The bcr1- and bcr3- promyelocytic leukemia/retinoic acid receptor α (PML/RARα) are the two major fusion proteins expressed in acute promyelocytic leukemia (APL) patients. These proteins, which are present in different lengths of PML (amino acids 1-552 and 1-394, respectively), contain most of the functional domains of PML and RARα, bind all-trans-retinoic acid (t-RA), and act as t-RA–dependent transcription factors. T-RA is an effective inducer of clinical remission only in patients carrying the t(15; 17) and expressing the PML/RARα products. However, in APL patients achieving complete remission with t-RA therapy the bcr3-PML/RARα product has been found associated with a poorer prognosis than bcr1-PML/RARα. In the present study we have investigated the structural and functional properties of the bcr3-PML/RARα in comparison to the previously characterized bcr1-PML/RARα. In particular, we have measured the binding properties of the two endogenous ligands t-RA and 9-cis-RA to both of these isoforms. T-RA binding analysis of nuclear and cytosolic extracts prepared from bcr3-PML/RARα APL patients and from bcr3-PML/RARα COS-1 transfected cells indicates that this protein is present only as high-molecular-weight nuclear complexes. Using saturation binding assays and Scatchard analyses we found that t-RA binds with slightly less affinity to the bcr3-PML/RARα receptor than to bcr1-PML/RARα or RARα (Kd = 0.4 nmol/L, 0.13 nmol/L or 0.09 nmol/L, respectively). Moreover, two different high-affinity 9-cis-RA binding sites (Kd = 0.45 and 0.075 nmol/L) were detectable in the bcr3-PML/RARα product but not in the bcr1-PML/RARα product (Kd = 0.77 nmol/L). By competition binding experiments we showed that 9-cis-RA binds with higher specificity to the bcr3-PML/RARα isoform than to the bcr1-PML/RARα or RARα. Consistent with these data, the binding of 9-cis-RA to the bcr3-PML/RARα product resulted in increased transcriptional activation of the RA-responsive element (RARE) TRE, but not of the βRARE, in transiently transfected COS-1 cells. These results provide evidence indicating that preferential retinoid binding to the different PML/RARα products can be measured.

THE CHARACTERISTIC balanced translocation t(15q+; 17q−) of acute promyelocytic leukemia (APL) fuses a portion of the promyelocytic leukemia (PML) gene on chromosome 15 to the second coding exon of the nuclear retinoic acid receptor α (RARα) on chromosome 17. As a result of this translocation a PML/RARα chimeric gene is formed which is transcriptionally active in all cases of APL.1,2 The chimeric transcript encodes a PML/RARα fusion protein that retains both the PML “ring finger” and dimerization domains and the RARα DNA-binding and RA-binding domains.3,4 The breakpoints on chromosome 15 are clustered at three different sites in the PML locus: in intron 6 (breakpoint cluster region 1; bcr1), exon 6 (bcr2), and intron 3 (bcr3), leading to three different fusion proteins.5-8 The most frequent products are the bcr1- (also known as “long” or type “B”) and bcr3- (or “short” or type “A”) PML/RARα,4,5 which present different lengths of PML (amino acids 1-552 and 1-394, respectively). In rare APL cases, the RARα gene fuses with the PLZF (promyelocytic leukemia zinc finger) gene on chromosome 119,10 or with the nucleophosmin gene on 5,11 suggesting that aberrant RARα gene expression may be associated with leukemogenesis.

APL has become the first model of differentiation therapy targeted to a defined genetic defect.3,4,12 All-trans-retinoic acid (t-RA) induces disease remission of APL patients by triggering terminal differentiation of malignant cells.3,4,12 However, there is diversity of t-RA sensitivity in APL cases: t-RA induces cell differentiation and clinical remission in patients with the t(15; 17) translocation,4,12 but APL patients and cultured cells from patients with the t(11; 17) fail to differentiate in response to t-RA. In the single t(5; 17) APL case a reduced response to t-RA therapy compared to PML/RARα cases10 was found. In addition, within APLs with the t(15; 17), evaluation of patients in remission after treatment with t-RA alone or with t-RA followed by chemotherapy showed that the expression of the bcr3-PML/RARα isoform is significantly associated with a poorer prognosis compared to those with the bcr1-PML/RARα isoform.12-14 

The mechanisms underlying the heterogeneous t-RA response in APL are unclear. Retinoids activate two classes of nuclear receptor proteins of the steroid and thyroid hormone superfamily, the RARs (α, β, and γ), and the retinoid X receptors (RXRs α, β, and γ), for gene transcriptional activation.15,16 T-RA binds only to RARs, whereas 9-cis-retinoic acid (9-cis-RA) is a bifunctional ligand capable of binding to both RARs and RXRs.17-19 Whether 9-cis-RA has a different spectrum of biologic activity from t-RA is presently unknown.

With respect to retinoid binding properties, we and others6,20 have previously reported that the bcr1-PML/RARα isoform binds retinoids with the same affinity and specificity as the wild-type RARα receptor and is present predominantly in high-molecular-weight nuclear complexes.20 

In the present investigation we have characterized the structural and functional properties of the bcr3-PML/RARα and PLZF/RARα in comparison to those of the bcr1-PML/RARα product. We show that in cells from bcr3- PML/RARα APL patients and bcr3-PML/RARα or PLZF/RARα COS-1–transfected cells, these fusion proteins are present in high-molecular-weight nuclear complexes. Moreover, our results provide evidence for preferential retinoid binding to the different PML/RARα products. In fact, we found that 9-cis-RA binds with higher affinity and specificity to the bcr3-PML/RARα isoform than to the bcr1-PML/RARα or RARα. This effect was also associated with a differential activation of retinoic acid responsive elements in COS-1 cells transfected with bcr3-PML/RARα and either TRE (thyroid response element) or βRARE (RARβ response element). Taken together, these results indicate that preferential retinoid binding to different gene fusion products can be measured and suggest the possibility of specific differentiation therapy in APL patients.

9-cis-[10-3H]retinoic acid (24 Ci/mmol) and unlabeled retinoids used in this study were synthesized by the Department of Medicinal Chemistry, Hoffmann-La Roche, Nutley, NJ. All-trans-[3H]-retinoic acid (50.7 Ci/mmol) was purchased from DuPont/NEN (Boston, MA). COS-1 cells were obtained from American Type Culture Collection (ATCC; Rockville, MD). Rabbit anti-RARα polyclonal serum RPα(F )21 was kindly provided by Prof P. Chambon (Strasbourg, France).

Patients samples and processing.Fresh leukemic cells were obtained from peripheral blood (PB) and bone marrow (BM) aspirates of newly diagnosed, informed APL patients presenting an initial percentage of blasts that was more than 80%. Patients were classified as M3 or M3-variant according to the French-American-British (FAB) classification.22 Characterization of the PML/RARα isoform (bcr1-bcr2-bcr3) was performed by reverse transcriptase-polymerase chain reaction (RT-PCR) assay as previously described.23 Leukemic cells were isolated by Ficoll-Hypaque density gradients (Pharmacia, Uppsala, Sweden).

RNA preparation and Northern blot analysis.Total RNA was prepared by the guanidinium isothiocyanate-CsCl procedure24 from Ficoll-Hypaque–isolated APL blasts washed twice with calcium-magnesium free phosphate solution. Total RNA (10 μg/sample) was electrophoresed through a 1.2% denaturing agarose-formaldeyde gel. After electrophoresis, RNA was transferred to Nytran (Schleicher & Schuell, Hayward, CA) by capillary blotting and then cross-linked by UV irradiation. The cDNA probes were radiolabeled with [α-32P]dCTP (3,000 Ci/mmol; DuPont-NEN) via random priming using the kit and protocols supplied by Bethesda Research Laboratories. The cDNA probes for human RARα, β, and γ25,26 were obtained from Prof P. Chambon. The cDNA probes for human RXRα27 and for mouse RXRβ and γ28 were obtained from Dr R.M. Evans (The Salk Institute, San Diego, CA). The cDNA probe for chicken glyceraldehyde-3-dehydrogenase (GAPDH)29 was used as control probe to standardize the level of gene expression.

Cell culture, transfection, and retinoic acid binding assays.COS-1 cells were transiently transfected by electroporation as previously described19 with pSG5 expression vectors containing cDNAs for human RARα, bcr1-, and bcr3-PML/RARα,30 or with a pMT2 expression vector24 containing a cDNA encoding the human PLZF/RARα.9 The PLZF/RARα cDNA was reconstructed from wild-type RARα and PLZF cDNAs by PCR and conventional cloning strategies (M. Ruthardt and P.G. Pelicci, manuscript submitted). After transfection (48 to 72 hours), cells (3 to 7 × 107) were removed by trypsinization and collected by centrifugation. Nuclear and cytosolic extracts were prepared from COS transfected cells and from fresh blasts from APL patients as previously described.19,31,32 Extracts were incubated with 10 nmol/L [3H]-t-RA for 18 hours at 4°C. [3H]-t-RA binding was analyzed by high-performance liquid chromatography (HPLC) performed at 4°C using a size exclusion column Superose 6 HR 10/30 (Pharmacia). The eluent was PTG buffer containing 0.4 mol/L KCl and the flow rate was 0.4 mL/min.20 

Apparent molecular weights (MW) were calculated on the basis of the elution times of a series of marker proteins used to calibrate the system, such as: blue dextran, MW 2,000,000; thyroglobulin, MW 669,000; apoferritin, MW 443,000; β-amylase MW 200,000; alcohol dehydrogenase, MW 150,000; bovine albumin, MW 66,000; ovalbumin, MW 45,000; carbonic anhydrase, MW 29,000; and lactalbumin, MW 14,200.

For saturation binding studies as well as competitive binding assays, bound RA was separated from free radioactivity using PD10 desalting columns (Pharmacia) as described.19,20 In saturation binding studies, incubations were performed, for 18 hours at 4°C, in the presence of increasing concentrations of [3H]-t-RA or [3H]-9-cis-retinoic acid. Binding in the presence of 200-fold molar excess of unlabeled t-RA or 9-cis-RA was defined as nonspecific binding. Specific binding resulted from total binding minus nonspecific binding. Linear least-square analysis of the Scatchard plot was performed with the aid of the computer program BDATA-EMF.33 A fixed concentration of [3H]-t-RA (10 nmol/L) and increasing concentrations of unlabeled competing ligand (ranging between 0.5 nmol/L and 1μmol/L) were used in incubations for competitive binding assays. The EC50 values (retinoid concentrations that inhibit 50% of the total specific RA binding) were calculated using the nonlinear least squares regression analysis program ALLFIT.34 

Sodium dodecyl sulfate (SDS)-gel electrophoresis and Western immunoblotting.Nuclear extracts were diluted 1:1 with 2× SDS sample buffer (2% SDS, 0.125 mol/L Tris-HCl, pH 6.8, 20% glycerol, 0.02% bromophenol blue, and 10% 2-mercaptoethanol). Proteins were then fractionated by electrophoresis on an 8% SDS polyacrylamyde gel and electroblotted to nitrocellulose membrane (Hybond C Super; Amersham Life Science Inc, Arlington Heights, IL). Proteins that reacted with the anti-RARα RPα(F ) antibody21 (used at a 1:500 dilution) were detected using the ECL Western blotting detection kit (Amersham) with the reagents provided according to the manufacturer's instructions.

Transient cotransfection of COS-1 cells and transactivation assays.COS-1 cells were plated in six-well 35-mm culture dishes at a density of 2.5 × 105 cells/well. Twenty-four hours after plating cells were transiently transfected using the Lipofectamine reagent (GIBCO-BRL, Gaithersburg, MD). Each well received 0.5 μg of the pSG5 expression vectors containing the coding regions for human RARα, bcr1- or bcr3-PML/RARα, 0.5 μg (βRARE)3-tk-LUC,35 or TRE2-tk-LUC,36 or the RARβ pr-LUC reporter,8,35 and the plasmid encoding β-galactosidase (pSV-βGal) (0.3 μg) was cotransfected to monitor transfection efficiency and for normalization of reactions. After 5 hours, transfecting medium was removed and cells were incubated in Dulbecco′s modified Eagle′s medium containing 5% heat-inactivated, charcoal stripped fetal calf serum. The next day retinoids were added for 16 hours to minimize retinoid isomerisation. Cells were washed twice with calcium-magnesium–free phosphate solution and lysate using a Reporter Lysis Buffer (Promega Corp, Madison, WI). Luciferase activity was measured using a luminometer (Berthold, Wildbad, Germany).37 Reporter gene expression was calculated in arbitrary units, relative to β-galactosidase expression. The EC50 values (retinoid concentrations which induced 50% of maximal response) were calculated using the nonlinear least squares regression analysis program ALLFIT.34 

Retinoic acid binding in bcr3-PML/RARα APL patients.Previous retinoid binding studies performed in the APL cell line NB4, which express the bcr1-PML/RARα product,6 showed the presence of specific [3H]-t-RA binding corresponding to CRABP, RARs, monomeric form of bcr1-PML/RARα (110,000), and to the high-molecular-weight complexes formed by the interaction of bcr1-PML/RARα with itself or with other nuclear proteins.20,38 We have now investigated the t-RA binding properties of fresh APL blasts cells obtained from patients who expressed the bcr3 isoform of the PML/RARα transcript. Nuclear and cytosolic extracts were labeled with 10 nmol/L [3H]-t-RA in the absence or in the presence of 200-fold mol/L excess of unlabeled t-RA to determine nonspecific binding, and the t-RA binding was analyzed by HPLC size exclusion chromatography. The HPLC profile of nuclear extracts prepared from these cells showed the presence of three main peaks of specific [3H]-t-RA binding eluting at 36, 42, and 47 minutes, corresponding to molecular weights of about 500,000, 50,000, and 18,000, respectively (Fig 1). Similar HPLC profiles were consistently found in blasts isolated either from BM or from PB of bcr3-PML/RARα APL patients. The specific t-RA binding peaks corresponding to molecular weights of 50,000 and 18,000 probably represent binding to RARs and CRABP, respectively. The presence of CRABP in cytosol extracts prepared from these cells was established by the presence of a main peak of specific binding eluting at 47 minutes and corresponding to a molecular weight of 18,000. In addition, in cytosolic extracts, two small peaks of specific t-RA binding activity eluting at a retention time of 28 and 34 minutes, which probably represented contamination of cytosol with nuclear extract, could be detected.

Fig. 1.

Size exclusion HPLC analysis of cytosolic (A) and nuclear (B) extracts prepared from fresh blasts from the PB of an APL patient expressing the bcr3-PML/RARα isoform. Extracts (200 μL) were incubated with 10 nmol/L [3H]-t-RA in the absence (•) or in the presence (○) of 200-fold mol/L excess of unlabeled t-RA for 18 hours at 4°C. The samples were then fractionated over a Superose 6 HR 10/30 size exclusion column as described in Materials and Methods.

Fig. 1.

Size exclusion HPLC analysis of cytosolic (A) and nuclear (B) extracts prepared from fresh blasts from the PB of an APL patient expressing the bcr3-PML/RARα isoform. Extracts (200 μL) were incubated with 10 nmol/L [3H]-t-RA in the absence (•) or in the presence (○) of 200-fold mol/L excess of unlabeled t-RA for 18 hours at 4°C. The samples were then fractionated over a Superose 6 HR 10/30 size exclusion column as described in Materials and Methods.

Close modal

Retinoic acid binding in bcr3-PML/RARα-COS-1 transfected cells and immunoblot analysis.To easily characterize the t-RA ligand binding properties of the bcr3-PML/RARα isoform in comparison to the previously reported RARα and bcr1-PML/RARα, COS-1 cells were transiently transfected with the cDNAs for RARα, bcr1-PML/RARα, bcr3-PML/RARα, or expression plasmid (mock). Nuclear extracts prepared from these cells were first examined by immunoblot analysis using the anti-RARα RPα(F ) antibody21 to confirm the production of intact receptors. Immunoblot analysis showed the presence of immunoreactive proteins corresponding to the approximate MW calculated from their respective cDNA sequences30,39,40 (RARα 50,000; bcr1-PML/RARα 105,000; bcr3-PML/RARα 90,000) (Fig 2A). As previously described,20 endogenous RARα was not immunodetectable in nuclear extracts from mock-transfected cells (Fig 2A). In pSG5/bcr1-PML/RARα extracts a small degree of degradation was detected, as evidenced by an immunoreactive band showing an MW of about 40,000. We next examined the binding of [3H]-t-RA to these proteins by HPLC size exclusion analysis (Fig 2B). The HPLC profile obtained from nuclear extracts prepared from pSG5/bcr3-PML/RARα transiently transfected COS-1 cells showed the presence of two main peaks of specific t-RA binding activity eluting at 31 and 37 minutes and then corresponding to an MW of more than 1,000,000 and of about 550,000, respectively. These HPLC profiles were very similar to those of nuclear extracts prepared from blasts obtained from bcr3-PML/RARα APL patients. Conversely to RARα and pSG5/bcr1-PML/RARα, a complete absence of the monomeric form of this receptor was found (Fig 2B). In fact, t-RA specific binding activity was not present in fractions corresponding to lower MW proteins. We concluded that the pSG5/bcr3-PML/RARα product is present only as macromolecular complexes that may represent pSG5/bcr3-PML/RARα homodymers and/or high-molecular-weight complexes with other nuclear proteins.

Fig. 2.

Immunoblot analysis and HPLC analysis of [3H]-t-RA binding in nuclear extracts prepared from mock, pSG5/RARα, pSG5/bcr1-PML/RARα and pSG5/bcr3-PML/RARα transiently transfected COS-1 cells. (A) Immunoblot analysis: Nuclear extracts were subjected to 8% polyacrylamide slab gel electrophoresis followed by immunoblotting using the anti-RARα RPα(F ) antibody21 as described in Materials and Methods. (B) Size exclusion HPLC analysis. Nuclear extracts were analyzed as described in the legend of Fig 1. (•) [3H]-t-RA alone; (○) [3H]-t-RA plus 200-fold mol/L excess of unlabeled t-RA. Arrows indicate the elution times of marker proteins (in thousands) used to calibrate the Superose 6 HR 10/30 size exclusion column as indicated in Materials and Methods.

Fig. 2.

Immunoblot analysis and HPLC analysis of [3H]-t-RA binding in nuclear extracts prepared from mock, pSG5/RARα, pSG5/bcr1-PML/RARα and pSG5/bcr3-PML/RARα transiently transfected COS-1 cells. (A) Immunoblot analysis: Nuclear extracts were subjected to 8% polyacrylamide slab gel electrophoresis followed by immunoblotting using the anti-RARα RPα(F ) antibody21 as described in Materials and Methods. (B) Size exclusion HPLC analysis. Nuclear extracts were analyzed as described in the legend of Fig 1. (•) [3H]-t-RA alone; (○) [3H]-t-RA plus 200-fold mol/L excess of unlabeled t-RA. Arrows indicate the elution times of marker proteins (in thousands) used to calibrate the Superose 6 HR 10/30 size exclusion column as indicated in Materials and Methods.

Close modal

The HPLC profile of cytosolic extracts prepared from COS-1 transfected cells showed the presence of a specific t-RA binding component corresponding to an MW of 18,000 and probably representing the endogenous CRABP and similar peaks of specific binding present in nuclear extracts probably resulting from a lysis of a small percentage of nuclei during the preparation of cytosolic extracts (data not shown).

Saturation binding and Scatchard analysis of t-RA and 9-cis-RA.Previous studies20 indicated that the bcr1-PML/RARα isoform bound t-RA with similar affinity as RARα (Kd 0.13 nmol/L and 0.09 nmol/L, respectively). To characterize the binding affinities of t-RA to the bcr3-PML/RARα product we first performed saturation binding and Scatchard analysis. Nuclear extracts prepared from pSG5/bcr3-PML/RARα COS-1 transfected cells were labeled for 18 hours with increasing concentrations of [3H]-t-RA. Bound and free radiolabeled were separated with PD10 desalting columns and counted as described in Materials and Methods. A typical binding curve and Scatchard plot for [3H]-t-RA binding to the bcr3-PML/RARα is shown in Fig 3. The binding of t-RA to the bcr3-PML/RARα was specific, saturable, and exhibited high affinity similar to that described for RARα and bcr1-PML/RARα isoform.20 Scatchard analysis yielded a linear plot consisting of the presence of a single class of binding sites (r = −.91). The Kd value obtained was 0.47 ± 0.01 nmol/L. These data indicate that t-RA binds with relative lower affinity (threefold to fivefold) to bcr3-PML/RARα compared with the previously reported Kd values measured for RARα and bcr1-PML/RARα receptors.20 

Fig. 3.

Saturation binding (A) and Scatchard plot analyses (B) for the binding of [3H]-t-RA to bcr3-PML/RARα. Nuclear extracts from pSG5/bcr3-PML/RARα transiently transfected COS-1 cells were incubated with the indicated concentrations of [3H]-t-RA and analysed using PD10 desalting columns as described in Materials and Methods. Nonspecific binding activity (○) was subtracted from total binding activity (•) to calculate specific binding activity (▪). Specific t-RA binding activity (▪) was also plotted in the form of a Scatchard plot (B).

Fig. 3.

Saturation binding (A) and Scatchard plot analyses (B) for the binding of [3H]-t-RA to bcr3-PML/RARα. Nuclear extracts from pSG5/bcr3-PML/RARα transiently transfected COS-1 cells were incubated with the indicated concentrations of [3H]-t-RA and analysed using PD10 desalting columns as described in Materials and Methods. Nonspecific binding activity (○) was subtracted from total binding activity (•) to calculate specific binding activity (▪). Specific t-RA binding activity (▪) was also plotted in the form of a Scatchard plot (B).

Close modal

It has been shown that 9-cis-RA binds with high affinity to each of the members of the RXR and RAR subfamilies.17,41 Scatchard analyses of the binding of [3H]-9-cis-RA to RARα performed by different groups yield Kd values of 0.24 to 0.31 nmol/L.17,41 We examined the binding of [3H]-9-cis-RA to the bcr1-PML/RARα and bcr3-PML/RARα present in nuclear extracts prepared from COS-1 cells transfected with their respective cDNAs. The [3H]-9-cis-RA binding to both bcr1-PML/RARα and bcr3-PML/RARα was specific and saturable (Fig 4). However, saturation kinetics and Scatchard analyses of the binding of [3H]-9-cis-RA to the bcr1-PML/RARα yield kd value of 0.77 nmol/L (r = −.97), whereas two kd values of 0.45 nmol/L (r = −.98) and 0.075 nmol/L (r = −.96) could be calculated for the bcr3-PML/RARα. These data indicated that 9-cis-RA binds with a lower affinity to the bcr1-PML/RARα than to the bcr3-PML/RARα. Moreover, two sites of 9-cis-RA binding with different affinities are present in the bcr3-PML/RARα fusion product. However, the analysis of the maximum binding showed that low-affinity sites were involved in the majority of this binding activity.

Fig. 4.

Saturation kinetics for the binding of [3H]-9-cis-RA to pSG5/bcr1-PML/RARα (A) and pSG5/bcr3-PML/RARα (B) transiently transfected COS-1 cells. Specific t-RA binding activity (▪) is defined as total binding (•) minus nonspecific binding (○). Scatchard plot analysis of the saturation binding for bcr1-PML/RARα (C) and bcr3-PML/RARα (D).

Fig. 4.

Saturation kinetics for the binding of [3H]-9-cis-RA to pSG5/bcr1-PML/RARα (A) and pSG5/bcr3-PML/RARα (B) transiently transfected COS-1 cells. Specific t-RA binding activity (▪) is defined as total binding (•) minus nonspecific binding (○). Scatchard plot analysis of the saturation binding for bcr1-PML/RARα (C) and bcr3-PML/RARα (D).

Close modal

Competition binding.To further characterize the ligand binding properties of the bcr3-PML/RARα isoform, and the different binding affinities of t-RA and 9-cis-RA for the two major PML/RARα products, competition binding experiments were performed in COS-transfected cells using different unlabeled retinoids such as t-RA, 9-cis-RA, CH55,20,42 AM580, and TTNPB.15,16 In these studies, nuclear fractions prepared from pSG5/RARα, pSG5/bcr1-PML/RARα, and pSG5/bcr3-PML/RARα COS-1 transfected cells were incubated for 18 hours at 4°C with 10 nmol/L [3H]-t-RA in the presence of varying concentrations of the unlabeled retinoid (Table 1). According with previous observations,20 t-RA exhibited similar ability to compete for [3H]-t-RA binding to either RARα and bcr1-PML/RARα (EC50 4.9 and 4.8 nmol/L, respectively). For the bcr3-PML/RARα isoform, the calculated EC50 value for competition binding by t-RA was 7.5 nmol/L. TTNPB and AM580 showed a slightly different specificity (about 1.5- to 2-fold) for competing to the three receptors, whereas CH55 had similar EC50 values. Conversely, competition binding of unlabeled 9-cis-RA to RARα, bcr1-PML/RARα and bcr3-PML/RARα yield EC50 values of 29.5 ± 5.5 nmol/L, 27.5 ± 4.1nmol/L, and 4.8 ± 1.3 nmol/L, respectively. These results indicated that 9-cis-RA binds with a higher specificity to the bcr3-PML/RARα isoform than to the RARα or the bcr1-PML/RARα.

Table 1.

Retinoid Binding Specificity of RARα, bcr1-, and bcr3-PML/RARα

RetinoidEC50 (nmol/L)
RARαPML/RARαPML/RARα
bcr1bcr3
t-RA 4.9 ± 0.7 4.8 ± 0.3 7.5 ± 0.2 
9-cis-RA 29.5 ± 5.5 27.5 ± 4.1 4.8 ± 1.3 
Ch55 15.9 ± 7.1 19.7 ± 4.5 18.3 ± 5.2 
AM580 18.1 ± 1.3 39.1 ± 3.2 22.2 ± 5.4 
TTNPB 52.6 ± 8.3 21.5 ± 9.2 33.1 ± 7.4 
RetinoidEC50 (nmol/L)
RARαPML/RARαPML/RARα
bcr1bcr3
t-RA 4.9 ± 0.7 4.8 ± 0.3 7.5 ± 0.2 
9-cis-RA 29.5 ± 5.5 27.5 ± 4.1 4.8 ± 1.3 
Ch55 15.9 ± 7.1 19.7 ± 4.5 18.3 ± 5.2 
AM580 18.1 ± 1.3 39.1 ± 3.2 22.2 ± 5.4 
TTNPB 52.6 ± 8.3 21.5 ± 9.2 33.1 ± 7.4 

Nuclear extracts prepared from RARα, bcr1-, and bcr3-PML/RARα transiently transfected COS-1 cells were labeled for 18 hours at 4°C with 10 nmol/L [3H]-tRA in the presence of increasing concentrations (0.5 nmol/L to 1 μmol/L) of the indicated unlabeled retinoid. RA binding was analysed using PD10 desalting columns (Pharmacia). The EC50 values were calculated using the nonlinear least squares regression analysis program ALLFIT. Values are means for at least three determinations ± SD.

Functional transactivation assay in cotransfected COS-1 cells.We next investigated whether the higher affinity and specificity of 9-cis-RA for the bcr3-PML/RARα isoform resulted in a different ability of this protein to transactivate two different retinoic acid-response elements (RARE) such as the βRARE and the TRE, compared with RARα and/or bcr1-PML/RARα. The βRARE is the direct repeat sequence (DR5) of the RARβ2 promoter. The TRE is a palindromic sequence that also mediates thyroid hormone transactivation. In COS-1 cells, when RARα, bcr1-PML/RARα or bcr3-PML/RARα expression vectors were cotransfected with either the (βRARE)3-tk-LUC or TRE2-tk-LUC reporters, treatment with t-RA and 9-cis-RA induced luciferase activity in a dose-dependent manner. However, 9-cis-RA was less potent than t-RA in activating these repoter genes (Table 2). The magnitude of the (βRARE)3-tk-LUC induction by t-RA was similar for the three receptors (EC50 ranging between 0.4 to 0.9 nmol/L). 9-cis-RA was able to induce (βRARE)3-tk-LUC about fourfold more efficiently in RARα expressing cells than in bcr1- or bcr3-PML/RARα-COS-1 transfected cells (Table 2). However, under these conditions 9-cis-RA had very similar EC50 for the two PML/RARα isoforms. The different transactivation activity between t-RA and 9-cis-RA was abolished when the RARβ-pr LUC,8,35 a reporter plasmid containing the RARβ gene promoter region (−5 kb to +155) fused to the promoterless luciferase gene was used (Table 3). As recently shown,43 it is possible that the mechanism of ligand-dependent transcription is modulated by a number of positive and negative factors that may not be functional when direct repeat sequences are used.

Table 2.

Transactivation Properties of Different Retinoids

TRE2-tk-LUC(βRARE)3-tk-LUC
EC50 (nmol/L)EC50 (nmol/L)
t-RA9cisRAAM580TTNPBt-RA9cisRAAM580TTNPB
RARα 1.6 367.9 0.2 0.03 0.4 6.6 0.4 0.002 
bcr1-PML/RARα 0.5 302.6 0.2 0.02 0.8 28.9 0.5 0.002 
bcr3-PML/RARα 0.3 37.5 0.4 0.15 0.9 28.9 2.7 0.015 
TRE2-tk-LUC(βRARE)3-tk-LUC
EC50 (nmol/L)EC50 (nmol/L)
t-RA9cisRAAM580TTNPBt-RA9cisRAAM580TTNPB
RARα 1.6 367.9 0.2 0.03 0.4 6.6 0.4 0.002 
bcr1-PML/RARα 0.5 302.6 0.2 0.02 0.8 28.9 0.5 0.002 
bcr3-PML/RARα 0.3 37.5 0.4 0.15 0.9 28.9 2.7 0.015 

Extracts were prepared from transiently cotransfected COS-1 cells and analyzed as described in Materials and Methods. The EC50 values (retinoid concentrations required to induce 50% of the maximal response) were calculated using the nonlinear test squares regression analysis program ALLFIT.

Table 3.

Transactivation Properties of All-trans-RA and 9-cis-RA on the Reporter Plasmid Containing the RARβ Gene Promoter Region −5 kb to +155 (RARβ pr-LUC)

RARβ pr-LUC
EC50 (nmol/L)
t-RA9cisRA
RARα 
bcr1-PML/RARα 175 110 
bcr3-PML/RARα 128 166 
RARβ pr-LUC
EC50 (nmol/L)
t-RA9cisRA
RARα 
bcr1-PML/RARα 175 110 
bcr3-PML/RARα 128 166 

Extracts were prepared and analyzed as described in the legend of Table 2.

T-RA was about threefold to fivefold more potent in activating the TRE2-tk-LUC reporter gene in COS-1 cells expressing the bcr1- and bcr3-PML/RARα than in COS-1 cells expressing RARα. In contrast, 9-cis-RA treatment was equally potent in activating this reporter gene in RARα and bcr1-PML/RARα expressing cells. Remarkably, 9-cis-RA was about 10-fold more active in activating the TRE2-tk-LUC in the bcr3-PML/RARα than in the RARα or in the bcr1-PML/RARα COS-1 transfected cells. These results suggested that the higher affinity and specificity of the binding of 9-cis-RA to the bcr3-PML/RARα product resulted in an increased ability of this protein to specifically transactivate the TRE2-tk-LUC, and not the (βRARE)3-tk-LUC, in transiently transfected COS-1 cells.

In agreement with this, preliminary results obtained from blast cells directly isolated from two bcr3-PML/RARα patients showed that 9-cis-RA was more potent than t-RA in inducing cell differentiation, as evaluated by morphology, by the nitroblue tetrazolium (NBT) dye reduction assay and by type II transglutaminase activity induction.38 In fact, the calculated EC50 for these effects were estimated to be approximately 1.8 nmol/L for t-RA and 0.1 nmol/L for 9-cis-RA, respectively.

Additional differences between binding specifity and transcriptional activity were also detected for the RARα- and RAR-selective retinoids AM580 and TTNPB. AM580 and TTNPB in fact, resulted to be very effective in transactivating either TRE2-LUC or (βRARE)3-tk-LUC in cotransfected COS-1 cells. However, these retinoids were consistently found to be less active in transactivating these RAREs in the bcr3-PML/RARα COS-1 transfected cells (Table 2).

Retinoic acid binding properties of PLZF/RARα.We then compared the t-RA binding properties of PML/RARα isoforms with that of the PLZF/RARα, the fusion protein generated by the less frequent chromosomal rearrangement t(11; 17) present in APL and associate with refractoriness to t-RA treatment. Nuclear and cytosolic extracts were prepared from COS-1 cells transfected with a pMT2 expression vector containing a cDNA encoding the human PLZF/RARα. PLZF/RARα was able to bind t-RA and t-RA specific binding activity was found mainly associated with the nuclear fraction (Fig 5). Similarly to PML/RARα, PLZF/RARα was found present in high-molecular-weight nuclear complexes eluting with apparent MW of 950,000 and 500,000. Similar HPLC profile was obtained in nuclear extracts prepared from myeloid U937 precursor cells expressing PLZF/RARα (data not shown). Moreover, as in the case of RARα and bcr1-PML/RARα,17,20,31,41 a single site of specific high-affinity binding could be detected for PLZF/RARα by saturation kinetics and Scatchard analyses. The calculated binding affinities were 0.17 nmol/L for t-RA and 2.7 nmol/L for 9-cis-RA (data not shown).

Fig. 5.

Size exclusion HPLC analysis of [3H]-t-RA binding to nuclear (A) and cytosolic (B) extracts prepared from pMT2/PLZF/RARα transiently transfected COS-1 cells. (•) [3H]-t-RA alone; (○) [3H]-t-RA plus 200-fold mol/L excess of unlabeled t-RA. The Superose 6 HR 10/30 size exclusion column was calibrated with the indicated marker proteins (in thousands) as described in Materials and Methods.

Fig. 5.

Size exclusion HPLC analysis of [3H]-t-RA binding to nuclear (A) and cytosolic (B) extracts prepared from pMT2/PLZF/RARα transiently transfected COS-1 cells. (•) [3H]-t-RA alone; (○) [3H]-t-RA plus 200-fold mol/L excess of unlabeled t-RA. The Superose 6 HR 10/30 size exclusion column was calibrated with the indicated marker proteins (in thousands) as described in Materials and Methods.

Close modal

Expression of RARs and RXRs in APL patients.Retinoid induced differentiation involves activation of multiple RA-dependent signaling pathways, including RARs and RXRs. Therefore, we have analyzed the pattern of expression of retinoid receptors in leukemic blasts expressing bcr1 or bcr3 isoforms of PML/RARα by Northern blotting analysis of RNA samples from 4 bcr1-PML/RARα and 5 bcr3-PML/RARα APL patients (Fig 6). As previously described,1,44,45 three RARα transcripts of 2.1, 3.0, and 4.8 kb were detectable in mRNAs prepared from all the APL patients. 3.1-kb RARγ and 5.6-kb RXRα transcripts were relatively highly expressed in APL patients whereas RARβ and RXRγ transcripts were undetectable. As expected, samples from bcr1- and bcr3-PML/RARα APL patients expressed the same subtypes of RAR or RXR receptor mRNAs. RXRβ transcript was about equally expressed in all the samples whereas slightly different levels of expression of RARα, RARγ, and RXRα transcripts were detectable. However, a correlation between the expression level of RARs and RXRs and the presence of a particular PML/RARα isoform was not found.

Fig. 6.

Expression of RAR's and RXR's mRNAs in fresh cells from APL patients. Total RNA (10 μg) prepared from Ficoll-Hypaque–isolated APL blasts was fractionated, transferred to Nytran membrane, and hybridized to [32P]-labeled cDNA probes for RAR-α, -β, -γ, RXR-α, -β, -γ. Hybridization to GAPDH was used as a control.

Fig. 6.

Expression of RAR's and RXR's mRNAs in fresh cells from APL patients. Total RNA (10 μg) prepared from Ficoll-Hypaque–isolated APL blasts was fractionated, transferred to Nytran membrane, and hybridized to [32P]-labeled cDNA probes for RAR-α, -β, -γ, RXR-α, -β, -γ. Hybridization to GAPDH was used as a control.

Close modal

The combination of t-RA and conventional cytotoxic agents has been found to provide a more effective overall therapy for APL than conventional chemotherapy or retinoid therapy alone.3,4,12,46 Therapies including t-RA induced terminal differentiation of the leukemic promyelocytes, which are then replaced by normal hematopoiesis.3,4 However, the precise molecular mechanism of RA-induced differentiation in APL blasts has yet to be elucidated. It has been shown that the ligand binding domains of RARs and of the other members of the nuclear receptor superfamily also includes homodimerizing and heterodimerizating interfaces, ligand-dependent transcriptional activation function 2 (AF-2), and in some cases, hormone reversible transcriptional repression functions.15,43 All PML/RARα isoforms retain most of the RARα functional domains.3,4 In addition, APL blasts containing bcr1- and bcr3-PML/RARα also express transcripts encoding for RARα, RARγ, RXRα, and RXRβ. Moreover, specific retinoid binding probably corresponding to RARs and to CRABPs was detectable in these cells. These data indicate that in APL cells multiple retinoid signaling pathways are probably involved in RA-induced differentiation.

T-RA is an effective inducer of clinical remission only in patients carrying the t(15; 17) and expressing the PML/RARα product.3,4,12,47 APL patients defined as M3 by FAB criteria,22 but lacking the t(15; 17) or presenting t(11; 17), do not respond to t-RA–based treatments.10,47 Recent clinical studies indicate a different prognosis in APL patients treated with t-RA alone or with t-RA followed by chemotherapy on the basis of the fusion product present.13,14,48 In APL, the bcr3-PML/RARα isoform has been associated with the more aggressive microgranular APL variant, to CD2 expression, to higher leukocyte counts, and to poorer prognosis.12-14,49 However, in patients expressing the two major isoforms of PML/RARα treated with combined t-RA and chemotherapy no statistical differences in overall survival and complete remission duration was found.50 These findings stress the importance of a complete analysis of the molecular and clinical characteristics of APL patients to define t-RA sensitivity and response to differentiation therapy.

In this study we investigated the molecular mechanism of t-RA response in APL by analyzing functional properties such as retinoid binding and transcriptional activation of bcr1- and bcr3-PML/RARα, the two predominant fusion proteins present in APL.

In particular, we measured the binding properties of the two endogenous ligands t-RA and 9-cis-RA to bcr1- and bcr3-PML/RARα isoforms. By Scatchard analysis we showed that t-RA binds to the bcr3-PML/RARα isoform with slightly less affinity (Kd = 0.4 nmol/L) than to bcr1 PML/RARα or RARα (Kd = 0.13 nmol/L or 0.09 nmol/L, respectively). A relatively lower t-RA binding specificity for the bcr3-PML/RARα could also be measured in competitive binding analysis. Moreover, these experiments showed that 9-cis-RA is able to bind with about eightfold higher specificity to bcr3-PML/RARα than to RARα or bcr1-PML/RARα (EC50 values of 4.8 nmol/L, 33.5 nmol/L, and 37.5 nmol/L, respectively). This finding was also supported by saturation kinetics studies which indicated that [3H]-9-cis-RA binds with a lower affinity to the bcr1-PML/RARα than to the bcr3-PML/RARα. Two sites of 9-cis-RA binding with different affinities were detectable in the bcr3-PML/RARα fusion product. In fact, for bcr1-PML/RARα a Kd value of 0.77 nmol/L was measured, whereas two Kd values of 0.45 nmol/L and 0.075 nmol/L could be calculated for bcr3-PML/RARα.

Both PML/RARα isoforms retain the RING domain of PML which is probably involved in protein-protein interactions and regions involved in DNA binding.49 However, the bcr3-PML/RARα product lacks the C-terminal proline/serine–rich region of PML.7 It has been shown that human RARα contains overlapping but distinct binding pockets for t-RA and 9-cis-RA in a region which corresponds to the ligand-dependent transcriptional activation function 2 (AF-2 ) domain.51 This region in RARα is predicted to form an amphipatic α-helix that is well conserved among nuclear receptors.52 Using alanine scanning mutagenesis, specific amino acids that constitute the high-affinity binding of 9-cis-RA have been defined within this region.53

Our hypothesis of functional differences between the two isoforms is also supported by the fact that different protein complexes are probably formed by the two PML/RARα fusion products. At variance to RARα,20,31 both PML/RARα isoforms were found in high molecular nuclear complexes by HPLC size exclusion analysis of nuclear extracts prepared from bcr1-PML/RARα and bcr3-PML/RARα transiently transfected COS-1 cells. However, the bcr3-PML/RARα product was present only in protein-protein complexes as shown by the absence of any RA-specific binding corresponding to the monomeric form of this protein either in COS-1 transfected cells as well as in APL patients nuclear extracts. In similar experimental conditions, RA-specific binding activity corresponding to the monomeric form of the bcr1-PML/RARα product was always measurable in NB4 and in COS-1 transfected cells.20,38 In addition, identical HPLC profiles were obtained if nuclear extracts from mock, RARα, bcr1-, and bcr3-PML/RARα COS-1 transfected cells were labeled with [3H]-9cis-RA (data not shown).

It has been shown that both PML/RARα and PLZF/RARα fusion proteins contain identical portions of RARα and retain the ability to act as RA-dependent transcription factors, to bind to RA-response elements, and to antagonize the function of the wild-type RARα.5-9,54 Therefore, it appears that they both have the potential to directly influence the RARα-dependent endogenous pathway that control terminal myeloid differentiation. The introduction of PLZF/RARα cDNA into COS-1 cells by transient transfection resulted in high levels of nuclear t-RA specific binding activity. The affinity of t-RA binding to PLZF/RARα was similar to that of PML/RARα. However, APL patients with the t(11; 17) expressing the PLZF/RARα are refractory to t-RA–induced differentiation9,10,54 and the expression of PLZF/RARα in hematopoietic precursor cells blocks terminal differentiation (M. Ruthardt and P.G. Pelicci, manuscript submitted). Therefore, it seems that despite PML/RARα and PLZF/RARα having similar retinoid-binding properties they have different effects in vivo on the RA-signaling pathway. The biochemical consequences of PML or PLZF sequences on the in vivo activity of the two RARα fusion proteins might differ and underlie their different biologic activities. However, the presence of both PML/RARα and PLZF/RARα in similar high-molecular-weight nuclear complexes suggests the involvement of these complexes in the block of promyelocytic differentiation.

Previous reports indicate only minor differences between the two PML/RARα isoforms in the activation of various RAREs in a variety of target cells.5-8 In this study we extended these previous findings, most of which had been obtained using a single t-RA concentration, by analyzing the t-RA and 9-cis-RA dose-dependent activation of different RAREs in RARα, bcr1-PML/RARα and bcr3-PML/RARα COS-1 transfected cells. We found that bcr1-PML/RARα and bcr3-PML/RARα overexpressed in COS-1 cells had similar alterations in activating (βRARE)3-tk-LUC and RARβ pr-LUC when compared with wild-type RARα. However, the higher affinity and specificity of 9-cis-RA binding to the bcr3-PML/RARα product resulted in an increased ability (about 10-fold) of this fusion product to specifically transactivate the TRE2-tk-LUC. It should be noted that the TRE is usually present in few RA target genes and is less responsive to RA than the DR-5 motif, thus far the strongest and the most abundant class of RARE present in the promoter of the RARβ and in the promoters of several RA-responsive genes.43,55 In agreement with these results, we found the 9-cis-RA to be more potent than t-RA in the induction of differentiation of blast cells from two bcr3-PML/RARα APL patients in culture (data not shown).

In conclusion, our results indicate that distinct functional properties are associated with the two major PML/RARα isoforms. This in turn may provide a rationale for targeting differential retinoid therapy at the specific molecular lesion of APL patients.

The manuscript by M. Ruthardt and P.G. Pelicci listed as submitted is now in press (Ruthardt M, Testa U, Nervi C, Ferrucci PF, Grignani F, Puccetti E, Grignani F, Peschle C, Pelicci PG: Mol Cell Biol, 1997).

We thank Dr. P. Chambon for providing RARα antibody and RARs cDNA and Dr R.M. Evans for providing RXRs cDNA. The RARβ pr-luc was kindly provided by Dr A. Dejean.

Supported by grants from the European Economic Community (Biomed and Biotech), from the Associazione Italiana per la Ricerca sul Cancro (AIRC), from Ministero Università e Ricerca Scientifica e Tecnologica (MURST), and from CNR special project A.C.R.O., CT 95.00.454.39.

Address reprint requests to Clara Nervi, MD, PhD, Department of Histology and Medical Embryology, University of Rome “La Sapienza,” Via A. Scarpa 14, 00161 Rome, Italy.

1
Longo
L
Pandolfi
PP
Biondi
A
Rambaldi
A
Mencarelli
A
Lo
Coco F
Diverio
D
Pegoraro
L
Avanzi
G
Tabilio
A
Zangrilli
D
Alcalay
M
Donti
E
Grignani
F
Pelicci
PG
Rearrangements and aberrant expression of the retinoic acid receptor α gene in acute promyelocytic leukemia.
J Exp Med
172
1990
1571
2
Miller
WH Jr
Warrell
RP Jr
Frankel
SR
Jakubowski
A
Gabrilove
JL
Muindi
J
Dmitrovsky
E
Novel retinoic acid receptor-α transcripts in acute promyelocytic leukemia responsive to all-trans-retinoic acid.
J Natl Cancer Inst
82
1990
1932
3
Warrell
RP Jr
de Thé
H
Wang
ZY
Degos
L
Acute promyelocytic leukemia.
N Engl J Med
329
1993
177
4
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
5
Pandolfi
PP
Alcalay
M
Mencarelli
A
Biondi
A
Lo
Coco F
Grignani
F
Pelicci
PG
Structure and origin of the acute promyelocytic leukemia myl/RARα cDNA and characterization of its retinoid-binding and transactivation properties.
Oncogene
6
1991
1285
6
Kastner
P
Perez
A
Lutz
Y
Rochette-Egly
C
Gaub
MP
Durand
B
Lanotte
M
Berger
R
Chambon
P
Structure, localization and transcriptional properties of two classes of retinoic acid receptor α fusion proteins in acute promyelocytic leukemia (APL): Structural similarities with a new family of oncoproteins.
EMBO J
11
1992
629
7
Kakizuka
A
Miller
WH Jr
Umesono
K
Warrell
RP Jr
Frankel
SR
Murty
VVVS
Dmitrovsky
E
Evans
RM
Chromosomal translocation t(15; 17) in human acute promyelocytic leukemia fuses RARα with a novel putative transcription factor, PML.
Cell
66
1991
663
8
de Thé
H
Lavau
C
Marchio
A
Chomienne
C
Degos
L
Dejean
A
The PML-RARα fusion mRNA generated by the t(15; 17) translocation in acute promyelocytic leukemia encodes a functionally altered RAR.
Cell
66
1991
675
9
Chen
Z
Brand
NJ
Chen
A
Chen
S-J
Tong
JH
Wang
Z-Y
Waxman
S
Zelent
A
Fusion between a novel Kruppel-like zinc finger gene and retinoic acid receptor-α locus due to a variant t(11; 17) translocation associated with acute promyelocytic leukemia.
EMBO J
12
1993
1161
10
Licht
JD
Chomienne
C
Goy
A
Chen
A
Scott
AA
Head
DR
Michaux
JL
Wu
Y
Deblasio
A
Miller
WH Jr
Zelenetz
AD
Willman
CL
Chen
Z
Chen
SJ
Zelent
A
Macintyre
E
Veil
A
Cortes
J
Kantarjian
H
Waxman
S
Clinical and molecular characterization of a rare syndrome of acute promyelocytic leukemia associated with translocation (11; 17).
Blood
85
1995
1083
11
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
12
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
13
Huang
W
Sun
GL
Li
XS
Cao
Q
Lu
Y
Jang
GS
Zhang
FQ
Chai
JR
Wang
Z
Waxman
S
Chen
Z
Chen
SJ
Acute promyelocytic leukemia: Clinical relevance of two major PML-RARα isoforms and detection of minimal residual disease by retrotranscriptase/polimerase chain reaction to predict relapse.
Blood
82
1993
1264
14
Vahdat
L
Maslak
P
Miller
WH Jr
Eardley
A
Heller
G
Scheinberg
DA
Warrell
RP Jr
Early mortality and the retinoic acid syndrome in acute promyelocytic leukemia: Impact of leukocytosis, low dose chemotherapy, PMN/RAR-α isoform, and CD13 expression in patients treated with all-trans retinoic acid.
Blood
84
1994
3843
15
Sporn MB, Roberts AB, Goodman DS: The Retinoids: Biology, Chemistry and Medicine. New York, NY, Raven, 1994
16
Blomhoff R: Vitamin A in Health and Disease. New York, NY, Dekker, 1994
17
Allenby
G
Bocquel
T
Saunders
M
Kazmer
S
Speck
J
Rosenberger
M
Lovey
A
Kastner
P
Grippo
JF
Chambon
P
Levin
AA
Retinoic acid receptors and retinoid X receptors: Interactions with endogenous retinoic acids.
Proc Natl Acad Sci USA
90
1993
30
18
Heyman
RA
Mangelsdorf
DJ
Dyck
JA
Stein
RB
Eichele
G
Evans
RM
Thaller
C
9-cis retinoic acid is a high affinity ligand for the retinoid X receptor.
Cell
68
1992
397
19
Levin
AA
Sturzenbecker
LJ
Kazmer
S
Bosakowsky
T
Huselton
C
Allenby
G
Speck
J
Kratzeisen
C
Rosenberger
M
Lovey
A
Grippo
JF
9-cis retinoic acid stereoisomer binds and activates the nuclear receptor RXRα.
Nature
355
1992
359
20
Nervi
C
Poindexter
EC
Grignani
F
Pandolfi
PP
Lo
Coco F
Avvisati
G
Pelicci
PG
Jetten
AM
Characterization of the PML-RARα chimeric product of the acute promyelocytic leukemia specific t(15; 17) translocation.
Cancer Res
52
1992
3687
21
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
22
Bennett
JM
Catovsky
D
Daniel
MT
Flandrin
G
Galton
DA
Gralnick
HR
Sultan
C
Proposed revised criteria for the classification of acute myeloid leukemia. A report of the French-American-British Cooperative Group.
Ann Intern Med
103
1985
620
23
Biondi
A
Rambaldi
A
Alcalay
M
Pandolfi
PP
Lo
Coco F
Diverio
D
Rossi
V
Mencarelli
A
Longo
L
Zangrilli
D
Masera
G
Barbui
T
Mandelli
F
Grignani
F
Pelicci
PG
RARα rearrangements as genetic markers for diagnosis and monitoring in acute promyelocytic leukemia.
Blood
77
1991
1418
24
Sambrook J, Fritsch EF, Maniatis T: Molecular Cloning: A Laboratory Manual. Cold Spring Harbor, NY, Cold Spring Harbor Laboratory, 1989
25
Brand
N
Petkovich
M
Krust
A
Chambon
P
de Thé
H
Marchio
A
Tiollais
P
Dejean
A
Identification of a second human retinoic acid receptor.
Nature
332
1988
850
26
Krust
A
Kastner
P
Petkovich
M
Zelent
A
Chambon
P
A third human retinoic acid receptor, hRAR-gamma.
Proc Natl Acad Sci USA
86
1989
5310
27
Mangelsdorf
DJ
Ong
ES
Dyck
JA
Evans
RM
Nuclear receptor that identifies a novel retinoic acid response pathway.
Nature
345
1990
224
28
Mangelsdorf
DJ
Borgmeyer
U
Heyman
RA
Zhou
JY
Ong
ES
Oro
AE
Kakizuka
A
Evans
RM
Characterization of three RXR genes that mediate the action of 9-cis retinoic acid.
Genes Dev
6
1992
329
29
Dugaicyzk
AD
Haron
JA
Stone
EM
Dennison
OE
Rothblum
KN
Schwartz
RJ
Cloning and sequencing of a ribonucleic acid copy of glyceraldehyde-3-phosphate dehydrogenase messenger ribonucleic acid isolated from chicken muscle.
Biochemistry
22
1983
1605
30
Pandolfi
PP
Alcalay
M
Fagioli
M
Zangrilli
D
Mencarelli
A
Diverio
D
Biondi
A
Lo
Coco F
Rambaldi
A
Grignani
F
Rochette-Egly
C
Gaub
MP
Chambon
P
Pelicci
PG
Genomic variability and alternative splicing generate multiple PML/RARa transcripts that encode aberrant PML proteins and PML/RARα isoforms in acute promyelocitic leukaemia.
EMBO J
11
1992
1397
31
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
32
Jetten AM, Grippo JF, Nervi C: Isolation and binding characteristics of nuclear retinoic acid receptors, in Packer L (ed): Methods in Enzymology, vol “Retinoids.” San Diego, CA, Academic, 1990, p 248
33
Rodbard D: Mathematics of hormone-receptor interaction. Basic principles, in O'Malley BW, Means AR (eds): Receptors for Reproductive Hormones. New York, NY, Plenum, 1973, p 289
34
DeLean A, Munson PJ, Rodbard D: Simultaneous analysis of families of sigmoidal dose curves: Application to bioassay, radioligand assay, and physiological dose-response curves. Am J Physiol 235:E97, 1973
35
de Thé
H
delMar Vivanco-Ruiz M
Tiollais
P
Stunnenberg
H
Dejean
A
Identification of a retinoic acid response element in the retinoic acid receptor β gene.
Nature
343
1990
177
36
de Verneuil
H
Metzger
D
The lack of transcriptional activation of the v-erbA oncogene is part due to a mutation present in the binding domain of the protein.
Nucleic Acids Res
18
1990
4489
37
Giguere V: Application of the firefly luciferase reporter gene, in Murray EJ (ed): Gene Transfer and Expression Protocols. Methods in Molecular Biology. Clifton, NJ, Humana, 1991, p 237
38
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
39
Petkovich
M
Brand
NJ
Krust
A
Chambon
P
A human retinoic acid receptor which belongs to the family of nuclear receptors.
Nature
330
1987
444
40
Giguere
V
Ong
ES
Segui
P
Evans
RM
Identification of a new class of steroid hormone receptors.
Nature
331
1988
91
41
Allegretto
EA
McClurg
MR
Lazarchik
SB
Clemm
DL
Kerner
SA
Elgort
MG
Boehm
MF
White
SK
Pike
JW
Heyman
RA
Transactivation properties of retinoic acid and retinoid X receptors in mammalian cells and yeast.
J Biol Chem
268
1993
26625
42
Nervi
C
Vollberg
TM
Grippo
JF
Lucas
DA
George
MD
Sherman
MI
Shudo
K
Jetten
AM
Expression of nuclear retinoic acid receptors in wilde type and mutant embryonal carcinoma PCC4.aza1R cells.
Cell Growth Differ
1
1990
535
43
Mangelsdorf
DJ
Evans
RM
The RXR heterodimers and orphan receptors.
Cell
83
1995
841
44
de Thé
H
Chomienne
C
Lanotte
M
Degos
L
Dejean
A
The t(15; 17) translocation of acute promyelocytic leukemia fuses the retinoic acid receptor α gene to a novel transcribed locus.
Nature
347
1990
558
45
Goddard
AD
Borrow
J
Freemont
P
Solomon
E
Characterization of a zinc finger gene disrupted by the t(15; 17) in acute promyelocytic leukemia.
Science
254
1991
1371
46
Avvisati
G
Lo
Coco F
Diverio
D
Falda
M
Ferrara
F
Lazzarino
M
Russo
D
Petti
MC
Mandelli
F
AIDA (all-trans-retinoic acid plus Idarubicin) in newly diagnosed acute promyelocytic leukemia: A GIMEMA pilot study.
Blood
88
1996
1390
47
Miller
WH
Jr
Kakizuka
A
Frankel
SR
Warrell
RP Jr
Levine
K
Evans
RM
Dmitrovsky
E
Reverse transcriptase/polymerase chain reaction amplification for PML/RAR-α clarifies diagnosis and detects minimal residual disease in acute promyelocytic leukemia.
Proc Natl Acad Sci USA
89
1992
2694
48
Gallagher
RE
Li
YP
Rao
S
Paietta
E
Andersen
J
Etkind
P
Bennett
JM
Tallman
MS
Wiernik
PH
Characterization of acute promyelocytic leukemia cases with PML-RARα break/fusion sites in PML exon 6: Identification of a subgroup with decreased in vitro responsiveness to all-trans-retinoic acid.
Blood
86
1995
1540
49
Claston
DF
Reading
CL
Nagarajan
L
Tsujimoto
Y
Andersson
BS
Estey
E
Cork
A
Huh
YO
Trujillo
J
Deisseroth
AB
Correlation of CD2 expression with PML gene breakpoints in patients with acute promyelocitic leukemia.
Blood
80
1992
582
50
Fukutani
H
Naoe
T
Ohno
R
Yoshida
H
Miyawaki
S
Shimazaki
C
Miyake
T
Nakayama
Y
Kobayashi
H
Goto
S
Takeshita
A
Kobayashi
S
Kato
Y
Shiraishi
K
Sasada
M
Ohtake
S
Murakami
H
Kobayashi
M
Endo
N
Shindo
H
Matsushita
K
Hasegawa
S
Tsuji
K
Ueda
Y
Tominaga
N
Furuya
H
Inoue
Y
Takeuchi
J
Morishita
H
Iida
H
Isoforms of PML-retinoic acid receptor alpha fused transcripts affect neither clinical features of acute promyelocytic leukemia nor prognosis after treatment with all-trans-retinoic acid.
Leukemia
9
1995
1478
51
Tate
BF
Allenby
G
Janocha
R
Kazmer
S
Speck
J
Sturzenbecker
LJ
Abarzùa
P
Levin
AA
Grippo
JF
Distinct binding determinants for 9-cis-retinoic acid are located within AF-2 of retinoic acid receptor α.
Mol Cell Biol
14
1994
2323
52
Danielian
PS
White
R
Lees
JA
Parker
MG
Identification of a conserved region required for hormone dependent transcriptional activation by steroid hormone receptors.
EMBO J
11
1992
1025
53
Tate
BF
Grippo
JF
Mutagenesis of the ligand binding domain of the human retinoic acid receptor α identifies critical residues for 9-cis-retinoic acid binding.
J Biol Chem
270
1995
20258
54
Chen
Z
Guidez
F
Rousselot
P
Agadira
A
Chen
S-J
Wang
Z-Y
Degos
L
Zelent
A
Waxman
S
Chomienne
C
PLZF-RARα 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 USA
91
1994
1178
55
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
Sign in via your Institution