Chronic myelogenous leukemia (CML) invariably progresses to blast crisis, which represents the most proliferative phase of the disease. The BCR-ABL1 oncogene stimulates growth and survival pathways by phosphorylating numerous substrates, including various Src family members. Here we describe up-regulation, in contrast to activation, of the ubiquitously expressed Src kinase, Fyn, by BCR-ABL1. In a tissue microarray, Fyn expression was significantly increased in CML blast crisis compared with chronic phase. Cells overexpressing BCR-ABL1 in vitro and in vivo display an up-regulation of Fyn protein and mRNA. Knockdown of Fyn with shRNA slows leukemia cell growth, inhibits clonogenicity, and leads to increased sensitivity to imatinib, indicating that Fyn mediates CML cell proliferation. In severe combined immunodeficient (SCID) mice injected with Fyn shRNA–expressing cells, myeloid-derived cell numbers dropped by 50% and death from leukemia was delayed. Taken together, these results encourage the development of therapies targeting Fyn expression.

The management of chronic myelogenous leukemia (CML) has been revolutionized by kinase inhibitors that were developed in response to cues from biologic studies of the BCR-ABL1 oncogene. However two challenging problems persist: the progression of the disease to blast crisis and resistance to kinase inhibition.1  Continued investigation of BCR-ABL1 kinase signaling will provide insight into these problems. Members of the Src kinase family, which regulate proliferation, differentiation, and motility,2  are known downstream targets of BCR-ABL1. In myeloid cell lines, BCR-ABL1 activates Lyn and Hck.3,4  Several reports link growth, survival, and imatinib resistance of Philadelphia chromosome–positive (Ph+) leukemias to Lyn kinase expression and activation.5,6  However, reports examining Fyn, a ubiquitously expressed Src family member, are sparse. Of note, phase-specific gene expression in CML using microarray analyses revealed that Fyn gene expression was linked to imatinib relapse.7  In addition, a separate study using combined systems biology and gene expression approaches in Ph+ acute lymphoblastic leukemia (ALL) specimens identified Fyn as a hub for signaling.8 

Here we show that Fyn protein expression is increased in patients with blast-crisis CML compared with chronic-phase disease. By examining effects of silencing Fyn using shRNA, we find that Fyn transduces a mitogenic signal. Collectively, our results identify a novel effect of BCR-ABL1—up-regulation of Fyn—and delineate consequences of the observed up-regulation.

Patient specimens were used for this study and were collected after informed consent was obtained in accordance with the Declaration of Helsinki. The tissue microarray studies were initiated after approval from the University of Texas M. D. Anderson Cancer Center Institutional Review Board. Animal experiments were Institutional Animal Care and Use Committee–approved.

Antibodies, chemicals, and cell lines

Antibodies were purchased from sources outlined in Document S1 (available on the Blood website; see the Supplemental Materials link at the top of the online article). Imatinib was kindly provided by Dr Elisabeth Buchdunger at Novartis Pharmaceuticals (Basel, Switzerland). Murine growth factor–dependent pro-B lymphoid BaF3 cell lines transformed with vector, wild-type BCR-ABL1, or imatinib-resistant mutant BCR-ABL1 were kindly provided by Dr Charles Sawyers9  and were cultured as previously described.9  K562 cells, TonB210 cells stably expressing a tetracycline-inducible BCR-ABL1 expression vector (kindly provided by Dr George Daley, Children's Hospital Boston, Harvard Medical School, MA),10  and mouse 32D and 32Dp210 cells were maintained in RPMI1640 medium with 10% FBS supplemented. Mouse 32D cells were supplemented with 10% WEHI-cultured conditioned medium as a source of interleukin-3 (IL-3) in addition to 10% FBS.

Design of shRNA to Fyn

K562 cells were transfected with Fyn shRNA and control vectors (TranSilent human shRNA from Panomics, Redwood, CA) using the Nucleofector system kit V and transfection program T-16 (Amaxa Biosystems, Cologne, Germany). Lentiviral knockdown of Fyn and rescue design is detailed in Document S1. To generate the rescue construct, 4 nucleotides in shRNA target no. 1 and target no. 2 sequence regions in wild-type Fyn cDNA were replaced, thereby encoding the same amino acid as wild-type Fyn, but containing different nucleotide sequences in the Fyn shRNA target region.

Assessment of doubling time, clonogenic potential, and DNA fragmentation

Doubling time was measured either 24 hours or 48 hours after plating the indicated number of Fyn shRNA– or scrambled shRNA–containing cells; total cell numbers were counted using a Vi-Cell Viability Analyzer (Beckman Coulter, Fullerton, CA). Clonogenicity was measured using MethoCult GF H4434 “Complete” Methylcellulose medium with recombinant cytokines from StemCell Technologies (Vancouver, BC) and was used in accordance with the manufacturer's instructions. Flow cytometric analysis was performed to determine the percentage of subdiploid cells as described previously.11 

Detection of Fyn mRNA, protein, and kinase activity

Standard polymerase chain reaction (PCR), Western blotting, and kinase assay procedures were used as described in Document S1.

In vivo experiments

Transplantation and shRNA in vivo experiments are described in Document S1.

Immunohistochemistry of CML tissue microarray

Staining for Fyn was conducted in bone marrow samples as detailed in Document S1.

Although the phosphorylation status and contribution to leukemogenesis of the Src family members Lyn, Hck, and Fgr has been studied in CML,3,4  little is known regarding Fyn. We examined Fyn protein expression in a tissue microarray containing bone marrow from healthy non-CML donors, and chronic-, accelerated-, and blastic-phase CML patients (Figure 1A). The average percentage of cells that stained positive for Fyn in blast crisis patients was higher than in chronic-phase or accelerated-phase patients (68.8% vs 18%; P = .007). In 11 of 16 blast crisis patients, 100% of the cells were positive for Fyn. In comparison, the majority of the combined chronic and accelerated specimens were negative for Fyn (11/18) and only one sample contained 90% Fyn-positive cells. In non-CML healthy donor bone marrow, 3 of 5 specimens were negative for Fyn and the average percentage of Fyn-positive cells was the lowest seen in any cohort (2.5%). Our results indicate that Fyn expression was lowest in non-CML marrow, slightly higher in chronic- and accelerated-phase CML, and significantly elevated in blast crisis patients. Furthermore, 2 patients in this cohort were evaluated for Fyn expression while in chronic phase and then after progression to blast crisis. In both patients, disease progression was accompanied by an increased percentage of cells staining positive for Fyn as shown by immunohistochemistry in one representative patient who progressed to myeloid blast crisis (Figure 1B).

Figure 1

Fyn is up-regulated in chronic-phase CML and in BCR-ABL1–expressing cells. (A) Fyn protein expression is increased in blast-crisis CML. A tissue microarray containing bone marrow specimens from 10 chronic-phase patients, 8 accelerated-phase patients, and 16 blastic-phase patients was stained using an anti-Fyn–directed antibody as described in detail in Document S1. The percentage of Fyn-positive cells in each sample was counted and is depicted graphically. Eleven of 16 bone marrow specimens from blast crisis CML patients were 100% positive for Fyn. Numeric percentages next to the horizontal bars refer to average percentage of Fyn-positive cells in each group. (B) Fyn expression increases from chronic phase to blast crisis in the same patient. Paired bone marrow specimens from a patient who progressed from chronic phase to blastic myeloid blast crisis were acquired and stained for Fyn as described in Document S1. Images were acquired on a Nikon Microphot FXA microscope and 20×/0.75 numeric aperture objective lens (total magnification 200×; Nikon Instruments, Melville, NY), with an Olympus DP70 camera (Olympus America, Melville, NY) running DP controller software v.1.2.1.108 (Olympus). Images were processed with Adobe Photoshop CS v.8.0.1 (Adobe Systems, San Jose, CA). (C) Fyn protein levels are increased in BCR-ABL1–expressing cells. Fyn protein was assessed by Western blotting in K562 cells that are imatinib sensitive (lane 1) or imatinib resistant (lane 2), in BaF3 cells transduced with vector (lane 3), wild-type BCR-ABL1 (lane 4), or imatinib-resistant mutant BCR-ABL1 (lanes 5,6) and in parental 32D cells or 32D cells stably transfected with p210 BCR-ABL1. The same membrane was reprobed with antiactin antibody, and the ratio of Fyn/actin protein expression was calculated by densitometry. Numeric values listed below the bands represent a ratio of Fyn protein expression to actin protein expression and are normalized relative to Fyn levels in the BaF3 vector cells, which contain the lowest level of Fyn. Results shown were reproduced in 3 independent experiments. (D) Fyn protein is up-regulated in TonB cells transfected with a tetracycline-inducible BCR-ABL1 expression vector. Western blotting for p210 BCR-ABL1 (top panel) and Fyn (middle panel) in TonB cells transfected with an inducible expression vector after gene induction with 1 μg/mL doxycycline for 12 hours, 24 hours, 36 hours, or 48 hours. Actin protein levels (bottom panel) indicate equal loading. (E) Fyn kinase activity is increased in BCR-ABL1–expressing cells. (Top panel) Immunoprecipitation of Fyn and subsequent Western blotting with a phospho-Src antibody (labeled p-Fyn) or with Fyn antibody (labeled Fyn) shows increased expression of both in BCR-ABL1–expressing cells. Actin was probed in total cell lysates prior to immunoprecipitation. (Bottom panel) Kinase assay for Fyn was conducted in BaF3 vector and BaF3 p210 cells as described in Document S1 using Sam 68 as the substrate. A band corresponding to autophosphorylation of Fyn is also highlighted. Numeric values listed below the bands represent the densitometric analysis of the band corresponding to phosphorylated Sam 68 normalized relative to levels in the BaF3 vector cells (assigned a value of 1). (F) Fyn mRNA is up-regulated in an animal model for CML. Fyn mRNA levels were examined using Fyn-specific primers in cells sorted from a recipient mouse and transplanted with bone marrow from a donor mouse's stem cells transduced with MigR1-GFP-BCR-ABL1. Fyn mRNA was qualitatively measured by RT-PCR in GFP-sorted cells (indicating successful transduction) in the left panel. Also in the left panel, as a positive control for Fyn mRNA expression, BaF3 vector and BaF3 p210 cells were used to show that mRNA for Fyn was increased in BCR-ABL1–expressing cell lines. In the right panel, Fyn mRNA was compared in mice that received a transplant of mouse bone marrow infected with empty MigR1 vector or MigR1 p210-expressing vector. Actin was also amplified. (G) Inhibition of BCR-ABL1 decreases Fyn protein expression. K562 cells were treated with diluent or 0.25 μM imatinib for 24 hours, and levels of Fyn, p210 BCR-ABL1, p145 c-abl, and actin protein were measured by Western blotting. Results are representative of 3 independent experiments.

Figure 1

Fyn is up-regulated in chronic-phase CML and in BCR-ABL1–expressing cells. (A) Fyn protein expression is increased in blast-crisis CML. A tissue microarray containing bone marrow specimens from 10 chronic-phase patients, 8 accelerated-phase patients, and 16 blastic-phase patients was stained using an anti-Fyn–directed antibody as described in detail in Document S1. The percentage of Fyn-positive cells in each sample was counted and is depicted graphically. Eleven of 16 bone marrow specimens from blast crisis CML patients were 100% positive for Fyn. Numeric percentages next to the horizontal bars refer to average percentage of Fyn-positive cells in each group. (B) Fyn expression increases from chronic phase to blast crisis in the same patient. Paired bone marrow specimens from a patient who progressed from chronic phase to blastic myeloid blast crisis were acquired and stained for Fyn as described in Document S1. Images were acquired on a Nikon Microphot FXA microscope and 20×/0.75 numeric aperture objective lens (total magnification 200×; Nikon Instruments, Melville, NY), with an Olympus DP70 camera (Olympus America, Melville, NY) running DP controller software v.1.2.1.108 (Olympus). Images were processed with Adobe Photoshop CS v.8.0.1 (Adobe Systems, San Jose, CA). (C) Fyn protein levels are increased in BCR-ABL1–expressing cells. Fyn protein was assessed by Western blotting in K562 cells that are imatinib sensitive (lane 1) or imatinib resistant (lane 2), in BaF3 cells transduced with vector (lane 3), wild-type BCR-ABL1 (lane 4), or imatinib-resistant mutant BCR-ABL1 (lanes 5,6) and in parental 32D cells or 32D cells stably transfected with p210 BCR-ABL1. The same membrane was reprobed with antiactin antibody, and the ratio of Fyn/actin protein expression was calculated by densitometry. Numeric values listed below the bands represent a ratio of Fyn protein expression to actin protein expression and are normalized relative to Fyn levels in the BaF3 vector cells, which contain the lowest level of Fyn. Results shown were reproduced in 3 independent experiments. (D) Fyn protein is up-regulated in TonB cells transfected with a tetracycline-inducible BCR-ABL1 expression vector. Western blotting for p210 BCR-ABL1 (top panel) and Fyn (middle panel) in TonB cells transfected with an inducible expression vector after gene induction with 1 μg/mL doxycycline for 12 hours, 24 hours, 36 hours, or 48 hours. Actin protein levels (bottom panel) indicate equal loading. (E) Fyn kinase activity is increased in BCR-ABL1–expressing cells. (Top panel) Immunoprecipitation of Fyn and subsequent Western blotting with a phospho-Src antibody (labeled p-Fyn) or with Fyn antibody (labeled Fyn) shows increased expression of both in BCR-ABL1–expressing cells. Actin was probed in total cell lysates prior to immunoprecipitation. (Bottom panel) Kinase assay for Fyn was conducted in BaF3 vector and BaF3 p210 cells as described in Document S1 using Sam 68 as the substrate. A band corresponding to autophosphorylation of Fyn is also highlighted. Numeric values listed below the bands represent the densitometric analysis of the band corresponding to phosphorylated Sam 68 normalized relative to levels in the BaF3 vector cells (assigned a value of 1). (F) Fyn mRNA is up-regulated in an animal model for CML. Fyn mRNA levels were examined using Fyn-specific primers in cells sorted from a recipient mouse and transplanted with bone marrow from a donor mouse's stem cells transduced with MigR1-GFP-BCR-ABL1. Fyn mRNA was qualitatively measured by RT-PCR in GFP-sorted cells (indicating successful transduction) in the left panel. Also in the left panel, as a positive control for Fyn mRNA expression, BaF3 vector and BaF3 p210 cells were used to show that mRNA for Fyn was increased in BCR-ABL1–expressing cell lines. In the right panel, Fyn mRNA was compared in mice that received a transplant of mouse bone marrow infected with empty MigR1 vector or MigR1 p210-expressing vector. Actin was also amplified. (G) Inhibition of BCR-ABL1 decreases Fyn protein expression. K562 cells were treated with diluent or 0.25 μM imatinib for 24 hours, and levels of Fyn, p210 BCR-ABL1, p145 c-abl, and actin protein were measured by Western blotting. Results are representative of 3 independent experiments.

Close modal

To clarify the relationship between BCR-ABL1 and Fyn expression, we examined Fyn protein levels in numerous CML cell lines. Western blotting revealed a 4-fold increase in Fyn protein in BaF3 p210 cells compared with BaF3 vector–transduced cells (Figure 1C lanes 3,4). Imatinib-resistant point mutants of BCR-ABL1 (T315I and E255K)12  retained the up-regulation of Fyn (Figure 1C lanes 5,6). K562 cells, which also express p210 BCR-ABL1, were positive for Fyn protein expression. Imatinib-resistant versions of these cells (which do not carry point mutations)13  also retained Fyn protein expression (Figure 1C lanes 1,2). 32D cells stably transfected with p210 BCR-ABL1 similarly showed heightened Fyn protein expression compared with parental 32D cells. Confirmation of Fyn up-regulation was further provided by TonBp210 cells transfected with a tetracycline-inducible Bcr-Abl expression vector. After exposure to 1 μg/mL doxycycline, BCR-ABL1 and Fyn expression increased in a time-dependent manner (Figure 1D). The up-regulated Fyn was found to be active as a kinase by two methods. Phospho-Fyn was detected by immunoprecipitation with a Fyn-specific antibody and Western blotting with a phospho-Src antibody (Figure 1E top panel). Total Fyn in the immunoprecipitates was increased to the same degree in p210-expressing BaF3 cells. Total lysates, collected prior to immunoprecipitation, were used to measure actin levels, which were roughly equivalent across samples, indicating equal protein loading. A Fyn kinase assay was also performed in BaF3 vector and BaF3 p210 cells using Sam 68 as a substrate, and showed a 50% increase in p210-expressing cells (Figure 1E bottom panel). Autophosphorylation of Fyn was also seen as indicated by the lower band in the bottom panel of Figure 1E. The immunoprecipitation data show a more robust increase in phospho-Fyn than the kinase assay data, likely due to different amounts of protein lysate used for the assays. One milligram of protein was used for the kinase assay, whereas 100 μg was used for the immunoprecipitation. Since these are both semiquantitative methods, it is likely that the kinase assay signal was nearly saturated in the BaF3 vector cells, thus the difference between Fyn kinase activity in the vector versus p210-expressing cells is less obvious.

Fyn up-regulation was also evident in an in vivo CML model system. Bone marrow was harvested from donor mice and infected with MigR1-GFP-p210. Infected cells were injected intravenously into irradiated recipient mice. RNA was isolated from GFP-sorted peripheral blood cells of the recipient mice. GFP-positive cells expressed Fyn mRNA, whereas GFP-negative cells did not (Figure 1F lanes 1,2). Furthermore, mice that received a transplant of bone marrow infected with vector (Figure 1F, MigR1 vector, second panel) did not up-regulate Fyn mRNA. As a control, RNA from BaF3 vector– and p210-transduced cell lines was also subjected to reverse-transcription (RT)–PCR (Figure 1F lanes 3,4) and illustrate that in cell lines, Fyn mRNA is also increased. Together, these results indicate that Fyn up-regulation occurs in BCR-ABL1–expressing cells in vivo (Figure 1F) as well as in vitro (Figure 1C,D) at the mRNA and protein level.

To place all of these events within the context of BCR-ABL1 kinase activity, we measured Fyn protein levels after exposure to 0.25 μM imatinib for 24 hours (Figure 1G). Fyn protein up-regulation was blunted, demonstrating that BCR-ABL1 kinase activity contributes to heightened Fyn protein expression.

RNA interference-based knockdown of Fyn was conducted to determine the functional significance of increased Fyn levels. Lentiviral and plasmid shRNA approaches were used. Fyn protein (Figure 2A) and mRNA (Figure 2A) expression were inhibited by more than 80% in Fyn shRNA–transfected K562 cells compared with the control scrambled vector–transfected K562 cells. Similarly, lentiviral shRNA with 2 sequences (U613 and U67) to Fyn decreased protein expression, whereas a rescue construct overexpressing a nondegradable form of Fyn partially restored expression. Protein levels of neither Lyn nor c-abl were altered in the Fyn shRNA–expressing cells (Figure 2A). Interestingly, BCR-ABL1 kinase activity was significantly repressed as measured by phosphorylation of the substrate Crk. This finding is consistent with a recent report showing that Fyn is capable of phosphorylating the SH3-SH2 region of BCR-ABL1 in vitro.14 

Figure 2

Knockdown of Fyn slows cell growth, reduces clonogenicity, inhibits growth of myeloid cells in vivo, and sensitizes cells to imatinib. (A) Cells expressing Fyn shRNA achieve knockdown of Fyn but not Lyn or c-abl. K562 cells were stably transfected with Fyn shRNA as described in “Design of shRNA to Fyn.” Levels of Fyn (top panel), Lyn, and c-abl (bottom panel) were assessed by Western blotting in cells transfected with Fyn shRNA plasmid mix (left panels) or lentivirally expressed shRNAs (U613 + U67). Numeric values indicate the ratio of Fyn/actin normalized to the scrambled/noncoding shRNA-expressing K562 cells. In the left bottom panel, Fyn mRNA was detected by RT-PCR in K562 cells transfected with scrambled shRNA or shRNA plasmids directed toward Fyn. Numeric values represent the ratio of band intensity corresponding to Fyn mRNA to actin band intensity and are not normalized. Right panel shows a kinase assay for BCR-ABL1 in K562 cells infected with nonspecific shRNA (LV) or Fyn-targeted shRNAs (U613 + U67). Phosphorylation of the BCR-ABL1 substrate, p-Crk, is lost in Fyn shRNA–expressing cells. (B) Fyn knockdown slows cell growth significantly. K562 cells transfected with scrambled shRNA or Fyn shRNA (left graph) were plated at a density of 200 000 cells/mL. After 24 hours and 48 hours, the numbers of cells were counted using a ViCell Coulter Counter (Beckman Coulter). At 48 hours, there was a statistically significant difference (P < .01) in cell number. Overexpression of a mutant Fyn not degraded by Fyn shRNA restores cell numbers (right graph). K562 cells were plated at a density of 750 000 cells/mL, and cell numbers were counted at 24 and 48 hours. At 48 hours, there was a statistically significant difference in cell number (P < .04). (C) Fyn knockdown significantly diminishes clonogenic growth. K562 cells infected with nothing (K562), lentivirus with nonspecific shRNA (LV), lentivirus with Fyn shRNAs (U613 + U67), or mutant Fyn not degraded by shRNA were plated in Methocult media, and the numbers of colonies were counted after 5 days. There was a significant decrease in colony number in Fyn shRNA–expressing cells (P < .05), whereas the rescue construct restored clonogenic potential (P < .05). (D) Fyn knockdown sensitizes cells to imatinib-induced apoptosis. K562 cells infected with nothing (K562), lentivirus with nonspecific shRNA (LV), lentivirus with Fyn shRNAs (U613 + U67), or mutant Fyn not degraded by shRNA were treated with 5 μM imatinib for 48 hours. Subdiploid percentage of cells, indicative of DNA fragmentation, was assessed by PI staining and subsequent analysis by flow cytometry. Bar graph depicts 3 experiments. (E) Myeloid cell growth in vivo is slowed in SCID mice injected with Fyn shRNA–expressing cells. SCID mice were injected with 20 million K562 cells expressing scrambled or Fyn-directed shRNA. Injected cells were more than 90% viable as determined by trypan blue staining. After 4 weeks, peripheral blood from 10 mice (5 reconstituted with scrambled shRNA and 5 reconstituted with Fyn shRNA) was collected and CBCs were conducted. Percentages of segmented cells are shown. Error bars represent SD. (F) Survival of SCID mice reconstituted with Fyn shRNA–expressing cells versus scrambled shRNA–expressing cells. Kaplan-Meier analysis of mice described in panel 2E (P = .079).

Figure 2

Knockdown of Fyn slows cell growth, reduces clonogenicity, inhibits growth of myeloid cells in vivo, and sensitizes cells to imatinib. (A) Cells expressing Fyn shRNA achieve knockdown of Fyn but not Lyn or c-abl. K562 cells were stably transfected with Fyn shRNA as described in “Design of shRNA to Fyn.” Levels of Fyn (top panel), Lyn, and c-abl (bottom panel) were assessed by Western blotting in cells transfected with Fyn shRNA plasmid mix (left panels) or lentivirally expressed shRNAs (U613 + U67). Numeric values indicate the ratio of Fyn/actin normalized to the scrambled/noncoding shRNA-expressing K562 cells. In the left bottom panel, Fyn mRNA was detected by RT-PCR in K562 cells transfected with scrambled shRNA or shRNA plasmids directed toward Fyn. Numeric values represent the ratio of band intensity corresponding to Fyn mRNA to actin band intensity and are not normalized. Right panel shows a kinase assay for BCR-ABL1 in K562 cells infected with nonspecific shRNA (LV) or Fyn-targeted shRNAs (U613 + U67). Phosphorylation of the BCR-ABL1 substrate, p-Crk, is lost in Fyn shRNA–expressing cells. (B) Fyn knockdown slows cell growth significantly. K562 cells transfected with scrambled shRNA or Fyn shRNA (left graph) were plated at a density of 200 000 cells/mL. After 24 hours and 48 hours, the numbers of cells were counted using a ViCell Coulter Counter (Beckman Coulter). At 48 hours, there was a statistically significant difference (P < .01) in cell number. Overexpression of a mutant Fyn not degraded by Fyn shRNA restores cell numbers (right graph). K562 cells were plated at a density of 750 000 cells/mL, and cell numbers were counted at 24 and 48 hours. At 48 hours, there was a statistically significant difference in cell number (P < .04). (C) Fyn knockdown significantly diminishes clonogenic growth. K562 cells infected with nothing (K562), lentivirus with nonspecific shRNA (LV), lentivirus with Fyn shRNAs (U613 + U67), or mutant Fyn not degraded by shRNA were plated in Methocult media, and the numbers of colonies were counted after 5 days. There was a significant decrease in colony number in Fyn shRNA–expressing cells (P < .05), whereas the rescue construct restored clonogenic potential (P < .05). (D) Fyn knockdown sensitizes cells to imatinib-induced apoptosis. K562 cells infected with nothing (K562), lentivirus with nonspecific shRNA (LV), lentivirus with Fyn shRNAs (U613 + U67), or mutant Fyn not degraded by shRNA were treated with 5 μM imatinib for 48 hours. Subdiploid percentage of cells, indicative of DNA fragmentation, was assessed by PI staining and subsequent analysis by flow cytometry. Bar graph depicts 3 experiments. (E) Myeloid cell growth in vivo is slowed in SCID mice injected with Fyn shRNA–expressing cells. SCID mice were injected with 20 million K562 cells expressing scrambled or Fyn-directed shRNA. Injected cells were more than 90% viable as determined by trypan blue staining. After 4 weeks, peripheral blood from 10 mice (5 reconstituted with scrambled shRNA and 5 reconstituted with Fyn shRNA) was collected and CBCs were conducted. Percentages of segmented cells are shown. Error bars represent SD. (F) Survival of SCID mice reconstituted with Fyn shRNA–expressing cells versus scrambled shRNA–expressing cells. Kaplan-Meier analysis of mice described in panel 2E (P = .079).

Close modal

Proliferation of Fyn shRNA–containing cells versus scrambled shRNA–containing cells was assessed by plating equivalent cell numbers and comparing cell counts after 24 hours and 48 hours (Figure 2B left graph). Cells transfected with Fyn shRNA grew slower than counterparts transfected with scrambled shRNA (P < .01). A rescue construct, encoding a mutant Fyn not susceptible to shRNA degradation, stimulated cell growth (P < .04) compared with Fyn shRNA–expressing cells (Figure 2B right graph). The effects of Fyn knockdown on cell survival were assessed using clonogenic assays. Significantly fewer colonies were apparent after Fyn shRNA transduction (P < .05), whereas transduction with the rescue construct restored clonogenic growth (P < .05; Figure 2C). Fyn shRNA also sensitized K562 cells to imatinib treatment. Treatment with 5 μM imatinib for 48 hours elicited higher levels of DNA fragmentation in K562 cells expressing Fyn shRNA than nontargeted shRNA (P < .005) or Fyn rescue construct (P < .05; Figure 2D).

To test whether Fyn knockdown impacted CML growth in vivo, we injected severe combined immunodeficient (SCID) mice with K562 cells containing shRNA directed toward Fyn or toward an unrelated sequence. Four weeks later, peripheral blood cell counts were assessed. Percentages of segmented cells were lower in mice injected with Fyn shRNA–containing cells (P = .05; Figure 2E). There was a trend toward prolonged survival in mice carrying Fyn shRNA compared with those carrying nonspecific shRNA. However, statistical analyses of this small cohort indicated it did not meet statistical significance (P = .079; Figure 2F).

While existing therapies, such as dasatinib, target activation of Src family members and indeed inhibit Fyn kinase activity,15  our data suggest that increased Fyn protein expression is a feature of CML cells. Furthermore, our data corroborate those of others suggesting that Fyn expression may control BCR-ABL1 kinase activity thereby making Fyn an attractive target for CML therapies. Taken together, our data highlight the importance of Fyn expression in BCR-ABL1–expressing cells and have diagnostic implications for improved prediction of patients who might be transforming to blast crisis and require a therapeutic intervention.

The online version of this article contains a data supplement.

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 USC section 1734.

We thank Gary Gallick, PhD, for sharing his expertise on Src family members and for thoughtful discussions and feedback.

This work was supported by grants from the National Institutes of Health (RO1 CA115811 to J.C) and from the Children's Leukemia Research Foundation (J.C.).

National Institutes of Health

Contribution: K.B., Y.G., X.L., and Q.L. performed research; H.M.A. and M.M. analyzed data; A.H. and C.M. performed research and analyzed data; R.B.A. and M.B.-E. contributed reagents; and J.C. designed research, analyzed data, and wrote the paper.

Conflict-of-interest disclosure: The authors declare no competing financial interests.

Correspondence: Joya Chandra, PhD, Box 853, Pediatrics Research, University of Texas M. D. Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030; e-mail: jchandra@mdanderson.org.

1
Van Etten
RA
Oncogenic signaling: new insights and controversies from chronic myeloid leukemia.
J Exp Med
2007
204
461
465
2
Palacios
EH
Weiss
A
Function of the Src-family kinases, Lck and Fyn, in T-cell development and activation.
Oncogene
2004
23
7990
8000
3
Danhauser-Riedl
S
Warmuth
M
Druker
BJ
Emmerich
B
Hallek
M
Activation of Src kinases p53/56lyn and p59hck by p210bcr/abl in myeloid cells.
Cancer Res
1996
56
3589
3596
4
Hu
Y
Liu
Y
Pelletier
S
et al
Requirement of Src kinases Lyn, Hck and Fgr for BCR-ABL1-induced B-lymphoblastic leukemia but not chronic myeloid leukemia.
Nat Genet
2004
36
453
461
5
Donato
NJ
Wu
JY
Stapley
J
et al
BCR-ABL independence and LYN kinase overexpression in chronic myelogenous leukemia cells selected for resistance to STI571.
Blood
2003
101
690
698
6
Ptasznik
A
Nakata
Y
Kalota
A
Emerson
SG
Gewirtz
AM
Short interfering RNA (siRNA) targeting the Lyn kinase induces apoptosis in primary, and drug-resistant, BCR-ABL1(+) leukemia cells.
Nat Med
2004
10
1187
1189
7
Radich
JP
Dai
H
Mao
M
et al
Gene expression changes associated with progression and response in chronic myeloid leukemia.
Proc Natl Acad Sci U S A
2006
103
2794
2799
8
Juric
D
Lacayo
NJ
Ramsey
MC
et al
Differential gene expression patterns and interaction networks in BCR-ABL-positive and -negative adult acute lymphoblastic leukemias.
J Clin Oncol
2007
25
1341
1349
9
Chandra
J
Tracy
J
Loegering
D
et al
Adaphostin-induced oxidative stress overcomes BCR/ABL mutation-dependent and -independent imatinib resistance.
Blood
2006
107
2501
2506
10
Klucher
KM
Lopez
DV
Daley
GQ
Secondary mutation maintains the transformed state in BaF3 cells with inducible BCR/ABL expression.
Blood
1998
91
3927
3934
11
Chandra
J
Hackbarth
J
Le
S
et al
Involvement of reactive oxygen species in adaphostin-induced cytotoxicity in human leukemia cells.
Blood
2003
102
4512
4519
12
Gorre
ME
Mohammed
M
Ellwood
K
et al
Clinical resistance to STI-571 cancer therapy caused by BCR-ABL gene mutation or amplification.
Science
2001
293
876
880
13
Mow
BM
Chandra
J
Svingen
PA
et al
Effects of the Bcr/abl kinase inhibitors STI571 and adaphostin (NSC 680410) on chronic myelogenous leukemia cells in vitro.
Blood
2002
99
664
671
14
Meyn
MA
III
Wilson
MB
Abdi
FA
et al
Src family kinases phosphorylate the Bcr-Abl SH3-SH2 region and modulate Bcr-Abl transforming activity.
J Biol Chem
2006
281
30907
30916
15
Doggrell
SA
BMS-354825: a novel drug with potential for the treatment of imatinib-resistant chronic myeloid leukaemia.
Expert Opin Investig Drugs
2005
14
89
91

Author notes

K.B. and Y.G. contributed equally to this work.

Sign in via your Institution