The PML gene is frequently fused to the retinoic acid receptor α (RARα) gene in acute promyelocytic leukemia (APL), generating a characteristic PML-RARα oncogenic chimera. PML-RARα disrupts the discrete nuclear speckles termed nuclear bodies, which are formed in PML, suggesting that nuclear body disruption is involved in leukemogenesis. Nuclear body formation that relies upon PML oligomerization and its stabilization of the hypoxia-inducible protein kinase (HIPK)-2 is disrupted by expression of the PML-RARα chimera. Here, we report that disruption of nuclear bodies is also mediated by PML-RARα inhibition of PML oligomerization. PKA-mediated phosphorylation of PML-RARα blocked its ability to inhibit PML oligomerization and destabilize HIPK2. Our results establish that both PML oligomerization and HIPK2 stabilization at nuclear bodies are important for APL cell differentiation, offering insights into the basis for the most common prodifferentiation therapies of APL used clinically. Cancer Res; 73(14); 4278–88. ©2013 AACR.

In human leukemia, specific chromosomal translocations result in the expression of fusion proteins promoting malignancy (1, 2). The PML gene is the target of the t(15;17) chromosome translocation that is observed in more than 90% of acute promyelocytic leukemia (APL) cases, in which fusion of the PML and retinoic acid receptor α (RARα) genes leads to the expression of the aberrant PML-RARα fusion protein (3–6). The PML protein normally forms discrete nuclear speckles in the nucleus called PML nuclear bodies; in these, PML recruits other proteins, including transcription factors such as p53 and acute myeloid leukemia 1 (7–9), transcription coactivators such as hypoxia-inducible protein kinase (HIPK)-2 and p300 (10, 11), SUMO (12), and DAXX (13, 14). Nuclear bodies have been implicated in the regulation of apoptosis, cellular senescence, and antiviral responses. It has been reported that PML stabilizes transcription coactivators, such as HIPK2 and p300, to assemble transcription factor/coactivator complexes within nuclear bodies (15). In contrast, nuclear bodies are disrupted in t(15;17) APL (12, 16–19); in the presence of the PML-RARα fusion protein, nuclear bodies appear as dispersed microspeckles in which HIPK2 is destabilized (15). All-trans-retinoic acid (ATRA) and As2O3, which are used clinically in APL, restore the normal appearance of nuclear bodies (12, 17, 19–21). In t(15;17) APL, it remains unclear that the disruption of nuclear bodies is related to leukemogenesis and that their restoration would lead to therapy. The molecular mechanism by which PML-RARα disrupts nuclear bodies has remained elusive, hampering progress in the understanding of leukemogenesis.

The present study reveals that PML-RARα blocks PML oligomerization and disrupts nuclear bodies, and that the effect is reversed upon cAMP/PKA-dependent phosphorylation of PML-RARα. In addition, pharmacologic activation of adenylyl cyclase by forskolin restores PML nuclear bodies and promotes ATRA-induced APL cell differentiation. Furthermore, nuclear body restoration induced HIPK2 stabilization. These results suggest that nuclear body formation is regulated by PML-RARα phosphorylation, and that the restoration of nuclear bodies is important for APL cell differentiation.

Cell culture, infection, and antibodies

293FT cells, which were purchased from Invitrogen, and U2OS cells were cultured in Dulbecco's Modified Eagle Medium (DMEM) supplemented with 10% fetal calf serum (FCS). Plat-E cells were obtained from Dr. T. Kitamura (University of Tokyo, Tokyo, Japan) and were cultured in DMEM supplemented with 10% FCS, 10 μg/mL blasticidin, and 1 μg/mL puromycin. NB4 cells and K562 cells were cultured in RPMI-1640 medium supplemented with 10% FCS. Anti-HIPK2 antibody was described previously (15). Other antibodies were purchased commercially: anti-HA (3F10, Roche and Y11, Santa Cruz), anti-FLAG (M2, Sigma), anti-Myc (9E10, Upstate), anti-tubulin (H235, Santa Cruz), anti-PML (16.1–104, Upstate; H238, Santa Cruz; 001, MBL), anti-SUMO (Zymed), anti-DAXX (Exbio), and anti-RARα (C20, Santa Cruz).

Plasmids

The PML, PML-RARα, and HIPK2 expression vectors were generated as described previously (15). PML and PML-RARα deletion mutants were generated by PCR using pLNCX-HA-PML IV or pLNCX-HA-PML-RARα as the template.

Immunoprecipitation and Western blotting

293FT cells were transfected with the desired vectors and lysed as described previously (15). Cell lysates and immunoprecipitates were fractionated on SDS-polyacrylamide gels and transferred onto nitrocellulose membranes (Amersham). The membranes were incubated with primary antibodies and horseradish peroxidase-conjugated secondary antibodies. The immune complexes were visualized by the enhanced chemiluminescence (ECL) or ECL-Plus technique (Amersham), and the images were analyzed by ImageGauge (FUJIFILM).

Immunofluorescence

U2OS cells were cultured in 4-well chamber slides and transfected as described previously (15). The cells were exposed to 50 μmol/L forskolin for 18 hours. After forskolin exposure, the cells were fixed and incubated with antibodies as described previously. PML-RARα–expressing mouse c-kit+ cells were exposed to 50 μmol/L forskolin for 24 hours. NB4 cells were exposed to 50 μmol/L forskolin or 1 μmol/L ATRA for 24 or 72 hours. The cells were analyzed by the same techniques as U2OS cells. Images were captured on an Olympus microscope and analyzed by deconvolution.

Serial replating assay

pMSCV, pMSCV-HA-PML-RARα, pMSCV-HA-PML-RARα ΔE, pMSCV-HA-PML-RARα S704A, pMSCV-HA-PML-RARα S704D, and pMSCV-HA-HIPK2 were transfected into Plat-E cells using GeneJuice (Novagen), and supernatants containing retrovirus were collected 48 hours after transfection. C-kit+ cells were selected from the femurs of C57BL/6 mice using CD117-specific MicroBeads (Miltenyi Biotech), transduced with retroviruses using RetroNectin (Takara), and plated in methylcellulose medium (M3434, StemCell Technologies). The cells were cultured and replated every 4 to 6 days in methylcellulose medium under G418 selection. The cells from the first round (empty vector and PML-RARα ΔE) or the third round (PML-RARα point mutants) of colonies were harvested and analyzed by using immunofluorescence as described above. With respect to HIPK2 stabilization, c-kit+ cells were infected with retroviruses containing HA-HIPK2. The next day, the cells were infected with retroviruses containing wild-type or mutants of HA-PML-RARα. The cells were cultured in methylcellulose medium under G418 and puromycin selection. The cells of the third-round colonies were harvested and subjected to Western blot analysis.

RT-PCR

Reverse transcriptase PCR was conducted as described previously (15). HIPK2 mRNA expression was analyzed using the following primers: forward (5′-CCCCTCAAATACATTCGCCC-3′) and reverse (5′-TGGTGTCTTCAGTCTCCACAAAGG-3′). The glyceraldehyde-3-phosphate dehydrogenase primer set was described previously (15).

Flow cytometric analysis

Before incubation with anti-Mac1-FITC (M1/70, eBioscience), NB4 cells were preincubated with immunoglobulin G from rat serum (Sigma) to prevent nonspecific binding of the antibody. The stained cells were analyzed by JSAN (Bay Bioscience), and the results were analyzed using FLOWJO software.

PML oligomerization is required for nuclear body formation

The coiled-coil domain of PML mediates homo-oligomerization and hetero-oligomerization (18, 22–24). We have previously shown that PML IV interacts with 2 transcription factors important for granulocytic differentiation, PU.1 and CAAT/enhancer-binding protein (C/EBPϵ; refs. 25, 26). Therefore, we used PML IV and a PML mutant lacking the coiled-coil domain (PML IV ΔCC; Fig. 1A). As expected, PML IV ΔCC could not form homo-oligomers (Supplementary Fig. S1A). Immunofluorescence was conducted to assess the localization of the PML IV ΔCC mutant. As shown previously (21, 27), wild-type PML IV localized to the nucleus and formed nuclear bodies of normal appearance; however, the PML IV ΔCC mutant was expressed uniformly throughout the nucleus and did not form nuclear bodies (Supplementary Fig. S1B). These results suggest that PML oligomerization is associated with nuclear body formation. HIPK2 is stabilized by PML in nuclear bodies and destabilized by PML-RARα (15). To test whether the PML mutant destabilized HIPK2, FLAG-tagged HIPK2 was cotransfected with HA-tagged PML IV or PML IV ΔCC. Results showed that PML IV stabilized HIPK2, which is consistent with previous observations (15); however, PML IV ΔCC destabilized HIPK2 (Fig. 1B). The destabilization was rescued by a proteasome inhibitor, MG132 (Fig. 1B). These results suggest that PML oligomerization is required for PML-mediated HIPK2 stabilization.

Figure 1.

The coiled-coil domain of PML is required for HIPK2 stabilization. A, diagram of the PML deletion mutant. The proline-rich region (Pro), the ring-finger domain (RING), the coiled-coil domain (CC), the nuclear-import signal (NLS), and the serine-proline–rich region (SP) are indicated. B, PML IV ΔCC destabilizes HIPK2. 293FT cells were transfected with pLNCX-FLAG-HIPK2 and empty vector, pLPCX-HA-PML IV, or pLPCX-HA-ΔCC. The cells were treated with or without 10 μmol/L MG132 for 16 hours. The expression of HIPK2 (top), PML IV, or PML IV ΔCC (middle) and tubulin (bottom) was detected by immunoblotting using anti-FLAG, anti-HA and anti-tubulin antibodies, respectively.

Figure 1.

The coiled-coil domain of PML is required for HIPK2 stabilization. A, diagram of the PML deletion mutant. The proline-rich region (Pro), the ring-finger domain (RING), the coiled-coil domain (CC), the nuclear-import signal (NLS), and the serine-proline–rich region (SP) are indicated. B, PML IV ΔCC destabilizes HIPK2. 293FT cells were transfected with pLNCX-FLAG-HIPK2 and empty vector, pLPCX-HA-PML IV, or pLPCX-HA-ΔCC. The cells were treated with or without 10 μmol/L MG132 for 16 hours. The expression of HIPK2 (top), PML IV, or PML IV ΔCC (middle) and tubulin (bottom) was detected by immunoblotting using anti-FLAG, anti-HA and anti-tubulin antibodies, respectively.

Close modal

PML-RARα disrupts nuclear bodies; however, the molecular mechanism underlying this effect remains unclear. The defect in nuclear body formation by the oligomerization-deficient PML IV ΔCC mutant led to the hypothesis that PML-RARα may prevent PML oligomerization. To test this hypothesis, the effect of PML-RARα on nuclear body disruption was evaluated by immunofluorescence. Wild-type PML and PML-RARα are expressed in APL cells. The localization of PML is diffused in cells expressing PML-RARα (18). We transfected U2OS cells with expression vectors encoding FLAG-tagged PML IV and hemagglutinin (HA)-tagged PML IV or HA-tagged PML-RARα. PML nuclear bodies were detected using anti-FLAG antibody. As shown in Supplementary Fig. S2B, PML IV formed large, discrete, and distinct nuclear foci in the absence of PML-RARα (top) but small and disperse foci in the presence of PML-RARα (bottom). The oligomerization capacity of PML was subsequently assessed in the presence and absence of PML-RARα. As shown in Supplementary Fig. S2C, PML-RARα inhibited PML–PML interaction, suggesting that PML-RARα disrupts PML nuclear bodies by inhibiting PML oligomerization. The N-terminal or the coiled-coil domain of PML-RARα has been reported to be important for nuclear body disruption (28). PML 1–394, which is the PML moiety of PML-RARα and lacks the PML C-terminal, did not disrupt PML nuclear bodies (Supplementary Fig. S2D), suggesting that the RARα moiety of the fusion protein is also important for nuclear body disruption.

The ligand-binding domain of the RARα moiety of PML-RARα is essential for nuclear body disruption

Because the RARα moiety of PML-RARα is important for the disruption of PML nuclear bodies, PML-RARα deletion mutants were generated (Fig. 2A) to identify the region of RARα required for the effect. HA-tagged deletion mutants were cotransfected with FLAG-tagged PML IV. Immunofluorescence analysis indicated that PML-RARα 1–748 led to dispersed microspeckles, as did wild-type PML-RARα, but that deletion mutants lacking the ligand-binding domain, such as PML-RARα 1–567, 1–492, and 1–420, did not (Fig. 2B). These results suggest that the ligand-binding domain of RARα plays an important role in PML-RARα–mediated nuclear body disruption. The ΔE PML-RARα mutant, which lacks the ligand-binding domain (Fig. 2A), failed to disrupt nuclear bodies (Fig. 2B). The ΔC PML-RARα mutant, which lacks the DNA-binding domain, disrupted nuclear bodies (Fig. 2B). All of the PML-RARα deletion mutants were expressed at similar levels (Fig. 2C). We also tested the effect of the wild-type PML-RARα and the ΔE PML-RARα mutant on endogenous nuclear body formation in normal myeloid stem/progenitor cells. As the cells transduced with PML-RARα ΔE were not immortalized (Fig. 2D), we assayed for PML localization using the cells of first-round colonies with an anti-PML antibody specific for mouse Pml. Nuclear bodies were not disrupted in PML-RARα ΔE–infected cells (Fig. 2E). The localization of ΔE was different from that of wild-type PML-RARα. The ligand-binding domain of PML-RARα might be important for its dispersion. These results suggest that the ligand-binding domain of PML-RARα is indeed important for nuclear body disruption.

Figure 2.

The ligand-binding domain of PML-RARα is essential for nuclear body disruption. A, diagram of PML-RARα deletion mutants. B, the ligand-binding domain of PML-RARα is required for the disruption of PML nuclear bodies. U2OS cells were transfected with pLNCX-FLAG-PML IV and pLNCX-HA-PML-RARα deletion constructs or only with pLNCX-HA-PML-RARα deletion constructs as described. PML nuclear bodies were analyzed using anti-FLAG antibody. The white bar represents 10 μm. C, expression of PML-RARα deletion mutants. The expression of PML IV (top), PML-RARα deletion mutants (middle), and tubulin (bottom) in U2OS cells was detected by immunoblotting using anti-FLAG, anti-HA, and anti-tubulin antibodies, respectively. D, the cells expressing PML-RARα ΔE are not immortalized. C-kit+ mouse bone marrow cells were infected with empty vector (mock), pMSCV-HA-PML-RARα wild-type, or ΔE and cultured in methylcellulose medium. The colony number from the third to the fifth round of colonies is indicated (top). Values represent mean ± SEM from 3 independent experiments. E, PML-RARα ΔE does not inhibit nuclear body formation. C-kit+ mouse bone marrow cells were infected with empty vector, pMSCV-HA-PML-RARα wild-type, or ΔE, and plated in methylcellulose medium. Endogenous murine PML nuclear bodies of the cells at first round of colonies were analyzed using mouse Pml-specific antibody (16.1–104). The white bar represents 10 μm. E, effect of PML-RARα deletion mutants on HIPK2 stability. 293FT cells were transfected with pLNCX-FLAG-HIPK2 and either empty vector or pLNCX-HA-PML-RARα deletion constructs. The expression of HIPK2 (top), PML-RARα deletion mutants (middle), and tubulin (bottom) was detected by immunoblotting using anti-FLAG, anti-HA, and anti-tubulin antibodies, respectively. G, effect of PML-RARα deletion mutants on PML oligomerization. 293FT cells were transfected with pLNCX-Myc-PML IV and either pLNCX-HA-PML-RARα deletion constructs or pLNCX-FLAG-PML IV (empty vectors were used as negative controls). The expression of Myc-tagged PML IV and HA-tagged PML-RARα deletion mutants in the lysates of transfectants was detected by immunoblotting using anti-Myc and anti-HA antibodies, respectively (input). The lysates of transfectants were incubated with anti-FLAG antibody. The immunoprecipitates were analyzed by immunoblotting using anti-Myc, anti-HA, and anti-FLAG antibodies (IP). The values of IP/input intensity of Myc-PML IV were quantified with ImageGauge and normalized to the value of Myc-PML IV/empty vector/FLAG-PML IV. DAPI, 4′, 6-diamidino-2-phenylindole; IP, immunoprecipitation.

Figure 2.

The ligand-binding domain of PML-RARα is essential for nuclear body disruption. A, diagram of PML-RARα deletion mutants. B, the ligand-binding domain of PML-RARα is required for the disruption of PML nuclear bodies. U2OS cells were transfected with pLNCX-FLAG-PML IV and pLNCX-HA-PML-RARα deletion constructs or only with pLNCX-HA-PML-RARα deletion constructs as described. PML nuclear bodies were analyzed using anti-FLAG antibody. The white bar represents 10 μm. C, expression of PML-RARα deletion mutants. The expression of PML IV (top), PML-RARα deletion mutants (middle), and tubulin (bottom) in U2OS cells was detected by immunoblotting using anti-FLAG, anti-HA, and anti-tubulin antibodies, respectively. D, the cells expressing PML-RARα ΔE are not immortalized. C-kit+ mouse bone marrow cells were infected with empty vector (mock), pMSCV-HA-PML-RARα wild-type, or ΔE and cultured in methylcellulose medium. The colony number from the third to the fifth round of colonies is indicated (top). Values represent mean ± SEM from 3 independent experiments. E, PML-RARα ΔE does not inhibit nuclear body formation. C-kit+ mouse bone marrow cells were infected with empty vector, pMSCV-HA-PML-RARα wild-type, or ΔE, and plated in methylcellulose medium. Endogenous murine PML nuclear bodies of the cells at first round of colonies were analyzed using mouse Pml-specific antibody (16.1–104). The white bar represents 10 μm. E, effect of PML-RARα deletion mutants on HIPK2 stability. 293FT cells were transfected with pLNCX-FLAG-HIPK2 and either empty vector or pLNCX-HA-PML-RARα deletion constructs. The expression of HIPK2 (top), PML-RARα deletion mutants (middle), and tubulin (bottom) was detected by immunoblotting using anti-FLAG, anti-HA, and anti-tubulin antibodies, respectively. G, effect of PML-RARα deletion mutants on PML oligomerization. 293FT cells were transfected with pLNCX-Myc-PML IV and either pLNCX-HA-PML-RARα deletion constructs or pLNCX-FLAG-PML IV (empty vectors were used as negative controls). The expression of Myc-tagged PML IV and HA-tagged PML-RARα deletion mutants in the lysates of transfectants was detected by immunoblotting using anti-Myc and anti-HA antibodies, respectively (input). The lysates of transfectants were incubated with anti-FLAG antibody. The immunoprecipitates were analyzed by immunoblotting using anti-Myc, anti-HA, and anti-FLAG antibodies (IP). The values of IP/input intensity of Myc-PML IV were quantified with ImageGauge and normalized to the value of Myc-PML IV/empty vector/FLAG-PML IV. DAPI, 4′, 6-diamidino-2-phenylindole; IP, immunoprecipitation.

Close modal

The effect of these PML-RARα mutations on HIPK2 stability was assessed. As shown in Fig. 2F, wild-type PML-RARα and PML-RARα 1–748, which disrupted nuclear bodies, destabilized HIPK2, whereas the other PML-RARα mutants, which did not disrupt nuclear bodies, did not destabilize HIPK2. To assess the effect of the same PML-RARα mutations on PML oligomerization, 293FT cells were cotransfected with Myc-tagged and FLAG-tagged PML IV together with HA-tagged PML-RARα mutants. As shown in Fig. 2G, immunoprecipitation and Western blotting indicated that wild-type PML-RARα and 1–748 inhibited the interaction between Myc-tagged and FLAG-tagged PML, whereas the other PML-RARα mutants did not. These results support the essential role of the ligand-binding domain of PML-RARα in the disruption of PML nuclear bodies.

The PKA phosphorylation site of PML-RARα is required for nuclear body disruption

Experiments were carried out to identify the sites responsible for PML nuclear body inhibition within the ligand-binding domain. As shown in Supplementary Fig. S3A, HA-tagged PML-RARα 1–708 or 1–661 was cotransfected with FLAG-tagged PML IV. PML-RARα 1–708 led to the formation of microspeckle PML nuclear bodies, whereas PML-RARα 1–661 did not (Supplementary Fig. S3B). These results indicate that the PML-RARα region spanning amino acids 662 to 708 is required for PML nuclear body inhibition. RARα and PML-RARα are phosphorylated by PKA (29, 30). Because serine 704, the PKA-dependent phosphorylation site of PML-RARα, is located within the region required for nuclear body disruption (Fig. 3A), mutants were generated in which serine 704 was substituted by alanine and aspartate to simulate dephosphorylated and phosphorylated serine, respectively. The effect of these mutants on PML nuclear body formation was assessed by the transfection of U2OS cells. Representative immunofluorescence data are shown in Fig. 3B (left), and the expression levels of PML and PML-RARα are shown in Fig. 3C. Quantification was done by counting the number of cells that formed large, discrete, and distinct PML nuclear foci in all transfected cells (Fig. 3D). Interestingly, wild-type PML-RARα and the alanine mutant (S704A) disrupted nuclear bodies, as shown by the presence of microspeckles, whereas the aspartate mutant (S704D) did not. Both mutants interacted with PML IV as strongly as did wild-type PML-RARα and the ΔE mutant (Supplementary Fig. S4), suggesting that the inability to disrupt nuclear bodies is not due to any deficiency in binding to PML. Forskolin was then used to activate adenylyl cyclase to determine whether cAMP/PKA might restore nuclear bodies by phosphorylating the serine residue of PML-RARα. In the presence of forskolin, wild-type PML-RARα did not affect nuclear bodies (Fig. 3B and D); however, the S704A mutant, which lacks the serine residue phosphorylated by PKA, inhibited PML nuclear body formation even in the presence of forskolin.

Figure 3.

The PKA-dependent phosphorylation site of PML-RARα regulates PML nuclear body formation. A, diagram of the location of the PKA-dependent phosphorylation site of PML-RARα. B, phosphorylation of serine 704 is important for the restoration of PML nuclear bodies. FLAG-tagged PML IV and HA-tagged PML-RARα point mutants were coexpressed in U2OS cells. Cells were exposed to 50 μmol/L forskolin. PML nuclear bodies were analyzed using an anti-FLAG antibody. The white bar represents 10 μm. C, expression of PML-RARα wild-type and point mutants. The expression of PML IV (top), PML, PML-RARα wild-type or point mutants (middle), and tubulin (bottom) in U2OS cells was detected by immunoblot analysis with anti-FLAG, anti-HA, and anti-tubulin antibodies, respectively. D, quantification of nuclear body formation. The number of cells with PML nuclear bodies was counted. Values represent the mean ± SEM of 4 independent experiments. *, P < 0.01 compared with the PML-RARα value without forskolin. DAPI, 4′, 6-diamidino-2-phenylindole; NB, nuclear body.

Figure 3.

The PKA-dependent phosphorylation site of PML-RARα regulates PML nuclear body formation. A, diagram of the location of the PKA-dependent phosphorylation site of PML-RARα. B, phosphorylation of serine 704 is important for the restoration of PML nuclear bodies. FLAG-tagged PML IV and HA-tagged PML-RARα point mutants were coexpressed in U2OS cells. Cells were exposed to 50 μmol/L forskolin. PML nuclear bodies were analyzed using an anti-FLAG antibody. The white bar represents 10 μm. C, expression of PML-RARα wild-type and point mutants. The expression of PML IV (top), PML, PML-RARα wild-type or point mutants (middle), and tubulin (bottom) in U2OS cells was detected by immunoblot analysis with anti-FLAG, anti-HA, and anti-tubulin antibodies, respectively. D, quantification of nuclear body formation. The number of cells with PML nuclear bodies was counted. Values represent the mean ± SEM of 4 independent experiments. *, P < 0.01 compared with the PML-RARα value without forskolin. DAPI, 4′, 6-diamidino-2-phenylindole; NB, nuclear body.

Close modal

We also tested the effect of wild-type and mutant PML-RARα in normal myeloid stem/progenitor cells. C-kit+ mouse myeloid stem/progenitor cells were infected with a retrovirus encoding HA-tagged PML-RARα, HA-tagged S704A, or HA-tagged S704D and cultured in methylcellulose medium. The location of Pml in the immortalized cells was assessed using an anti-PML antibody specific for mouse Pml. Representative immunofluorescence data are shown in Fig. 4A, and quantification is shown in Fig. 4B. Nuclear body formation was maintained in S704D-expressing cells but was largely disrupted in wild-type PML-RARα- and S704A-expressing cells. The cells transduced with S704D were immortalized as well as those transduced with PML-RARα and S704A (Fig. 4C). Wild-type PML-RARα, S704A, and S704D were expressed at similar levels in the immortalized cells (Fig. 4D). Forskolin restored PML nuclear bodies in the wild-type PML-RARα–expressing cells but could hardly restore nuclear bodies in the S704A-expressing cells (Fig. 4E). These results suggest that the ability of PML-RARα to disrupt nuclear bodies is inhibited by cAMP/PKA-mediated phosphorylation of PML-RARα at serine 704.

Figure 4.

Nuclear body formation is maintained in cells stably expressing PML-RARα S704D. A, PML-RARα S704D does not inhibit nuclear body formation. C-kit+ mouse bone marrow cells were infected with pMSCV-HA-PML-RARα wild-type, S704A, or S704D. Endogenous murine PML nuclear bodies of cells at the third round of colonies were analyzed using mouse Pml-specific antibody (16.1–104). The white bar represents 10 μm. The thick arrow represents PML nuclear bodies, arrowhead represents intermediate nuclear bodies, and thin arrow represents microspeckles. B, quantification of nuclear body formation. The number of cells with PML nuclear bodies, intermediate nuclear bodies, and microspeckles was counted. Values represent the average of 4 independent experiments. C, the cells expressing PML-RARα S704D are immortalized. C-kit+ mouse bone marrow cells were infected with empty vector (mock), pMSCV-HA-PML-RARα wild-type, S704A, or S704D and cultured in methylcellulose medium. The colony number from the third to the fifth round of colonies is indicated (top). Values represent mean ± SEM from 3 independent experiments. The cells at the third round of colonies were stained with May–Giemsa stain (bottom). D, expression of PML-RARα wild-type, S704A, and S704D. The expression of wild-type PML-RARα and mutants and of tubulin in cells at the third round of colonies was analyzed by immunoblotting using anti-PML and anti-tubulin antibodies, respectively. E, quantification of nuclear body restoration. The cells expressing wild-type PML-RARα and S704A at the third round of colonies were collected and exposed to 50 μmol/L forskolin for 24 hours. Endogenous murine PML nuclear bodies were analyzed as described in A. The number of cells with PML nuclear bodies, intermediate nuclear bodies, or microspeckles was counted. Values represent the average of 4 independent experiments. DAPI, 4′, 6-diamidino-2-phenylindole; NB, nuclear body; WT, wild-type.

Figure 4.

Nuclear body formation is maintained in cells stably expressing PML-RARα S704D. A, PML-RARα S704D does not inhibit nuclear body formation. C-kit+ mouse bone marrow cells were infected with pMSCV-HA-PML-RARα wild-type, S704A, or S704D. Endogenous murine PML nuclear bodies of cells at the third round of colonies were analyzed using mouse Pml-specific antibody (16.1–104). The white bar represents 10 μm. The thick arrow represents PML nuclear bodies, arrowhead represents intermediate nuclear bodies, and thin arrow represents microspeckles. B, quantification of nuclear body formation. The number of cells with PML nuclear bodies, intermediate nuclear bodies, and microspeckles was counted. Values represent the average of 4 independent experiments. C, the cells expressing PML-RARα S704D are immortalized. C-kit+ mouse bone marrow cells were infected with empty vector (mock), pMSCV-HA-PML-RARα wild-type, S704A, or S704D and cultured in methylcellulose medium. The colony number from the third to the fifth round of colonies is indicated (top). Values represent mean ± SEM from 3 independent experiments. The cells at the third round of colonies were stained with May–Giemsa stain (bottom). D, expression of PML-RARα wild-type, S704A, and S704D. The expression of wild-type PML-RARα and mutants and of tubulin in cells at the third round of colonies was analyzed by immunoblotting using anti-PML and anti-tubulin antibodies, respectively. E, quantification of nuclear body restoration. The cells expressing wild-type PML-RARα and S704A at the third round of colonies were collected and exposed to 50 μmol/L forskolin for 24 hours. Endogenous murine PML nuclear bodies were analyzed as described in A. The number of cells with PML nuclear bodies, intermediate nuclear bodies, or microspeckles was counted. Values represent the average of 4 independent experiments. DAPI, 4′, 6-diamidino-2-phenylindole; NB, nuclear body; WT, wild-type.

Close modal

Disruption of PML nuclear bodies by PML-RARα is strongly correlated with the destabilization of HIPK2 and the inhibition of PML oligomerization by PML-RARα

As shown in Fig. 1B and Supplementary Fig. S1B, PML IV ΔCC did not form nuclear bodies and destabilized HIPK2. Similarly, the PML-RARα deletion mutants that inhibited PML nuclear body formation also destabilized HIPK2 (Fig. 2F). These results suggest a correlation between nuclear body formation and HIPK2 stability. Therefore, the PML-RARα point mutants described above were assessed for their effect on HIPK2 stability. As shown in Fig. 5A, the level of HIPK2 decreased when HIPK2 was cotransfected with wild-type PML-RARα or S704A but not when HIPK2 was cotransfected with S704D. The effect of the point mutants on HIPK2 stability was also assessed in a stable system as shown in Fig. 4. Endogenous mouse HIPK2 could not be detected (data not shown). The expression of HA-tagged human HIPK2, introduced in mouse c-kit+ cells, was detected only in S704D-expressing cells (Fig. 5B) of the third-round colonies. However, HIPK2 mRNA levels in cells expressing wild-type PML-RARα, S704A, or S704D were not appreciably different (Supplementary Fig. S5). These data support the hypothesis that HIPK2 destabilization is associated with PML nuclear body disruption. The point mutants were then assessed for their effect on PML oligomerization. S704A inhibited PML homo-oligomerization, as did wild-type PML-RARα, whereas S704D did not (Fig. 5C), implying that the protection of PML oligomerization from PML-RARα promotes PML nuclear body formation.

Figure 5.

HIPK2 destabilization and inhibition of PML oligomerization are correlated with PML nuclear body disruption by PML-RARα. A, effect of the PML-RARα point mutants on HIPK2 stability. FLAG-tagged HIPK2 was expressed with either empty vector or HA-tagged PML-RARα point mutants. The expression of HIPK2 (top), PML-RARα point mutants (middle), and tubulin (bottom) was detected by immunoblotting using anti-FLAG, anti-HA, and anti-tubulin antibodies, respectively. B, effect of the PML-RARα point mutants on HIPK2 stability in a stable expression system. C-kit+ mouse bone marrow cells were infected with pMSCV-HA-HIPK2 and pMSCV-HA-PML-RARα encoding either wild-type, S704A, or S704D. The expression of HIPK2, PML-RARα (wild-type and mutants), and tubulin at the third round of colonies was analyzed by immunoblotting using anti-HIPK2, anti-PML, and anti-tubulin antibodies, respectively. C, effect of the PML-RARα point mutants on PML oligomerization. 293FT cells were transfected with pLNCX-Myc-PML IV and either pLNCX-HA-PML-RARα substitution constructs or pLNCX-FLAG-PML IV (empty vectors were used as negative constructs). The expression of Myc-tagged PML IV and HA-tagged PML-RARα substitution mutants in the lysates of transfectants was detected by immunoblotting using anti-Myc and anti-HA antibodies, respectively (Input). The lysates of transfectants were incubated with anti-FLAG antibody. The immunoprecipitates were analyzed by immunoblotting using anti-Myc, anti-HA, and anti-FLAG antibodies (IP). The values of IP/Input intensity of Myc-PML IV were quantified with ImageGauge, and normalized to the value of Myc-PML IV/empty vector/FLAG-PML IV. IP, immunoprecipitation.

Figure 5.

HIPK2 destabilization and inhibition of PML oligomerization are correlated with PML nuclear body disruption by PML-RARα. A, effect of the PML-RARα point mutants on HIPK2 stability. FLAG-tagged HIPK2 was expressed with either empty vector or HA-tagged PML-RARα point mutants. The expression of HIPK2 (top), PML-RARα point mutants (middle), and tubulin (bottom) was detected by immunoblotting using anti-FLAG, anti-HA, and anti-tubulin antibodies, respectively. B, effect of the PML-RARα point mutants on HIPK2 stability in a stable expression system. C-kit+ mouse bone marrow cells were infected with pMSCV-HA-HIPK2 and pMSCV-HA-PML-RARα encoding either wild-type, S704A, or S704D. The expression of HIPK2, PML-RARα (wild-type and mutants), and tubulin at the third round of colonies was analyzed by immunoblotting using anti-HIPK2, anti-PML, and anti-tubulin antibodies, respectively. C, effect of the PML-RARα point mutants on PML oligomerization. 293FT cells were transfected with pLNCX-Myc-PML IV and either pLNCX-HA-PML-RARα substitution constructs or pLNCX-FLAG-PML IV (empty vectors were used as negative constructs). The expression of Myc-tagged PML IV and HA-tagged PML-RARα substitution mutants in the lysates of transfectants was detected by immunoblotting using anti-Myc and anti-HA antibodies, respectively (Input). The lysates of transfectants were incubated with anti-FLAG antibody. The immunoprecipitates were analyzed by immunoblotting using anti-Myc, anti-HA, and anti-FLAG antibodies (IP). The values of IP/Input intensity of Myc-PML IV were quantified with ImageGauge, and normalized to the value of Myc-PML IV/empty vector/FLAG-PML IV. IP, immunoprecipitation.

Close modal

PKA-dependent phosphorylation of PML-RARα restores nuclear bodies and promotes ATRA-induced APL cell differentiation

As shown in Figs. 3–5, PKA-dependent phosphorylation of PML-RARα may be the switch that restores nuclear bodies. Cyclic AMP (cAMP) alone has no effect on nuclear body restoration, but cAMP and ATRA cooperatively restore nuclear bodies in NB4-R1 cells, which are ATRA-maturation–resistant cell lines (31). These data suggest that the cAMP/PKA pathway plays an important role in nuclear body formation. To determine whether the cAMP/PKA pathway actually regulates nuclear body formation in APL cells, APL-derived NB4 cells, which express endogenous PML-RARα, were exposed to forskolin alone. Nuclear bodies were detected using an anti-PML antibody; nuclear bodies were disrupted in NB4 cells, and became clear in the presence of forskolin (Supplementary Fig. S6A). Forskolin did not induce NB4 cell differentiation (Supplementary Fig. S6A). To characterize the forskolin-induced clear particles, the localization of SUMO and DAXX, which are recruited to nuclear bodies, was assessed using anti-SUMO and anti-DAXX antibodies. In the absence of forskolin or ATRA, the colocalization of SUMO and PML, or DAXX and PML, was very limited (Supplementary Fig. S6B). In contrast, ATRA restored nuclear bodies and recruited SUMO and DAXX to nuclear bodies. Forskolin changed the appearance of PML nuclear bodies from microspeckles to clear particles. In the clear particles, PML colocalized with SUMO and DAXX (Supplementary Fig. S6B). Thus, forskolin alone, like ATRA, restored nuclear bodies. ATRA induced PML-RARα degradation, whereas forskolin did not (Supplementary Fig. S6C). These data might reflect the smaller size of nuclear bodies in the presence of forskolin compared with that of ATRA. These data also suggest that PML-RARα phosphorylated by cAMP/PKA inhibits nuclear body disruption.

Because forskolin restored nuclear bodies in NB4 cells, the stability of endogenous HIPK2 was assessed in NB4 cells exposed to forskolin. As shown in Fig. 6A, forskolin increased HIPK2 protein levels but not HIPK2 mRNA levels. In contrast, forskolin did not increase HIPK2 protein levels in the non-APL K562 cells (Fig. 6B). HIPK2 protein levels increased in NB4 cells exposed to ATRA for 24 hours (Fig. 6C). Time-course analysis indicated that the increase in HIPK2 expression was correlated with nuclear body restoration (Fig. 6C and D). These results suggest that HIPK2 is stabilized in nuclear bodies restored upon cAMP/PKA-mediated phosphorylation of PML-RARα.

Figure 6.

Nuclear body restoration promotes ATRA-induced APL cell differentiation. A, forskolin increases HIPK2 protein expression in NB4 cells. NB4 cells were treated with forskolin for 72 hours. The expression of HIPK2 and tubulin was analyzed by immunoblotting (right) using anti-HIPK2 and anti-tubulin antibodies, respectively. B, forskolin does not increase HIPK2 protein expression in K562 cells. The expression of HIPK2 and tubulin in K562 cells was analyzed as described in A. C, effect of forskolin or ATRA on HIPK2 expression. NB4 cells were treated with forskolin or ATRA for 24, 48, and 72 hours and were harvested. The lysates were analyzed as described in A. D, effect of forskolin or ATRA on nuclear body restoration. NB4 cells were exposed to forskolin or ATRA for 24 or 72 hours and stained with anti-PML antibody. The white bar represents 10 μm. E, growth curve of NB4 cells exposed to ATRA and/or forskolin. Cells were counted every other day for 6 days. Values were normalized to the value obtained at the zero time point. Values represent the mean ± SEM of 4 independent experiments. F, forskolin increases ATRA-induced NB4 cell differentiation. NB4 cells were stained with May–Giemsa stain 5 days after adding ATRA and/or forskolin. G, the combination of ATRA and forskolin increases the expression of Mac-1 in NB4 cells. NB4 cells were treated with ATRA and/or forskolin for 5 days, and incubated with anti-Mac-1-FITC. The cells were analyzed by flow cytometry.

Figure 6.

Nuclear body restoration promotes ATRA-induced APL cell differentiation. A, forskolin increases HIPK2 protein expression in NB4 cells. NB4 cells were treated with forskolin for 72 hours. The expression of HIPK2 and tubulin was analyzed by immunoblotting (right) using anti-HIPK2 and anti-tubulin antibodies, respectively. B, forskolin does not increase HIPK2 protein expression in K562 cells. The expression of HIPK2 and tubulin in K562 cells was analyzed as described in A. C, effect of forskolin or ATRA on HIPK2 expression. NB4 cells were treated with forskolin or ATRA for 24, 48, and 72 hours and were harvested. The lysates were analyzed as described in A. D, effect of forskolin or ATRA on nuclear body restoration. NB4 cells were exposed to forskolin or ATRA for 24 or 72 hours and stained with anti-PML antibody. The white bar represents 10 μm. E, growth curve of NB4 cells exposed to ATRA and/or forskolin. Cells were counted every other day for 6 days. Values were normalized to the value obtained at the zero time point. Values represent the mean ± SEM of 4 independent experiments. F, forskolin increases ATRA-induced NB4 cell differentiation. NB4 cells were stained with May–Giemsa stain 5 days after adding ATRA and/or forskolin. G, the combination of ATRA and forskolin increases the expression of Mac-1 in NB4 cells. NB4 cells were treated with ATRA and/or forskolin for 5 days, and incubated with anti-Mac-1-FITC. The cells were analyzed by flow cytometry.

Close modal

Finally, NB4 cells were exposed to forskolin or/and ATRA to determine whether nuclear body restoration may promote NB4 cell differentiation. As shown previously, forskolin was not sufficient to induce NB4 cell differentiation (Fig. 6E and F); however, forskolin enhanced ATRA-induced differentiation (Fig. 6E and F). The combination of ATRA and forskolin resulted in the differentiation of NB4 cells into segmented granulocytes (Fig. 6F) and increased the expression of the differentiation marker Mac-1 (Fig. 6G) more efficiently than either drug alone. Other studies have also shown the efficacy of cAMP against APL (32–35). These results and reports suggest that cAMP/PKA promotes ATRA-induced APL cell differentiation by restoring nuclear bodies.

In more than 90% of APL cases, the PML-RARα fusion protein is generated by the t(15;17) chromosomal translocation. PML-RARα disrupts PML nuclear bodies by a mechanism that was not understood in detail. The present study reveals that PML-RARα blocks PML oligomerization, resulting in the disruption of nuclear bodies, and that cAMP/PKA phosphorylation of PML-RARα restores nuclear bodies. Our results suggest that nuclear body restoration enhances APL cell differentiation.

PML nuclear body disruption and restoration

Nuclear bodies are disrupted in APL cells harboring the t(15;17) chromosomal translocation (16, 17, 19). Results showed that wild-type PML-RARα blocked PML oligomerization (Supplementary Fig. S2C). Deletion analysis showed that PML-RARα mutants that block PML oligomerization also disrupt nuclear bodies (Figs. 2B and G, 3B, and 5C). The blocking of PML oligomerization by PML-RARα occurs independent of their interaction (Figs 2G and 5C). These results suggest that the inability of inhibition of PML oligomerization by PML-RARα mutants is not due to the inability of interaction with PML IV, and imply that PML-RARα–induced nuclear body disruption is due to impaired PML oligomerization.

RARα and PML-RARα are phosphorylated by the cAMP/PKA pathway at a site located within the ligand-binding domain (29, 30). The PML-RARα S704D mutant, which is expected to simulate phosphorylated PML-RARα, did not block PML oligomerization and did not disrupt nuclear bodies. Moreover forskolin restored nuclear bodies disrupted in U2OS cells expressing wild-type PML-RARα (Fig. 3B and D), in mouse myeloid stem/progenitor cells expressing wild-type PML-RARα (Fig. 4E), or in APL-derived NB4 cells (Fig. 6D). Our results indicate that the ligand-binding domain of PML-RARα is key to the inhibition of PML oligomerization and to the disruption of PML nuclear bodies, and that PKA-dependent phosphorylation of the serine residue in that region reverses those effects and restores nuclear body formation. Phosphorylated PML-RARα is known to be easily degraded by ATRA (30); however, in the absence of ATRA, phosphorylated PML-RARα was stable (Figs. 4D, 5A and B, and Supplementary Fig. S6C). The mechanism by which phosphorylation of PML-RARα prevents its inhibition of PML oligomerization remains unclear. However, there are precedents for this; the phosphorylation of RARα by PKA enhances the interaction between RARα and cyclin H/cdk2 (29), suggesting that phosphorylation of RARα increases its interaction with cyclin H/cdk2. Thus, PML might find it easier to access a complex of phosphorylated PML-PML-RARα than a complex of dephosphorylated PML-PML-RARα.

Nuclear body restoration is important for differentiation of APL cells

The PKA phosphorylation site of PML-RARα regulates nuclear body formation. Forskolin restored nuclear bodies in APL-derived NB4 cells (Fig. 6D). Although forskolin alone was not sufficient to induce NB4 cell differentiation, it promoted ATRA-induced differentiation (Fig. 6G). Published studies showed that cAMP enhances retinoic acid-induced APL differentiation and PML-RARα transactivation (32–35). These studies also showed that PKA dissociates RARα from SMRT and activates transcription. Moreover, a recent report indicated that cAMP-dependent phosphorylation of PML-RARα was crucial for the eradication of APL-initiating cells (30). Taken together, these reports and the present data suggest that nuclear body restoration is one of the reasons why cAMP/PKA could be useful as an APL therapy.

We previously showed that HIPK2 is stabled in PML nuclear bodies and degraded outside of nuclear bodies by SCFFbx3, suggesting that HIPK2 is destabilized by disruption of nuclear bodies (15). In this paper, we showed that PML IV ΔCC, wild-type PML-RARα, PML-RARα 1–748, and S704A, which disrupted PML nuclear bodies, also destabilized HIPK2. In contrast, PML, PML-RARα 1–567, 1–492, 1–420, ΔE, and S704D, which did not disrupt nuclear bodies, did not destabilize HIPK2. Forskolin and ATRA, which restore nuclear bodies in NB4 cells, increased HIPK2 expression (Fig. 6A and C), and the increase in HIPK2 expression was correlated with nuclear body restoration (Fig. 6D). These data indicate that HIPK2 destabilization is strongly correlated with nuclear body disruption and that nuclear body formation is important for HIPK2 stabilization. HIPK2 is important for PML-dependent transcriptional activation (15). We have also found that PML stabilizes the PU.1/p300 complex to regulate PU.1-dependent transcription and myeloid differentiation (26). Mutations of HIPK2 are found in AML and myelodysplastic syndrome (36). PML-RARα also disrupts PU.1/p300 complexes and inhibits myeloid differentiation (26). Therefore, nuclear body formation by PML oligomerization may lead to the recruitment of transcription factors/coactivators and to their stabilization for transcriptional activation and regulation of granulopoiesis. As suggested by the results of the present study, this might be because cAMP/PKA-dependent nuclear body restoration enhances APL cell differentiation.

No potential conflicts of interest were disclosed.

Conception and design: Y. Shima, I. Kitabayashi

Development of methodology: Y. Shima

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): Y. Shima, Y. Honma

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): Y. Shima, I. Kitabayashi

Writing, review, and/or revision of the manuscript: Y. Shima, I. Kitabayashi

The authors thank Dr. de Thé for helpful discussions.

This work was supported in part by Grants-in-Aid from the Ministry of Health, Labor, and Welfare, the Ministry of Education, Culture, Sports, Science, and Technology, and National Cancer Center Research and Development Fund.

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.
Gilliland
DG
. 
Molecular genetics of human leukemia
.
Leukemia
1998
;
12
Suppl 1
:
S7
12
.
2.
Look
AT
. 
Oncogenic transcription factors in the human acute leukemias
.
Science
1997
;
278
:
1059
64
.
3.
de Thé
H
,
Chomienne
C
,
Lanotte
M
,
Degos
L
,
Dejean
A
. 
The t(15;17) translocation of acute promyelocytic leukaemia fuses the retinoic acid receptor alpha gene to a novel transcribed locus
.
Nature
1990
;
347
:
558
61
.
4.
de Thé
H
,
Lavau
C
,
Marchio
A
,
Chomienne
C
,
Degos
L
,
Dejean
A
. 
The PML-RAR alpha fusion mRNA generated by the t(15;17) translocation in acute promyelocytic leukemia encodes a functionally altered RAR
.
Cell
1991
;
66
:
675
84
.
5.
Goddard
AD
,
Borrow
J
,
Freemont
PS
,
Solomon
E
. 
Characterization of a zinc finger gene disrupted by the t(15;17) in acute promyelocytic leukemia
.
Science
1991
;
254
:
1371
4
.
6.
Kakizuka
A
,
Miller
WH
 Jr
,
Umesono
K
,
Warrell
RP
 Jr
,
Frankel
SR
,
Murty
VV
, et al
Chromosomal translocation t(15;17) in human acute promyelocytic leukemia fuses RAR alpha with a novel putative transcription factor, PML
.
Cell
1991
;
66
:
663
74
.
7.
Guo
A
,
Salomoni
P
,
Luo
J
,
Shih
A
,
Zhong
S
,
Gu
W
, et al
The function of PML in p53-dependent apoptosis
.
Nat Cell Biol
2000
;
2
:
730
6
.
8.
Nguyen
LA
,
Pandolfi
PP
,
Aikawa
Y
,
Tagata
Y
,
Ohki
M
,
Kitabayashi
I
. 
Physical and functional link of the leukemia-associated factors AML1 and PML
.
Blood
2005
;
105
:
292
300
.
9.
Pearson
M
,
Carbone
R
,
Sebastiani
C
,
Cioce
M
,
Fagioli
M
,
Saito
S
, et al
PML regulates p53 acetylation and premature senescence induced by oncogenic Ras
.
Nature
2000
;
406
:
207
10
.
10.
Möller
A
,
Sirma
H
,
Hofmann
TG
,
Rueffer
S
,
Klimczak
E
,
Droge
W
, et al
PML is required for homeodomain-interacting protein kinase 2 (HIPK2)-mediated p53 phosphorylation and cell cycle arrest but is dispensable for the formation of HIPK domains
.
Cancer Res
2003
;
63
:
4310
4
.
11.
von Mikecz
A
,
Zhang
S
,
Montminy
M
,
Tan
EM
,
Hemmerich
P
. 
CREB-binding protein (CBP)/p300 and RNA polymerase II colocalize in transcriptionally active domains in the nucleus
.
J Cell Biol
2000
;
150
:
265
73
.
12.
Weis
K
,
Rambaud
S
,
Lavau
C
,
Jansen
J
,
Carvalho
T
,
Carmo-Fonseca
M
, et al
Retinoic acid regulates aberrant nuclear localization of PML-RAR alpha in acute promyelocytic leukemia cells
.
Cell
1994
;
76
:
345
56
.
13.
Ishov
AM
,
Sotnikov
AG
,
Negorev
D
,
Vladimirova
OV
,
Neff
N
,
Kamitani
T
, et al
PML is critical for ND10 formation and recruits the PML-interacting protein daxx to this nuclear structure when modified by SUMO-1
.
J Cell Biol
1999
;
147
:
221
34
.
14.
Torii
S
,
Egan
DA
,
Evans
RA
,
Reed
JC
. 
Human Daxx regulates Fas-induced apoptosis from nuclear PML oncogenic domains (PODs)
.
EMBO J
1999
;
18
:
6037
49
.
15.
Shima
Y
,
Shima
T
,
Chiba
T
,
Irimura
T
,
Pandolfi
PP
,
Kitabayashi
I
. 
PML activates transcription by protecting HIPK2 and p300 from SCFFbx3-mediated degradation
.
Mol Cell Biol
2008
;
28
:
7126
38
.
16.
Daniel
MT
,
Koken
M
,
Romagne
O
,
Barbey
S
,
Bazarbachi
A
,
Stadler
M
, et al
PML protein expression in hematopoietic and acute promyelocytic leukemia cells
.
Blood
1993
;
82
:
1858
67
.
17.
Dyck
JA
,
Maul
GG
,
Miller
WH
 Jr
,
Chen
JD
,
Kakizuka
A
,
Evans
RM
. 
A novel macromolecular structure is a target of the promyelocyte-retinoic acid receptor oncoprotein
.
Cell
1994
;
76
:
333
43
.
18.
Kastner
P
,
Perez
A
,
Lutz
Y
,
Rochette-Egly
C
,
Gaub
MP
,
Durand
B
, et al
Structure, localization and transcriptional properties of two classes of retinoic acid receptor alpha fusion proteins in acute promyelocytic leukemia (APL): structural similarities with a new family of oncoproteins
.
EMBO J
1992
;
11
:
629
42
.
19.
Koken
MH
,
Puvion-Dutilleul
F
,
Guillemin
MC
,
Viron
A
,
Linares-Cruz
G
,
Stuurman
N
, et al
The t(15;17) translocation alters a nuclear body in a retinoic acid-reversible fashion
.
EMBO J
1994
;
13
:
1073
83
.
20.
Chen
GQ
,
Shi
XG
,
Tang
W
,
Xiong
SM
,
Zhu
J
,
Cai
X
, et al
Use of arsenic trioxide (As2O3) in the treatment of acute promyelocytic leukemia (APL): I. As2O3 exerts dose-dependent dual effects on APL cells
.
Blood
1997
;
89
:
3345
53
.
21.
Zhu
J
,
Koken
MH
,
Quignon
F
,
Chelbi-Alix
MK
,
Degos
L
,
Wang
ZY
, et al
Arsenic-induced PML targeting onto nuclear bodies: implications for the treatment of acute promyelocytic leukemia
.
Proc Natl Acad Sci U S A
1997
;
94
:
3978
83
.
22.
Lin
RJ
,
Evans
RM
. 
Acquisition of oncogenic potential by RAR chimeras in acute promyelocytic leukemia through formation of homodimers
.
Mol Cell
2000
;
5
:
821
30
.
23.
Minucci
S
,
Maccarana
M
,
Cioce
M
,
De Luca
P
,
Gelmetti
V
,
Segalla
S
, et al
Oligomerization of RAR and AML1 transcription factors as a novel mechanism of oncogenic activation
.
Mol Cell
2000
;
5
:
811
20
.
24.
Perez
A
,
Kastner
P
,
Sethi
S
,
Lutz
Y
,
Reibel
C
,
Chambon
P
. 
PMLRAR homodimers: distinct DNA binding properties and heteromeric interactions with RXR
.
EMBO J
1993
;
12
:
3171
82
.
25.
Tagata
Y
,
Yoshida
H
,
Nguyen
LA
,
Kato
H
,
Ichikawa
H
,
Tashiro
F
, et al
Phosphorylation of PML is essential for activation of C/EBP epsilon and PU.1 to accelerate granulocytic differentiation
.
Leukemia
2008
;
22
:
273
80
.
26.
Yoshida
H
,
Ichikawa
H
,
Tagata
Y
,
Katsumoto
T
,
Ohnishi
K
,
Akao
Y
, et al
PML-retinoic acid receptor alpha inhibits PML IV enhancement of PU.1-induced C/EBPepsilon expression in myeloid differentiation
.
Mol Cell Biol
2007
;
27
:
5819
34
.
27.
Jeanne
M
,
Lallemand-Breitenbach
V
,
Ferhi
O
,
Koken
M
,
Le Bras
M
,
Duffort
S
, et al
PML/RARA oxidation and arsenic binding initiate the antileukemia response of As2O3
.
Cancer Cell
2010
;
18
:
88
98
.
28.
Grignani
F
,
Testa
U
,
Rogaia
D
,
Ferrucci
PF
,
Samoggia
P
,
Pinto
A
, et al
Effects on differentiation by the promyelocytic leukemia PML/RARalpha protein depend on the fusion of the PML protein dimerization and RARalpha DNA binding domains
.
EMBO J
1996
;
15
:
4949
58
.
29.
Gaillard
E
,
Bruck
N
,
Brelivet
Y
,
Bour
G
,
Lalevee
S
,
Bauer
A
, et al
Phosphorylation by PKA potentiates retinoic acid receptor alpha activity by means of increasing interaction with and phosphorylation by cyclin H/cdk7
.
Proc Natl Acad Sci U S A
2006
;
103
:
9548
53
.
30.
Nasr
R
,
Guillemin
MC
,
Ferhi
O
,
Soilihi
H
,
Peres
L
,
Berthier
C
, et al
Eradication of acute promyelocytic leukemia-initiating cells through PML-RARA degradation
.
Nat Med
2008
;
14
:
1333
42
.
31.
Duprez
E
,
Lillehaug
JR
,
Naoe
T
,
Lanotte
M
. 
cAMP signalling is decisive for recovery of nuclear bodies (PODs) during maturation of RA-resistant t(15;17) promyelocytic leukemia NB4 cells expressing PML-RAR alpha
.
Oncogene
1996
;
12
:
2451
9
.
32.
Altucci
L
,
Rossin
A
,
Hirsch
O
,
Nebbioso
A
,
Vitoux
D
,
Wilhelm
E
, et al
Rexinoid-triggered differentiation and tumor-selective apoptosis of acute myeloid leukemia by protein kinase A-mediated desubordination of retinoid X receptor
.
Cancer Res
2005
;
65
:
8754
65
.
33.
Duprez
E
,
Lillehaug
JR
,
Gaub
MP
,
Lanotte
M
. 
Differential changes of retinoid-X-receptor (RXR alpha) and its RAR alpha and PML-RAR alpha partners induced by retinoic acid and cAMP distinguish maturation sensitive and resistant t(15;17) promyelocytic leukemia NB4 cells
.
Oncogene
1996
;
12
:
2443
50
.
34.
Guillemin
MC
,
Raffoux
E
,
Vitoux
D
,
Kogan
S
,
Soilihi
H
,
Lallemand-Breitenbach
V
, et al
In vivo activation of cAMP signaling induces growth arrest and differentiation in acute promyelocytic leukemia
.
J Exp Med
2002
;
196
:
1373
80
.
35.
Kamashev
D
,
Vitoux
D
,
De Thé
H
. 
PML-RARA-RXR oligomers mediate retinoid and rexinoid/cAMP cross-talk in acute promyelocytic leukemia cell differentiation
.
J Exp Med
2004
;
199
:
1163
74
.
36.
Li
XL
,
Arai
Y
,
Harada
H
,
Shima
Y
,
Yoshida
H
,
Rokudai
S
, et al
Mutations of the HIPK2 gene in acute myeloid leukemia and myelodysplastic syndrome impair AML1- and p53-mediated transcription
.
Oncogene
2007
;
26
:
7231
9
.