Purpose: The double-strand breaks elicited by sapacitabine, a clinically active nucleoside analogue prodrug, are repaired by RAD51 and the homologous recombination repair (HR) pathway, which could potentially limit its toxicity. We investigated the mechanism by which histone deacetylase (HDAC) inhibitors targeted RAD51 and HR to sensitize acute myelogenous leukemia (AML) cells to sapacitabine.

Experimental Design: Chromatin immunoprecipitation identified the role of HDACs in silencing miR-182 in AML. Immunoblotting, gene expression, overexpression, or inhibition of miR-182 and luciferase assays established that miR-182 directly targeted RAD51. HR reporter assays, apoptotic assays, and colony-forming assays established that the miR-182, as well as the HDAC inhibition–mediated decreases in RAD51 inhibited HR repair and sensitized cells to sapacitabine.

Results: The gene repressors, HDAC1 and HDAC2, became recruited to the promoter of miR-182 to silence its expression in AML. HDAC inhibition induced miR-182 in AML cell lines and primary AML blasts. miR-182 targeted RAD51 protein both in luciferase assays and in AML cells. Overexpression of miR-182, as well as HDAC inhibition–mediated induction of miR-182 were linked to time- and dose-dependent decreases in the levels of RAD51, an inhibition of HR, increased levels of residual damage, and decreased survival after exposure to double-strand damage-inducing agents.

Conclusions: Our findings define the mechanism by which HDAC inhibition induces miR-182 to target RAD51 and highlights a novel pharmacologic strategy that compromises the ability of AML cells to conduct HR, thereby sensitizing AML cells to DNA-damaging agents that activate HR as a repair and potential resistance mechanism. Clin Cancer Res; 22(14); 3537–49. ©2016 AACR.

Translational Relevance

Sapacitabine is a clinically active nucleoside analogue prodrug that elicits double-strand DNA breaks. RAD51 is a key protein in the homologous recombination repair (HR) pathway that repairs the DNA damage caused by sapacitabine to potentially limit its toxicity. In this study, we identify that miR-182 targets RAD51 and that miR-182 is silenced by the histone deacetylases (HDAC) in acute myelogenous leukemia (AML). HDAC inhibition induces miR-182 to cause reciprocal decreases in RAD51 and inhibits HR to sensitize AML cells to sapacitabine. Our findings highlight a novel pharmacologic strategy that targets RAD51 to compromise the ability of AML cells to conduct HR, and can be used to sensitize cells to DNA-damaging agents that activate HR repair as a potential resistance mechanism.

Sapacitabine is an orally available nucleoside analogue prodrug that is structurally related to cytarabine. It is active against acute myelogenous leukemia (AML) both in elderly patients and in relapsed, refractory disease (1, 2). Sapacitabine is metabolized into its active form 2′-C-Cyano-2′-deoxy-1-β-D-arabino-pentofuranosylcytosine (CNDAC) by amidases. Following intracellular phosphorylation, the resulting triphosphate is incorporated into replicating DNA. In contrast to other nucleoside analogues, this does not terminate replication. Rather, after additional nascent DNA strand elongation, a β-elimination reaction occurs, which causes the analogue to rearrange to a 2′, 3′-dideoxyconfiguration. This terminates the 3′-end resulting in a single-stranded nick, which becomes converted to a double-strand break (DSB) upon a subsequent round of DNA replication (3, 4). The DNA DSBs generated by CNDAC (5) or ionizing radiation (6) are partly repaired by the homologous recombination repair (HR) pathway. HR is governed by multiple proteins such as ATM, the MRE11–RAD50–NBS1 complex (7), BRCA1/2 and RAD51, a key protein that directs the homology search, and DNA strand exchange that is critical for successful HR repair (8). High levels of RAD51 are linked to resistance to DNA-damaging therapies, relapsing disease, and poor survival (9, 10). Conversely, cells lacking RAD51 or its paralog RAD51D have an increased sensitivity to DNA-damaging agents (11) including CNDAC (5). Unlike breast and ovarian tumors that often harbor intrinsic defects in HR, AML cells usually exhibit a functional HR pathway (12), which may allow them to limit the toxicity of DNA DSB-inducing agents.

The histone deacetylases (HDACs) are a class of chromatin-modulating proteins that become recruited to target promoters to compact chromatin and silence gene expression. Conversely, inhibitors of HDACs (HDACi) function by promoting the acetylation of proteins, including histones at gene promoters to induce a transcriptionally active chromatin configuration and reverse HDAC-mediated gene expression (13, 14). HDACis such as vorinostat (SAHA) and belinostat are approved for the treatment of cutaneous T-cell leukemia (15), whereas panobinostat is approved for the treatment of refractory multiple myeloma (16, 17). In AML, HDACi's showed synergy with CNDAC in xenograft models (18) and were effective in phase I/II trials when combined with demethylating (19) or other chemotherapeutic agents (20). Mechanistically, panobinostat exposure led to decreases in the levels of ATM, BRCA1, and RAD51 to sensitize cells to cytarabine, daunorubicin, or ionizing radiation in solid tumor cell lines (21, 22). HDACi treatment also leads to the depression an important class of regulatory genes, the miRNAs (23–25). miRNAs bind to complementary sequences in target RNA to either destabilize it or prevent its transcription in a cell- and context-specific manner (26, 27). Specific miRNA such as miR-182 are overexpressed in breast cancer and intrinsically compromise HR in those cells (28). Other miRNAs, such as miR-103, miR-107, miR-96, and miR-182 target RAD51 and other components of the HR pathway in solid tumors (28–31); however, it is not known whether these miRNA target RAD51 in AML. As RAD51 is well expressed in AML, an investigation of the expression levels of miRNAs that potentially target RAD51 and the mechanisms that regulate their expression becomes important.

In this study, we investigated whether HDAC inhibition upregulated the expression of miRNA genes that targeted RAD51, and whether the resultant impairment in HR sensitized cells to sapacitabine, a clinically relevant DSB-inducing drug in AML.

Materials

Panobinostat (PS) was provided by Novartis. Vorinostat (SAHA) was purchased from Cayman Chemical), and decitabine (DAC) was from Sigma. All reagents were dissolved in 100% DMSO (Burdick & Jackson) to a stock concentration of 10−2 mol/L and stored at −80 °C. CNDAC was supplied by Dr. A. Matsuda (Hokkaido University, Sapporo, Japan).

Cell lines

OCI-AML3 and MV4-11 AML cells were obtained from ATCC and maintained in RPMI supplemented with 5% FCS and 1% glutamine in a 37°C incubator containing 5% CO2. OCI-AML3 cells were authenticated at the Characterized Cell Line Core facility at The University of Texas MD Anderson Cancer Center (Houston, TX). MV4-11 and HeLa-DR-13 cell lines were authenticated by sequencing at the Ohio State University (OSU; Columbus, OH). The HeLa-DR-13 (HeLa-DR) and HEK cells were a gift from Dr. Parvin (Ohio State University, Columbus, OH) and maintained as described previously (32). Primary AML cells and nucleic acids from normal bone marrow were collected under an Institutional review board–approved protocol and obtained from the Leukemia tissue bank at OSU. Primary AML blasts were maintained in RPMI with 20% FCS and 1X Stem Span CC100 (Stem Cell Technologies).

Cytotoxicity assays

OCI-AML3 and MV4-11 cells were exposed to 2 μmol/L CNDAC and increasing concentrations of panobinostat or SAHA for 48 hours following which Annexin assays were conducted by incubating cells with Annexin V–fluorescein isothiocyanate and propidium iodide (BD Biosciences) for 15 minutes and accumulating the fluorescence of at least 10,000 cells on a BD Biosciences FACSCalibur flow cytometer to determine the percentage of apoptotic and viable cells. Drug interactions were analyzed using the CalcuSyn (BioSoft). A combination index (CI) value of 1 indicated an additive drug interaction, whereas a CI value >1 suggested antagonism and a value <1 denoted synergism (33).

miRNA expression array

miRNA microarray analysis was carried out by LC sciences (http://www.lcsciences.com/) as described in Supplementary methods.

Immunoblotting

Immunoblotting was performed with antibodies against RAD51 (Millipore), H3K9Ac (Millipore), PARP (Cell Signaling Technology), cleaved caspase-3 (Cell Signaling Technology), γ-H2AX and H2AX (Cell Signaling Technology), or GAPDH (Cell Signaling Technology).

Chromatin immunoprecipitation and protein immunoprecipitation

Equal amounts of cross-linked chromatin from OCI-AML3 or primary AML cells were used to carry out chromatin immunoprecipitation (ChIP) directed against HDAC1, HDAC2 (Millipore), H3K4me3, and H3K9AC. Rabbit IgG was used as a nonspecific control (Jackson Laboratories; ref. 34). DNA eluted from immunoprecipitates were purified and analyzed by PCR probes specific for the miR-182 promoter. For immunoprecipitation, HDAC1, HDAC2, and IgG immune complexes were prepared by incubating extracts from OCI-AML3 cells with antiserum for 1 hour, followed by 45-minute precipitation with protein A agarose beads, washed, and assayed for the presence of HDAC1 or HDAC2.

Real-time PCR

Total RNA was extracted from cells using the mir-Vana RNA extraction kit (Applied Biosystems). The expression of miR-182 was determined using 5 ng of total RNA, the step loop reverse primer, and the mir-Vana Reverse transcription kit followed by qPCR using primer probes from Applied Biosystems. Expression levels were quantitated by the comparative Ct method using snRNA RNU48 levels for normalization. Similarly, the levels of RAD51 were measured and normalized to GAPDH using the one-step, real-time (RT)-PCR procedure probes from Applied Biosystems.

miRNA, anti-miR, and siRNA transfection

Pre-miR-182, anti-miR-182, and Pre-miR–negative control #1 were obtained from Life Technologies. Oligonucleotides were transfected into OCI-AML3 or HeLa-DR cells at 150 nmol/L using the Nucleofection system, (Solution T, X-001; ref. 35; Amaxa) following manufacturer's instructions for 72 to 96 hours, after which the cells were harvested for RNA and protein analysis.

Colony forming assay

OCI-AML3 were exposed to drugs for 24 hours, washed with PBS (37°C), and plated in methylcellulose medium in triplicate. Clonogenicity was determined after 7 days. OCI-AML3 cells were exposed to scrambled (scr), miR-182, or anti-miR-182 for 48 hours, and then exposed to CNDAC for an additional 24 hours before being assayed for the ability to form colonies after 7 days.

HR-directed repair assay

HeLa-DR cells were transfected with 300 or 500 nmol/L miR-182 or scr for 48 hours before being transfected with the pCBASceI plasmid. For the experiments with miR-182 and scr, we performed a second round of transfection along with the PCBASce1 plasmid. After 96 hours, cells expressing green fluorescence were measured using a Gallios FACSCalibur instrument as described previously (32). For experiments testing HDAC inhibitors, cells were exposed to 10 or 15 nmol/L panobinostat for 24 hours before transfection with PCBASce1 and assayed for HDR as described previously (32).

Luciferase assay

The pmiRTarget vector with RAD51 WT insert (full-length 3′-UTR) or mutated RAD51 3′-UTR (containing a deletion of the miR-182 target site) were cotransfected into HeLa or HEK cells with miR-182 or scr in HeLa cells. Cells were lysed 48 hours after transfection and luciferase and red fluoresence protein intensity (used as an transfection control) were measured for 36 to 48 hours and data expressed as the mean ± SEM from triplicate determinations from two to four independent transfections.

Statistical analysis

Quantitative data are shown as the mean ± SEM for atleast three independent experiments. Comparative statistics used the paired Student t test or one-way ANOVA with Tukey post hoc tests to evaluate differences between experimental groups. P < 0.05 and lower was considered statistically significant.

HDAC inhibitors sensitize AML cells to the cytotoxic action of CNDAC

The pan-HDAC inhibitor, vorinostat (SAHA), has been shown to synergize with CNDAC, the active metabolite of sapacitabine, in killing MV4-11 AML cells both in vitro and in a xenograft mouse model (18). To determine whether panobinostat, a related HDACi, would synergize with CNDAC in AML cells, we exposed OCI-AML3 and MV4-11 cells to either 2 μmol/L CNDAC, 0.1, 0.2, or 0.5 nmol/L panobinostat, or to 0.1, 0.2, or 0.5 μmol/L of SAHA and observed minimal cytotoxicity (Fig. 1A and B and Supplementary Figs. S1A and S1B). However, when these cells were exposed to the combination of 2 μmol/L CNDAC with increasing concentrations of panobinostat up to 0.5 nmol/L (Fig. 1A and Supplementary Fig. S1A) or SAHA up to 0.5 μmol/L (Fig. 1B and Supplementary Fig. S1B) there was a greater than additive loss in cell survival (P value between P < 0.01 and P < 0.05). Drug interactions were analyzed by the Chou–Talalay method; the combination index (CI) values in OCI-AML3 cells for the interaction of 2 μmol/L CNDAC with 0.1, 0.2, or 0.5 nmol/L panobinostat and 0.1, 0.2, or 0.5 μmol/L SAHA were below 0.68 ± 0.24 (Supplementary Table S1). Similarly, the CI value in MV4-11 cells for the interaction of 2 μmol/L CNDAC with 0.1, 0.2, or 0.5 nmol/L panobinostat and 0.1, 0.2, or 0.5 μmol/L SAHA were below 0.72 ± 0.02 indicating synergy (Supplementary Table S1). We then chose the highest concentrations of panobinostat (0.5 nmol/L) and SAHA (0.5 μmol/L) that were synergistic with CNDAC in the apoptotic assays and determined whether they could synergize with CNDAC in colony forming assays. Concentrations of 0.5 μmol/L CNDAC were chosen for the cloning assay because exposure to 1 μmol/L or higher CNDAC inhibited colony growth in the OCI-AML3 cell line (5) attesting to the higher sensitivity of the clonogenic assays. OCI-AML3 cells were exposed to 0.5 μmol/L CNDAC, 0.5 nmol/L panobinostat, 0.5 μmol/L SAHA, or to combinations of CNDAC and panobinostat or CNDAC and SAHA for 24 hours. Our results indicate that the combination of 0.5 nmol/L panobinostat as well as 0.5 μmol/L SAHA was synergistic with 0.5 μmol/L CNDAC in inhibiting colony growth (P < 0.0001 and P < 0.001, respectively; Fig. 1C and D). Taken together, our data indicate that HDACis synergize with CNDAC to limit the survival of AML cell lines.

Figure 1.

HDAC inhibitors synergize with CNDAC in OCI-AML3 cells and cause decreases in RAD51. A, OCI-AML3 cells were treated with 0.1, 0.2, or 0.5 nmol/L panobinostat (PS) alone, 2 μmol/L CNDAC alone or a combination of panobinostat and CNDAC for 48 hours after which cell death was determined by determining the percentage of Annexin V/PI–positive cells. Data represent mean ± SEM of six independent experiments (*,P < 0.05; **, P < 0.01, one-way ANOVA). B, OCI-AML3 cells were treated with 0.1, 0.2, or 0.5 μmol/L vorinistat (SAHA) alone, 2 μmol/L CNDAC alone, or a combination of SAHA and CNDAC for 48 hours after which cell death was determined by determining the percentage of Annexin V/PI–positive cells. Data represent mean ± SEM of three independent experiments (*,P < 0.05; **, P < 0.01, one-way ANOVA). C, OCI-AML3 cells were exposed to 0.5 nmol/L panobinostat, 0.5 μmol/L CNDAC or both drugs for 24 hours in after which they were plated onto methylcellulose and monitored for colony growth. Graph represents mean ± SEM of three independent experiments in triplicate; the difference between CNDAC, panobinostat and the combination was significant (****, P < 0.0001; one-way ANOVA). D, OCI-AML3 cells were exposed to 0.5 μmol/L SAHA, 0.5 μmol/L CNDAC or both drugs for 24 hours in after which they were plated onto methylcellulose and monitored for colony growth. Graph represents mean ± SEM of three independent experiments in triplicate; the difference between CNDAC, SAHA and the combination was significant (***, P < 0.001; one-way ANOVA). E, action of increasing concentrations of panobinostat or SAHA in the presence or absence of 2 μmol/L CNDAC on RAD51. GAPDH was used a loading control and H3K9/14Ac was used to measure the hyperacetylation of histones following HDAC inhibition. RAD51 and GAPDH were quantitated to derive the ratios of RAD51/GAPDH. Figure and ratios of RAD51/GAPDH are representative of three independent experiments, F, action of 0.2 nmol/L and 0.5 nmol/L panobinostat for varying times on the levels of RAD51, cleaved caspase-3 (C-Casp-3), full-length and cleaved PARP and GAPDH in OCI-AML3 cells. Figure is representative of three independent experiments.

Figure 1.

HDAC inhibitors synergize with CNDAC in OCI-AML3 cells and cause decreases in RAD51. A, OCI-AML3 cells were treated with 0.1, 0.2, or 0.5 nmol/L panobinostat (PS) alone, 2 μmol/L CNDAC alone or a combination of panobinostat and CNDAC for 48 hours after which cell death was determined by determining the percentage of Annexin V/PI–positive cells. Data represent mean ± SEM of six independent experiments (*,P < 0.05; **, P < 0.01, one-way ANOVA). B, OCI-AML3 cells were treated with 0.1, 0.2, or 0.5 μmol/L vorinistat (SAHA) alone, 2 μmol/L CNDAC alone, or a combination of SAHA and CNDAC for 48 hours after which cell death was determined by determining the percentage of Annexin V/PI–positive cells. Data represent mean ± SEM of three independent experiments (*,P < 0.05; **, P < 0.01, one-way ANOVA). C, OCI-AML3 cells were exposed to 0.5 nmol/L panobinostat, 0.5 μmol/L CNDAC or both drugs for 24 hours in after which they were plated onto methylcellulose and monitored for colony growth. Graph represents mean ± SEM of three independent experiments in triplicate; the difference between CNDAC, panobinostat and the combination was significant (****, P < 0.0001; one-way ANOVA). D, OCI-AML3 cells were exposed to 0.5 μmol/L SAHA, 0.5 μmol/L CNDAC or both drugs for 24 hours in after which they were plated onto methylcellulose and monitored for colony growth. Graph represents mean ± SEM of three independent experiments in triplicate; the difference between CNDAC, SAHA and the combination was significant (***, P < 0.001; one-way ANOVA). E, action of increasing concentrations of panobinostat or SAHA in the presence or absence of 2 μmol/L CNDAC on RAD51. GAPDH was used a loading control and H3K9/14Ac was used to measure the hyperacetylation of histones following HDAC inhibition. RAD51 and GAPDH were quantitated to derive the ratios of RAD51/GAPDH. Figure and ratios of RAD51/GAPDH are representative of three independent experiments, F, action of 0.2 nmol/L and 0.5 nmol/L panobinostat for varying times on the levels of RAD51, cleaved caspase-3 (C-Casp-3), full-length and cleaved PARP and GAPDH in OCI-AML3 cells. Figure is representative of three independent experiments.

Close modal

HDAC inhibition results in a decrease in the levels of RAD51 protein

In colon cancer cells, HDAC inhibition was linked to decreases in RAD51 and an inability to complete repair by HR (21). To determine the action of HDAC inhibition on RAD51 in AML, we exposed OCI-AML3 cells to increasing concentrations of panobinostat, SAHA, or combinations of panobinostat or SAHA with 2 μmol/L CNDAC for 48 hours. HDAC inhibition resulted in the hyperacetylation of histones H3 and H4 at lysine residues (K9/14). This was accompanied by dose-dependent decreases in the levels of the RAD51 protein in cells exposed to either panobinostat, SAHA, and to combinations of panobinostat or SAHA with 2 μmol/L CNDAC (Fig. 1E), whereas cells exposed to CNDAC alone did not significantly decrease RAD51 protein. Similar results were obtained when MV4-11 cells were exposed to increasing concentrations of panobinostat either alone or in combination with 2 μmol/L CNDAC (Supplementary Fig. S1C). We then conducted time course experiments where OCI-AML3 and MV4-11 cells were exposed to 2 μmol/L CNDAC or 2 μmol/L CNDAC in combination with 0.5 nmol/L panobinostat for up to 48 hours or to 0.5 nmol/L panobinostat alone for 48 hours. Our results demonstrate that cells exposed to CNDAC minimally decreased the levels of RAD51 in both cell lines. However, those exposed to either a combination of CNDAC with panobinostat or panobinostat alone significantly decreased RAD51 levels (Supplementary Fig. S2A and S2B) indicating that it was HDAC inhibition that was responsible for the observed losses in RAD51 protein.

Multiple mechanisms such as downregulation of mRNA transcript (36), caspase-3–mediated cleavage (37) and miRNA-mediated targeting regulate the abundance of any given protein (36). To identify the mechanisms by which HDAC inhibition decreased RAD51 protein, we exposed OCI-AML3 to 0.2 or 0.5 nmol/L panobinostat for varying times. Exposure of 0.2 nmol/L panobinostat or 0.5 nmol/L panobinostat for up to 48 hours led to minimal decreases in the levels of RAD51 mRNA (Supplementary Fig. S3A). The levels of RAD51 protein, on the other hand, decreased by 20% by 48 hours in response to 0.2 nmol/L panobinostat and by over 60% within 24 hours in response to 0.5 nmol/L panobinostat (Fig. 1F and Supplementary Fig. S3B). Loss of RAD51 was accompanied by the appearance of cleaved caspase-3 and some PARP cleavage in cells exposed to 0.5 nmol/L but not 0.2 nmol/L panobinostat (Fig. 1F). In contrast to RAD51, the levels of the closely related HR repair proteins RAD50, ATM, and NBS1 remained constant for up to 48 hours (Supplementary Figs. S3B and S4) suggesting that within the HR pathway, RAD51 was selectively targeted by HDAC inhibition. Because RAD51 is a known substrate of caspase-3 (37) and cells exposed to 0.5 nmol/L panobinostat showed processing of caspase-3, we exposed OCI-AML3 cells to 0.5 nmol/L panobinostat with or without the presence of the pan-caspase inhibitor, z-vad-fmk. The results indicated that levels of RAD51 declined to the same extent regardless of inhibition in caspase-3 processing, indicating that caspase-3–mediated cleavage was not a major mechanism for the observed declines in RAD51 in response to HDAC inhibition (Supplementary Fig. S3C and S3D).

HDAC inhibition induces a miRNA signature that target proteins of the HR pathway

Antagonizing the action of the HDACs leads to a reversal of silencing of a large number of genes, including those of miRNA (23, 24). To determine whether HDAC inhibition could induce the expression of epigenetically silenced miRNA that were predicted to target RAD51, we exposed OCI-AML3 cells to 0.5 nmol/L panobinostat for 6 hours. The expression of a total of 163 miRNA was altered in response to HDAC inhibition; the miRNA significantly up- or downregulated (P < 0.01) are shown in Fig. 2A. Many of the miRNA induced in response to panobinostat putatively targeted multiple genes of the HR repair pathway, such as RAD51, its paralogs (RAD51D, RAD51B, XRCC2/3), BRCA1 and NBS1 (Supplementary Table S2); miRNA predicted to target RAD51 in silico by a minimum of two algorithms (Target Scan and miRanda) and are highlighted in bold in Supplementary Table S2. To validate the results of the microarray, we chose three miRNAs, miR-182, miR-211, and miR-320, that were induced after HDAC inhibition as well as predicted to target RAD51 for further evaluation. Following exposure of OCI-AML3 cells for varying times with 0.5 nmol/L panobinostat, the expression of these miRNA was induced between four- and sixfold in response to HDAC inhibition (Fig. 2B–D).

Figure 2.

HDAC inhibition results in an induction of miRNA expression. A, OCI-AML3 cells were exposed to 0.5 nmol/L panobinostat for 6 hours following which the miRNA from two independent experiments were isolated and hybridized in duplicate to an oligonucleotide array to determine the effect of panobinostat exposure on miRNA expression levels. The figure shows the miRNA that were either upregulated or downregulated with a P <0.01. B–D, validation of microarray results. OCI-AML3 cells were exposed to 0.5 nmol/L panobinostat for 8, 12, or 18 hours after which the induction of miR-211, miR-320, and miR-182 was determined. Assays were done thrice in duplicate. E, MV4-11 cells were exposed to 0.5 μmol/L decitabine (DAC), 10 nmol/L panobinostat, or a combination of DAC and panobinostat for increasing times after which the levels of miR-182 were quantitated. Graph represents mean ± SEM of three experiments. F, MV4-11 cells were exposed to 0.5 μmol/L DAC, 10 nmol/L panobinostat or a combination of DAC and panobinostat for increasing times after which the levels of RAD51 were determined. GAPDH was used as a loading control and H3K9/14Ac was used to measure the hyperacetylation of histones following HDAC inhibition. Figure is representative of three experiments.

Figure 2.

HDAC inhibition results in an induction of miRNA expression. A, OCI-AML3 cells were exposed to 0.5 nmol/L panobinostat for 6 hours following which the miRNA from two independent experiments were isolated and hybridized in duplicate to an oligonucleotide array to determine the effect of panobinostat exposure on miRNA expression levels. The figure shows the miRNA that were either upregulated or downregulated with a P <0.01. B–D, validation of microarray results. OCI-AML3 cells were exposed to 0.5 nmol/L panobinostat for 8, 12, or 18 hours after which the induction of miR-211, miR-320, and miR-182 was determined. Assays were done thrice in duplicate. E, MV4-11 cells were exposed to 0.5 μmol/L decitabine (DAC), 10 nmol/L panobinostat, or a combination of DAC and panobinostat for increasing times after which the levels of miR-182 were quantitated. Graph represents mean ± SEM of three experiments. F, MV4-11 cells were exposed to 0.5 μmol/L DAC, 10 nmol/L panobinostat or a combination of DAC and panobinostat for increasing times after which the levels of RAD51 were determined. GAPDH was used as a loading control and H3K9/14Ac was used to measure the hyperacetylation of histones following HDAC inhibition. Figure is representative of three experiments.

Close modal

DNA hypermethylation acts in conjunction with the histone deacetylases to epigenetically silence gene expression (38). To determine the contribution of DNA hypermethylation to the silencing of miR-182 in AML, we exposed MV4-11 AML cells to 0.5 μmol/L decitabine (DAC, a DNA methylation inhibitor), 10 nmol/L panobinostat, or a combination of panobinostat and DAC for up to 24 hours. We exposed MV4-11 cells to 10 nmol/L panobinostat based on the concentrations utilized to demonstrate synergy with other DNA-damaging agents in this cell line (39). The results show that exposure to DAC alone for up to 48 hours induced miR-182 by less than twofold (Fig. 2E and Supplementary Fig. S5). However, exposure to panobinostat alone or in combination with DAC induced miR-182 levels by sevenfold within 24 hours, indicating that HDACs and not DNA promoter hypermethylation played a dominant role in the silencing of miR-182 in AML (Fig. 2E). Correspondingly, the panobinostat -induced increases in miR-182 in cells exposed to panobinostat alone or in combination with DAC led to reciprocal decreases in the levels of RAD51 (Fig. 2F).

miR-182 targets RAD51 in AML

The ability of a miRNA to target a given mRNA is highly context and tissue specific (26, 27) making it important to validate the interaction between miRNA and its targets in the relevant tissue of interest. Therefore, we focused on 2 miRNAs, miR-320 and miR-182 that had highly conserved binding sites clustered within the central portion of the 3′-UTR of RAD51. The RAD51 gene 3′- UTR has two binding sites for miR320a/c and a single binding site for miR-182 (Fig. 3A). Mimetics for miR-182 and miR-320 were transfected into OCI-AML3 cells along with the nontargeting controls (scr) for 48 hours. Of these, miR-320 did not reduce RAD51, whereas miR-182 consistently reduced the levels of RAD51 by 40% to 50% after 48 hours compared with cells expressing scr oligonucleotides (Fig. 3B–E; P < 0.01, paired t test, n = 6). Time course experiments then confirmed that cells expressing miR-182 targeted RAD51 protein even up to 72 hours (Fig. 3C). Next, to definitively determine the role of miR-182 in targeting RAD51, we overexpressed antagomirs to miR-182 for 48 hours. Our data indicate that in the presence of anti-miR-182, the levels of RAD51 were preserved, whereas expression of miR-182 caused the expected loss in RAD51 levels (Fig. 3D and E). Finally, to test whether miR-182 directly targeted RAD51, HeLa-DR as well as HEK cells were transfected with the pmiRTarget luciferase reporter containing either the full-length RAD51 3′-UTR or one with the miR-182-binding site deleted with miR-182 (Fig. 3F, underlined) or nontargeting controls at 300 nmol/L. Ectopic expression of miR-182 caused a 30% decrease in luciferase activity (P < 0.05, paired t test, n = 4) in HeLa cells and a 70% decrease in luciferase activity (P < 0.05, paired t test, n = 2) in HEK cells in comparison with the scr, whereas deletion of the 3′-UTR binding site for miR-182 restored the luciferase activity in both cell types (Fig. 3G and H). This indicates that miR-182 directly targets RAD51.

Figure 3.

miR-182 targets RAD51. A, represents the predicted location of the binding sites for miR182 and miR-320a/c within the RAD51 3′-UTR. B, action of ectopic expression of the miRNA mimics, miR-182 and miR-320 for 48 hours in OCI-AML3 cells on levels of RAD51. scrambled (scr) sequence was used for comparison and GAPDH was measured as a loading control. Figure is representative of six experiments. C, action of miR-182 on RAD51 in OCI-AML3 cells at 48 and 72 hours. RAD51 levels after transfection with scr sequence and in untreated cells (C) were used for comparison. Figure is representative of three experiments. D and E, action of anti-miR-182 in blocking the miR-182–mediated decrease of RAD51 protein in OCI-AML3 cells; the graph represents the quantitated values for RAD51 protein from six experiments (**, P < 0.01, paired t test). F, the putative binding site of miR-182 on the RAD51 3′-UTR. G, the pmiRTarget vector with RAD51 WT insert (full-length 3′-UTR) or mutated RAD51 3′-UTR (containing a deletion of the miR-182 target site) were cotransfected with miR-182 or Scr in HeLa cells. Luciferase activity was recorded after 36 to 48 hours. The luciferase counts between Scr and miR-182 are statistically significant and represents the mean ± SEM from triplicate determinations from four independent transfections (*, P < 0.05, paired t test). H, the pmiRTarget vector with RAD51 WT insert (full-length 3′-UTR) or mutated RAD51 3′-UTR (containing a deletion of the miR-182 target site as underlined) were cotransfected with miR-182 or Scr in HEK cells. Luciferase activity was recorded after 24 hours. Data represent the mean ± SEM from triplicate determinations from two independent transfections (*, P < 0.05, paired t test).

Figure 3.

miR-182 targets RAD51. A, represents the predicted location of the binding sites for miR182 and miR-320a/c within the RAD51 3′-UTR. B, action of ectopic expression of the miRNA mimics, miR-182 and miR-320 for 48 hours in OCI-AML3 cells on levels of RAD51. scrambled (scr) sequence was used for comparison and GAPDH was measured as a loading control. Figure is representative of six experiments. C, action of miR-182 on RAD51 in OCI-AML3 cells at 48 and 72 hours. RAD51 levels after transfection with scr sequence and in untreated cells (C) were used for comparison. Figure is representative of three experiments. D and E, action of anti-miR-182 in blocking the miR-182–mediated decrease of RAD51 protein in OCI-AML3 cells; the graph represents the quantitated values for RAD51 protein from six experiments (**, P < 0.01, paired t test). F, the putative binding site of miR-182 on the RAD51 3′-UTR. G, the pmiRTarget vector with RAD51 WT insert (full-length 3′-UTR) or mutated RAD51 3′-UTR (containing a deletion of the miR-182 target site) were cotransfected with miR-182 or Scr in HeLa cells. Luciferase activity was recorded after 36 to 48 hours. The luciferase counts between Scr and miR-182 are statistically significant and represents the mean ± SEM from triplicate determinations from four independent transfections (*, P < 0.05, paired t test). H, the pmiRTarget vector with RAD51 WT insert (full-length 3′-UTR) or mutated RAD51 3′-UTR (containing a deletion of the miR-182 target site as underlined) were cotransfected with miR-182 or Scr in HEK cells. Luciferase activity was recorded after 24 hours. Data represent the mean ± SEM from triplicate determinations from two independent transfections (*, P < 0.05, paired t test).

Close modal

HDAC1 and HDAC2 bind the miR-182 promoter in OCI-AML cells and primary AML blasts

We compared the expression of miR-182 in normal bone marrow and AML (Supplementary Table S3) and found that the expression of miR-182 was 10- to 1,000-fold lower in AML blasts (Fig. 4A). As we had determined that HDAC inhibition led to an upregulation of miR-182 in AML, we evaluated whether HDAC1 and HDAC2 were recruited to the miR-182 promoter to silence its expression. miR-182 is an intergenic miRNA transcribed as part of a cluster (40) and contains a major (S1) recruitment site for HDAC1 and HDAC2 (Fig. 4B), the major class I HDACs. HDAC1 and HDAC2 are found together in repressive transcriptional complexes which suggest a high degree of functional redundancy between the two proteins (41). Reciprocal immunoprecipitations from nuclear extracts using antibodies against HDAC1, HDAC2, or IgG were resolved on gels and immunoblotted for the presence of HDAC1 or HDAC2. The results demonstrated that HDAC1 and HDAC2 form a complex in nuclei (Fig. 4C). ChIP assays demonstrated that HDAC1 and HDAC2 were recruited specifically over IgG at the miR-182 promoter in OCI-AML3 cells (Fig. 4D). Conversely, HDAC inhibition led to the accumulation of transcriptionally permissive chromatin modifications such as the trimethylation of histone H3 (H3K4me3) over nonspecific IgG at the miR-182 promoter (Fig. 4E). This was accompanied by a six- to eightfold increase in the expression of miR-182 by 18 hours (Fig. 2D).

Figure 4.

Role of HDACs in regulating miR-182 expression in AML cell lines and primary blasts. A, comparison of miR-182 expression in normal bone marrow (BM; n = 4) and AML blasts (n = 7). B, schematic depicting the putative binding sites for HDAC1 and HDAC2 on the miR-182 promoter. C, coimmunoprecipitation of HDAC1 and HDAC2 from the nuclei of OCI-AML3 cells. Figure is representative of three experiments. D, recruitment of HDAC1 and HDAC2 at the miR-182 promoter in OCI-AML3 cells by ChIP. Data represent mean ± SEM of four independent experiments (***, P < 0.001, paired t test). E, accumulation of the transcriptionally permissive mark H3K4me3 at the miR-182 promoter in OCI-AML3 cells after exposure to 0.5 nmol/L panobinostat (PS) for 6 hours. Data represent mean ± SEM of four independent experiments (****, P < 0.0001, paired t test). F, recruitment of HDAC1 and HDAC2 at the miR-182 at the miR-182 promoter in primary AML blasts by ChIP. Data represent mean ± SEM from three independent samples in triplicate (***, P < 0.001, paired t test). G, accumulation of H3K4me3 at the miR-182 promoter in primary AML blasts after exposure to 10 nmol/L panobinostat for 6 hours. Data represent mean ± SEM from three independent samples in triplicate (****, P < 0.0001, paired t test). H, induction in miR-182 expression in primary AML blasts exposed to panobinostat for varying times. Assays were done in duplicate for each sample. I, decrease in the levels of RAD51 in AML blasts treated as in GAPDH was used a loading control and H3K9Ac was used to measure the hyperacetylation of histones as a positive measure of HDAC inhibition. Figure representative of duplicate immunoblots.

Figure 4.

Role of HDACs in regulating miR-182 expression in AML cell lines and primary blasts. A, comparison of miR-182 expression in normal bone marrow (BM; n = 4) and AML blasts (n = 7). B, schematic depicting the putative binding sites for HDAC1 and HDAC2 on the miR-182 promoter. C, coimmunoprecipitation of HDAC1 and HDAC2 from the nuclei of OCI-AML3 cells. Figure is representative of three experiments. D, recruitment of HDAC1 and HDAC2 at the miR-182 promoter in OCI-AML3 cells by ChIP. Data represent mean ± SEM of four independent experiments (***, P < 0.001, paired t test). E, accumulation of the transcriptionally permissive mark H3K4me3 at the miR-182 promoter in OCI-AML3 cells after exposure to 0.5 nmol/L panobinostat (PS) for 6 hours. Data represent mean ± SEM of four independent experiments (****, P < 0.0001, paired t test). F, recruitment of HDAC1 and HDAC2 at the miR-182 at the miR-182 promoter in primary AML blasts by ChIP. Data represent mean ± SEM from three independent samples in triplicate (***, P < 0.001, paired t test). G, accumulation of H3K4me3 at the miR-182 promoter in primary AML blasts after exposure to 10 nmol/L panobinostat for 6 hours. Data represent mean ± SEM from three independent samples in triplicate (****, P < 0.0001, paired t test). H, induction in miR-182 expression in primary AML blasts exposed to panobinostat for varying times. Assays were done in duplicate for each sample. I, decrease in the levels of RAD51 in AML blasts treated as in GAPDH was used a loading control and H3K9Ac was used to measure the hyperacetylation of histones as a positive measure of HDAC inhibition. Figure representative of duplicate immunoblots.

Close modal

Similarly, HDAC1 and HDAC2 were specifically recruited over IgG to the promoter for miR-182 in primary AML blasts (Fig. 4F). The recruitment of HDAC1 and HDAC2 at the miR-182 promoter did not change significantly before or after exposure to panobinostat in AML cell lines or primary samples (Supplementary Fig. S6). The IC50 of panobinostat against AML blasts is less than 20 nmol/L (39). On the basis of this finding and other published results (42), we exposed AML blasts to 10 nmol/L panobinostat which led increases in the levels of the activating chromatin mark, AcH3K9, in the primary AML samples (Fig. 4G), a 10- to 50-fold increase in the levels of miR-182 in primary AML samples (Fig. 4H) as well as reciprocal and sustained losses in the levels of RAD51 in all primary AML cells (Fig. 4I).

Targeting of RAD51 by miR-182 or HDAC inhibition impairs HR and delays DNA repair in AML

Because the loss of RAD51 was associated with an attenuation of DNA repair in solid tumor cells (21), we evaluated the consequences of miR-182 overexpression or of HDAC inhibition on the HR repair process using the HDR assay. We utilized HeLa-DR cells that had the p-DR-GFP (with 2 inactive copies of GFP) integrated into its genome (43). Transfection with the I-SceI endonuclease caused a single DSB, which, if repaired using homologous sequences from the donor GFP gene, resulted in a functional GFP (32). HeLa-DR cells were transfected with scrambled (scr) or miR-182 oligonucleotides at 300 nmol/L or 500 nmol/L for 48 hours before being transfected with the I-SceI endonuclease. Overexpression of miR-182 but not scr resulted in a >50% decrease in RAD51 in HeLa-DR cells (Fig. 5A). Correspondingly, transfection of the scr oligos together with I-SceI endonuclease resulted in 20% GFP-positive cells indicative of successful HR repair (Fig. 5B, C, and E). In contrast, transfection of miR-182 in conjunction with the I-SceI endonuclease led to a >50% inhibition of HR as measured by a reduction in the percentage of GFP-positive cells (Fig. 5B, D, and F; P < 0.001). Inhibition of HR would be expected to lead to higher levels of unrepaired damaged DNA, which can be assayed by measuring the levels of γ-H2AX that persist after DNA damage (44). Therefore, OCI-AML3 cells were exposed to either 0.5 μmol/L CNDAC, or to miR-182 or scr oligonucleotides for 48 hours before being challenged with 0.5 μmol/L CNDAC for 24 hours. Cells exposed to CNDAC alone or those expressing scr did not decrease RAD51, but responded with some increases in γ-H2AX after exposure to CNDAC for 24 hours (Fig. 5G). In contrast, cells transfected with miR-182 showed the expected decrease in RAD51 and exhibited higher levels of γ-H2AX when exposed to CNDAC for 24 hours (Fig. 5G). We then tested the consequence of the miR-182–mediated decrease in RAD51 and inhibition of DNA repair on the survival of cells exposed to CNDAC. OCI-AML3 cells were left untreated, exposed to 0.5 μmol/L CNDAC for 24 hours, or transfected with scr, miR-182, or anti-miR-182 oligonucleotides after which a portion was exposed to 0.5 μmol/L CNDAC for 24 hours before being assayed for colony growth. Untreated cells or those expressing scrambled showed similar numbers of colonies (280 ± 41 and 282 ± 14; Fig. 5H). Expression of miR-182 decreased colony formation by 23% to 218 ± 7 in comparison with cells expressing scr (P < 0.05, two-tailed p test), whereas expression of anti-miR-182 increased the number of colonies to 243 ± 6 in comparison with those expressing miR-182 (P < 0.05, two-tailed p test). Untreated cells or those expressing scr showed a 40% decrease in colony formation to 60 ± 9 and 62 ± 7 after exposure to CNDAC (P < 0.001, two-tailed p test). However, exposure of cells expressing miR-182 CNDAC caused a greater than 65% inhibition in colony formation to 38 ± 9 in comparison with cells expressing scr that were exposed to CNDAC (P < 0.001, two-tailed p test). Finally, exposure of cells expressing anti-miR-182 to CNDAC supported 185 ± 6 colonies (decrease of 24% vs. 65% in cells exposed to miR-182+CNDAC; P < 0.01, two-tailed p test; Fig. 5H). Finally, to determine whether the miR-182–mediated loss in RAD51 augmented the toxicity of panobinostat in combination with CNDAC, we exposed OCI-AML3 cells to scrambled oligonucleotides, miR-182 or anti-miR-182 for 48 hours before challenging cells to a combination of 0.5 nmol/L panobinostat with 0.5 μmol/L CNDAC. Transfection with scrambled oligonucleotides elicited 17% ± 2% Annexin-positive cells. In comparison, cells transfected with miR-182 showed an increase in Annexin-positive cells to 28% ± 7%, whereas those transfected with anti-miR-182 showed decreases in cell death to 13% ± 11%. Addition of panobinostat and to cells transfected with scrambled, miR-182, and anti-miR-182 increased Annexin positivity to 52% ± 2%, 66% ± 12%, and 32% ± 22%, respectively (P = ns, Supplementary Fig. S7).

Figure 5.

miR-182 targets RAD51 to impair HR. A, HeLa-DR cells were treated with miR-182 or scrambled (scr) sequence to determine effects on RAD51. GAPDH was used as a loading control. B, quantitation of the % inhibition of HR in cells overexpressing miR-182 (300 or 500 nmol/L) in comparison with scrambled in four independent experiments conducted in triplicate (***, P < 0.001, paired t test). C–F, HR assay showing impairment of the HR process owing to miR-182 overexpression. HeLa-DR-13 cells were left untransfected, or transfected with the nontargeting miRNA controls (scr) and miR-182 at 300 nmol/L and 500 nmol/L for 48 hours before being transfected with the I-SceI endonuclease to induce a defined DSB. The percentage of GFP-positive cells was measured by flow cytometry after 48 hours after transfecting with I-SceI endonuclease. Figure representative of four independent experiments. G, cells were transfected with Scr or miR-182 for 48 hours before being challenged with 2 μmol/L CNDAC for 24 hours after which the levels of RAD51 were measured to confirm the RAD51 targeting action of miR-182. Following this, the levels of γ-H2AX was assessed as a measure of DNA damage. GAPDH was used as a loading control. Figure is representative of three experiments. H, OCI-AML3 cells were left untreated, or transfected with scr oligonucleotides, miR-182 and anti-miR-182 for 48 hours before a portion from each condition was exposed to 0.5 μmol/L CNDAC for 24 hours. Cells were then washed and plated onto methylcellulose and monitored for colony growth. Graph represents mean ± SEM of three independent experiments in triplicate (*, P < 0.05; **, P < 0.01;***, P < 0.001; two-tailed p tests).

Figure 5.

miR-182 targets RAD51 to impair HR. A, HeLa-DR cells were treated with miR-182 or scrambled (scr) sequence to determine effects on RAD51. GAPDH was used as a loading control. B, quantitation of the % inhibition of HR in cells overexpressing miR-182 (300 or 500 nmol/L) in comparison with scrambled in four independent experiments conducted in triplicate (***, P < 0.001, paired t test). C–F, HR assay showing impairment of the HR process owing to miR-182 overexpression. HeLa-DR-13 cells were left untransfected, or transfected with the nontargeting miRNA controls (scr) and miR-182 at 300 nmol/L and 500 nmol/L for 48 hours before being transfected with the I-SceI endonuclease to induce a defined DSB. The percentage of GFP-positive cells was measured by flow cytometry after 48 hours after transfecting with I-SceI endonuclease. Figure representative of four independent experiments. G, cells were transfected with Scr or miR-182 for 48 hours before being challenged with 2 μmol/L CNDAC for 24 hours after which the levels of RAD51 were measured to confirm the RAD51 targeting action of miR-182. Following this, the levels of γ-H2AX was assessed as a measure of DNA damage. GAPDH was used as a loading control. Figure is representative of three experiments. H, OCI-AML3 cells were left untreated, or transfected with scr oligonucleotides, miR-182 and anti-miR-182 for 48 hours before a portion from each condition was exposed to 0.5 μmol/L CNDAC for 24 hours. Cells were then washed and plated onto methylcellulose and monitored for colony growth. Graph represents mean ± SEM of three independent experiments in triplicate (*, P < 0.05; **, P < 0.01;***, P < 0.001; two-tailed p tests).

Close modal

We then tested the consequence of HDAC inhibition on the ability of Hela-DR cells to carry out HR. Cells transfected with the I-SceI endonuclease alone showed 24% GFP-positive cells indicating ongoing HR, whereas those preexposed to 10 and 15 nmol/L panobinostat responded with a 75% to 80% inhibition in HR (Fig. 6A–E). We exposed HeLa-DR-13 cells to concentrations of 10 and 15 nmol/L panobinostat because those concentrations decreased RAD51 protein by >50% (Fig. 6F). We then tested whether HDAC-mediated inhibition of HR would affect the ability of cells to resolve DNA damage. When OCI-AML3 cells were exposed to IR, an agent that causes instantaneous DSB, there was a rapid increase in the levels of γ-H2AX which decreased to baseline within 4 to 6 hours indicating the successful repair of DNA damage. However, if these cells were pretreated with 0.5 nmol/L panobinostat before being exposed to IR, the γ-H2AX signal persisted indicating a delay in DNA repair (Fig. 6G). In parallel, when OCI-AML cells were exposed to CNDAC which causes DSB after 2 replication cycles, there was an increase in the γ-H2AX starting at 24 to 48 hours. However, pretreatment with panobinostat resulted in γ-H2AX accumulating earlier (16 hours) and to a greater level suggesting that the HDACi-mediated loss in RAD51 led to a delay in DNA repair and higher levels of residual DNA damage (Fig. 6H).

Figure 6.

Exposure to panobinostat (PS) impairs homologous recombination and leads to a sustained increase in the intensity of γ-H2AX after IR or CNDAC-induced double strand breaks. A–D, HR assay shows impairment of the HR process when cells are exposed to panobinostat. HeLa-DR cells were left untransfected, transfected with the I-SceI endonuclease alone to induce a defined DSB or exposed to 10 nmol/L and 15 nmol/L panobinostat before transfecting with the I-SceI endonuclease after which the percentage of GFP positive cells was measured by flow cytometry at 48 hours. Figure representative of three independent experiments. E, the comparisons between HeLa-DR transfected with I-SceI and HeLa-DR transfected with sce-1 in the presence of 10 nmol/L or 15 nmol/L panobinostat are statistically significant (***, P < 0.001, paired t test) and represent the average values of triplicate determinations of three independent experiments. F, exposure to 10 nmol/L panobinostat leads to a decrease in the levels of RAD51 in the HeLa-DR cells. G, OCI-AML3 cells were exposed to 3 Gy ionizing radiation (IR) alone or to 0.5 nmol/L panobinostat for 18 hours before being challenged with 3 Gy IR after which the levels of γ-H2AX were determined over time. Total H2AX levels were assayed as a control. Figure representative of three independent experiments. H, OCI-AML3 cells were exposed to 2 μmol/L CNDAC alone, to 0.5 nmol/L panobinostat alone, or to a combination of both drugs and the levels of γ-H2AX were determined over time. Total H2AX levels were assayed as a control. Figure representative of three independent experiments.

Figure 6.

Exposure to panobinostat (PS) impairs homologous recombination and leads to a sustained increase in the intensity of γ-H2AX after IR or CNDAC-induced double strand breaks. A–D, HR assay shows impairment of the HR process when cells are exposed to panobinostat. HeLa-DR cells were left untransfected, transfected with the I-SceI endonuclease alone to induce a defined DSB or exposed to 10 nmol/L and 15 nmol/L panobinostat before transfecting with the I-SceI endonuclease after which the percentage of GFP positive cells was measured by flow cytometry at 48 hours. Figure representative of three independent experiments. E, the comparisons between HeLa-DR transfected with I-SceI and HeLa-DR transfected with sce-1 in the presence of 10 nmol/L or 15 nmol/L panobinostat are statistically significant (***, P < 0.001, paired t test) and represent the average values of triplicate determinations of three independent experiments. F, exposure to 10 nmol/L panobinostat leads to a decrease in the levels of RAD51 in the HeLa-DR cells. G, OCI-AML3 cells were exposed to 3 Gy ionizing radiation (IR) alone or to 0.5 nmol/L panobinostat for 18 hours before being challenged with 3 Gy IR after which the levels of γ-H2AX were determined over time. Total H2AX levels were assayed as a control. Figure representative of three independent experiments. H, OCI-AML3 cells were exposed to 2 μmol/L CNDAC alone, to 0.5 nmol/L panobinostat alone, or to a combination of both drugs and the levels of γ-H2AX were determined over time. Total H2AX levels were assayed as a control. Figure representative of three independent experiments.

Close modal

The HDAC inhibitors panobinostat and SAHA have been shown to synergize with cytarabine, daunorubicin (22), and CNDAC using cell viability assays (18). We utilized cell viability as well as colony forming assays to demonstrate that panobinostat and SAHA synergized with CNDAC in killing AML cells. Pharmacokinetic evaluations show that the maximum serum plasma concentrations of vorinostat are approximately 1.4 μmol/L, 1 to 3 hours postdosing in AML (45, 46) and approximately 19 nmol/L for panobinostat (46) indicating that the concentrations of vorinostat and panobinostat achieved in patients are more than sufficient to inhibit both the proliferative capacity as well as kill AML cells. Correlative studies during therapy would help validate the action of this combination on the proliferative index and survival of circulating AML blasts.

To elucidate the mechanism underlying the synergy between HDACi and CNDAC, we focused on RAD51, a key component of the homologous repair pathway in AML cell lines and primary AML lymphocytes. The one published study did not elucidate the mechanism underlying the observed synergy between SAHA and CNDAC (18), whereas two additional publications showed that panobinostat at concentrations of 10 nmol/L decreased RAD51, BRCA1, and ATM transcripts (22) and protein to inhibit HR (21). However, at the lower concentrations of panobinostat (0.5 nmol/L) used in our study, RAD51 transcript levels did not decrease. Rather, we identified an alternative mechanism whereby RAD51 protein levels decreased due to targeting by miR-182. miRNAs have been shown to regulate components of the HR pathway. In breast cancer cells, miR-182 was found to target BRCA1, P53BP1, as well as CHEK (28, 30). In addition, miR-182, miR-96, miR-103, and miR-106 exhibited high levels of expression in breast cancer cells to target RAD51 (29, 31) and intrinsically suppress HR (28, 31). AML differs from these solid tumors in that the miRNAs targeting RAD51 are underexpressed in AML leading to robust levels of expression of RAD51 as well as a functional HR pathway. In addition, miRNA target proteins in a context-dependent manner and miRNA usually express a reverse correlation with their cellular targets (29, 44). Therefore, although miR-103 and miR-106 targeted RAD51 in solid tumors (29), it was unlikely they would do the same in AML because they and their target, RAD51 were expressed at high levels in OCI-AML3 cells (Supplementary Table S2). This conclusion is supported by our findings with miR-320 which is expressed at high levels in OCI-AML3 cells (Supplementary Table S2) and failed to target RAD51 even though RAD51 was a predicted target.

Thus, we focused on miR-182 because it was expressed at low levels in AML, whereas its predicted target RAD51 had robust expression. Mechanistically, targeting the HDACs with panobinostat or SAHA rapidly reversed the silencing of miR-182, whereas targeting DNA hypermethylation did not indicating that while DNA promoter methylation has a key role in suppressing many miRNAs in AML (38), miR-182 is not regulated by this mechanism. miR-182 directly targeted RAD51 at the protein level and in luciferase assays. Targeting RAD51, either via HDACi exposure or ectopic expression of miR-182 inhibited HR and strongly sensitized AML cells to the cytotoxic action of CNDAC in colony forming assays highlighting the importance of RAD51 as a determinant of the sensitivity of AML cells to double-strand–damaging agents. Manipulating the levels of miR-182 with mimics and anti-miRs moderately increased or attenuated the cytotoxicity caused by the combination of panobinostat and CNDAC. This is not surprising given that the combination of panobinostat and CNDAC induces a very robust cell death response. However, this observation raises the possibility that miR-182 may have additional targets the loss of which may cooperate with panobinostat and CNDAC to impact AML survival.

In conclusion, our findings define the strategy of blocking HDACs to induce miR-182 and limit RAD51 expression. Furthermore, if miR-182 targets multiple components of the HR pathway in AML cells as shown for solid tumors (28, 30); epigenetic reactivation of miR-182 could be used to compromise HR at multiple levels. Because, HDAC inhibition induced miR-182 which targeted RAD51 in AML was sufficient to decrease HR by over 80% and sensitized cells to DNA strand breaking agents, it represents a novel pharmacologic strategy. HR is implicated in the repair of damage caused by diverse agents including topoisomerase I inhibitors (47), IR (6) DNA interstrand cross-linkers (48) as well as sapacitabine (5). This suggests that investigations in other malignancies could establish whether targeting RAD51 expression with HDAC inhibitors may also serve to augment the selective toxicity of DSB-inducing DNA-damaging agents across cancers. Thus, this mechanism-based strategy may have multiple applications for clinical evaluations.

No potential conflicts of interest were disclosed.

Conception and design: W. Plunkett, D. Sampath

Development of methodology: T.-H. Lai, B. Ewald, C. Liu, D. Sampath

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): T.-H. Lai, B. Ewald, A. Zecevic, C. Liu, M. Sulda, D. Papaioannou, R. Garzon, D. Sampath

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): T.-H. Lai, B. Ewald, W. Plunkett, D. Sampath

Writing, review, and/or revision of the manuscript: W. Plunkett, D. Sampath

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): J.S. Blachly

Study supervision: W. Plunkett, D. Sampath

The authors thank the Leukemia Tissue Bank at Ohio State University and Dr. John Byrd for the primary AML samples and intellectual discussions.

This study was supported by the National Cancer Institute, Department of Health and Human Services (R01-CA28596), The MD Anderson Cancer Center, and OSU Cancer Center Support grants (NCI-CA016672 and NCI-CA06058).

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

1.
Kantarjian
H
,
Garcia-Manero
G
,
O'Brien
S
,
Faderl
S
,
Ravandi
F
,
Westwood
R
, et al
Phase I clinical and pharmacokinetic study of oral sapacitabine in patients with acute leukemia and myelodysplastic syndrome
.
J Clin Oncol
2010
;
28
:
285
91
.
2.
Kantarjian
H
,
Faderl
S
,
Garcia-Manero
G
,
Luger
S
,
Venugopal
P
,
Maness
L
, et al
Oral sapacitabine for the treatment of acute myeloid leukaemia in elderly patients: a randomised phase 2 study
.
Lancet Oncol
2012
;
13
:
1096
104
.
3.
Liu
X
,
Kantarjian
H
,
Plunkett
W
. 
Sapacitabine for cancer
.
Expert Opinion Invest Drugs
2012
;
21
:
541
55
.
4.
Azuma
A
,
Huang
P
,
Matsuda
A
,
Plunkett
W
. 
2′-C-cyano-2′-deoxy-1-beta-D-arabino-pentofuranosylcytosine: a novel anticancer nucleoside analog that causes both DNA strand breaks and G(2) arrest
.
Mol Pharmacol
2001
;
59
:
725
31
.
5.
Liu
X
,
Wang
Y
,
Benaissa
S
,
Matsuda
A
,
Kantarjian
H
,
Estrov
Z
, et al
Homologous recombination as a resistance mechanism to replication-induced double-strand breaks caused by the antileukemia agent CNDAC
.
Blood
2010
;
116
:
1737
46
.
6.
Groth
P
,
Orta
ML
,
Elvers
I
,
Majumder
MM
,
Lagerqvist
A
,
Helleday
T
. 
Homologous recombination repairs secondary replication induced DNA double-strand breaks after ionizing radiation
.
Nucleic Acids Res
2012
;
40
:
6585
94
.
7.
Chapman
JR
,
Taylor
MR
,
Boulton
SJ
. 
Playing the end game: DNA double-strand break repair pathway choice
.
Mol Cell
2012
;
47
:
497
510
.
8.
Sung
P
,
Robberson
DL
. 
DNA strand exchange mediated by a RAD51-ssDNA nucleoprotein filament with polarity opposite to that of RecA
.
Cell
1995
;
82
:
453
61
.
9.
Barbano
R
,
Copetti
M
,
Perrone
G
,
Pazienza
V
,
Muscarella
LA
,
Balsamo
T
, et al
High RAD51 mRNA expression characterize estrogen receptor-positive/progesteron receptor-negative breast cancer and is associated with patient's outcome
.
Int J Cancer
2011
;
129
:
536
45
.
10.
Li
Y
,
Yu
H
,
Luo
RZ
,
Zhang
Y
,
Zhang
MF
,
Wang
X
, et al
Elevated expression of Rad51 is correlated with decreased survival in resectable esophageal squamous cell carcinoma
.
J Surg Oncol
2011
;
104
:
617
22
.
11.
Huehls
AM
,
Wagner
JM
,
Huntoon
CJ
,
Karnitz
LM
. 
Identification of DNA repair pathways that affect the survival of ovarian cancer cells treated with a poly(ADP-ribose) polymerase inhibitor in a novel drug combination
.
Mol Pharmacol
2012
;
82
:
767
76
.
12.
Seedhouse
CH
,
Hunter
HM
,
Lloyd-Lewis
B
,
Massip
AM
,
Pallis
M
,
Carter
GI
, et al
DNA repair contributes to the drug-resistant phenotype of primary acute myeloid leukaemia cells with FLT3 internal tandem duplications and is reversed by the FLT3 inhibitor PKC412
.
Leukemia
2006
;
20
:
2130
6
.
13.
Scott
GK
,
Mattie
MD
,
Berger
CE
,
Benz
SC
,
Benz
CC
. 
Rapid alteration of microRNA levels by histone deacetylase inhibition
.
Cancer Res
2006
;
66
:
1277
81
.
14.
Mitsiades
CS
,
Mitsiades
NS
,
McMullan
CJ
,
Poulaki
V
,
Shringarpure
R
,
Hideshima
T
, et al
Transcriptional signature of histone deacetylase inhibition in multiple myeloma: biological and clinical implications
.
Proc Natl Acad Sci U S A
2004
;
101
:
540
5
.
15.
Zain
J
,
O'Connor
OA
. 
Targeting histone deacetyalses in the treatment of B- and T-cell malignancies
.
Invest New Drugs
2010
;
28
:
S58
78
.
16.
Ashjian
E
,
Redic
K
. 
Multiple myeloma: updates for pharmacists in the treatment of relapsed and refractory disease
.
J Oncol Pharm Pract
2016
;
22
:
289
302
.
17.
De Souza
C
,
Chatterji
BP
. 
HDAC inhibitors as novel anti-cancer therapeutics
.
Recent Patents Anti-Cancer Drug Discov
2015
;
10
:
145
62
.
18.
Green
SR
,
Choudhary
AK
,
Fleming
IN
. 
Combination of sapacitabine and HDAC inhibitors stimulates cell death in AML and other tumour types
.
Br J Cancer
2010
;
103
:
1391
9
.
19.
Garcia-Manero
G
,
Kantarjian
HM
,
Sanchez-Gonzalez
B
,
Yang
H
,
Rosner
G
,
Verstovsek
S
, et al
Phase 1/2 study of the combination of 5-aza-2′-deoxycytidine with valproic acid in patients with leukemia
.
Blood
2006
;
108
:
3271
9
.
20.
Garcia-Manero
G
,
Tambaro
FP
,
Bekele
NB
,
Yang
H
,
Ravandi
F
,
Jabbour
E
, et al
Phase II trial of vorinostat with idarubicin and cytarabine for patients with newly diagnosed acute myelogenous leukemia or myelodysplastic syndrome
.
J Clin Oncol
2012
;
30
:
2204
10
.
21.
Adimoolam
S
,
Sirisawad
M
,
Chen
J
,
Thiemann
P
,
Ford
JM
,
Buggy
JJ
. 
HDAC inhibitor PCI-24781 decreases RAD51 expression and inhibits homologous recombination
.
Proc Natl Acad Sci U S A
2007
;
104
:
19482
7
.
22.
Xie
C
,
Drenberg
C
,
Edwards
H
,
Caldwell
JT
,
Chen
W
,
Inaba
H
, et al
Panobinostat enhances cytarabine and daunorubicin sensitivities in AML cells through suppressing the expression of BRCA1, CHK1, and Rad51
.
PLoS One
2013
;
8
:
e79106
.
23.
Sampath
D
,
Calin
GA
,
Puduvalli
VK
,
Gopisetty
G
,
Taccioli
C
,
Liu
CG
, et al
Specific activation of microRNA106b enables the p73 apoptotic response in chronic lymphocytic leukemia by targeting the ubiquitin ligase Itch for degradation
.
Blood
2009
;
113
:
3744
53
.
24.
Sampath
D
,
Liu
C
,
Vasan
K
,
Sulda
M
,
Puduvalli
VK
,
Wierda
WG
, et al
Histone deacetylases mediate the silencing of miR-15a, miR-16, and miR-29b in chronic lymphocytic leukemia
.
Blood
2012
;
119
:
1162
72
.
25.
Wendtner
CM
. 
Cocktail of eternity: HDAC meets miR
.
Blood
2012
;
119
:
1095
6
.
26.
Croce
CM
. 
Causes and consequences of microRNA dysregulation in cancer
.
Nat Rev Genet
2009
;
10
:
704
14
.
27.
Tili
E
,
Michaille
JJ
,
Gandhi
V
,
Plunkett
W
,
Sampath
D
,
Calin
GA
. 
miRNAs and their potential for use against cancer and other diseases
.
Future Oncol
2007
;
3
:
521
37
.
28.
Moskwa
P
,
Buffa
FM
,
Pan
Y
,
Panchakshari
R
,
Gottipati
P
,
Muschel
RJ
, et al
miR-182-mediated downregulation of BRCA1 impacts DNA repair and sensitivity to PARP inhibitors
.
Mol Cell
2011
;
41
:
210
20
.
29.
Huang
JW
,
Wang
Y
,
Dhillon
KK
,
Calses
P
,
Villegas
E
,
Mitchell
PS
, et al
Systematic screen identifies mirnas that target rad51 and rad51d to enhance chemosensitivity
.
Mol Cancer Res
2013
;
11
:
1564
73
.
30.
Krishnan
K
,
Steptoe
AL
,
Martin
HC
,
Wani
S
,
Nones
K
,
Waddell
N
, et al
MicroRNA-182–5p targets a network of genes involved in DNA repair
.
Rna
2013
;
19
:
230
42
.
31.
Wang
Y
,
Huang
JW
,
Calses
P
,
Kemp
CJ
,
Taniguchi
T
. 
MiR-96 downregulates REV1 and RAD51 to promote cellular sensitivity to cisplatin and PARP inhibition
.
Cancer Res
2012
;
72
:
4037
46
.
32.
Kotian
S
,
Liyanarachchi
S
,
Zelent
A
,
Parvin
JD
. 
Histone deacetylases 9 and 10 are required for homologous recombination
.
J Biol Chem
2011
;
286
:
7722
6
.
33.
Chou
TC
,
Talalay
P
. 
Quantitative analysis of dose-effect relationships: the combined effects of multiple drugs or enzyme inhibitors
.
Adv Enzyme Regul
1984
;
22
:
27
55
.
34.
Komashko
VM
,
Acevedo
LG
,
Squazzo
SL
,
Iyengar
SS
,
Rabinovich
A
,
O'Geen
H
, et al
Using ChIP-chip technology to reveal common principles of transcriptional repression in normal and cancer cells
.
Genome Res
2008
;
18
:
521
32
.
35.
Zhang
W
,
Konopleva
M
,
Burks
JK
,
Dywer
KC
,
Schober
WD
,
Yang
JY
, et al
Blockade of mitogen-activated protein kinase/extracellular signal-regulated kinase kinase and murine double minute synergistically induces Apoptosis in acute myeloid leukemia via BH3-only proteins Puma and Bim
.
Cancer Res
2010
;
70
:
2424
34
.
36.
Wu
X
,
Brewer
G
. 
The regulation of mRNA stability in mammalian cells: 2.0
.
Gene
2012
;
500
:
10
21
.
37.
Huang
Y
,
Nakada
S
,
Ishiko
T
,
Utsugisawa
T
,
Datta
R
,
Kharbanda
S
, et al
Role for caspase-mediated cleavage of Rad51 in induction of apoptosis by DNA damage
.
Mol Cell Biol
1999
;
19
:
2986
97
.
38.
Liu
S
,
Wu
LC
,
Pang
J
,
Santhanam
R
,
Schwind
S
,
Wu
YZ
, et al
Sp1/NFkappaB/HDAC/miR-29b regulatory network in KIT-driven myeloid leukemia
.
Cancer Cell
2010
;
17
:
333
47
.
39.
Maiso
P
,
Colado
E
,
Ocio
EM
,
Garayoa
M
,
Martin
J
,
Atadja
P
, et al
The synergy of panobinostat plus doxorubicin in acute myeloid leukemia suggests a role for HDAC inhibitors in the control of DNA repair
.
Leukemia
2009
;
23
:
2265
74
.
40.
Landgraf
P
,
Rusu
M
,
Sheridan
R
,
Sewer
A
,
Iovino
N
,
Aravin
A
, et al
A mammalian microRNA expression atlas based on small RNA library sequencing
.
Cell
2007
;
129
:
1401
14
.
41.
Grozinger
CM
,
Schreiber
SL
. 
Deacetylase enzymes: biological functions and the use of small-molecule inhibitors
.
Chem Biol
2002
;
9
:
3
16
.
42.
Fiskus
W
,
Sharma
S
,
Qi
J
,
Shah
B
,
Devaraj
SG
,
Leveque
C
, et al
BET protein antagonist JQ1 is synergistically lethal with FLT3 tyrosine kinase inhibitor (TKI) and overcomes resistance to FLT3-TKI in AML cells expressing FLT-ITD
.
Mol Cancer Ther
2014
;
13
:
2315
27
.
43.
Ransburgh
DJ
,
Chiba
N
,
Ishioka
C
,
Toland
AE
,
Parvin
JD
. 
Identification of breast tumor mutations in BRCA1 that abolish its function in homologous DNA recombination
.
Cancer Res
2010
;
70
:
988
95
.
44.
Gasparini
P
,
Lovat
F
,
Fassan
M
,
Casadei
L
,
Cascione
L
,
Jacob
NK
, et al
Protective role of miR-155 in breast cancer through RAD51 targeting impairs homologous recombination after irradiation
.
Proc Natl Acad Sci USA
2014
;
111
:
4536
41
.
45.
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
.
46.
Slingerland
M
,
Hess
D
,
Clive
S
,
Sharma
S
,
Sandstrom
P
,
Loman
N
, et al
A phase I, open-label, multicenter study to evaluate the pharmacokinetics and safety of oral panobinostat in patients with advanced solid tumors and various degrees of hepatic function
.
Cancer Chemother Pharmacol
2014
;
74
:
1089
98
.
47.
Maede
Y
,
Shimizu
H
,
Fukushima
T
,
Kogame
T
,
Nakamura
T
,
Miki
T
, et al
Differential and common DNA repair pathways for topoisomerase I- and II-targeted drugs in a genetic DT40 repair cell screen panel
.
Mol Cancer Ther
2014
;
13
:
214
20
.
48.
Hinz
JM
. 
Role of homologous recombination in DNA interstrand crosslink repair
.
Environ Mol Mutagen
2010
;
51
:
582
603
.