ES-differentiated day 7 hematopoietic progenitors were analyzed by May-Grünwald/Giemsa-Wright staining using the HEMA 3 solution according to the manufacturer's protocol (Biochemical Sciences, Swedesboro, NJ).

BCR-ABL kinase inhibition studies in ES-derived hematopoietic progenitors

On day 5 of ES cell differentiation, the BCR-ABL tyrosine kinase inhibitor imatinib mesylate was added to cultures at a final concentration of 1 μM and 10 μM. On day 7 of ES differentiation, cells were analyzed.

Early-stage apoptosis analysis in ES-derived hematopoietic progenitors

ES-differentiated day 7 progenitors were stained with annexin V–PE (BD Pharmingen) to determine early stages of apoptosis. 7-Amino-actinomycin D (7-AAD) was used to exclude dead cells. The manufacturer's (BD Pharmingen) protocol was followed and samples were analyzed using a FACScan (Becton Dickinson).

Retroviral vectors and generation of virus stocks

The retroviral vector pMSCV33 containing either 5′ long terminal repeat (LTR)–driven BCR-ABLP210internal ribosome entry site (IRES) EGFP or 5′ LTR-driven IRES EGFP was used to generate high-titer helper-free retrovirus stocks prepared by transient cotransfection of 293T cells.34 

Generation of BCR-ABL–expressing BaF3 cells and IL-3 stimulation

The interleukin 3 (IL-3)–dependent BaF3 cell line was maintained in Iscove medium supplemented with 10% fetal bovine serum (FBS) and 10% WEHI-3B conditioned medium (WEHI) as a source of IL-3. Cells (5 × 105) were infected in 3 mL virus supplemented with 10% WEHI and 8 μg/mL polybrene for 3 hours. After 3 hours, 7 mL BaF3 medium was added. At 48 hours after infection, EGFP+cells were sorted from BaF3 cells infected with either murine stem cell virus (MSCV) IRES EGFP or MSCV BCR-ABLP210 IRES EGFP retrovirus to more than 95% purity on a FACSVantage (Becton Dickinson).

For IL-3 stimulation, BaF3 cells were washed 3 times in BaF3 medium lacking WEHI. Washed BaF3 cells were starved for IL-3 for 6 to 8 hours in BaF3 media lacking WEHI. WEHI was added to a final concentration of 10% to IL-3–deprived cells for 10 minutes to stimulate the IL-3 signal transduction pathway. Ten minutes after IL-3 stimulation cells were harvested for analyses.

RT-PCR analyses

EGFP was used as a surrogate marker to sort for BCR-ABL–expressing ES-differentiated day 7 hematopoietic progenitors to more than 95% purity using the FACSVantage cell sorter (Becton Dickinson). EGFP, BCR-ABL–uninduced ES-differentiated day 7 hematopoietic progenitors were similarly purified. OP9 cells were harvested by trypsinization. Total RNA was extracted from 106 cells using the RNeasy Mini Kit (Qiagen, Valencia, CA) and 1 μg RNA was used to generate cDNA using SuperScript reverse transcriptase (RT; Gibco-BRL, Carlsbad, CA) and oligo(dT) primer according to the manufacturer's protocol. One microliter cDNA was used per polymerase chain reaction (PCR) with stem cell factor (SCF) primers sense 5′-GACTGTGTGCTCTCTTCAAC-3′ and antisense 5′-CTTGCAAAACCTCCAGAGTC-3′ generating a cDNA fragment of 592 bp and a genomic DNA band of about 1500 bp; HPRT housekeeping gene primers sense 5′-CACAGGACTAGAACACCTGC-3′ and antisense 5′-GCTGGTGAAAAGGACCTCT-3′ generating a cDNA fragment of 249 bp and a genomic DNA band of about 1100 bp.35 

Protein analyses by Western blotting

BCR-ABL–expressing and control ES-differentiated day 7 hematopoietic progenitors were similarly sorted as in the RT-PCR analyses. BaF3 cells expressing BCR-ABL IRES EGFP or EGFP were similarly sorted to more than 95% purity 48 hours after infection. Cell lysates were prepared as described.36 Protein lysate (10 μg) was analyzed by immunoblotting with the following antibodies according to the manufacturer's protocol unless otherwise stated: anti-ABL was diluted 1:500,37 antiphosphotyrosine, clone 4G10 (05-321; Upstate Biotechnology, Lake Placid, NY), BCL-2 (610538; BD Transduction Laboratories, Lexington, KY), rabbit polyclonal antibodies AKT1/2 (sc-8312), ERK2 (sc-154), STAT5b (sc-835; Santa Cruz Biotechnology, Santa Cruz, CA), BCL-XL (2762), p38 MAPK (9212), phospho-p38 MAPK (Thr180/Tyr182; 9211), phospho-STAT5 (Tyr694; 9351), phospho-AKT (Ser473; 9271) (Cell Signaling Technology, Beverly, MA).

p38 MAPK inhibition studies during ES cell–derived hematopoietic progenitor development

The p38 mitogen–activated protein kinase (MAPK)–specific inhibitor SB 203580 (559398; Calbiochem, San Diego, CA) was added daily starting on day 5 of ES cell in vitro differentiation at concentrations of 0.1 μM, 1 μM, and 10 μM. An equivalent volume of dimethyl sulfoxide (DMSO) was added to control cells. ES day 7 differentiated hematopoieitic progenitors were harvested and evaluated for p38 MAPK inhibitory effects.

BCR-ABL expression at the onset of hematopoiesis expands multipotent and myeloid progenitors with suppression of differentiation into erythroblasts

We developed an ES cell in vitro differentiation system with TET-regulated BCR-ABL expression to study its effects during defined stages of hematopoietic differentiation.31,32 ES cells were induced to differentiate on an OP9 stromal cell line such that by day 5 of ES cell differentiation, the majority of cells were composed of mesodermal cells and hematopoietic precursors as defined by FLK-1 expression in 60% to 80% of the cells, VE-cadherin expression in 9% to 10% of the cells, and a lack of CD45 and TER-119 expression (Figure1A).31,32,38,39 Five to 10% of day 5 differentiated ES cells remained undifferentiated and expressed E-cadherin (Figure 1A). Individual FLK-1+, VE-cadherin–negative day 4 and day 5 differentiated ES cells, and individual FLK-1+, VE-cadherin–positive day 5 differentiated ES cells have been shown to develop into both hematopoietic and endothelial cells, demonstrating that ES differentiated day 4 and day 5 cells with these markers represent mesodermal cells and hematopoietic precursors.39BCR-ABL expression was induced by TET removal on day 5 mesodermal cells and hematopoietic precursors (Figure 1A). Turning on BCR-ABL expression at earlier time points than day 5 of ES-OP9 differentiation severely hampered subsequent hematopoietic differentiation and was not a feasible approach to study BCR-ABL effects in the cellular context of hematopoietic progenitors. For example, turning on BCR-ABL expression on day 0 of ES-OP9 differentiation abrogated differentiation of ES cells into day 5 FLK-1+ and day 7 hematopoietic cells. Turning on BCR-ABL expression on day 3 of ES-OP9 differentiation resulted in 10% to 20% of day 5 cells expressing FLK-1+with most cells remaining undifferentiated and incapable of hematopoietic development (data not shown). After turning on BCR-ABL expression on day 5 of ES-OP9 differentiation, its effects during the development of hematopoietic progenitors were studied by analyzing day 7 BCR-ABL–expressing hematopoietic progenitors and control cells (Figure 1A). Nonadherent day 7 hematopoietic progenitors were collected by gentle pipetting and adherent endothelial cells were not collected and remained on the culture dish (Figure 1A). To identify BCR-ABL–expressing cells, BCR-ABL was linked to an IRES EGFP (Figure 1A-B). EGFP intensity corresponded to the level of BCR-ABL expression (Figure 1A-B). In this system, removal of TET induced BCR-ABL expression in 40% to 60% of cells as demonstrated by EGFP fluorescence and confirmed by protein expression (Figure 1A-B). EGFP cells in BCR-ABL–induced cultures did not express BCR-ABL and were used as a control population (Figure 1A-B).

We took advantage of multiparameter cell surface marker analysis to define specific progenitor populations affected by BCR-ABL activation. C-KIT and CD34 receptors have been used in combination with lineage-depleted cells to define hematopoietic progenitors capable of in vivo multilineage reconstitution.40,41 This same set of markers was used to define multipotent hematopoietic progenitors in ES day 7 differentiated cells (Figure 2B). B- and T-cell progenitors were not generated by day 7 of ES cell in vitro differentiation and erythroblasts did not express CD34 (data not shown).31,32,38 Consequently, most experiments used the myeloid-specific CD11b integrin receptor as the sole marker for exclusion of lineage-committed progenitors from C-KIT and CD34+ multipotent progenitors (Figure 2B). Lineage-committed myeloid progenitors were defined by coexpression of C-KIT and CD34 along with CD11b (Figure 2B). Erythroblasts were identified by expression of TER-119. This was sufficient to define erythroblasts because all day 7 ES cells contain a nucleus demonstrating that no mature erythrocytes have yet formed (Figure2A).

Fig. 2.

BCR-ABL expression at the onset of hematopoiesis expands multipotent and myeloid progenitors with suppression of differentiation toward erythroblasts.

(A) Morphology of day 7 ES-differentiated hematopoietic progenitors appeared unaltered on BCR-ABL induction. Cytospin preparations were stained with May-Grünwald/Giemsa-Wright. Slides were photographed at ×1000. (B) BCR-ABL expression does not skew hematopoietic differentiation toward multipotent and myeloid progenitors. The absolute percentages of multipotent and myeloid progenitors on day 7 of ES in vitro differentiation in the presence and absence of BCR-ABL induction were determined by flow cytometry using EGFP as a surrogate marker for BCR-ABL expression. Multipotent progenitors were defined by coexpression of cell surface markers C-KIT and CD34 and lack of CD11b expression or lack of lineage panel TER-119, B220, CD4, CD8, CD11b, Gr-1 expression. Myeloid progenitors were defined with cell surface markers C-KIT, CD34, and CD11b. The ratio of multipotent to myeloid progenitors was calculated from the absolute percentages in the presence and absence of BCR-ABL induction. (C) BCR-ABL tyrosine kinase activity is required to expand ES-derived hematopoietic progenitors. The effect of the BCR-ABL tyrosine kinase inhibitor imatinib mesylate on progenitor expansion was analyzed by counting the total number of ES day 7 cells after imatinib mesylate addition on ES day 5 differentiation, in the presence and absence of BCR-ABL induction. BCR-ABL–induced cultures led to BCR-ABL expression in 54% of the cells by EGFP levels, and cell counts were normalized to 100% BCR-ABL expression. Imatinib mesylate was added at both 1 μM and 10 μM concentrations with similar results. Results represent the mean ± SEM of an experiment conducted in triplicate. Similar results were obtained in 3 independent experiments. Statistical analysis (paired t test) of BCR-ABL OFF versus BCR-ABL ON total cell population, multipotent progenitors, and myeloid progenitors was P < .01. Statistical analysis (paired ttest) of BCR-ABL OFF versus BCR-ABL ON and BCR-ABL ON plus imatinib mesylate versus BCR-ABL ON ES day 7 cultures wereP < .01. (D) Selective expansion of multipotent and myeloid progenitors but not erythroblasts on BCR-ABL induction. Total cell counts of day 7 ES-derived hematopoietic progenitors combined with percentages of multipotent progenitors (panel B), myeloid progenitors (panel B), and erythroblasts (defined by expression of TER-119) were used to obtain cell numbers of specific populations. BCR-ABL–induced cultures led to BCR-ABL expression in 54% of the cells by EGFP levels and cell counts were normalized to 100% BCR-ABL expression. Results represent the mean ± SEM of an experiment conducted in triplicate. Similar results were obtained in 3 independent experiments. Statistical analysis (paired t test) of BCR-ABL OFF versus BCR-ABL ON multipotent and myeloid progenitors wasP < .01. Statistical analysis (paired t test) of BCR-ABL OFF versus BCR-ABL ON erythroblasts wasP = .18, thereby representing an insignificant difference. (E) The tyrosine kinase activity of BCR-ABL is required to suppress hematopoietic differentiation toward erythroblasts. The effect of the BCR-ABL tyrosine kinase inhibitor imatinib mesylate on erythroblast differentiation was analyzed by examining the percentage of TER-119+ erythroblasts generated after addition of imatinib mesylate on ES day 5 differentiation, in the presence and absence of BCR-ABL induction. BCR-ABL–induced cultures led to its expression in 67% of the cells by EGFP levels and TER-119 percentages were normalized to 100% BCR-ABL expression. Imatinib mesylate was added at both 1 μM and 10 μM concentrations with similar results. Representative result of an experiment conducted in triplicate wells. Similar results were obtained in another 2 independent experiments. Statistical analysis (paired t test) of BCR-ABL OFF versus BCR-ABL ON TER-119+ erythroblasts was P < .01. X indicates fold.

Fig. 2.

BCR-ABL expression at the onset of hematopoiesis expands multipotent and myeloid progenitors with suppression of differentiation toward erythroblasts.

(A) Morphology of day 7 ES-differentiated hematopoietic progenitors appeared unaltered on BCR-ABL induction. Cytospin preparations were stained with May-Grünwald/Giemsa-Wright. Slides were photographed at ×1000. (B) BCR-ABL expression does not skew hematopoietic differentiation toward multipotent and myeloid progenitors. The absolute percentages of multipotent and myeloid progenitors on day 7 of ES in vitro differentiation in the presence and absence of BCR-ABL induction were determined by flow cytometry using EGFP as a surrogate marker for BCR-ABL expression. Multipotent progenitors were defined by coexpression of cell surface markers C-KIT and CD34 and lack of CD11b expression or lack of lineage panel TER-119, B220, CD4, CD8, CD11b, Gr-1 expression. Myeloid progenitors were defined with cell surface markers C-KIT, CD34, and CD11b. The ratio of multipotent to myeloid progenitors was calculated from the absolute percentages in the presence and absence of BCR-ABL induction. (C) BCR-ABL tyrosine kinase activity is required to expand ES-derived hematopoietic progenitors. The effect of the BCR-ABL tyrosine kinase inhibitor imatinib mesylate on progenitor expansion was analyzed by counting the total number of ES day 7 cells after imatinib mesylate addition on ES day 5 differentiation, in the presence and absence of BCR-ABL induction. BCR-ABL–induced cultures led to BCR-ABL expression in 54% of the cells by EGFP levels, and cell counts were normalized to 100% BCR-ABL expression. Imatinib mesylate was added at both 1 μM and 10 μM concentrations with similar results. Results represent the mean ± SEM of an experiment conducted in triplicate. Similar results were obtained in 3 independent experiments. Statistical analysis (paired t test) of BCR-ABL OFF versus BCR-ABL ON total cell population, multipotent progenitors, and myeloid progenitors was P < .01. Statistical analysis (paired ttest) of BCR-ABL OFF versus BCR-ABL ON and BCR-ABL ON plus imatinib mesylate versus BCR-ABL ON ES day 7 cultures wereP < .01. (D) Selective expansion of multipotent and myeloid progenitors but not erythroblasts on BCR-ABL induction. Total cell counts of day 7 ES-derived hematopoietic progenitors combined with percentages of multipotent progenitors (panel B), myeloid progenitors (panel B), and erythroblasts (defined by expression of TER-119) were used to obtain cell numbers of specific populations. BCR-ABL–induced cultures led to BCR-ABL expression in 54% of the cells by EGFP levels and cell counts were normalized to 100% BCR-ABL expression. Results represent the mean ± SEM of an experiment conducted in triplicate. Similar results were obtained in 3 independent experiments. Statistical analysis (paired t test) of BCR-ABL OFF versus BCR-ABL ON multipotent and myeloid progenitors wasP < .01. Statistical analysis (paired t test) of BCR-ABL OFF versus BCR-ABL ON erythroblasts wasP = .18, thereby representing an insignificant difference. (E) The tyrosine kinase activity of BCR-ABL is required to suppress hematopoietic differentiation toward erythroblasts. The effect of the BCR-ABL tyrosine kinase inhibitor imatinib mesylate on erythroblast differentiation was analyzed by examining the percentage of TER-119+ erythroblasts generated after addition of imatinib mesylate on ES day 5 differentiation, in the presence and absence of BCR-ABL induction. BCR-ABL–induced cultures led to its expression in 67% of the cells by EGFP levels and TER-119 percentages were normalized to 100% BCR-ABL expression. Imatinib mesylate was added at both 1 μM and 10 μM concentrations with similar results. Representative result of an experiment conducted in triplicate wells. Similar results were obtained in another 2 independent experiments. Statistical analysis (paired t test) of BCR-ABL OFF versus BCR-ABL ON TER-119+ erythroblasts was P < .01. X indicates fold.

Close modal

Short-term 48-hour BCR-ABL expression expanded the total population of day 7 ES cells by 3.7-fold (Figure 2C). This BCR-ABL effect was dependent on its tyrosine kinase activity because addition of the inhibitor imatinib mesylate suppressed progenitor expansion (Figure2D). Specifically, BCR-ABL expression expanded multipotent and myeloid progenitors by 3.1-fold and 3.0-fold, respectively (Figure 2D). The selective expansion of multipotent and myeloid progenitors could be due to a skewing of hematopoietic differentiation toward these cell types on BCR-ABL induction. However, BCR-ABL expression did not increase the ratio of multipotent and myeloid progenitors generated (Figure2B).

In contrast, BCR-ABL expression suppressed the development of erythroblasts with a 3.8-fold decrease in the percentage of erythroblasts generated (Figure 2E). This BCR-ABL effect was also dependent on its tyrosine kinase activity because addition of the inhibitor imatinib mesylate restored the percentage of erythroblasts generated (Figure 2E). However, suppression in erythroblast development did not reduce the absolute number of erythroblasts generated because BCR-ABL expression increased the total number of day 7 ES progenitors by 3.7-fold, which is to a similar degree as the 3.8-fold decrease in erythroblast development (Figure 2C,E). SCF has been shown to synergize with BCR-ABL leading to an expansion of granulocyte-macrophage colony-stimulating factor (GM-CSF) colonies27 and erythroid cells.42-44 The lack of BCR-ABL–induced erythroid expansion in ES-OP9 cultures may be due to a lack of SCF production. By RT-PCR, OP9 cells and ES-derived hematopoietic progenitors express SCF (Figures 1C and 2).45 Addition of SCF from day 5 to day 7 of ES-OP9 differentiation did not lead to a further increase in any day 7 hematopoietic cell populations in the presence or absence of BCR-ABL expression (data not shown).

BCR-ABL expands hematopoietic progenitors by suppression of programmed cell death

In cell line studies BCR-ABL expression has been shown to expand cells by suppressing programmed cell death in the face of apoptosis inducing agents such as tumor necrosis factor α (TNF-α), etoposide, or growth factor withdrawal.46-48 Primary Ph+progenitors are also resistant to growth factor withdrawal–induced cell death compared to normal progenitors.49 However, Ph+ progenitors remain susceptible to TNF-α–, ceramide-, or radiation-induced cell death.49-51 

We next determined if BCR-ABL expression expanded ES-derived hematopoietic progenitors by suppressing programmed cell death in an environment without exogenous apoptotic stimuli. Previous analyses on the quantitative role of apoptosis following BCR-ABL activation were ambiguous because coexpressed EGFP was not used as a marker to specifically define BCR-ABL+ cells in such cultures.31 

Early stages of apoptosis were measured by analyzing the level of phosphatidylserine on cell surfaces via annexin V staining. Cells that had already progressed to the stage of membrane integrity breakdown would leak out EGFP and were excluded by intercalation of 7-AAD into their DNA. BCR-ABL expression suppressed apoptosis by 3-fold, from 23% to 8% in ES-derived hematopoietic progenitors (Figure3). This was similar to the level of cell expansion mediated by BCR-ABL expression (Figure 2B). Addition of the BCR-ABL tyrosine kinase inhibitor imatinib mesylate abolished the antiapoptotic effect of BCR-ABL (Figure 3). This demonstrated that the tyrosine kinase activity of BCR-ABL was essential for its protection against programmed cell death in such progenitor cell populations.

Fig. 3.

BCR-ABL tyrosine kinase activity is required to suppress apoptosis in hematopoietic progenitors.

Percent apoptotic progenitors were analyzed by annexin V binding with progenitors already progressed to the stage of membrane permeability excluded by 7-AAD intercalation into DNA. Shown are representative results of an experiment done in triplicate cultures. Similar results were obtained in another independent experiment done in triplicate cultures.

Fig. 3.

BCR-ABL tyrosine kinase activity is required to suppress apoptosis in hematopoietic progenitors.

Percent apoptotic progenitors were analyzed by annexin V binding with progenitors already progressed to the stage of membrane permeability excluded by 7-AAD intercalation into DNA. Shown are representative results of an experiment done in triplicate cultures. Similar results were obtained in another independent experiment done in triplicate cultures.

Close modal

Late-stage apoptosis as measured by bromodeoxyuridine (BrdU) incorporation at the 3′-hydroxy termini of DNA strand breaks by terminal transferase (TUNEL assay from Phoenix Flow Systems, San Diego, CA) was low in these cultures (data not shown). Fluorescence-activated cell sorted (FACS) EGFP+ and EGFP ES day 7 progenitors were permeabilized and stained with 7-AAD in combination with BrdU to determine the level of apoptosis at both the G1 and S/G2/M phases of the cell cycle. BCR-ABL expression suppressed the modest percent of late-stage apoptotic cells predominantly at the G1 phase of the cell cycle (data not shown). There was no significant increase in the percentage of cells in the S/G2/M phase of the cell cycle on BCR-ABL induction, suggesting that an increase in cell proliferation was not a major mechanism by which BCR-ABL expanded ES-derived progenitors (data not shown). These results demonstrate that antiapoptosis was the predominant mechanism by which acute BCR-ABL expression expanded ES-derived hematopoietic progenitors.

BCR-ABL suppresses programmed cell death in a cell autonomous manner

There is conflicting evidence on whether all BCR-ABL effects are cell autonomous or can occur in a nonautonomous fashion. BCR-ABL cell autonomous effects have been demonstrated in cell cocultures in which BCR-ABL–expressing cells, and not their normal counterparts, continue to expand under growth factor–limiting conditions.27,52BCR-ABL noncell autonomous effects have been demonstrated in Ph+ progenitors that secrete IL-3,53-56 in BCR-ABL–induced murine myeloproliferative disorder cells that express high levels of IL-3 and GM-CSF,57 and in medium from BCR-ABL–expressing embryoid bodies capable of supporting growth of IL-3–dependent cell lines such as BaF3 and 32D.30 

To distinguish between BCR-ABL cell autonomous versus noncell autonomous effects, EGFP+ cells were selected and compared to EGFP cells within the same ES-derived hematopoietic progenitor culture. EGFP+ BCR-ABL–expressing progenitors had a 6% level of annexin V staining, whereas EGFPBCR-ABL–nonexpressing progenitors had a 21% level of annexin V staining within the same culture (Figure4). When comparing the level of apoptotic suppression mediated by BCR-ABL expression in separate cultures either expressing BCR-ABL or not, the same 3-fold reduction was seen (Figure3A). A range of 2.7-fold to 3.5-fold reduction in apoptosis was observed in 3 independent experiments, each done in triplicate cell cultures under BCR-ABL ON compared to BCR-ABL OFF conditions. These results demonstrate that BCR-ABL expression suppressed apoptosis in a cell autonomous manner.

Fig. 4.

BCR-ABL suppresses programmed cell death in a cell autonomous manner.

Percent apoptotic progenitors were analyzed as in Figure 3. To determine BCR-ABL cell autonomous versus noncell autonomous effects, the percent of apoptotic progenitors was analyzed within a culture containing both BCR-ABL–expressing and control cells by gating on EGFP+ and EGFP cells, respectively. A representative result of an experiment done in triplicate cultures is shown. Similar results were obtained in 3 independent experiments done in triplicate cultures.

Fig. 4.

BCR-ABL suppresses programmed cell death in a cell autonomous manner.

Percent apoptotic progenitors were analyzed as in Figure 3. To determine BCR-ABL cell autonomous versus noncell autonomous effects, the percent of apoptotic progenitors was analyzed within a culture containing both BCR-ABL–expressing and control cells by gating on EGFP+ and EGFP cells, respectively. A representative result of an experiment done in triplicate cultures is shown. Similar results were obtained in 3 independent experiments done in triplicate cultures.

Close modal

Of a select group of antiapoptotic molecules, only BCL-XL is up-regulated by BCR-ABL in hematopoietic progenitors

Programmed cell death generally requires the loss of mitochondrial membrane potential to release apoptogenic factors such as cytochromec into the cytoplasm. These factors activate protein caspases that degrade cellular proteins leading to apoptosis. Up-regulation of the BCL-2 family of antiapoptotic proteins such as BCL-2 and BCL-XL can prevent loss of mitochondrial membrane potential, cytochrome c release, and apoptosis. In addition, BCL-2 expression can inhibit apoptosis in a cytochromec/Apaf-1/caspase-9 “apoptosome”-independent manner.58 Conversely, induction of BCL-2 family proapoptotic proteins such as BAX and BAD can inactivate BCL-2 family antiapoptotic proteins leading to apoptosis (for a review, see Huang59). Cell death–suppressing signal transduction pathways such as the AKT and STAT5 pathways up-regulate BCL-2 antiapoptotic family members such as BCL-2 and BCL-XL to inhibit apoptosis.60-62 

A broad range of antiapoptotic pathways have been connected to BCR-ABL expression in various cell systems. We examined a subset of these antiapoptotic molecules including AKT, STAT5, BCL-2, and BCL- XL. AKT and STAT5 are hyperphosphorylated in various cell lines expressing BCR-ABL60,63,64 and overexpression of dominant-negative AKT or STAT5 suppresses BCR-ABL–induced transformation of 32D cells.60,65 Both BCL-2 and BCL-XL are up-regulated by BCR-ABL expression in BaF3 cells.66,67 Antisense BCL-2 RNA induces cell death in BCR-ABL–expressing BaF3 cells.66 BCL-XLoverexpression suppresses cell death induced by imatinib mesylate in the Ph+ K562 cell line.62 

ES-derived hematopoietic progenitors expressing BCR-ABL and control cultures were sorted by FACS to more than 95% purity for EGFP+ and EGFP cells, respectively. BaF3 cells expressing BCR-ABL have been shown to up-regulate all 4 antiapoptotic molecules AKT, STAT5, BCL-2, and BCL-XL60,61,63,64,66 and were included as positive controls. Stimulating BaF3 cells with IL-3 generated controls for AKT and STAT5 activation independent of BCR-ABL expression.

Our overall results demonstrate that AKT, STAT5, and BCL-2 were not up-regulated by BCR-ABL expression in ES-derived hematopoietic progenitors (Figure 5A). This was in contrast to control BaF3 cells and other continuous cell lines where similar levels of AKT and STAT5 protein were hyperphosphorylated at their respective active sites on BCR-ABL induction (Figure5A).60,63,64 BCL-2 was also up-regulated by BCR-ABL expression in BaF3 cells as previously reported (Figure5A).66 These results demonstrate that the response to BCR-ABL expression in ES-derived hematopoietic progenitors and BaF3 cells are dramatically different and therefore stress the importance of cell context–specific activation of signaling pathways.

Fig. 5.

BCR-ABL antiapoptotic signaling is dramatically different in ES-derived hematopoietic progenitors compared with BaF3 cells.

(A) Major antiapoptotic signaling molecules regulated by BCR-ABL expression in the pro-B BaF3 cell line are not in ES-derived hematopoietic progenitors. ES-derived hematopoietic progenitor and BaF3 cell lysates expressing BCR-ABL and control samples were prepared and equivalent protein amounts were immunoblotted with site-specific phospho-antibodies and total protein antibodies for STAT5 and AKT. IL-3–stimulated BaF3 cell lysates were included as positive controls for STAT5 and AKT activation. The levels of BCL-2 and BCR-ABL were measured with their respective antibodies, and for BCR-ABL activity a total phosphotyrosine antibody was used. Each molecular pathway was examined twice from 2 independent experiments with identical results. (B) BCL-XL is up-regulated on BCR-ABL induction in both ES-derived hematopoietic progenitors and BaF3 cells. Conditions are identical to panel A, with a BCL-XL antibody. This molecular pathway was analyzed 3 times from 3 separate experiment samples with identical results.

Fig. 5.

BCR-ABL antiapoptotic signaling is dramatically different in ES-derived hematopoietic progenitors compared with BaF3 cells.

(A) Major antiapoptotic signaling molecules regulated by BCR-ABL expression in the pro-B BaF3 cell line are not in ES-derived hematopoietic progenitors. ES-derived hematopoietic progenitor and BaF3 cell lysates expressing BCR-ABL and control samples were prepared and equivalent protein amounts were immunoblotted with site-specific phospho-antibodies and total protein antibodies for STAT5 and AKT. IL-3–stimulated BaF3 cell lysates were included as positive controls for STAT5 and AKT activation. The levels of BCL-2 and BCR-ABL were measured with their respective antibodies, and for BCR-ABL activity a total phosphotyrosine antibody was used. Each molecular pathway was examined twice from 2 independent experiments with identical results. (B) BCL-XL is up-regulated on BCR-ABL induction in both ES-derived hematopoietic progenitors and BaF3 cells. Conditions are identical to panel A, with a BCL-XL antibody. This molecular pathway was analyzed 3 times from 3 separate experiment samples with identical results.

Close modal

In ES-derived hematopoietic progenitors there was a basal level of BCL-2 expression and AKT phosphorylation; however, no STAT5 phosphorylation was detectable (Figure 5A). These results demonstrate that although BCL-2 and AKT up-regulation were not required for suppression of programmed cell death by BCR-ABL, a basal level of these proteins may be required for the maintenance of ES-derived hematopoietic progenitors. STAT5 phosphorylation, however, was not required either for BCR-ABL–induced cell growth or the generation of ES-derived hematopoietic progenitors.

BCL-XL was the only antiapoptotic molecule examined that was up-regulated by BCR-ABL expression in ES-derived hematopoietic progenitors (Figure 5B). Previous reports have demonstrated that BCR-ABL induction up-regulates BCL-XL in different cell types ranging from pro-B BaF3 cells, myeloid 32D cells, to the Ph+ blast crisis K562 erythroid cell line.61,67-70 BCL-XL may be a general antiapoptotic molecule that contributes to the ability of BCR-ABL to suppress cell death in different cell lineages.

BCR-ABL expression suppresses phosphorylation of p38 MAPK at its Thr180-Gly-Tyr182 active site in hematopoietic progenitors

Inhibiting the expansion of Ph+ progenitors and cell lines with chemotherapeutic agents such as interferon α (IFN-α) and butyrate correlates with phosphorylation of p38 MAPK at its Thr180-Gly-Tyr182 active site.71,72 Addition of the p38 MAPK–specific inhibitor SB 203580 is sufficient to restore in vitro expansion of Ph+ cells in the face of these chemotherapeutic agents.71,72 These results demonstrate that p38 MAPK inhibition plays a critical role in the expansion of Ph+ cells. Whether p38 MAPK suppression is directly due to BCR-ABL expression or is an event that occurs during the evolution of a Ph+ leukemic clone has not been examined.

Acute BCR-ABL expression suppressed p38 MAPK phosphorylation at its Thr180-Gly-Tyr182 active site in both ES-derived hematopoietic progenitors and BaF3 cells (Figure 6A). The total amount of p38 MAPK protein, however, was significantly elevated on BCR-ABL induction in ES-derived hematopoietic progenitors (Figure 6A). These results suggest that suppression of p38 MAPK activation may prevent p38 MAPK degradation leading to an accumulation of inactive p38 MAPK in BCR-ABL–expressing ES-derived hematopoietic progenitors.

Fig. 6.

p38 MAPK inhibition can account for most of the overall antiapoptotic effect mediated by BCR-ABL expression in hematopoietic progenitors.

(A) BCR-ABL expression suppresses p38 MAPK activity in both ES-derived hematopoietic progenitors and BaF3 cells. Conditions are identical to Figure 5 with an activation site–specific p38 MAPK phospho-antibody and total p38 MAPK antibody. This molecular pathway was analyzed 3 times from 3 independent experiments with similar results. (B) The p38 MAPK small molecule inhibitor SB 203580 expands ES-derived hematopoietic progenitors in a dose-dependent manner. Doses of 0.1, 1.0, and 10 μM of SB 203580 or control DMSO were added daily from day 5 to day 7 of ES in vitro differentiation cultures. ES-differentiated day 7 total cell numbers on SB 203580 addition were compared to those in the presence and absence of BCR-ABL induction. Results represent the mean ± SEM of an experiment conducted in triplicate. Similar results were obtained in another independent experiment done in triplicate cultures. Statistical analysis (paired t test) of BCR-ABL OFF versus BCR-ABL ON total cell population wasP < .01. Statistical analysis (paired t test) of BCR-ABL OFF plus 10 μM SB 203580 versus BCR-ABL OFF total cell population was P < .01. (C) The p38 MAPK small molecule inhibitor SB 203580 suppresses apoptosis in ES-derived hematopoietic progenitors similar to BCR-ABL expression. SB 203580 (10 μM) or control DMSO (10 μM) was added daily from day 5 to day 7 of ES in vitro differentiation cultures. The level of apoptosis as measured by annexin V levels (refer to Figure 3) on SB 203580 addition was compared to that in the presence and absence of BCR-ABL induction. A representative result of an experiment done in triplicate cultures is shown. Similar results were obtained in 2 independent experiments done in triplicate cultures.

Fig. 6.

p38 MAPK inhibition can account for most of the overall antiapoptotic effect mediated by BCR-ABL expression in hematopoietic progenitors.

(A) BCR-ABL expression suppresses p38 MAPK activity in both ES-derived hematopoietic progenitors and BaF3 cells. Conditions are identical to Figure 5 with an activation site–specific p38 MAPK phospho-antibody and total p38 MAPK antibody. This molecular pathway was analyzed 3 times from 3 independent experiments with similar results. (B) The p38 MAPK small molecule inhibitor SB 203580 expands ES-derived hematopoietic progenitors in a dose-dependent manner. Doses of 0.1, 1.0, and 10 μM of SB 203580 or control DMSO were added daily from day 5 to day 7 of ES in vitro differentiation cultures. ES-differentiated day 7 total cell numbers on SB 203580 addition were compared to those in the presence and absence of BCR-ABL induction. Results represent the mean ± SEM of an experiment conducted in triplicate. Similar results were obtained in another independent experiment done in triplicate cultures. Statistical analysis (paired t test) of BCR-ABL OFF versus BCR-ABL ON total cell population wasP < .01. Statistical analysis (paired t test) of BCR-ABL OFF plus 10 μM SB 203580 versus BCR-ABL OFF total cell population was P < .01. (C) The p38 MAPK small molecule inhibitor SB 203580 suppresses apoptosis in ES-derived hematopoietic progenitors similar to BCR-ABL expression. SB 203580 (10 μM) or control DMSO (10 μM) was added daily from day 5 to day 7 of ES in vitro differentiation cultures. The level of apoptosis as measured by annexin V levels (refer to Figure 3) on SB 203580 addition was compared to that in the presence and absence of BCR-ABL induction. A representative result of an experiment done in triplicate cultures is shown. Similar results were obtained in 2 independent experiments done in triplicate cultures.

Close modal

Inhibition of p38 MAPK activity expands hematopoietic progenitors by suppressing apoptosis analogous to BCR-ABL expression

Inhibition of p38 MAPK suppression by SB 203580 was sufficient to expand ES-derived hematopoietic progenitors in a dose-dependent manner (Figure 6B). At the highest dose of inhibitor, 10 μM SB 203580, the level of cell expansion was 80% of that observed with BCR-ABL expression (Figure 6B). p38 MAPK inhibition has been shown to suppress cell death.73,74 In ES-derived hematopoietic progenitors, 10 μM SB 203580 induced cell expansion by suppressing apoptosis from 21% to 7% as measured by annexin V binding (Figure 6C). This 3-fold reduction was of the same magnitude as the BCR-ABL level of apoptosis inhibition (Figure 6C). These results suggest that inhibition of the p38 MAPK pathway can account for a remarkably large fraction of the overall antiapoptotic effect mediated by BCR-ABL expression in ES-derived hematopoietic progenitors.

BCR-ABL suppresses p38 MAPK activity and up-regulates BCL-XL independent of each other

We examined whether inhibition of p38 MAPK led to an up-regulation of BCL-XL as a possible mechanism for suppressing apoptosis in ES-derived hematopoietic progenitors. Inhibition of p38 MAPK with 10 μM SB 203580 did not up-regulate BCL-XL in the absence of BCR-ABL expression (Figure 7). Furthermore, p38 MAPK inhibition by SB 203580 did not lead to an accumulation of total p38 MAPK unlike BCR-ABL expression (Figure 7). Similar amounts of protein lysate were examined in each condition as verified by equal levels of total ERK 2 (Figure 7). These results suggest that BCR-ABL–induced suppression of p38 MAPK activity and up-regulation of BCL-XL occur independently of each other.

Fig. 7.

BCR-ABL suppresses p38 MAPK activity and up-regulates BCL-XL independent of each other.

Conditions are identical to Figure 6, panels A and C, with an additional total ERK2 antibody. These molecular pathways were analyzed from 2 independent experiments with similar results.

Fig. 7.

BCR-ABL suppresses p38 MAPK activity and up-regulates BCL-XL independent of each other.

Conditions are identical to Figure 6, panels A and C, with an additional total ERK2 antibody. These molecular pathways were analyzed from 2 independent experiments with similar results.

Close modal

We have examined the early effects of BCR-ABL expression on the growth and development of multilineage hematopoietic progenitors. Using a BCR-ABL–inducible ES in vitro differentiation system, we demonstrate that BCR-ABL expression selectively expands multipotent and myeloid progenitors with suppression of differentiation into erythroblasts. These BCR-ABL effects require its tyrosine kinase activity and are independent of its reported scaffolding functions.75 This suppression along the erythroid lineage is extended to a reduction in mature erythrocyte development in this system.31 Together these results suggest that BCR-ABL expression may suppress hematopoietic development for the erythroid lineage leading to mild anemia in most patients with CML. The Ph+ erythroblast K562 cell line requires BCR-ABL activity to suppress hemoglobinization, providing further evidence to support this proposed mechanism.47 However, BCR-ABL expression in CD34+ progenitors can exclusively lead to an increase in erythroid colonies42-44 or GM-CSF colonies.26,27 Furthermore, embryoid body–derived BCR-ABL hematopoietic progenitors exclusively develop into erythroid cells in vitro, but differentiate into myeloid, T, and B cells in vivo.76 These apparently conflicting results suggest that microenvironmental conditions such as stromal cell contacts and growth factor levels may play pivotal roles in determining the specific lineage of cells that BCR-ABL expression affects. In our ES-OP9 differentiation system, the microenvironment may be sufficient to support expansion of BCR-ABL–expressing multipotent and myeloid progenitors and, as a consequence, reduce the development of BCR-ABL–expressing erythroblasts.

The expansion of ES-derived multipotent and myeloid progenitors was mediated by a rapid antiapoptotic effect and not by enhanced cell proliferation. Examining major antiapoptotic pathways connected to BCR-ABL expression from cell line studies revealed that AKT, STAT5, and BCL-2 were not up-regulated on BCR-ABL induction in ES-derived hematopoietic progenitors. In fact, STAT5 phosphorylation at its active site was undetectable in ES-derived hematopoietic progenitors. STAT5 has also been shown to be dispensable for generating a BCR-ABL–induced myeloproliferative disorder in vivo.77 These findings contrast with results from BCR-ABL expression in cell lines and highlight the importance of evaluating signaling in the specific cellular context of hematopoietic progenitors. In vitro ES-differentiated progenitors can contribute to long-term, multilineage hematopoiesis in vivo78 and BCR-ABL–expressing ES-derived hematopoietic progenitor clones generate an acute leukemia in vivo.30 These results suggest that ES-derived hematopoietic progenitors may be a potential cell population that can be used to study BCR-ABL effects in vivo. BCR-ABL cell context–dependent signaling has been suggested in a study where 12 genes regulated by BCR-ABL expression in a Ph+ pre-B cell line are largely unaffected in Ph+ myeloid cell lines.79 

The antiapoptotic molecule BCL-XL is up-regulated in BCR-ABL–expressing ES-derived hematopoietic progenitors. Unexpectedly, this up-regulation occurred independently of STAT5 or AKT activation. Both STAT5 and AKT activation have been linked to BCL-XLup-regulation by BCR-ABL.61,62,70,80 In fact, dominant-negative forms of STAT5 have been shown to suppress BCL-XL up-regulation in cell lines expressing BCR-ABL.61,62 However, STAT5AB-deficient pro-B cells express BCL-XL at similar levels to wild-type pro-B cells, demonstrating that STAT5 is not required for BCL-XLexpression (V. Sexl, written personal communication, December 2002). Other signaling pathways that up-regulate BCL-XL include RAS,81 ERK,82 and nuclear factor κB (NF-κB).83 These signal transducers, RAS,84 ERK,85 and NF-κB,86 are activated by BCR-ABL expression in cell line studies and could contribute to BCL-XL up-regulation in ES-derived hematopoietic progenitors.

BCL-XL inhibitors have been used to examine the contribution of BCL-XL signaling in BCR-ABL–mediated suppression of apoptosis. Cell permeable peptides targeted against the BH3 domain of BCL-XL, the natural compound tetrocarcin A, the antibiotic antimycin A, and the low-molecular-weight organic compound HA14-1 have all been shown to inhibit BCL-XL (for a review, see Huang59). BCL-XL is up-regulated in a wide range of cell types expressing BCR-ABL.61,67-70BCL-XL inhibitors could be an effective treatment for patients with CML resistant to imatinib mesylate by suppressing BCR-ABL effects in all cell types.

There is conflicting evidence on whether targeting secreted factors from Ph+ cells would be a rationale treatment option for patients with CML resistant to imatinib mesylate.27,30,52-57 In our hematopoietic progenitor cultures, BCR-ABL expression suppresses apoptosis in a cell autonomous manner and does not appear to have noncell autonomous effects.

We demonstrate that suppression of the p38 MAPK pathway can largely account for the acute reduction in apoptosis mediated by BCR-ABL expression in ES-derived hematopoietic progenitors. These results are in agreement with studies showing that p38 MAPK inhibition restores BCR-ABL transformation in the face of chemotherapeutic agents such as IFN-α and butyrate.71,72 

p38 MAPK inhibition may suppress apoptosis by up-regulating BCL-XL. Addition of the p38 MAPK inhibitor SB 203580 to ES-derived hematopoietic progenitors does not up-regulate BCL-XL, unlike BCR-ABL expression. These results suggest that BCR-ABL suppresses p38 MAPK and up-regulates BCL-XLindependently of each other.

BCR-ABL expression could suppress p38 MAPK at different points along its activation pathway. We did not observe any diminution in the phosphorylation of its upstream kinase MKK3/6 (data not shown). p38 MAPK phosphatases could be up-regulated on BCR-ABL induction. This is a difficult issue to examine because other MAPKs such as ERK and JNK can also be substrates for these phosphatases (for a review, see Lee et al87). BCR-ABL could also suppress TAB1 expression, a scaffolding protein that can bind to and autophosphorylate p38 MAPK.88 Alternatively, p38 MAPK suppression can occur on RAS activation in fibroblasts.89 BCR-ABL expression has been shown to increase the levels of active RAS-GTP84making this a likely pathway to be used by BCR-ABL for p38 MAPK suppression.

We have shown that BCR-ABL expression suppresses p38 MAPK activation in the face of IL-3 stimulation in BaF3 cells (Figure 6A). IL-3 is an activator of p38 MAPK in BaF3 cells (Figure 6A).90 IL-3 is also up-regulated on BCR-ABL induction30,53 with its production correlating to expansion of Ph+progenitors.55 BCR-ABL expression may enhance effects of IL-3–induced cell expansion by suppressing p38 MAPK-mediated apoptosis. This potential mechanism would give Ph+progenitors a selective advantage over normal cells to expand in the presence of IL-3.

We examined whether p38 MAPK inhibition was sufficient to expand both ES-derived hematopoietic progenitors and mature myeloid cells, similar to BCR-ABL expression. Although addition of the p38 MAPK inhibitor SB 203580 expanded ES-derived hematopoietic progenitors, mature myeloid cell production was severely suppressed (data not shown). These results demonstrate that BCR-ABL must regulate other signaling pathways in addition to p38 MAPK inhibition to expand myeloid cells. A comparison of signaling components regulated solely by p38 MAPK inhibition versus BCR-ABL activation in ES-derived hematopoietic progenitors may help to identify signaling pathways used by BCR-ABL to selectively expand myeloid elements.

We are grateful to Dr Veronica Sexl (University of Austria, Vienna) for providing unpublished results; Drs Arnie Berk, John Colicelli, Harvey Herschman, Caius Radu, Charles Sawyers, and Larry Zipursky (UCLA) for helpful discussions and critical reading of the manuscript; to Fiona Willis for technical expertise on the FACSVantage cell sorter; and to J. C. White for preparation of the manuscript.

O.N.W. is an Investigator of the Howard Hughes Medical Institute.

Prepublished online as Blood First Edition Paper, January 9, 2003; DOI 10.1182/blood-2002-11-3376.

Supported in part by grant CA76204 from the National Institutes of Health (O.N.W.).

The publication costs of this article were defrayed in part by page charge payment. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 U.S.C. section 1734.

1
Hanahan
D
Weinberg
RA
The hallmarks of cancer.
Cell.
100
2000
57
70
2
Jain
M
Arvanitis
C
Chu
K
et al
Sustained loss of a neoplastic phenotype by brief inactivation of MYC.
Science.
297
2002
102
104
3
Felsher
DW
Bishop
JM
Reversible tumorigenesis by MYC in hematopoietic lineages.
Mol Cell.
4
1999
199
207
4
Nowell
PC
Hungerford
DA
A minute chromosome in human chronic granulocytic leukemia [letter].
Science.
132
1960
1497
5
Rowley
JD
A new consistent chromosomal abnormality in chronic myelogenous leukemia identified by quinacrine fluorescence and Giemsa staining.
Nature.
243
1973
290
293
6
Groffen
J
Stephenson
JR
Heisterkamp
N
de Klein
A
Bartram
CR
Grosveld
G
Philadelphia chromosomal breakpoints are clustered within a limited region, bcr, on chromosome 22.
Cell.
36
1984
93
99
7
Stam
K
Heisterkamp
N
Grosveld
G
et al
Evidence of a new chimeric bcr/c-abl mRNA in patients with chronic myelocytic leukemia and the Philadelphia chromosome.
N Engl J Med.
313
1985
1429
1433
8
Shtivelman
E
Lifshitz
B
Gale
RP
Roe
BA
Canaani
E
Fused transcript of abl and bcr genes in chronic myelogenous leukaemia.
Nature.
315
1985
550
554
9
Ben-Neriah
Y
Daley
GQ
Mes-Masson
A-M
Witte
ON
Baltimore
D
The chronic myelogenous leukemia-specific P210 protein is the product of the bcr/abl hybrid gene.
Science.
233
1986
212
214
10
Heisterkamp
N
Stam
K
Groffen
J
de Klein
A
Grosveld
G
Structural organization of the bcr gene and its role in the Ph' translocation.
Nature.
315
1985
758
761
11
Konopka
JB
Watanabe
SM
Witte
ON
An alteration of the human c-abl protein in K562 leukemia cells unmasks associated tyrosine kinase activity.
Cell.
37
1984
1035
1042
12
Lugo
TG
Pendergast
A
Muller
AJ
Witte
ON
Tyrosine kinase activity and transformation potency of bcr-abl oncogene products.
Science.
247
1990
1079
1082
13
Pear
WS
Miller
JP
Xu
L
et al
Efficient and rapid induction of a chronic myelogenous leukemia-like myeloproliferative disease in mice receiving P210 bcr/abl-transduced bone marrow.
Blood.
92
1998
3780
3792
14
Wong
S
Witte
ON
Modeling Philadelphia chromosome positive leukemias.
Oncogene.
20
2001
5644
5659
15
Druker
BJ
Talpaz
M
Resta
DJ
et al
Efficacy and safety of a specific inhibitor of the BCR-ABL tyrosine kinase in chronic myeloid leukemia.
N Engl J Med.
344
2001
1031
1037
16
Kantarjian
H
Sawyers
C
Hochhaus
A
et al
Hematologic and cytogenetic responses to imatinib mesylate in chronic myelogenous leukemia.
N Engl J Med.
346
2002
645
652
17
Talpaz
M
Silver
RT
Druker
BJ
et al
Imatinib induces durable hematologic and cytogenetic responses in patients with accelerated phase chronic myeloid leukemia: results of a phase 2 study.
Blood.
99
2002
1928
1937
18
Gorre
ME
Mohammed
M
Ellwood
K
et al
Clinical resistance to STI-571 cancer therapy caused by BCR-ABL gene mutation or amplification.
Science.
293
2001
876
880
19
Branford
S
Rudzki
Z
Walsh
S
et al
High frequency of point mutations clustered within the adenosine triphosphate-binding region of BCR/ABL in patients with chronic myeloid leukemia or Ph-positive acute lymphoblastic leukemia who develop imatinib (STI571) resistance.
Blood.
99
2002
3472
3475
20
Mitelman
F
The cytogenetic scenario of chronic myeloid leukemia.
Leuk Lymphoma.
11
1993
11
15
21
Di Bacco
A
Keeshan
K
McKenna
SL
Cotter
TG
Molecular abnormalities in chronic myeloid leukemia: deregulation of cell growth and apoptosis.
Oncologist.
5
2000
405
415
22
Fialkow
PJ
Gartler
SM
Yoshida
A
Clonal origin of chronic myelocytic leukemia in man.
Proc Natl Acad Sci U S A.
58
1967
1468
1471
23
Kabarowski
JHS
Witte
ON
Consequences of BCR-ABL expression within the haematopoietic stem cell in chronic myeloid leukemia.
Stem Cells.
18
2000
399
408
24
Negrin
RS
Weissman
I
Hematopoietic stem cells in normal and malignant states.
Marrow Transplant Rev.
2
1992
23
26
25
Holyoake
T
Jiang
X
Eaves
C
Eaves
A
Isolation of a highly quiescent subpopulation of primitive leukemic cells in chronic myeloid leukemia.
Blood.
94
1999
2056
2064
26
Zhao
RC
Jiang
Y
Verfaillie
CM
A model of human p210(bcr/ABL)-mediated chronic myelogenous leukemia by transduction of primary normal human CD34(+) cells with a BCR/ABL-containing retroviral vector.
Blood.
97
2001
2406
2412
27
Moore
S
Haylock
DN
Levesque
JP
et al
Stem cell factor as a single agent induces selective proliferation of the Philadelphia chromosome positive fraction of chronic myeloid leukemia CD34(+) cells.
Blood.
92
1998
2461
2470
28
Ghaffari
S
Daley
GQ
Lodish
HF
Growth factor independence and BCR/ABL transformation: promise and pitfalls of murine model systems and assays.
Leukemia.
13
1999
1200
1206
29
Pierce
A
Spooncer
E
Wooley
S
et al
Bcr-Abl protein tyrosine kinase activity induces a loss of p53 protein that mediates a delay in myeloid differentiation.
Oncogene.
19
2000
5487
5497
30
Peters
DG
Klucher
KM
Perlingeiro
RC
Dessain
SK
Koh
EY
Daley
GQ
Autocrine and paracrine effects of an ES-cell derived, BCR/ABL-transformed hematopoietic cell line that induces leukemia in mice.
Oncogene.
20
2001
2636
2646
31
Era
T
Witte
ON
Regulated expression of P210 Bcr-Abl during embryonic stem cell differentiation stimulates multipotential progenitor expansion and myeloid cell fate.
Proc Natl Acad Sci U S A.
97
2000
1737
1742
32
Era
T
Wong
S
Witte
ON
Analysis of Bcr-Abl function using an in vitro embryonic stem cell differentiation system.
Methods in Molecular Biology.
Turksen
K
185
2001
83
95
Humana Press
Totowa, NJ
33
Hawley
RG
Lieu
FHL
Fong
ZC
Hawley
TS
Versatile retroviral vectors for potential use in gene therapy.
Gene Ther.
1
1994
136
138
34
Pear
W
Transient transfection methods for preparation of high-titer retroviral supernatants.
Current Protocols in Molecular Biology.
Ausubel
FM
Brent
R
Kingston
RE
et al
2
1996
9.11.11
9.11.18
Wiley
Boston, MA
35
Keller
G
Kennedy
M
Papayannopoulou
T
Wiles
MV
Hematopoietic commitment during embryonic stem cell differentiation in culture.
Mol Cell Biol.
13
1993
473
486
36
Lim
Y-M
Wong
S
Lau
G
Witte
ON
Colicelli
J
BCR/ABL inhibition by an escort/phosphatase fusion protein.
Proc Natl Acad Sci U S A.
97
2000
12233
12238
37
Schiff-Maker
L
Burns
MC
Konopka
JB
Clark
S
Witte
ON
Rosenberg
N
Monoclonal antibodies specific for v-abl- and c-abl-encoded molecules.
J Virol.
57
1986
1182
1186
38
Nakano
T
Kodama
H
Honjo
T
Generation of lymphohematopoietic cells from embryonic stem cells in culture.
Science.
265
1994
1098
1101
39
Nishikawa
SI
Nishikawa
S
Hirashima
M
Matsuyoshi
N
Kodama
H
Progressive lineage analysis by cell sorting and culture identifies FLK1+VE-cadherin+ cells at a diverging point of endothelial and hemopoietic lineages.
Development.
125
1998
1747
1757
40
Yoder
MC
Hiatt
K
Dutt
P
Mukherjee
P
Bodine
DM
Orlic
D
Characterization of definitive lymphohematopoietic stem cells in the day 9 murine yolk sac.
Immunity.
7
1997
335
344
41
Ito
T
Tajima
F
Ogawa
M
Developmental changes of CD34 expression by murine hematopoietic stem cells.
Exp Hematol.
28
2000
1269
1273
42
Issaad
C
Vainchenker
W
Growth of erythroid colonies in chronic myelogenous leukemia is independent of erythropoietin only in the presence of steel factor.
Blood.
84
1994
3447
3456
43
Fogli
M
Amabile
M
Martinelli
G
et al
Selective expansion of normal haemopoietic progenitors from chronic myelogenous leukaemia marrow.
Br J Haematol.
101
1998
119
129
44
Chalandon
Y
Jiang
X
Hazlewood
G
et al
Modulation of p210(BCR-ABL) activity in transduced primary human hematopoietic cells controls lineage programming.
Blood.
99
2002
3197
3204
45
Cho
SK
Webber
TD
Carlyle
JR
Nakano
T
Lewis
SM
Zuniga-Pflucker
JC
Functional characterization of B lymphocytes generated in vitro from embryonic stem cells.
Proc Natl Acad Sci U S A.
96
1999
9797
9802
46
Amarante-Mendes
GP
Naekyung Kim
C
Liu
L
et al
Bcr-Abl exerts its antiapoptotic effect against diverse apoptotic stimuli through blockage of mitochondrial release of cytochrome C and activation of caspase-3.
Blood.
91
1998
1700
1705
47
Fang
G
Kim
CN
Perkins
CL
et al
CGP57148B (STI-571) induces differentiation and apoptosis and sensitizes Bcr-Abl-positive human leukemia cells to apoptosis due to antileukemic drugs.
Blood.
96
2000
2246
2253
48
Bedi
A
Zehnbauer
BA
Barber
JP
Sharkis
SJ
Jones
RJ
Inhibition of apoptosis by BCR-ABL in chronic myeloid leukemia.
Blood.
83
1994
2038
2044
49
Maguer-Satta
V
Petzer
AL
Eaves
AC
Eaves
CJ
BCR-ABL expression in different subpopulations of functionally characterized Ph+ CD34+ cells from patients with chronic myeloid leukemia.
Blood.
88
1996
1796
1804
50
Amos
TA
Lewis
JL
Grand
FH
Gooding
RP
Goldman
JM
Gordon
MY
Apoptosis in chronic myeloid leukaemia: normal responses by progenitor cells to growth factor deprivation, X-irradiation and glucocorticoids.
Br J Haematol.
91
1995
387
393
51
Albrecht
T
Schwab
R
Henkes
M
Peschel
C
Huber
C
Aulitzky
WE
Primary proliferating immature myeloid cells from CML patients are not resistant to induction of apoptosis by DNA damage and growth factor withdrawal.
Br J Haematol.
95
1996
501
507
52
Daley
GQ
Baltimore
D
Transformation of an interleukin 3-dependent hematopoietic cell line by the chronic myelogenous leukemia-specific P210bcr/abl protein.
Proc Natl Acad Sci U S A.
85
1988
9312
9316
53
Anderson
SM
Mladenovic
J
The BCR-ABL oncogene requires both kinase activity and src-homology 2 domain to induce cytokine secretion.
Blood.
87
1996
238
244
54
Holyoake
TL
Jiang
X
Jorgensen
HG
et al
Primitive quiescent leukemic cells from patients with chronic myeloid leukemia spontaneously initiate factor-independent growth in vitro in association with up-regulation of expression of interleukin-3.
Blood.
97
2001
720
728
55
Jiang
X
Lopez
A
Holyoake
T
Eaves
A
Eaves
C
Autocrine production and action of IL-3 and granulocyte colony-stimulating factor in chronic myeloid leukemia.
Proc Natl Acad Sci U S A.
96
1999
12804
12809
56
Hariharan
IK
Adams
JM
Cory
S
BCR/ABL oncogene renders myeloid cell line factor independent: potential autocrine mechanism in chronic myeloid leukemia.
Oncol Res.
3
1988
387
399
57
Zhang
X
Ren
R
Bcr-Abl efficiently induces a myeloproliferative disease and production of excess interleukin-3 and granulocyte-macrophage colony-stimulating factor in mice: a novel model for chronic myelogenous leukemia.
Blood.
92
1998
3829
3840
58
Marsden
VS
O'Connor
L
O'Reilly
LA
et al
Apoptosis initiated by Bcl-2-regulated caspase activation independently of the cytochrome c/Apaf-1/caspase-9 apoptosome.
Nature.
419
2002
634
637
59
Huang
Z
Bcl-2 family proteins as targets for anticancer drug design.
Oncogene.
19
2000
6627
6631
60
Skorski
T
Bellacosa
A
Nieborowska-Skorska
M
et al
Transformation of hematopoietic cells by BCR/ABL requires activation of a PI-3k/Akt-dependent pathway.
EMBO J.
16
1997
6151
6161
61
Gesbert
F
Griffin
JD
Bcr/Abl activates transcription of the Bcl-X gene through STAT5.
Blood.
96
2000
2269
2276
62
Horita
M
Andreu
EJ
Benito
A
et al
Blockade of the Bcr-Abl kinase activity induces apoptosis of chronic myelogenous leukemia cells by suppressing signal transducer and activator of transcription 5-dependent expression of Bcl-xL.
J Exp Med.
191
2000
977
984
63
Shuai
K
Halpern
J
ten Hoeve
J
Rao
X
Sawyers
CL
Constitutive activation of STAT5 by the BCR-ABL oncogene in chronic myelogenous leukemia.
Oncogene.
13
1996
247
254
64
Ilaria
RL
Jr
Van Etten
RA
P210 and P190 (BCR/ABL) induce the tyrosine phosphorylation and DNA binding activity of multiple specific STAT family members.
J Biol Chem.
271
1996
31704
31710
65
Nieborowska-Skorska
M
Wasik
MA
Slupianek
A
et al
Signal transducer and activator of transcription (STAT)5 activation by BCR/ABL is dependent on intact src homology (SH)3 and SH2 domains of BCR/ABL and is required for leukemogenesis.
J Exp Med.
189
1999
1229
1242
66
Sanchez-Garcia
I
Grutz
G
Tumorigenic activity of the BCR-ABL oncogenes is mediated by BCL2.
Proc Natl Acad Sci U S A.
92
1995
5287
5291
67
Amarante-Mendes
GP
McGahon
AJ
Nishioka
WK
Afar
DE
Witte
ON
Green
DR
Bcl-2-independent Bcr-Abl-mediated resistance to apoptosis: protection is correlated with up-regulation of Bcl-xL.
Oncogene.
16
1998
1383
1390
68
Hoover
RR
Gerlach
MJ
Koh
EY
Daley
GQ
Cooperative and redundant effects of STAT5 and Ras signaling in BCR/ABL transformed hematopoietic cells.
Oncogene.
20
2001
5826
5835
69
Gelfanov
VM
Burgess
GS
Litz-Jackson
S
et al
Transformation of interleukin-3-dependent cells without participation of Stat5/bcl-xL: cooperation of akt with raf/erk leads to p65 nuclear factor kappaB-mediated antiapoptosis involving c-IAP2.
Blood.
98
2001
2508
2517
70
Salomoni P, Condorelli F, Sweeney SM, Calabretta B. Versatility of BCR/ABL-expressing leukemic cells in circumventing proapoptotic BAD effects. Blood. 2000;96
71
Witt
O
Sand
K
Pekrun
A
Butyrate-induced erythroid differentiation of human K562 leukemia cells involves inhibition of ERK and activation of p38 MAP kinase pathways.
Blood.
95
2000
2391
2396
72
Mayer
IA
Verma
A
Grumbach
IM
et al
The p38 MAPK pathway mediates the growth inhibitory effects of interferon-alpha in BCR-ABL-expressing cells.
J Biol Chem.
276
2001
28570
28577
73
Kummer
JL
Rao
PK
Heidenreich
KA
Apoptosis induced by withdrawal of trophic factors is mediated by p38 mitogen-activated protein kinase.
J Biol Chem.
272
1997
20490
20494
74
Schwenger
P
Bellosta
P
Vietor
I
Basilico
C
Skolnik
EY
Vilcek
J
Sodium salicylate induces apoptosis via p38 mitogen-activated protein kinase but inhibits tumor necrosis factor-induced c-Jun N-terminal kinase/stress-activated protein kinase activation.
Proc Natl Acad Sci U S A.
94
1997
2869
2873
75
Wertheim
JA
Forsythe
K
Druker
BJ
Hammer
D
Boettiger
D
Pear
WS
BCR-ABL-induced adhesion defects are tyrosine kinase-independent.
Blood.
99
2002
4122
4130
76
Perlingeiro
RC
Kyba
M
Daley
GQ
Clonal analysis of differentiating embryonic stem cells reveals a hematopoietic progenitor with primitive erythroid and adult lymphoid-myeloid potential.
Development.
128
2001
4597
4604
77
Sexl
V
Piekorz
R
Moriggl
R
et al
Stat5a/b contribute to interleukin 7-induced B-cell precursor expansion, but abl- and bcr/abl-induced transformation are independent of stat5.
Blood.
96
2000
2277
2283
78
Kyba
M
Perlingeiro
RC
Daley
GQ
HoxB4 confers definitive lymphoid-myeloid engraftment potential on embryonic stem cell and yolk sac hematopoietic progenitors.
Cell.
109
2002
29
37
79
Deininger
MW
Vieira
S
Mendiola
R
Schultheis
B
Goldman
JM
Melo
JV
BCR-ABL tyrosine kinase activity regulates the expression of multiple genes implicated in the pathogenesis of chronic myeloid leukemia.
Cancer Res.
60
2000
2049
2055
80
Neshat
MS
Raitano
AB
Wang
HG
Reed
JC
Sawyers
CL
The survival function of the Bcr-Abl oncogene is mediated by Bad-dependent and -independent pathways: roles for phosphatidylinositol 3-kinase and Raf.
Mol Cell Biol.
20
2000
1179
1186
81
Rosen
K
Rak
J
Leung
T
Dean
NM
Kerbel
RS
Filmus
J
Activated Ras prevents downregulation of Bcl-X(L) triggered by detachment from the extracellular matrix: a mechanism of Ras-induced resistance to anoikis in intestinal epithelial cells.
J Cell Biol.
149
2000
447
456
82
Jost
M
Huggett
TM
Kari
C
Boise
LH
Rodeck
U
Epidermal growth factor receptor-dependent control of keratinocyte survival and Bcl-xL expression through a MEK-dependent pathway.
J Biol Chem.
276
2001
6320
6326
83
Chen
C
Edelstein
LC
Gelinas
C
The Rel/NF-kappaB family directly activates expression of the apoptosis inhibitor Bcl-x(L).
Mol Cell Biol.
20
2000
2687
2695
84
Mandanas
RA
Leibowitz
DS
Gharehbaghi
K
et al
Role of p21 RAS in p210 bcr-abl transformation of murine myeloid cells.
Blood.
82
1993
1838
1847
85
Cortez
D
Reuther
G
Pendergast
AM
The Bcr-Abl tyrosine kinase activates mitogenic signaling pathways and stimulates G1-to-S phase transition in hematopoietic cells.
Oncogene.
15
1997
2333
2342
86
Hamdane
M
David-Cordonnier
MH
D'Halluin
JC
Activation of p65 NF-kappaB protein by p210BCR-ABL in a myeloid cell line (P210BCR-ABL activates p65 NF-kappaB).
Oncogene.
15
1997
2267
2275
87
Lee
JC
Kumar
S
Griswold
DE
Underwood
DC
Votta
BJ
Adams
JL
Inhibition of p38 MAP kinase as a therapeutic strategy.
Immunopharmacology.
47
2000
185
201
88
Ge
B
Gram
H
Di Padova
F
et al
MAPKK-independent activation of p38alpha mediated by TAB1-dependent autophosphorylation of p38alpha.
Science.
295
2002
1291
1294
89
Wolfman
JC
Palmby
T
Der
CJ
Wolfman
A
Cellular N-Ras promotes cell survival by downregulation of Jun N-terminal protein kinase and p38.
Mol Cell Biol.
22
2002
1589
1606
90
Rausch
O
Marshall
CJ
Cooperation of p38 and extracellular signal-regulated kinase mitogen-activated protein kinase pathways during granulocyte colony-stimulating factor-induced hemopoietic cell proliferation.
J Biol Chem.
274
1999
4096
4105

Author notes

Owen N. Witte, HHMI/UCLA, 675 Charles E. Young Dr S, Room 5720, Los Angeles, CA 90095; e-mail:owenw@microbio.ucla.edu.

Sign in via your Institution