Purpose: Adult T-cell leukemia/lymphoma (ATLL) is an aggressive human T-cell malignancy induced by human T-lymphotrophic virus-1 (HTLV-1) infection. The genetic alterations in infected cells that lead to transformation have not been completely elucidated, thus hindering the identification of effective therapeutic targets for ATL. Here, we present the first assessment of MYB proto-oncogene dysregulation in ATL and an exploration of its role in the onset of ATL.

Experimental Design: We investigated the expression patterns of MYB splicing variants in ATL. The molecular characteristics of the c-Myb-9A isoform, which was overexpressed in ATL cells, were examined using chromatin immunoprecipitation and promoter assays. We further examined the biologic impacts of abnormal c-Myb overexpression in ATL using overall c-Myb knockdown with shRNA or c-Myb-9A knockdown with morpholino oligomers.

Results: Both total c-Myb and c-Myb-9A, which exhibited strong transforming activity, were overexpressed in ATL cells in a leukemogenesis- and progression-dependent manner. Knockdown of either total c-Myb or c-Myb-9A induced ATL cell death. c-Myb transactivates nine genes that encode essential regulators of cell proliferation and NF-κB signaling. c-Myb-9A induced significantly stronger transactivation of all tested genes and stronger NF-κB activation compared with wild-type c-Myb.

Conclusions: Our data demonstrate that c-Myb pathway overactivation caused by unbalanced c-Myb-9A overexpression is associated with disorders in cellular homeostasis and consequently, accelerated transformation, cell proliferation, and malignancy in ATL cells. These data support the notion of the c-Myb pathway as a promising new therapeutic target for ATL. Clin Cancer Res; 22(23); 5915–28. ©2016 AACR.

Translational Relevance

Adult T-cell leukemia/lymphoma (ATL), one of the most aggressive T-cell malignancies, is caused by human T-lymphotrophic virus type 1 (HTLV-1) infection. As HTLV-1 pathogenesis and the genetic disorders responsible for ATL onset and maintenance are poorly understood, there is currently no effective cure. In the current study, we present an initial exploration of the c-Myb proto-oncogene pathway dysregulation as a novel therapeutic target in ATL for the first time. We found that an oncogenic c-Myb isoform with a strong transforming capacity, c-Myb-9A, is overexpressed in ATL cells. We demonstrated that c-Myb-9A induces significantly higher transactivation of its target genes, including critical regulators of cell proliferation and NF-κB activity compared with wild-type c-Myb. Our data indicate a close correlation between c-Myb pathway dysregulation due to c-Myb-9A overexpression and disorders in cellular homeostasis and, consequently, ATL.

The proto-oncogene MYB has been identified as the cellular counterpart of v-Myb; the latter oncogene is present in the avian myeloblastosis virus (AMV) and E26, the product of which induces myeloid and lymphoid leukemia in infected birds (1). In normal cells, c-Myb functions physiologically as a transcription factor and is involved in the regulation of hematopoietic cell development and proliferation. The primary structure and functional mechanisms of c-Myb have been well described (2–4).

The c-Myb protein has a molecular weight of approximately 75 kDa and consists of three major domains: an N-terminal DNA-binding domain (DBD), a central transactivation domain (TAD), and a C-terminal–negative regulatory domain (NRD). The TAD and DBD are essential for the function of c-Myb as a transcription factor, whereas the NRD is a critical regulator of c-Myb activity (2, 5). MYB mRNA contains several natural alternative splicing variants produced by the usage of alternative exons. Woo and colleagues first demonstrated that one MYB splicing variant encoded a c-Myb isoform bearing an additional 121 amino acids not present in wild-type (WT) c-Myb (6). Subsequently, Kumar and colleagues demonstrated that expression of the 89-kDa c-Myb isoform, encoded by a MYB transcript variant containing extra 363 base pairs between exons 9 and 10, induced higher cell viability than did WT c-Myb (7). Most recently, O'Rourke and Ness identified a series of MYB splicing variants formed using the alternative exons 8A, 9A, 9B, 10A, 13A, and 14A and demonstrated the expression patterns of these variants in hematopoietic cell lines (Jurkat, KG-1, and K562), as well as in primary acute myeloid leukemia (AML) and T-cell acute lymphoblastic leukemia (ALL) cells (8). The authors observed overall increases in total MYB RNA levels, as well as MYB-9A and -10A mRNA levels, in patients with AML and ALL. In particular, up to 180-fold increases in MYB-9A mRNA levels were observed in T-cell ALL samples compared with normal bone marrow samples. The authors also found that the MYB-9A and -10A mRNAs, which included premature termination codons (PTCs) within exons 9A and 10A, respectively, encoded c-Myb isoforms (c-Myb-9A and -10A, respectively) that lacked the C-terminal NRD region. Both isoforms, which lack NRD to a similar degree as that observed in v-Myb, exhibited transactivity levels similar to or even exceeding that of v-Myb.

Notably, given its roles in hematopoietic cell differentiation and proliferation, c-Myb is expressed only in immature myeloid and lymphoid T cells but not in normal mature T and B cells (9–13). Importantly, Maurice and colleagues reported that c-Myb plays a critical role in the determination of T-helper cell fate by directly targeting the GATA3 gene (14); another study subsequently investigated in detail the molecular regulatory mechanism of GATA3 gene expression by c-Myb (15). Conversely, c-Myb overexpression and dysregulation have been frequently observed in hematopoietic tumors, particularly AML, ALL, and lymphomas, and the implications of c-Myb overexpression/deregulation and leukemogenesis have been extensively investigated (4, 5). Despite the accumulation of knowledge about the physiologic roles of c-Myb in normal and tumor cells, the biologic significance of c-Myb overexpression in leukemic cells has not been completely clarified.

Adult T-cell leukemia/lymphoma (ATLL), one of the most aggressive T-cell malignancies, is caused by human T-lymphotrophic virus type 1 (HTLV-1) infection. More than a million HTLV-1 carriers have been reported in areas where HTLV-1 is endemic, such as Japan (∼1% of the national population; ref. 16). Although the lifetime incidence of ATL among HTLV-1 carriers is approximately 5%, estimations suggest that annually, approximately 1,000 deaths in Japan are attributable to ATL. HTLV-1 was discovered to be the causative agent of ATL in the early 1980s (17–19). HTLV-1 is mainly transmitted to infants from infected mothers through breastfeeding. HTLV-1 then leads to the immortalization of infected cells, and the accumulation of genetic disorders triggers malignant transformation in these cells, followed by a monoclonal expansion of ATL leukemic cells. Nevertheless, the underlying molecular mechanism of HTLV-1 pathogenesis and the genetic disorders responsible for the onset of ATL are poorly understood. Identification of the key molecules responsible for ATL leukemogenesis is necessary for the identification of new therapeutic targets for ATL.

Our laboratory previously conducted comparative analyses of the gene expression patterns in ATL cells (n = 52) and normal CD4+ T cells (n = 21); these revealed MYB mRNA overexpression and upregulation of the c-Myb pathway in ATL cells (GSE33615). As described previously, c-Myb expression is normally suppressed in mature T cells. Consequently, we hypothesized that the abnormal overexpression of c-Myb might allow this protein to function when it is not required, thus dysregulating cellular homeostasis in ATL cells. In the current study, we observed for the first time the overexpression of not only total c-Myb but also an oncogenic splicing variant, MYB-9A, in primary ATL cells. We demonstrated that c-Myb targets various genes that encode essential regulators of cell proliferation and that c-Myb-9A induces significantly higher levels of transactivation of these genes compared with WT-c-Myb. Our data suggest that c-Myb pathway dysregulation consequent to the unbalanced overexpression of c-Myb-9A, which overproduces and thus overactivates c-Myb target genes, might underlie the wide range of abnormalities and unlimited cell growth observed in ATL cells.

Cell lines and cells

All cell lines used in the current study were obtained and maintained as previously reported (20, 21). Briefly, Jurkat (T-ALL patient-derived T-cell line), TL-Om1 (ATL patient-derived T-cell line), and MT-2 (HTLV-1 immortalized T-cell line) were maintained in RPMI (Gibco) containing 10% FBS (Gibco) at 37°C with 5% CO2. HeLa (cervical cancer–derived epithelial cell line), 293T (human embryonic kidney–derived cell line containing the SV40 T-antigen), and 293FT (derivative of 293T) cells were cultured in DMEM (Nissui) containing 10% FBS (Gibco) at 37°C with 5% CO2.

The cell lines used in this study were obtained and authenticated as follows. TL-Om1, derived from malignant T cells from a patient with ATL, was kindly provided by Dr. K. Sugamura in Tohoku University (Tokyo, Japan). We authenticated within 6 months of this study by FISH and HTLV-1 provirus–specific real-time PCR and confirmed that this cell line contained 1 copy/cell of HTLV-1 provirus at the site of 1p13 of chromosome 1 in the gDNA, maintaining the original characteristics of TL-Om1 (22). MT-2, HTLV-1 transformed T cell line, was kindly provided by Dr. H. Hoshino in Gunma University (Tokyo, Japan). We authenticated within 6 months of this study by the proviral integration site sequencing technique (23) and confirmed that this cell line contains 10 copies of HTLV-1 provirus/cell. We also confirmed that this cell line produced infectious HTLV-1 viral particles and expressed viral Gag-Tax fusion protein, which are major characteristics of MT-2 cells. Jurkat was obtained within 6 month of this study from ATCC that authenticated this cell line by short tandem repeat (STR) analysis. 293FT was obtained more than 6 months before this study from Riken Cell Bank that conducted a series of authentication analysis on this cell line including growth curve analysis, cell adhesion analysis, isozyme analysis, and STR analysis. Just after arrival to our laboratory, 293FT cells were cultured for several days in the conditioned medium and divided to 20 cryo-tubes as live cell stock, then stored in liquid N2 until use. HeLa and 293T cells were gifted by the Japanese Foundation for Cancer Research (JFCR) more than 6 months prior to this study. We authenticated these cell lines by karyotype analysis within 6 months of this study.

Peripheral blood mononuclear cells (PBMCs) from patients with ATL and healthy volunteers were a part of those collected with an informed consent as a collaborative project of the Joint Study on Prognostic Factors of ATL Development (JSPFAD). The project was approved by the Human Genome Research Ethics Committee in the Institute of Medical Sciences, the University of Tokyo (IMSUT; Tokyo, Japan). Clinical information of patients with ATL and healthy volunteers enrolled in the current study is shown in Supplementary Table S6.

Quantitative real-time PCR

Total RNA was extracted using Isogen (Nippon Gene Co., Ltd.) following the manufacturer's protocol. Extracted total RNA samples were subjected to reverse transcription using SuperScript II (Life Technologies, Thermo Fisher Scientific), followed by quantitative real-time PCR using SYBR Premix Ex Taq and thermal cycler dice (both from Takara Bio Inc.) or by semiquantitative PCR. Primer sequences used for real-time PCR are shown in Supplementary Table S1.

c-Myb protein expression plasmids

c-Myb expression plasmids (c-Myb-8A, 9A, 9B, 10A, 13A, and 14A in pCDNA3 mammalian expression vector) were generous gifts from Dr. O'Rourke (8). FLAG-tagged c-Myb expression plasmids (WT-c-Myb, 9A, and 10A) were generated by inserting PCR-amplified cDNA fragments for each isoform into pME-FLAG vector at XhoI and SpeI sites.

Western blotting and antibodies

For detection of proteins in Western blotting, the following primary antibodies were purchased from the indicated companies: c-Myb (#ab45150, Abcam), FoxM1 (#sc-500, Santa Cruz Biotechnology), AURKA (#121005, Cell Signaling Technology), EZH2 (#3147S, Cell Signaling Technology), NIK (#4994S, Cell Signaling Technology), NF-κB2-p52 (#sc-7386, Santa Cruz Biotechnology), NF-κB1-p50 (#sc-1190X, Santa Cruz Biotechnology), IκBα (#sc-371, Santa Cruz Biotechnology), P-IκBα (#9246S, Cell Signaling Technology), FLAG (F3165; Sigma-Aldrich Corporation), and β-actin (sc-69879; Santa Cruz Biotechnology, Inc.). For the secondary antibodies conjugated with alkaline phosphatase, anti-mouse IgG (S372B; Promega) and anti-rabbit IgG (S373B; Promega) were used depending on the host species of the primary antibody.

Chromatin immunoprecipitation assays

Chromatin immunoprecipitation (ChIP) against endogenous c-Myb was conducted in Jurkat and TL-Om1 cells, and ChIP against FLAG-tagged c-Mybs (WT, c-Myb-9A, and c-Myb-10A) was conducted in 293FT cells transiently overexpressing each FLAF-c-Myb. The ChIP procedures is described elsewhere (24) with minor modifications. For immunoprecipitation of c-Myb in Jurkat and TL-Om1, rabbit monoclonal c-Myb antibody (#ab45150, Abcam) was used. Normal rabbit IgG (#2729P, Cell Signaling Technology) was used as the negative control. For precipitation of FLAG-tagged c-Myb in 293FT cells, anti-FLAG M2 Affinity Gel (#A2220, Sigma) was used. For the negative control, the lysate was incubated with a normal mouse IgG (#I5381, Sigma), followed by precipitation with ProteinG 4 Fast Flow sepharose (#17-0618-01, GE Healthcare). Primer sequences for detection of c-Myb target promoters by real-time PCR are listed in Supplementary Table S2.

Detection of activated NF-κB p65 by immunocytochemistry

Activated NF-κB p65 was detected by immunocytochemistry in HeLa cells overexpressing c-Myb isoforms (WT, 8A, 9A, 9B, 10A, 13A, and 14A). Fixed HeLa cells, transiently overexpressing c-Myb isoforms (WT, c-Myb-8A, 9A, 9B, 10A, 13A, and 14A) or mock-transfected with the empty vector as the negative control, were fixed in 4% paraformaldehyde for 5 minutes, permeabilized in 0.1% Triton X-100 in PBS for 10 minutes, blocked in 3% BSA in PBS for 1 hour, and incubated with an mAb against activated subunit of NF-κB-p65 (#MAB3026, Millipore) followed by Alexa Fluor 546–conjugated goat anti-mouse IgG (A11030, Invitrogen). Cells were then observed by Zeiss710 laser scanning microscope.

Electrophoretic mobility shift assay

Detection of NF-κB activity by electrophoretic mobility shift assay (EMSA) in nuclear extract of cells was conducted as described elsewhere (21). Briefly, nuclear extracts from Jurkat cells with c-Myb overexpression or knockdown and TL-Om1 cells with c-Myb knockdown were prepared and incubated with [γ-32P]ATP-labeled oligo DNA probes for NF-κB (#E3291, Promega) and for Oct1 (#E3241, Promega). The samples were separated by electrophoresis with 4% acrylamide gels at 150 V for 1.5 hours and completely dried at 65°C for 3 hours. The band intensity of 32P-labeled probe, binding to nuclear proteins, was detected by autoradiography.

Transformation assays

Soft agar assays were conducted in NIH3T3 cells stably expressing WT-c-Myb, c-Myb-9A, or c-Myb-10A. NIH3T3 cells stably expressing WT-c-Myb, c-Myb-9A, or c-Myb-10A were established by transfecting c-Myb-pCDNA3 plasmid and selecting by addition of 0.8 mg/mL G418 for 10 days. HA-pCDNA3 plasmid was transfected as negative control. Selected c-Myb expressing NIH3T3 cells were seeded at a density of 1 × 104 cells in 1 mL of 0.3% agarose in DMEM with 10% FBS over a cushion of 2-mL 0.5% agarose in DMEM with 10% FBS per well in 6-well plate. After the top agar was set, 500 μL/well of DMEM was gently overlaid as feeding medium. After culturing at 37°C with 5% CO2 for 2 weeks, colonies were scored and photographed.

Construction of c-Myb target gene promoter plasmids for luciferase-based reporter assays

PCR-amplified promoter fragment of c-Myb target genes was subcloned into pGL4.10 plasmid (Promega). Primer sequences for PCR amplification of promoter region are shown in Supplementary Table S3. For FoxM1, the promoter reporter clone for human FoxM1 (HPRM13918) was purchased from GeneCopoeia, and the BglII/HindIII digested FoxM1 promoter fragment was inserted to pGL4.10.

Luciferase-based reporter assays

In the current study, luciferase-based reporter assays were conducted for assessing (i) c-Myb target promoter activity, (ii) NF-κB activity, and (iii) HTLV-1 LTR activity. HEK293T cells were seeded at the concentration of 8 × 104 cells/mL and 200 mL/well in 48-well culture plate 1 day before transfection. At 24 hours after seeding, 10 ng reporter plasmid (c-Myb target promoter-pGL4.10, 6× NF-κB–binding sites pGL4.10, or HTLV-1-LTR-pGL4.10), 5 ng RSV-Renilla-luciferase plasmid, and 200 ng c-Myb-pCDNA3 for a well were cotransfected by Lipofectamine 2000 (Invitrogen). At 24 hours after transfection, Renilla and firefly luciferase activities were measured using the Dual Luciferase Assay System (Promega) with the Centro LB 960 luminometer (Berthold Technologies). Detected firefly luciferase activity was divided by corresponding Renilla-luciferase activity to normalize transfection efficiency.

c-Myb knockdown by sh-c-Myb transduced by recombinant lentiviruses

In the current study, two shRNAs targeting 3′ UTR and exon 8 of MYB mRNA were prepared. Two sets of sense- and antisense-DNA oligos targeting 3′ UTR and exon8 of MYB mRNA were annealed by boiling for 5 minutes followed by cooling down in room temperature for more than 1 hour. Annealed fragments were inserted at BglII/XbaI in pENT4-H1 vector (Invitrogen). The sh-c-Myb fragment in pENT4-H1 was further transferred to the destination vector, CS-RfA-EvBsd, by incubation with LR-clonaseII. For preparation of sh-Myb lentiviruses, 293FT cells were seeded at 1 × 105 cells/mL and 10 mL/10-cm dish. At 24 hours after seeding, 17 ng sh-Myb-#1 or -#2- CS-RfA-EvBsd, 10 ng CAG-HIV-gp, and 10 ng pCMV-VSV-G/RSV-Rev were cotransfected by phosphate-Ca2+ method. At 72 hours after transfection, the culture medium was filtrated through 45-μm filter, and recombinant lentiviruses were precipitated by centrifugation at 10,000 rpm for 3 hours at 4°C. Precipitated viruses were resuspended in 100 μL of RPMI (FBS) and stored at −80°C until use.

For infection, 2 × 106 cells of Jurkat, TL-Om1, or primary ATL cells were resuspended in 100 μL sh-Myb-lentivirus stock and processed for centrifugation at 2000 rpm for 3 hours at 35°C to enhance infection. After centrifugation, cells were resuspended in 2 mL of RPMI and incubated at 37°C with 5% CO2 for 72 hours before being used for experiments. Sh-c-Myb oligos used in the current study are shown in Supplementary Table S4.

FACS analyses

Cells for FACS analyses were collected and resuspended in FACS buffer (PBS containing 2% FBS) prior to analysis. For apoptosis assays, cells were stained with PE-Annexin V in the binding buffer (all from PE AnnexinV Apoptosis Detection Kit I, #559763, BD Pharmingen) for 20 minutes at room temperature before analyses. All FACS analyses were conducted with FACSCalibur (BD Biosciences), with appropriate instrument settings and compensation, if necessary. Obtained data were analyzed by FlowJo ver.10 (TreeStar Inc.).

c-Myb-9A–specific downregulation by morpholino oligos

On the basis of the report by Marcos (25), which thoroughly examined and reviewed employment of morpholino oligos for regulation of alternative splicing, we designed FITC-conjugated 2 morpholino oligos, targeting the exon9A/intoron9 border (MYB-MO-#1) and exon9B/intron10 border (MYB-MO-#2) of MYB mRNA, respectively, with assistance from GENE TOOLS, LLC. The custom morpholino oligos, as well as the standard oligos (i.e., the control MO) were purchased from GENE TOOLS, LLC. The morpholino oligos were introduced at 10 μmol/L concentration to Jurkat, TL-Om1, MT-2 cells, and primary ATL cells at concentration of 1 × 106 cells in 400 μL Cytomix buffer (25 mmol/L HEPES, 120 mmol/L KCl, 10 mmol/L KPO4, 2 mmol/L EGTA, 0.15 mmol/L CaCl2, 5 mmol/L MgCl, pH 7.6, by KOH) by electroporation (220∼250 V, 1,050 F, 720 Ω) using Electro Cell Manipulator 600 (BTX). Morpholino oligo sequences are shown in Supplementary Table S5.

Statistical analyses

Throughout the current study, two-tailed paired Student t test was performed to test the statistical difference between the experimental groups. Asterisks and sharps in the figures indicate a significant difference between the tested groups (*, #, P < 0.05; **, ##, P < 0.01; and ***, ###, P < 0.001; n > 3).

ATL cells overexpress c-Myb

As shown in Fig. 1A, seven well-known transcript variants (NCBI: U22376.1, AJ606324.1) and isoforms of c-Myb have been reported [see the work of O'Rourke and Ness (ref. 8) for a detailed structure of each variant and encoded isoform]. First, the expression level of each MYB transcript variant was assessed using a qPCR analysis with variant-specific primers. All tested transcript variants, including MYB-9A and -10A, were overexpressed in ATL cells, particularly those isolated from patients with chronic and acute ATL (Fig. 1B; data not shown for MYB-8A, -9B, -13A, and -14A mRNAs). Moreover, Western blotting with a c-Myb–specific antibody revealed significantly increased c-Myb protein levels in ATL cells compared with normal CD4+ T cells (Fig. 1C).

Figure 1.

Overexpression of oncogenic MYB transcript variants in ATL cells. A, Seven well-known transcripts of c-Myb caused by alternative exon usages and their encoding isoforms (NCBI: U22376.1, AJ606324.1). B,MYB variant–specific qPCR revealed significant overexpression of total MYB, MYB-9A, and -10A mRNAs in ATL cells, especially from patients with chronic and acute ATL, relative to normal CD4+ T cells (N: normal, n = 7; S: smoldering-type ATL, n = 4; C: chronic-type ATL, n = 4; A: acute-type ATL, n = 4; *, P < 0.05; **, P < 0.01; ***, P < 0.001). C, Western blotting indicates elevated c-Myb protein levels in ATL cells from patients (C5: chronic-type ATL, A5–A7: acute-type ATL) compared with normal CD4+ T cells (N8–N10). The graph shows the integrated density values of each band (c-Myb/β-actin). The mean intensity of the c-Myb bands was significantly higher in ATL primary cells than in normal cells.

Figure 1.

Overexpression of oncogenic MYB transcript variants in ATL cells. A, Seven well-known transcripts of c-Myb caused by alternative exon usages and their encoding isoforms (NCBI: U22376.1, AJ606324.1). B,MYB variant–specific qPCR revealed significant overexpression of total MYB, MYB-9A, and -10A mRNAs in ATL cells, especially from patients with chronic and acute ATL, relative to normal CD4+ T cells (N: normal, n = 7; S: smoldering-type ATL, n = 4; C: chronic-type ATL, n = 4; A: acute-type ATL, n = 4; *, P < 0.05; **, P < 0.01; ***, P < 0.001). C, Western blotting indicates elevated c-Myb protein levels in ATL cells from patients (C5: chronic-type ATL, A5–A7: acute-type ATL) compared with normal CD4+ T cells (N8–N10). The graph shows the integrated density values of each band (c-Myb/β-actin). The mean intensity of the c-Myb bands was significantly higher in ATL primary cells than in normal cells.

Close modal

c-Myb-9A exhibits strong transforming activity

Like the v-Myb oncoprotein, c-Myb-9A is expected to exhibit strong transforming activity. In soft agar assays using NIH3T3 cells engineered to overexpress various c-Myb isoforms (Fig. 2A), c-Myb-9A exhibited the strongest colony formation activity, whereas WT-c-Myb did not exhibit significant transforming activity (Fig. 2B).

Figure 2.

c-Myb-9A exhibits a strong transforming activity. A, Schematic presentation of the soft agar assay employed in the current study. Right, overexpression of c-Myb isoforms in NIH3T3 cells from the soft agar assay. B, c-Myb-9A induced the strongest colony formation activity, whereas WT-c-Myb did not have such a strong influence on NIH3T3 cells. The graph indicates the numbers of colonies per well from three independent experiments (mean ± SD, n = 3; *, P < 0.05).

Figure 2.

c-Myb-9A exhibits a strong transforming activity. A, Schematic presentation of the soft agar assay employed in the current study. Right, overexpression of c-Myb isoforms in NIH3T3 cells from the soft agar assay. B, c-Myb-9A induced the strongest colony formation activity, whereas WT-c-Myb did not have such a strong influence on NIH3T3 cells. The graph indicates the numbers of colonies per well from three independent experiments (mean ± SD, n = 3; *, P < 0.05).

Close modal

c-Myb knockdown induces cell death

The impact of c-Myb overexpression on cell growth was further evaluated by knocking down total c-Myb at the protein level. Jurkat and TL-Om1 cells were infected with recombinant lentiviruses to induce the overexpression of shRNAs against all MYB transcripts. The top panels of Fig. 3A show that both shRNAs effectively knocked down c-Myb at the protein level. Subsequently, Venus (+) cell populations (i.e., shRNA-expressing populations) were assessed for approximately 2 weeks after infection. We observed a specific reduction in Venus (+) cell populations, indicating selective cell death in c-Myb knockdown cells (Fig. 3A). Primary ATL cells were also subjected to c-Myb knockdown (n = 4). Again, both shRNAs effectively suppressed c-Myb protein expression (Fig. 3B, top left). The proportions of Annexin V (+) populations increased, particularly in response to sh-c-Myb-#2, indicating the induction of apoptosis following c-Myb knockdown in primary ATL cells (Fig. 3B). It may be noteworthy that the protein levels of both WT-c-Myb and c-Myb-9A were reduced following treatment with the shRNAs described herein (Fig. 3B).

Figure 3.

c-Myb knockdown induces cell death. A, c-Myb–dependent cell survival was assessed in Jurkat and TL-Om1 cells engineered to overexpress c-Myb. Both sh-c-Myb-#1 and -#2 effectively downregulated c-Myb protein expression in Jurkat and TL-Om1. Moreover, FACS analyses revealed a decreased proportion of Venus (+) cells over time, indicating the selective cell death of c-Myb–knockdown cells. The data are representative of three independent experiments. B, c-Myb knockdown in primary ATL cells from the PBMCs of four patients with ATL (ATL#1–#4). sh-c-Myb-#1 and -#2 effectively downregulated the expression of both WT-c-Myb and c-Myb-9A proteins. FACS analyses of cells from three patients with ATL revealed significant increases in the proportions of Annexin V (+) cells among c-Myb knockdown cells, particularly those treated with sh-c-Myb-#2. The graph indicates the relative numbers of apoptotic cells among ATL cells treated with the indicated shRNA (means ± SD, n = 3; *, P < 0.05).

Figure 3.

c-Myb knockdown induces cell death. A, c-Myb–dependent cell survival was assessed in Jurkat and TL-Om1 cells engineered to overexpress c-Myb. Both sh-c-Myb-#1 and -#2 effectively downregulated c-Myb protein expression in Jurkat and TL-Om1. Moreover, FACS analyses revealed a decreased proportion of Venus (+) cells over time, indicating the selective cell death of c-Myb–knockdown cells. The data are representative of three independent experiments. B, c-Myb knockdown in primary ATL cells from the PBMCs of four patients with ATL (ATL#1–#4). sh-c-Myb-#1 and -#2 effectively downregulated the expression of both WT-c-Myb and c-Myb-9A proteins. FACS analyses of cells from three patients with ATL revealed significant increases in the proportions of Annexin V (+) cells among c-Myb knockdown cells, particularly those treated with sh-c-Myb-#2. The graph indicates the relative numbers of apoptotic cells among ATL cells treated with the indicated shRNA (means ± SD, n = 3; *, P < 0.05).

Close modal

c-Myb-9A–specific knockdown induces apoptosis

Thus far, our results demonstrate that c-Myb is responsible for sustainable ATL cell growth. To assess the effects of the overexpression of c-Myb-9A, which harbors strong transforming activity, we attempted a c-Myb-9A–specific knockdown using morpholino oligomers. We tested 2 FITC-labeled morpholino oligomers (Supplementary Table S5) and found that MYB-MO-#2 effectively suppressed the expression of both c-Myb-9A and -9B (see Supplementary Fig. S1C). Consequently, we employed MYB-MO-#2 for the specific suppression of c-Myb-9A (Fig. 4A). First, we introduced MYB-MO-#2 to Jurkat, TL-Om1, and MT-2 cells. Exon-specific qPCR confirmed significant decreases in MYB-9A mRNA levels in all cell lines following MYB-MO-#2 treatment (Fig. 4B). FACS analyses of apoptotic cells demonstrated that a significantly higher proportion of FITC (+) cells (i.e., c-Myb-9A knockdown cells) were also Annexin V (+) compared with control Morpholino oligomer–transfected cells (Fig. 4B). Figure 4C demonstrates the influence of MYB-MO-#2–mediated c-Myb-9A–specific knockdown in primary ATL cells (n = 4). FACS analyses demonstrated that c-Myb-9A–specific knockdown also induced apoptosis in primary ATL cells. These results provide evidence for a major role of c-Myb-9A overexpression in ATL cell survival.

Figure 4.

c-Myb-9A–specific knockdown using morpholino oligos (MO) induces apoptosis. A, Schematic design of MYB-MO-#2 against the MYB gene. B, c-Myb-9A–specific knockdown using MYB-MO-#2 in Jurkat, TL-Om1, and MT-2 cells. Exon-specific qPCR confirmed the successful reduction in MYB-9A mRNA levels (mean ± SD, n > 3; *, P < 0.05; ***, P < 0.001). FACS analyses demonstrated that c-Myb-9A–specific knockdown induced apoptosis at a significantly higher rate compared with control MO–transfected cells. The graphs demonstrate the fold increase in Annexin V (+) cells among MYB-MO-#2–transfected cells relative to control MO–transfected cells (means ± SD, n > 3; *, P < 0.05; **, P < 0.01; ***, P < 0.001). C, c-Myb-9A–specific knockdown by MYB-MO-#2 in primary ATL cells from four patients with ATL (ATL#5–#8). Exon-specific qPCR confirmed the successful reduction in MYB-9A mRNA levels (means ± SD, n = 4; **, P < 0.01). FACS analyses demonstrated a significant increase in Annexin V (+) cells among c-Myb-9A–knockdown cells compared with control cells. Graphs demonstrate the fold increase in Annexin V (+) cells among MYB-MO-#2–transfected cells compared with control MO–transfected cells (means ± SD, n = 4; *, P < 0.05).

Figure 4.

c-Myb-9A–specific knockdown using morpholino oligos (MO) induces apoptosis. A, Schematic design of MYB-MO-#2 against the MYB gene. B, c-Myb-9A–specific knockdown using MYB-MO-#2 in Jurkat, TL-Om1, and MT-2 cells. Exon-specific qPCR confirmed the successful reduction in MYB-9A mRNA levels (mean ± SD, n > 3; *, P < 0.05; ***, P < 0.001). FACS analyses demonstrated that c-Myb-9A–specific knockdown induced apoptosis at a significantly higher rate compared with control MO–transfected cells. The graphs demonstrate the fold increase in Annexin V (+) cells among MYB-MO-#2–transfected cells relative to control MO–transfected cells (means ± SD, n > 3; *, P < 0.05; **, P < 0.01; ***, P < 0.001). C, c-Myb-9A–specific knockdown by MYB-MO-#2 in primary ATL cells from four patients with ATL (ATL#5–#8). Exon-specific qPCR confirmed the successful reduction in MYB-9A mRNA levels (means ± SD, n = 4; **, P < 0.01). FACS analyses demonstrated a significant increase in Annexin V (+) cells among c-Myb-9A–knockdown cells compared with control cells. Graphs demonstrate the fold increase in Annexin V (+) cells among MYB-MO-#2–transfected cells compared with control MO–transfected cells (means ± SD, n = 4; *, P < 0.05).

Close modal

Determination of c-Myb target genes by ChIP assays

To evaluate the influence of c-Myb deregulation on transcriptional regulation, a series of ChIP assays was conducted to identify c-Myb target genes. Figure 5A presents the results of ChIP assays performed against endogenous c-Myb in Jurkat and TL-Om1 cells. Notably, c-Myb accumulated on the promoter regions of the tested genes. Consistent with the possibility that c-Myb may regulate those genes, the levels of the associated transcripts, except for IL6 mRNA (Supplementary Fig. S2), were upregulated in patients with ATL (n = 52), compared with healthy donors (n = 21), as determined in our laboratory using gene expression microarray analyses (GSE33615). These data indicate that c-Myb acts as a positive regulator of these target genes in ATL cells.

Figure 5.

Confirmation of c-Myb target genes using ChIP and promoter reporter assays. A, ChIP assays of Jurkat and TL-Om1 cells demonstrated the accumulation of c-Myb on the promoter regions of the tested genes (indicated in graphs). Little or no accumulation of c-Myb was observed on the GAPDH promoter region. B, c-Myb isoform–specific ChIP assays were conducted using 293FT cells that transiently overexpressed FLAG-tagged WT-c-Myb, c-Myb-9A, or c-Myb-10A. Mock-transfected cells (transfection with empty vector) were prepared as a negative control. c-Myb-9A was enriched on the tested target promoters at significantly higher levels compared with WT-c-Myb. The data are representative of three independent ChIP assays. C, Luciferase-based promoter reporter plasmids were prepared and used to analyze the transactivity of c-Myb isoforms (WT-c-Myb, c-Myb-9A, and c-Myb-10A) in 293T cells that transiently overexpressed these c-Myb isoforms. Mock-transfected cells (transfection with empty vector) were prepared as a negative control. For all tested promoters, WT-c-Myb exhibited a significantly higher level of reporter activity compared with the mock control. c-Myb-9A induced the highest level of promoter transactivation (n = 6, means ± SD; **, P < 0.01; ***, P < 0.001 compared with the mock control; #, P < 0.05; ##, P < 0.01, and ###, P < 0.001 compared with WT-c-Myb).

Figure 5.

Confirmation of c-Myb target genes using ChIP and promoter reporter assays. A, ChIP assays of Jurkat and TL-Om1 cells demonstrated the accumulation of c-Myb on the promoter regions of the tested genes (indicated in graphs). Little or no accumulation of c-Myb was observed on the GAPDH promoter region. B, c-Myb isoform–specific ChIP assays were conducted using 293FT cells that transiently overexpressed FLAG-tagged WT-c-Myb, c-Myb-9A, or c-Myb-10A. Mock-transfected cells (transfection with empty vector) were prepared as a negative control. c-Myb-9A was enriched on the tested target promoters at significantly higher levels compared with WT-c-Myb. The data are representative of three independent ChIP assays. C, Luciferase-based promoter reporter plasmids were prepared and used to analyze the transactivity of c-Myb isoforms (WT-c-Myb, c-Myb-9A, and c-Myb-10A) in 293T cells that transiently overexpressed these c-Myb isoforms. Mock-transfected cells (transfection with empty vector) were prepared as a negative control. For all tested promoters, WT-c-Myb exhibited a significantly higher level of reporter activity compared with the mock control. c-Myb-9A induced the highest level of promoter transactivation (n = 6, means ± SD; **, P < 0.01; ***, P < 0.001 compared with the mock control; #, P < 0.05; ##, P < 0.01, and ###, P < 0.001 compared with WT-c-Myb).

Close modal

c-Myb-9A accumulates on target genes at significantly higher levels compared with WT-c-Myb

Among the well-known c-Myb isoforms, c-Myb-9A has the highest level of transactivity (8). We speculate that this increased transactivity might be attributable to the greater recruitment of c-Myb-9A to target regions compared with other isoforms. Therefore, we prepared plasmids that would express FLAG-tagged WT-c-Myb, c-Myb-9A, and c-Myb-10A and subjected 293FT cells overexpressing FLAG-c-Myb proteins to ChIP assays using a FLAG antibody. FLAG-c-Myb-9A was indeed detected at significantly higher levels on the tested target promoters compared with FLAG-WT-c-Myb (Fig. 5B). Furthermore, the accumulation of c-Myb on some target genes was enhanced under conditions of DNA hypomethylation (Supplementary Fig. S3).

c-Myb-9A exhibits higher target gene transactivation compared with WT-c-Myb

Using a luciferase-based promoter reporter system for the indicated c-Myb target genes, we tested the activation of these reporters with the following c-Myb isoforms: WT, 9A, and 10A (Fig. 5C). Consistent with the ChIP assays (Fig. 5A and B), WT-c-Myb exhibited significantly higher reporter activities relative to the mock control on all tested promoters. Moreover, c-Myb-9A and -10A exhibited significantly higher transactivation of these promoters compared with WT-c-Myb. In addition, c-Myb-9A exhibited the highest transactivity for HTLV-1 LTR among the 7 c-Myb isoforms (Supplementary Fig. S4), which is known to be regulated by c-Myb (26).

c-Myb activates NF-κB, which is overruled by c-Myb-9A

Thus far, our results indicate that c-Myb upregulates several genes, the products of which play critical regulatory roles in cell proliferation. In addition, c-Myb-9A exhibits a hyperactive capacity for transactivation compared with WT-c-Myb. Among the tested c-Myb target genes, NIK, FoxM1, and AURKA are positive regulators of the NF-κB pathway. Thus, c-Myb might influence cellular NF-κB activity by regulating these components. Indeed, the activation of NF-κB by WT-c-Myb was confirmed using NF-κB reporter assays (Fig. 6A) and immunocytochemical analysis of activated p65 (Supplementary Fig. S5). Moreover, c-Myb-9A induced the highest level of NF-κB pathway activity among the tested c-Myb isoforms (Fig. 6A). In agreement with these results, both the protein and mRNA levels of NIK, FoxM1, and AURKA, together with the protein level of P-IκBα, were decreased following c-Myb knockdown in Jurkat and TL-Om1 cells (Fig. 6B and Supplementary Fig. S6). Conversely, the overexpression of c-Myb-9A in Jurkat cells increased the expression levels of those proteins, as well as of downstream NF-κB components such as NF-κB2-p52, NF-κB1-p50, and P-IκBα (Fig. 6B). Finally, we used an EMSA to test NF-κB activities in nuclear extracts from c-Myb overexpressing or knocked down Jurkat and TL-Om1 cells. In agreement with the data in Fig. 6B, NF-κB activity was increased most strongly in response to the overexpression of c-Myb-9A, followed by c-Myb-10A. In contrast, NF-κB activity was suppressed in Jurkat and TL-Om1 cells following c-Myb knockdown (Fig. 6C).

Figure 6.

c-Myb–induced NF-κB pathway activation is overruled by c-Myb-9A. A, NF-κB pathway activation in response to c-Myb isoforms (WT-c-Myb, c-Myb-8A, -9A, -9B, -10A, -13A, and -14A) was assessed using luciferase-based NF-κB reporter assays. Bottom, ectopic expression of c-Myb isoforms in 293T cells. WT-c-Myb transactivated the NF-κB reporter at a significantly higher level relative to the mock-transfected control. Moreover, c-Myb-9A, -10A, -13A, and -14A exhibited significantly higher reporter activity compared with WT-c-Myb, and c-Myb-9A exhibited the highest level of activity (means ± SD, n = 6; ***, P < 0.001 compared with the mock-transfected control; ##, P < 0.01; ###, P < 0.001 compared with WT-c-Myb). B, Protein levels of NIK, FoxM1, AURKA, and EZH2, as well as P-IkBα, were decreased following c-Myb knockdown in Jurkat cells. Compared with WT-c-Myb–expressing cells, the protein levels of NIK, FoxM1, AURKA, and EZH2 were most strongly increased in Jurkat cells overexpressing c-Myb-9A, followed by c-Myb-10A. The expression levels of downstream NF-κB pathway components, such as NF-κB2-p52, NF-κB1-p50, and P-IκBα, were also increased in c-Myb-9A–overexpressing cells. The data are representative of three independent experiments. C, NF-κB activity in nuclear extracts from c-Myb–overexpressing or knocked down Jurkat and TL-Om1 cells was assessed via EMSA. NF-κB activity increased most strongly in response to the overexpression of c-Myb-9A, followed by c-Myb-10A. In contrast, NF-κB activity was suppressed by c-Myb knockdown in both Jurkat and TL-Om1 cells. Jurkat cells were treated with TNFα (10 μmol/L, 1 hour) as a positive control. Data are representative of more than two independent experiments. D, Summary of c-Myb deregulation and its effects on ATL cells. The expression levels of total c-Myb and c-Myb-9A increase during the leukemogenesis and progression of ATL. c-Myb-9A, which lacks a negative regulatory domain, exhibited significantly higher transactivity against c-Myb target genes encoding proteins such as FoxM1, AURKA, and NIK and a strong transforming activity relative to WT-c-Myb. These proteins are positive regulators of cell proliferation and NF-κB activity; accordingly, the overexpression of c-Myb-9A can accelerate multiple disorders in cellular pathways, thus providing a basis for cell proliferation, constitutive NF-κB activation, and transformation in ATL cells.

Figure 6.

c-Myb–induced NF-κB pathway activation is overruled by c-Myb-9A. A, NF-κB pathway activation in response to c-Myb isoforms (WT-c-Myb, c-Myb-8A, -9A, -9B, -10A, -13A, and -14A) was assessed using luciferase-based NF-κB reporter assays. Bottom, ectopic expression of c-Myb isoforms in 293T cells. WT-c-Myb transactivated the NF-κB reporter at a significantly higher level relative to the mock-transfected control. Moreover, c-Myb-9A, -10A, -13A, and -14A exhibited significantly higher reporter activity compared with WT-c-Myb, and c-Myb-9A exhibited the highest level of activity (means ± SD, n = 6; ***, P < 0.001 compared with the mock-transfected control; ##, P < 0.01; ###, P < 0.001 compared with WT-c-Myb). B, Protein levels of NIK, FoxM1, AURKA, and EZH2, as well as P-IkBα, were decreased following c-Myb knockdown in Jurkat cells. Compared with WT-c-Myb–expressing cells, the protein levels of NIK, FoxM1, AURKA, and EZH2 were most strongly increased in Jurkat cells overexpressing c-Myb-9A, followed by c-Myb-10A. The expression levels of downstream NF-κB pathway components, such as NF-κB2-p52, NF-κB1-p50, and P-IκBα, were also increased in c-Myb-9A–overexpressing cells. The data are representative of three independent experiments. C, NF-κB activity in nuclear extracts from c-Myb–overexpressing or knocked down Jurkat and TL-Om1 cells was assessed via EMSA. NF-κB activity increased most strongly in response to the overexpression of c-Myb-9A, followed by c-Myb-10A. In contrast, NF-κB activity was suppressed by c-Myb knockdown in both Jurkat and TL-Om1 cells. Jurkat cells were treated with TNFα (10 μmol/L, 1 hour) as a positive control. Data are representative of more than two independent experiments. D, Summary of c-Myb deregulation and its effects on ATL cells. The expression levels of total c-Myb and c-Myb-9A increase during the leukemogenesis and progression of ATL. c-Myb-9A, which lacks a negative regulatory domain, exhibited significantly higher transactivity against c-Myb target genes encoding proteins such as FoxM1, AURKA, and NIK and a strong transforming activity relative to WT-c-Myb. These proteins are positive regulators of cell proliferation and NF-κB activity; accordingly, the overexpression of c-Myb-9A can accelerate multiple disorders in cellular pathways, thus providing a basis for cell proliferation, constitutive NF-κB activation, and transformation in ATL cells.

Close modal

In this study, we have demonstrated for the first time the abnormal and unbalanced MYB transcript overexpression in ATL cells. We have demonstrated increases in the expression of not only overall c-Myb but also of c-Myb-9A, an isoform with a “gain -of-function oncogenic characteristics,” in ATL cells. In tumors, MYB gene overexpression and its influence on cellular homeostasis have been extensively studied (27–35). Conversely, limited information is available regarding the expression profiles of c-Myb isoforms, each of which possesses a considerably different transactivation capacity and transforming activity. In the current study, we explored the molecular impacts of c-Myb dysregulation consequent to unbalanced c-Myb isoform expression on the malignancy of ATL cells.

A variant-specific qPCR analysis of MYB mRNA revealed increased expression levels of all tested variants (data not shown for MYB-8A, 9B, 13A, and 14A) in patients with ATL compared with healthy volunteers; this tendency was most significant for MYB-9A mRNA (Fig. 1B). In addition, the magnitude of increased expression of this transcript variant was even greater in ATL cells from patients with chronic- and acute-type ATL compared with those from patients with smoldering-type ATL. In other words, c-Myb-9A levels might correlate with the risk of ATL progression. In contrast to WT-c-Myb, c-Myb-9A exhibited strong transforming activity in soft agar assays that used NIH-3T3 cells as an indicator of transformation (Fig. 2), thus confirming the “gain-of-function oncogenic characteristics” of this isoform.

The mechanism of MYB-9A mRNA overexpression has not yet been elucidated. We recently reported that abnormal splicing patterns of HELIOS mRNA, another hematopoietic transcription factor, were observed frequently in ATL cells (20). Moreover, we reported the drastic dysregulation of miRNA expression in ATL cells (21). Significant downregulation of miR-150, a well-known negative regulator of MYB mRNA expression (36), was observed in our previous miRNA microarray analysis of patients with ATL (GSE31629). Changes in transcriptional activity, mRNA stability, and splicing preferences might contribute to the overexpression of oncogenic MYB transcript variants.

The overall suppression of c-Myb–induced death in both ATL cell lines and primary ATL cells (Fig. 3). Moreover, the specific morpholino oligomer–induced knockdown of c-Myb-9A resulted in a significant increase in apoptotic cell rates in T-cell lines, regardless of HTLV-1 infection status, as well as primary ATL cells (Fig. 4). These data suggest that c-Myb-9A plays a crucial role in cell survival in c-Myb–overexpressing cells, including primary ATL cells.

Extensive attempts have been made to identify c-Myb target genes (14, 15, 37–40). A list of hundreds of putative c-Myb target genes was generated using a combination of c-Myb knockdown and gene expression microarray analyses. In the current study, we conducted ChIP analyses of nine putative c-Myb target genes that encode regulators of cell proliferation. Figure 5A demonstrates the enrichment of c-Myb on the promoters of all tested target genes. A gene expression microarray analysis (GSE33615) demonstrated that the expression levels of all tested c-Myb target genes, except for IL6 mRNA, were elevated in ATL cells (Supplementary Fig. S2), further supporting the notion that c-Myb is a positive regulator of these genes in ATL cells.

c-Myb-9A, which lacks a C-terminal NRD and is structurally similar to the v-Myb AMV oncoprotein, is known to exhibit a significantly higher transactivation capacity relative to that of WT-c-Myb (8). We wondered whether this higher transactivation capacity could be attributed to enhanced recruitment to target promoters. In a series of previous studies, C-terminal deletion mutants of c-Myb led to significant increases in DNA-binding activity, transactivation, and transformation activity, indicating the importance of the NRD in the negative self-regulation of c-Myb (41–45). Molvaersmyr and colleagues identified SUMOylation sites for degradation within the NRD (46). Our ChIP assays demonstrated significantly higher levels of FLAG-c-Myb-9A enrichment on the tested target promoters compared with FLAG-WT-c-Myb enrichment (Fig. 5B). In agreement with the ChIP assay results, luciferase-based promoter reporter assays revealed significantly higher levels of promotor transactivation with c-Myb-9A than with WT-c-Myb (Fig. 5C and Supplementary Fig. S4). Furthermore, c-Myb was generally capable of accessing the target regions more efficiently under conditions of Aza-dC treatment–induced DNA hypomethylation; under the same conditions, c-Myb-9A exhibited even more efficient recruitment, especially to GATA3, FOXM1, AURKA, EZH2, and IL-2Rα (Supplementary Fig. S3). In agreement with our results, Wang and colleagues demonstrated that WNT5A gene hypomethylation led to increased c-Myb recruitment (47). DNA methylation is a known marker of epigenetic silencing (48). Therefore, c-Myb-9A might have a stronger impact when the target gene is not silenced. Collectively, these data led us to speculate that the enhanced transactivation capacity of c-Myb-9A leads to the overexpression of c-Myb target genes in cells with elevated c-Myb-9A expression.

Finally, we revealed that c-Myb plays an important role in NF-κB pathway activation. Of the c-Myb target gene products evaluated in the current study, NIK is a key positive regulator of the noncanonical NF-κB pathway (49). AURKA is known to phosphorylate IκBα, leading to its degradation and activating NF-κB (50). FoxM1 transactivates AURKA gene expression to a similar extent as c-Myb and is also known as a strong positive regulator of cell cycle and cell proliferation (51). Recently, c-Myb and FoxM1 were found to share a considerable number of target genes and are expected to function synergistically (51). Constant activation of the NF-κB pathway, as well as its pro-oncogenic influences, has been well-documented and reviewed in ATL cells (52). Figure 6A shows that c-Myb indeed activates the NF-κB pathway and that c-Myb-9A induces the highest level of activation. In Fig. 6B and Supplementary Fig. S6, we demonstrate that NIK, FoxM1, and AURKA protein/mRNA levels, as well as the levels of downstream NF-κB components, were regulated in a manner dependent on the c-Myb expression level. In particular, c-Myb-9A overexpression enhanced the expression levels of the abovementioned proteins in Jurkat cells (Fig. 6B). In c-Myb-9A–overexpressing cells, the molecular impact of c-Myb-9A might be accelerated in a positive feedback manner by the activation of c-Myb itself, as well as synergistic activation between the c-Myb and FoxM1 pathways. This dysregulation of critical transcription factors at multiple stages might be responsible for the constitutive and drastic NF-κB activation observed in ATL cells. Using EMSA, we ultimately tested whether a direct relationship existed between the c-Myb expression level and NF-κB activity level in Jurkat and ATL-derived TL-Om1 cells. Figure 6C shows that the overexpression of c-Myb, especially c-Myb-9A, activates NF-κB in Jurkat cells, whereas shRNA-mediated c-Myb knockdown suppressed NF-κB activity in both Jurkat and TL-Om1 cells. Altogether, these data suggest that c-Myb plays a critical role in the regulation of NF-κB activity by controlling the expression of proteins involved in the NF-κB pathway, such as FoxM1, AURKA, and NIK. Moreover, it is highly possible that in the absence of a self-regulatory domain, c-Myb-9A can overproduce these gene products and thus hyperactivate downstream NF-κB activity compared with WT-c-Myb (Fig. 6D).

In summary, this study highlights for the first time the impact of c-Myb dysregulation on the malignant characteristics of ATL cells. Moreover, we have revealed that the aberrantly expressed oncogenic c-Myb isoform, c-Myb-9A, is responsible for extensive c-Myb pathway dysregulation. Our current study data suggest that the expression levels of c-Myb isoforms, including c-Myb-9A, are increased during the leukemogenesis and progression of ATL. This overexpression of c-Myb can trigger the dysregulation of cellular homeostasis; c-Myb-9A, which lacks self-regulatory machinery, further augments this disorder by inducing the undesirable overexpression of cellular proteins involved in cell proliferation, NF-κB activation, and transformation (Fig. 6D). Because the c-Myb pathway is profoundly involved in the regulation of T-cell function and composition, dysregulation of this pathway may be an effective therapeutic target for ATL. Further investigations to elucidate the molecular basis of c-Myb pathway dysregulation in ATL cells will be fundamental to the establishment of an effective therapeutic approach.

No potential conflicts of interest were disclosed.

Conception and design: K. Nakano, K. Yamaguchi, T. Watanabe

Development of methodology: K. Nakano, T. Watanabe

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): K. Nakano, K. Uchimaru, A. Utsunomiya, T. Watanabe

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): K. Nakano, T. Watanabe

Writing, review, and/or revision of the manuscript: K. Nakano, K. Uchimaru, A. Utsunomiya, T. Watanabe

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

Study supervision: T. Watanabe

The authors thank Dr. O'Rourke in University of New Mexico for his generous gifts of c-Myb expression plasmids. They also appreciate Dr. S. Firouzi in the University of Tokyo for the integration site analysis of MT-2 cells; Dr. H. Sato in the University of Tokyo for the karyotype analysis of HeLa and 293T cells; and Drs. H. Miyoshi and A. Miyawaki in RIKEN, Japan, for providing the Venus-encoding lentivirus vectors.

This work was supported by Grants-in-Aid for Scientific Research from the Ministry of Education, Culture, Sports, Science, and Technology of Japan, to T. Watanabe (No. 26293226 and No. 23659484) and to K. Nakano (No. 24501304).

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Westin
E
,
Gallo
R
,
Arya
S
. 
Differential expression of the amv gene in human hematopoietic cells
.
Proc Natl Acad Sci U S A
1982
;
79
:
2194
8
.
2.
Ness
SA
. 
The Myb oncoprotein: regulating a regulator
.
Biochim Biophys Acta
1996
;
1288
:
F123
39
.
3.
Greig
KT
,
Carotta
S
,
Nutt
SL
. 
Critical roles for c-Myb in hematopoietic progenitor cells
.
Semin Immunol
2008
;
20
:
247
56
.
4.
Ramsay
RG
,
Gonda
TJ
. 
MYB function in normal and cancer cells
.
Nat Rev Cancer
2008
;
8
:
523
34
.
5.
Pattabiraman
DR
,
Gonda
TJ
. 
Role and potential for therapeutic targeting of MYB in leukemia
.
Leukemia
2013
;
27
:
269
77
.
6.
Woo
C
,
Sopchak
L
,
Lipsick
J
. 
Overexpression of an alternatively spliced form of c-Myb results in increases in transactivation and transforms avian myelomonoblasts
.
J Virol
1998
;
72
:
6813
21
.
7.
Kumar
A
,
Baker
SJ
,
Lee
CM
,
Reddy
EP
. 
Molecular mechanisms associated with the regulation of apoptosis by the two alternatively spliced products of c-Myb
.
Mol Cell Biol
2003
;
23
:
6631
45
.
8.
O'Rourke
JP
,
Ness
SA
. 
Alternative RNA splicing produces multiple forms of c-Myb with unique transcriptional activities
.
Mol Cell Biol
2008
;
28
:
2091
101
.
9.
Hu
T
,
Simmons
A
,
Yuan
J
,
Bender
TP
,
Alberola-Ila
J
. 
The transcription factor c-Myb primes CD4+CD8+ immature thymocytes for selection into the iNKT lineage
.
Nat Immunol
2010
;
11
:
435
41
.
10.
Orkin
SH
,
Zon
LI
. 
Hematopoiesis: an evolving paradigm for stem cell biology
.
Cell
2008
;
132
:
631
44
.
11.
Sandberg
ML
,
Sutton
SE
,
Pletcher
MT
,
Wiltshire
T
,
Tarantino
LM
,
Hogenesch
JB
, et al
c-Myb and p300 regulate hematopoietic stem cell proliferation and differentiation
.
Dev Cell
2005
;
8
:
153
66
.
12.
Yuan
J
,
Crittenden
RB
,
Bender
TP
. 
c-Myb promotes the survival of CD4+CD8+ double-positive thymocytes through upregulation of Bcl-xL
.
J Immunol
2010
;
184
:
2793
804
.
13.
Nakata
Y
,
Shetzline
S
,
Sakashita
C
,
Kalota
A
,
Rallapalli
R
,
Rudnick
SI
, et al
c-Myb contributes to G2/M cell cycle transition in human hematopoietic cells by direct regulation of cyclin B1 expression
.
Mol Cell Biol
2007
;
27
:
2048
58
.
14.
Maurice
D
,
Hooper
J
,
Lang
G
,
Weston
K
. 
c-Myb regulates lineage choice in developing thymocytes via its target gene Gata3
.
EMBO J
2007
;
26
:
3629
40
.
15.
Nakata
Y
,
Brignier
AC
,
Jin
S
,
Shen
Y
,
Rudnick
SI
,
Sugita
M
, et al
c-Myb, Menin, GATA-3, and MLL form a dynamic transcription complex that plays a pivotal role in human T helper type 2 cell development
.
Development
2010
;
116
:
1280
90
.
16.
Iwanaga
M
,
Watanabe
T
,
Yamaguchi
K
. 
AdultT-cell leukemia: a review of epidemiological evidence
.
Front Microbiol
2012
;
3
:
Article 322
.
17.
Poiesz
BJ
,
Ruscetti
FW
,
Gazdar
AF
,
Bunn
PA
,
Minna
JD
,
Gallo
RC
. 
Detection and isolation of type C retrovirus particles from fresh and cultured lymphocytes of a patient with cutaneous T-cell lymphoma
.
Proc Natl Acad Sci U S A
1980
;
77
:
7415
9
.
18.
Poiesz
BJ
,
Ruscetti
FW
,
Reitz
MS
,
Kalyanaraman
VS
,
Gallo
RC
. 
Discovery of new type C retrovirus (HTLV-1)
.
Nature
1981
;
294
:
268
71
.
19.
Yoshida
M
,
Miyoshi
I
,
Yorio
H
. 
Isolation and characterization of retrovirus from cell lines of human adult T-cell leukemia and its implications in the disease
.
Proc Natl Acad Sci U S A
1982
;
79
:
2031
5
.
20.
Asanuma
S
,
Yamagishi
M
,
Kawanami
K
,
Nakano
K
,
Sato-Otsubo
A
,
Muto
S
, et al
Adult T-cell leukemia cells are characterized by abnormalities of Helios expression that promote T cell growth
.
Cancer Sci
2013
;
104
:
1097
106
21.
Yamagishi
M
,
Nakano
K
,
Miyake
A
,
Yamochi
T
,
Kagami
Y
,
Tsutsumi
A
, et al
Polycomb-mediated loss of miR-31 activates NIK-dependent NF-κB pathway in adult T cell leukemia and other cancers
.
Cancer Cell
2012
;
21
:
121
35
.
22.
Kuramitsu
M
,
Okuma
K
,
Yamagishi
M
,
Yamochi
T
,
Firouzi
S
,
Momose
H
, et al
Identification of TL-Om1, an adult T-cell leukemia (ATL) cell line, as reference material for quantitative PCR for human T-lymphotropic virus 1
.
J Clin Microbiol
2015
;
53
:
587
96
.
23.
Firouzi
S
,
López
Y
,
Suzuki
Y
,
Nakai
K
,
Sugano
S
,
Yamochi
T
, et al
Development and validation of a new high-throughput method to investigate the clonality of HTLV-1-infected cells based on provirus integration sites
.
Genome Med
2014
;
6
:
46
.
24.
Suzuki
K
,
Juelich
T
,
Lim
H
,
Ishida
T
. 
Closed chromatin architecture is induced by an RNA duplex targeting the HIV-1 promoter region
.
J Biol Chem
2008
;
283
:
23353
63
.
25.
Morcos
PA
. 
Achieving targeted and quantifiable alteration of mRNA splicing with Morpholino oligos
.
Biochem Biophys Res Commun
2007
;
358
:
521
7
.
26.
Dasgupta
P
,
Reddy
CD
,
Saikumar
P
,
Reddy
EP
. 
The cellular proto-oncogene product Myb acts as transcriptional activator of the long terminal repeat of human T-lymphotropic virus type I
.
J Virol
1992
;
66
:
270
6
.
27.
Clappier
E
,
Cuccuini
W
. 
The C-MYB locus is involved in chromosomal translocation and genomic duplications in human T-cell acute leukemia (T-ALL), the translocation defining a new T-ALL subtype in very young children
.
Blood
2007
;
110
:
1251
61
.
28.
Dassé
E
,
Volpe
G
,
Walton
DS
,
Wilson
N
,
Del Pozzo
W
,
O'Neill
LP
, et al
Distinct regulation of c-myb gene expression by HoxA9, Meis1 and Pbx proteins in normal hematopoietic progenitors and transformed myeloid cells
.
Blood Cancer J
2012
;
2
:
e76
.
29.
Drabsch
Y
,
Hugo
H
. 
Mechanism of and requirement for estrogen-regulated MYB expression in estrogen-receptor-positive breast cancer cells
.
Proc Natl Acad Sci U S A
2007
;
104
:
13762
7
.
30.
Jin
S
,
Zhao
H
,
Yi
Y
,
Nakata
Y
,
Kalota
A
,
Gewirtz
AM
. 
c-Myb binds MLL through menin in human leukemia cells and is an important driver of MLL-associated leukemogenesis
.
J Clin Inv
2010
;
120
:
593
606
.
31.
Lidonnici
MR
,
Corradini
F
,
Waldron
T
,
Bender
TP
,
Calabretta
B
. 
Requirement of c-Myb for p210 BCR/ABL -dependent transformation of hematopoietic progenitors and leukemogenesis
.
Bone Marrow Transplant
2008
;
111
:
4771
9
.
32.
Manzotti
G
,
Mariani
SA
,
Corradini
F
,
Bussolari
R
,
Cesi
V
,
Vergalli
J
, et al
Expression of p89c-Mybex9b, an alternatively spliced form of c-Myb, is required for proliferation and survival of p210BCR/ABL-expressing cells
.
Blood Cancer J
2012
;
2
:
e71
.
33.
Sampurno
S
,
Bijenhof
A
,
Cheasley
D
,
Xu
H
,
Robine
S
,
Hilton
D
, et al
The Myb-p300-CREB axis modulates intestine homeostasis, radiosensitivity and tumorigenesis
.
Cell Death Dis
2013
;
4
:
e605
.
34.
Stenman
G
,
Andersson
MK
,
Andren
Y
. 
New tricks from an old oncogene: gene fusion and copy number alterations of MYB in human cancer
.
Cell Cycle
2010
;
9
:
2986
95
.
35.
Volpe
G
,
Walton
DS
,
Del Pozzo
W
,
Garcia
P
,
Dassé
E
,
O'Neill
LP
, et al
C/EBPα and MYB regulate FLT3 expression in AML
.
Leukemia
2013
;
27
:
1487
96
.
36.
Lin
Y-C
,
Kuo
M-W
,
Yu
J
,
Kuo
H-H
,
Lin
R-J
,
Lo
W-L
, et al
c-Myb is an evolutionary conserved miR-150 target and miR-150/c-Myb interaction is important for embryonic development
.
Mol Biol Evol
2008
;
25
:
2189
98
.
37.
Lei
W
,
Rushton
JJ
,
Davis
LM
,
Liu
F
,
Ness
SA
. 
Positive and negative determinants of target gene specificity in myb transcription factors
.
J Biol Chem
2004
;
279
:
29519
27
.
38.
Lorenzo
PI
,
Brendeford
EM
,
Gilfillan
S
,
Gavrilov
AA
,
Leedsak
M
,
Razin
SV
, et al
Identification of c-Myb target genes in K562 cells reveals a role for c-Myb as a master regulator
.
Genes Cancer
2011
;
2
:
805
17
.
39.
Quintana
AM
,
Liu
F
,
O'Rourke
JP
,
Ness
SA
. 
Identification and regulation of c-Myb target genes in MCF-7 cells
.
BMC Cancer
2011
;
11
:
30
.
40.
Zhao
L
,
Glazov
EA
,
Pattabiraman
DR
,
Al-Owaidi
F
,
Zhang
P
,
Brown
MA
, et al
Integrated genome-wide chromatin occupancy and expression analyses identify key myeloid pro-differentiation transcription factors repressed by Myb
.
Nucleic Acids Res
2011
;
39
:
4664
79
.
41.
Gonda
T
,
Buckmaster
C
,
Ramsay
R
. 
Activation of c-myb by carboxy-terminal truncation: relationship to transformation of murine haemopoietic cells in vitro
.
EMBO J
1989
;
8
:
1777
83
.
42.
Sakura
H
,
Kanei-Ishii
C
. 
Delineation of three functional domains of the transcriptional activator encoded by the c-myb protooncogene
.
Proc Natl Acad Sci U S A
1989
;
86
:
5758
62
.
43.
Grässer
F
,
Graf
T
,
Lipsick
J
. 
Protein truncation is required for the activation of the c-myb proto-oncogene
.
Mol Cell Biol
1991
;
11
:
3987
96
.
44.
Press
R
,
Reddy
E
,
Ewert
D
. 
Overexpression of C-terminally but not N-terminally truncated Myb induces fibrosarcomas: a novel nonhematopoietic target cell for the myb oncogene
.
Mol Cell Biol
1994
;
14
:
2278
90
.
45.
Wang
DM
,
Dubendorff
JW
,
Woo
CH
,
Lipsick
JS
. 
Functional analysis of carboxy-terminal deletion mutants of c-Myb
.
J Virol
1999
;
73
:
5875
86
.
46.
Molvaersmyr
A-K
,
Saether
T
,
Gilfillan
S
,
Lorenzo
PI
,
Kvaløy
H
,
Matre
V
, et al
A SUMO-regulated activation function controls synergy of c-Myb through a repressor-activator switch leading to differential p300 recruitment
.
Nucleic Acids Res
2010
;
38
:
4970
84
.
47.
Wang
Q
,
Williamson
M
,
Bott
S
,
Brookman-Amissah
N
,
Freeman
A
,
Nariculam
J
, et al
Hypomethylation of WNT5A, CRIP1 and S100P in prostate cancer
.
Oncogene
2007
;
26
:
6560
5
.
48.
Johns
PA
. 
Functions of DNA methylation: islands, start sites, gene bodies and beyond
.
Nat Rev Genet
2012
;
13
:
484
92
.
49.
Sun
S
. 
Non-canonical NF-κB signaling pathway
.
Cell Res
2011
;
21
:
71
85
.
50.
Briassouli
P
,
Chan
F
,
Savage
K
,
Reis-Filho
JS
,
Linardopoulos
S
. 
Aurora-A regulation of nuclear factor-kappaB signaling by phosphorylation of IkappaBalpha
.
Cancer Res
2007
;
67
:
1689
95
.
51.
Lefebvre
C
,
Rajbhandari
P
,
Alvarez
MJ
,
Bandaru
P
,
Lim
WK
,
Sato
M
, et al
A human B-cell interactome identifies MYB and FOXM1 as master regulators of proliferation in germinal centers
.
Mol Syst Biol
2010
;
6
:
377
.
52.
Sun
S-C
,
Yamaoka
S
. 
Activation of NF-kB by HTLV-I and implications for cell transformation
.
Oncogene
2005
;
24
:
5952
64
.

Supplementary data