The most common chromosomal translocation in acute promyelocytic leukemia (APL), t15;17(q22;q21), creates PMLRAR andRARPML fusion genes. We previously developed a mouse model of APL by expressing PMLRAR in murine myeloid cells. In order to examine the mechanisms by which PMLRAR can initiate leukemia, we have now generated transgenic mice expressingPMLRARm4 and RARm4, proteins that are unable to activate transcription in response to retinoic acid.PMLRARm4 transgenic mice developed myeloid leukemia, demonstrating that transcriptional activation by PMLRAR is not required for leukemic transformation. The characteristics of the leukemias arising in the PMLRARm4 transgenic mice varied from those previously observed in our PMLRAR transgenic mice, indicating that ligand responsiveness may influence the phenotype of the leukemic cells. The leukemias that arose in PMLRARm4transgenic mice did not differentiate in response to retinoic acid therapy. This result supports the hypothesis that a major therapeutic effect of retinoic acid is mediated directly through thePMLRAR protein. However, a variable effect on survival suggested that this agent may be of some benefit in APL even when leukemic cells are resistant to its differentiative effects. Transgenic mice expressing high levels of RARm4 have not developed leukemia, providing evidence that the PML domain ofPMLRAR plays a specific and critical role in the pathogenesis of APL.

Retinoids are signaling molecules with significant roles in development and differentiation.1,2 These biologic effects led to the hypothesis that retinoids might be useful agents in the treatment of human malignancies. Hence, retinoids have been evaluated as possible therapies for a variety of human neoplasms including leukemias, skin cancers, cervical cancer, and neuroblastomas.3,4 Among myeloid leukemias, acute promyelocytic leukemia (APL) was found to be particularly sensitive to retinoic acid5 and more than a decade has passed since the demonstration that all-trans retinoic acid (tRA) could induce remission in patients with APL by stimulating differentiation of the leukemic cells.6,7 Understanding the pathogenesis and retinoid responsiveness of APL is important for expanding the application of retinoids in cancer treatment and for developing additional differentiation therapies.

In 1977 Rowley and colleagues8 described a specific association of APL with a t(15;17) chromosomal translocation. Subsequent to the demonstration of the therapeutic benefit of tRA in APL, the breakpoint on chromosome 17 was identified to be within a gene encoding a retinoic acid receptor, RARα.9-12 The breakpoint on chromosome 15 was identified to be within a novel gene,PML.13-17 Expression of the PMLRARαfusion is a consistent feature of the disease in the vast majority of APL patients.18 Since these discoveries, efforts have been directed at understanding the role of PMLRARα in leukemogenesis and response to therapy.

In addition to the common t(15;17) translocation, other chromosomal translocations have been identified in rare cases of APL. These translocations also result in fusions to RARα and include fusions with PLZF in t(11;17)(q23;q21),19,NPM in t(5;17)(q32;q21),20 and NuMA in t(11;17)(q13;q21).21 The partners of RARαin the APL fusions are all nuclear but otherwise have limited commonality. This fact raises the possibility that all the translocations contribute to APL pathogenesis by generating abnormal retinoic acid receptors that share common transcriptional properties.

RARα is a ligand inducible transcription factor. In the absence of its ligand, tRA, RARα generally acts as a transcriptional repressor by recruiting corepressor molecules, including SMRT and N-CoR, which in turn recruit histone deacetylases. In the presence of ligand, RARα generally acts to induce transcription by releasing corepressor molecules and recruiting coactivators.22 When compared with RARα,PMLRARα has context dependent effects on transcription.13,15,17,23-25 For example, depending on cell type and the transcriptional element assayed, PMLRARα can decrease or increase basal transcription in the absence of ligand. Similarly, PMLRARα can exhibit both dominant negative activity and superactivation in the presence of tRA. Whether transcriptional activation and/or transcriptional repression byPMLRARα is necessary or sufficient for leukemogenesis has not been experimentally addressed.

Although almost all APL patients respond to tRA therapy, resistance to this agent often develops in patients so treated.26,27 It has been suggested that enhanced metabolism of tRA, increased expression of the cellular retinoic acid binding protein II, and increased expression of the multidrug resistance gene product may contribute to clinical tRA resistance (reviewed in Ding et al28 and Imaizumi et al29). However, alterations of the PMLRARα protein itself have been described in some retinoic acid resistant subclones of the NB4 APL cell line, as well as in some patients with a disease that was clinically resistant to tRA.28-31 The observed mutations in PMLRARαincluded amino acid changes that impair the ability of the protein to bind retinoic acid and to activate transcription. These findings provided evidence that loss of ligand responsiveness byPMLRARα can play a role in clinical tRA resistance.

We previously developed a murine myeloid leukemia model that recapitulates many of the features of APL.32 We have now generated additional transgenic mice to assess the role of hormone responsiveness by PMLRARα in both leukemogenesis and tRA response, as well as the sufficiency of transcriptional repression at retinoic acid response elements (RAREs) in initiation of leukemia.

Preparation of plasmid constructs

The m4 mutation was introduced into the PMLRARα andRARα open-reading frames by a 2-step polymerase chain reaction (PCR) mutagenesis protocol, using 2 mutagenic primers, 5′-GATCACGCCGAAGATGGAGATCCC-3′ and 5′-ATCTTCGGC GTGATCACCCGCTC-3′. The resulting PCR-generated fragment was digested with Bcl I and Xba I and was transferred into the pSG5-RARα or pSG5-PMLRARα background. The results of mutagenesis were verified by sequencing. A pGEX-KG construct was used to express either glutathione S-transferase (GST) or a GST-SMRT fusion (encoding codons 751-1495 of human SMRT) inEscherichia coli.33 

Protease resistance assay

35S-radiolabeled PMLRARα andPMLRARαm4 mutant proteins were synthesized in vitro by the TnT procedure. For the trypsin assay, 1 μL aliquots of in vitro translation product were diluted to a final volume of 20 μL each with 50 mmol/L Tris-Cl (pH 7.4) containing either tRA or an equivalent volume of ethanol carrier. Proteolysis was initiated by adding from 0 to 8 μgs of trypsin-TPCK per sample (trypsin pretreated with tosyl-L-phenylalanine chloromethyl ketone). The samples were then incubated at room temperature for 10 minutes. The proteolysis was terminated by addition of 14 μL of 5 × denaturing polyacrylamide gel electrophoresis (PAGE) sample buffer and the samples were rapidly frozen on dry ice. The samples were subsequently thawed, boiled for 10 minutes, resolved by denaturing PAGE, and visualized by autoradiography.

Transient transfections

CV-1 cell transfections were performed by a lipofection method as recommended by the manufacturer (Lipofectin, Gibco-BRL). Approximately 7 × 104 cells were transfected with 25 ng of the pSG5-RARα or pSG5-PMLRARα plasmids (representing “wild-type” or the m4 mutant), 100 ng of pCMV-lacZ (used as an internal normalization control for the efficiency of the transfection procedure) and 100 ng of the ptk-luciferase-βRARE reporter. Five hours after transfection, the cells were transferred into media either lacking or containing 1 μmol tRA. Cells were harvested 48 hours after transfection and the levels of luciferase and β-galactosidase were determined.34,35 

In vitro receptor/corepressor binding assays

GST-fusion proteins were expressed in E coli and were purified and immobilized by binding to glutathione agarose as previously described.34,35S-methionine–labeled full-length RARα, RARαm4, PMLRARα, andPMLRARαm4 proteins were synthesized by a coupled in vitro transcription and translation system (Promega TnT kit, Promega, Madison, WI). The radiolabeled proteins were subsequently incubated with the immobilized GST fusion proteins in HEMG buffer in the presence or absence of tRA, the agarose matrix was extensively washed and bound proteins were eluted with free glutathione and analyzed by denaturing PAGE.33 The electrophoretograms were visualized and quantified by phosphorimager analysis (Molecular Dynamics STORM system, Molecular Dynamics, Sunnyvale, CA).

Generation of transgenic mice

The human PMLRARαm4 and RARαm4 cDNAs were cloned into the hMRP8 expression cassette.36 Transgenic animals were prepared following standard procedures37 from inbred FVB/N mice.38 

Western blotting and immunofluorescence

Western blotting was performed as previously described with a rabbit polyclonal antiserum raised against a GST-fusion protein, encompassing amino acids 420-462 of the human RARα protein (anti-RARαF).32,39 Whole-cell lysates of bone marrow from control and transgenic mice were subjected to denaturing PAGE on 8% or 12% SDS-polyacrylamide gels and were transferred to nitrocellulose. Immunofluorescence analysis of bone marrow cells was performed essentially as described40 but using the anti-RARαF antiserum at a 1:150 dilution.

Isolation of cells from tissues, cell staining, and fluorescence-activated cell sorting

These were performed as previously described.32,40 In addition, Sudan Black B staining was performed using reagents from Sigma, according the manufacturer's directions.

Peripheral blood counts

Blood was analyzed on a Hemavet veterinary hematology analyzer to assess white blood cell counts, hemoglobin, and platelet counts. White blood cell differential counts were performed on peripheral blood smears.

Methylcellulose cultures

Bone marrow cells were cultured in duplicate in 35 mm petri dishes in Methocult M3230 methylcellulose medium (StemCell Technologies, Vancouver, BC) supplemented with either 50 units/mL G-CSF (Boehringer Mannheim), or 2.5 ng/mL GM-CSF (StemCell Technologies) plus 2% Xg63Ag8-653-IL341 conditioned medium. One milliliter cultures contained 5 × 104 viable bone marrow cells. Analysis was as previously described.40 

Transplantations

Cells isolated from bone marrow and spleens of leukemic animals were resuspended in buffered saline and injected into the tail veins of 6- to 12-week-old FVB/N mice, 5 × 106 viable cells/recipient. Nonleukemic bone marrow isolated from PMLRARαm4transgenic founder #4048 was transplanted into lethally irradiated FVB/N mice as previously described.32 

Treatment with all-trans retinoic acid

Leukemic mice were treated by subcutaneous implantation of 21-day release pellets containing 5 mg tRA or placebo (Innovative Research of America). Morphologic differentiation by tRA was assessed on days 4 and 11 of therapy.

Generation of transgenic mice

We generated transgenic mice expressing a PMLRARα unable to activate transcription as well as transgenic mice expressing anRARα with dominant negative activity. For this purpose, we introduced a Leu to Pro mutation at amino acid 398 of RARαinto cDNAs encoding PMLRARα andRARα (Figure 1A). This mutation was originally identified by Shao and colleagues31in a retinoic acid resistant subclone of human APL cells (NB4-R4) and was designated the m4 mutation. The m4 mutation impairs ligand binding, abrogates ligand-induced transcriptional activation, and blocks ligand-induced release of SMRT corepressor.31,42Furthermore, PMLRARαm4 and RARαm4 act as dominant negative inhibitors of tRA-induced transcription.31 

Fig. 1.

PMLRARm4 and RARm4.

A, PMLRARαm4: the Leu to Pro point mutation at codon 398 ofRARα was introduced into a human PMLRARα cDNA whose chromosome 15 breakpoint lies in breakpoint cluster 1.16The PML portion of the fusion is shown with hatching with selected structural domains labeled and shown in white. The B-F domains that encompass the RARα portion of the fusion are labeled and functional regions are noted. RARαm4: the Leu to Pro point mutation was introduced into a human RARα1 cDNA. B, Hormone binding by PMLRARα (“Wild-type,” WT) andPMLRARαm4 (m4 mutant). Radiolabeled proteins synthesized by in vitro transcription and translation were incubated without or with increasing amounts of trypsin (indicated above the panels) in the absence or presence of 1 μmol tRA. The protein products were resolved by denaturing PAGE and visualized by autoradiography. The arrows show the position of the full-length undigested proteins. Smaller bands represent partially degraded products. C, Dominant negative activity of the m4 mutant proteins. CV-1 cells were transiently transfected with pSG5 constructs containing no exogenous receptor, RARα (WT),RARαm4, PMLRARα (WT), and PMLRARαm4. Luciferase activity expressed from a cotransfected βRARE-luciferase reporter gene was normalized to β-galactosidase activity from a cotransfected pCH210-LacZ plasmid. D, Decreased hormone-induced dissociation of SMRT corepressor by the m4 mutant proteins. GST-SMRT fusion protein was synthesized in E coli and was immobilized on glutathione agarose. The different receptor proteins were synthesized by in vitro transcription and translation and were incubated with the immobilized GST-SMRT in the absence or presence of 1 μmol tRA, as indicated below the panels. Equivalent amounts of GST-SMRT and radiolabeled receptor protein were used for each panel. Nonrecombinant GST, immobilized on glutathione agarose, was used in parallel as a negative control. The radiolabeled receptors remaining bound to the GST or GST-SMRT matrix after washing were eluted, were resolved by denaturing PAGE, and were visualized and quantified by phosphorimager analysis. The amount of radiolabeled receptor bound to the GST or GST-SMRT matrix, relative to the amount of receptor input in each binding reaction, is displayed beneath each panel.

Fig. 1.

PMLRARm4 and RARm4.

A, PMLRARαm4: the Leu to Pro point mutation at codon 398 ofRARα was introduced into a human PMLRARα cDNA whose chromosome 15 breakpoint lies in breakpoint cluster 1.16The PML portion of the fusion is shown with hatching with selected structural domains labeled and shown in white. The B-F domains that encompass the RARα portion of the fusion are labeled and functional regions are noted. RARαm4: the Leu to Pro point mutation was introduced into a human RARα1 cDNA. B, Hormone binding by PMLRARα (“Wild-type,” WT) andPMLRARαm4 (m4 mutant). Radiolabeled proteins synthesized by in vitro transcription and translation were incubated without or with increasing amounts of trypsin (indicated above the panels) in the absence or presence of 1 μmol tRA. The protein products were resolved by denaturing PAGE and visualized by autoradiography. The arrows show the position of the full-length undigested proteins. Smaller bands represent partially degraded products. C, Dominant negative activity of the m4 mutant proteins. CV-1 cells were transiently transfected with pSG5 constructs containing no exogenous receptor, RARα (WT),RARαm4, PMLRARα (WT), and PMLRARαm4. Luciferase activity expressed from a cotransfected βRARE-luciferase reporter gene was normalized to β-galactosidase activity from a cotransfected pCH210-LacZ plasmid. D, Decreased hormone-induced dissociation of SMRT corepressor by the m4 mutant proteins. GST-SMRT fusion protein was synthesized in E coli and was immobilized on glutathione agarose. The different receptor proteins were synthesized by in vitro transcription and translation and were incubated with the immobilized GST-SMRT in the absence or presence of 1 μmol tRA, as indicated below the panels. Equivalent amounts of GST-SMRT and radiolabeled receptor protein were used for each panel. Nonrecombinant GST, immobilized on glutathione agarose, was used in parallel as a negative control. The radiolabeled receptors remaining bound to the GST or GST-SMRT matrix after washing were eluted, were resolved by denaturing PAGE, and were visualized and quantified by phosphorimager analysis. The amount of radiolabeled receptor bound to the GST or GST-SMRT matrix, relative to the amount of receptor input in each binding reaction, is displayed beneath each panel.

Close modal

Before producing transgenic animals, we validated the characteristics of our PMLRARαm4 and RARαm4 cDNA constructs, comparing our results with those previously reported. The effects of the m4 mutation on ligand binding had been assessed by evaluating the binding of 3H-tRA to PMLRARαm4 orRARαm4 in nuclear extracts of transiently transfected Cos-1 cells.31 In these assays, no ligand binding was observed. We used a sensitive protease-resistance assay to determine whether the m4 mutation fully abolished ligand binding. Condensation of a hormone-binding domain of a nuclear receptor around the hormone ligand can result in a protease-resistant core. Gain of protease resistance has been seen with many nuclear hormone receptors and has been used as a measure of ligand occupancy.43,44 RadiolabeledPMLRARα and PMLRARαm4 protein were incubated with increasing amounts of trypsin in the absence or presence of 1 μmol tRA. The products were then resolved by denaturing PAGE and visualized by phosphor imaging (Figure 1B). PMLRARα was readily degraded by proteolytic treatment in the absence of hormone, but produced a protease-resistant polypeptide in the presence of hormone.PMLRARαm4 also exhibited some protease resistance in the presence of 1 μmol tRA, although it was less resistant thanPMLRARα. Ten- to 20-fold higher concentrations of hormone were required to produce a resistant polypeptide fromPMLRARαm4 than from PMLRARα (data not shown).

Although tRA was able to bind weakly to PMLRARαm4, when we examined the effects of the m4 mutation on transcriptional activity and association with SMRT corepressor, our results were similar to published analyses. PMLRARα, PMLRARαm4,RARα, and RARαm4 were transiently expressed in CV-1 cells and transcription from a βRARE response element in the absence or presence of tRA was assessed. In contrast to PMLRARα andRARα, the m4 mutant proteins strongly inhibited transcriptional activation (Figure 1C). In cell-free protein assays to measure receptor interaction with SMRT, the m4 mutants, in contrast toRARα, were unable to release the corepressor on addition of 1 μmol tRA (Figure 1D). As has been previously reported,42,45 PMLRARα was itself somewhat less efficient at releasing corepressor than was RARα.

The PMLRARαm4 and RARαm4 cDNAs were cloned into theMRP8 expression vector we had used to generatePMLRARα transgenic mice.32 As with our earlier experiments, transgenic mice were produced in the FVB/N inbred background.38 Injections yielded 7MRP8-PMLRARαm4 and 8 MRP8-RARαm4 transgenic animals.

Expression of the transgenes

The MRP8 promoter element can drive transgene expression in myeloid cells, including myeloblasts, neutrophils, and monocytes.32,36,40 Western blotting of bone marrow was performed using a rabbit polyclonal antiserum raised to humanRARαF.32 The results are summarized in Tables1 and 2, and representative data are shown (Figure 2A and B). Although the murine peptide differs from the human by only 4 of 43 amino acids, the antiserum recognizes murine RARα poorly and as a result endogenous murine RARα is not seen on these blots. PMLRARαm4 protein was present in 5 of 7 lines ofMRP8-PMLRARαm4 transgenic mice analyzed. Levels of expression in 2 of the lines appeared comparable to levels of PMLRARα in our highest expressing MRP8-PMLRARα mice (Figure 2A).RARαm4 protein was present in 5 of 7 lines ofMRP8-RARαm4 transgenic mice analyzed. Levels of expression in 2 of the lines appeared to exceed the levels of PMLRARα in our highest expressing MRP8-PMLRARα mice (Figure 2B).

Table 1.

PMLRARm4 transgenic mice

Mouse Transgene Expression in Bone Marrow Age at Death (days) Reason for Death
4042  +  93 Leukemia  
4048* ++  37  Abnormal Skin Growth 
 4048.2   189  Leukemia  
 4048.3   215 Leukemia  
 4048.4   259  Leukemia  
 4048.5  328  Leukemia  
 4048.6   341  Abdominal Sarcoma 
4099  ++  156  Leukemia  
4100  − 548  Study1-153 
4104  +  235  Leukemia  
4105  235  Abnormal Skin Growth  
4114  −  235  Abnormal Skin Growth 
Mouse Transgene Expression in Bone Marrow Age at Death (days) Reason for Death
4042  +  93 Leukemia  
4048* ++  37  Abnormal Skin Growth 
 4048.2   189  Leukemia  
 4048.3   215 Leukemia  
 4048.4   259  Leukemia  
 4048.5  328  Leukemia  
 4048.6   341  Abdominal Sarcoma 
4099  ++  156  Leukemia  
4100  − 548  Study1-153 
4104  +  235  Leukemia  
4105  235  Abnormal Skin Growth  
4114  −  235  Abnormal Skin Growth 

Relative levels based on visual comparison of western blots. ++: expression equal to or greater than PMLRARα expression in the highest expressing line of MRP8-PMLRARα transgenic mice (line 556). +: expression present but less than PMLRARα expression in line 556. +/−: minimal expression. −: no signal on western blot.

*

When nonleukemic founder 4048 was euthanized at 37 days of age, its bone marrow was transplanted into lethally irradiated nontransgenic FVB/N mice. These mice were designated as 4048.1-4048.6. Mouse 4048.1 was euthanized for assessment of transgene expression and the other 5 recipient mice were monitored for illness.

Founders 4042, 4048, 4099, 4105, and 4114 exhibited abnormal skin growth similar to that seen in our MRP8-PMLRARα transgenic mice (data not shown).

Radiation can induce sarcomas in FVB/N recipients of transgenic marrow.

F1-153

This founder was euthanized at 18 months of age for examination and assessment of transgene expression.

Table 2.

RARm4 transgenic mice

Founder Transgene Expression in Bone Marrow Cohort
4142  ++  26 mice. 10-18 mo  
4151  +  10 mice. 10-18 mo  
4155  ND  Founder, 13 mo  
  No offspring. 
4157  +/−  Founder, 9 months* 
4159  +  2 mice, 11 & 18 mo  
4160  −  Founder, 9 mo* 
4192  ++  5 mice, 10-18 mo  
4195  −  Founder, 9 mo* 
Founder Transgene Expression in Bone Marrow Cohort
4142  ++  26 mice. 10-18 mo  
4151  +  10 mice. 10-18 mo  
4155  ND  Founder, 13 mo  
  No offspring. 
4157  +/−  Founder, 9 months* 
4159  +  2 mice, 11 & 18 mo  
4160  −  Founder, 9 mo* 
4192  ++  5 mice, 10-18 mo  
4195  −  Founder, 9 mo* 

See note to Table 1.

*

Founders 4157, 4160, and 4195 (and their offspring) were monitored for illness for up to 9 months. Monitoring was then discontinued given very low to absent transgene expression.

ND: not determined.

Fig. 2.

Expression of the transgenes.

A, B, Whole cell lysates of bone marrow were subjected to denaturing PAGE and Western blotting using a rabbit polyclonal antihumanRARαF domain antibody. Signals corresponding to transgenically expressed PMLRARα and RARα proteins are indicated by arrows. Locations of size markers are indicated by lines. (A) Protein expression in nonleukemic bone marrow of the highest expressing MRP8-PMLRARα transgenic line (Tg556-PR) and in 2 of the MRP8-PMLRARαm4 mice, Tg4048-PRm4 (nonleukemic bone marrow) and Tg4099-PRm4 (leukemic bone marrow). 8% SDS-polyacrylamide. (B) Protein expression in nonleukemic bone marrow Tg556-PR and in the marrows of healthy MRP8-RARαm4 transgenic mice from 3 lines, Tg4142-Rm4, Tg4151-Rm4, and Tg4192-Rm4. 12% SDS-polyacrylamide. C, Immunofluorescence analysis of bone marrow neutrophilic cells, anti-RARαF antiserum and Hoechst 33258, 1300 × .

Fig. 2.

Expression of the transgenes.

A, B, Whole cell lysates of bone marrow were subjected to denaturing PAGE and Western blotting using a rabbit polyclonal antihumanRARαF domain antibody. Signals corresponding to transgenically expressed PMLRARα and RARα proteins are indicated by arrows. Locations of size markers are indicated by lines. (A) Protein expression in nonleukemic bone marrow of the highest expressing MRP8-PMLRARα transgenic line (Tg556-PR) and in 2 of the MRP8-PMLRARαm4 mice, Tg4048-PRm4 (nonleukemic bone marrow) and Tg4099-PRm4 (leukemic bone marrow). 8% SDS-polyacrylamide. (B) Protein expression in nonleukemic bone marrow Tg556-PR and in the marrows of healthy MRP8-RARαm4 transgenic mice from 3 lines, Tg4142-Rm4, Tg4151-Rm4, and Tg4192-Rm4. 12% SDS-polyacrylamide. C, Immunofluorescence analysis of bone marrow neutrophilic cells, anti-RARαF antiserum and Hoechst 33258, 1300 × .

Close modal

To further substantiate that the transgenes were expressed in myeloid cells, bone marrow was also analyzed by immunofluorescence using the anti-RARαF antiserum. Cells with ring-shaped nuclei, as revealed by a fluorescent DNA-binding dye, are primarily neutrophilic. Transgene expression in such neutrophilic cells was observed in bothPMLRARαm4 and RARαm4 transgenic animals (Figure2C). A range of staining intensity was seen and neutrophilic cells without visibly detectable protein were also present. The speckled nuclear staining present in PMLRARαm4 transgenic mice was similar to that seen in PMLRARα transgenic mice. The nuclear staining observed in RARαm4 mice lacked the distinct speckles present in the other transgenics.

PMLRARm4 transgenic mice develop leukemia

Leukemias developed in 4 of 7 founders/lines ofMRP8-PMLRARαm4 transgenic mice (Table 1), a frequency similar to that encountered previously in MRP8-PMLRARα transgenic mice.32 The latency until leukemia onset, 3 to 11 months, was also comparable to that seen in our MRP8-PMLRARα mice (Table 1; see also Brown et al32). Assessment of leukemia penetrance was hampered by the fact that because of early illness, poor reproduction, or lack of transgene transmission, we did not obtain transgenic offspring for any of the lines in which leukemias developed. Nevertheless, it was apparent that PMLRARαm4 could readily initiate leukemia: 3 of 7 independent founder mice developed leukemia and 4 of 5 mice that were reconstituted with the nonleukemic bone marrow of a fourth independent founder also developed leukemia (Table1).

The leukemias arising in the PMLRARαm4 transgenic mice were acute leukemias with promyelocytic features. The peripheral blood of the leukemic animals was characterized by anemia, thrombocytopenia, and the presence of leukemic cells at the blast/promyelocyte stage of neutrophilic differentiation (Table 3). The leukemic bone marrows had large numbers of early myeloid cells, many of which had numerous azurophilic primary granules (Figure3B). These early cells also stained strongly with Sudan Black B (Figure 3D). The morphology and strong Sudan Black B staining were indicative of the promyelocytic character of the leukemic cells. The leukemias caused hepatomegaly and splenomegaly and were invasive, being present not only in the periportal areas of the liver but also invading the liver parenchyma (Figure 3F).

Table 3.

PMLRARm4 leukemias blood

MiceWBC HGB PLT Lymph Blast + Pro Imm NeutMat NeutMonoEosin
Control  3.0 ± 2.1 13.6 ± 0.3  1064 ± 69  89.9 ± 5.4   0  7.9 ± 3.9 1.7 ± 2.0 0.4 ± 0.2 
PR  4.5 ± 5.0   7.8 ± 1.8 334 ± 69  87.2 ± 7.2   6.8 ± 4.7 3.2 ± 2.2 2.0 ± 1.5 0.5 ± 0.8 0.3 ± 0.8 
PRm4  67.7 ± 50.7   7.8 ± 1.2  483 ± 192  48.8 ± 25.3  16.9 ± 14.0 13.9 ± 10.5 17.6 ± 19.1 2.4 ± 3.5 0.3 ± 0.5 
Leukemia  
 4042  101.33-150 8.73-150 6313-150 52 11  11  17 1  
 4048.2  3.73-150 93-150 3173-150 88  8  3  0  
 4048.3 Not increased  NA  NA  23  20  47 1  
 4048.5  6.6  7.7  389  61 13.5  16.5  3.5 5.5 0  
 4099  1413-150 7.43-150 7423-150 49  45  3  0  
 4104 86.1  6.1  335  20  15  30  34 
MiceWBC HGB PLT Lymph Blast + Pro Imm NeutMat NeutMonoEosin
Control  3.0 ± 2.1 13.6 ± 0.3  1064 ± 69  89.9 ± 5.4   0  7.9 ± 3.9 1.7 ± 2.0 0.4 ± 0.2 
PR  4.5 ± 5.0   7.8 ± 1.8 334 ± 69  87.2 ± 7.2   6.8 ± 4.7 3.2 ± 2.2 2.0 ± 1.5 0.5 ± 0.8 0.3 ± 0.8 
PRm4  67.7 ± 50.7   7.8 ± 1.2  483 ± 192  48.8 ± 25.3  16.9 ± 14.0 13.9 ± 10.5 17.6 ± 19.1 2.4 ± 3.5 0.3 ± 0.5 
Leukemia  
 4042  101.33-150 8.73-150 6313-150 52 11  11  17 1  
 4048.2  3.73-150 93-150 3173-150 88  8  3  0  
 4048.3 Not increased  NA  NA  23  20  47 1  
 4048.5  6.6  7.7  389  61 13.5  16.5  3.5 5.5 0  
 4099  1413-150 7.43-150 7423-150 49  45  3  0  
 4104 86.1  6.1  335  20  15  30  34 

Data are shown as arithmetic means ± SD. WBC: White blood cell count in 1000/μL. HGB: Hemoglobin in gms/dL. PLT: Platelet Count in 1000/μL. Percentages of nucleated leukocytes were derived from 200 cell differential counts of Wright's Giemsa stained blood smears. Lymph: lymphocytes. Blast + Pro: blasts & promyelocytes. Imm Neut: neutrophilic myelocytes & metamyelocytes. Mat Neut: neutrophilic band, mature ring, & polymorphonuclear forms. Mono: monocytes. Eosin: eosinophils. WBC, HGB, and PLT: Control n = 7, PR (MRP8-PMLRARα) primary leukemias n = 10, PRm4 (MRP8-PMLRARαm4) n = 5 (leukemic mice 4104 & 4048.5 and transplant recipients of leukemias 4042, 4048.2, and 4099). Differential counts: Control n = 7, PR primary leukemias n = 6, PRm4 primary leukemias n = 6.

F3-150

WBC, HGB, PLT obtained on initial transplant recipients. Peripheral blood was not obtained from leukemic mouse 4048.4. NA: not available.

Fig. 3.

Acute leukemia in MRP8-PMLRARm4 transgenic mice.

(A, C, E) Samples from control mice. (B, D, F) Samples from leukemicPMLRARαm4 mice. (B) Founder 4099. (D) Transplanted leukemia 4048.2. (F) founder 4042. (A, B) Bone marrow, Wright's Giemsa stain, 500 × . (C, D) Bone marrow, Sudan Black B stain, 965 × . (E, F) Liver, Hematoxylin and eosin stain, 200 × .

Fig. 3.

Acute leukemia in MRP8-PMLRARm4 transgenic mice.

(A, C, E) Samples from control mice. (B, D, F) Samples from leukemicPMLRARαm4 mice. (B) Founder 4099. (D) Transplanted leukemia 4048.2. (F) founder 4042. (A, B) Bone marrow, Wright's Giemsa stain, 500 × . (C, D) Bone marrow, Sudan Black B stain, 965 × . (E, F) Liver, Hematoxylin and eosin stain, 200 × .

Close modal

The leukemias were readily transplantable to histocompatible normal mice. Four of the leukemias were each transplanted by intravenous injection into 6 healthy unirradiated nontransgenic FVB/N animals. The cells engrafted and leukemias developed in all 24 recipient mice. The leukemias were subsequently maintained by serial transplantation in vivo and by cryopreservation.

The PMLRARαm4 leukemias exhibited variability. White blood cell counts ranged from normal to markedly elevated (Table 3). Peripheral blood leukocytes included significant numbers of maturing neutrophilic cells in some but not all cases. Similarly, although the bone marrows of some mice were effaced with promyelocytes, neutrophilic cells maturing beyond the promyelocyte stage were present in other animals (Table 4). Flow cytometric analysis with Gr-1 and Mac-1 markers revealed that leukemia 4099 had the low-level expression pattern typical of leukemias in ourPMLRARα transgenic mice, but that the 4042 and 4048.2 leukemias expressed these markers at moderately high levels (Figure4). The observation that some of thePMLRARαm4 leukemias were associated with increased white blood cell counts and significant numbers of maturing neutrophilic cells in the blood and bone marrow contrasts with the leukemic phenotype previously observed in our PMLRARα transgenic mice (Tables 3 and 4).

Table 4.

PMLRARm4 leukemias bone marrow

MiceBlast + Pro Imm Neut Mat Neut Erythroid LymphEosin
Control  2.0 ± 0.7  16.4 ± 2.4  27.0 ± 5.0   40.3 ± 6.5  12.1 ± 1.9 2.3 ± 0.9  
PR  81.3 ± 8.1   4.5 ± 2.6 0.7 ± 0.7   8.7 ± 7.7   4.5 ± 2.2 0.3 ± 0.3  
PRm4  65.7 ± 27.1  13.1 ± 14.4 12.7 ± 17.5   3.5 ± 2.4   4.4 ± 2.4 0.5 ± 1.1  
Leukemia  
 4042  44.5  37.5  5.5 4  6  2.5  
 4048.3  66.5  11  15.5  0.5 6.5  0  
 4048.5  94.5  0.5  0  3.5  1.5  
 4099  90  4.5  0.5  2.5  2  0  
 4104  33 12  42  7  6  
MiceBlast + Pro Imm Neut Mat Neut Erythroid LymphEosin
Control  2.0 ± 0.7  16.4 ± 2.4  27.0 ± 5.0   40.3 ± 6.5  12.1 ± 1.9 2.3 ± 0.9  
PR  81.3 ± 8.1   4.5 ± 2.6 0.7 ± 0.7   8.7 ± 7.7   4.5 ± 2.2 0.3 ± 0.3  
PRm4  65.7 ± 27.1  13.1 ± 14.4 12.7 ± 17.5   3.5 ± 2.4   4.4 ± 2.4 0.5 ± 1.1  
Leukemia  
 4042  44.5  37.5  5.5 4  6  2.5  
 4048.3  66.5  11  15.5  0.5 6.5  0  
 4048.5  94.5  0.5  0  3.5  1.5  
 4099  90  4.5  0.5  2.5  2  0  
 4104  33 12  42  7  6  

Data are shown as arithmetic means ± SD. Percentages of nucleated cells were derived from 400 cell differential counts of Wright's Giemsa stained bone marrow smears. Abbreviations: see legend to Table 3. Control n = 6, PR (MRP8-PMLRARα) primary leukemias n = 6, PRm4 (MRP8-PMLRARαm4) primary leukemias n = 5. Bone marrow smears were not available for leukemic mice 4048.2 and 4048.4.

Fig. 4.

Variable surface marker expression in PMLRARm4leukemias.

Bone marrow cells were stained with Gr-1 and Mac-1 antibodies that recognize myeloid surface antigens. Dead cells were eliminated from the analysis on the basis of staining with propidium iodide. (A) Control. (B) Leukemic mouse 4042 (C) Leukemic mouse 4099 (D) Transplanted leukemia 4048.2.

Fig. 4.

Variable surface marker expression in PMLRARm4leukemias.

Bone marrow cells were stained with Gr-1 and Mac-1 antibodies that recognize myeloid surface antigens. Dead cells were eliminated from the analysis on the basis of staining with propidium iodide. (A) Control. (B) Leukemic mouse 4042 (C) Leukemic mouse 4099 (D) Transplanted leukemia 4048.2.

Close modal

Although PMLRARαm4 leukemias were invasive transplantable diseases, they were not always associated with an aggressive clinical course. In our experience with PMLRARα leukemias, the interval between when a leukemic mouse appears ill and when it has progressed to a moribund condition is usually very short, on the order of 1 to 7 days. We were therefore surprised when we noted that mice ill with PMLRARαm4 leukemia did not necessarily exhibit rapid deterioration. Leukemic mice 4042 and 4099 were euthanized at the time their leukemias were initially apparent as determined by visible signs of illness. The primary transplant recipients of these 2 leukemias did not rapidly deteriorate after their leukemias became clinically apparent, living with their disease for weeks to months. Leukemic mice 4048.2 and 4104 were not euthanized at the time their leukemias were initially apparent, but were killed more than 2 weeks later. On transplantation, these leukemias were more rapidly fatal (Table5). Serial transplantation of the leukemias was, in some instances, accompanied by changes in features of the disease that may reflect the accumulation of additional genetic abnormalities. For example, although the first recipients of leukemia 4042 survived with the leukemia for an extended period and exhibited high peripheral white blood cell counts, mice that received the third serial transplant of this leukemia died rapidly without developing a peripheral blood leukocytosis.

Table 5.

Survival of primary recipients of PMLRAR and PMLRARm4 leukemias

Survival of Primary Transplant Recipients (days)
Mean Median Range
PMLRARα Leukemias  38  34  31-60  
PMLRARαm4 Leukemias  
 4042  135  136  119-154  
 4048.2  44 42  42-50  
 4099  71  73  60-85  
 4104  31 31  28-34 
Survival of Primary Transplant Recipients (days)
Mean Median Range
PMLRARα Leukemias  38  34  31-60  
PMLRARαm4 Leukemias  
 4042  135  136  119-154  
 4048.2  44 42  42-50  
 4099  71  73  60-85  
 4104  31 31  28-34 

Results obtained with 3 different PMLRARα leukemias and 4 different PMLRARαm4 leukemias that were transplanted into healthy FVB/N mice are shown (5-6 recipients for each leukemia). ThePMLRARα leukemias were similar to each other and the combined data are presented.

All-trans retinoic acid does not cause differentiation ofPMLRARm4 leukemias

Mice that were recipients of 3 different PMLRARαm4leukemias (4042, 4048.2, 4099) were treated with placebo or tRA to ascertain the effects of the Leu to Pro mutation on tRA responsiveness. For our PMLRARα leukemias, tRA generally causes a rapid rise in the peripheral leukocyte count as leukemic promyelocytes differentiate to mature neutrophils (25-fold average increase in leukocyte count on day 4 of therapy). Morphologic differentiation is readily apparent in the bone marrow of tRA-treated animals (Figure5A and B; see also Brown et al32) and regression of the leukemia is seen in histologic sections of liver (Figure 5C and D). In contrast, examination of the bone marrow and liver of tRA-treated PMLRARαm4 leukemic mice revealed that tRA did not induce morphologic differentiation and disease regression (Figure 5E through L). In addition, unlikePMLRARα leukemias, retinoic acid therapy did not cause a rapid rise in the peripheral white blood cell count (1.5-fold average increase in leukocyte count on day 4 of therapy).

Fig. 5.

Retinoic acid response of PMLRARm4 leukemias.

Leukemic mice were treated with placebo (A, C, E, G, I, K) or tRA (B, D, F, H, J, L). (A-D) PMLRARα expressing leukemia. (E-H)PMLRARαm4 leukemia 4099. (I-J) PMLRARαm4 leukemia 4042. (K-L) PMLRARαm4 leukemia 4048.2. (A, B, E, F, I-L) Bone marrow, Wright's Giemsa stain, 350 × . (C, D, G, H) Liver, Hematoxylin and eosin stain, 140 × . Effects of 11 days of tRA therapy are shown.

Fig. 5.

Retinoic acid response of PMLRARm4 leukemias.

Leukemic mice were treated with placebo (A, C, E, G, I, K) or tRA (B, D, F, H, J, L). (A-D) PMLRARα expressing leukemia. (E-H)PMLRARαm4 leukemia 4099. (I-J) PMLRARαm4 leukemia 4042. (K-L) PMLRARαm4 leukemia 4048.2. (A, B, E, F, I-L) Bone marrow, Wright's Giemsa stain, 350 × . (C, D, G, H) Liver, Hematoxylin and eosin stain, 140 × . Effects of 11 days of tRA therapy are shown.

Close modal

We also assessed the clinical effectiveness of tRA in the treatment of leukemias that arose in the PMLRARαm4 transgenic mice. To this end, we studied the effect of tRA on survival of mice that were recipients of 2 independent leukemias, leukemia 4048.2 and the aggressive variant of leukemia 4042 that arose on serial passaging. Mice that received leukemic cells by intravenous injection were treated with placebo or tRA when ill. Four recipients of leukemia 4042 treated with placebo died on days 5 and 6 of therapy and 5 mice treated with tRA also died rapidly, on days 5 to 10. Although the difference in survival was statistically significant (P = .04 by log-rank test), the rapid demise of tRA treated animals contrasted with our previous experience with PMLRARαleukemias.32,46 Unexpectedly, although tRA did not cause morphologic differentiation of leukemia 4048.2 (Figure 5K through L), it nevertheless substantially prolonged survival of the leukemic animals: 3 placebo-treated mice died on days 8 and 9 of therapy, whereas 3 tRA-treated mice were still alive at the end of 21 days of tRA treatment (P = .015 by log-rank test).

RARm4 transgenic mice are healthy

None of the MRP8-RARαm4 transgenic mice developed leukemia in up to 18 months of observation (Table 2). This result contrasts with our observations in the MRP8-PMLRARα andMRP8-PMLRARαm4 transgenic mice: in these mice, leukemias developed in more than half of the independently derived founders/lines beginning at 3 months of age, and by 10 months of age leukemia had appeared in one third of the mice of the highest expressingPMLRARα line32 and, as noted previously, in 4 of 5 mice derived from a high-expressing PMLRARαm4 founder.

Although leukemias did not develop in the RARαm4 transgenic mice, we investigated whether the RARαm4 protein altered neutrophil development in 1 of the highest expressing lines, line 4142. Expression of RARαm4 did not alter peripheral white blood cell counts. In the bone marrow, there was a trend toward increased immature neutrophilic cells, but this trend did not reach statistical significance (controls n = 9, line 4142 mice n = 6,P > .05, data not shown). In our previous work, we had observed that MRP8-PMLRARα and MRP8-PEBP2βMYH11transgenic mice exhibited a modest shift in the bone marrow toward immature neutrophilic cells that was accompanied by increased expression of the Mac-1 cell surface antigen (Kogan et al40and unpublished observations). When stained with Mac-1, the mean fluorescence of the myeloid cells in the bone marrow ofRARαm4 transgenic mice was 2.4-fold greater than that observed in controls (controls n = 5, line 4142 mice n = 5, data not shown). This increase was statistically significant (P = .002 by Student t test) and is consistent with the trend toward increased immature neutrophilic cells observed by morphologic examination. We also compared the colony-forming units present in the bone marrow of control and RARαm4 transgenic mice. Bone marrow cells from groups of 3 healthy untreated mice were grown in methylcellulose cultures in the presence of either G-CSF or a combination of GM-CSF and IL-3. Neither the number of colony-forming units nor the morphology of the cells as assessed on cytospins were significantly different between RARαm4 transgenic and control mice (data not shown).

We and others had demonstrated that directing expression of thePMLRARα fusion protein to immature mouse myeloid cells initiated leukemias with promyelocytic features.32,47,48 We have now similarly expressed altered forms of PMLRARα andRARα that are unable to respond to retinoic acid. The results show that retinoic acid responsiveness of PMLRARα, including transcriptional activation by tRA, is dispensable for leukemogenesis. In addition, we found that the ability of the PMLRARα protein to respond to tRA plays an important though perhaps not exclusive role in the therapeutic effects of retinoic acid. Furthermore, our finding that a RARα with dominant negative activity did not readily initiate leukemia suggests that the PML portion of PMLRARαplays an essential role in leukemic transformation.

Leukemogenesis cannot be explained by inappropriate transcriptional activation by PMLRAR

Alterations in transcription factors have been shown to play a central role in the pathogenesis of leukemias, lymphomas, and other malignancies.49,50 Changes in both transcriptional activation and repression can be important for the pathogenic effects of these alterations. PMLRARα retains the abilities ofRARα to repress and to activate transcription of retinoic acid receptor target genes. Although in some settings PMLRARαcan act to inappropriately repress transcription, when compared withRARα, it can also increase transcription in the absence or presence of ligand.13,15,17,23-25 The possible role of transcriptional activation in the pathogenesis of APL has not been previously tested.

By expressing PMLRARαm4 in the myeloid cells of transgenic mice, we directly assessed whether ligand-induced transcriptional activation by PMLRARα is required for leukemic transformation. The ability of PMLRARα variants to initiate leukemia has not been heretofore assessed. Grignani and colleagues45,51,52 examined the ability of altered forms ofPMLRARα to inhibit differentiation of the U937 promonocytic cell line. In this cell line, mutations that abolished the ability ofPMLRARα to interact with corepressors abrogated the ability of PMLRARα to inhibit differentiation.45 Although studies in U937 cells did not specifically address the role of transcriptional activation in inhibition of differentiation, thePMLRARα variants that were most effective at blocking differentiation retained the ability to act as strong transcriptional activators.52 PMLRARαm4 is unable to activate transcription. Our finding that it readily initiates leukemia demonstrates that although PMLRARα can enhance the transcription of RARα target genes, this ability to activate transcription in response to ligand plays no role in leukemogenesis.

Retinoic acid responsiveness may influence leukemic phenotype

Although RA binding, corepressor release, and transcriptional activation are not required for leukemogenesis, these activities of thePMLRARα protein may influence the characteristics of the leukemias initiated by PMLRARα. Leukemias that developed in our original PMLRARα transgenic mice were characterized by normal white blood cell counts, bone marrow effaced by cells at the promyelocyte stage of maturation, and a rapidly fatal course. In contrast, PMLRARαm4 leukemias were more variable: some of these leukemias were characterized by increased white blood cell counts, persistence of neutrophilic maturation beyond the promyelocyte stage, and an indolent clinical course. Because transgenes integrate randomly into the genome, we cannot fully exclude the possibility that the differences between PMLRARα and PMLRARαm4leukemias were due to variation in the level or pattern of protein expression in the independent founders. Furthermore, caution in interpreting such variation is warranted considering the modest number of leukemias that arose in this study. Nevertheless, the differences observed between the PMLRARα and PMLRARαm4leukemias might reflect an effect of endogenous ligands on the behavior of the leukemic cells. Physiologic levels of retinoic acid may bind toPMLRARα and thereby decrease repression or increase activation of target genes.

The possibility that ligand-responsiveness may influence leukemic phenotype is supported by a study of leukemias initiated by a PLZFRARα transgene. PLZFRARα is expressed as a result of a t(11;17)(q13;q21) translocation in rare APL patients who have a disease that is clinically resistant to tRA.53,54 Similar to PMLRARαm4,PLZFRARα does not activate transcription in response to tRA.25,55 Leukemias in Cathepsin G-PLZFRARαtransgenic mice resembled human chronic myeloid leukemia and displayed less of a block in neutrophilic differentiation than leukemias inCathepsin G-PMLRARα transgenic mice.56 The differences between Cathepsin G-PLZFRARα and Cathepsin G-PMLRARα leukemias are strikingly parallel to those we observed between MRP8-PMLRARαm4 and MRP8-PMLRARα leukemias. Given the view that retinoids play a role in fostering neutrophilic maturation, it was unexpected that the fusion proteins that are less responsive to retinoic acid were associated with leukemias with a greater degree of differentiation. Retinoids are not, however, simply differentiative agents. Depending on the experimental system and the maturational state of the cells, retinoids can stimulate or inhibit both proliferation and differentiation of myeloid cells (reviewed in Purton et al57). We speculate that retinoid-responsiveness of PMLRARα facilitates differentiation arrest at the promyelocyte stage of neutrophil maturation.

A dominant negative RAR does not appear sufficient to initiate leukemia

A number of lines of evidence support the hypothesis that dominant negative inhibition by PMLRARα of RARα may underlie the pathogenesis of APL. First, retinoic acid can enhance neutrophilic differentiation.58,59 Second, dominant negativeRARα can inhibit neutrophilic maturation of primary cells.60 Third, comparisons between PMLRARα andPLZFRARα have focused attention on the role of these proteins as transcriptional repressors that can interfere with normal activation of retinoic acid responsive genes.34,42,45,56,61 Fourth, the 4 described translocations involving RARα in APL result in fusions to PML, PLZF, NPM, and NuMA, proteins that do not appear to share common functions. In light of the evidence that transcriptional repression is important in the pathogenesis of APL,34,42,45,56,61 the lack of similarities between the 4RARα partners raises the possibility that the fusion proteins contribute to APL by acting as dominant negative RARαs. The fact that PMLRARα and PMLRARαm4 readily initiated leukemias, whereas RARαm4 did not, strongly suggests that the PML domain does more than simply confer dominant negative activity ontoRARα.

The PML domain of PMLRARα may also contribute to transformation by disrupting the normal function of PML, or, alternatively, by altering the DNA binding characteristics ofPMLRARα relative to RARα. PML can act as a growth suppressor62-65 and loss of PML predisposes mice to the development of a variety of tumors when the mice are treated with the tumor initiator dimethylbenzanthracene.66 Cells lacking PML grow more quickly than normal cells and are resistant to apoptosis induced by a variety of agents.66,67 These findings indicate that decreased PML function may contribute to transformation by allowing for increased proliferation and increased survival. In support of the hypothesis that disruption of PML activity byPMLRARα contributes to APL is a report that loss of PML increases the penetrance of leukemia in Cathepsin G-PMLRARαtransgenic mice.68 An alternative explanation for the differences we have observed in the leukemogenicity ofPMLRARαm4 and RARαm4 is that the PML domain changes binding site specificity69,70: The genes whose transcription is repressed by PMLRARαm4 may differ from those repressed by RARαm4 and such differences could account for the absence of leukemia in RARαm4 transgenic mice.

A recent study of retrovirally infected murine bone marrow demonstrated that transcriptional repression by RARα fusion proteins, dominant negative RARα, or unliganded normal RARαcan inhibit neutrophilic differentiation and immortalize primary hematopoietic cells.71 These observations support a role for transcriptional repression in APL pathogenesis and raised the possibility that dominant negative RARα would act as potent leukemogen. In vitro immortalization does not, however, always correspond with leukemic behavior in vivo. For example, although HRX-ENL both immortalizes cells and causes leukemia in mice,72 immortalized murine myeloid cell lines such as FDC-P cells are nonleukemic when injected into histocompatible animals.73 The fact that MRP8-PMLRARαm4 but not MRP8-RARαm4 mice developed leukemia is not inconsistent with the hypothesis that transcriptional repression is necessary for leukemic transformation. Furthermore, although our results show that the leukemogenic potential of PMLRARα extends beyond its ability to inhibit RARα function, our findings cannot be interpreted to indicate that dominant negative RARα would not contribute to leukemia when expressed in mice under conditions other than those we have used.

The expression of RARαm4 in the bone marrow cells of our transgenic mice had a modest impact on neutrophil differentiation. The lack of a strong effect of the MRP8-RARαm4 transgene compared with prior in vitro studies of retrovirally expressed dominant negativeRARα could reflect a number differences in experimental methods, including protein expression levels in hematopoietic progenitors, in vitro growth conditions, and mouse strain. In addition, expression of the transgene was heterogeneous and its effects on differentiation of total bone marrow may consequently have been masked by the presence of neutrophilic cells with low or absentRARαm4 protein. Our results are, however, concordant with studies of mice that lack RARα1 andRARγ (which comprise the vast majority ofRARs expressed in myeloid cells):RARα1−/−, RARγ−/− mice displayed no apparent hematopoietic defect in vivo and the neutrophilic cells from these mice showed a modest maturation defect in vitro.74 

Retinoic acid binding to PMLRAR is essential for retinoic acid-induced differentiation of leukemic promyelocytes

The leukemias that arose in the PMLRARαm4 transgenic mice did not differentiate in response to retinoic acid. The m4 mutation was originally described in a subclone of the NB4 cell line selected to grow in retinoic acid. Our results parallel the previously observed association of the m4 mutation with resistance to differentiation, and further demonstrate that the ability of tRA to cause differentiation of leukemic cells requires direct effects of tRA on the PMLRARαfusion protein.

Interestingly, tRA may have been of some therapeutic benefit despite the fact that differentiation of the leukemic blasts was not observed. Such a therapeutic effect of tRA has been observed in at least 1 case of human APL exhibiting marked in vitro resistance to the differentiative effects of tRA.28 The mechanism by which tRA was of benefit in these individual murine and human leukemias is not known. However, the possibility that tRA might improve survival even when it does not cause morphologic differentiation draws attention to the fact that novel therapies may have unanticipated effects that contribute to their clinical impact. This finding also suggests that tRA therapy may, in combination with other agents, continue to be of some benefit to APL patients identified as having resistant disease.

We thank Daphne Haas-Kogan and H. Jeffrey Lawrence for critical reading of the manuscript, and Meijuan Zhou for technical assistance.

Supported by grants CA 4338 and CA 75985 from the National Institutes of Health and by funds from the G.W. Hooper Research Foundation. S.C.K. is a recipient of a Burroughs Wellcome Fund Career Award.

Reprints:Scott C. Kogan, Department of Laboratory Medicine, Room M524, Box 0100, 505 Parnassus Ave, University of California, San Francisco, CA 94143-0100.

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

1
Sporn
MB
Roberts
AB
Goodman
DS
The Retinoids: Biology, Chemistry, and Medicine.
2nd ed.
1994
Raven
New York, NY
2
Kastner
P
Mark
M
Chambon
P
Nonsteroid nuclear receptors: what are genetic studies telling us about their role in real life?
Cell.
83
1995
859
869
3
Smith
MA
Parkinson
DR
Cheson
BD
Friedman
MA
Retinoids in cancer therapy.
J Clin Oncol.
10
1992
839
864
4
Redfern
CP
Lovat
PE
Malcolm
AJ
Pearson
AD
Gene expression and neuroblastoma cell differentiation in response to retinoic acid: differential effects of 9-cis and all-trans retinoic acid.
Eur J Cancer.
31A
1995
486
494
5
Breitman
TR
Collins
SJ
Keene
BR
Terminal differentiation of human promyelocytic leukemic cells in primary culture in response to retinoic acid.
Blood.
57
1981
1000
1004
6
Huang
ME
Ye
YC
Chen
SR
et al
All-trans retinoic acid with or without low dose cytosine arabinoside in acute promyelocytic leukemia. Report of 6 cases.
Chin Med J (Engl).
100
1987
949
953
7
Huang
ME
Ye
YC
Chen
SR
et al
Use of all-trans retinoic acid in the treatment of acute promyelocytic leukemia.
Blood.
72
1988
567
572
8
Rowley
JD
Golomb
HM
Dougherty
C
15/17 translocation, a consistent chromosomal change in acute promyelocytic leukaemia.
Lancet.
1
1977
549
550
9
de The
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.
347
1990
558
561
10
Borrow
J
Goddard
AD
Sheer
D
Solomon
E
Molecular analysis of acute promyelocytic leukemia breakpoint cluster region on chromosome 17.
Science.
249
1990
1577
1580
11
Alcalay
M
Zangrilli
D
Pandolfi
PP
et al
Translocation breakpoint of acute promyelocytic leukemia lies within the retinoic acid receptor alpha locus.
Proc Natl Acad Sci U S A.
88
1991
1977
1981
12
Lemons
RS
Eilender
D
Waldmann
RA
et al
Cloning and characterization of the t(15;17) translocation breakpoint region in acute promyelocytic leukemia.
Genes Chromosomes Cancer.
2
1990
79
87
13
Kakizuka
A
Miller
W
Jr
Umesono
K
et al
Chromosomal translocation t(15;17) in human acute promyelocytic leukemia fuses RAR alpha with a novel putative transcription factor, PML.
Cell.
66
1991
663
674
14
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.
254
1991
1371
1374
15
de The
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.
66
1991
675
684
16
Pandolfi
PP
Alcalay
M
Fagioli
M
et al
Genomic variability and alternative splicing generate multiple PML/RAR alpha transcripts that encode aberrant PML proteins and PML/RAR alpha isoforms in acute promyelocytic leukaemia.
Embo J.
11
1992
1397
1407
17
Kastner
P
Perez
A
Lutz
Y
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.
11
1992
629
642
18
Pandolfi
P
PML, PLZF and NPM genes in the molecular pathogensis of acute promyelocytic leukemia.
Haematologica.
81
1996
472
482
19
Chen
Z
Brand
NJ
Chen
A
et al
Fusion between a novel Kruppel-like zinc finger gene and the retinoic acid receptor-alpha locus due to a variant t(11;17) translocation associated with acute promyelocytic leukaemia.
Embo J.
12
1993
1161
1167
20
Redner
RL
Rush
EA
Faas
S
Rudert
WA
Corey
SJ
The t(5;17) variant of acute promyelocytic leukemia expresses a nucleophosmin-retinoic acid receptor fusion.
Blood.
87
1996
882
886
21
Wells
RA
Catzavelos
C
Kamel-Reid
S
Fusion of retinoic acid receptor alpha to NuMA, the nuclear mitotic apparatus protein, by a variant translocation in acute promyelocytic leukaemia.
Nat Genet.
17
1997
109
113
22
Mangelsdorf
DJ
Evans
RM
The RXR heterodimers and orphan receptors.
Cell.
83
1995
841
850
23
Rousselot
P
Hardas
B
Patel
A
et al
The PML-RAR alpha gene product of the t(15;17) translocation inhibits retinoic acid-induced granulocytic differentiation and mediated transactivation in human myeloid cells.
Oncogene.
9
1994
545
551
24
Pandolfi
PP
Grignani
F
Alcalay
M
et al
Structure and origin of the acute promyelocytic leukemia myl/RAR alpha cDNA and characterization of its retinoid-binding and transactivation properties.
Oncogene.
6
1991
1285
1292
25
Ruthardt
M
Testa
U
Nervi
C
et al
Opposite effects of the acute promyelocytic leukemia PML-retinoic acid receptor alpha (RAR alpha) and PLZF-RAR alpha fusion proteins on retinoic acid signalling.
Mol Cell Biol.
17
1997
4859
4869
26
Degos
L
Dombret
H
Chomienne
C
et al
All-trans-retinoic acid as a differentiating agent in the treatment of acute promyelocytic leukemia.
Blood.
85
1995
2643
2653
27
Warrell
RP
Jr
Maslak
P
Eardley
A
et al
Treatment of acute promyelocytic leukemia with all-trans retinoic acid: an update of the New York experience.
Leukemia.
8
1994
929
933
28
Ding
W
Li
YP
Nobile
LM
et al
Leukemic cellular retinoic acid resistance and missense mutations in the PML-RARalpha fusion gene after relapse of acute promyelocytic leukemia from treatment with all-trans retinoic acid and intensive chemotherapy.
Blood.
92
1998
1172
1183
29
Imaizumi
M
Suzuki
H
Yoshinari
M
et al
Mutations in the E-domain of RAR portion of the PML/RAR chimeric gene may confer clinical resistance to all-trans retinoic acid in acute promyelocytic leukemia.
Blood.
92
1998
374
382
30
Kitamura
K
Kiyoi
H
Yoshida
H
Saito
H
Ohno
R
Naoe
T
Mutant AF-2 domain of PML-RARalpha in retinoic acid-resistant NB4 cells: differentiation induced by RA is triggered directly through PML-RARalpha and its down-regulation in acute promyelocytic leukemia.
Leukemia.
11
1997
1950
956
31
Shao
W
Benedetti
L
Lamph
WW
Nervi
C
Miller
WH
Jr
A retinoid-resistant acute promyelocytic leukemia subclone expresses a dominant negative PML-RAR alpha mutation.
Blood.
89
1997
4282
4289
32
Brown
D
Kogan
S
Lagasse
E
et al
A PMLRAR alpha transgene initiates murine acute promyelocytic leukemia.
Proc Natl Acad Sci U S A.
94
1997
2551
2556
33
Sande
S
Privalsky
ML
Identification of TRACs (T3 receptor-associating cofactors), a family of cofactors that associate with, and modulate the activity of, nuclear hormone receptors.
Mol Endocrinol.
10
1996
813
825
34
Hong
SH
David
G
Wong
CW
Dejean
A
Privalsky
ML
SMRT corepressor interacts with PLZF and with the PML-retinoic acid receptor alpha (RARalpha) and PLZF-RARalpha oncoproteins associated with acute promyelocytic leukemia.
Proc Natl Acad Sci U S A.
94
1997
9028
9033
35
Hong
SH
Privalsky
ML
Retinoid isomers differ in the ability to induce release of SMRT corepressor from retinoic acid receptor-alpha.
J Biol Chem.
274
1999
2885
2892
36
Lagasse
E
Weissman
IL
bcl-2 inhibits apoptosis of neutrophils but not their engulfment by macrophages.
J Exp Med.
179
1994
1047
1052
37
Hogan
B
Constantini
F
Lacy
E
Manipulating the Mouse Embryo: A Laboratory Manual.
2nd ed.
1994
Cold Spring Harbor Laboratory Press
Plainview, NY
38
Taketo
M
Schroeder
AC
Mobraaten
LE
et al
FVB/N: an inbred mouse strain preferable for transgenic analyses.
Proc Natl Acad Sci U S A.
88
1991
2065
2069
39
Robbins
SM
Quintrell
NA
Bishop
JM
Myristoylation and differential palmitoylation of the HCK protein-tyrosine kinases govern their attachment to membranes and association with caveolae.
Mol Cell Biol.
15
1995
3507
3515
40
Kogan
SC
Lagasse
E
Atwater
S
et al
The PEBP2betaMYH11 fusion created by Inv(16)(p13;q22) in myeloid leukemia impairs neutrophil maturation and contributes to granulocytic dysplasia.
Proc Natl Acad Sci U S A.
95
1998
11,863
11
868.
41
Karasuyama
H
Melchers
F
Establishment of mouse cell lines which constitutively secrete large quantities of interleukin 2, 3, 4 or 5, using modified cDNA expression vectors.
Eur J Immunol.
18
1988
97
104
42
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
814
43
Keidel
S
LeMotte
P
Apfel
C
Different agonist- and antagonist-induced conformational changes in retinoic acid receptors analyzed by protease mapping.
Mol Cell Biol.
14
1994
287
298
44
Leng
X
Tsai
SY
O'Malley
BW
Tsai
MJ
Ligand-dependent conformational changes in thyroid hormone and retinoic acid receptors are potentially enhanced by heterodimerization with retinoic X receptor.
J Steroid Biochem Mol Biol.
46
1993
643
661
45
Grignani
F
De Matteis
S
Nervi
C
et al
Fusion proteins of the retinoic acid receptor-alpha recruit histone deacetylase in promyelocytic leukaemia.
Nature.
391
1998
815
818
46
Lallemand-Breitenbach
V
Guillemin
MC
Janin
A
et al
Retinoic acid and arsenic synergize to eradicate leukemic cells in a mouse model of acute promyelocytic leukemia.
J Exp Med.
189
1999
1043
1052
47
Grisolano
JL
Wesselschmidt
RL
Pelicci
PG
Ley
TJ
Altered myeloid development and acute leukemia in transgenic mice expressing PML-RAR alpha under control of cathepsin G regulatory sequences.
Blood.
89
1997
376
387
48
He
LZ
Tribioli
C
Rivi
R
et al
Acute leukemia with promyelocytic features in PML/RARalpha transgenic mice.
Proc Natl Acad Sci U S A.
94
1997
5302
5307
49
Rabbitts
TH
Chromosomal translocations in human cancer.
Nature.
372
1994
143
149
50
Look
AT
Oncogenic transcription factors in the human acute leukemias.
Science.
278
1997
1059
1064
51
Grignani
F
Ferrucci
PF
Testa
U
et al
The acute promyelocytic leukemia-specific PML-RAR alpha fusion protein inhibits differentiation and promotes survival of myeloid precursor cells.
Cell.
74
1993
423
431
52
Grignani
F
Testa
U
Rogaia
R
et al
Effects on differentiation by the promyelocytic leukemia PML/RAR alpha protein depend on the fusion of the PML protein dimerization and RAR alpha DNA binding domain.
Embo J.
15
1996
4949
4958
53
Chen
SJ
Zelent
A
Tong
JH
et al
Rearrangements of the retinoic acid receptor alpha and promyelocytic leukemia zinc finger genes resulting from t(11;17)(q23;q21) in a patient with acute promyelocytic leukemia.
J Clin Invest.
91
1993
2260
2267
54
Licht
JD
Chomienne
C
Goy
A
et al
Clinical and molecular characterization of a rare syndrome of acute promyelocytic leukemia associated with translocation (11;17).
Blood.
85
1995
1083
1094
55
Chen
Z
Guidez
F
Rousselot
P
et al
PLZF-RAR alpha fusion proteins generated from the variant t(11;17)(q23;q21) translocation in acute promyelocytic leukemia inhibit ligand-dependent transactivation of wild-type retinoic acid receptors.
Proc Natl Acad Sci U S A.
91
1994
1178
1182
56
He
LZ
Guidez
F
Tribioli
C
et al
Distinct interactions of PML-RARalpha and PLZF-RARalpha with co-repressors determine differential responses to RA in APL.
Nat Genet.
18
1998
126
135
57
Purton
LE
Bernstein
ID
Collins
SJ
All-trans retinoic acid delays the differentiation of primitive hematopoietic precursors (lin-c-kit+Sca-1(+)) while enhancing the terminal maturation of committed granulocyte/monocyte progenitors.
Blood.
94
1999
483
495
58
Douer
D
Koeffler
HP
Retinoic acid enhances colony-stimulating factor-induced clonal growth of normal human myeloid progenitor cells in vitro.
Exp Cell Res.
138
1982
193
198
59
Gratas
C
Menot
ML
Dresch
C
Chomienne
C
Retinoid acid supports granulocytic but not erythroid differentiation of myeloid progenitors in normal bone marrow cells.
Leukemia.
7
1993
1156
1162
60
Tsai
S
Collins
SJ
A dominant negative retinoic acid receptor blocks neutrophil development at the promyelocyte stage.
Proc Natl Acad Sci U S A.
90
1993
7153
7157
61
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-RARalpha underlie molecular pathogenesis and treatment of acute promyelocytic leukemia.
Blood.
91
1998
2634
2642
62
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
6867
63
Koken
MH
Linares-Cruz
G
Quignon
F
et al
The PML growth-suppressor has an altered expression in human oncogenesis.
Oncogene.
10
1995
1315
1324
64
Borden
KL
Campbell-Dwyer
EJ
Salvato
MS
The promyelocytic leukemia protein PML has a pro-apoptotic activity mediated through its RING domain.
FEBS Lett.
418
1997
30
34
65
Le
XF
Vallian
S
Mu
ZM
Hung
MC
Chang
KS
Recombinant PML adenovirus suppresses growth and tumorigenicity of human breast cancer cells by inducing G1 cell cycle arrest and apoptosis.
Oncogene.
16
1998
1839
1849
66
Wang
ZG
Delva
L
Gaboli
M
et al
Role of PML in cell growth and the retinoic acid pathway.
Science.
279
1998
1547
1551
67
Wang
ZG
Ruggero
D
Ronchetti
S
et al
PML is essential for multiple apoptotic pathways.
Nat Genet.
20
1998
266
272
68
Wang
ZG
Rego
E
Penuzzi
D
He
LZ
Pandolfi
PP
Loss of PML function enhances the frequency and onset of APL in PML-RARalpha transgenic mice.
Blood.
92
1998
S1,479a
69
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.
12
1993
3171
3182
70
Jansen
JH
Mahfoudi
A
Rambaud
S
Lavau
C
Wahli
W
Dejean
A
Multimeric complexes of the PML-retinoic acid receptor alpha fusion protein in acute promyelocytic leukemia cells and interference with retinoid and peroxisome-proliferator signaling pathways.
Proc Natl Acad Sci U S A.
92
1995
7401
7405
71
Du
C
Redner
RL
Cooke
MP
Lavau
C
Overexpression of wild-type retinoic acid receptor alpha (RARalpha) recapitulates retinoic acid-sensitive transformation of primary myeloid progenitors by acute promyelocytic leukemia RARalpha-fusion genes.
Blood.
94
1999
793
802
72
Lavau
C
Szilvassy
SJ
Slany
R
Cleary
ML
Immortalization and leukemic transformation of a myelomonocytic precursor by retrovirally transduced HRX-ENL.
Embo J.
16
1997
4226
4237
73
Dexter
TM
Garland
J
Scott
D
Scolnick
E
Metcalf
D
Growth of factor-dependent hemopoietic precursor cell lines.
J Exp Med.
152
1980
1036
1047
74
Labrecque
J
Allan
D
Chambon
P
Iscove
NN
Lohnes
D
Hoang
T
Impaired granulocytic differentiation in vitro in hematopoietic cells lacking retinoic acid receptors alpha1 and gamma.
Blood.
92
1998
607
615
Sign in via your Institution