All-trans-retinoic acid (RA) treatment induces morphological remission in acute promyelocytic leukemia (APL) patients carrying the t(15;17) and expressing the PML/RARα product by inducing terminal differentiation of the leukemic clone. RA treatment induces downregulation of PML/RARα and reorganization of the PML-nuclear bodies. These events have been proposed to be essential for the induction of APL cell differentiation by RA. Here, we show that in the APL-derived NB4 cell line as well as in myeloid precursor U937 cells expressing the PML/RARα (U937/PR9) and in blasts from APL patients, the PML/RARα fusion protein is cleaved by a caspase 3–like activity induced by RA treatment. In fact, a caspase 3–like activity is detectable in PML/RARα expressing cells after RA treatment, and selective caspase inhibitor peptides are able to prevent the RA-induced degradation of the fusion protein in vivo and in vitro. Using recombinant caspases and PML/RARα deletion mutants we mapped a caspase 3 cleavage site (Asp 522) within the α-helix region of the PML component of the fusion protein. The extent of PML/RARα cleavage directly correlates with the ability of RA to restore the normal PML nuclear bodies (NBs) pattern. However, RA-induced differentiation is not prevented by the persistence of the fusion product and occurs in the absence of normally structured PML NBs. These results indicate that PML/RARα is directly involved in conferring RA sensitivity of APL cells and that the RA-induced reassembly of PML NBs is the consequence of the disappearance of PML/RARα.

THE ACUTE PROMYELOCYTIC leukemia (APL) phenotype depends on the expression of the PML/RARα fusion product, which results from the recombination between the promyelocytic leukemia (PML) gene on chromosome 15 and the retinoic acid (RA) receptor α gene (RARα) on chromosome 17 (t15;17).1,2 PML, a member of the RING finger family, is ubiquitously expressed in tissues within large multiprotein nuclear structures, termed nuclear bodies (NBs), and acts as a growth suppressor in vitro.3-6 RARα is one of the nuclear retinoid receptors that mediates RA action on myeloid differentiation.7-10 PML/RARα retains large portions of the parental proteins, including the RING, B1-B2, and coiled-coil regions of PML and the DNA and ligand binding domains of RARα.1,2 

The mechanism of the PML/RARα leukemogenic activity is poorly understood. PML/RARα expression into hematopoietic precursor cell lines induces differentiation block and promotes survival.11 Both phenotypes depend on the integrity and fusion of PML and RARα sequences.12 It has been proposed that PML/RARα exerts a dominant negative action on wild-type PML and RXR, an RARα cofactor, because expression of PML/RARα provokes PML and RXR delocalization.3-5 Indeed, in APL cells and in bone marrow cells from leukemic PML/RARα transgenic mice, the PML NBs are disrupted into a microspeckled pattern.3-5,13-15 Based on these findings it has been suggested that the integrity of PML NBs is important for normal myeloid differentiation and that PML/RARα may cause leukemia by interfering with either the RAR- or PML-dependent differentiation pathways.3-5,16 

APL is a unique model for differentiation therapy, as indicated by the fact that RA induces terminal differentiation of PML/RARα-expressing cells both in vivo and in vitro.1,2,16,17 Treatment with RA induces downregulation of the fusion protein, disappearance of the PML/RARα microspeckles, reorganization of PML NBs pattern, and relocalization of PML to its physiological site, therefore suggesting that the main effect of RA in APL cells is to release the dominant negative effect of PML/RARα on wild-type PML.3-5,18,19However, expression of PML/RARα into different myeloid cell lines increases RA sensitivity and alterations of PML/RARα correlates with RA resistance in APL-derived NB4 cell lines.11,20-22 These findings would indicate that the fusion protein is actively involved in conferring RA sensitivity and that the integrity of the PML NBs plays no role.

Downregulation of PML/RARα by RA occurs at the posttranslational level18 and is prevented by proteasome inhibitors.19 Recent findings show that PML is associated with ubiquitin-related proteins,23,24 and a possible role for the proteasome pathway in the RA-induced degradation of PML/RARα has been suggested.19 The proteasome is also involved in cellular pathways leading to caspase activation and apoptosis.25,26 Caspases are a family of cysteine proteases with aspartic acid substrate specificity, thought to be key effectors of cellular apoptosis in multicellular organisms.27,28 

Here, we show that caspases mediate the RA-induced degradation of PML/RARα. Using different caspase inhibitor peptides, we were able to prevent RA-induced PML/RARα degradation and test directly the contribution of PML/RARα degradation and PML NBs reorganization to RA response of APL cells.

Chemicals, antibodies, and plasmids.

All-trans-retinoic acid was obtained from Sigma (Milano, Italy). z-Al-Ala-DL-Asp-fluorometylketone (zVAD), Ac-Asp-Glu-Val-aspartic acid aldehyde (DEVD), and Ac-Tyr-Val-Ala-Asp-chloromethylketone (YVAD) were obtained from Bachem, Switzerland. RARα antibody was obtained from Dr P. Chambon (Strasbourg, France)29 and from Santa Cruz Biotechnology (Santa Cruz, CA). PG-M3 and α-PML3 are anti-PML antibodies.12,30 

PML/RARα and the mutants ΔC P/R, ΔH P/R, and HA-tagged PML/RARα were previously described.11,12 The pSG5/H4-RAR mutant was constructed by polymerase chain reaction (PCR) amplification and fusion of the PML heptad region with the RARα moiety of the PML/RARα fusion protein (F. Grignani and P.G. Pelicci, manuscript in preparation). MBP-PML/RARα construct was generated by PCR/subcloning in the pMal vector (New England Biolabs, Beverly, MA) and expressed in Escherichia coli B21 cells by induction with isopropyl-1-thio-β-D-galactopyranoside (IPTG) for 1 to 3 hours at 23°C. Poly(ADP-ribose) polymerase (PARP) cDNA31 was subcloned into a pBluescript vector (Stratagene, La Jolla, CA). [35S]-PARP, -PML/RARα, and -mutant proteins were prepared by in vitro transcription-translation using the TNT T7 coupled reticulocyte lysate system (Promega, Madison, WI). [35S]-methionine was purchased from Amersham (Arlington Heights, IL).

Cell culture and differentiation.

The human APL cell line NB4 was obtained from Dr M. Lanotte, INSERM, Paris, France32; the U937/PR9 and the U937/HA-PR1 are described subclones of the U937 promonocytic cell line.11,12 Leukemic cells from informed, newly diagnosed APL patients were isolated and cultured as previously described.33 Cells were maintained in RPMI 1640 medium supplemented with 10% fetal calf serum (FCS). Cell differentiation was evaluated as described.11,12 

Immunofluorescence staining and immunoblot analysis.

Cells were collected, cytocentrifuged, and fixed in 100% methanol at room temperature for 5 minutes, followed by acetone for 2 minutes at −20°C. PML staining was performed with the PGM3 or with the anti-HA monoclonal antibodies as described.12,30 The immunofluorescence was detected using an Olympus BX-60 fluoromicroscope equipped with a chilled 3CCD digital color camera (C5810 Hamamatsu Photonics, Hamamatsu City, Japan).

Immunoblot analysis was performed on total cell homogenates as previously described.12,18 Immunoreactivity was determined using the ECL method (Amersham).

In vitro cleavage assays.

Cells were collected by centrifugation and resuspended in 100 μL of a lysis buffer containing 50 mmol/L NaCl, 2 mmol/L MgCl2, 40 mmol/L β-glycerophosphate, 5 mmol/L EGTA, and 10 mmol/L HEPES pH 7.0. Cleavage reactions were performed in a volume of 36 μL containing 25 mmol/L HEPES, pH 7.5, 100 mmol/L NaCl, 2 mmol/L MgCl2, 5 mmol/L dithiotreitol, 0.1% Triton X, 1 mmol/L phenylmethylsulfonyl fluoride, and 2 μg/mL of each aprotinin, leupeptin, and pepstatin with 3 μL of [35S] methionine–labeled PARP and 15 μL of the cell lysates. Each reaction was incubated for 1 hour at 37°C. To map the PML/RARα caspase cleavage site, cell lysates were substituted with bacterially expressed GST or GST caspase1, or GST–caspase 3 fusion proteins.31 The reactions were fractionated on sodium dodecyl sulfate (SDS)-PAGE. The gels were fixed (acetic acid 60%, methanol 40%, and glycerol 5%), treated with the Amplify solution (Amersham), and dried. The cleavage products were visualized after an overnight exposure at −80°C or by immunoblotting as described above.

Inhibition of caspases prevents RA-induced PML/RARα degradation in vivo.

To investigate the involvement of caspases in the RA-induced degradation of PML/RARα, NB4 cells, fresh APL blasts, and the U937/PR9 cell line, a subclone of the promonocytic leukemia cell line U937 stably transfected with the PML/RARα cDNA under the control of a Zinc (Zn)-inducible promoter were treated with RA, in the presence or absence of the broad spectrum caspase inhibitor oligopeptide zVAD,34 and analyzed for PML/RARα expression by immunoblotting using the anti-RARα RPα(F) antibody.29 As shown in Fig 1A, RA-induced degradation of PML/RARα was inhibited by the addition of 100 μmol/L zVAD. Densitometric analysis of the Western blots shown in Fig 1A revealed that zVAD is able to inhibit about 40% to 50% of the RA-induced PML/RARα degradation in NB4 cells and in PML/RARα expressing U937/PR9 cells, whereas in APL blasts inhibition by zVAD is 87%. In addition, treatment of NB4 and APL blasts with zVAD in the absence of RA often resulted into increased levels of PML/RARα. This effect of zVAD was not detectable in PML/RARα expressing AML cells (U937/PR9 cells), suggesting that caspases are involved in the steady-state regulation of PML/RARα stability in APL cells.

Fig. 1.

Effect of the caspase inhibitors zVAD, DEVD, and YVAD in preventing RA-induced PML/RARα degradation. (A) Cells were treated with 1 μmol/L RA for 96 hours. U937/PR9 cells were induced to express PML/RARα by treatment with 100 μmol/L Zn for 16 hours. zVAD (100 μmol/L) was added for 1 hour before RA treatment. (B) Zn-induced U937/PR9 cells were treated with the indicated concentrations of DEVD or YVAD. Immunoblot analysis was performed on total cellular proteins (50 μg) using the anti-RARα RPα(F) antibody. Immunoreactivity to β-tubulin was used for loading control.

Fig. 1.

Effect of the caspase inhibitors zVAD, DEVD, and YVAD in preventing RA-induced PML/RARα degradation. (A) Cells were treated with 1 μmol/L RA for 96 hours. U937/PR9 cells were induced to express PML/RARα by treatment with 100 μmol/L Zn for 16 hours. zVAD (100 μmol/L) was added for 1 hour before RA treatment. (B) Zn-induced U937/PR9 cells were treated with the indicated concentrations of DEVD or YVAD. Immunoblot analysis was performed on total cellular proteins (50 μg) using the anti-RARα RPα(F) antibody. Immunoreactivity to β-tubulin was used for loading control.

Close modal

Caspase inhibitors containing the peptide-recognition elements of endogenous substrates may reveal the involvement of specific classes of caspases.27,28 The tetrapeptide YVAD is a potent inhibitor of the caspase 1–like subfamily and a poor inhibitor of CED-3–like caspases. Conversely, the tetrapeptide DEVD effectively inhibits caspase 3–like activities. Increasing concentrations of YVAD or DEVD were added for 1 hour, before 1 μmol/L RA treatment for 20 hours. Degradation of PML/RARα was almost completely blocked by DEVD (60% to 80%), whereas YVAD was less effective (Fig 1B). Similar results were obtained in NB4 cells (not shown). These results suggested that a caspase 3–like protease is preferentially involved in the degradation of PML/RARα by RA in vivo.

RA induces caspase 3–like activity in NB4 cells.

To evaluate whether caspase 3–like activity was induced by RA treatment, we tested NB4 cellular lysates for their capacity to induce cleavage of the nuclear enzyme poly(ADP-ribose) polymerase (PARP), an endogenous substrate for caspase 3–like proteases.28Incubation of in vitro–translated PARP with cell lysates from untreated NB4 cells resulted in a partial PARP cleavage, as shown by the appearance of the 85-kD cleavage product (Fig 2, arrow). However, PARP cleavage was greatly enhanced by lysates from RA-treated NB4 cells (Fig 2A). In both cases, this cleavage was prevented by DEVD in vitro, the inhibitor peptide mimicking the PARP cleavage site (Fig 2A). These results indicate that caspase 3–like activity is endogenously present in NB4 cells and that it is further enhanced after RA treatment.

Fig. 2.

Cleavage of in vitro–translated PARP. [35S]-PARP was incubated for 1 hour at 37°C with cell lysates from untreated or from 1μmol/L RA-treated (A) NB4 and (B) U937/PR9 cells induced (+) or not (−) by 100 μmol/L Zn to express PML/RARα. One hundred micromoles per liter DEVD was added as indicated. Arrow indicates the 85-kD PARP cleavage product.

Fig. 2.

Cleavage of in vitro–translated PARP. [35S]-PARP was incubated for 1 hour at 37°C with cell lysates from untreated or from 1μmol/L RA-treated (A) NB4 and (B) U937/PR9 cells induced (+) or not (−) by 100 μmol/L Zn to express PML/RARα. One hundred micromoles per liter DEVD was added as indicated. Arrow indicates the 85-kD PARP cleavage product.

Close modal
RA-induced caspase 3–like activity requires PML/RARα expression.

We investigated the role of PML/RARα on the induction of RA-dependent PARP cleavage activity using cell lysates from the U937/PR9 clone. In these cells treatment with 100 μmol/L Zn results in the expression of the PML/RARα protein.11 Cell extracts prepared from untreated U937/PR9 cells, or from cells treated with RA or Zn, failed to cleave in vitro–translated PARP. However, PARP cleavage was induced by lysates from U937/PR9 cells treated with Zn and RA. PARP cleavage was prevented by DEVD (Fig 2B). Simultaneous treatment of parental U937 cells with Zn and RA did not induce caspase 3 activity (not shown). Therefore, PML/RARα expression is required for the induction of caspase 3–like activity that follows RA treatment.

Mapping of a major caspase 3 cleavage site within the α-helix region of PML.

We then directly tested whether PML/RARα could be cleaved by caspase 3 in vitro. Incubation of in vitro–translated or bacterially expressed (MBP-PML/RARα; see Materials and Methods) PML/RARα with recombinant GST caspase 3 for 1 hour induced the formation of ∼50 kD proteolytic fragment (Fig 3B and C). This cleavage product was specifically induced by caspase 3 because it was blocked in vitro by DEVD and was not observed using GST or GST caspase 1 (Fig 3B and C and data not shown). Anti-RARα RPα(F) antibody specifically reacted with p50 (Fig 3B), thereby suggesting that GST caspase 3 induces a p50 proteolytic fragment that contains the C-terminal F region of RARα.

Fig. 3.

(A) Schematic representation of the amino acid sequences of PML/RARα and mutants. P, proline-rich region; R, RING finger domain; B1 and B2, B-boxes; C.C., coiled-coil region; α-H, α-helix region; SP, the α-helix serine and proline-rich region; BP, PML/RARα junction. (B through F) RARα functional regions. (B) MBP-PML/RARα (MBP-P/R) or (C) in vitro–translated [35S]-PML/RARα (P/R) or (D) [35S]-▵C P/R, (E) [35S]-▵H P/R, and (F) [35S]-H4-RAR were incubated with GST or GST caspase 3 for 1 hour at 37°C. One hundred micromoles per liter DEVD was added where indicated. Samples were analyzed by discontinous 8% to 15% SDS-PAGE and visualized by autoradiography or by immunoblotting (B) using an anti-RARα antibody (Santa Cruz Biotechnology). Molecular weight markers are indicated. Arrows indicate the p50 proteolytic fragment.

Fig. 3.

(A) Schematic representation of the amino acid sequences of PML/RARα and mutants. P, proline-rich region; R, RING finger domain; B1 and B2, B-boxes; C.C., coiled-coil region; α-H, α-helix region; SP, the α-helix serine and proline-rich region; BP, PML/RARα junction. (B through F) RARα functional regions. (B) MBP-PML/RARα (MBP-P/R) or (C) in vitro–translated [35S]-PML/RARα (P/R) or (D) [35S]-▵C P/R, (E) [35S]-▵H P/R, and (F) [35S]-H4-RAR were incubated with GST or GST caspase 3 for 1 hour at 37°C. One hundred micromoles per liter DEVD was added where indicated. Samples were analyzed by discontinous 8% to 15% SDS-PAGE and visualized by autoradiography or by immunoblotting (B) using an anti-RARα antibody (Santa Cruz Biotechnology). Molecular weight markers are indicated. Arrows indicate the p50 proteolytic fragment.

Close modal

Because in vitro–translated RARα was not cleaved by GST caspase 3 or 1 (data not shown) and the RARα component of the fusion protein is ∼46 kD, the caspase 3 cleavage site was expected to map within the C-terminal portion of the PML component of PML/RARα fusion product. To directly show it, we tested the stability of various in vitro–translated PML/RARα deletion mutants (shown in Fig 3A) in the presence of GST caspase 3. ΔC P/R is a PML/RARα deletion mutant in which the N-terminal RING and B1+ B2 regions were deleted.12 In the ΔH P/R mutant, the coiled-coil region was deleted.12 The two regions deleted in ΔC P/R and ΔH P/R are adjacent in PML. Figure 3D and E shows that p50 is released following the incubation of GST caspase 3 with in vitro–translated ΔC P/R and ΔH P/R mutants, thereby suggesting that the caspase 3 cleavage site maps downstream to PML coil-coiled region, within the α-helix region. Notably, recombinant caspase 3 was not able to cleave the H4-RAR mutant obtained by fusing the PML heptad repeats directly to the RARα portion of PML/RARα (Fig 3F). Thus, a caspase 3 cleavage site maps within the α-helix region of PML retained in both ΔC P/R and ΔH P/R mutants but not in H4-RAR. This region contains only one aspartic acid (Asp) at position 522, within a sequence (PHLD:GP) permissive to caspases cleavage. The predicted molecular weight for the 434 amino acids proteolytic product (∼49.9 kD) is consistent with the observed p50 fragment and with Asp 522 being the cleaved residue.

Inhibition of PML/RARα degradation does not prevent RA-induced myeloid differentiation.

To investigate the role of PML/RARα degradation in RA-induced terminal differentiation, we took advantage of the possibility to prevent PML/RARα downregulation using caspase inhibitors. NB4 and Zn-induced U937/PR9 cells were pretreated with caspase inhibitors (DEVD or zVAD) or control medium for 1 hour before the addition of 1 μmol/L RA for 4 days. Immunoblot analysis revealed a marked effect of caspase inhibitor DEVD on the stability of PML/RARα in both NB4 (Fig 4A) and U937/PR9 (Fig 4B) cells. Treatment with RA and caspase inhibitors, either DEVD or zVAD, induced differentiation of both NB4 and Zn-induced U937/PR9 cells, as shown by the nitroblue tetrazolium (NBT) dye reduction test (Fig4C) and the quantitative fluorescence-activated cell sorter (FACS) analysis of the expression of the CD11a surface differentiation antigen (Fig 4D). In control experiments performed in the absence of RA, caspase inhibitors (DEVD or zVAD) had no effect on the differentiation properties of these cells (not shown). Comparison of differentiation in cells treated with RA or RA plus caspase inhibitors revealed a slight, but consistent, synergistic effect of caspase inhibitors (Fig 4C and D). Remarkably, similar results were obtained using leukemic blasts from peripheral blood of newly diagnosed APL patients (Fig 5).

Fig. 4.

Time-dependent inhibition of 1 μmol/L RA-induced PML/RARα degradation by 100 μmol/L DEVD in (A) NB4 cells and (B) Zn-induced U937/PR9 cells. Immunoblot analysis was performed on total cellular proteins using an anti-RARα antibody (Santa Cruz Biotechnology). β-tubulin antibody was used for loading control. (C and D) Effect of DEVD and zVAD on cell differentiation in cells grown in the absence (black bars) or in the presence of 1 μmol/L RA for 96 hours (diagonal bars) evaluated by NBT reduction test (C) or quantitative expression of CD11a antigen (D).

Fig. 4.

Time-dependent inhibition of 1 μmol/L RA-induced PML/RARα degradation by 100 μmol/L DEVD in (A) NB4 cells and (B) Zn-induced U937/PR9 cells. Immunoblot analysis was performed on total cellular proteins using an anti-RARα antibody (Santa Cruz Biotechnology). β-tubulin antibody was used for loading control. (C and D) Effect of DEVD and zVAD on cell differentiation in cells grown in the absence (black bars) or in the presence of 1 μmol/L RA for 96 hours (diagonal bars) evaluated by NBT reduction test (C) or quantitative expression of CD11a antigen (D).

Close modal
Fig. 5.

Leukemic cells isolated from a newly diagnosed APL patient were treated for 96 hours with either 1 μmol/L RA or 100 μmol/L zVAD or with the combination of both agents as indicated. (A) Immunoblot analysis performed using the anti-RARα RPα(F) antibody; (B) percentage of differentiated cells determined by NBT reduction assay.

Fig. 5.

Leukemic cells isolated from a newly diagnosed APL patient were treated for 96 hours with either 1 μmol/L RA or 100 μmol/L zVAD or with the combination of both agents as indicated. (A) Immunoblot analysis performed using the anti-RARα RPα(F) antibody; (B) percentage of differentiated cells determined by NBT reduction assay.

Close modal
Inhibition of PML/RARα degradation by caspase inhibitors prevents reorganization of PML NBs.

RA treatment of APL blasts induces progressive disappearance of PML/RARα microspeckles, reorganization of PML NBs, and in consequence, relocalization of PML to its physiological site. Whether these phenomena are involved in RA differentiation and the role of PML/RARα degradation is not clear. Therefore, we analyzed the effect of DEVD on the subcellular localization of PML and PML/RARα in NB4 and in U937/PR9 cells by indirect immunofluorescence using the PG-M3 anti-PML monoclonal antibody.30 NB4 and Zn-treated U937/PR9 cells showed the microspeckled pattern of PML and PML/RARα expression typical of APL cells (Fig 6a and d). RA treatment restored the speckled pattern of PML NBs in both cell lines (Fig 6b and e). However, this effect was blocked by caspase 3 inhibitor DEVD (Fig 6c and f). In fact, cells treated with both RA and DEVD revealed a microspeckled pattern of anti-PML staining (Fig 6c and f). It appears that inhibition of PML/RARα degradation prevents the reorganization of PML NBs. These results imply that PML/RARα localization is not sensitive to RA and that restructuring of the PML NBs induced by RA might be the consequence of the degradation of PML/RARα within the microspeckles, rather than its relocalization to PML NBs. To directly test this hypothesis, we analyzed the effects of RA treatment independently on the localization of PML and PML/RARα using an U937 clone expressing the HA-tagged PML/RARα protein (clone HA-PR1), where PML/RARα and the endogenous PML stainings can be recognized by double immunostaining using α-HA and α-PML3 antibodies, respectively. The anti-PML3 polyclonal antibody recognizes a carboxyterminal, isoform-specific PML3 epitope not retained within PML/RARα.12 Immunofluorescence analysis performed in untreated U937-HA-PR1 cells showed that both PML and PML/RARα are expressed into a microspeckled pattern (Fig7a and b). After RA treatment, the immunofluorescence analysis of HA-PML/RARα–expressing cells using the α-HA antibody revealed a progressive disappearance of the labeling (Fig 7c). Immunostaining of the same cells with the α-PML3 antibody demonstrated the simultaneous reformation of the PML NBs pattern (compare the microspeckled pattern of staining in control cells with the speckled pattern in RA-treated cells, Fig 7b and d, respectively). These data confirm that PML/RARα expression is downregulated by RA and indicate that RA-induced PML/RARα degradation takes place into the microspeckles and that there is minimal, if any, RA-induced PML/RARα relocalization to the PML NBs.

Fig. 6.

Immunofluorescence analysis in NB4 (a through c) and Zn-induced U937/PR9 cells (e through g) using the PG-M3 anti-PML monoclonal antibody. Control cells (a and d) and cells treated for 96 hours with 1 μmol/L RA (b and e) or 1 μmol/L RA + 100 μmol/L DEVD (c and f).

Fig. 6.

Immunofluorescence analysis in NB4 (a through c) and Zn-induced U937/PR9 cells (e through g) using the PG-M3 anti-PML monoclonal antibody. Control cells (a and d) and cells treated for 96 hours with 1 μmol/L RA (b and e) or 1 μmol/L RA + 100 μmol/L DEVD (c and f).

Close modal
Fig. 7.

Immunofluorescence analysis in Zn-induced U937/HA-PR1 cells analyzed after 24 hours in the absence (a and b) or in the presence (c and d) of 1 μmol/L RA using the monoclonal anti-HA (a and c) or the polyclonal anti-PML3 antibodies (b and d).

Fig. 7.

Immunofluorescence analysis in Zn-induced U937/HA-PR1 cells analyzed after 24 hours in the absence (a and b) or in the presence (c and d) of 1 μmol/L RA using the monoclonal anti-HA (a and c) or the polyclonal anti-PML3 antibodies (b and d).

Close modal

It has been recently proposed that the PML/RARα fusion protein is degraded through the proteasome during RA treatment of APL cells and that this event is required for the differentiative activity of RA.16,18,19 Recent studies show that a proteasomal pathway can be upstream to caspase activation in thymocytes and neurons induced by different stimuli,25,26 suggesting a possible linkage between these two pathways.

In this study, we have elucidated the molecular mechanism and the biological significance of PML/RARα degradation by RA. We first show that a member of the caspase 3 family is responsible for PML/RARα proteolysis during RA treatment of APL cells. In fact, RA-induced degradation of PML/RARα can be prevented in vivo with the broad spectrum caspase inhibitor zVAD and the caspase 3 selective inhibitor tetrapeptide DEVD. Degradation of the fusion protein can be reconstituted in a cell-free system by using in vitro–translated PML/RARα and recombinant caspases. One major caspase cleavage site maps within the PML component of the fusion protein (α-helix; Asp 522), and the proteolytic event leaves the RARα component intact and potentially able to mediate RA-dependent transcriptional events. Notably, Asp 522 is not retained in a shorter variant of PML/RARα (bcr3-PML/RARα isoform)1,2 found in approximately 35% of patients.35,36 This short PML/RARα isoform is indeed resistant to RA-induced degradation.37 

The mechanisms involved in the triggering of caspase-mediated degradation of PML/RARα by RA treatment remain unclear. A caspase 3–like activity responsible for the proteolysis of PARP was detectable after treatment with RA in NB4 cells as well as in U937/PR9 cells induced to express PML/RARα. However, caspase activation also depends on PML/RARα expression, as shown by the finding that RA induces caspase activation in U937 cells only when the fusion protein is expressed. These findings are in agreement with recent data that shows that PARP polypeptide and its enzymatic activity decline dramatically during RA-induced differentiation in NB4 cells but not in HL-60 cells, a myeloblastic cell line that does not express PML/RARα.38 It appears, therefore, that the PML/RARα target protein is also required for RA-induced caspase activation in APL cells.

Caspase activation is a complex and still obscure process that is triggered by apoptotic signals.28,39 Bcl2 family members control the mitochondrial APAFs and the activation of caspase 3.28,39-41 Downregulation of Bcl2 by RA has been reported for some myeloid cells,42-44 and it is a constant and more pronounced phenomenon in PML/RARα-expressing cells.37,42,45 Therefore, Bcl2 might be a direct molecular target of PML/RARα, and its RA-induced downregulation might contribute to caspase activation and consequent PML/RARα degradation. Notably, RA-induced downregulation of Bcl2 and caspase activation is not followed by apoptotic cell death in APL cells,33,45-47thereby suggesting that additional molecular events are required to complete the apoptotic program.

Finally, these results show that APL cell differentiation might occur despite the persistence of PML/RARα expression. Oligopeptides that are selective caspase inhibitors could effectively prevent RA-induced PML/RARα degradation without impairing RA-induced differentiation in NB4 cells, in U937/PR9 cells, and in cells derived from APL patients, as evaluated by morphology, NBT reduction, and quantitative FACS analysis of the CD11a surface antigen expression. Indeed, in the presence of caspase inhibitors, we observed increased expression of the fusion protein and a parallel increased differentiative effect of RA, thereby suggesting an active contribution of PML/RARα to RA-induced differentiation. This is supported by the fact that RA induces cell differentiation and clinical remission also in APL patients expressing the short isoform of PML/RARα (bcr3),35,36 which appears to be resistant to RA-induced degradation.37 

Inhibition of PML/RARα degradation largely maintained the microspeckled localization of the fusion protein preventing the reorganization of the PML NBs. Thus, in APL cells, RA-induced reassembly of NBs seems to be the consequence of the disappearance of PML/RARα within microspeckles and may not be required for myeloid differentiation. Taken together these data suggest that RA might convert PML/RARα to an active inducer of myeloid differentiation, possibly by triggering its trascriptional activator function on specific RA-target genes.

It has been recently shown that unligated nuclear receptors, including RARs, are associated with coregulatory proteins such as N-CoR and SMRT that can act as transcriptional corepressors. SMRT and N-CoR associate with the corepressor Sin3 and the histone deacetylase protein to form a transcriptional repressor complex.48,49 RA binding releases this complex and recruits the multisubunit activation complex, which possesses histone acetyltransferase activity.48 PML/RARα has been found to be associated with transcriptional corepressors that can dissociate from the fusion products at much higher RA concentrations than from wild-type RARα.50-54 In agreement with these findings, we propose that in APL cells, RA binding to PML/RARα induces allosteric changes of the fusion protein that release the corepressors and recruit the activator complex resulting in histone acetylation, modification of gene expression, and cell differentiation. The same allosteric changes in PML/RARα conformation might also allow specific PML/RARα amino acid sequences to be cleaved by caspases.

We gratefully acknowledge Wilson Miller, Giulio Cossu, and Angelika Rosenauer for helpful suggestions and discussions. We also thank P. Chambon, M. Lanotte, K. Schulze-Osthoff, R. Beyaert, and Sara Droetto for providing reagents.

Supported by grants from the Associazione Italiana per la Ricerca sul Cancro (AIRC), Ministero Università e Ricerca Scientifica e Tecnologica (MURST), CNR Biotechnology and European Community (Biomed).

Address reprint requests to Clara Nervi, MD, PhD, Dipartimento di Istologia ed Embriologia Medica, University of Rome “La Sapienza,” Via A. Scarpa 14, 00161 Rome, Italy; e-mail:nervi@axrma.uniroma1.it.

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

1
Grignani
F
Fagioli
M
Alcalay
M
Longo
L
Pandolfi
PP
Donti
E
Biondi
A
Grignani
F
Lo Coco
F
Pelicci
PG
Acute promyelocytic leukemia: From genetics to treatment.
Blood
83
1994
10
2
Warrell RP Jr
de Thè
H
Wang
ZY
Degos
L
Acute promyelocytic leukemia.
New Engl J Med
329
1993
177
3
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
76
1994
333
4
Weis
K
Rambaud
S
Lavau
C
Jansen
J
Carvalho
T
Carmo-Fonseca
M
Lamond
A
Dejean
A
Retinoic acid regulates aberrant nuclear localization of PML-RARα in acute promyelocytic leukemia cells.
Cell
76
1994
345
5
Koken
MHM
Puvio-Dutilleul
F
Guillemin
MC
Viron
G
Cruz-Linares
G
Stuurman
N
de Jong
L
Szostecki
C
Calvo
F
Chomienne
C
Degos
L
Puvion
E
de Thè
H
The t(15;17) translocation alters a nuclear body in a retinoic acid-reversible fashion.
EMBO J
13
1994
1073
6
Mu
ZM
Chin
KV
Liu
JH
Lozano
G
Chang
KS
PML, a growth suppressor disrupted in acute promyelocytic leukemia.
Mol Cell Biol
14
1994
6858
7
Mangelsdorf DJ, Umesono K, Evans RM: The retinoid receptors, in Sporn MB, Roberts AB, Goodman DS (eds): The Retinoids: Biology, Chemistry, and Medicine. New York, NY, Raven, 1994, p 319
8
Kastner P, Leid M, Chambon P: Role of nuclear retinoid receptors in the regulation of gene expression, in Blomhoff R (ed): Vitamin A in Health and Disease. New York, NY, Marcel Dekker, 1994, p 189
9
Nervi
C
Grippo
JF
Sherman
MI
George
MD
Jetten
AM
Identification and characterization of nuclear retinoic acid-binding activity in human myeloblastic leukemia HL-60 cells.
Proc Natl Acad Sci USA
86
1989
5854
10
Tsai
S
Bartelmez
S
Heyman
RA
Damm
K
Evans
RM
Collins
SJ
A mutated retinoic acid receptor-α exhibiting dominant-negative activity alters the lineage development of a multipotent hematopoietic cell line.
Genes Dev
6
1992
2258
11
Grignani
F
Ferrucci
PF
Testa
U
Talamo
G
Fagioli
M
Alcalay
M
Mencarelli
A
Peschle
C
Nicoletti
I
Pelicci
PG
The acute promyelocytic leukaemia specific PML/RARα fusion protein inhibits differentiation and promotes survival of myeloid precursor cells.
Cell
74
1993
423
12
Grignani
F
Testa
U
Rogaia
D
Ferrucci
PF
Samoggia
P
Pinto
A
Aldinucci
D
Gelmetti
V
Fagioli
M
Alcalay
M
Seeler
J
Grignani
F
Nicoletti
I
Peschle
C
Pelicci
PG
Effects on differentiation by the promyelocytic leukemia PML/RARα protein depend on the fusion of the PML protein dimerization and RARα DNA binding domains.
EMBO J
15
1996
4949
13
Brown
D
Scott
K
Lagasse
E
Weissman
I
Alcalay
M
Pelicci
PG
Atwater
S
Bishop
M
A PML/RARα transgene initiates murine acute promyelocytic leukemia.
Proc Natl Acad Sci USA
945
1997
2551
14
Grisolano
JL
Wesselschmidt
RL
Pelicci
PG
Ley
TJ
Altered myeloid development and acute leukemia in transgenic mice expressing PML/RARα under control of catepsin G regulatory sequences.
Blood
89
1997
376
15
He
LI
Tribioli
C
Rivi
R
Peruzzi
D
Pelicci
PG
Soares
V
Cattoretti
G
Pandolfi
PP
Acute leukemia with promyelocytic features in PML/RARα transgenic mice.
Proc Natl Acad Sci USA
94
1997
5302
16
Fenaux
P
Degos
L
Differentiation therapy for acute promyelocytic leukemia.
New Engl J Med
337
1997
1076
17
Degos
L
Dombret
H
Chomienne
C
Daniel
MT
Micléa
JM
Chastang
C
Castaigne
S
Fenaux
P
All-trans-retinoic acid as a differentiating agent in the treatment of acute promyelocytic leukemia.
Blood
85
1995
2643
18
Raelson
JV
Nervi
C
Rosenauer
A
Benedetti
L
Monczak
Y
Pearson
M
Pelicci
PG
Miller WH Jr
The PML/RARα oncoprotein is a direct molecular target of retinoic acid in acute promyelocytic leukemia cells.
Blood
88
1996
2826
19
Yoshida
H
Kitamura
K
Tanaka
K
Omura
S
Miyazaki
T
Hachiya
T
Ohno
R
Naoe
T
Accelerated degradation of PML-retinoic acid receptor α (PML-RARA) oncoprotein by all-trans-retinoic acid in acute promyelocytic leukemia: Possible role of the proteasome pathway.
Cancer Res
38
1996
2945
20
Rosenauer
A
Raelson
JV
Nervi
C
Eydoux
P
Deblasio
A
Miller WH Jr
Alteration in expression, binding to ligand and DNA, and transcriptional activity of rearranged and wild-type retinoid receptors in retinoid-resistant acute promyelocytic leukemia cell lines.
Blood
88
1996
2671
21
Dermine
S
Grignani
F
Clerici
M
Nervi
C
Sozzi
G
Talamo
GP
Marchesi
E
Formelli
F
Parmiani
G
Pelicci
PG
Gambacorti-Passerini
C
The occurrence of resistance to retinoic acid in the acute promyelocytic leukemia cell line NB4 is associated with altered expression of the PML/RARα protein.
Blood
82
1993
1573
22
Shao
W
Benedetti
L
Lamph
WW
Nervi
C
Miller WH Jr
A retinoid-resistant acute promyelocytic leukemia subclone expresses a dominant negative PML-RARα mutation.
Blood
89
1997
4282
23
Boddy
M
Howe
K
Etkin
LD
Solomon
E
Freemont
PS
PIC-1 a novel ubiquitin-like protein which interacts with the PML component of a multiprotein complex that is disrupted in acute promyelocytic leukemia.
Oncogene
13
1996
971
24
Muller
S
Matunis
MJ
Dejean
A
Conjugation with the ubiquitin-related modifier SUMO-1 regulates the partitioning of PML within the nucleus.
EMBO J
17
1998
61
25
Sadoul
R
Fernandez
PA
Quiquerez
AL
Martinou
I
Maki
M
Schroter
M
Becherer
JD
Irmler
M
Tschopp
J
Martinou
JC
Involvment of the proteasome in the programmed cell death of NGF-deprived sympathetic neurons.
EMBO J
15
1996
3845
26
Grimm
LM
Goldberg
AL
Poirer
GG
Schwartz
LM
Osborne
BA
Proteasomes play an essential role in thymocyte apoptosis.
EMBO J
15
1996
3835
27
Alnemri
ES
Livingston
DJ
Nicholson
DW
Salvesen
G
Thornberry
NA
Wong
WW
Yuan
J
Human ICE/CED-3 Protease Nomenclature.
Cell
87
1996
171
28
Nicholson
DW
Thornberry
NA
Caspases: Killer proteases.
Trends Biochem Sci
22
1997
299
29
Rochette-Egly
C
Lutz
Y
Saunders
M
Gaub
MP
Chambon
P
Retinoid acid receptor gamma specific immunodetection and phosphorylation.
J Cell Biol
115
1991
535
30
Flenghi
L
Fagioli
M
Tomassoni
L
Pileri
S
Gambacorta
M
Pacini
R
Grignani
F
Casini
T
Ferrucci
PF
Martelli
MF
Pelicci
PG
Falini
B
Characterization of a new monoclonal antibody (PG-M3) directed against the aminoterminal portion of the PML gene product: Immunocytochemical evidence for high expression of PML proteins on activated macrophages, endothelial cells, and epithelia.
Blood
85
1995
1871
31
Beyaert
R
Kidd
VJ
Cornelis
S
Van de Craen
M
Denecker
G
Lahti
JM
Gururajan
R
Vandeabeele
P
Fiers
W
Cleavage of PITSLRE kinases by ICE/CASP-1 and CPP32/CASP-3 during apoptosis induced by tumor necrosis factor.
J Biol Chem
272
1997
11694
32
Lanotte
M
Martin-Thouvenin
V
Najman
S
Ballerini
P
Valensi
F
Berger
R
NB4, a maturation inducible cell line with t(15;17) marker isolated from a human acute promyelocytic leukemia (M3).
Blood
77
1991
1080
33
Benedetti
L
Grignani
F
Scicchitano
BM
Jetten
AM
Diverio
D
Lo Coco
F
Avvisati
G
Gambacorti-Passerini
C
Adamo
S
Levin
AA
Pelicci
PG
Nervi
C
Retinoid-induced differentiation of acute promyelocytic leukemia involves PML-RARα-mediated increase of type II transglutaminase.
Blood
87
1996
1939
34
Pronk
GJ
Ramer
K
Amiri
P
Williams
LT
Requirement of an ICE-like protease for induction of apoptosis and ceramide generation by REAPER.
Science
271
1996
808
35
Gallagher
RE
Willman
CL
Slack
JL
Andersen
J
Li
YP
Viswanatha
D
Bloomfield
CD
Appelbaum
FR
Schiffer
CA
Tallman
MS
Wiernik
PH
Association of PML-RARα fusion mRNA type with pretreatment hematologic characteristics but not treatment outcome in acute promyelocytic leukemia: An intergroup molecular study.
Blood
90
1997
1656
36
Mandelli
F
Diverio
D
Avvisati
G
Luciano
A
Barbui
T
Bernasconi
C
Broccia
G
Cerri
R
Falda
M
Fioritoni
G
Leoni
F
Liso
V
Petti
MC
Rodeghiero
F
Saglio
G
Vegna
ML
Visani
G
Jehn
U
Willemze
R
Muus
P
Pelicci
PG
Biondi
A
Lo Coco
F
Molecular remission in PML/RARα-positive acute promyelocytic leukemia by combined all-trans-retinoic acid and idarubicin (AIDA) therapy.
Blood
90
1997
1014
37
Slack
JL
Yu
M
Constitutive expression of the promyelocytic leukemia-associated oncogene PML-RARα in TF1 cells: Isoform-specific and retinoic acid-dependent effects on growth, bcl-2 expression, and apoptosis.
Blood
91
1998
3347
38
Bhatia
M
Kirkland
JB
Meckling-Gill
KA
Modulation of poly(ADP-ribose) polymerase during neutrophilic and monocytic differentiation of promyelocytic (NB4) and myelocytic (HL-60) leukaemia cells.
Biochem J
308
1995
131
39
Rao
L
White
E
Bcl-2 and the ICE family of apoptotic regulators: Making a connection.
Curr Opin Genet Dev
7
1997
52
40
Vaux
DL
CED-4. The third horseman of apoptosis.
Cell
90
1997
389
41
Reed
JC
Double identity for proteins of the Bcl-2 family.
Nature
387
1997
773
42
Bruel
A
Benoit
G
De Nay
D
Brown
S
Lanotte
M
Distinct apoptotic response in maturation sensitive and resistant t(15;17) acute promyelocytic leukemia NB4 cells. 9-cis retinoic acid induces apoptosis independent of maturation and Bcl-2 expression.
Leukemia
9
1995
1173
43
Park
JR
Robertson
K
Hickstein
DD
Schickwann
T
Hockenbery
DM
Collins
SJ
Dysregulated bcl-2 expression inhibits apoptosis but not differentiation of retinoic acid-induced HL-60 granulocytes.
Blood
84
1994
440
44
Hu
ZB
Minden
MD
McCulloch
EA
Direct evidence for the partecipation of bcl-2 in the regulation by retinoic acid of the Ara-C sensitivity of leukemic stem cells.
Leukemia
9
1995
1667
45
Calbresse
C
Barbey
S
Venturini
L
Balitrand
N
Degos
L
Fenaux
P
Chomienne
C
In vitro treatment with retinoids or the topoisomerase inhibitor, VP-16, evidences different functional apoptotic pathways in acute promyelocytic leukemic cells.
Leukemia
9
1995
2049
46
Shao
W
Fanelli
M
Ferrara
FF
Riccioni
R
Rosenauer
A
Davison
K
Lamph
WW
Waxman
S
Pelicci
PG
Lo Coco
F
Avvisati
G
Testa
U
Peschle
C
Gambacorti-Passerini
C
Nervi
C
Miller WH Jr
Arsenic trioxide as an inducer of apoptosis and loss of PML/RARα protein in acute promyelocytic leukemia cells.
J Natl Cancer Inst
90
1998
124
47
Zhu
J
Koken
MHM
Quignon
F
Chelbi-Alix
M
Degos
L
Wang
ZY
Chen
Z
de Thè
H
Arsenic-induced PML targeting onto nuclear bodies: Implications for the treatment of acute promyelocytic leukemia.
Proc Natl Acad Sci USA
94
1997
3978
48
Glass
CK
Rose
DW
Rosenfeld
MG
Nuclear receptor coactivators.
Curr Opin Cell Biol
9
1997
222
49
Horlein
AJ
Naar
AM
Heinzel
T
Torchia
J
Gloss
B
Kurokawa
R
Ryan
A
Kamei
Y
Soderstrom
M
Glass
CK
Rosenfeld
MG
Ligand-dependent repression by the thyroid hormone receptor mediated by a nuclear receptor co-repressor.
Nature
377
1995
397
50
Hong
SH
David
G
Wong
CW
Dejean
A
Privalsky
ML
SMRT corepressor interacts with PLZF and with the PML-retinoic acid receptor α (RARα) and PLZF/RARα oncoproteins associated with acute promyelocytic leukemia.
Proc Natl Acad Sci USA
94
1997
9028
51
Grignani
F
De Matteis
S
Nervi
C
Tomassoni
L
Gelmetti
V
Cioce
M
Fanelli
M
Ruthardt
M
Ferrara
FF
Zamir
I
Seiser
C
Grignani
F
Lazar
MA
Minucci
S
Pelicci
PG
Fusion proteins of the retinoic acid receptor-α recruit histone deacetylase in promyelocytic leukaemia.
Nature
391
1998
815
52
Lin
RJ
Nagy
L
Inoue
S
Shao
W
Miller WH Jr
Evans
RM
Role of the histone deacetylase complex in acute promyelocytic leukaemia.
Nature
391
1998
811
53
He
LZ
Guidez
F
Tribioli
C
Peruzzi
D
Ruthardt
M
Zelent
A
Pandolfi
PP
Distinct interactions of PML-RARα and PLZF-RARα with co-repressors determine differential response to RA in APL.
Nat Genet
18
1998
126
54
Guidez
F
Ivins
S
Zhu
J
Soderstrom
M
Waxman
S
Zelent
A
Reduced retinoic acid-sensitivities of nuclear receptor corepressor binding to PML- and PLZF-RARα underlie molecular pathogenesis and treatment of acute promyelocytic leukemia.
Blood
91
1998
2634
Sign in via your Institution