8
©2012 Landes Bioscience. Do not distribute. www.landesbioscience.com Cell Cycle 3219 Cell Cycle 11:17, 3219-3226; September 1, 2012; © 2012 Landes Bioscience REPORT REPORT *Correspondence to: Gesine Bug; Email: [email protected] Submitted: 02/21/12; Revised: 07/19/12; Accepted: 07/20/12 http://dx.doi.org/10.4161/cc.21565 Introduction Acute myeloid leukemia (AML) is an aggressive malignant disor- der affecting mostly older patients. In spite of intensive treatment, the majority of AML patients relapse and die of their disease. As residual leukemic stem and progenitor cells (LSC) are a potential reservoir for AML relapse, their response has to be taken into account when evaluating the efficacy of novel therapies. LSC are functionally defined by their capacity to initiate the disease upon inoculation into irradiated mice due to their extensive prolifera- tion and self-renewal capacity. 1,2 Thus, targeting of transplant- able LSC while sparing normal hematopoietic stem cell function is a matter of intensive research. Deacetylase inhibitors (DACi) are promising drugs that lead to growth inhibition, cell cycle arrest, premature senescence and apoptosis of malignant cells. 3 The underlying molecular Acute myeloid leukemia (AML) is a highly malignant disease that is not curable in the majority of patients. Numerous non-random genetic abnormalities are known, among which several translocations such as PLZF/RARα or AML1/ETO are known to aberrantly recruit histone deacetylases. Deacetylase inhibitors (DACi) are promising drugs leading to growth inhibition, cell cycle arrest, premature senescence and apoptosis in malignant cells. It is believed that DACi may have clinical efficacy by eradicating the most primitive population of leukemic stem and progenitor cells, possibly by interfering with self-renewal. The aim of the study was to investigate the effects of DACi on leukemic stem and progenitor cells using murine trans- duction-transplantation models of hematopoietic cells harboring the leukemia-associated fusion proteins (LAFP) PLZF/ RARα or a truncated AML1/ETO protein (AML1/ETO exon 9). We show that the self-renewal and short-term repopulation capacity of AML1/ETO- or PLZF/RARα-expressing Sca1 + /lin - stem and progenitor cells are profoundly inhibited by clini- cally applicable concentrations of the DACi dacinostat and vorinostat. To further investigate the mechanisms underlying these effects, we examined the impact of DACi on the transcription factor c-MYC and the Polycomb group protein BMI1, which are induced by LAFP and involved in leukemic transformation. In AML1/ETO or PLZF/RARα-positive 32D cells, DACi- mediated antiproliferative effects were associated with downregulation of BMI1 and c-MYC protein levels. Similar effects were demonstrated in primary samples of cytogenetically defined high-risk AML patients. In conclusion, DACi may be ef- fective as maintenance therapy by negatively interfering with signaling pathways that control survival and proliferation of leukemic stem and progenitor cells. Deacetylase inhibitors modulate proliferation and self-renewal properties of leukemic stem and progenitor cells Annette Romanski, 1,† Kerstin Schwarz, 1,† Maren Keller, 1 Sarah Wietbrauk, 1 Anja Vogel, 1 Jessica Roos, 1 Claudia Oancea, 1 Boris Brill, 2 Oliver H. Krämer, 3 Hubert Serve, 1 Martin Ruthardt 1 and Gesine Bug 1, * 1 Department of Medicine II; Hematology and Oncology; Johann Wolfgang Goethe-University; Frankfurt/Main, Germany; 2 Institute for Biomedical Research Georg Speyer- Haus; Frankfurt/Main, Germany; 3 Institute of Biochemistry and Biophysics; Center for Molecular Biomedicine; Friedrich-Schiller University; Jena, Germany These authors contributed equally to this work. Keywords: acute myeloid leukemia, leukemic stem cells, deacetylase inhibitor, BMI1, self-renewal, short-term repopulation, dacinostat, vorinostat mechanisms include epigenetic reprogramming of the transcrip- tome by alteration of the acetylation status of core histones and thus modification of the chromatin structure. DACi additionally induce selective proteasomal degradation of histone decetylase 2 (HDAC2), DNA methyltransferases and other proteins respon- sible for aberrant gene repression and signaling. 4-6 This effect equally alters the cellular program, but to date, the underlying mechanisms remain elusive and have not yet been studied in the leukemic stem and progenitor compartment. Balanced translocations such as t(8;21) and t(11;15) and the corresponding leukemia-associated fusion proteins (LAFP) AML1/ETO and PLZF/RARα, respectively, are a hallmark of AML. Ectopic expression of PLZF/RARα or a truncated AML1/ ETO protein (AML1/ETO exon 9) in murine hematopoietic stem cells and transplantation into syngeneic mice recapitulates the leukemic phenotype characterized by a differentiation block

Deacetylase inhibitors modulate proliferation and self-renewal properties of leukemic stem and progenitor cells

  • Upload
    gesine

  • View
    214

  • Download
    1

Embed Size (px)

Citation preview

Page 1: Deacetylase inhibitors modulate proliferation and self-renewal properties of leukemic stem and progenitor cells

©20

12 L

ande

s B

iosc

ienc

e. D

o no

t dis

tribu

te.

www.landesbioscience.com Cell Cycle 3219

Cell Cycle 11:17, 3219-3226; September 1, 2012; © 2012 Landes Bioscience

RepoRt RepoRt

*Correspondence to: Gesine Bug; Email: [email protected]: 02/21/12; Revised: 07/19/12; Accepted: 07/20/12http://dx.doi.org/10.4161/cc.21565

Introduction

Acute myeloid leukemia (AML) is an aggressive malignant disor-der affecting mostly older patients. In spite of intensive treatment, the majority of AML patients relapse and die of their disease. As residual leukemic stem and progenitor cells (LSC) are a potential reservoir for AML relapse, their response has to be taken into account when evaluating the efficacy of novel therapies. LSC are functionally defined by their capacity to initiate the disease upon inoculation into irradiated mice due to their extensive prolifera-tion and self-renewal capacity.1,2 Thus, targeting of transplant-able LSC while sparing normal hematopoietic stem cell function is a matter of intensive research.

Deacetylase inhibitors (DACi) are promising drugs that lead to growth inhibition, cell cycle arrest, premature senescence and apoptosis of malignant cells.3 The underlying molecular

Acute myeloid leukemia (AML) is a highly malignant disease that is not curable in the majority of patients. Numerous non-random genetic abnormalities are known, among which several translocations such as pLZF/RARα or AML1/eto are known to aberrantly recruit histone deacetylases. Deacetylase inhibitors (DACi) are promising drugs leading to growth inhibition, cell cycle arrest, premature senescence and apoptosis in malignant cells. It is believed that DACi may have clinical efficacy by eradicating the most primitive population of leukemic stem and progenitor cells, possibly by interfering with self-renewal.

the aim of the study was to investigate the effects of DACi on leukemic stem and progenitor cells using murine trans-duction-transplantation models of hematopoietic cells harboring the leukemia-associated fusion proteins (LAFp) pLZF/RARα or a truncated AML1/eto protein (AML1/eto exon 9). We show that the self-renewal and short-term repopulation capacity of AML1/eto- or pLZF/RARα-expressing Sca1+/lin- stem and progenitor cells are profoundly inhibited by clini-cally applicable concentrations of the DACi dacinostat and vorinostat. to further investigate the mechanisms underlying these effects, we examined the impact of DACi on the transcription factor c-MYC and the polycomb group protein BMI1, which are induced by LAFp and involved in leukemic transformation. In AML1/eto or pLZF/RARα-positive 32D cells, DACi-mediated antiproliferative effects were associated with downregulation of BMI1 and c-MYC protein levels. Similar effects were demonstrated in primary samples of cytogenetically defined high-risk AML patients. In conclusion, DACi may be ef-fective as maintenance therapy by negatively interfering with signaling pathways that control survival and proliferation of leukemic stem and progenitor cells.

Deacetylase inhibitors modulate proliferation and self-renewal properties of leukemic

stem and progenitor cellsAnnette Romanski,1,† Kerstin Schwarz,1,† Maren Keller,1 Sarah Wietbrauk,1 Anja Vogel,1 Jessica Roos,1 Claudia oancea,1

Boris Brill,2 oliver H. Krämer,3 Hubert Serve,1 Martin Ruthardt1 and Gesine Bug1,*

1Department of Medicine II; Hematology and oncology; Johann Wolfgang Goethe-University; Frankfurt/Main, Germany; 2Institute for Biomedical Research Georg Speyer-Haus; Frankfurt/Main, Germany; 3Institute of Biochemistry and Biophysics; Center for Molecular Biomedicine; Friedrich-Schiller University; Jena, Germany

†these authors contributed equally to this work.

Keywords: acute myeloid leukemia, leukemic stem cells, deacetylase inhibitor, BMI1, self-renewal, short-term repopulation, dacinostat, vorinostat

mechanisms include epigenetic reprogramming of the transcrip-tome by alteration of the acetylation status of core histones and thus modification of the chromatin structure. DACi additionally induce selective proteasomal degradation of histone decetylase 2 (HDAC2), DNA methyltransferases and other proteins respon-sible for aberrant gene repression and signaling.4-6 This effect equally alters the cellular program, but to date, the underlying mechanisms remain elusive and have not yet been studied in the leukemic stem and progenitor compartment.

Balanced translocations such as t(8;21) and t(11;15) and the corresponding leukemia-associated fusion proteins (LAFP) AML1/ETO and PLZF/RARα, respectively, are a hallmark of AML. Ectopic expression of PLZF/RARα or a truncated AML1/ETO protein (AML1/ETO exon 9) in murine hematopoietic stem cells and transplantation into syngeneic mice recapitulates the leukemic phenotype characterized by a differentiation block

Page 2: Deacetylase inhibitors modulate proliferation and self-renewal properties of leukemic stem and progenitor cells

©20

12 L

ande

s B

iosc

ienc

e. D

o no

t dis

tribu

te.

3220 Cell Cycle Volume 11 Issue 17

by PLZF/RARα or a truncated form of AML1/ETO (AML1/ETO exon 9), respectively.7,8 Both leukemia-associated fusion proteins (LAFP) were cloned into proviral constructs and retrovi-rally transduced into 32D cells. Expression of the fusion proteins was confirmed by western blot (Fig. 1A and B). The LAFP-transduced 32D cells were cultured for 7 d with dacinostat (10 and 20 nM) or vorinostat (1 and 2 μM). Selection of the respec-tive DACi doses was based on previously proven inhibition of deacetylase activity in primary AML progenitor cells and clinical relevance.4,19 DACi treatment efficiently inhibited proliferation of LAFP- as well as mock-transduced 32D cells by the factor 5–21 compared with untreated controls (Fig. 1C). This anti-prolifera-tive effect was dose-dependent, but not statistically significantly different between LAFP- and mock-transduced 32D cells.

Deacetylase inhibitors impair short-term engraftment potential of leukemic stem cells. To determine whether DACi are targeting the leukemic stem and progenitor cell compartment, we utilized a transduction-transplantation mouse model (Fig. 2A). PLZF/RARα and AML1/ETO were retrovirally transduced into Sca1+/lin- hematopoietic stem cells (HSC). FACS analysis of GFP positive cells confirmed constant transduction efficien-cies of > 60–70% (Fig. 2B). Addition of dacinostat (20 nM) or vorinostat (2 μM) to a 7-day culture of 2.5 x 104 mock- or LAFP-expressing Sca1+/lin- cells resulted in a six- to 22-fold reduction of cell numbers compared with untreated controls (n = 3; Fig. 2C). Importantly, the antiproliferative effect of the DACi on the bulk of mock- or LAFP-transduced HSC did not vary significantly.

Engraftment and in vivo proliferation capacity of the DACi-pretreated leukemic stem and progenitor cells were assessed in

and an increased self-renewal capacity.7,8 AML1/ETO as well as PLZF/RARα have been found to induce the transcription factor c-MYC,9,10 and ectopic expression of c-MYC in murine bone marrow elicited an aggressive myeloid leukemia by con-ferring self-renewal capacity to committed myeloid progenitor cells.11,12 The Polycomb group (PcG) protein BMI1 is essential for the leukemic transformation property of PLZF/RARα and a target gene of c-MYC.13,14 The PcG family of proteins main-tains the repressed transcriptional state of their target genes und thus contributes to regulation of stem cell renewal.15 As part of the Polycomb-repressive complex-1 (PRC1), BMI1 mediates the interaction between PLZF/RARα and PRC1, resulting in repres-sion of retinoic acid responsive genes.

As aberrant recruitment of histone deacetylase activity is a common oncogenic feature of LAFP, we have analyzed the impact of DACi on leukemic AML1/ETO and PLZF/RARα-positive stem and progenitor cells using the potent DACi dacinostat and vorinostat. Both DACi belong to the group of hydroxamic acid derivatives which have shown antitumor activity in pre-clinical models and proven its efficacy in patients with hematologic malignancies including AML.16-18 The aim of the study was to investigate the effects of DACi on leukemic stem and progenitor cells using doses that are effective and allow long-term treatment.

Results

Deacetylase inhibitors suppress proliferation of AML fusion protein-expressing 32D cells. We studied the impact of DACi using two well characterized models of acute leukemia induced

Figure 1. effect of DACi treatment on proliferation of 32D cells expressing AML1/eto or pLZF/RARα. expression of the oncofusion genes AML1/eto exon 9 (A) and pLZF/RARα (B) in 32D cells was demonstrated by western blotting using the indicated antibodies. Lysates of Kasumi and AML1/eto transduced 293t cells were used as positive controls for AML1/eto. (C) the 32D cells expressing mock, AML1/eto or pLZF/RARα were treated with dacinostat (10 nM, 20 nM) or vorinostat (1 μM, 2 μM) for 7 days, and proliferation was determined by trypan blue exclusion of viable cells. Results are given as fold change reduction in cell growth of DACi-treated cells compared with untreated controls. the means of three separate experiments performed in duplicates +/- SeM are given.

Page 3: Deacetylase inhibitors modulate proliferation and self-renewal properties of leukemic stem and progenitor cells

©20

12 L

ande

s B

iosc

ienc

e. D

o no

t dis

tribu

te.

www.landesbioscience.com Cell Cycle 3221

To determine if dacinostat and vorinostat inhibit the self-renewal potential of the LAFP-positive HSC, serial replating experiments were performed as shown in Figure 3A. We have previously reported that LAFP such as PML/RARα and PLZF/RARα con-fer an increased replating efficiency to Sca1+/lin- HSC, which correlates with an aberrant self-renewal potential.21 Here, Sca1+/lin- stem cells carrying AML1/ETO or PLZF/RARα had a serial replating capacity far exceeding that of mock infected controls (n = 3; more than six vs. three rounds of plating, Fig. 3B–D), which was significantly impaired by DACi treatment. In the presence of dacinostat or vorinostat, colony formation of AML1/ETO-positive cells was significantly reduced from the third plating on and completely abrogated at the sixth plating. Using PLZF/RARα-positive cells, the same results were obtained for

spleen colony-forming unit (CFU-S) assays20 by transplanting all progeny grown from 2.5 × 104 mock or LAFP-transduced Sca1+/lin- cells, i.e., 15–30 × 104 untreated and 1–6 × 104 DACi-treated cells. Spleen colony formation was not affected by exposure to DACi in mice receiving mock-transduced cells (Fig. 2D). In con-trast, DACi-pretreatment of LAFP-transduced Sca1+/lin- cells led to a significant reduction in CFU-S numbers of PLZF/RARα- or AML1/ETO-positive cells in the spleen of transplanted mice as analyzed by RT-PCR (Fig. 2D and E). Quantitative RT-PCR of spleen cells confirmed a 4–7 log reduction of LAFP-positive cells (Fig. 2F) suggesting profound depletion of short-term LSC by the potent DACi dacinostat and vorinostat.

Deacetylase inhibitors exhaust in vitro self-renewal poten-tial of murine AML1/ETO- and PLZF/RARα-positive HSC.

Figure 2. effect of DACi for regulation of leukemia-initiating potential of AML1/eto and pLZF/RARα positive stem cells. (A) experimental strategy for studying the influence of DACi dacinostat and vorinostat on the biology of murine HSC. Sca1+/lin- bone marrow (BM) cells were infected with the indicated retroviruses and maintained for one week in liquid culture supplemented with the indicated growth factors in the presence of DACi. All cells were inoculated into lethally irradiated recipients that were then sacrificed at day 12 after transplantation. (B) GFp reporter gene expression is given by FACS analysis of mock, AML1/eto and pLZF/RARα infected Sca1+/lin- cells. one out of three representative experiments is shown. (C) proliferation of mock, AML1/eto and pLZF/RARα expressing Sca1+/lin- HSC exposed to DACi (20 nM dacinostat, 2 μM vorinostat). the cells were treated with indicated DACi for seven days and proliferation was determined by trypan blue exclusion of viable cells. Reported is the mean of fold change reduction of cell number with SeM compared with untreated controls in vitro (n = 3). (D) Numbers of spleen colonies are given as fold change reduction in CFU-S com-pared with untreated controls (n = 3). the colony numbers show the mean of three independent CFU-S12 experiments with SeM of mock, AML1/eto and pLZF/RARα expressing Sca1+/lin- HSC exposed to DACi (dacinostat, vorinostat) for seven days in vitro and transplanted into three mice per group. p values: p = 0.02 (dacinostat-treated AML1/eto expressing Sca1+/lin- HSC), p = 0.005 (vorinostat-treated pLZF/RAR Sca1+/lin- HSC) compared with mock-treated cells. (e) the expression of the leukemia-associated fusion proteins (LAFp) AML1/eto and pLZF/RARα in the spleen colonies was assessed by Rt-pCR. β-actin was used as a control. (n = 2) (F) For verification of the Rt-pCR analyses, one quantitative real-time pCR analysis of the transgenes AML1/eto and pLZF/RARα of the spleen colonies was performed. Internal reference gene was GApDH. Results are represented as 2-ΔΔCt values.

Page 4: Deacetylase inhibitors modulate proliferation and self-renewal properties of leukemic stem and progenitor cells

©20

12 L

ande

s B

iosc

ienc

e. D

o no

t dis

tribu

te.

3222 Cell Cycle Volume 11 Issue 17

associated with a good prognosis.24 The same holds true for the rare PLZF/RARα-positive acute promyelocytic leukemia.25 To assess the potential clinical implications of our findings, we collected leukemic samples from AML patients with genetically defined high-risk disease, who often fail standard chemotherapy.26,27

Figure 5 displays the results of several AML samples representing the most frequent and clinically relevant high-risk features: complex cytogenetic abnormalities, i.e., three or more chromosomal aberrations (FFM03), mono-somy of chromosome 7 (FFM12), a monosomal karyotype, i.e., presence of one single monosomy in addition to a complex aberrant karyotype (FFM05), and a normal karyotype with pres-ence of an internal tandem duplication in the FMS-related tyrosine kinase 3 gene (FLT3-ITD; FFM23).24 Here, we show that primary AML samples contain primitive leukemic progenitor cells that are capable of in vitro proliferation for several weeks in presence of the cytokines inter-leukin-3, stem cell factor, flt3-ligand and throm-bopoietin. Within six weeks, an exponential cell growth resulting in a 2–4 log expansion was observed in all three AML samples tested (Fig. 5A).

In the presence of dacinostat or vorinostat, the AML cultures consistently declined, confirming our results obtained in AML1/ETO or PLZF/RARα-expressing 32D cells. Hyperacetylation of histone H3 and induction of p21 served as a

proof of the DAC inhibitory activity in FFM05 cells (Fig. 5B). Importantly, downregulation of c-MYC and BMI1 by the potent DACi dacinostat and vorinostat was also verified in this primary AML sample.

Discussion

In the majority of AML patients, a small population of leukemic stem and progenitor cells persists after intensive chemotherapy and represents a reservoir of cells that may lead to recurrent disease. Targeting these LSC may improve patient outcome by reducing the probability of AML relapse.

In our study, we demonstrate that the DACi dacinostat and vorinostat effectively diminish self-renewal and short-term repop-ulating capacity of AML1/ETO- and PLZF/RARα-positive stem and progenitor cells. The well characterized LAFP AML1/ETO exon 9 (referred to as AML1/ETO) and PLZF/RARα are known to confer LSC properties to normal HSC28 and induce an acute leukemia in C57BL/6N mice,8,29 rendering these mouse mod-els excellent read-outs for leukemogenesis. We confirmed that Sca1+/lin- HSC carrying AML1/ETO or PLZF/RARα possess an almost indefinite replating capacity30 and give rise to leukemic colonies in the CFU-S assay, thus demonstrating engraftment and in vivo proliferation of the transplanted LSC.

dacinostat, while residual colony formation was observed in the presence of vorinostat. Thus, prolonged treatment with moderate doses of DACi may severely inhibit or even eliminate self-renew-ing leukemic stem and progenitor cells.

DACi decrease BMI1- und c-MYC protein expression in LAFP-positive 32D cells. We next aimed to examine the effect of DACi on c-MYC as a potential mechanism underlying LSC targeting in LAFP-positive cells. Due to a limited amount of Sca1+/lin- HSC, AML1/ETO- or PLZF/RARα-transduced 32D cells were used for western blot analyses. Treatment with daci-nostat or vorinostat for 48 h led to a dose-dependent reduction of c-MYC protein levels (Fig. 4A). The polycomb group protein BMI1 is another key player involved in regulation of the prolif-erative potential of leukemic stem and progenitor cells.22 Similar to c-MYC, BMI1 was also consistently reduced following DACi treatment. These inhibitory effects were observed in both LAFP- and mock-transduced 32D cells (Fig. 4A). As the cell cycle regulatory gene p21 is a known target of DACi treatment,23 we analyzed its expression and found it to be increased in LAFP-positive and mock 32D as expected (Fig. 4B).

Effect of dacinostat and vorinostat on primary patient samples of high-risk AML. Risk classification of AML is based on cytogenetic and molecular genetic aberrations. Accordingly, AML1/ETO which is present in about 12% of AML cases is

Figure 3. effect of DACi treatment on the replating efficiency of Sca1+/lin- HSC expressing AML1/eto and pLZF/RARα. (A) experimental strategy for studying the influence of the indicated DACi on the biology of murine HSC. Sca1+/lin- BM cells were infected with the indicated retroviruses and plated in semi-solid medium with the indicated growth factors to determine the serial plating potential in the presence of either dacinostat or vorino-stat. Replating efficiency of murine Sca1+/lin- HSC expressing mock (B), AML1/eto (C) and pLZF/RARα (D) upon exposure to dacinostat (10 nM, 20 nM) or vorinostat (1 μM, 2 μM). Numbers of platings (first to sixth) and CFU numbers are provided. error bars show means of triplicates with SeM, n = 3.

Page 5: Deacetylase inhibitors modulate proliferation and self-renewal properties of leukemic stem and progenitor cells

©20

12 L

ande

s B

iosc

ienc

e. D

o no

t dis

tribu

te.

www.landesbioscience.com Cell Cycle 3223

ETO-positive hematopoietic cells and, by another group, in various breast cancer cell lines.34 Downregulation of c-MYC by DACi has been reported previously35-38 and may also account for inhibition of LSC capacity. Here, a reduced protein level of c-MYC was recognized as early as 6 h after addition of DACi (data not shown), suggesting that c-MYC repression is a direct effect of DACi treatment. In contrast, the regulation of the c-MYC target gene BMI113 by DACi was found to be indirect and independent from c-MYC in breast cancer.34 Further stud-ies will have to show if BMI1 is a direct target of c-MYC in our leukemic models.

In summary, we present evidence that DACi interfere with signaling pathways controlling survival and proliferation of leukemic stem and progenitor cells. Thus, abrogation or severe reduction of in vitro self-renewal by the hydroxamic acid analogs dacinostat and vorinostat may be associated with beneficial clini-cal effects, e.g., inducing sensitivity of LSC to chemotherapy.39 Our results have potential impact on the clinical development of DACi. To date, DACi have not been approved for the treat-ment of AML patients. Although complete remissions have been reported, most patients do not enjoy a satisfactory and sustained antileukemic effect and/or suffer from considerable dose-depen-dent side effects. According to our data showing a substantial

After treatment of the LAFP-transduced HSC with dacinostat or vorinostat, only few PLZF/RARα- or AML1/ETO-expressing spleen cells could be detected by qRT-PCR, suggesting profound depletion of leukemia-initiating cells. Expression of LAFP in the spleens was routinely evaluated to discriminate between leukemic and residual normal cells, which were present in the transplanted population as we did not isolate trans-duced Sca1+/lin- cells in view of con-sistently high transduction efficiencies exceeding 60–70%.

These results are in agreement with our previously published data showing considerable loss of normal committed progenitor cells in response to DACi, as measured by cell proliferation, while largely sparing the stem cell frac-tion giving rise to CFU-S.19 Here, we provide evidence that leukemic short-term repopulating stem cells are more susceptible to DACi-mediated damage than their normal counterparts.

In methylcellulose culture, our HSC exhausted their proliferative capacity after the third plating, while LAFP-transduced HSC form colonies for more than three passages as previ-ously reported.30 Serial replating and day 12 spleen colony-forming unit assays (CFU-S) are thus both suitable to detect functional, self-renewing LSC.20 We observed a dose-dependent suppression of the self-renewal capacity of AML1/ETO-positive cells by dacino-stat and vorinostat. This observation is consistent with a gradual loss of self-renewing LSC over time and is in agreement with our previous report demonstrating a diminishing replating capacity of PLZF/RARα-positive LSC upon dacinostat treatment.31

As primary LSC are rare, pathway analysis was performed using transduced IL-3 dependent myeloid 32D cells as a well-accepted model system. Functional impairment of murine 32D but also of human AML cells was associated with downregula-tion of c-MYC and BMI1. The proliferative potential of leuke-mic stem and progenitor cells lacking BMI1 is compromised, because they eventually undergo proliferation arrest and show signs of differentiation and apoptosis, leading to failure of the leukemic cells to initiate disease in transplanted syngeneic hosts.22 Dacinostat and vorinostat reduced BMI1 protein lev-els leading to reversion of the leukemic phenotype. As BMI1 is expressed in all AML cells analyzed so far, it may thus sup-port proliferation and self-renewal of leukemic and progenitor cells independently of PLZF/RARα.32,33 Accordingly, its deple-tion may be associated with a more general antileukemic or even antitumor effect of DACi, based on our results in AML1/

Figure 4. effect of DACi on Wnt target genes in Mock, AML1/eto and pLZF/RARα expressing 32D cells. Infected 32D cells (mock, AML1/eto and pLZF/RARα) were cultured in the presence or absence of dacinostat (10 and 20 nM) or vorinostat (1 and 2 μM) for 48 h. (A) Nuclear extracts were prepared. protein expression of BMI1, c-MYC and Ac-H3 was assessed by western blot analysis with the indicated antibodies. (B) Q-Rt-pCR analysis of p21 expression. Results are represented as 2-ΔΔCt values. Internal reference gene was GApDH. one representative experiment is shown.

Page 6: Deacetylase inhibitors modulate proliferation and self-renewal properties of leukemic stem and progenitor cells

©20

12 L

ande

s B

iosc

ienc

e. D

o no

t dis

tribu

te.

3224 Cell Cycle Volume 11 Issue 17

colony assay, cells harvested from suspension culture were plated in methylcellulose (Methocult® GF H4434, CellSystems Biotech) ± dacinostat or vorinostat. Colonies (> 20 cells) were counted after 12 d and replated if possible. Data were given as mean ± SEM and compared by the Student’s t or Mann-Whitney U-test, as appropriate. p values < 0.05 were considered as significant.

Isolation of Sca1+/lin- murine HSCs. Sca1+/lin- HSCs were isolated from female C57BL/6J mice from 6 to 12 weeks of age (Janvier) killed by cervical dislocation. Bone marrow (BM) was harvested from femora and tibiae by flushing the bones with a syringe and 26-gauge needle. Cells were “lineage depleted” using the Lineage Cell Depletion Kit (Miltenyi Biotech GmbH) according to the manufacturer’s instructions. Sca1+ cells were purified by immunomagnetic beads using EasySep column-free system according to the manufacturer’s instruction (StemCell Technologies, Inc.). Purified cells were pre-stimulated prior to further use for two days in medium containing mIL-3 (20 ng/mL), mIL-6 (20 ng/mL) and murine stem cell factor (mSCF: 100 ng/mL; Peprotech).

Retroviral infection. Phoenix cells were transfected with ret-roviral vectors as described before.41 Retroviral supernatant was collected at days two and three after transfection. Target cells (32D or Sca1+/lin- murine HSCs) were plated onto retronectin-coated (Takara-Shuzo) nontissue culture-treated 24-well plates and exposed to the retroviral supernatant for three hours at 37°C, in the presence of polybrene if 32D cells were used. Cells were centrifuged at 600 g for 45 min. Infection was repeated four times and infection efficiency had to be 70%, as assessed by the

inhibition of LSC, potent DACi may be more relevant in the set-ting of minimal residual disease requiring prolonged treatment.

Materials and Methods

Cells lines and reagents. The hematopoietic progenitor cells 32D were maintained in RPMI-1640 (Invitrogen) plus 10% fetal calf serum (FCS) (Invitrogen) supplemented with 10 ng/ml of m-IL3 (Peprotech). The ecotropic Phoenix packaging cell line was cultured in Dulbecco’s modified Eagle’s medium (DMEM) (Invitrogen) containing 10% FCS. Dacinostat was kindly pro-vided by Novartis Pharmaceuticals and vorinostat (suberoylani-lide hydroxamic acid, SAHA) by Biozol.

AML samples. Peripheral blood samples were obtained from AML patients at diagnosis or relapse. All patients gave written informed consent. Collection of patient samples was approved by the ethics committee of the Goethe-University of Frankfurt. Baseline morphology, cytogenetics and cell surface antigen anal-ysis were performed as part of the routine clinical evaluation of the patients. Isolation of mononuclear cells and the subsequent immunomagnetic selection of CD34+ or CD34+CD38- cells were performed as previously described.40

Culture and analysis of leukemic progenitor cells. CD34+ cells were maintained in X-Vivo (Lonza) supplemented with 10% FCS (Hyclone), 1% L-Glutamine (Invitrogen), interleu-kin-3, thrombopoietin (25 ng/mL each), stem cell factor and Flt-3 ligand (50 ng/mL each, Peprotech) for seven days. Either dacinostat or vorinostat were added to the cultured cells. Flow cytometry was performed as previously described.40 For the

Figure 5. AML cells after DACi treatment. CD34+/CD38- progenitors isolated from AML patients and enriched by magnetic column separation were cultured in the presence or absence of DACi (dacinostat (20 nM) or vorinostat (2 μM). proliferation was determined by trypan dye exclusion assay. (A) exponential cell proliferation of untreated and DACi-treated cells of FFM03, FFM12, FFM23 and FFM05 cells. (B) Regulation of stem-cell specific proteins by DACi. AML cells were cultured in the presence or absence of dacinostat (10 and 20 nM) or vorinostat (1 and 2 μM) for 48 h, and protein lysates were prepared. Altered expression of ac-Histone-3, p21, c-MYC and BMI1 was observed by western blot analysis for FFM05.

Page 7: Deacetylase inhibitors modulate proliferation and self-renewal properties of leukemic stem and progenitor cells

©20

12 L

ande

s B

iosc

ienc

e. D

o no

t dis

tribu

te.

www.landesbioscience.com Cell Cycle 3225

The expression data were evaluated using the ΔΔCT method.42 Analysis of AML1/ETO and PLZF/RARα in the spleen cells was performed in reference laboratories in Frankfurt (Dr. Heike Pfeifer, Department of Medicine II, Johann Wolfgang Goethe-University) and Munich (PD Dr. Susanne Schnittger) by quantitative real-time PCR (qRT-PCR) using GAPDH as a housekeeping gene, as previously described.

RT-PCR. Total RNA and cDNA were obtained according to standard protocols. For detection of the AML1/ETO and PLZF/RARα fusion transcripts by RT-PCR, the following primer pairs were used with an annealing temperature of 55°C; for AML1/ETO: AML1b_fwd (TTG TCG GTC GAA GTG GAA GA) and ETO_rev (GCG CCA TTC AAG GCT GTA G) and for PLZF/RARα: PLZF_fwd (AGG CTG TGG AGC AGC AGC ACA GGA AG) and RAR_rev (TCT GGA TGC TGC GGC GGA AGA A).

Disclosure of Potential Conflicts of Interest

G.B. has received honoraria and travel grants from Novartis Pharma GmbH.

Acknowledgments

The authors are grateful to PD Dr. Susanne Schnittger and Dr. Heike Pfeifer for performing the qRT-PCR for PLZF/RARα and AML1/ETO. The authors thank Novartis Pharmaceuticals for providing dacinostat. This work was supported by the Deutsche Krebshilfe (grant no. 107693 to G.B.) and the Alfred und Angelika Gutermuth-Stiftung.

Authorship

Contribution: A.R., K.S., M.K., S.W., A.V., J.R., C.O. and B.B. performed experiments; G.B., O.H.K., H.S. and M.R. designed the study and analyzed and interpreted the data; A.R., G.B., O.H.K. and M.R. wrote the paper; and all authors reviewed/revised the manuscript and gave their final approval of the ver-sion to be published.

detection of green fluorescent protein (GFP)-positive cells by fluorescence-enhanced cell sorting (FACS).

Colony assays, replating efficiency, murine Sca1+/lin- cells. At day two after infection, Sca1+/lin- cells were plated at 5,000 cells/ml in methylcellulose (Methocult® GF M3534, CellSystems Biotech StemCell) and treated with DACi (dacinostat, vorino-stat). On day 12 after plating, the colony number was counted. After removing the methylcellulose, 5,000 cells/ml were plated again in fresh methylcellulose treated with DACi. Replating effi-ciency was determined by serial plating and counting.

Day 12 spleen colony-forming unit assay (CFU-S12), murine Sca1+/lin- cells. After seven days of culture of Mock, AML1/ETO and PLZF/RARα infected Sca1+/lin- cells (treated with DACi: dacinostat, vorinostat), all grown cells, from initially 25,000 Sca1+/lin- cells, isolated from C57B/6 mice (Ly5.2), were injected into lethally irradiated (12 Gy) female C57b/6 mice (Ly5.2), eight to 12 weeks of age. Three mice per group were transplanted mice and euthanized 12 d later. Spleens were either fixed in Tellesnizky’s fixative for five minutes and subsequently transferred to 70% ethanol or used for RNA isolation and subse-quent RT-PCR analysis.

Western blotting. Western blotting was done according to widely used protocols with the following antibodies: anti-β-actin, anti-acetyl-Histone-3 (from Cell Signaling Technology), anti-c-MYC, anti-BMI1 (from Santa Cruz Biotechnologies). All antibodies were diluted in 5% low fat dry milk or 5% BSA. Blocking was performed in 5% low fat dry milk; washing was performed in TBS containing 0.1% Tween 20 (TBS-T).

Real-time PCR-TaqMan (qRT-PCR). Total RNA and first strand DNA were obtained according to standard protocols. TaqMan-PCR was performed in triplicate using the ABI PRISM 7700 (Applied Biosystems). The related “assays-on-demand” for p21 transcripts were used according to the manufacturer’s instructions (Assay-ID: p21 - Mm00432448_m1) (Applied Biosystems). Internal reference gene was glyceraldehyde-3-phos-phate dehydrogenase (GAPDH) (Assay-ID: Mm99999915_g1).

References1. Lapidot T, Sirard C, Vormoor J, Murdoch B, Hoang T,

Caceres-Cortes J, et al. A cell initiating human acute myeloid leukaemia after transplantation into SCID mice. Nature 1994; 367:645-8; PMID:7509044; http://dx.doi.org/10.1038/367645a0

2. Bonnet D, Dick JE. Human acute myeloid leukemia is organized as a hierarchy that originates from a primitive hematopoietic cell. Nat Med 1997; 3:730-7; PMID:9212098; http://dx.doi.org/10.1038/nm0797-730

3. Romanski A, Bacic B, Bug G, Pfeifer H, Gul H, Remiszewski S, et al. Use of a novel histone deacetylase inhibitor to induce apoptosis in cell lines of acute lym-phoblastic leukemia. Haematologica 2004; 89:419-26; PMID:15075075

4. Bug G, Ritter M, Wassmann B, Schoch C, Heinzel T, Schwarz K, et al. Clinical trial of valproic acid and all-trans retinoic acid in patients with poor-risk acute myeloid leukemia. Cancer 2005; 104:2717-25; PMID:16294345; http://dx.doi.org/10.1002/cncr.21589

5. Fiskus W, Buckley K, Rao R, Mandawat A, Yang Y, Joshi R, et al. Panobinostat treatment depletes EZH2 and DNMT1 levels and enhances decitabine mediated de-repression of JunB and loss of survival of human acute leukemia cells. Cancer Biol Ther 2009; 8:939-50; PMID:19279403; http://dx.doi.org/10.4161/cbt.8.10.8213

6. Buchwald M, Krämer OH, Heinzel T. HDACi--targets beyond chromatin. Cancer Lett 2009; 280:160-7; PMID:19342155; http://dx.doi.org/10.1016/j.can-let.2009.02.028

7. Zheng X, Beissert T, Kukoc-Zivojnov N, Puccetti E, Altschmied J, Strolz C, et al. Gamma-catenin contrib-utes to leukemogenesis induced by AML-associated translocation products by increasing the self-renewal of very primitive progenitor cells. Blood 2004; 103:3535-43; PMID:14739224; http://dx.doi.org/10.1182/blood-2003-09-3335

8. Yan M, Burel SA, Peterson LF, Kanbe E, Iwasaki H, Boyapati A, et al. Deletion of an AML1-ETO C-terminal NcoR/SMRT-interacting region strongly induces leukemia development. Proc Natl Acad Sci USA 2004; 101:17186-91; PMID:15569932; http://dx.doi.org/10.1073/pnas.0406702101

9. Müller-Tidow C, Steffen B, Cauvet T, Tickenbrock L, Ji P, Diederichs S, et al. Translocation products in acute myeloid leukemia activate the Wnt signaling pathway in hematopoietic cells. Mol Cell Biol 2004; 24:2890-904; PMID:15024077; http://dx.doi.org/10.1128/MCB.24.7.2890-2904.2004

10. Rice KL, Hormaeche I, Doulatov S, Flatow JM, Grimwade D, Mills KI, et al. Comprehensive genom-ic screens identify a role for PLZF-RARalpha as a positive regulator of cell proliferation via direct regulation of c-MYC. Blood 2009; 114:5499-511; PMID:19855079; http://dx.doi.org/10.1182/blood-2009-03-206524

11. Luo H, Li Q, O’Neal J, Kreisel F, Le Beau MM, Tomasson MH. c-Myc rapidly induces acute myeloid leukemia in mice without evidence of lymphoma-asso-ciated antiapoptotic mutations. Blood 2005; 106:2452-61; PMID:15972450; http://dx.doi.org/10.1182/blood-2005-02-0734

12. Xiang Z, Luo H, Payton JE, Cain J, Ley TJ, Opferman JT, et al. Mcl1 haploinsufficiency protects mice from Myc-induced acute myeloid leukemia. J Clin Invest 2010; 120:2109-18; PMID:20484815; http://dx.doi.org/10.1172/JCI39964

Page 8: Deacetylase inhibitors modulate proliferation and self-renewal properties of leukemic stem and progenitor cells

©20

12 L

ande

s B

iosc

ienc

e. D

o no

t dis

tribu

te.

3226 Cell Cycle Volume 11 Issue 17

33. Rizo A, Olthof S, Han L, Vellenga E, de Haan G, Schuringa JJ. Repression of BMI1 in normal and leukemic human CD34(+) cells impairs self-renewal and induces apoptosis. Blood 2009; 114:1498-505. PMID:19556423; http://dx.doi.org/10.1182/blood-2009-03-209734

34. Bommi PV, Dimri M, Sahasrabuddhe AA, Khandekar JD, Dimri GP. The polycomb group protein BMI1 is a transcriptional target of HDAC inhibitors. Cell Cycle 2010; 9:2663-73; PMID:20543557; http://dx.doi.org/10.4161/cc.9.13.12147

35. Wang R, Brunner T, Zhang L, Shi Y. Fungal metabolite FR901228 inhibits c-Myc and Fas ligand expression. Oncogene 1998; 17:1503-8; PMID:9794227; http://dx.doi.org/10.1038/sj.onc.1202059

36. Skov S, Rieneck K, Bovin LF, Skak K, Tomra S, Michelsen BK, et al. Histone deacetylase inhibitors: a new class of immunosuppressors targeting a novel signal pathway essential for CD154 expression. Blood 2003; 101:1430-8; PMID:12393479; http://dx.doi.org/10.1182/blood-2002-07-2073

37. Xu Y, Voelter-Mahlknecht S, Mahlknecht U. The his-tone deacetylase inhibitor suberoylanilide hydroxamic acid down-regulates expression levels of Bcr-abl, c-Myc and HDAC3 in chronic myeloid leukemia cell lines. Int J Mol Med 2005; 15:169-72; PMID:15583844

38. Wang LG, Liu XM, Fang Y, Dai W, Chiao FB, Puccio GM, et al. De-repression of the p21 promoter in prostate cancer cells by an isothiocyanate via inhibition of HDACs and c-Myc. Int J Oncol 2008; 33:375-80; PMID:18636159

39. Kadia TM, Yang H, Ferrajoli A, Maddipotti S, Schroeder C, Madden TL, et al. A phase I study of vorinostat in combination with idarubicin in relapsed or refractory leukaemia. Br J Haematol 2010; 150:72-82; PMID:20456355

40. Rossmanith T, Schröder B, Bug G, Müller P, Klenner T, Knaus R, et al. Interleukin 3 improves the ex vivo expansion of primitive human cord blood progeni-tor cells and maintains the engraftment potential of scid repopulating cells. Stem Cells 2001; 19:313-20; PMID:11463951; http://dx.doi.org/10.1634/stem-cells.19-4-313

41. Puccetti E, Obradovic D, Beissert T, Bianchini A, Washburn B, Chiaradonna F, et al. AML-associated translocation products block vitamin D(3)-induced dif-ferentiation by sequestering the vitamin D(3) receptor. Cancer Res 2002; 62:7050-8; PMID:12460926

42. Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)). Method. Methods 2001; 25:402-8; PMID:11846609; http://dx.doi.org/10.1006/meth.2001.1262

24. Döhner H, Estey EH, Amadori S, Appelbaum FR, Büchner T, Burnett AK, et al. European LeukemiaNet. Diagnosis and management of acute myeloid leukemia in adults: recommendations from an international expert panel, on behalf of the European LeukemiaNet. Blood 2010; 115:453-74; PMID:19880497; http://dx.doi.org/10.1182/blood-2009-07-235358

25. Grimwade D, Biondi A, Mozziconacci MJ, Hagemeijer A, Berger R, Neat M, et al. Characterization of acute promyelocytic leukemia cases lacking the clas-sic t(15;17): results of the European Working Party. Groupe Français de Cytogénétique Hématologique, Groupe de Français d’Hematologie Cellulaire, UK Cancer Cytogenetics Group and BIOMED 1 European Community-Concerted Action “Molecular Cytogenetic Diagnosis in Haematological Malignancies”. Blood 2000; 96:1297-308; PMID:10942371

26. Estey E. High cytogenetic or molecular genetic risk acute myeloid leukemia. Hematology Am Soc Hematol Educ Program 2010; 2010:474-80; PMID:21239839.

27. Schlenk RF, Döhner K, Krauter J, Fröhling S, Corbacioglu A, Bullinger L, et al.; German-Austrian Acute Myeloid Leukemia Study Group. Mutations and treatment outcome in cytogenetically normal acute myeloid leukemia. N Engl J Med 2008; 358:1909-18; PMID:18450602; http://dx.doi.org/10.1056/NEJMoa074306

28. Huntly BJ, Shigematsu H, Deguchi K, Lee BH, Mizuno S, Duclos N, et al. MOZ-TIF2, but not BCR-ABL, confers properties of leukemic stem cells to committed murine hematopoietic progenitors. Cancer Cell 2004; 6:587-96; PMID:15607963; http://dx.doi.org/10.1016/j.ccr.2004.10.015

29. He LZ, Guidez F, Tribioli C, Peruzzi D, Ruthardt M, Zelent A, et al. Distinct interactions of PML-RARalpha and PLZF-RARalpha with co-repressors determine differential responses to RA in APL. Nat Genet 1998; 18:126-35; PMID:9462740; http://dx.doi.org/10.1038/ng0298-126

30. Lavau C, Szilvassy SJ, Slany R, Cleary ML. Immortalization and leukemic transformation of a myelomonocytic precursor by retrovirally trans-duced HRX-ENL. EMBO J 1997; 16:4226-37; PMID:9250666; http://dx.doi.org/10.1093/emboj/16.14.4226

31. Puccetti E, Zheng X, Brambilla D, Seshire A, Beissert T, Boehrer S, et al. The integrity of the charged pocket in the BTB/POZ domain is essential for the phenotype induced by the leukemia-associated t(11;17) fusion protein PLZF/RARalpha. Cancer Res 2005; 65:6080-8; PMID:16024608; http://dx.doi.org/10.1158/0008-5472.CAN-04-3631

32. van Gosliga D, Schepers H, Rizo A, van der Kolk D, Vellenga E, Schuringa JJ. Establishing long-term cultures with self-renewing acute myeloid leukemia stem/progenitor cells. Exp Hematol 2007; 35:1538-49; PMID:17889721; http://dx.doi.org/10.1016/j.exphem.2007.07.001

13. Guo WJ, Datta S, Band V, Dimri GP. Mel-18, a polycomb group protein, regulates cell proliferation and senescence via transcriptional repression of Bmi-1 and c-Myc oncoproteins. Mol Biol Cell 2007; 18:536-46; PMID:17151361; http://dx.doi.org/10.1091/mbc.E06-05-0447

14. Boukarabila H, Saurin AJ, Batsché E, Mossadegh N, van Lohuizen M, Otte AP, et al. The PRC1 Polycomb group complex interacts with PLZF/RARA to mediate leukemic transformation. Genes Dev 2009; 23:1195-206; PMID:19451220; http://dx.doi.org/10.1101/gad.512009

15. Sparmann A, van Lohuizen M. Polycomb silencers control cell fate, development and cancer. Nat Rev Cancer 2006; 6:846-56; PMID:17060944; http://dx.doi.org/10.1038/nrc1991

16. Ottmann O, Spencer A, Prince H, Bhalla K, Fischer T, Liu A, et al. Phase IA/II Study of Oral Panobinostat (LBH589), a Novel Pan- Deacetylase Inhibitor (DACi) Demonstrating Efficacy in Patients with Advanced Hematologic Malignancies. Blood 2008; 112:a958.

17. Burbury KL, Bishton MJ, Johnstone RW, Dickinson MJ, Szer J, Prince HM. MLL-aberrant leukemia: complete cytogenetic remission following treatment with a histone deacetylase inhibitor (HDACi). Ann Hematol 2011; 90:847-9; PMID:20949272; http://dx.doi.org/10.1007/s00277-010-1099-6

18. Garcia-Manero G, Yang H, Bueso-Ramos C, Ferrajoli A, Cortes J, Wierda WG, et al. Phase 1 study of the histone deacetylase inhibitor vorinostat (suber-oylanilide hydroxamic acid [SAHA]) in patients with advanced leukemias and myelodysplastic syndromes. Blood 2008; 111:1060-6; PMID:17962510; http://dx.doi.org/10.1182/blood-2007-06-098061

19. Schwarz K, Romanski A, Puccetti E, Wietbrauk S, Vogel A, Keller M, et al. The deacetylase inhibitor LAQ824 induces notch signalling in haematopoi-etic progenitor cells. Leuk Res 2011; 35:119-25; PMID:20674020; http://dx.doi.org/10.1016/j.leu-kres.2010.06.024

20. Coulombel L. Identification of hematopoietic stem/progenitor cells: strength and drawbacks of functional assays. Oncogene 2004; 23:7210-22; PMID:15378081; http://dx.doi.org/10.1038/sj.onc.1207941

21. Zheng X, Seshire A, Puccetti E, Gul H, Beissert T, Hoelzer D, et al. The acute promyelocytic leukemia-associated fusion protein PML/RAR blocks t-RA-induced differentiation in a subset of cells with stem cell potential. Blood 2004; 11:333a.

22. Lessard J, Sauvageau G. Bmi-1 determines the prolif-erative capacity of normal and leukaemic stem cells. Nature 2003; 423:255-60; PMID:12714970; http://dx.doi.org/10.1038/nature01572

23. Richon VM, Sandhoff TW, Rifkind RA, Marks PA. Histone deacetylase inhibitor selectively induces p21WAF1 expression and gene-associated histone acetylation. Proc Natl Acad Sci USA 2000; 97:10014-9; PMID:10954755; http://dx.doi.org/10.1073/pnas.180316197