Glioma stem cells (GSC) are essential for tumor maintenance, invasiveness, and recurrence. Using a global epigenetic screening with an shRNA library, we identified HDAC3 as an essential factor for GSC stemness. Here, we demonstrated that GSCs poorly respond to an HDAC3 inhibitor, RGFP966 (HDAC3i), owing to the production of IL6 and STAT3 activation. To enhance GSC sensitivity to HDAC3i, we explored whether cotreatment with a BRD4 inhibitor, JQ1 (BRD4i), in GSCs produced a better antitumor effect. BRD4i synergistically inhibits GSC growth in association with HDAC3i. HDAC3 inhibition upregulated the acetylation of H3K27, which allowed the recruitment of BRD4 to the GLI1 gene promoter and induced its expression. GLI1, a transcription factor, turned on the expression of IL6, which led to the activation of STAT3 signaling pathways. However, BRD4i inhibited transcription of the GLI1 gene, thereby blocking the GLI1/IL6/STAT3 pathway. In vivo, the HDAC3i/BRD4i combination caused stronger tumor growth suppression than either drug alone. Thus, HDAC3i/BRD4i might provide promising therapies for GBM.

The incidence of glioma, the most common malignant brain tumor, is on the rise (1). Glioblastoma multiforme (GBM) is the most aggressive form of malignant astrocytoma (2, 3). Less than 5% of patients diagnosed with gliomas survive despite the currents standard treatment including resection, radiotherapy, and chemotherapy (3, 4). Studies have also suggested that the residual therapy-resistant and slow-dividing malignant cells, known as glioma stem cells (GSC; refs. 5–7). There is progress to reinitiate tumor formation and promote invasiveness and recurrence. However, current therapeutic approaches fail to target and eliminate GSCs. Thus, there is a critical need for developing new and effective therapeutics for treating GSCs.

Acetylation and deacetylation of histone are critical determinants in the control of gene expression. The level of histone acetylation is dependent on the recruitment of histone acetyltransferases (HAT) and histone deacetylases (HDAC; refs. 8, 9). Many studies have shown that HDACs are generally considered suppressors of gene expression and HDAC inhibitors exert potent anticancer effects (8, 9). Among members of the HDAC family, HDAC3 plays a key role in cell-cycle regulation, DNA damage control, and genomic stability maintenance (10, 11). Meanwhile, our data showed that inhibition of HDAC3 could lead to suppress GSC proliferation using global epigenetic screening with a shRNA library capable of targeting 243 known regulators of chromatin remodeling. However, we have noted that HDAC3 inhibition promoted tyrosyl phosphorylation of STAT3 (pY705), which potentially limited the inhibitory effects of HDAC3i on GSC growth.

Some studies showed that the combination of pan-HDACi, SAHA, and a BRD4i, JQ1, augmented cell death and more potently than the individual drugs, and suppressed the growth of advanced pancreatic ductal adenocarcinoma (12). In addition, SAHA poorly inhibited breast cancer cell growth owing to increased STAT3 pY705 and owing to its ability to upregulate the expression of receptor for the leukemia-inhibitory factor (LIFR; ref. 13). BRD4 was essential for LIFR transcription and the downstream activation of STAT3, via JAK1 by LIFR. Thus, inhibition of HDAC in association with JAK1 or BRD4 inhibitors potentially affords therapeutic benefit for patients with breast cancer (13). In the current study, we found that a HDAC3-specific inhibitor, RGFP966, was unable to suppress growth due to an elevated activation of STAT3-dependent signaling in GBM. However, a BRD4 inhibitor, JQ1, strongly blocked STAT3 signaling. These observations prompted us that BRD4 inhibitor may enhance GSC sensitivity to HDAC3 inhibitor. Our study demonstrated that upon cotreatment of cells with a HDAC3i, RGFP966 (14), and a BRD4i, JQ1 (15), strongly blocked STAT3 signaling and promoted GSC growth suppression, indicating that combination inhibition of HDAC3 with BRD4 as potential therapies for glioma progression.

Cell culture

The human GSCs lines TS543 and TS576 were provided by Cameron Brenner, Holland, and Mellinghoff laboratories, Memorial Sloan-Kettering Cancer Center (New York, NY). Murine GSCs, CSC2078, CSC1534, and CSC1589 were isolated from hGFAPCre+ p53L/L PtenL/+ mice as described previously (16). They were cultured in neurobasal media (Thermo Fisher Scientific, Inc.) supplemented with EGF (20 ng/mL) and basic FGF (bFGF; 10 ng/mL) or differentiation medium (neurobasal media with 1% FBS). BRD4i and HDAC3i (MedChemExpress) were dissolved in DMSO.

shRNA library screening and score

CSC2078, CSC1534, and CSC1589 were three independent GSC lines from hGFAPCre+ p53L/L PtenL/+ mice, which displayed high self-renewal, partial differentiation, and strong tumor initiation capacities. A shRNA library was established and blocked the expression of 243 known chromatin regulators (17) in these cells and a screening score standard for the candidate genes was as followed: a negative control shRNA (shRluc.713) was set as “Score 4” and those with positive control (shRpa3.455) was set as “Score 1”. Notably, the presence of GFP fluorescence in the vector could enable distinguishing of shRNA-expressing cells from their surrounding noninfected control cells. If the size of GFP-positive colonies is smaller than the negative control, the knockdown of this gene can suppress glioma cell growth. So, according to the size and number of GFP+ clones, target genes whose inactivation by the cognate shRNA that scored in the range of “Score 1–2” in at least two of the three glioma lines used were found out.

Transient transfection

RNAi was used for knocking down the expression of specific genes. The sequences of HDAC3 shRNA, STAT3 shRNA, and other siRNAs were shown in Tables 1 and 2. shRNAs capable of targeting STAT3 (18) and HDAC3 (17) were used as published previously. The siRNAs specific for BRD4, GLI1, and IL6 (Genepharma) were transfected using the reagent (Thermo Fisher Scientific, Inc.) following the manufacturer's protocol. Cells were collected after transfection for 48 to 72 hours.

Table 1.

siRNA oligonucleotides.

BRD4 F: GCCUGAAGAGCCAGUUGUUTT 
GLI1 F: AAACGCTATACAGATCCTA 
IL6 F: GGAGUUUGAGGUAUACCUATT 
siControl F: UUCUCCGAACGUGUCACGUTT 
BRD4 F: GCCUGAAGAGCCAGUUGUUTT 
GLI1 F: AAACGCTATACAGATCCTA 
IL6 F: GGAGUUUGAGGUAUACCUATT 
siControl F: UUCUCCGAACGUGUCACGUTT 
Table 2.

shRNA sequence.

shHDAC3 TGCTGTTGACAGTGAGCGCGAGGAACTTCCCTATAGTGAATAGTGAAGCCACAGATGTATTCACTATAGGGAAGTTCCTCATGCCTACTGCCTCGGA 
shSTAT3 TGCTGTTGACAGTGAGCGACTGGATCTTATGCAGCGGGAATAGTGAAGCCACAGATGTATTCCCGCTGCATAAGATCCAGCTGCCTACTGCCTCGGA 
EV TGCTGTTGACAGTGAGCGCAGGAATTATAATGCTTATCTATAGTGAAGCCACAGATGTATAGATAAGCATTATAATTCCTATGCCTACTGCCTCGGA 
shHDAC3 TGCTGTTGACAGTGAGCGCGAGGAACTTCCCTATAGTGAATAGTGAAGCCACAGATGTATTCACTATAGGGAAGTTCCTCATGCCTACTGCCTCGGA 
shSTAT3 TGCTGTTGACAGTGAGCGACTGGATCTTATGCAGCGGGAATAGTGAAGCCACAGATGTATTCCCGCTGCATAAGATCCAGCTGCCTACTGCCTCGGA 
EV TGCTGTTGACAGTGAGCGCAGGAATTATAATGCTTATCTATAGTGAAGCCACAGATGTATAGATAAGCATTATAATTCCTATGCCTACTGCCTCGGA 

Cell assays

Cell viability

Cell viability was assessed after various treatments using a Cell Counting Kit-8 (MedChemExpress) per the manufacturer's instructions. Cells were seeded in 96-well plates (5,000 cells/well) in 100 μL media. After allowing them to adhere to the plates overnight, various treatments were given.

Combination index (CI) scores were calculated as previously described using CompuSyn software, which was published by the ComboSyn, Inc. Cell proliferation data obtained in the presence of DMSO (i.e., vehicle) single drugs and their combination were used for calculating CI values for each combination point. Quantitatively, the following define the metrics used for drug impact on cells: additivity (CI = 1), synergy (CI < 1), and antagonism (CI > 1). The resulting values were used to construct a plot of CI values over a range of fractions affected. On the basis of the CI values that exhibited best synergy, we chose 500 nmol/L of HDAC3i and 100 nmol/L of BRD4i for treating CSC2078, CSC1589, and TS576 cells in all further experiments. The effective drug concentrations of HDAC3i and BRD4i for TS543 cells were 500 and 200 nmol/L, respectively.

Cell proliferation

Cells were seeded on 6-well plates after treatment at a concentration of 5 × 104 cells/well and number of cells. Cell numbers were counted under a microscope at 24, 48, and 72 hours posttreatment with the indicated agents. Each condition had 3 replicates.

Colony formation assay

After a 0.6% soft agar (prepared in growth media) feeder layer was solidified, cells (2,500 cells/well) were plated in an upper layer consisting of 0.4% soft agar, and then treated with BRD4i (100 nmol/L) or HDAC3i (500 nmol/L). After 15 days, cells were stained with 0.5% crystal violet and photographed.

Self-renewal assay

Self-renewal capacity of GSCs was measured by neural sphere formation assays. Cells (500/well) were seeded into 6-well with proliferation media. After treatment with the indicated agents for 10 days, the number of neural spheres formed in each condition was measured by counting of five random fields under IX71 inverted fluorescence microscope (Olympus).

Cell-cycle analyses

Cells were collected and washed twice with cold PBS. They were suspended in Cell Cycle Detection Kit (BestBio), and the numbers of cells in each cycle phase were determined using a C6 flow cytometer (Becton Dickinson). Each condition had 3 replicates.

Apoptosis assays

Cells (1 × 105) were plated into 6-well dishes and harvested for analyses after various treatments. They were stained with a FITC-Annexin V using a commercially available kit (BD Biosciences) and the FITC+ cells were scored using a Becton Dickinson C6 flow cytometer. Each condition had 3 replicates.

Protein extracts and Western blot analyses

Equal quantities of total cellular proteins were subjected to Western blot analyses as previously described earlier (19). Antibodies used were as follows: pSTAT3 (Tyr705; cat no. #9145), cleaved caspase-3 (cat no. #9664), and acetylated-lysine (cat no. #9814s) from Cell Signaling Technology; BCL-2 (cat no. 26593–1-AP), GLI1 (cat no. 66905–1-AP), and β-actin (cat no. 66009–1-Ig) were from Proteintech Group; STAT3 (cat no. sc-482), HDAC3 (cat no. sc-11417), and Mcl-1 (cat no. sc-12756) were from Santa Cruz Biotechnology and H3K27ac (cat no. ab4729) was from Abcam. Each condition had 3 replicates.

qRT-PCR

RNA was isolated using the TRIzol reagent (Thermo Fisher Scientific, Inc.) and was reverse transcribed using the Thermo Scientific RevertAid First Strand cDNA Synthesis Kit (cat no. #K1622). qPCR was performed using the SYBR Premix Ex TaqTM II (Tli RNaseH Plus; cat no. #RR820A, TaKaRa) by Bio-Rad CFX96 PCR machines (Bio-Rad) The 2−ΔΔCt method was used for quantifying specific transcripts. β-Actin expression was used as internal control for normalization. The primers used were shown in Table 3. Each condition had 5 replicates.

Table 3.

qRT-PCR primer information.

BIM F: GACAGAACCGCAAGACAGGAG 
 R: GGACTTGGGGTTTGTGTTGAC 
Bax F: AGACAGGGGCCTTTTTGCTAC 
 R: AATTCGCCGGAGACACTCG 
Bcl-XL F: ACATCCCAGCTTCACATAACCC 
 R: CCATCCCGAAAGAGTTCATTCAC 
Bcl-2 F: GAGAGCGTCAACAGGGAGATG 
 R: CCAGCCTCCGTTATCCTGGA 
Cyclin D1 F: GCGTACCCTGACACCAATCTC 
 R: ACTTGAAGTAAGATACGGAGGGC 
p21 F: CGAGAACGGTGGAACTTTG 
 R: CCAGGGCTCAGGTAGACCTT 
c-Myc F: ATGCCCCTCAACGTGAACTTC 
 R: GTCGCAGATGAAATAGGGCTG 
β-Actin F: GTGACGTTGACATCCGTAAAGA 
 R: GCCGGACTCATCGTACTCC 
NESTIN F: CGAATATTGTCAATGTGCAGTT 
 R: TTTTCTTGGACCGTTTCTGC 
S-100β F: TGGTTGCCCTCATTGATGTCT 
 R: CCCATCCCCATCTTCGTCC 
Olig2 F: TCATCTTCCTCCAGCACCTC 
 R: CCGTAGATCTCGCTCACCAG 
Tuj-1 F: TAGACCCCAGCGGCAACTAT 
 R: GTTCCAGGTTCCAAGTCCACC 
HDAC3 F: ACGTGGGCAACTTCCACTAC 
 R: GACTCTTGGTGA AGCCTTGC 
BIM F: GACAGAACCGCAAGACAGGAG 
 R: GGACTTGGGGTTTGTGTTGAC 
Bax F: AGACAGGGGCCTTTTTGCTAC 
 R: AATTCGCCGGAGACACTCG 
Bcl-XL F: ACATCCCAGCTTCACATAACCC 
 R: CCATCCCGAAAGAGTTCATTCAC 
Bcl-2 F: GAGAGCGTCAACAGGGAGATG 
 R: CCAGCCTCCGTTATCCTGGA 
Cyclin D1 F: GCGTACCCTGACACCAATCTC 
 R: ACTTGAAGTAAGATACGGAGGGC 
p21 F: CGAGAACGGTGGAACTTTG 
 R: CCAGGGCTCAGGTAGACCTT 
c-Myc F: ATGCCCCTCAACGTGAACTTC 
 R: GTCGCAGATGAAATAGGGCTG 
β-Actin F: GTGACGTTGACATCCGTAAAGA 
 R: GCCGGACTCATCGTACTCC 
NESTIN F: CGAATATTGTCAATGTGCAGTT 
 R: TTTTCTTGGACCGTTTCTGC 
S-100β F: TGGTTGCCCTCATTGATGTCT 
 R: CCCATCCCCATCTTCGTCC 
Olig2 F: TCATCTTCCTCCAGCACCTC 
 R: CCGTAGATCTCGCTCACCAG 
Tuj-1 F: TAGACCCCAGCGGCAACTAT 
 R: GTTCCAGGTTCCAAGTCCACC 
HDAC3 F: ACGTGGGCAACTTCCACTAC 
 R: GACTCTTGGTGA AGCCTTGC 

TUNEL assay

Cells grown on glass coverslips or tissue sections were treated with 4% paraformaldehyde for 30 minutes at room temperature and were then stained using a One Step TUNEL Apoptosis Assay Kit (Beyotime Biotechnology Inc.) per the manufacturer's instructions. FITC-labeled TUNEL-positive cells were observed and enumerated using the IX71 fluorescence microscope (Olympus), and images were captured. Each condition had 3 replicates.

RNA-seq and data analyses

Total RNA was isolated using the TRIzol Reagent (Thermo Fisher Scientific, Inc.). Sequencing libraries were generated using the TruSeq RNA Sample Preparation Kit (Illumina) and then were purified according to AMPure XP system (Beckman Coulter). Purified products were subjected to paired-end (PE) sequencing based on the Illumina HiSeq sequencing platform using next-generation sequencing (NGS). The raw data (Raw Data) were filtered out based on 200 bp cDNA fragments, and the resultant high-quality sequence data (Clean Data) was aligned to the reference genome of the species. On the basis of the alignment results, the expression level of each gene was calculated. On this basis, the samples were further analyzed for differential analysis, enrichment analysis, and cluster analysis on a Hiseq platform (Illumina, Inc.) by Shanghai Personal Biotechnology Co. Ltd. Sequence Read Archive (SRA)/NCBI: Coordinates have been deposited with accession code SUB7884448.

Chromatin immunoprecipitation

Chromatin immunoprecipitation (ChIP) assay was performed using the EZ-Magna ChIP A/G Chromatin Immunoprecipitation Kit (cat no. 17–10086, Millipore) according to the procedures provided by the manufacturer. Following antibodies were used for immunoprecipitation: IgG (cat no.17–10086, Millipore), H3K27ac (cat no. ab4729, Abcam), BRD4 (cat no. #13440, Cell Signaling Technology), and GLI1 (cat no. NB600–600, Novus Biologicals). Quantitative PCR analyses of the DNA fragments recovered from the immunoprecipitated products were performed using a Bio-Rad CFX96 real-time PCR machine (Bio-Rad), using primers that encompass GLI1 promoter. The primers of GLI1 transcriptional start site and IL6 promoter were listed in Table 4. Each condition had 3 replicates.

Table 4.

ChIP-qPCR primer information.

GLI1-TSS-1 F: TTTTTCTCGCTGTTGCCACC 
 R: GAACGGTCCGAAGGAAGGAT 
GLI1-TSS-2 F: TATGGGGTTGGGAGAGTTTG 
 R: AAAGAGACCTGGGACAGACAC 
IL6-TSS-1 F: GCAGTGGGATCAGCACTAACAGAT 
 R: CCTGGACAACAGACAGTAATGTTG 
GLI1-TSS-1 F: TTTTTCTCGCTGTTGCCACC 
 R: GAACGGTCCGAAGGAAGGAT 
GLI1-TSS-2 F: TATGGGGTTGGGAGAGTTTG 
 R: AAAGAGACCTGGGACAGACAC 
IL6-TSS-1 F: GCAGTGGGATCAGCACTAACAGAT 
 R: CCTGGACAACAGACAGTAATGTTG 

Immunofluorescence staining

Cells were fixed in 4% paraformaldehyde and then permeabilized with 0.3% Triton X-100 for 15 minutes. After blocking with 5% BSA, they were incubated overnight with select primary antibodies at 4°C in dark. After washing the sections with PBS, samples were stained with diluted green Alexa Fluor 488–conjugated secondary antibody (AS035; ABclonal Biotech Co., Ltd.) and diluted red Alexa Fluor 647–conjugated secondary antibody for 30 minutes in the dark at room temperature. Finally, cells were treated with DAPI for 2 minutes, and images were acquired using a BX53 fluorescence microscope (Olympus).

IHC

Mouse tumor xenografts were immersed in 4% paraformaldehyde for at least 4 hours at room temperature, followed by dehydration, dipping in wax, and paraffin embedding for the preparation of paraffin sections. The sections were incubated with 2% BSA for 30 minutes first and then with the select primary antibodies and subsequently secondary antibodies. Images were captured using an Olympus BX53 microscope fitted with a digital camera.

ELISA

ELISA was used to measure HDAC3 activity and IL6 level (MEIMIAN) in cell culture supernatants following the manufacturer's protocol. ELISA data were collected using a microplate reader (FLUO star Omega) at absorbance 450 nm. The assay sensitivity ranges of ELISA for HDAC3 activity and IL6 were 1.6 to 65U/L and 10 to 240 pg/mL, respectively.

Animal experiments

BALB/c nude mice were purchased from Beijing Vital River Laboratory Animal Technology. Mice were housed in an air-conditioned room at 22°C ± 2°C with ample supply of food and water. All animal experiments were conducted in accordance with the NIH Guide for the Care and Use of Laboratory Animals, with the approval of the Scientific Investigation Board of the College of Basic Medicine, Jilin University (Changchun, China). Mice were subcutaneously transplanted with TS543 cells (1 × 107/mouse) and tumors were allowed to establish. Tumor-bearing mice were randomized into four groups (n = 6 each) and treatment was initiated when average tumor size reached 4 mm3. HDAC3i (25 mg/kg once every other day) via subcutaneous injection and BRD4i (50 mg/kg once daily) via intraperitoneal injection were given for the number of days indicated. Both drugs were given concurrently if a drug combination was used. The size of tumor was measured every 3 days and tumor volume was calculated with the following formula: V = (length) × (width)2/2. After 21 days, mice were euthanized, and tumor tissues were collected and prepared for Western blot or IHC staining.

Statistical analyses

All analyses were performed using the Prism GraphPad 6.00 software. One-way ANOVA with a Newman–Keuls multiple comparison test was performed to discern the significance of the difference between groups. The comparisons of two sets of data were analyzed using Student t test. A P value <0.05 was considered statistically significant. Data are presented as mean ± SD in each case.

HDAC3 inhibition results in limited proliferation block and feedback activation of STAT3 signaling in GSCs

In an effort to identify the critical chromatin regulators required for stemness of CSCs, we employed RNAi with a shRNA library capable of knocking down 243 known genes and identified as HDAC3 as important factor required for self-renewal and proliferation of GSCs (Supplementary Fig. S1A and S1B). We found that inhibition of HDAC3 suppressed GSCs from hGFAP-Cre+ p53L/L PtenL/+ mice (16). To further verify the role of HDAC3 in GSCs growth, we treated CSC2078 cells with various doses of a HDAC3i, RGFP966 (up to 500 nmol/L). Cell viability analyses showed that HDAC3i did not strongly block cell viability. Its effects were marginal at 1,000 nmol/L, the highest dose tested in this study (Fig. 1A). The drug was active, because it was able to block HDAC3 activity as shown by an ELISA (Fig. 1B). The ineffectiveness of HDAC3i on CSC2078 cell growth at least did not due to a failure of HDAC3 inhibition. Puzzlingly, Western blot analyses showed that STAT3 phosphorylation was increased in a time- and dose-dependent manner in response to HDAC3i treatment. Therefore, we measured levels of antiapoptotic proteins such as Bcl-2 and Mcl-1, the downstream targets of STAT3. Indeed, Bcl-2 and Mcl-1 were also increased in the presence of HDAC3i (Fig. 1C). To determine the importance of STAT3 in this process, we knocked it down using a specific shRNA. Indeed, depletion of STAT3 significantly restored the growth-inhibitory effects of HDAC3 inhibitor (Fig. 1D). STAT3 shRNA itself only slightly elevated the proapoptotic activated caspase-3, which was further increased robustly after HDAC3i treatment (Fig. 1E). Consistent with this, in the presence of STAT3 shRNA, HDAC3i strongly inhibited cell proliferation (Fig. 1F) and self-renewal capacity of GSCs (Fig. 1G). HDAC3i was incapable of blocking growth strongly and the study confirmed that JQ1 can effectively inhibit the STAT3 activation and synergistically enhance the sensitivity of breast cancer to HDAC inhibitor SAHA to achieve a better therapeutic effect (13), so we tested another inhibitor that blocks the acetylation reader BRD4. Interestingly, we found that BRD4i significantly reduced STAT3 phosphorylation (Fig. 1H). On the basis of the above studies, we selected BRD4i to overcome the poor efficacy of HDAC3i and evaluated whether a combination of HDAC3i and BRD4i could cause a more durable suppression of GSC growth.

Figure 1.

HDAC3 inhibition results in limited proliferation block and feedback activation of STAT3 signaling in GSCs. A, Viability of CSC2078 cells was tested using Cell Counting Kit-8 after RGFP966 treatment for various times (n = 3 independent replicates). B, HDAC3 activity measured by ELISA (n = 5 independent replicates). C, CSC2078 cells were treated with different concentrations of RGFP966. D and E, Cells were treated with STAT3 shRNA and RGFP966 treatment for 48 hours. STAT3 phosphorylation and antiapoptotic signaling expression were monitored using Western blot analysis (n = 3 independent replicates). F, Downregulation of STAT3 sensitizes cells to growth inhibition by RGFP966. A cell proliferation assay was conducted with CSC2078 cells, and conditions were indicated. RGFP966 was at 200 μmol/L (n = 9 independent replicates). G, Sphere-forming ability of CSC2078 cells after RGFP966 treatment. Scale bars, 50 μm. H, STAT3 phosphorylation is inhibited by JQ1. Values, mean ± SD. One-way ANOVA was carried out to test significance.

Figure 1.

HDAC3 inhibition results in limited proliferation block and feedback activation of STAT3 signaling in GSCs. A, Viability of CSC2078 cells was tested using Cell Counting Kit-8 after RGFP966 treatment for various times (n = 3 independent replicates). B, HDAC3 activity measured by ELISA (n = 5 independent replicates). C, CSC2078 cells were treated with different concentrations of RGFP966. D and E, Cells were treated with STAT3 shRNA and RGFP966 treatment for 48 hours. STAT3 phosphorylation and antiapoptotic signaling expression were monitored using Western blot analysis (n = 3 independent replicates). F, Downregulation of STAT3 sensitizes cells to growth inhibition by RGFP966. A cell proliferation assay was conducted with CSC2078 cells, and conditions were indicated. RGFP966 was at 200 μmol/L (n = 9 independent replicates). G, Sphere-forming ability of CSC2078 cells after RGFP966 treatment. Scale bars, 50 μm. H, STAT3 phosphorylation is inhibited by JQ1. Values, mean ± SD. One-way ANOVA was carried out to test significance.

Close modal

HDAC3i/BRD4i synergistically inhibits GSC growth

Cotreatment with HDAC3i and BRD4i synergistically suppressed the viability of CSC2078, CSC1589, TS543, and TS576 cells (Fig. 2A; Supplementary Fig. S2A–S2C), as demonstrated by a marked CI values (Fig. 2B; Supplementary Fig. S2D–S2F). Similarly, cell growth was significantly suppressed by HDAC3i in the presence of a BRD4-siRNA (Supplementary Fig. S2G). Kinetic analyses following treatment of cells with HDAC3i in the presence of either JQ1 or siBRD4 showed similar robust effects on CSC2078 cell growth (Fig. 2C and D). In addition, similar patterns of growth inhibition were noted with other GSCs, CSC1589 (Supplementary Fig. S2H), TS543 (Supplementary Fig. S2J), and TS576 cells (Supplementary Fig. S2I). GSCs form undifferentiated stem-like clones in soft agar growth media. Therefore, we next tested whether silencing of HDAC3 and BRD4 also synergized to suppress colony formation. As shown in Supplementary Fig. S2K, cotreatment with HDAC3i/BRD4i caused a marked suppression of colony formation. Figure 2E and Supplementary Fig. S2L–S2N showed quantification of these data. A very similar effect on self-renewal capacity was noted when siBRD4-transfected cells were treated with HDAC3i (Fig. 2F and G; Supplementary Fig. S2O).

Figure 2.

HDAC3i/BRD4i synergistically inhibits GSC growth. A, CSC2078 cell growth was measured using the CCK8 Assay Kit after treating the cells with the indicated agents for 48 hours (n = 5 independent replicates). B, CI for RGFP966 and JQ1 in CSC2078 cells were calculated based on cell growth data using CompuSyn software. C and D, Growth of CSC2078 cells after BRD4 and HDAC3 inhibition (n = 9 independent replicates). E, Soft agar assay for colony formation of CSC2078 cells by RGFP966 and JQ1 (n = 5 independent replicates, means ± SD of five fields). F, Self-renewal ability of CSC2078 cells after treatment with RGFP966 (100 nmol/L)/JQ1 (200 nmol/L). Scale bars, 50 μm. G, Neurosphere formation after BRD4 siRNA and RGFP966 in CSC2078. Scale bars, 50 μm. H, Cell-cycle distribution as measured by flow cytometry in CSC2078 cells after various treatments for 48 hours. I, Quantification of apoptotic death after FITC-Annexin-V staining of CSC2078 cells treated with RGFP966 (100 nmol/L)/JQ1 (200 nmol/L) for 48 hours. J, qPCR analyses of the BIM, Bax, Bcl-XL, Bcl-2, Cyclin D1, p21, c-Myc, and HDAC3 mRNAs in CSC2078 cells (n = 5 independent replicates). Values, mean ± SD. One-way ANOVA was carried out to test significance.

Figure 2.

HDAC3i/BRD4i synergistically inhibits GSC growth. A, CSC2078 cell growth was measured using the CCK8 Assay Kit after treating the cells with the indicated agents for 48 hours (n = 5 independent replicates). B, CI for RGFP966 and JQ1 in CSC2078 cells were calculated based on cell growth data using CompuSyn software. C and D, Growth of CSC2078 cells after BRD4 and HDAC3 inhibition (n = 9 independent replicates). E, Soft agar assay for colony formation of CSC2078 cells by RGFP966 and JQ1 (n = 5 independent replicates, means ± SD of five fields). F, Self-renewal ability of CSC2078 cells after treatment with RGFP966 (100 nmol/L)/JQ1 (200 nmol/L). Scale bars, 50 μm. G, Neurosphere formation after BRD4 siRNA and RGFP966 in CSC2078. Scale bars, 50 μm. H, Cell-cycle distribution as measured by flow cytometry in CSC2078 cells after various treatments for 48 hours. I, Quantification of apoptotic death after FITC-Annexin-V staining of CSC2078 cells treated with RGFP966 (100 nmol/L)/JQ1 (200 nmol/L) for 48 hours. J, qPCR analyses of the BIM, Bax, Bcl-XL, Bcl-2, Cyclin D1, p21, c-Myc, and HDAC3 mRNAs in CSC2078 cells (n = 5 independent replicates). Values, mean ± SD. One-way ANOVA was carried out to test significance.

Close modal

To further examine the mechanisms of growth suppression, we evaluated the cell-cycle status and cell death using flow cytometry. The combination of HDAC3i with either BRD4i or BRD4 siRNA (Supplementary Fig. S3A–S3G) forced more cells into G1 arrest (Fig. 2H) and caused increased apoptosis in cells (Fig. 2I), than those treated with either HDAC3i or BRD4i alone. Consistent with these observations, qPCR analyses showed that the expression of c-Myc, and Cyclin D1 (cell-cycle progression), and Bcl-2 and Bcl-XL (anti-apoptotic) was significantly inhibited, while those of p21 (a CDK-inhibitor) and Bim and Bax (proapoptotic) were significantly upregulated after treatment with HDAC3i and BRD4i in CSC2078 cells (Fig. 2J). A similar effect was noted in human TS543 cells (Supplementary Fig. S3H). To measure apoptosis in these cells, we used TUNEL assays. HDAC3i/BRD4i caused significantly more TUNEL-positive (high fluorescence) in CSC2078 and TS543 cells (Supplementary Fig. S3I and S3J), compared with either the HDAC3i or BRD4i alone. Collectively, these observations indicated that the HDAC3i/BRD4i combination strongly suppresses GSCs proliferation via cycle arrest and promotes apoptosis.

HDAC3i/BRD4i synergistically inhibits GBM progression in vivo

To investigate whether the HDAC3i/BRD4i combination blocks tumor growth in vivo, we used a glioma xenograft mouse model (Fig. 3A). HDAC3i and BRD4i alone produced some significant growth inhibition compared with the untreated control in vivo. However, their combination dramatically reduced tumor volume and growth rate (Fig. 3B and C), compared with either drug alone. Importantly, there was no loss of weight (Supplementary Fig. S4A) and no organ tissue damage (Supplementary Fig. S4B) in mice following any of the treatments, indicating no significant side effects. Tumors resected from the treatment groups were used for IHC analyses to determine the changes in the expression of certain growth-associated gene products. The HDAC3i/BRD4i combination strongly suppressed the expression of growth-promoting proteins PCNA, BCL-2, c-Myc, and STAT3 pY705. These tumors also expressed significantly more BAX (Fig. 3D). TUNEL staining of tumor sections revealed that the HDAC3i/BRD4i combination induced more apoptosis when compared with the controls treated with either drug alone (Fig. 3D). Indeed, qPCR analyses of tumor tissues revealed that corresponding alterations in RNAs coding for Bim, Bax, Bcl-2, Bcl-XL, c-Myc, Cyclin D1, and p21 (Fig. 3E). In addition, Western blot analyses of tumor showed that the combination strongly blocked the pY705 of STAT3 and increased caspase-3 activation (Fig. 3F). Thus, the HDACi/BRD4i combination robustly suppresses tumor growth via an induction of apoptosis.

Figure 3.

HDAC3i/BRD4i synergistically inhibits GBM progression in vivo. A, Schematic representation of treatment schedule for TS543 xenografts. Mice were transplanted with the TS543 cells (1 × 107). After 5 days, tumor-bearing mice (n = 6/group) were randomly assigned for various treatment groups. Tumor growth was quantified over 21 days. Various treatments used in the experiment were indicated. B, Image of the tumor tissues of the transplanted xenogeneic model. C, Tumor growth curve of mice for 21-day treatment (n = 6 independent replicates). D, IHC analyses of tumor tissues for the expression of c-Myc, PCNA, BAX, BCL-2, and STAT3 (pY705), and TUNEL assay (for apoptosis; n = 6 independent replicates). Scale bars, 20 μm. E, Fold changes in the percentage of BIM, Bax, Bcl-XL, Bcl-2, Cyclin D1, p21, and c-Myc transcripts as based on qPCR (n = 5 independent replicates). F, STAT3 (pY705), STAT3, MCL-1, BCL-2, and activated caspase-3 protein levels were determined using Western blot analyses (n = 3 independent replicates). Values, mean ± SD. One-way ANOVA was carried out to test significance.

Figure 3.

HDAC3i/BRD4i synergistically inhibits GBM progression in vivo. A, Schematic representation of treatment schedule for TS543 xenografts. Mice were transplanted with the TS543 cells (1 × 107). After 5 days, tumor-bearing mice (n = 6/group) were randomly assigned for various treatment groups. Tumor growth was quantified over 21 days. Various treatments used in the experiment were indicated. B, Image of the tumor tissues of the transplanted xenogeneic model. C, Tumor growth curve of mice for 21-day treatment (n = 6 independent replicates). D, IHC analyses of tumor tissues for the expression of c-Myc, PCNA, BAX, BCL-2, and STAT3 (pY705), and TUNEL assay (for apoptosis; n = 6 independent replicates). Scale bars, 20 μm. E, Fold changes in the percentage of BIM, Bax, Bcl-XL, Bcl-2, Cyclin D1, p21, and c-Myc transcripts as based on qPCR (n = 5 independent replicates). F, STAT3 (pY705), STAT3, MCL-1, BCL-2, and activated caspase-3 protein levels were determined using Western blot analyses (n = 3 independent replicates). Values, mean ± SD. One-way ANOVA was carried out to test significance.

Close modal

Inhibition of HDAC3 and BRD4 potently induces GSC differentiation

Given the positive effects of HDAC3 on differentiation (20, 21), we next examined whether HDAC3i/BRD4i also promotes GSC differentiation, which typically limits proliferation. Data from qPCR analyses showed that the inhibition of HDAC3 and BRD4 suppressed the expression of NESTIN (a stem cell marker) and increased that of S-100β (a mature astrocyte marker) in CSC2078 cell growth in a differentiation medium (Fig. 4A) and in vivo (Fig. 4B). Because the levels of Olig2 (an oligodendrocyte marker) and Tuj1 (a neuronal precursor marker) were inconsistent between in vitro and in vivo studies (Fig. 4A and B), we did not pursue them in further studies. Immunofluorescence imaging for cell differentiation makers in HDAC3i/BRD4i–treated cells showed a pattern similar to that of qPCR. Nestin levels were decreased, while those of S-100β were increased (Fig. 4C and D; Supplementary Fig. S5A and S5B) after treatment with HDAC3i/BRD4i. These data were also confirmed in tumor tissues using IHC analyses (Fig. 4E and F). Treatment with HDAC3i or BRD4i alone did not cause such obvious changes.

Figure 4.

Inhibition of HDAC3 and BRD4 potently induces GSC differentiation. A and B, The mRNA level of NESTIN (stem cell marker), S-100β (astrocyte marker), Olig2 (oligodendrocyte marker), and Tuj1 (the neuronal precursor marker) from CSC2078 cells with treatment of RGFP966 and JQ1 for 48 hours cultured in differentiation and tumor tissues (n = 5 independent replicates, means ± SD of five fields). C and D, Immunofluorescent staining of NESTIN and S-100β in CSC2078 with combination therapy (n = 3 independent replicates, means ± SD of five fields). Scale bars, 20 μm. E and F, IHC staining, including NESTIN and S-100β, of tumor tissues from combination therapy (n = 6 independent replicates, means ± SD of five fields). Scale bars, 20 μm. Values, mean ± SD. One-way ANOVA was carried out to test significance.

Figure 4.

Inhibition of HDAC3 and BRD4 potently induces GSC differentiation. A and B, The mRNA level of NESTIN (stem cell marker), S-100β (astrocyte marker), Olig2 (oligodendrocyte marker), and Tuj1 (the neuronal precursor marker) from CSC2078 cells with treatment of RGFP966 and JQ1 for 48 hours cultured in differentiation and tumor tissues (n = 5 independent replicates, means ± SD of five fields). C and D, Immunofluorescent staining of NESTIN and S-100β in CSC2078 with combination therapy (n = 3 independent replicates, means ± SD of five fields). Scale bars, 20 μm. E and F, IHC staining, including NESTIN and S-100β, of tumor tissues from combination therapy (n = 6 independent replicates, means ± SD of five fields). Scale bars, 20 μm. Values, mean ± SD. One-way ANOVA was carried out to test significance.

Close modal

These results suggest that inhibition of HDAC3 and BRD4 could suppress the “stemness” of GSCs into and could promote their differentiation into astrocytic lineage.

GLI1 is a downstream target of HDAC3/BRD4 and depletion of GLI1 suppresses proliferation and promotes death in GSCs

To investigate a molecular basis for the induction of cell death, we analyzed gene expression profiles using RNA sequencing (RNA-seq) in CSC2078 cells treated for 48 hours with either HDAC3i or BRD4i alone, or their combination. These studies revealed alterations in the expression of 2,811 genes (down: 1,484; up: 1327) in BRD4i-treated cells; 2,562 genes (down: 880; up: 1,682) in HDAC3i-treated cells; 2,463 (down: 1,272; up: 1,191) in HDAC3i/BRD4i–treated cells (Supplementary Fig. S6A) and also revealed that there were 1,294 overlapping genes between HDAC3i- and HDAC3i/BRD4i–treated cells (Supplementary Fig. S6B), compared with the sham control. Among the 1,294 genes, approximately 35 genes were increased by HDAC3i, which then were significantly inhibited in the presence of BRD4i (Fig. 5A). The synergistic upregulation indices of such genes were shown in Fig. 5B. Among the highly induced 35 genes, GLI1 was scored at the highest synergy index. On the basis of these observations, we hypothesized that GLI1 may be a key promoter of GSC growth.

Figure 5.

GLI1 is a downstream target of HDAC3/BRD4, and depletion of GLI1 suppresses proliferation and promotes death in GSCs. A, RNA-seq study reveals the 35 genes, which are raised by the HDAC3 inhibitor and fell significantly after JQ1 was added in CSC2078 cells (n = 3 independent replicates). B, Synergistic index of 35 genes. C, Western blot analyses of CSC2078 cells after repressing HDAC3 and BRD4 (n = 3 independent replicates). D, The GLI1 protein expression in BRD4i-treated cells (n = 3 independent replicates). E, The protein level of CSC2078 administrated with GLI1 siRNA (n = 3 independent replicates). F and G, Neurosphere formation (F) and cell growth (G) measured by cell counting of CSC2078 with siGLI1 transfection and HDAC3i after 24, 48, and 72 hours. Values, mean ± SD. Significance of differences was tested by t test. H, FITC-Annexin V/PI staining and data analyses indicated increased apoptosis in CSC2078 cells with GLI1 and HDAC3 inhibition (n = 3 independent replicates). Scale bars, 50 μm. Values, mean ± SD. Significance of differences was tested by t test.

Figure 5.

GLI1 is a downstream target of HDAC3/BRD4, and depletion of GLI1 suppresses proliferation and promotes death in GSCs. A, RNA-seq study reveals the 35 genes, which are raised by the HDAC3 inhibitor and fell significantly after JQ1 was added in CSC2078 cells (n = 3 independent replicates). B, Synergistic index of 35 genes. C, Western blot analyses of CSC2078 cells after repressing HDAC3 and BRD4 (n = 3 independent replicates). D, The GLI1 protein expression in BRD4i-treated cells (n = 3 independent replicates). E, The protein level of CSC2078 administrated with GLI1 siRNA (n = 3 independent replicates). F and G, Neurosphere formation (F) and cell growth (G) measured by cell counting of CSC2078 with siGLI1 transfection and HDAC3i after 24, 48, and 72 hours. Values, mean ± SD. Significance of differences was tested by t test. H, FITC-Annexin V/PI staining and data analyses indicated increased apoptosis in CSC2078 cells with GLI1 and HDAC3 inhibition (n = 3 independent replicates). Scale bars, 50 μm. Values, mean ± SD. Significance of differences was tested by t test.

Close modal

Therefore, we next verified whether alterations in the expression of GLI1 occurred in GSCs treated with HDAC3i/BRD4i. GLI1 expression and STAT3 pY705 at the protein levels were blocked in GSCs treated with HDAC3i/BRD4i (Fig. 5C; Supplementary Fig. S6C), compared with those GSCs treated with HDAC3i alone. Similarly, in the presence of siBRD4/HDAC3i and BRD4i/shHDAC3, GLI1 and STAT3 activation induced by HDAC3i was strongly suppressed (Supplementary Fig. 6D and E). Furthermore, the expression of antiapoptotic proteins Mcl-1 and Bcl-2, two known downstream targets of activated STAT3, was also suppressed in the presence of HDAC3i/BRD4i. However, it was interesting that BRD4i, on its own, did not affect GLI1 expression compared with DMSO or siControl (Fig. 5D), consistent with RNA-seq results.

To verify whether GLI1 inhibition was sufficient to promote apoptosis of GSCs, we depleted the endogenous GLI1 in cells using RNAi and then treated with HDAC3i. Western blot analyses showed that GLI1-knockdown suppressed STAT3 activation and antiapoptotic protein expression (Fig. 5E; Supplementary Fig. S6F). Silencing of GLI1 alone significantly inhibited self-renewal (Fig. 5F), cell proliferation (Fig. 5G), and induced apoptosis (Fig. 5H) in GSCs. Strikingly, depletion of GLI1 was sufficient to promote sensitivity of GSCs to HDAC3i.

Taken together, these results showed that HDAC3i/BRD4i strongly blocked STAT3 activation and cell proliferation, and promoted apoptosis, owing to a suppression of GLI1 expression.

Coinhibition of BRD4 and HDAC3 represses GSC growth via GLI1/IL6/STAT3 signaling

To provide evidence that GLI1 is regulated by histone acetylation and BRD4 binding, we performed a ChIP assay in CSC2078 cells. One of the positive regulatory chromatin makers is the acetylation of histone H3 at K27 position (H3K27ac; ref. 22). Using antibodies specific for this marker, we performed a ChIP assay to detect its binding at the GLI1 promoter. The experiment process of ChIP was shown in Fig. 6A. Two distinct sets of primers were used for this assay. DNA recovered in the ChIP products was used as a template for qPCR with GLI1-specific primers. HDAC3i treatment significantly increased the deposition of H3K27ac at the GLI1 enhancer (Fig. 6B). We therefore suspected that BRD4 may be involved in GLI1 transcription in HDAC3i-treated cells. Therefore, we performed a similar ChIP assay with a BRD4-specific antibody. ChIP-qPCR showed a recruitment of BRD4 to GLI1 promoter in the presence of HDAC3i (Fig. 6C).

Figure 6.

Combined inhibition of BRD4 and HDAC3 represses GSC growth via GLI1/IL6/STAT3 signaling. A, Experiment process of ChIP. B, Histone acetylation in GLI1 promoter. CSC2078 cells were treated with RGFP966 (500 nmol/L) for 48 hours before being subjected to ChIP assay using antiacetylhistone H3K27 (Ac-H3K27) antibody followed by qPCR analyses using primers targeting indicated GLI1 promoter region (n = 3 independent replicates). Values, mean ± SD. One-way ANOVA was carried out to test significance. C, BRD4 enrichment in GLI1 promoter. CSC2078 cells were treated with RGFP966 (500 nmol/L) for 48 hours before being subjected to ChIP assay using anti-BRD4 antibody. QPCR analyses were performed using the primers targeting indicated GLI1 promoter region (n = 3 independent replicates). Values, mean ± SD. One-way ANOVA was carried out to test significance. D, The mRNA level of IL6 in CSC2078 cells with transfection siGLI1 (n = 3 independent replicates). Values, mean ± SD. Significance of differences was tested by t test. E, ChIP assay to define IL6 as the direct target of GLI1 (n = 3 independent replicates). Values, mean ± SD. One-way ANOVA was carried out to test significance. F and G, The protein level of IL6 in CSC2078 cells with transfection siGLI1 (n = 3 independent replicates). Values, mean ± SD. Significance of differences was tested by t test. H, IL6 secretion measured by ELISA with GLI1 silence (n = 5 independent replicates). Values, mean ± SD. Significance of differences was tested by t test. I, Western blot analyses of CSC2078 cells with depletion of IL6 using siRNA or RGFP966 (n = 3 independent replicates). J, Protein alteration in CSC2078 treated with siGLI1or exogenous IL6 (10 μmol/L; n = 3 independent replicates). K, Proposed working model.

Figure 6.

Combined inhibition of BRD4 and HDAC3 represses GSC growth via GLI1/IL6/STAT3 signaling. A, Experiment process of ChIP. B, Histone acetylation in GLI1 promoter. CSC2078 cells were treated with RGFP966 (500 nmol/L) for 48 hours before being subjected to ChIP assay using antiacetylhistone H3K27 (Ac-H3K27) antibody followed by qPCR analyses using primers targeting indicated GLI1 promoter region (n = 3 independent replicates). Values, mean ± SD. One-way ANOVA was carried out to test significance. C, BRD4 enrichment in GLI1 promoter. CSC2078 cells were treated with RGFP966 (500 nmol/L) for 48 hours before being subjected to ChIP assay using anti-BRD4 antibody. QPCR analyses were performed using the primers targeting indicated GLI1 promoter region (n = 3 independent replicates). Values, mean ± SD. One-way ANOVA was carried out to test significance. D, The mRNA level of IL6 in CSC2078 cells with transfection siGLI1 (n = 3 independent replicates). Values, mean ± SD. Significance of differences was tested by t test. E, ChIP assay to define IL6 as the direct target of GLI1 (n = 3 independent replicates). Values, mean ± SD. One-way ANOVA was carried out to test significance. F and G, The protein level of IL6 in CSC2078 cells with transfection siGLI1 (n = 3 independent replicates). Values, mean ± SD. Significance of differences was tested by t test. H, IL6 secretion measured by ELISA with GLI1 silence (n = 5 independent replicates). Values, mean ± SD. Significance of differences was tested by t test. I, Western blot analyses of CSC2078 cells with depletion of IL6 using siRNA or RGFP966 (n = 3 independent replicates). J, Protein alteration in CSC2078 treated with siGLI1or exogenous IL6 (10 μmol/L; n = 3 independent replicates). K, Proposed working model.

Close modal

GLI-1 is known to regulate STAT3 (23). Therefore, we next investigated the mechanism by which GLI-1 activates STAT3. We hypothesized that GLI-1 induces the expression of a cytokine that induces STAT3. IL6 is a positive regulator of Y705 phosphorylation of STAT3 (23). To examine the possible connections between GLI1, IL6, and STAT3 activation, we first examined IL6 expression in GSCs transfected with siGLI1. qPCR analyses demonstrated that IL6 transcript level was significantly downregulated upon silencing of GLI1 (Fig. 6D). ChIP assays revealed that GLI1 binding to the IL6 gene promoter was significantly reduced in the presence of siGLI1, compared withg the siControl (Fig. 6E). Consistent with these observations, silencing of GLI1 also suppressed IL6 protein expression as measured by Western blots (Fig. 6F and G). Similarly, secretion of IL6 into the culture media was downregulated as measured by an ELISA (Fig. 6H). Thus, IL6 appeared to be a direct target of GLI1. Using Western blots, we further demonstrated that depletion of IL6 using siRNA blocked STAT3 activation (Fig. 6I), compared with the control siRNA. In GLI1-depleted cells, IL6 loss accompanied with a loss of STAT3 activation, which culminated in the inhibition of the expression of antiapoptotic proteins. Conversely, treatment with IL6 not only reactivated STAT3 but also induced the antiapoptotic proteins in GLI1-depleted cells (Fig. 6J).

The poor survival rate of patients with GBM is a major clinical problem. Most therapeutics developed to date do not provide a long-lasting benefit to these patients (1, 3, 4). GSCs pose a major hurdle to the currently available therapeutic glioma maintenance, invasiveness, and recurrence. Hence, there is an urgent need for developing new and effective therapies of malignant gliomas from the perspective of GSCs.

Although the pan-HDAC inhibitors have shown to afford some antitumor effects in some solid tumors, their effectiveness is limited because of STAT3 activation or the drug-associated side effects caused by global inhibition of all HDACs affecting host processes in pancreatic ductal adenocarcinoma (12) and breast cancer, thus limiting their use (13). Hence, the expression of specific HDACs and their association with disease must be empirically determined in each disease before targeting them for inhibition in a therapeutic setting. shRNA screening library identified that HDAC3 is critical for maintaining stemness in GSCs in cell culture models. Consistent with this, among the HDACs, HDAC3 is overexpressed in gliomas and its expression is associated with growth and differentiation of GSCs. However, we have found that HDAC3i was not highly effective at inhibiting GSC growth due to its ability to activate STAT3. HDAC3 inhibitors are known to open chromatin structure and recruit an “acetyl-lysine reader” to the acetylation site to regulate downstream target gene expression (24). BRD4, as a histone acetylation reader, is required for transcriptional induction (25), and BRD4i inhibited STAT3 pY705 of GSCs in our study. Hence, we hypothesize that histone acetylation level alters STAT3 signaling and the simultaneous inhibition of HDAC3 and BRD4 will effectively block the aberrant growth by inhibiting STAT3 signaling. Indeed, this combination had robustly quelled proliferation and stemness, enforced differentiation and apoptosis in GSCs (Figs. 2 and 4), thereby increasing possibility of clinical promotion for glioma therapy. Mechanistically, HDAC3 inhibition alone sustained acetylation of H3K27, which allowed the recruitment of BRD4 to the GLI1 gene promoter and its transcription. GLI1 then induced IL6 expression. Such IL6 in an autocrine manner appears to activate STAT3 in GSCs. Thus, the combination of HDAC3i and BRD4i exerts a potent synergistic tumor-suppressive effect through an inhibition of the GLI1/IL6/STAT3 signaling axis glioma.

Recently, HDAC3 has been reported to regulate differentiation in hepatic cells and certain leukemia (20, 21). It promoted inflammatory gene expression by deacetylating NF-κB p65, in chronic obstructive pulmonary disease (26). HDAC3 inhibitor can effectively prevent deacetylation of primarily chromatin proteins, which ultimately impact cell proliferation, differentiation, and apoptosis by altering gene transcription (8, 27). However, very little is known about the role of HDAC3 in GSCs. RGFP966, the HDAC3i used in this study can cross the blood–brain barrier (28) and has been used in preclinical testing in other models of neuronal degeneration (29). Some studies reported that Coptis chinensis downregulated STAT3 phosphorylation by reducing the expression of HDAC3, which ultimately inhibited the invasion, migration, and proliferation of glioma cells (30, 31). In mouse embryonic stem cells (mESC), STAT3 pY705 induces the transcription of various antiapoptotic genes such as Mcl-1 and Bcl-2. STAT3 pS727 induces neuronal differentiation of mESCs (32). Importantly, we confirmed that STAT3 pY705 was the main mechanism for a lack of sensitivity of GSCs to HDAC3i treatment. The resistance to HDAC3i could be overcome by using BRD4 inhibitor.

GLI1 proteins participate in the Hedgehog (Hh) signaling pathways. Hh signaling pathway is composed of Hh ligands, Hh receptors (Ptch), smoothened (Smo), and GLI proteins and participated in multifarious processes including human tumorigenesis, malignancy, and metastasis (33, 34). Activation of this pathway results induces the zinc-finger transcription factors GLI1, GLI2, and GLI3. GLI1 controls the genes downstream of the Hh pathway (33, 35), which is associated with a poor prognosis in some diseases such as pancreatic and breast cancer. Indeed, induction of GLI1 by HDAC3i was consistent with a higher level H3K27ac deposition in GSCs. Such induction of GLI1 appears occur via a noncanonical pathway (36, 37). Previous studies reported that the genomic footprint of BRD4 overlaps with that of GLI1 in medulloblastomas and Hh-driven tumors are inhibited by BRD4i (25, 38), although mechanisms were not understood. In this study, we provided a direct evidence that BRD4 was recruited to the GLI1 promoter by acetylated lysine residues (in the presence of HDAC3i) and transactivated its expression.

Further investigations will be necessary to determine how GLI1 and STAT3 pathways are linked. In the medulloblastoma, GLI1 directly associates with STAT3 and regulates STAT3 activity (39). Alternatively, GLI1 could maintain the levels of activated STAT3 through the modulation of IL6 signaling, which has been demonstrated in a mouse pancreatic cancer model (40). IL6 binding to the transmembrane receptor IL6R has been shown to be a critical factor in promoting malignancy (41). Conversely, Dsg2, a desmosomal cadherin, induces STAT3 phosphorylation in basal cell carcinoma, which further increased GLI1 levels in both an autocrine and paracrine manner (42). Even so, the cross-talk between Hh/GLI1 and STAT3 pathways is relatively unknown in GSCs. The increased IL6 levels in gliomas are associated with the grade of malignancy and contribute to the maintenance of tumor heterogeneity (43, 44). The IL6/STAT3 signaling pathway promotes survival and the stemness potential of GBM (44). Therefore, we focused on the IL6 alteration. Indeed, the downregulation of GLI1 significantly lowered the expression of IL6 and the stemness of GSCs. We also confirmed that GLI1 depletion inhibited IL6 and STAT3 activation, which suggested that GLI1 acted as upstream of IL6. In summary, our studies identified a GLI1/IL6/STAT3 signaling axis as a potential target for HDC3i/BRD4i to block GSCs (Fig. 6K). Thus, targeting the GLI1/IL6/STAT3 pathway may provide therapeutic benefit by inhibiting tumor malignancy in GSCs.

No potential conflicts of interest were disclosed.

Q. Wang: Conceptualization, data curation, formal analysis, methodology, writing-original draft. S. Jia: Data curation, formal analysis. D. Wang: Data curation, formal analysis. X. Chen: Data curation. D.V. Kalvakolanu: Writing-review and editing, administrative, technical, or material support. H. Zheng: Writing-review and editing, administrative, technical, or material support. X. Wei: Formal analysis. N. Wen: Formal analysis. H. Liang: Formal analysis. B. Guo: Conceptualization, supervision, funding acquisition, methodology, writing-review and editing, administrative, technical, or material support. L. Zhang: Conceptualization, supervision, funding acquisition, writing-review and editing, administrative, technical, or material support.

The current study was funded by the Fundamental Research Funds for the Central Universities, JLU and the National Natural Science Foundation of China (grant numbers 81773217 to L. Zhang), Research Fund of Jilin Provincial Science and Technology Department (grant numbers 20190701065GH and 20200404120YY to L. Zhang), Jilin Province Health Technology Innovation Project (grant numbers 2019J030 to L. Zhang), and Chunhui International Research Project of Ministry of Education. D.V. Kalvakolanu is supported by the Cigarette Restitution Funds of the University of Maryland Greenebaum NCI Comprehensive Cancer Center. We thank Dr. Cameron Brenner at the Holland and Mellinghoff laboratories (MSKCC) for providing the human GSC lines TS543 and TS576.

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.
Ohgaki
H
,
Kleihues
P
. 
Genetic alterations and signaling pathways in the evolution of gliomas
.
Cancer Sci
2009
;
100
:
2235
41
.
2.
Zhang
X
,
Zhang
W
,
Mao
XG
,
Zhen
HN
,
Cao
WD
,
Hu
SJ
. 
Targeting role of glioma stem cells for glioblastoma multiforme
.
Curr Med Chem
2013
;
20
:
1974
84
.
3.
Aldape
K
,
Zadeh
G
,
Mansouri
S
,
Reifenberger
G
,
von Deimling
A
. 
Glioblastoma: pathology, molecular mechanisms and markers
.
Acta Neuropathol
2015
;
129
:
829
48
.
4.
Iacob
G
,
Dinca
EB
. 
Current data and strategy in glioblastoma multiforme
.
J Med Life
2009
;
2
:
386
93
.
5.
Ahmed
AU
,
Auffinger
B
,
Lesniak
MS
. 
Understanding glioma stem cells: rationale, clinical relevance and therapeutic strategies
.
Expert Rev Neurother
2013
;
13
:
545
55
.
6.
Bao
S
,
Wu
Q
,
McLendon
RE
,
Hao
Y
,
Shi
Q
,
Hjelmeland
AB
, et al
Glioma stem cells promote radioresistance by preferential activation of the DNA damage response
.
Nature
2006
;
444
:
756
60
.
7.
Polyak
K
,
Hahn
WC
. 
Roots and stems: stem cells in cancer
.
Nat Med
2006
;
12
:
296
300
.
8.
Sun
Y
,
Sun
Y
,
Yue
S
,
Wang
Y
,
Lu
F
. 
Histone deacetylase inhibitors in cancer therapy
.
Curr Top Med Chem
2018
;
18
:
2420
8
.
9.
Ropero
S
,
Esteller
M
. 
The role of histone deacetylases (HDACs) in human cancer
.
Mol Oncol
2007
;
1
:
19
25
.
10.
Bhaskara
S
,
Knutson
SK
,
Jiang
G
,
Chandrasekharan
MB
,
Wilson
AJ
,
Zheng
S
, et al
Hdac3 is essential for the maintenance of chromatin structure and genome stability
.
Cancer Cell
2010
;
18
:
436
47
.
11.
Bhaskara
S
,
Chyla
BJ
,
Amann
JM
,
Knutson
SK
,
Cortez
D
,
Sun
ZW
, et al
Deletion of histone deacetylase 3 reveals critical roles in S phase progression and DNA damage control
.
Mol Cell
2008
;
30
:
61
72
.
12.
Mazur
PK
,
Herner
A
,
Mello
SS
,
Wirth
M
,
Hausmann
S
,
Sánchez-Rivera
FJ
, et al
Combined inhibition of BET family proteins and histone deacetylases as a potential epigenetics-based therapy for pancreatic ductal adenocarcinoma
.
Nat Med
2015
;
21
:
1163
71
.
13.
Zeng
H
,
Qu
J
,
Jin
N
,
Xu
J
,
Lin
C
,
Chen
Yi
, et al
Feedback activation of leukemia inhibitory factor receptor limits response to histone deacetylase inhibitors in breast cancer
.
Cancer Cell
2016
;
30
:
459
73
.
14.
Malvaez
M
,
McQuown
SC
,
Rogge
GA
,
Astarabadi
M
,
Jacques
V
,
Carreiro
S
, et al
HDAC3-selective inhibitor enhances extinction of cocaine-seeking behavior in a persistent manner
.
Proc Natl Acad Sci U S A
2013
;
110
:
2647
52
.
15.
Wang
H
,
Huang
W
,
Liang
M
,
Shi
Y
,
Zhang
C
,
Li
Q
, et al
(+)-JQ1 attenuated LPS-induced microglial inflammation via MAPK/NFκB signaling
.
Cell Biosci
2018
;
8
:
60
.
16.
Zheng
H
,
Ying
H
,
Yan
H
,
Kimmelman
AC
,
Hiller
DJ
,
Chen
AJ
, et al
p53 and Pten control neural and glioma stem/progenitor cell renewal and differentiation
.
Nature
2008
;
455
:
1129
33
.
17.
Zuber
J
,
Shi
J
,
Wang
E
,
Rappaport
AR
,
Herrmann
H
,
Sison
EA
, et al
RNAi screen identifies Brd4 as a therapeutic target in acute myeloid leukaemia
.
Nature
2011
;
478
:
524
8
.
18.
Zhang
L
,
Gao
L
,
Zhao
L
,
Guo
B
,
Ji
K
,
Tian
Y
, et al
Intratumoral delivery and suppression of prostate tumor growth by attenuated Salmonella enterica serovar typhimurium carrying plasmid-based small interfering RNAs
.
Cancer Res
2007
;
67
:
5859
64
.
19.
Wang
Q
,
Sun
Y
,
Li
T
,
Liu
L
,
Zhao
Y
,
Li
L
, et al
Function of BRD4 in the pathogenesis of high glucoseinduced cardiac hypertrophy
.
Mol Med Rep
2019
;
19
:
499
507
.
20.
Li
S
,
Li
M
,
Liu
X
,
Yang
Y
,
Wei
Y
,
Chen
Y
, et al
Genetic and chemical screenings identify HDAC3 as a key regulator in hepatic differentiation of human pluripotent stem cells
.
Stem Cell Reports
2018
;
11
:
22
31
.
21.
Bolden
JE
,
Peart
MJ
,
Johnstone
RW
. 
Anticancer activities of histone deacetylase inhibitors
.
Nat Rev Drug Discov
2006
;
5
:
769
84
.
22.
Creyghton
MP
,
Cheng
AW
,
Welstead
GG
,
Kooistra
T
,
Carey
BW
,
Steine
EJ
, et al
Histone H3K27ac separates active from poised enhancers and predicts developmental state
.
Proc Natl Acad Sci U S A
2010
;
107
:
21931
6
.
23.
Desrivières
S
,
Kunz
C
,
Barash
I
,
Vafaizadeh
V
,
Borghouts
C
,
Groner
B
. 
The biological functions of the versatile transcription factors STAT3 and STAT5 and new strategies for their targeted inhibition
.
J Mammary Gland Biol Neoplasia
2006
;
11
:
75
87
.
24.
Musselman
CA
,
Lalonde
M-E
,
Côté
J
,
Kutateladze
TG
. 
Perceiving the epigenetic landscape through histone readers
.
Nat Struct Mol Biol
2012
;
19
:
1218
27
.
25.
Tang
Y
,
Gholamin
S
,
Schubert
S
,
Willardson
MI
,
Lee
A
,
Bandopadhayay
P
, et al
Epigenetic targeting of Hedgehog pathway transcriptional output through BET bromodomain inhibition
.
Nat Med
2014
;
20
:
732
40
.
26.
Pelzel
HR
,
Schlamp
CL
,
Waclawski
M
,
Shaw
MK
,
Nickells
RW
. 
Silencing of Fem1cR3 gene expression in the DBA/2J mouse precedes retinal ganglion cell death and is associated with histone deacetylase activity
.
Invest Ophthalmol Vis Sci
2012
;
53
:
1428
35
.
27.
Bassett
SA
,
Barnett
MP
. 
The role of dietary histone deacetylases (HDACs) inhibitors in health and disease
.
Nutrients
2014
;
6
:
4273
301
.
28.
Leus
NGJ
,
van der Wouden
PE
,
van den Bosch
T
,
Hooghiemstra
WTR
,
Ourailidou
ME
,
Kistemaker
LEM
, et al
HDAC 3-selective inhibitor RGFP966 demonstrates anti-inflammatory properties in RAW 264.7 macrophages and mouse precision-cut lung slices by attenuating NF-kappaB p65 transcriptional activity
.
Biochem Pharmacol
2016
;
108
:
58
74
.
29.
Gottesfeld
JM
,
Rusche
JR
,
Pandolfo
M
. 
Increasing frataxin gene expression with histone deacetylase inhibitors as a therapeutic approach for Friedreich's ataxia
.
J Neurochem
2013
;
126
(
Suppl 1
):
147
54
.
30.
Gupta
M
,
Han
JJ
,
Stenson
M
,
Wellik
L
,
Witzig
TE
. 
Regulation of STAT3 by histone deacetylase-3 in diffuse large B-cell lymphoma: implications for therapy
.
Leukemia
2012
;
26
:
1356
64
.
31.
Li
J
,
Ni
L
,
Li
B
,
Wang
M
,
Ding
Z
,
Xiong
C
, et al
Coptis Chinensis affects the function of glioma cells through the down-regulation of phosphorylation of STAT3 by reducing HDAC3
.
BMC Complement Altern Med
2017
;
17
:
524
.
32.
Huang
G
,
Yan
H
,
Ye
S
,
Tong
C
,
Ying
QL
. 
STAT3 phosphorylation at tyrosine 705 and serine 727 differentially regulates mouse ESC fates
.
Stem Cells
2014
;
32
:
1149
60
.
33.
Cui
Y
,
Cui
CA
,
Yang
ZT
,
Ni
WD
,
Jin
Y
,
Xuan
YH
. 
Gli1 expression in cancer stem-like cells predicts poor prognosis in patients with lung squamous cell carcinoma
.
Exp Mol Pathol
2017
;
102
:
347
53
.
34.
Mao
J
,
Ligon
KL
,
Rakhlin
EY
,
Thayer
SP
,
Bronson
RT
,
Rowitch
D
, et al
A novel somatic mouse model to survey tumorigenic potential applied to the Hedgehog pathway
.
Cancer Res
2006
;
66
:
10171
8
.
35.
Robbins
DJ
,
Fei
DL
,
Riobo
NA
. 
The Hedgehog signal transduction network
.
Sci Signal
2012
;
5
:
re6
.
36.
Colavito
SA
,
Zou
MR
,
Yan
Q
,
Nguyen
DX
,
Stern
DF
. 
Significance of glioma-associated oncogene homolog 1 (GLI1) expression in claudin-low breast cancer and crosstalk with the nuclear factor kappa-light-chain-enhancer of activated B cells (NFkappaB) pathway
.
Breast Cancer Res
2014
;
16
:
444
.
37.
Han
B
,
Qu
Y
,
Jin
Y
,
Yu
Yi
,
Deng
N
,
Wawrowsky
K
, et al
FOXC1 activates smoothened-independent hedgehog signaling in basal-like breast cancer
.
Cell Rep
2015
;
13
:
1046
58
.
38.
Lee
EY
,
Ji
H
,
Ouyang
Z
,
Zhou
B
,
Ma
W
,
Vokes
SA
, et al
Hedgehog pathway-regulated gene networks in cerebellum development and tumorigenesis
.
Proc Natl Acad Sci U S A
2010
;
107
:
9736
41
.
39.
Lim
Y
,
Gondek
L
,
Li
Li
,
Wang
Q
,
Ma
H
,
Chang
E
, et al
Integration of Hedgehog and mutant FLT3 signaling in myeloid leukemia
.
Sci Transl Med
2015
;
7
:
291ra96
.
40.
Song
Z
,
Ren
D
,
Xu
X
,
Wang
Y
. 
Molecular cross-talk of IL-6 in tumors and new progress in combined therapy
.
Thorac Cancer
2018
;
9
:
669
75
.
41.
Teoh
PJ
,
Chung
TH
,
Chng
PYZ
,
Toh
SHM
,
Chng
WJ
. 
IL6R-STAT3-ADAR1 (P150) interplay promotes oncogenicity in 1q21(amp) multiple myeloma
.
Haematologica
2020
;
105
:
1391
404
.
42.
Brennan-Crispi
DM
,
Overmiller
AM
,
Tamayo-Orrego
L
,
Marous
MR
,
Sahu
J
,
McGuinn
KP
, et al
Overexpression of desmoglein 2 in a mouse model of gorlin syndrome enhances spontaneous basal cell carcinoma formation through STAT3-mediated Gli1 expression
.
J Invest Dermatol
2019
;
139
:
300
7
.
43.
Qiu
Bo
,
Zhang
D
,
Wang
Y
,
Ou
S
,
Wang
J
,
Tao
J
, et al
Interleukin-6 is overexpressed and augments invasiveness of human glioma stem cells in vitro
.
Clin Exp Metastasis
2013
;
30
:
1009
18
.
44.
Li
Z
,
Huang
Q
,
Chen
H
,
Lin
Z
,
Zhao
M
,
Jiang
Z
. 
Interferon regulatory factor 7 promoted glioblastoma progression and stemness by modulating IL-6 expression in microglia
.
J Cancer
2017
;
8
:
207
19
.