Acute promyelocytic leukemia (APL) is always associated with chromosomal translocations that disrupt the retinoic acid receptor α (RARα) gene. Whether these translocations relate to a role for endogenous RARα in normal granulopoiesis remains uncertain because most studies addressing this question have used non-physiological overexpression systems. Granulocyte differentiation in cells derived from RARα-deficient (RARα−/−) mice was studied and evaluated in the context of agonist-bound and ligand-free RARα. Our results demonstrate that RARα is dispensable for granulopoiesis, as RARα−/− mice have a normal granulocyte population despite an impaired ability to respond to retinoids. However, although it is not absolutely required, RARα can bidirectionally modulate granulopoiesis. RARα stimulates differentiation in response to exogenous retinoic acid. Furthermore, endogenous retinoids control granulopoiesis in vivo, as either vitamin A–deficient mice or animals treated with an RAR antagonist accumulate more immature granulocytes in their bone marrow. Conversely, RARα acts to limit differentiation in the absence of ligand because granulocyte precursors from RARα−/− mice differentiate earlier in culture. Thus, the block in granulopoiesis exerted by RARα fusion proteins expressed in APL cells may correspond to an amplification of a normal function of unliganded RARα.

The retinoic acid receptor α (RARα) gene is altered by chromosomal translocations in all cases of acute promyelocytic leukemia (APL). These translocations lead to the synthesis of chimeric proteins, resulting from the fusion of novel N-terminal sequences to RARα amino acids 60-462.1-3 To date, 5 distinct fusion partners for RARα have been identified, all of which belong to distinct protein families. PML, the most frequent partner, is a nuclear protein containing a RING finger and a coiled coil. PML has been implicated in regulating the formation and function of nuclear bodies, nuclear substructures whose function is not completely understood.4 Less frequent partners include PLZF (a zinc finger transcription factor),5 NuMA (a mitotic apparatus protein),6 nucleophosmin,7 and STAT5b.8 Forced expression of either PML/RARα or PLZF/RARα induces an APL-like disease in transgenic mice and establishes the causal role of these fusion proteins in APL leukemogenesis.9-11 At the molecular level, both PML/RARα and PLZF/RARα are strong repressors, efficiently recruiting a complex containing the nuclear receptor corepressors N-CoR/SMRT and histone deacetylase to target promoters.9,12-15 Importantly, these repressive properties appear to be closely linked with their leukemogenic potential, as histone deacetylase inhibitors can reverse the leukemic phenotype and lead to remission in transgenic mouse models of APL.9 

Despite accumulating data implicating the fusion proteins in leukemia, relatively little is known about how their action relates to a normal function of RARα during granulopoiesis, the process by which immature myeloid cells, such as promyelocytes, differentiate to mature neutrophils. In the hematopoietic system, RARα is expressed at significant levels in granulocytes, indicating a possible function within this lineage.16-18 RA has been shown to act as a differentiating agent for both normal and transformed immature myeloid cells,19,20 suggesting that the fusion proteins might antagonize normal RA-dependent differentiation. Conversely, overexpression of wild type (WT) RARα (as well as dominant negative forms of this receptor) has also been shown to block promyelocyte differentiation in culture.21-23 Thus, at limiting RA concentrations, an increase in the proportion of unliganded RAR molecules may lead to a dominant-negative inhibition of RA-controlled differentiation. Lastly, RA appears to orient the differentiation of pluripotent hematopoietic progenitors toward the granulocyte lineage, indicating that RA may be involved in granulocyte fate specification.24 These various studies, however, which employ either overexpressed receptors or exogenous ligands, fall short of demonstrating a physiological role for endogenous RARα in granulopoiesis. By studying cells derived from RARα-deficient mice, we demonstrate here that RARα is a bidirectional modulator of granulocyte differentiation: In the absence of RA, RARα acts to limit granulocyte differentiation; in the presence of RA, RARα stimulates differentiation.

Mice

RARα- and RARγ-deficient mice were described previously.25,26 Mice were genotyped by polymerase chain reaction (PCR) (primers available upon request). All mice used in this study were between 2 and 6 months of age. Vitamin A deficiency (VAD) in cellular retinol-binding protein type I (CRBPI)−/− mice was produced by maintaining CRBPI−/− mice (all males) on a VAD diet for at least 16 weeks after birth.27 VAD was confirmed by the observation of several VAD-related phenotypes: keratinization of the seminal vesicle and prostate epithelia, testis degeneration, and retinal defects.27 These symptoms were specifically observed in CRBPI−/− mice fed a VAD diet, not in control mice (ie, WT mice fed a VAD diet or CRBPI−/− mice fed a normal diet). All-trans-retinoic acid (ATRA) (Sigma Chemical Co, St Louis, MO) and the pan-RAR antagonist BMS493 were dissolved in ethanol and then placed in an emulsion with corn oil by vigorous vortexing. The emulsion was administered to test animals by gavage every 12 hours (BMS493) or every 36 hours (ATRA) for 3 days. Bone marrow (BM) cells were harvested by flushing femurs with medium. Fetal liver (FL) cell suspensions were prepared by sequential passage of dissected FLs through 18-, 22-, and 25-gauge needles.

Cell culture

Liquid myeloid cultures were performed by seeding 2 × 106 BM cells in 10 mL Iscoves modified Dulbecco medium supplemented with 20% fetal bovine serum (Gibco-BRL Life Technologies, Grand Island, NJ), 5 ng/mL granulocyte colony-stimulating factor (G-CSF), and 5 ng/mL stem cell factor (SCF) (R&D Systems, Minneapolis, MN), penicillin/streptomycin, and L-glutamine (2 mM final concentration). Ligands were added in 10 μL volumes from a 10−3 M solution in ethanol. Labeling with 5- and 6-carboxy fluorescein diacetate, succinimidyl ester (CFSE) (Molecular Probes, Eugene, OR) was performed by incubating BM cells for 10 minutes at 37°C at a concentration of 107 cells per mL in medium supplemented with 2 μL/mL of a 10 mg/mL CFSE solution in dimethyl sulfoxide followed by 2 washes with 10 mL medium. Clonogenic cultures were performed with commercial methylcellulose media (Methocult 3434; Stem Cell Technologies, Vancouver, BC, Canada). The media contained erythropoietin (EPO), interleukin-3 (IL-3), IL-6, and SCF for the experiments described in Figure1C and Figure2, and the media contained Methocult 3234 supplemented with granulocyte-macrophage (GM)–CSF (R&D Systems) at a final concentration of 50 ng/mL for the experiments described in Figure4.

Fig. 1.

RARs are dispensable for granulopoiesis.

(A) Fluorescence-activated cell sorter (FACS) profile of WT and RARα−/−RARγ−/− double mutant E14.5 FL cells stained for GR-1 and Mac-1 expression. (B) RT-PCR analysis of granulocyte marker expression in E14.5 FL from WT or RARα−/−RARγ−/− mice. (C) May-Grünwald Giemsa staining of cells from granulocyte-macrophage colonies derived from E14.5 WT and RARα−/−RARγ−/− FL cells showing morphologically normal neutrophils and macrophages. (D) Absence of RARβ expression in FL or pooled colonies (C) derived from WT or RARα−/−RARγ−/− mutants. cDNA from E8.5 embryos (E) was used as a positive control.

Fig. 1.

RARs are dispensable for granulopoiesis.

(A) Fluorescence-activated cell sorter (FACS) profile of WT and RARα−/−RARγ−/− double mutant E14.5 FL cells stained for GR-1 and Mac-1 expression. (B) RT-PCR analysis of granulocyte marker expression in E14.5 FL from WT or RARα−/−RARγ−/− mice. (C) May-Grünwald Giemsa staining of cells from granulocyte-macrophage colonies derived from E14.5 WT and RARα−/−RARγ−/− FL cells showing morphologically normal neutrophils and macrophages. (D) Absence of RARβ expression in FL or pooled colonies (C) derived from WT or RARα−/−RARγ−/− mutants. cDNA from E8.5 embryos (E) was used as a positive control.

Close modal
Fig. 2.

RARα−/− myeloid progenitors are resistant to the inhibitory effects of 9C-RA.

(A) Numbers of colonies developing from 5 × 104 BM cells from WT or RARα−/− mice in the presence or absence of 10−6 M 9C-RA. Each bar represents the average of duplicate samples from 3 mice. The cytokines used to stimulate growth of hematopoietic progenitors were SCF, IL-6, IL-3, and EPO. (B) Representative colonies (day 7) showing the reduction in size of WT granulocyte colonies grown in the presence of 9C-RA and the lack of such an effect in RARα−/− mutants.

Fig. 2.

RARα−/− myeloid progenitors are resistant to the inhibitory effects of 9C-RA.

(A) Numbers of colonies developing from 5 × 104 BM cells from WT or RARα−/− mice in the presence or absence of 10−6 M 9C-RA. Each bar represents the average of duplicate samples from 3 mice. The cytokines used to stimulate growth of hematopoietic progenitors were SCF, IL-6, IL-3, and EPO. (B) Representative colonies (day 7) showing the reduction in size of WT granulocyte colonies grown in the presence of 9C-RA and the lack of such an effect in RARα−/− mutants.

Close modal

Antibodies and flow cytometry

The following antibodies were used: fluorescein isothiocyanate (FITC)–labeled GR-1 (Caltag, South San Francisco, CA) or phycoerythrin-labeled GR-1 (Pharmingen, San Diego, CA) and biotin-labeled Mac-1 (CD11b, Pharmingen). Streptavidin-Cy5 was purchased from Jackson Immunoresearch Laboratories, West Grove, PA. Antibody stainings were performed on 106 BM or FL cells and analyzed (Coulter Elite cytometer; Coulter Electronics, Hialeah, FL). Listmode data were collected on 3 × 104 cells.

Reverse transcriptase–PCR

Reverse transcriptase (RT)–PCR reactions were performed on 50 ng complementary DNA (cDNA) using the following oligonucleotides: cathepsin G: 5′-GATTGTAATCAGGATGGCGG-3′, 5′-CTGACTAAGCAACGGTTCTGG-3′; myeloperoxidase: 5′-ATGCAGTGGGGACAGTTTCTG-3′, 5′-GTCGTTGTAGGATCGGTACTG-3′; elastase: 5′-CACCATCAGTCAGGTCTTCC-3′, 5′-AGTCTCCGAAGCATATGCC-3′; β-actin: 5′-GTGACGAGGCCCAGAGCAAGAG-3′, 5′-AGGGGCCGGACTCATCGTACTC-3′; RARβ: 5′-CTCGTCCCGAGCCCACCATCTCCACTT-3′, 5′-GAGGTCGTCTAGCTCCGCTGTCATCTC-3′; and hypoxanthine phosphoribosyltransferase (HPRT): 5′-GTAATGATCAGTCAACG-3′, 5′-CCAGCAAGCTTGCAACCTTAACCA-3′. RARβ and HPRT amplification products (Figure 1D) were detected after Southern blot analysis and hybridization with phosphorous 32 (32P)-labeled oligonucleotide probes (5′-CCATAGTGGTAGCC-3′ and 5′-CACTGGTAAAACAATGCAAA-3′, respectively).

RARs are dispensable for granulopoiesis

Granulocyte populations appeared normal in RARα−/− mice at the level of blood neutrophil counts (data not shown) and in the BM (GR-1+Mac-1+population; Figure 5C, upper right panel, and data not shown). To investigate whether functional compensation by other RARs occurs in RARα−/− mutants, we examined RARα−/−RARγ−/− double mutants, as RARγ is also expressed in granulocytes.17 Because RARα−/−RARγ−/− mutants die in utero,28 granulopoiesis was studied in the FL. Granulocytes developed normally in RARα−/−RARγ−/− E14.5 FLs as tested by several criteria: double-mutant myeloid cells exhibited a normal GR-1+Mac-1+ phenotype (Figure 1A); differentiated cells (containing ring-shaped or segmented nuclei) were detected at similar frequencies on cytospin preparations from mutant and WT FLs (data not shown); granulocyte-specific markers were expressed at similar levels (Figure 1B); and morphologically normal neutrophils were generated in myeloid colonies in vitro (Figure 1C). Because RARβ expression was not detected in freshly isolated or cultured FL cells (Figure 1D), granulopoiesis can thus proceed efficiently in the absence of any RAR.

To determine whether granulocyte progenitors from RARα−/− mice could still respond to retinoids, we compared the response of WT and RARα−/− BM cells to 9-cis retinoic acid (9C-RA, a pan-RAR and pan-RXR [retinoid X receptor] agonist) in a myeloid colony-forming assay. As previously reported,29 the total number of colonies developing from WT BM cells, as well as the size of pure granulocyte colonies, were markedly reduced in the presence of 9C-RA (Figure 2A,B). Neither of these effects were observed in cultures of RARα−/− BM cells (Figure 2) (P < .01), indicating that RARα−/− granulocyte progenitors are impaired in their response to retinoids. These results, together with the apparent normal granulopoiesis in RARα−/−RARγ−/− mutants, demonstrate that RARs and retinoids are dispensable for granulopoiesis.

Accelerated granulocyte differentiation in the absence of RARα

Although the RA/RAR pathway is not required, it can clearly modulate granulocyte differentiation, as shown by the response of WT cells to retinoids. Consequently, we analyzed the effects of RAR agonists and antagonists on granulocyte differentiation in liquid cultures of WT BM cells stimulated with G-CSF and SCF. To monitor cells that had undergone division during the culture period, freshly isolated BM cells were cytoplasmically labeled with the vital dye CFSE (a fluorescent compound that is gradually diluted upon cell division) prior to culture, and analysis was confined to CFSE−/lowcells (Figure 3A). Increased levels of GR-1 and Mac-1 expression were used to measure differentiation.30 9C-RA induced an increase in the proportion of de novo GR-1high cells in cultures of WT cells (Figure 3B, upper right panel, gate C, and Figure 3C) as well as a decrease in the proportion of CFSE−/low cells (data not shown), which is consistent with the premise that RA limits the proliferation of granulocyte precursors and induces their differentiation.

Fig. 3.

Accelerated differentiation of RARα−/− granulocytes in liquid cultures stimulated with G-CSF and SCF.

Cultures of CFSE-labeled BM cells were harvested 3 days after their initiation and stained for Mac-1 and GR-1 expression. The RAR antagonist BMS493 or the RXR/RAR agonist 9C-RA was added at the onset of the culture at a final concentration of 10−6 M, where indicated. (A) The analysis was limited to CFSElow cells, which correspond to cells that have arisen during the culture from proliferation of immature granulocytes. Note that CFSEhighcells correspond exclusively to mature granulocytes with a Mac-1highGR-1high phenotype (data not shown). (B) Representative GR-1/Mac-1 profiles of CFSElow cells. Note the shift toward the more immature phenotype in the BMS493-treated WT sample and the similarity of the phenotypes of all RARα−/− samples with that of the 9C-RA–treated WT sample. (C) Quantification in percent of all cells present in gates B and C of immature (gate B in panel A) and mature (gate C in panel A) cells from the experiment displayed in panel B. The experiment described in this figure was performed in duplicate for each of the 2 WT and 2 RARα−/− mice. For a given set of culture conditions and mouse genotype, variations in the percentage of the gated populations were within 1.5% of those indicated.

Fig. 3.

Accelerated differentiation of RARα−/− granulocytes in liquid cultures stimulated with G-CSF and SCF.

Cultures of CFSE-labeled BM cells were harvested 3 days after their initiation and stained for Mac-1 and GR-1 expression. The RAR antagonist BMS493 or the RXR/RAR agonist 9C-RA was added at the onset of the culture at a final concentration of 10−6 M, where indicated. (A) The analysis was limited to CFSElow cells, which correspond to cells that have arisen during the culture from proliferation of immature granulocytes. Note that CFSEhighcells correspond exclusively to mature granulocytes with a Mac-1highGR-1high phenotype (data not shown). (B) Representative GR-1/Mac-1 profiles of CFSElow cells. Note the shift toward the more immature phenotype in the BMS493-treated WT sample and the similarity of the phenotypes of all RARα−/− samples with that of the 9C-RA–treated WT sample. (C) Quantification in percent of all cells present in gates B and C of immature (gate B in panel A) and mature (gate C in panel A) cells from the experiment displayed in panel B. The experiment described in this figure was performed in duplicate for each of the 2 WT and 2 RARα−/− mice. For a given set of culture conditions and mouse genotype, variations in the percentage of the gated populations were within 1.5% of those indicated.

Close modal

In contrast, the pan-RAR antagonist BMS493 (H. Gronemeyer and P.C., unpublished observation, February 1996) induced an increase in the proportion of immature granulocytes (Figure 3B, upper middle panel, gate B, and Figure 3C), a finding corroborated by the higher frequency of promyelocytes/myelocytes on cytospin preparations from these cultures (data not shown). Interestingly, the magnitude of the response to BMS493 greatly exceeded that to 9C-RA, suggesting that retinoids in the serum might play an important part in controlling the differentiation that occurs in the absence of exogenous ligands. When RARα−/− BM cells were grown under identical culture conditions, the ratio of immature versus mature cells generated was unaffected by treatment with either 9C-RA or BMS493 (Figure 3B,C), confirming the resistance of RARα−/− granulocytes to the effects of RAR ligands. In addition, the proportions of mature cells were consistently increased when compared with WT samples in the control and BMS493-treated cultures to levels usually observed in 9C-RA–treated WT cultures (Figure 3B,C). Thus granulocyte differentiation appears to be accelerated in the absence of RARα.

This apparent acceleration of maturation was also seen in clonogenic methylcellulose cultures stimulated with GM-CSF (Figure 4). After 5 days of culture, granulocytes from pooled RARα−/−colonies were more mature than those from WT colonies (compare Figure4A with B and inset). By day 8, there was a marked underrepresentation of granulocytes in the mutant cultures, which now consisted mainly of macrophages (Figure 4C,D), suggesting that the prematurely differentiated neutrophils seen on day 5 of culture had died off over time. At the colony level, dense granulocyte clusters were less abundant and smaller in RARα−/− cultures after 8 days (Figure 4E,F), a phenotype that reliably allowed for the blind separation of dishes containing WT or RARα−/− cells. It is noteworthy that both types of experimental cultures used different cytokines (G-CSF and SCF for the liquid cultures and GM-CSF for the methylcellulose cultures), thus excluding a cytokine-specific effect. These observations provide evidence that lack of RARα relieves a physiological restraint on granulocyte differentiation normally exerted by this receptor in the absence of ligand.

Fig. 4.

Accelerated differentiation of RARα−/− granulocytes in GM-CSF–dependent clonogenic cultures.

We plated 5 × 104 BM cells in methylcellulose medium containing 50 ng/mL GM-CSF. (A-D) Pooled cells from all the colonies developing on a dish (about 70 colonies) were cytocentrifuged onto a glass slide and stained with May-Grünwald-Giemsa. Arrowheads in panel A indicate promyelocytes that are numerous on slides from day-5 WT cultures, but are less frequent in mutant samples. The inset in panels A and B corresponds to the scoring of approximately 400 cells of the granulocyte lineage for each case. (E,F) Typical colonies from culture dishes at day 8; note the reduction in the number and size of dense cellular aggregates (arrows), which correspond to granulocytes.

Fig. 4.

Accelerated differentiation of RARα−/− granulocytes in GM-CSF–dependent clonogenic cultures.

We plated 5 × 104 BM cells in methylcellulose medium containing 50 ng/mL GM-CSF. (A-D) Pooled cells from all the colonies developing on a dish (about 70 colonies) were cytocentrifuged onto a glass slide and stained with May-Grünwald-Giemsa. Arrowheads in panel A indicate promyelocytes that are numerous on slides from day-5 WT cultures, but are less frequent in mutant samples. The inset in panels A and B corresponds to the scoring of approximately 400 cells of the granulocyte lineage for each case. (E,F) Typical colonies from culture dishes at day 8; note the reduction in the number and size of dense cellular aggregates (arrows), which correspond to granulocytes.

Close modal

Retinoids control granulopoiesis in vivo

To understand how retinoids may modulate granulopoiesis in vivo, we evaluated granulocyte differentiation in WT mice treated with BMS493. Of 11 treated WT mice, 7 mice exhibited a clear increase in the proportion of immature granulocytes in the BM (Mac-1low/+GR-1−/low, Figure 5A). As this increase was not seen in vehicle-treated or untreated animals, its occurrence in BMS493-treated mice most likely reflects a role for endogenous retinoids in regulating granulocyte differentiation. To investigate further, we analyzed VAD mice using CRBPI-deficient mice that, owing to low vitamin A stores, become readily VAD when starved for vitamin A.27 In all VAD CRBPI−/− mice tested (n = 6), there was a marked expansion in the size of the entire granulocyte population (Figure 5B, upper right panel) when compared with control animals (ie, CRBPI−/− mice on a normal diet or CRBPI+/+ mice on a VAD diet) as well as an increase in the proportion of immature Mac-1low/+GR-1−/low cells (Figure 5B, lower right panel). This effect was accompanied by an increase of neutrophils in the spleen and blood (data not shown), which is consistent with a previous report showing enhanced neutrophil numbers in VAD rats.31 Importantly, the VAD-induced increase in the immature granulocyte compartment of the BM mirrors that seen following BMS493 administration (Figure 5A), underscoring a role for endogenous retinoids in the positive control of granulopoiesis in vivo.

Fig. 5.

Effect of retinoid excess and VAD on granulopoiesis in vivo.

BM granulocyte populations were analyzed by flow cytometry with the Mac-1 and GR-1 markers. Granulocyte differentiation proceeds along a curve-shaped path in which increasing levels of Mac-1 and GR-1 are acquired as the cells mature. Thus, Mac-1low/+GR-1low cells correspond mainly to promyelocytes, myelocytes, and metamyelocytes, whereas Mac-1+GR-1high cells correspond mainly to band and segmented neutrophils30 (data not shown). (A) WT mice were treated for 3 days with 10 mg/kg BMS493 or vehicle. In the right panels the GR-1 profile of the granulocyte population (Mac-1+ cells, gated in the left panels) is shown, and the percentage of GR-1low immature granulocytes within that population is indicated. (B) Granulocyte populations in VAD mice. Note the overall increase of the granulocyte population (gated in the top panels) and the increase, within that population, of the immature granulocyte population (GR-1low cells, bottom panels) that occurs specifically in the VAD mice (ie, CRBPI−/− mice fed a VAD diet). (C) Effect of ATRA administration. Mice were treated with 100 mg/kg ATRA or vehicle for 72 hours. Note the shift to the right of GR-1 expression within the immature granulocyte population (gated) in ATRA-treated WT mice, producing a comet-like profile, which is not seen in the ATRA-treated RARα−/− mice. Note that a reduction in the number of the mature (GR-1high) BM granulocytes was also observed in ATRA-treated WT mice, which probably reflects an enhanced efflux of neutrophils from the BM, as peripheral blood neutrophil numbers were increased 1.5- to 3-fold (data not shown).

Fig. 5.

Effect of retinoid excess and VAD on granulopoiesis in vivo.

BM granulocyte populations were analyzed by flow cytometry with the Mac-1 and GR-1 markers. Granulocyte differentiation proceeds along a curve-shaped path in which increasing levels of Mac-1 and GR-1 are acquired as the cells mature. Thus, Mac-1low/+GR-1low cells correspond mainly to promyelocytes, myelocytes, and metamyelocytes, whereas Mac-1+GR-1high cells correspond mainly to band and segmented neutrophils30 (data not shown). (A) WT mice were treated for 3 days with 10 mg/kg BMS493 or vehicle. In the right panels the GR-1 profile of the granulocyte population (Mac-1+ cells, gated in the left panels) is shown, and the percentage of GR-1low immature granulocytes within that population is indicated. (B) Granulocyte populations in VAD mice. Note the overall increase of the granulocyte population (gated in the top panels) and the increase, within that population, of the immature granulocyte population (GR-1low cells, bottom panels) that occurs specifically in the VAD mice (ie, CRBPI−/− mice fed a VAD diet). (C) Effect of ATRA administration. Mice were treated with 100 mg/kg ATRA or vehicle for 72 hours. Note the shift to the right of GR-1 expression within the immature granulocyte population (gated) in ATRA-treated WT mice, producing a comet-like profile, which is not seen in the ATRA-treated RARα−/− mice. Note that a reduction in the number of the mature (GR-1high) BM granulocytes was also observed in ATRA-treated WT mice, which probably reflects an enhanced efflux of neutrophils from the BM, as peripheral blood neutrophil numbers were increased 1.5- to 3-fold (data not shown).

Close modal

In contrast, ATRA treatment of WT mice (n = 7) elicited an increase in the levels of GR-1 and Mac-1 expressed by the Mac-1low/+GR-1−/low immature granulocyte population (Figure 5C, gated region, compare upper and lower left panels), an effect that did not occur in RARα−/− mice (n = 4) (Figure 5C, right panels). This indicates that RARα is the direct mediator of exogenous (and, by inference, endogenous) retinoid-induced differentiation. Significantly, the fact that all 7 ATRA-treated WT mice responded in a similar manner also suggests that under normal dietary conditions, RARα proteins are not fully liganded in vivo, thus placing them in an appropriate state to negatively regulate differentiation.

RARα: a bidirectional modulator of granulopoiesis

We have shown here that (1) endogenous RARα is a key mediator of the effects of retinoids on granulopoiesis, and (2) the RARα receptor can bidirectionally modulate granulopoiesis as a differentiation factor when liganded to RA or as an inhibitor in the absence of ligand (or when liganded to an antagonist), probably by acting as a transcriptional repressor.32-34 The fact that RA is a differentiating agent for granulocytes in culture has been well established for both normal and leukemic cells.19,20,35However, the role of retinoids during normal granulocyte differentiation in vivo has so far not been documented. The increase in the immature granulocyte populations, seen both in the VAD and BMS493-treated mice here, show for the first time that retinoid-controlled differentiation actively contributes to the regulation of granulocyte numbers in vivo. Our VAD results are in agreement with those of Kuwata et al,36 who recently reported a collective increase in granulocyte numbers during murine VAD as well as with an earlier report documenting elevated neutrophils in the peripheral blood of VAD rats.31 Although Kuwata et al36 did not study the relative contributions of immature versus mature cells in their granulocyte population, their VAD mice also seemed to accumulate more cells with a GR-1lowphenotype, as seen in the Kuwata et al Figure 3A,36 and may therefore exhibit a phenotype similar to that of our VAD mice. We have noted that optimal resolution of the GR-1low and GR-1high populations appears to depend on the fluorochrome to which the anti–GR-1 antibody is conjugated; FITC-conjugated anti–GR-1 (as used in our study) provided the best resolution, while other conjugated forms of this antibody appeared to give more compact stainings similar to those shown by Kuwata et al.36 These authors also found less apoptosis within the granulocyte population of their VAD mice, an observation that they used to explain their increase in granulocyte numbers. Although we cannot exclude that retinoids directly regulate cell survival, it is possible that the decrease in apoptosis seen by Kuwata and colleagues is an indirect consequence of a shift toward the more immature stages because apoptosis is the ultimate endpoint of differentiation.

The finding that endogenous unliganded RARα negatively regulates differentiation is consistent with results from previous studies showing that overexpression of WT RARα blocks promyelocyte differentiation in vitro.22 23 However, the non-physiological levels of receptor expressed in these systems have prevented any clear conclusion about the role of the endogenous molecule. In fact, these latter studies often claimed that the overexpressed receptors functioned in a dominant-negative manner by titrating out retinoids present in the medium, thereby countering the differentiating role of these compounds. Our present results suggest otherwise; the differentiation block exerted by overexpressed RARα corresponds to an amplification of an intrinsic inhibitory activity of the unliganded receptor. It is important to stress that in contrast to the overexpression studies, blocking the activation of the endogenous receptor in vivo or in vitro (with VAD or BMS493) never resulted in a complete arrest in differentiation such as the arrest achieved by overexpressed receptors, thereby reflecting the possibility that RARα is expressed in limiting amounts.

Intriguingly, our results appear to contradict those of Labrecque et al,17 who reported that granulocytes from myeloid colonies derived from RARα1−/−RARγ−/−neonatal BM cells tend to be more immature than those derived from WT cells (RARα1 being one of the 2 major isoforms produced from the RARα gene).37 As we never observed such a block in colonies derived from RARα−/−RARγ−/− FL cells, the apparent difference in the behavior of the RARα1−/−RARγ−/− cells might be due to the persistent expression of RARα2, an isoform not present in our double mutants. It should be noted that we used FL cells in our study, while Labrecque and colleagues17 used BM cells. However, we observed a similar premature differentiation in GM-CSF–dependent colonies derived from either FL or adult BM RARα−/−cells (unpublished observations, February 1998). Thus, it appears unlikely that RARs control granulopoiesis differently in FL and BM cells. With respect to RARγ, its role appears to be minor at best, as cells lacking only RARα are already resistant to the effects of retinoids.

The present results demonstrate that granulocyte differentiation can proceed in the absence of RARs. The RA/RARα pathway is therefore not an obligatory regulator of granulopoiesis, but rather a modulator of this differentiation. Both retinoid excess and deficiency affect granulopoiesis, suggesting that a balance between liganded and unliganded receptor molecules exists in vivo under normal conditions. The question therefore arises as to which physiological or pathological conditions modulate retinoid levels. Dietary VAD, a condition that is associated with higher susceptibility to bacterial infections, is an obvious candidate. The negative regulation of granulopoiesis by unliganded RARα (which ultimately translates into an elevated granulocyte pool) might thus represent an innate response used to off-set VAD-associated infections. In this view, an important role of RARα during granulopoiesis could paradoxically be to mediate the effect of ligand deficiency. Analysis of the VAD response in RARα−/− mice should help to clarify this issue.

At the molecular level, several downstream candidates might mediate the actions of RARα. A prime candidate is C-EBPε, an RA-target gene induced by RA in myeloid cell lines38,39 and shown to be essential for terminal granulocyte differentiation.40However, because granulocytes can fully differentiate in the absence of RARs or during retinoid deficiency, it is unlikely that retinoids are the only regulators of C-EBPε expression in vivo. In this respect we have found that overall, C-EBPε transcript levels were not altered in the BM of RARα−/− mice (unpublished data, March 1998). Other interesting candidates are the cdk inhibitors p21 and p27, whose increases have been shown to be associated with the transition between immature, proliferating cells and post-mitotic differentiated cells for several cell types including myeloid cells.41-45Interestingly, p21 has been proposed to be a target gene for nuclear receptors, RARs included.41,46 How these proteins are regulated in cells lacking RARα thus merits further investigation. Hox genes, which are regulated by retinoic acid in the embryo, might also contribute to mediating RAR function during hematopoiesis47; their precise role during granulopoiesis, however, and their possible regulation by RARs remain unknown. Finally, Mad1 has been shown to positively regulate cell cycle exit during granulopoiesis, possibly by antagonizing the effects of myc proteins.48 Whether the Myc/Mad and the RAR pathways are parallel or convergent modulators of granulopoiesis remains to be seen.

The role of retinoid signaling during hematopoiesis is likely to extend beyond its role of regulating granulocyte differentiation described here. RA has also been suggested to act as a commitment factor for the granulocyte lineage, as exogenous RA is able to redirect erythroid-, monocyte-, or eosinophil-committed progenitors toward a granulocyte fate.24,49 Although our study does not directly address this issue, it is clear that other factors besides RA can influence granulocyte lineage commitment, as commitment was not impaired in RARα−/−RARγ−/− double mutants or upon VAD. However, the proportion of granulocytes was often reduced in myeloid colonies derived from RARα−/− BM cells, and the frequency of granulocyte-only colonies was diminished (unpublished data, October 1998), phenotypes that could indicate defective granulocyte commitment in the mutants. The present effects of RAR agonists and antagonists during terminal granulocyte differentiation also contrast with those observed when similar compounds were used on hematopoietic stem cells, where RAR agonists and antagonists antagonized and promoted their differentiation, respectively.50 These differences suggest a complex pattern of retinoid receptor function during hematopoiesis.

Implications for APL pathogenesis

RARα chromosomal rearrangements represent the genetic hallmark of APL. The tight association between RARα alterations and APL strongly suggests that a normal RARα-dependent function must be altered to transform promyelocytes. Generally, it has been assumed that the APL RARα fusion proteins act as dominant-negative inhibitors of retinoid-dependent differentiation.51,52 However, because granulocyte differentiation is only partially blocked when retinoid signaling through RAR is prevented, dominant-negative inhibition of retinoid-induced differentiation cannot account for the complete differentiation block seen in APL cells. Our data suggest an alternative mechanism of action for the RAR fusion proteins: enhancement of the normal inhibitory function of unliganded RARα. This enhancement may be due to an overexpression of the fusion proteins vis-à-vis WT RARα, as is often observed in APL cells,53,54 and/or to the more potent repressive properties of the fusion proteins themselves. In this respect, PML-RARα and PLZF-RARα are powerful transcriptional repressors that efficiently recruit histone deacetylases to target promoters.9,12-15 Interestingly, overexpression of thyroid hormone receptor α (TRα) in erythroid precursors blocks their differentiation,55 suggesting that the differentiation arrest mediated by v-erbA, the oncogenic form of TRα, during erythropoiesis might also correspond to an enhancement of a normal property of TRα. Different nuclear receptors may therefore control differentiation and leukemogenesis in distinct hematopoietic lineages via similar mechanisms.

If interference with RARα-regulated differentiation appears important for APL pathogenesis, it probably represents only one facet of the action of RARα fusion proteins in APL. Fusion of RARα with PML or PLZF has been shown to be required to provoke APL.11Dominant interference with other functions, such as those ofPML (a tumor suppressor gene that controls apoptosis and p53 acetylation), is likely to be important.1,4,56-58 In this respect the coiled coil moiety of PML appears to mediate the formation of high molecular weight oligomeric complexes containing both PML-RARα and PML, complexes which could disrupt normal PML function.59 Simultaneous targeting of several cellular functions by the RARα fusion proteins might therefore cooperate to transform promyelocytes.

We thank P. Reczek (Bristol-Myers Squibb, Princeton, NJ) for providing BMS493 and M. Sporn for 9cis-RA; C. Bronn, B. Bondeau, and B. Feret for excellent technical assistance; C. Ebel for help with flow cytometry; and A. Pailleux, F. Fischer, and P. Michel for maintaining the mice.

Supported by grants from the Association pour la Recherche sur le Cancers; the Centre National pour la Recherche Scientifique; the Institut National de la Santé et de la Recherche Médicale; the Hôpital Universitaire de Strasbourg; the Collège de France, Illkirch Cedex, France; and Bristol-Myers Squibb, Princeton, NJ. H.J.L. is the recipient of a VA Career Development Award. BMS493 was provided by Bristol-Myers Squibb.

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
Melnick
A
Licht
JD
Deconstructing a disease: RARalpha, its fusion partners, and their roles in the pathogenesis of acute promyelocytic leukemia.
Blood.
93
1999
3167
3215
2
Lin
RJ
Egan
DA
Evans
RM
Molecular genetics of acute promyelocytic leukemia.
Trends Genet.
15
1999
179
184
3
Kogan
SC
Bishop
JM
Acute promyelocytic leukemia: from treatment to genetics and back.
Oncogene.
18
1999
5261
5267
4
Zhong
S
Salomoni
P
Pandolfi
PP
The transcriptional role of PML and the nuclear body.
Nat Cell Biol.
2
2000
E85
E90
5
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
6
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
7
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
8
Arnould
C
Philippe
C
Bourdon
V
Gregoire
MJ
Berger
R
Jonveaux
P
The signal transducer and activator of transcription STAT5b gene is a new partner of retinoic acid receptor alpha in acute promyelocytic-like leukaemia.
Hum Mol Genet.
8
1999
1741
1749
9
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
10
Cheng
GX
Zhu
XH
Men
XQ
et al
Distinct leukemia phenotypes in transgenic mice and different corepressor interactions generated by promyelocytic leukemia variant fusion genes PLZF-RARalpha and NPM-RARalpha.
Proc Natl Acad Sci U S A.
96
1999
6318
6323
11
Kogan
SC
Hong
SH
Shultz
DB
Privalsky
ML
Bishop
JM
Leukemia initiated by PMLRARalpha: the PML domain plays a critical role while retinoic acid-mediated transactivation is dispensable.
Blood.
95
2000
1541
1550
12
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
13
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
14
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
15
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
16
Zelent
A
Zhu
J
Lanotte
M
et al
Differential expression of retinoid receptors during multilineage differentiation of haematopoietic progenitor cells: role of the RARα2 isoform in normal granulopoiesis and leukemia.
Blood.
90
1997
44a
17
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
18
Largman
C
Detmer
K
Corral
JC
Hack
FM
Lawrence
HJ
Expression of retinoic acid receptor alpha mRNA in human leukemia cells.
Blood.
74
1989
99
102
19
Breitman
TR
Selonick
SE
Collins
SJ
Induction of differentiation of the human promyelocytic leukemia cell line (HL-60) by retinoic acid.
Proc Natl Acad Sci U S A.
77
1980
2936
2940
20
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
21
Tsai
S
Collins
SJ
A dominant negative retinoic acid receptor blocks neutrophil differentiation at the promyelocyte stage.
Proc Natl Acad Sci U S A.
90
1993
7153
7157
22
Onodera
M
Kunisada
T
Nishikawa
S
Sakiyama
Y
Matsumoto
S
Overexpression of retinoic acid receptor alpha suppresses myeloid cell differentiation at the promyelocyte stage.
Oncogene.
11
1995
1291
1298
23
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
24
Tocci
A
Parolini
I
Gabbianelli
M
et al
Dual action of retinoic acid on human embryonic/fetal hematopoiesis: blockade of primitive progenitor proliferation and shift from multipotent/erythroid/monocytic to granulocytic differentiation program.
Blood.
88
1996
2878
2888
25
Lufkin
T
Lohnes
D
Mark
M
et al
High postnatal lethality and testis degeneration in retinoic acid receptor alpha mutant mice.
Proc Natl Acad Sci U S A.
90
1993
7225
7229
26
Lohnes
D
Kastner
P
Dierich
A
Mark
M
LeMeur
M
Chambon
P
Function of retinoic acid receptor gamma in the mouse.
Cell.
73
1993
643
658
27
Ghyselinck
NB
Bavik
C
Sapin
V
et al
Cellular retinol-binding protein I is essential for vitamin A homeostasis.
EMBO J.
18
1999
4903
4914
28
Lohnes
D
Mark
M
Mendelsohn
C
et al
Function of the retinoic acid receptors (RARs) during development (I): craniofacial and skeletal abnormalities in RAR double mutants.
Development.
120
1994
2723
2748
29
Jacobsen
SE
Fahlman
C
Blomhoff
HK
Okkenhaug
C
Rusten
LS
Smeland
EB
All-trans- and 9-cis-retinoic acid: potent direct inhibitors of primitive murine hematopoietic progenitors in vitro.
J Exp Med.
179
1994
1665
1670
30
Hestdal
K
Ruscetti
FW
Ihle
JN
et al
Characterization and regulation of RB6–8C5 antigen expression on murine bone marrow cells.
J Immunol.
147
1991
22
28
31
Nauss
KM
Mark
DA
Suskind
RM
The effect of vitamin A deficiency on the in vitro cellular immune response of rats.
J Nutrit.
109
1979
1815
1823
32
Nagy
L
Kao
HY
Chakravarti
D
et al
Nuclear receptor repression mediated by a complex containing SMRT, mSin3A, and histone deacetylase.
Cell.
89
1997
373
380
33
Chen
JD
Evans
RM
A transcriptional co-repressor that interacts with nuclear hormone receptors.
Nature.
377
1995
454
457
34
Horlein
AJ
Naar
AM
Heinzel
T
et al
Ligand-independent repression by the thyroid hormone receptor mediated by a nuclear receptor co-repressor.
Nature.
377
1995
397
404
35
Lawson
ND
Berliner
N
Neutrophil maturation and the role of retinoic acid.
Exp Hematol.
27
1999
1355
1367
36
Kuwata
T
Wang
IM
Tamura
T
et al
Vitamin A deficiency in mice causes a systemic expansion of myeloid cells.
Blood.
95
2000
3349
3356
37
Leroy
P
Krust
A
Zelent
A
et al
Multiple isoforms of the mouse retinoic acid receptor alpha are generated by alternative splicing and differential induction by retinoic acid.
EMBO J.
10
1991
59
69
38
Chih
DY
Chumakov
AM
Park
DJ
Silla
AG
Koeffler
HP
Modulation of mRNA expression of a novel human myeloid-selective CCAAT/enhancer binding protein gene (C/EBP epsilon).
Blood.
90
1997
2987
2994
39
Park
DJ
Chumakov
AM
Vuong
PT
et al
CCAAT/enhancer binding protein epsilon is a potential retinoid target gene in acute promyelocytic leukemia treatment.
J Clin Invest.
103
1999
1399
1408
40
Yamanaka
R
Barlow
C
Lekstrom-Himes
J
et al
Impaired granulopoiesis, myelodysplasia, and early lethality in CCAAT/enhancer binding protein epsilon-deficient mice.
Proc Natl Acad Sci U S A.
94
1997
13187
13192
41
Liu
M
Lee
MH
Cohen
M
Bommakanti
M
Freedman
LP
Transcriptional activation of the Cdk inhibitor p21 by vitamin D3 leads to the induced differentiation of the myelomonocytic cell line U937.
Genes Dev.
10
1996
142
153
42
Nakayama
K
Cip/Kip cyclin-dependent kinase inhibitors: brakes of the cell cycle engine during development.
Bioessays.
20
1998
1020
1029
43
Taniguchi
T
Endo
H
Chikatsu
N
et al
Expression of p21(Cip1/Waf1/Sdi1) and p27 (Kip1) cyclin-dependent kinase inhibitors during human hematopoiesis.
Blood.
93
1999
4167
4178
44
Muto
A
Kizaki
M
Yamato
K
et al
1,25-Dihydroxyvitamin D3 induces differentiation of a retinoic acid-resistant acute promyelocytic leukemia cell line (UF-1) associated with expression of p21(WAF1/CIP1) and p27(KIP1).
Blood.
93
1999
2225
2233
45
Yaroslavskiy
B
Watkins
S
Donnenberg
AD
Patton
TJ
Steinman
RA
Subcellular and cell-cycle expression profiles of CDK-inhibitors in normal differentiating myeloid cells.
Blood.
93
1999
2907
2917
46
Liu
M
Lavarone
A
Freedman
LP
Transcriptional activation of the human p21(WAF1/CIP1) gene by retinoic acid receptor: correlation with retinoid induction of U937 cell differentiation.
J Biol Chem.
271
1996
31723
31728
47
Lawrence
HJ
Sauvageau
G
Humphries
RK
Largman
C
The role of HOX homeobox genes in normal and leukemic hematopoiesis.
Stem Cells.
14
1996
281
291
48
Foley
KP
McArthur
GA
Queva
C
Hurlin
PJ
Soriano
P
Eisenman
RN
Targeted disruption of the MYC antagonist MAD1 inhibits cell cycle exit during granulocyte differentiation.
EMBO J.
17
1998
774
785
49
Paul
CC
Mahrer
S
Tolbert
M
et al
Changing the differentiation program of hematopoietic cells: retinoic acid-induced shift of eosinophil-committed cells to neutrophils.
Blood.
86
1995
3737
3744
50
Purton
LE
Bernstein
ID
Collins
SJ
All-trans retinoic acid enhances the long-term repopulating activity of cultured hematopoietic stem cells.
Blood.
95
2000
470
477
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
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
53
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
54
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
55
Bauer
A
Mikulits
W
Lagger
G
Stengl
G
Brosch
G
Beug
H
The thyroid hormone receptor functions as a ligand-operated developmental switch between proliferation and differentiation of erythroid progenitors.
EMBO J.
17
1998
4291
4303
56
Wang
ZG
Ruggero
D
Ronchetti
S
et al
PML is essential for multiple apoptotic pathways.
Nat Genet.
20
1998
266
272
57
Quignon
F
De Bels
F
Koken
M
Feunteun
J
Ameisen
JC
de The
H
PML induces a novel caspase-independent death process.
Nat Genet.
20
1998
259
265
58
Pearson
M
Carbone
R
Sebastiani
C
et al
PML regulates p53 acetylation and premature senescence induced by oncogenic Ras.
Nature.
406
2000
207
210
59
Minucci
S
Maccarana
M
Cioce
M
et al
Oligomerization of RAR and AML1 transcription factors as a novel mechanism of oncogenic activation.
Mol Cell.
5
2000
811
820

Author notes

Philippe Kastner and Susan Chan, Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS-INSERM-ULP-Collège de France, BP163, 67404 Illkirch Cedex, France; e-mail: scpk@igbmc.u-strasbg.fr.

Sign in via your Institution