Uncontrolled Hedgehog (Hh) signaling is the cause of several malignancies, including the pediatric cancer medulloblastoma, a neuroectodermal tumor affecting the cerebellum. Despite the development of potent Hh pathway antagonists, medulloblastoma drug resistance is still an unresolved issue that requires the identification of novel drug targets. Following up on our observation that histone deacetylase 6 (HDAC6) expression was increased in Hh-driven medulloblastoma, we found that this enzyme is essential for full Hh pathway activation. Intriguingly, these stimulatory effects of HDAC6 are partly integrated downstream of primary cilia, a known HDAC6-regulated structure. In addition, HDAC6 is also required for the complete repression of basal Hh target gene expression. These contrasting effects are mediated by HDAC6′s impact on Gli2 mRNA and GLI3 protein expression. As a result of this complex interaction with Hh signaling, global transcriptome analysis revealed that HDAC6 regulates only a subset of Smoothened- and Gli-driven genes, including all well-established Hh targets such as Ptch1 or Gli1. Importantly, medulloblastoma cell survival was severely compromised by HDAC6 inhibition in vitro and pharmacologic HDAC6 blockade strongly reduced tumor growth in an in vivo allograft model. In summary, our data describe an important role for HDAC6 in regulating the mammalian Hh pathway and encourage further studies focusing on HDAC6 as a novel drug target in medulloblastoma. Mol Cancer Ther; 14(3); 727–39. ©2014 AACR.

Medulloblastoma is a pediatric cancer affecting the cerebellum and representing the most frequent malignant brain tumor in children (1). Four molecular subtypes have been classified and one of these subgroups (which accounts for approximately one third of cases) is driven by excessive Hedgehog (Hh) signaling (2). In the developing cerebellum, Purkinje cell–derived Sonic Hh (SHH; one of three mammalian Hh ligands) serves as a potent mitogen for granule cell precursors (GCP), which transiently amplify and later constitute the internal granule cell layer (IGL) of the cerebellum. Persistent Hh signaling expands the number of GCPs in an uncontrolled manner, resulting in the formation of malignant medulloblastoma, a lethal condition if left untreated (3). Current treatment schemes using radiation, chemotherapy, and surgery lead to appreciable survival rates but are often associated with life-long impairments due to unspecific damage of the developing brain (1).

Mechanistically, the Hh ligands bind to Patched (PTCH1 or PTCH2) receptors situated in the membrane of a specialized organelle, the primary cilium, of receiving cells (4, 5). The immotile primary cilium represents a sensory structure present on most cell types and is held in place by a microtubule-based axoneme (6). Motor protein transport by intraflagellar transport (IFT) complexes along the microtubule tracks regulates the assembly, function, and the disassembly of this organelle (7). The tubulin subunits of the ciliary axoneme contain posttranslational modifications associated with long-lived microtubules, such as the acetylation of α-tubulin. An important modulator of tubulin acetylation is histone deacetylase 6 (HDAC6), a mainly cytoplasmic member of the HDAC family that uses tubulin as one of its major substrates (8, 9).

Binding of Hh ligands to PTCH1 allows for the entry of Smoothened (SMO) proteins into the cilium and the subsequent dissociation of SUFU (Suppressor of Fused; a cardinal negative regulator of Hh signaling) from GLI transcription factors (10). Specifically, free GLI2 and GLI3 are activated in the cilium and can subsequently enter the nucleus to induce target gene transcription (11, 12). The transcriptional targets include Hh pathway–specific genes, such as PTCH1 and GLI1, as well as more cell type–specific genes such as N-Myc, BMI1, or D-type Cyclins.

Recently, potent pharmacologic inhibitors targeting SMO, the key upstream bottleneck in the Hh cascade, have been introduced to the clinic (13). Although very promising first results have underscored the principle effectiveness of targeting Hh signaling in medulloblastoma, the rapid development of drug resistance has dampened the initial enthusiasm (14–16). These data encourage for the development of additional inhibitors that could be applied together with SMO inhibitors or as single-agent therapy in refractory situations. Because the major mechanisms of the development of drug resistance seems to involve SMO mutations affecting the drug binding site or the activation of SMO-bypassing downstream events (17), novel Hh pathway inhibitors would ideally target a non-SMO protein that is acting at a later step in the signaling cascade (18).

We found HDAC6 to be overexpressed in a murine model of Hh-driven medulloblastoma and were interested to see if this enzyme plays a role in the Hh pathway itself and in medulloblastoma. Our results show that HDAC6 significantly regulates Hh signaling, playing a dichotomous role: on the one hand, its enzymatic activity represses basal Hh target gene transcription; while, on the other hand, HDAC6 activity promotes the maximum expression of target genes. Using an allograft model of Hh-elicited medulloblastoma, we could show that despite this dichotomous effect of HDAC6 on the Hh pathway, specific inhibitors of this enzyme significantly reduced in vivo tumor growth. In light of the fact that the genetic knockout of Hdac6 does not show a prominent phenotype (19), selective HDAC6 antagonists represent an interesting new class of potential drugs in the treatment of medulloblastoma.

Reagents

SMO agonist SAG was purchased from Calbiochem. Trichostatin A (TSA) and SANT (SANT-1) were from Sigma. Tubacin was kindly provided by Ralph Mazitschek and Stuart Schreiber [Initiative for Chemical Genetics, National Cancer Institute (NCI), Bethesda, MD, USA] and was also purchased from Enzo Life Sciences. CAY-10603 was purchased from Biomol. ACY-1215 for cell culture experiments was purchased from Selleck Bio and larger amounts for animal studies were bought from Xcess Bio. Lists of antibodies, siRNA sequences, and quantitative PCR (qPCR) primers used in this study can be found in Supplementary Tables S1–S3, respectively.

Cell lines

NIH3T3, Hek293A, ShhL2, and C3H10T1/2 cell lines were purchased from the ATCC in 2009 and were expanded and frozen in aliquots as passage 3 to 5. Cells were used for experiments until passage 25. Cell line authentication was performed by the ATCC. All cell lines were cultured in Dulbecco's modified Eagle medium (DMEM; high glucose plus glutamine and pyruvate; Invitrogen) supplemented with 10% fetal bovine serum (FBS) and 1% penicillin/streptomycin at 37°C with 5% CO2. Immortalized wild-type (wt) and Sufu−/− mouse embryonic fibroblasts (MEF; 20) were a kind gift of Rune Toftgard (Karolinska Institute, Stockholm, Sweden). MEF[SHH], MEF[Smo*], MEF[Gli2dN], and Gli23−/− MEFs cells were kindly provided by Wade Bushman (Molecular and Environmental Toxicology Center, University of Wisconsin Medical School, Madison, WI; ref. 21). MEF lines (obtained in 2009 and frozen as aliquots) are regularly authenticated by us through functional testing (e.g., loss of Sufu in the case of Sufu−/− MEFs).

Immunoblotting

Separation of lysates by SDS–PAGE was followed by subsequent Western blot analysis. SDS–PAGE gels were blotted on Immobilon-PVDF membranes (Millipore) and incubated with the respective primary antibody, followed by a horseradish peroxidase (HRP)–coupled secondary antibody. Detection of the HRP signal was performed using Pierce ECL Western Blotting Substrate (Thermo Scientific) according to the manufacturer's protocol.

Immunohistochemistry

Immunohistochemistry on formaldehyde-fixed and paraffin-embedded tissue sections was performed using an HDAC6 antibody (1:50) from Santa Cruz Biotechnology (sc-11420).

RNA preparation, cDNA synthesis, and qPCR

Total RNA was extracted using NucleoSpin RNA II kit (Macherey-Nagel) according to the manufacturer's protocol. cDNA synthesis of 1 μg total RNA was performed using the iScript cDNA Synthesis Kit (Bio-Rad) following the manufacturer's guidelines. qPCR reactions were performed using the Absolute QPCR SYBR Green Mix (ABGene). qPCR reactions were performed on 96-well qPCR plates (ABGene) using either the Mx3000P or Mx3005P qPCR systems (Agilent). Results were calculated as relative mRNA expression (2ΔΔCt). Data were obtained from at least three independent experiments and are shown as the mean ± SD.

Luciferase reporter assays

ShhL2 cells were plated in triplicate and were grown to full confluence in solid white 96-well plates with clear bottom. Subsequently, cells were treated in full growth medium with 100 nmol/L SAG plus the indicated compounds for 48 hours. Cells were lysed in Passive Lysis Buffer (Promega) and Firefly and Renilla luciferase activity were measured using an Orion L microplate luminometer (Berthold Detection Systems) using Beetle- and Renilla-Juice reagents (both PJK, Kleinblittersdorf, Germany).

Osteogenic differentiation assay

The mesenchymal progenitor cell line C3H10T1/2 was plated in triplicates and grown to full confluence in 96-well plates. Subsequently, cells were exposed to experimental compounds in full growth medium for 4 days. Afterward, cells were lysed in Passive Lysis Buffer (Promega). A part of the lysate was used for protein quantification (Bio-Rad Protein Assay; Bio-Rad) while the remaining lysate was used for measuring alkaline phosphatase (AP) activity (Alkaline Phosphatase Blue Microwell Substrate; Sigma).

Mouse lines

The Neurod2-SmoA1 mouse line was purchased from JAX (stock number 008831) and is described previously (22).

Animal studies

Primary cells from SmoA1 medulloblastoma were obtained by mincing and trypsinization of tumor fragments. Subsequently, these cerebellum-derived cells were mixed 1:1 with Matrigel (Sigma E1270) and injected subcutaneously (s.c.) into C57BL/6J mice for amplification of the cells. After a few rounds of in vivo passaging, cells were pooled, mixed with Matrigel (one third of the volume), and injected s.c. into 15 C57BL/6J mice (3.5 months old/20 g weight). One week later, all mice had palpable s.c. tumors of approximately 25 mm2. Mice were randomly divided into three groups (5 animals each, day 0) and were injected s.c. on days 0, 1, 2, 3, 4, 7, 8, 9, 10, and 11 each with 200 μL of compound [50 mg/kg; Solvent, vismodegib (GDC-0449; LC Laboratories), and ACY-1215 (Rocilinostat; Xcess Bio (ACY)]. The injections were peritumoral with an average distance to the tumor mass of approximately 0.5 to 1 cm. Mice were sacrificed on day 12 by cervical dislocation and tumors were removed for further analysis.

Solubilization of compounds

The compounds were first dissolved in 100% DMSO at a concentration of 70 mg/mL. Then, compounds were diluted in 45% (2-hydroxypropyl)-β-cyclodextrin (Sigma; #332607) in PBS for a final concentration of 5 mg/mL (1 mg/200 μL). All animal studies were approved and were in agreement with institutional and federal state laws.

RNAi transfection and microarray

Subconfluent MEF[SHH] cells were transfected with 35 nmol/L siRNA (Dharmacon SMARTpools) on day 0 using Lipofectamine RNAiMAX (Invitrogen) as transfection reagent. On day 1 (24 hours after transfection), the cells were confluent and the siRNA solution was exchanged against full growth medium. Cells were grown for another 48 hours and were harvested for RNA preparation on day 3 (72 hours after transfection). The integrity of the isolated RNA was subsequently determined using the Experion Automated Electrophoresis System (Bio-Rad). The RNAs from two independent experiments were pooled in a 1:1 ratio. Labeling, hybridization to microarrays, and data analysis were performed by the IMT Genomics Core Facility. RNA was labeled with the two-color Quick-Amp Labeling kit (Agilent) and hybridized against Agilent-026655 microarrays. Raw microarray data were normalized using the “loess” method implemented within the marray package of R/Bioconductor. Agilent probes were reassigned to the Ensembl revision 70 genome annotations by aligning sequences with a short read aligner (Bowtie against both, the transcriptome and the genome) as described previously (23). The microarray data have been deposited at Array Express under the accession number E-MTAB-2440.

Statistical analysis

Data are shown as the mean of three independent experiments (unless otherwise indicated) ± SD. Statistical significance was calculated by applying a two-tailed Student t test (Microsoft Excel). *, P < 0.05; **, P < 0.005; ***, P< 0.0005. If not stated otherwise, the comparison is between the experimental condition and the corresponding control (DMSO, siCon).

HDAC6 is overexpressed in murine medulloblastoma

HDAC6 had been implicated in the etiology of several cancers and in primary cilium biology (9, 24–27), which prompted us to analyze the role of HDAC6 in Hh-driven medulloblastoma. In line with an earlier report (28), we found endogenous Hdac6 to be overexpressed in cerebellar tumors arising in a mouse model of Hh-dependent medulloblastoma (SmoA1 model; Fig. 1A; refs. 22, 29). In wt animals, HDAC6 protein expression can mostly be seen in the non-tumor Purkinje cells. In contrast, in the transgenic mice, granule cell–derived tumor cell nests show high HDAC6 immunoreactivity (Fig. 1B). These findings raised the question about the functional importance for HDAC6 in Hh-driven medulloblastoma.

Figure 1.

HDAC6 is overexpressed in a mouse model of medulloblastoma. A, Hdac6 mRNA expression in cerebellae from wt mice (∼4 months old; n = 9) and from medulloblastoma cerebellae (SmoA1 mice; n = 9). B, HDAC6 immunohistochemistry on paraffin-embedded sections from wt cerebellum (left) and SmoA1 medulloblastoma cerebellum (middle and right). Note that in the wt situation, the Purkinje cells (and not the granule cells in the IGL) show the greatest HDAC6 signal (brown, indicated by arrowheads), whereas in the tumor setting, the cancerous granule cell nests display the highest HDAC6 expression.

Figure 1.

HDAC6 is overexpressed in a mouse model of medulloblastoma. A, Hdac6 mRNA expression in cerebellae from wt mice (∼4 months old; n = 9) and from medulloblastoma cerebellae (SmoA1 mice; n = 9). B, HDAC6 immunohistochemistry on paraffin-embedded sections from wt cerebellum (left) and SmoA1 medulloblastoma cerebellum (middle and right). Note that in the wt situation, the Purkinje cells (and not the granule cells in the IGL) show the greatest HDAC6 signal (brown, indicated by arrowheads), whereas in the tumor setting, the cancerous granule cell nests display the highest HDAC6 expression.

Close modal

Pharmacologic HDAC6 inhibitors block active Hh signaling

To address the relevance of HDAC6 in Hh-evoked disease, we were first interested to see whether this enzyme plays a role in the Hh pathway itself. To this end, we used three structurally distinct, specific HDAC6 antagonists (Tubacin, CAY-10603, and ACY-1215), which had been described and characterized before (Fig. 2A; refs. 26, 30, 31). We treated fibroblasts with these inhibitors because they represent a good model system for the analysis of the Hh signaling cascade and are one of the major Hh-responsive cell type known. As shown in Fig. 2B, analysis of NIH3T3 fibroblasts that stably carry a Hh-responsive luciferase construct in their genome (ShhL2 cells), demonstrated that the Hh pathway induction by a synthetic agonist (SAG; (32) could be blunted by coapplication of increasing amounts of HDAC6 inhibitor. The negative effect of HDAC6 blockade on endogenous Hh signaling could be observed with all three compounds, arguing against unspecific off-target effects. To rule out negative effects of the compounds on ciliogenesis, we analyzed the presence of primary cilia in treated cells. In line with HDAC6 stabilizing rather than destabilizing cilia (9, 27), we found no significant reduction of ciliogenesis or change in cilia morphology in cells exposed to HDAC6 inhibitors, irrespective of the serum concentrations used (Supplementary Fig. S1A–S1C). Moreover, HDAC6 inhibition was not associated with gross cellular toxicity (Supplementary Fig. S2A).

Figure 2.

Targeting endogenous HDAC6 impairs Hh signaling. A, chemical structures of the HDAC6 inhibitors used in this study. B, luciferase reporter assay using ShhL2 cells. The concentrations of the respective compounds are given in [μmol/L] following the drug, for example, SANT_0.2 indicates 0.2 μmol/L of the SMO antagonist SANT-1 (positive control). C, osteogenic differentiation assay using C3H10T1/2 cells. Activating Hh signaling with SAG induces osteogenic differentiation, indicated by the blue AP staining. Treatment with HDAC6 inhibitors interferes with this process. D, quantification of the results from the osteogenic differentiation assay shown in C. The mean (± SD) of three independent biologic experiments is shown. Note that the pan-HDAC inhibitor TSA that also blocks nuclear HDAC activity results in AP induction even in the absence of SAG. E, measurement of Hh target gene expression (Gli1, Ptch1, and Ptch2) by means of qPCR in MEF[SHH] cells transfected with the indicated siRNA. RNAi targeting Gli2 was used as a positive control. *, P < 0.05; **, P < 0.005; ***, P < 0.0005. F, verification of siRNA-mediated knockdown of Hdac6 mRNA in MEF[SHH] cells. Inset, an indirect evidence of functional depletion of HDAC6 protein: increase of acetylated tubulin (AcTub) levels upon transfection with siHdac6. The expression levels of endogenous HDAC6 protein were too low to be detected in Western blotting using lysates from MEF cells (not shown).

Figure 2.

Targeting endogenous HDAC6 impairs Hh signaling. A, chemical structures of the HDAC6 inhibitors used in this study. B, luciferase reporter assay using ShhL2 cells. The concentrations of the respective compounds are given in [μmol/L] following the drug, for example, SANT_0.2 indicates 0.2 μmol/L of the SMO antagonist SANT-1 (positive control). C, osteogenic differentiation assay using C3H10T1/2 cells. Activating Hh signaling with SAG induces osteogenic differentiation, indicated by the blue AP staining. Treatment with HDAC6 inhibitors interferes with this process. D, quantification of the results from the osteogenic differentiation assay shown in C. The mean (± SD) of three independent biologic experiments is shown. Note that the pan-HDAC inhibitor TSA that also blocks nuclear HDAC activity results in AP induction even in the absence of SAG. E, measurement of Hh target gene expression (Gli1, Ptch1, and Ptch2) by means of qPCR in MEF[SHH] cells transfected with the indicated siRNA. RNAi targeting Gli2 was used as a positive control. *, P < 0.05; **, P < 0.005; ***, P < 0.0005. F, verification of siRNA-mediated knockdown of Hdac6 mRNA in MEF[SHH] cells. Inset, an indirect evidence of functional depletion of HDAC6 protein: increase of acetylated tubulin (AcTub) levels upon transfection with siHdac6. The expression levels of endogenous HDAC6 protein were too low to be detected in Western blotting using lysates from MEF cells (not shown).

Close modal

To extend our investigations, we tested the impact of HDAC6 inhibition on a physiologic Hh-driven process. Mesenchymal progenitor cells (C3H10T1/2 cells) differentiate along the osteogenic lineage upon treatment with Hh, a process that can be visualized by staining for AP activity, an osteogenic marker protein. As shown in Fig. 2C and D, the SAG-induced differentiation of C3H10T1/2 cells could be potently antagonized by application of the HDAC6 inhibitors CAY-10603 or Tubacin. Interestingly, the pan-HDAC inhibitor TSA also blocked SMO-induced cell differentiation, but in addition also resulted in an increase of basal induction (Fig. 2D). This suggests that nuclear HDAC family members repress basal Hh signaling and further underlines the specificity of the HDAC6 inhibitors used in this study. However, to demonstrate the requirement for HDAC6 in Hh signaling with a chemical compound-independent approach, we used a pool of four different RNAi constructs to selectively knockdown endogenous Hdac6 mRNA in MEFs stably transfected with SHH (MEF[SHH]; ref. 21). Transfection of these constitutively signaling cells with siHdac6 resulted in a clear reduction of expression of the established Hh target genes Gli1, Ptch1, and Ptch2; indicative of Hh pathway inhibition (Fig. 2E and F). To rule out unspecific effects by members of the siRNA pool, we tested the individual siRNAs contained in the pool and could show that three of the four RNAi sequences repressed Hh signaling, supporting the previous findings (Supplementary Fig. S2B). Furthermore, the Hdac6-specific siRNA pool did not reduce the expression levels of Hdac family members other than Hdac6 (Supplementary Fig. S2C), demonstrating that the observed effects are in fact due to selective interference with Hdac6. Moreover, RNAi against Hdac6 also significantly reduced SAG-induced C3H10T1/2 cell osteogenic differentiation (Supplementary Fig. S2D–S2F). Finally, HDAC6 action was important for Hh signaling irrespective of the serum concentration (10% FBS; see Fig. 2; 1% FBS; see Supplementary Fig. S3A) and of the nature of the ligand (see recombinant SHH instead of SAG in Supplementary Fig. S3B). Taken together, our data show that HDAC6 functions in the Hh pathway and is required to achieve full pathway activity.

Epistatic analysis of HDAC6 effects

Next, we were interested to learn at which point in the Hh transduction cascade the HDAC6 functions would be integrated. To this end, we made use of MEF cell lines that harbor activating alterations to stimulate Hh signaling at different levels of the pathway and measured the abundance of the Hh target genes Gli1, Ptch1, Ptch2, and Hip1. As shown in Fig. 3A and B, signaling in MEF[SHH] cells (which activate Hh signaling at the ligand step) could be blocked by application of different HDAC6 antagonists as demonstrated on the mRNA (Fig. 3A) and the protein level (Fig. 3B). Again, none of the used HDAC6 inhibitors significantly increased the levels of acetylated histone H3 (Fig. 3B and Supplementary Fig. S2G), in contrast to the pan-HDAC inhibitor TSA, arguing for HDAC6-selective mechanisms.

Figure 3.

Epistatic analysis of HDAC6 in the Hh cascade. Measurement of Hh target gene expression (Gli1, Ptch1, and Ptch2) by means of qPCR in cells exposed for 48 hours to different inhibitors (SANT, 0.2 μmol/L; ACY-1215, 10 μmol/L; CAY-10603, 4 μmol/L; Tubacin, 20 μmol/L; TSA, 0.5 μmol/L). A, Hh target gene expression in MEF[SHH] cells. B, Western blot analysis of protein expression in MEF[SHH] treated with the indicated compounds. Note that the acetylation of nuclear histone H3 is only increased by TSA treatment, but not by inhibition of HDAC6 with CAY-10603 or Tubacin. C, Hh target gene expression in MEF[Smo*] cells. D, Hh target gene expression in Sufu−/− MEF cells. E, Hh target gene expression in MEF[Gli2dN] cells. F, Hh reporter assay in Hek293T cells transfected with HA-GLI1 plus empty vector control (mock) or various (Flag-tagged) HDAC constructs. The Western blot analysis depicts the protein expression of the HDAC proteins in this assay. *, P < 0.05; **, P < 0.005; ***, P < 0.0005; ns, not significant.

Figure 3.

Epistatic analysis of HDAC6 in the Hh cascade. Measurement of Hh target gene expression (Gli1, Ptch1, and Ptch2) by means of qPCR in cells exposed for 48 hours to different inhibitors (SANT, 0.2 μmol/L; ACY-1215, 10 μmol/L; CAY-10603, 4 μmol/L; Tubacin, 20 μmol/L; TSA, 0.5 μmol/L). A, Hh target gene expression in MEF[SHH] cells. B, Western blot analysis of protein expression in MEF[SHH] treated with the indicated compounds. Note that the acetylation of nuclear histone H3 is only increased by TSA treatment, but not by inhibition of HDAC6 with CAY-10603 or Tubacin. C, Hh target gene expression in MEF[Smo*] cells. D, Hh target gene expression in Sufu−/− MEF cells. E, Hh target gene expression in MEF[Gli2dN] cells. F, Hh reporter assay in Hek293T cells transfected with HA-GLI1 plus empty vector control (mock) or various (Flag-tagged) HDAC constructs. The Western blot analysis depicts the protein expression of the HDAC proteins in this assay. *, P < 0.05; **, P < 0.005; ***, P < 0.0005; ns, not significant.

Close modal

Interfering with endogenous HDAC6 function also blocked Hh signaling in Ptch1−/− MEFs (Supplementary Fig. S3C) and in cells stably expressing a dominant active version of Smo (MEF[Smo*]; Fig. 3C; ref. 21). Despite the fact that inhibition could be observed in all cell lines, it appeared as if cells harboring wt Smo (i.e., MEF[SHH] and Ptch1−/− MEFs) were more potently inhibited than cells with mutant Smo (MEF[Smo*]).

Furthermore, we observed that in Ptch1−/− cells, expression of the Hh target gene Hip1 was strongly repressed only if a pan-HDAC inhibitor was used (TSA; Supplementary Fig. S3C). In contrast, SANT and the HDAC6 inhibitors only reduced Hip1 levels very modestly, indirectly pointing to HDAC6 antagonists having no potent effect on histone acetylation, as was shown before.

To investigate whether HDAC6 also plays a role downstream of Ptch1 and Smo, we used MEF cells harboring a genetic deletion of the Sufu gene, leading to strong, but ligand- and receptor-independent constitutive signaling (20, 33). Although the SMO-selective inhibitor SANT (32) was ineffective in these cells, HDAC6 inhibition by ACY-1215, CAY-10603, or Tubacin resulted in a suppression of Hh target gene expression, albeit not to the same extent as in the MEF[SHH] cells (Fig. 3D). Additional evidence for pathway inhibition in Sufu−/− cells was also found upon RNAi-mediated knockdown of Hdac6 (Supplementary Fig. S3D).

Finally, we used cells with stable expression of low levels of an activated mutant of GLI2, the transcription factor mediating the final steps of the Hh signaling cascade (MEF[Gli2dN] cells; ref. 21). As shown in Fig. 3E, HDAC6 blockade also repressed Hh target gene expression in these cells, whereas the upstream inhibitor SANT was inactive. Again, the inhibition achieved by HDAC6 blockade was not as pronounced as in cells with Hh pathway activation at the ligand step (e.g., MEF[SHH]). Because direct deacetylation by nuclear HDACs had been shown to stimulate GLI function (34), we were interested if a similar mechanism applied to HDAC6 as well. Whereas HDAC1 and HDAC2 significantly promoted the activity of transfected GLI1 in luciferase reporter assays, HDAC6 remained inactive, despite prominent protein expression (Fig. 3F). This result demonstrates that HDAC6 does apparently not act through direct deacetylation of the GLI1 transcription factor and that it most likely functions through indirect mechanisms (which were overwhelmed by the high levels of transfected GLI1 in this assay).

In summary, we conclude that HDAC6 positively regulates Hh signaling, potentially at two levels: first, at the receptor level (PTCH1/SMO), and second, at a downstream point in the cascade at the level of the transcription factors.

Global analysis of HDAC6 inhibition

After having observed the antagonistic effects of HDAC6 inhibition in specific Hh assays, we were interested to investigate the wider impact on the Hh-induced transcriptome. To this end, we treated MEF[SHH] cells with control siRNA (targeting Firefly luciferase, siLuc) or RNAi against Hdac6. As reference constructs, we used siRNA targeting Smo, Gli1, or Gli2. RNA from these cells was subjected to microarray analysis. Confirming previous data, established Hh target genes, such as Ptch2 or Hip1 (Hhip), were downregulated in all sample groups compared with the control siRNA (not shown).

To compare global expression differences between siSmo and siHdac6 transfected samples, we used a scatter plot to represent the data. On the basis of the observed gene expression changes, we classified the transcripts into eight different categories, which can be seen as color-coded spots in the scatter plot in Fig. 4A. To investigate how these genes behave in a subsequent comparison with siGli1 and siGli2 conditions, we transferred these transcripts (and their color) into siGli1 and siGli2 scatter plots (Fig. 4A, middle and right).

Figure 4.

Global analysis of gene expression changes. A, scatter plot representation depicting mRNA expression changes in MEF[SHH] cells transfected with the indicated siRNA sequences (e.g., siSmo_P indicates the pool of four different siRNAs targeting Smo). Left, comparison between siSmo and siHdac6 samples. Genes fall into eight color-coded categories depending on their change in expression. The center of the scatter plot is empty because of a defined cutoff of at least two-fold. Middle, comparison between siGli1 and siHdac6 samples. Shown are the genes of the left. Right, comparison between siGli2 and siHdac6 samples. Shown are the genes of the left. nc, no change. B, Venn diagram representation of the MEF[SHH] microarray data grouped into “Hh signature,” “Smo-regulated,” and “Hdac6-regulated” genes. By definition, the Hh signature fully overlaps with the Smo-regulated gene set. C, quantitative real-time PCR verification of expression data derived from the microarray experiment in A. MEF[SHH] cells were used.

Figure 4.

Global analysis of gene expression changes. A, scatter plot representation depicting mRNA expression changes in MEF[SHH] cells transfected with the indicated siRNA sequences (e.g., siSmo_P indicates the pool of four different siRNAs targeting Smo). Left, comparison between siSmo and siHdac6 samples. Genes fall into eight color-coded categories depending on their change in expression. The center of the scatter plot is empty because of a defined cutoff of at least two-fold. Middle, comparison between siGli1 and siHdac6 samples. Shown are the genes of the left. Right, comparison between siGli2 and siHdac6 samples. Shown are the genes of the left. nc, no change. B, Venn diagram representation of the MEF[SHH] microarray data grouped into “Hh signature,” “Smo-regulated,” and “Hdac6-regulated” genes. By definition, the Hh signature fully overlaps with the Smo-regulated gene set. C, quantitative real-time PCR verification of expression data derived from the microarray experiment in A. MEF[SHH] cells were used.

Close modal

Comparison of the expression changes induced upon siSmo and siHdac6 transfection revealed that, as expected, the well-established canonical Hh target genes (e.g., Ptch1 and Ptch2) fall into group 6 (Fig. 4A, left, purple spots) and were downregulated by both siRNAs. Not unexpectedly, these genes were also regulated in the same direction by siGli1 and siGli2 (purple and pink spots in Fig. 4A, middle and right). Interestingly, genes that were regulated in opposite directions by siSmo and siHdac6 (groups 1 and 8, orange and brown colored spots) were basically absent.

Another interesting set of genes fell into class 7 and was downregulated upon siHdac6, but was not affected by siSmo. However, a substantial fraction of these genes (red-colored spots in Fig. 4A) was shifted to the left in siGli1 and siGli2 plots, indicating that they were regulated by Gli transcription factors and by Hdac6, but not by Smo. Finally, sets of genes exist that were regulated by Smo, but not by Hdac6 (groups 4 and 5 (green and gray spots) in Fig. 4A). Intriguingly, a large fraction of these genes was not regulated by Gli1 or Gli2 either.

Given the fact that not all Smo-regulated genes were also affected by Gli1/2 (and vice versa), we defined a common set of genes termed the “Hh signature.” This gene set fulfilled the following stringent criteria: genes must have been (i) regulated in siSmo samples compared with siControl and (ii) in addition, siSmo-regulated genes had to be regulated in the same direction by either siGli1 or siGli2 transfection. This resulted in a set of genes encompassing 820 transcripts (the “Hh signature”). Comparing the Hh signature with genes regulated upon knockdown of Hdac6 revealed that 56% of the Hh signature were affected by loss of Hdac6 (Fig. 4B).

The expression data were validated by qPCR analysis of selected genes predicted to be regulated by “Gli1 and Hdac6” and by “Gli1 only.” As shown in Fig. 4C, canonical Hh targets, such as Hip1, Ptch1, and Ptch2, were all downregulated by siGli1 as well as by siHdac6 transfection in MEF[SHH] cells. In contrast, the expression of Hsd11b1, Rasl11b, and Foxj1 were selectively affected by Gli1, but not by Hdac6, demonstrating that HDAC6 affects a subset of Gli target genes, as predicted by the microarray experiment.

In conclusion, HDAC6 activity affected more than half of the genes included in the Hh signature, but it did not affect all genes in the Hh signature. Interestingly, HDAC6 impinged also on the regulation of genes, which are Smo-independent yet Gli-dependent, arguing for some degree of downstream pathway control by HDAC6.

Dichotomous impact of HDAC6 on Hh signaling

To learn more about the underlying molecular links between HDAC6 and Hh, we turned to GLI2 and GLI3, the principle downstream signal transducers in the cascade. Because HDAC6 had been implicated in protein degradation and clearance (35–41), we asked whether HDAC6 inhibition would affect the protein levels or the processing of GLI2/3. Treatment of NIH3T3 fibroblasts with the HDAC6 inhibitors ACY-1215 and CAY-10603 reduced the overall amount of full-length GLI2 protein as well as the amount of GLI3 activator (GLI3A) and repressor (GLI3R; Fig. 5A). On the mRNA level, only Gli2 expression was reduced, but Gli3 mRNA levels remained unchanged (Fig. 5B). These data suggest that HDAC6 plays a role in the stabilization of GLI3 protein and also takes part in the transcriptional control of Gli2 expression. In light of the finding that HDAC6 inhibition reduced the GLI3R form, we speculated that this could derepress Hh target gene expression in nonstimulated cells. Therefore, we investigated Gli1 expression in wt MEF cells exposed to HDAC6 inhibitors. As shown in Fig. 5C, treatment with ACY-1215 or CAY-10603 indeed weakly stimulated the Hh pathway even in the absence of any Hh ligand or SAG. The target gene expression observed could not be reversed by the coadministration of the SMO antagonist SANT, which was able to block SAG-induced signaling (Fig. 5C). In contrast to the latter finding but in line with observations made before (Figs. 2 and 3), HDAC6 inhibition also reduced the maximal target gene expression induced by SAG. Similar findings were observed when Ptch1 expression was analyzed (Supplementary Fig. S4). Moreover, the induction of Gli1 transcription by HDAC6 inhibitors could also been demonstrated on the protein level in unstimulated cells. In line with qPCR data (Fig. 5C), HDAC6 blockade led to a reduction of maximal GLI1 protein levels in SAG-stimulated cells (Fig. 5D).

Figure 5.

Molecular link between HDAC6 and Hh signaling. A, top, endogenous Gli2A (full-length Gli2) protein expression in NIH3T3 cells. Actin was used as a loading control. Bottom, endogenous Gli3A (full-length Gli3) and Gli3R (truncated repressor form) protein expression in NIH3T3 cells treated with the indicated compounds for 48 hours (ACY-1215, 10 μmol/L; CAY-10603, 4 μmol/L; SANT, 0.2 μmol/L; SAG, 0.1 μmol/L). B, Gli2 and Gli3 mRNA expression in NIH3T3 cells treated with the indicated compounds for 48 hours (ACY-1215, 10 μmol/L; CAY-10603, 4 μmol/L; SANT, 0.2 μmol/L). C, Gli1 mRNA expression in wt MEFs exposed to the depicted inhibitors (concentrations as in A). D, endogenous GLI1 protein expression in wt MEFs treated with the indicated compounds (concentrations as in A; TSA, 0.5 μmol/L). E, Gli1 mRNA expression in wt MEFs compared with the Gli1 expression in Gli23−/− MEFs, both exposed to the depicted inhibitors (concentrations as in A). For comparison, the DMSO samples were set to 1. F, Ptch1 and Ptch2 mRNA expression in Gli23−/− MEFs transfected with control siRNA or with siRNA targeting Gli1. Cells were treated with DMSO or with 10 μmol/L ACY-1215 for 48 hours. The inset shows an immunoblot verifying the Gli1 knockdown in these cells. *, P < 0.05; **, P < 0.005.

Figure 5.

Molecular link between HDAC6 and Hh signaling. A, top, endogenous Gli2A (full-length Gli2) protein expression in NIH3T3 cells. Actin was used as a loading control. Bottom, endogenous Gli3A (full-length Gli3) and Gli3R (truncated repressor form) protein expression in NIH3T3 cells treated with the indicated compounds for 48 hours (ACY-1215, 10 μmol/L; CAY-10603, 4 μmol/L; SANT, 0.2 μmol/L; SAG, 0.1 μmol/L). B, Gli2 and Gli3 mRNA expression in NIH3T3 cells treated with the indicated compounds for 48 hours (ACY-1215, 10 μmol/L; CAY-10603, 4 μmol/L; SANT, 0.2 μmol/L). C, Gli1 mRNA expression in wt MEFs exposed to the depicted inhibitors (concentrations as in A). D, endogenous GLI1 protein expression in wt MEFs treated with the indicated compounds (concentrations as in A; TSA, 0.5 μmol/L). E, Gli1 mRNA expression in wt MEFs compared with the Gli1 expression in Gli23−/− MEFs, both exposed to the depicted inhibitors (concentrations as in A). For comparison, the DMSO samples were set to 1. F, Ptch1 and Ptch2 mRNA expression in Gli23−/− MEFs transfected with control siRNA or with siRNA targeting Gli1. Cells were treated with DMSO or with 10 μmol/L ACY-1215 for 48 hours. The inset shows an immunoblot verifying the Gli1 knockdown in these cells. *, P < 0.05; **, P < 0.005.

Close modal

To delineate if the induction of Hh signaling by HDAC6 antagonists was mediated through GLI factors or through GLI-independent mechanisms, we used cells lacking functional Gli2 and Gli3 genes (double knockout Gli23−/− MEFs; ref. 21). Treating these cells with pharmacologic HDAC6 inhibitors revealed that the Gli1 induction seen in wt cells was significantly reduced in the absence of Gli2 and Gli3 (Fig. 5E), suggesting that in particular the HDAC6-mediated effects on the GLI3R protein levels might be important. However, positive effects through Gli1 might be needed for the ligand-independent Hh target gene induction as well. Therefore, we reduced endogenous Gli1 levels by RNAi-mediated knockdown of the remaining Gli1 expression in Gli23−/− cells. As shown in Fig. 5F, siGli1 transfection reduced the Hh target gene induction (Ptch1 and Ptch2) compared with siControl cells, arguing also for the requirement of GLI1 activator function (Fig. 5F).

Taken together, we found a dichotomous behavior of HDAC6 inhibitors on Hh signaling. On the one hand, HDAC6 blockade results in a suppression of maximal Hh target gene expression; while on the other hand, HDAC6 interference stimulates basal target gene transcription. We hypothesize that the first observation might be caused by the reduced Gli2 mRNA expression upon HDAC6 inhibitor exposure, whereas the latter effect most likely involves the reduced GLI3R protein in combination with GLI1 activator functions.

Pharmacologic HDAC6 inhibition has suppressive effects on in vivo medulloblastoma growth

Next, we were interested to investigate whether an HDAC6-directed approach could in principle be exploited as a future therapy option in the treatment of medulloblastoma. To address this issue, we exposed primary mouse medulloblastoma cells (MB99–1 cells derived from the SmoA1 mouse model; refs. 22, 29) to increasing concentrations of HDAC6 inhibitors in culture and measured surviving cells by the CellTiter assays (Fig. 6A–C). As shown in Fig. 6A–C, we observed a striking reduction in cell number upon 48 hours of HDAC6 inhibition that correlated well with the increase in tubulin acetylation [r2 = 0.927 (ACY-1215) and r2 = 0.989 (CAY-10603) for inhibition of cell growth vs. acetylated tubulin]. This cytotoxicity seen was not limited to mouse medulloblastoma cells and was also observed in human cells (Daoy; data not shown). Importantly and in line with our previous data, histone acetylation was not increased by the HDAC6 inhibitors used and did not correlate with growth inhibition [r2 = 0.377 (ACY-1215) and r2 = 0.686 (CAY-10603) for inhibition of cell growth vs. acetylated histone H3; Fig. 6A–C].

Figure 6.

In vivo effects of pharmacologic HDAC6 inhibition. A, immunoblot of MB99–1 cells treated with the indicated compound concentrations for 48 hours. Note that the HDAC6-selective inhibitors affect tubulin acetylation, but not histone H3 acetylation, whereas the pan-HDAC inhibitor TSA increases the acetylation of both. B and C, quantification of the results depicted in A. In addition, the figures contain data on cell growth (CellTiter assay; Promega) measured after 48 hours of incubation with the indicated compounds. D, in vivo allograft: tumor volume change relative to day 0 (volume = 100%). Shown is the mean of 5 mice. E, mean tumor weight (g) of resected allografts on day 12. F, cleaved caspase-3 immunohistochemistry (brown) on allograft tissue sections taken on day 12. SOL, solvent; VIS, vismodegib; ACY, ACY-1215. *, P < 0.05; **, P < 0.005.

Figure 6.

In vivo effects of pharmacologic HDAC6 inhibition. A, immunoblot of MB99–1 cells treated with the indicated compound concentrations for 48 hours. Note that the HDAC6-selective inhibitors affect tubulin acetylation, but not histone H3 acetylation, whereas the pan-HDAC inhibitor TSA increases the acetylation of both. B and C, quantification of the results depicted in A. In addition, the figures contain data on cell growth (CellTiter assay; Promega) measured after 48 hours of incubation with the indicated compounds. D, in vivo allograft: tumor volume change relative to day 0 (volume = 100%). Shown is the mean of 5 mice. E, mean tumor weight (g) of resected allografts on day 12. F, cleaved caspase-3 immunohistochemistry (brown) on allograft tissue sections taken on day 12. SOL, solvent; VIS, vismodegib; ACY, ACY-1215. *, P < 0.05; **, P < 0.005.

Close modal

These results prompted us to test the in vivo effects of small-molecule HDAC6 inhibition. To this end, we established s.c. allografts of primary SmoA1 medulloblastoma cells (MB99–1 cells). After tumors had reached a palpable size, mice (in groups of 5) were injected peritumoral s.c. with solvent or with 50 mg/kg of either vismodegib (GDC-0449, an FDA-approved SMO antagonist as a positive control) or ACY-1215 (see Materials and Methods for treatment scheme). Control (solvent-treated) tumors vastly increased in size over a 12-day period, whereas vismodegib- and ACY-1215-treated tumors remained small (Fig. 6D). This was also reflected in the mean tumor weight of dissected tumors taken at the end of the experiment (Fig. 6E). Notably, all drugs were well tolerated by the animals with no obvious signs of toxicity.

Immunohistologic examination of the tumor tissue revealed that in particular ACY-1215-treated tumors had high levels of cleaved caspase-3, a marker of apoptotic cell death (Fig. 6F), despite the fact that both drugs (vismodegib and ACY-1215) suppressed Hh pathway activity in the in vivo allografts, as measured by reduced Hh target gene expression (Supplementary Fig. S5).

HDAC6 belongs to the family of HDACs but mainly acts on nonhistone substrates. The enzyme can be found in the cytoplasm as well as in the nucleus of cells, depending on the differentiation status (42). In the cytosol, many substrates and interacting proteins have been described, including cortactin, Hsp90, and peroxiredoxins (43–49). One of the most prominent and best characterized protein substrate is the α-subunit of tubulin (8, 50), through which the enzyme regulates microtubule dynamics and vesicular transport (51–53). In addition, cytoplasmic functions of HDAC6 also include the formation of higher-order multiprotein complexes such as the protein-degrading aggresome as well as RNA/protein-containing stress granules (54, 55). In addition, HDAC6 is involved in the regulation of autophagy and several signal transduction cascades (39, 41, 56–59). Moreover, HDAC6 plays a role in the nucleus as a modifier of gene transcription (42, 60–62). Given the plethora of actions in which HDAC6 is involved, it is more than surprising to note that Hdac6-null animals are viable and healthy and have only a very gentle phenotype (19). It is currently not clear if the observed defects in immune response and the increased bone mineral density in these animals are linked to abnormal changes in Hh signaling, but it should be mentioned that this pathway plays a role in these processes.

In this study, we found HDAC6 to be overexpressed in Hh-induced medulloblastoma and we wondered whether this enzyme plays a functional role in the signal transduction of this pathway. In fact, HDAC6 overexpression has been observed in medulloblastoma before (28), but no link to the Hh system was made. Surprisingly, these authors saw no effect of the HDAC6 inhibitor Tubastatin on granule cell proliferation or allograft growth (28), which might be related to the relatively low concentrations used. In our hands, prominent Hh inhibition required the use of higher concentrations of HDAC6 inhibitors, indicative of the need to fully block HDAC6 to affect the Hh system. Despite the use of higher inhibitor concentrations, the compounds selectively inhibited HDAC6 and displayed no prominent effect on histone acetylation levels. We should point out that an allograft system always suffers from its artificial nature and that direct compound injections can potentially lead to high local inhibitor concentrations, although we tried to circumvent this issue by changing the injection sites and by not injecting directly into the tumor. It is worth noting that newer HDAC6 inhibitors are able to penetrate the blood–brain barrier and can therefore be tested under more physiologic conditions in autochthonous medulloblastoma mouse models in the future (63). Interestingly, these newer HDAC6 inhibitors have been shown to possess antidepressive characteristics, a phenomenon that we have linked to Hh inhibition before (64).

Collectively, our data revealed that endogenous HDAC6 exerts dual and opposing roles in Hh signaling: (i) HDAC6 is required for full pathway activation, while (ii) basal Hh signaling in the absence of ligands uses HDAC6 for full repression of target gene expression. This complex interplay between positive and negative actions might explain the limited phenotype in Hdac6 knockout animals. Furthermore, an induction of ligand-independent Hh signaling was not observed in the differentiation assays using C3H10T1/2 cells or in reporter assays with ShhL2 cells, suggesting either cell-type differences or that the HDAC6-induced derepression of Hh target genes is not sufficiently strong enough for certain biologic processes to occur. However, it is noteworthy that in our in vivo allograft model, mice receiving the SMO antagonist vismodegib presented with hair loss at the cutaneous injection sites (Supplementary Fig. S6). This is not unexpected as Hh signaling is well known to promote hair growth. The alopecia seen in vismodegib-treated mice was not observed in the ACY-1215 animal cohort, despite the fact that the Hh pathway was blocked. Our microarray analysis revealed that approximately half of the SMO/GLI–regulated genes are affected by HDAC6 and one hypothetical reason for the lack of alopecia might be that HDAC6 does not affect the Hh target genes required in hair cell biology. This observation warrants further investigation as it could be used to circumvent unwanted side effects associated with blocking Hh-dependent physiologic processes. In addition, an unexplored question as of now is whether HDAC6 inhibition affects SMO-dependent, but GLI-independent noncanonical signaling (65, 66).

We hypothesize that the impact of HDAC6 on Hh signaling could be linked to its effect on tubulin acetylation and is not mediated by direct deacetylation of GLI proteins through HDAC6. Posttranslational modifications of tubulin have been shown to affect motor protein-driven transport along microtubule. As Hh signaling critically requires IFT transport toward and from primary cilia (67–69), interfering with these transport processes might, in turn, suppress the precisely coordinated Hh signal transduction. As such, prolonged increased tubulin acetylation might functionally (not morphologically) resemble cilia defects such as those typically induced by loss-of-function of IFT components. In fact, certain HDAC6-related aspects of our data presented here, such as the reduced GLI3R levels and the ligand-independent pathway activation, are in agreement with previous reports on cilia defects (70–74). The fact that HDAC6 inhibition negatively affects the Hh pathway in Sufu/ cells, which signal cilium independently (75, 76), suggests that other intracellular transport processes might be required downstream of SMO and primary cilia.

An alternative scenario would involve the nuclear HDAC6 fraction that might impinge on transcriptional complexes governing Hh target gene expression, such as Gli2 transcription. Importantly, we could demonstrate the HDAC6 specificity of our reagents in several instances, and thus exclude the role of histone-directed nuclear members of the HDAC family, which also modulate Hh signaling (34).

Interestingly, recent work implicates cilia-like processes at the immunologic synapse (77), a cellular structure that had previously been linked to HDAC6 (78). However, further investigations are needed to elucidate a function for HDAC6 in immunologic synapse-triggered Hh signaling.

HDAC6 has been shown to possess oncogenic traits in several tumors (25, 79, 80). In light of these findings and the requirement for HDAC6 to achieve maximal Hh signaling, we evaluated the impact of an HDAC6-directed therapy in an in vivo allograft model of medulloblastoma. HDAC6 inhibition had striking effects on medulloblastoma cell survival in vitro and was as effective as the positive control vismodegib in vivo. In light of the fact that ACY-1215 (but not vismodegib) induced strong and widespread apoptosis, longer treatment times might even induce complete tumor eradication.

In summary, we could identify an important function of HDAC6 in regulating mammalian Hh signaling. This provides an interesting novel drug target for the treatment of Hh-driven malignancies.

No potential conflicts of interest were disclosed.

Conception and design: M. Lauth

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): P.S. Holz, V. Fendrich

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): P.S. Holz, F. Finkernagel, M. Lauth

Writing, review, and/or revision of the manuscript: P.S. Holz, F. Finkernagel, M. Lauth

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): P.S. Holz, V. Fendrich

Study supervision: M. Lauth

Other (performed experiments): P.K. Dhanyamraju

The authors thank Julia Dick for expert assistance in animal work and Aninja Baier for help with histology.

This work was supported by grants obtained from the German Research Foundation (DFG LA2829/1-1 and LA2829/6-1, to M. Lauth), the University Medical Center Giessen and Marburg (UKGM, to M. Lauth), and the Head–Neck Tumor Research Foundation (to M. Lauth).

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.
Bourdeaut
F
,
Miquel
C
,
Alapetite
C
,
Roujeau
T
,
Doz
F
. 
Medulloblastomas: update on a heterogeneous disease
.
Curr Opin Oncol
2011
;
23
:
630
7
.
2.
Northcott
PA
,
Jones
DT
,
Kool
M
,
Robinson
GW
,
Gilbertson
RJ
,
Cho
YJ
, et al
Medulloblastomics: the end of the beginning
.
Nat Rev Cancer
2012
;
12
:
818
34
.
3.
Kool
M
,
Jones
DT
,
Jager
N
,
Northcott
PA
,
Pugh
TJ
,
Hovestadt
V
, et al
Genome sequencing of SHH medulloblastoma predicts genotype-related response to smoothened inhibition
.
Cancer Cell
2014
;
25
:
393
405
.
4.
Rohatgi
R
,
Milenkovic
L
,
Scott
MP
. 
Patched1 regulates hedgehog signaling at the primary cilium
.
Science
2007
;
317
:
372
6
.
5.
Nozawa
YI
,
Lin
C
,
Chuang
PT
. 
Hedgehog signaling from the primary cilium to the nucleus: an emerging picture of ciliary localization, trafficking and transduction
.
Curr Opin Genet Dev
2013
;
23
:
429
37
.
6.
Kim
S
,
Dynlacht
BD
. 
Assembling a primary cilium
.
Current opinion in cell biology
2013
;
25
:
506
11
.
7.
Liem
KF
 Jr
,
Ashe
A
,
He
M
,
Satir
P
,
Moran
J
,
Beier
D
, et al
The IFT-A complex regulates Shh signaling through cilia structure and membrane protein trafficking
.
J Cell Biol
2012
;
197
:
789
800
.
8.
Hubbert
C
,
Guardiola
A
,
Shao
R
,
Kawaguchi
Y
,
Ito
A
,
Nixon
A
, et al
HDAC6 is a microtubule-associated deacetylase
.
Nature
2002
;
417
:
455
8
.
9.
Pugacheva
EN
,
Jablonski
SA
,
Hartman
TR
,
Henske
EP
,
Golemis
EA
. 
HEF1-dependent Aurora A activation induces disassembly of the primary cilium
.
Cell
2007
;
129
:
1351
63
.
10.
Tukachinsky
H
,
Lopez
LV
,
Salic
A
. 
A mechanism for vertebrate Hedgehog signaling: recruitment to cilia and dissociation of SuFu–Gli protein complexes
.
J Cell Biol
2010
;
191
:
415
28
.
11.
Wen
X
,
Lai
CK
,
Evangelista
M
,
Hongo
JA
,
de Sauvage
FJ
,
Scales
SJ
. 
Kinetics of hedgehog-dependent full-length Gli3 accumulation in primary cilia and subsequent degradation
.
Mol Cell Biol
2010
;
30
:
1910
22
.
12.
Kim
J
,
Kato
M
,
Beachy
PA
. 
Gli2 trafficking links Hedgehog-dependent activation of Smoothened in the primary cilium to transcriptional activation in the nucleus
.
Proc Natl Acad Sci U S A
2009
;
106
:
21666
71
.
13.
Low
JA
,
de Sauvage
FJ
. 
Clinical experience with Hedgehog pathway inhibitors
.
J Clin Oncol
2010
;
28
:
5321
6
.
14.
Metcalfe
C
,
de Sauvage
FJ
. 
Hedgehog fights back: mechanisms of acquired resistance against Smoothened antagonists
.
Cancer Res
2011
;
71
:
5057
61
.
15.
Rudin
CM
,
Hann
CL
,
Laterra
J
,
Yauch
RL
,
Callahan
CA
,
Fu
L
, et al
Treatment of medulloblastoma with hedgehog pathway inhibitor GDC-0449
.
N Engl J Med
2009
;
361
:
1173
8
.
16.
Von Hoff
DD
,
LoRusso
PM
,
Rudin
CM
,
Reddy
JC
,
Yauch
RL
,
Tibes
R
, et al
Inhibition of the hedgehog pathway in advanced basal-cell carcinoma
.
N Engl J Med
2009
;
361
:
1164
72
.
17.
Yauch
RL
,
Dijkgraaf
GJ
,
Alicke
B
,
Januario
T
,
Ahn
CP
,
Holcomb
T
, et al
Smoothened mutation confers resistance to a Hedgehog pathway inhibitor in medulloblastoma
.
Science
2009
;
326
:
572
4
.
18.
Buonamici
S
,
Williams
J
,
Morrissey
M
,
Wang
A
,
Guo
R
,
Vattay
A
, et al
Interfering with resistance to smoothened antagonists by inhibition of the PI3K pathway in medulloblastoma
.
Sci Transl Med
2010
;
2
:
51ra70
.
19.
Zhang
Y
,
Kwon
S
,
Yamaguchi
T
,
Cubizolles
F
,
Rousseaux
S
,
Kneissel
M
, et al
Mice lacking histone deacetylase 6 have hyperacetylated tubulin but are viable and develop normally
.
Mol Cell Biol
2008
;
28
:
1688
701
.
20.
Svard
J
,
Heby-Henricson
K
,
Persson-Lek
M
,
Rozell
B
,
Lauth
M
,
Bergstrom
A
, et al
Genetic elimination of Suppressor of fused reveals an essential repressor function in the mammalian Hedgehog signaling pathway
.
Dev Cell
2006
;
10
:
187
97
.
21.
Lipinski
RJ
,
Bijlsma
MF
,
Gipp
JJ
,
Podhaizer
DJ
,
Bushman
W
. 
Establishment and characterization of immortalized Gli-null mouse embryonic fibroblast cell lines
.
BMC Cell Biol
2008
;
9
:
49
.
22.
Hallahan
AR
,
Pritchard
JI
,
Hansen
S
,
Benson
M
,
Stoeck
J
,
Hatton
BA
, et al
The SmoA1 mouse model reveals that notch signaling is critical for the growth and survival of sonic hedgehog-induced medulloblastomas
.
Cancer Res
2004
;
64
:
7794
800
.
23.
Adhikary
T
,
Kaddatz
K
,
Finkernagel
F
,
Schonbauer
A
,
Meissner
W
,
Scharfe
M
, et al
Genomewide analyses define different modes of transcriptional regulation by peroxisome proliferator-activated receptor-beta/delta (PPARbeta/delta)
.
PLoS ONE
2011
;
6
:
e16344
.
24.
Aldana-Masangkay
GI
,
Sakamoto
KM
. 
The role of HDAC6 in cancer
.
J Biomed Biotechnol
2010
;
2011
:
875824
.
25.
Lee
YS
,
Lim
KH
,
Guo
X
,
Kawaguchi
Y
,
Gao
Y
,
Barrientos
T
, et al
The cytoplasmic deacetylase HDAC6 is required for efficient oncogenic tumorigenesis
.
Cancer Res
2008
;
68
:
7561
9
.
26.
Santo
L
,
Hideshima
T
,
Kung
AL
,
Tseng
JC
,
Tamang
D
,
Yang
M
, et al
Preclinical activity, pharmacodynamic, and pharmacokinetic properties of a selective HDAC6 inhibitor, ACY-1215, in combination with bortezomib in multiple myeloma
.
Blood
2012
;
119
:
2579
89
.
27.
Jacob
LS
,
Wu
X
,
Dodge
ME
,
Fan
CW
,
Kulak
O
,
Chen
B
, et al
Genome-wide RNAi screen reveals disease-associated genes that are common to Hedgehog and Wnt signaling
.
Sci Signal
2011
;
4
:
ra4
.
28.
Lee
SJ
,
Lindsey
S
,
Graves
B
,
Yoo
S
,
Olson
JM
,
Langhans
SA
. 
Sonic hedgehog-induced histone deacetylase activation is required for cerebellar granule precursor hyperplasia in medulloblastoma
.
PLoS ONE
2013
;
8
:
e71455
.
29.
Hatton
BA
,
Villavicencio
EH
,
Tsuchiya
KD
,
Pritchard
JI
,
Ditzler
S
,
Pullar
B
, et al
The Smo/Smo model: hedgehog-induced medulloblastoma with 90% incidence and leptomeningeal spread
.
Cancer Res
2008
;
68
:
1768
76
.
30.
Haggarty
SJ
,
Koeller
KM
,
Wong
JC
,
Grozinger
CM
,
Schreiber
SL
. 
Domain-selective small-molecule inhibitor of histone deacetylase 6 (HDAC6)-mediated tubulin deacetylation
.
Proc Natl Acad Sci U S A
2003
;
100
:
4389
94
.
31.
Kozikowski
AP
,
Tapadar
S
,
Luchini
DN
,
Kim
KH
,
Billadeau
DD
. 
Use of the nitrile oxide cycloaddition (NOC) reaction for molecular probe generation: a new class of enzyme selective histone deacetylase inhibitors (HDACIs) showing picomolar activity at HDAC6
.
J Med Chem
2008
;
51
:
4370
3
.
32.
Chen
JK
,
Taipale
J
,
Young
KE
,
Maiti
T
,
Beachy
PA
. 
Small molecule modulation of Smoothened activity
.
Proc Natl Acad Sci U S A
2002
;
99
:
14071
6
.
33.
Cooper
AF
,
Yu
KP
,
Brueckner
M
,
Brailey
LL
,
Johnson
L
,
McGrath
JM
, et al
Cardiac and CNS defects in a mouse with targeted disruption of suppressor of fused
.
Development
2005
;
132
:
4407
17
.
34.
Canettieri
G
,
Di Marcotullio
L
,
Greco
A
,
Coni
S
,
Antonucci
L
,
Infante
P
, et al
Histone deacetylase and Cullin3-REN(KCTD11) ubiquitin ligase interplay regulates Hedgehog signalling through Gli acetylation
.
Nat Cell Biol
2010
;
12
:
132
42
.
35.
Boyault
C
,
Zhang
Y
,
Fritah
S
,
Caron
C
,
Gilquin
B
,
Kwon
SH
, et al
HDAC6 controls major cell response pathways to cytotoxic accumulation of protein aggregates
.
Genes Dev
2007
;
21
:
2172
81
.
36.
Bazzaro
M
,
Lin
Z
,
Santillan
A
,
Lee
MK
,
Wang
MC
,
Chan
KC
, et al
Ubiquitin proteasome system stress underlies synergistic killing of ovarian cancer cells by bortezomib and a novel HDAC6 inhibitor
.
Clin Cancer Res
2008
;
14
:
7340
7
.
37.
Boyault
C
,
Gilquin
B
,
Zhang
Y
,
Rybin
V
,
Garman
E
,
Meyer-Klaucke
W
, et al
HDAC6-p97/VCP controlled polyubiquitin chain turnover
.
Embo J
2006
;
25
:
3357
66
.
38.
Hebron
ML
,
Lonskaya
I
,
Sharpe
K
,
Weerasinghe
PP
,
Algarzae
NK
,
Shekoyan
AR
, et al
Parkin ubiquitinates Tar-DNA binding protein-43 (TDP-43) and promotes its cytosolic accumulation via interaction with histone deacetylase 6 (HDAC6)
.
J Biol Chem
2013
;
288
:
4103
15
.
39.
Lee
JY
,
Koga
H
,
Kawaguchi
Y
,
Tang
W
,
Wong
E
,
Gao
YS
, et al
HDAC6 controls autophagosome maturation essential for ubiquitin-selective quality-control autophagy
.
Embo J
2010
;
29
:
969
80
.
40.
Pandey
UB
,
Batlevi
Y
,
Baehrecke
EH
,
Taylor
JP
. 
HDAC6 at the intersection of autophagy, the ubiquitin-proteasome system and neurodegeneration
.
Autophagy
2007
;
3
:
643
5
.
41.
Pandey
UB
,
Nie
Z
,
Batlevi
Y
,
McCray
BA
,
Ritson
GP
,
Nedelsky
NB
, et al
HDAC6 rescues neurodegeneration and provides an essential link between autophagy and the UPS
.
Nature
2007
;
447
:
859
63
.
42.
Chen
PB
,
Hung
JH
,
Hickman
TL
,
Coles
AH
,
Carey
JF
,
Weng
Z
, et al
Hdac6 regulates Tip60-p400 function in stem cells
.
Elife
2013
;
2
:
e01557
.
43.
Kovacs
JJ
,
Murphy
PJ
,
Gaillard
S
,
Zhao
X
,
Wu
JT
,
Nicchitta
CV
, et al
HDAC6 regulates Hsp90 acetylation and chaperone-dependent activation of glucocorticoid receptor
.
Mol Cell
2005
;
18
:
601
7
.
44.
Matthias
P
,
Yoshida
M
,
Khochbin
S
. 
HDAC6 a new cellular stress surveillance factor
.
Cell Cycle
2008
;
7
:
7
10
.
45.
Valenzuela-Fernandez
A
,
Cabrero
JR
,
Serrador
JM
,
Sanchez-Madrid
F
. 
HDAC6: a key regulator of cytoskeleton, cell migration and cell-cell interactions
.
Trends Cell Biol
2008
;
18
:
291
7
.
46.
Parmigiani
RB
,
Xu
WS
,
Venta-Perez
G
,
Erdjument-Bromage
H
,
Yaneva
M
,
Tempst
P
, et al
HDAC6 is a specific deacetylase of peroxiredoxins and is involved in redox regulation
.
Proc Natl Acad Sci U S A
2008
;
105
:
9633
8
.
47.
Zhang
X
,
Yuan
Z
,
Zhang
Y
,
Yong
S
,
Salas-Burgos
A
,
Koomen
J
, et al
HDAC6 modulates cell motility by altering the acetylation level of cortactin
.
Mol Cell
2007
;
27
:
197
213
.
48.
Wilson-Edell
KA
,
Kehasse
A
,
Scott
GK
,
Yau
C
,
Rothschild
DE
,
Schilling
B
, et al
RPL24: a potential therapeutic target whose depletion or acetylation inhibits polysome assembly and cancer cell growth
.
Oncotarget
2014
;
5
:
5165
76
.
49.
Kamemura
K
,
Ogawa
M
,
Ohkubo
S
,
Ohtsuka
Y
,
Shitara
Y
,
Komiya
T
, et al
Depression of mitochondrial metabolism by downregulation of cytoplasmic deacetylase, HDAC6
.
FEBS Lett
2012
;
586
:
1379
83
.
50.
Zhang
Y
,
Li
N
,
Caron
C
,
Matthias
G
,
Hess
D
,
Khochbin
S
, et al
HDAC-6 interacts with and deacetylates tubulin and microtubules in vivo
.
Embo J
2003
;
22
:
1168
79
.
51.
Dompierre
JP
,
Godin
JD
,
Charrin
BC
,
Cordelieres
FP
,
King
SJ
,
Humbert
S
, et al
Histone deacetylase 6 inhibition compensates for the transport deficit in Huntington's disease by increasing tubulin acetylation
.
J Neurosci
2007
;
27
:
3571
83
.
52.
Zilberman
Y
,
Ballestrem
C
,
Carramusa
L
,
Mazitschek
R
,
Khochbin
S
,
Bershadsky
A
. 
Regulation of microtubule dynamics by inhibition of the tubulin deacetylase HDAC6
.
J Cell Sci
2009
;
122
:
3531
41
.
53.
Chen
S
,
Owens
GC
,
Makarenkova
H
,
Edelman
DB
. 
HDAC6 regulates mitochondrial transport in hippocampal neurons
.
PLoS ONE
2010
;
5
:
e10848
.
54.
Kwon
S
,
Zhang
Y
,
Matthias
P
. 
The deacetylase HDAC6 is a novel critical component of stress granules involved in the stress response
.
Genes Dev
2007
;
21
:
3381
94
.
55.
Kawaguchi
Y
,
Kovacs
JJ
,
McLaurin
A
,
Vance
JM
,
Ito
A
,
Yao
TP
. 
The deacetylase HDAC6 regulates aggresome formation and cell viability in response to misfolded protein stress
.
Cell
2003
;
115
:
727
38
.
56.
Deribe
YL
,
Wild
P
,
Chandrashaker
A
,
Curak
J
,
Schmidt
MH
,
Kalaidzidis
Y
, et al
Regulation of epidermal growth factor receptor trafficking by lysine deacetylase HDAC6
.
Sci Signal
2009
;
2
:
ra84
.
57.
Spange
S
,
Wagner
T
,
Heinzel
T
,
Kramer
OH
. 
Acetylation of non-histone proteins modulates cellular signalling at multiple levels
.
Int J Biochem Cell Biol
2009
;
41
:
185
98
.
58.
Yang
MH
,
Laurent
G
,
Bause
AS
,
Spang
R
,
German
N
,
Haigis
MC
, et al
HDAC6 and SIRT2 regulate the acetylation state and oncogenic activity of mutant K-RAS
.
Mol Cancer Res
2013
;
11
:
1072
7
.
59.
Shan
B
,
Yao
TP
,
Nguyen
HT
,
Zhuo
Y
,
Levy
DR
,
Klingsberg
RC
, et al
Requirement of HDAC6 for transforming growth factor-beta1-induced epithelial-mesenchymal transition
.
J Biol Chem
2008
;
283
:
21065
73
.
60.
Palijan
A
,
Fernandes
I
,
Bastien
Y
,
Tang
L
,
Verway
M
,
Kourelis
M
, et al
Function of histone deacetylase 6 as a cofactor of nuclear receptor coregulator LCoR
.
J Biol Chem
2009
;
284
:
30264
74
.
61.
Ozaki
T
,
Wu
D
,
Sugimoto
H
,
Nagase
H
,
Nakagawara
A
. 
Runt-related transcription factor 2 (RUNX2) inhibits p53-dependent apoptosis through the collaboration with HDAC6 in response to DNA damage
.
Cell Death Dis
2013
;
4
:
e610
.
62.
Westendorf
JJ
,
Zaidi
SK
,
Cascino
JE
,
Kahler
R
,
van Wijnen
AJ
,
Lian
JB
, et al
Runx2 (Cbfa1, AML-3) interacts with histone deacetylase 6 and represses the p21(CIP1/WAF1) promoter
.
Mol Cell Biol
2002
;
22
:
7982
92
.
63.
Jochems
J
,
Boulden
J
,
Lee
BG
,
Blendy
JA
,
Jarpe
M
,
Mazitschek
R
, et al
Antidepressant-like properties of novel HDAC6-selective inhibitors with improved brain bioavailability
.
Neuropsychopharmacology
2014
;
39
:
389
400
.
64.
Lauth
M
,
Rohnalter
V
,
Bergstrom
A
,
Kooshesh
M
,
Svenningsson
P
,
Toftgard
R
. 
Antipsychotic drugs regulate hedgehog signaling by modulation of 7-dehydrocholesterol reductase levels
.
Mol Pharmacol
2010
;
78
:
486
96
.
65.
Bijlsma
MF
,
Damhofer
H
,
Roelink
H
. 
Hedgehog-stimulated chemotaxis is mediated by smoothened located outside the primary cilium
.
Sci Signal
2012
;
5
:
ra60
.
66.
Chinchilla
P
,
Xiao
L
,
Kazanietz
MG
,
Riobo
NA
. 
Hedgehog proteins activate pro-angiogenic responses in endothelial cells through non-canonical signaling pathways
.
Cell Cycle
2010
;
9
:
570
79
.
67.
Dorn
KV
,
Hughes
CE
,
Rohatgi
R
. 
A Smoothened-Evc2 complex transduces the Hedgehog signal at primary cilia
.
Dev Cell
2012
;
23
:
823
35
.
68.
Corbit
KC
,
Aanstad
P
,
Singla
V
,
Norman
AR
,
Stainier
DY
,
Reiter
JF
. 
Vertebrate Smoothened functions at the primary cilium
.
Nature
2005
;
437
:
1018
21
.
69.
Huangfu
D
,
Liu
A
,
Rakeman
AS
,
Murcia
NS
,
Niswander
L
,
Anderson
KV
. 
Hedgehog signalling in the mouse requires intraflagellar transport proteins
.
Nature
2003
;
426
:
83
7
.
70.
Han
YG
,
Kim
HJ
,
Dlugosz
AA
,
Ellison
DW
,
Gilbertson
RJ
,
Alvarez-Buylla
A
. 
Dual and opposing roles of primary cilia in medulloblastoma development
.
Nat Med
2009
;
15
:
1062
5
.
71.
Cervantes
S
,
Lau
J
,
Cano
DA
,
Borromeo-Austin
C
,
Hebrok
M
. 
Primary cilia regulate Gli/hedgehog activation in pancreas
.
Proc Natl Acad Sci U S A
2010
;
107
:
10109
14
.
72.
Ho
L
,
Ali
SA
,
Al-Jazrawe
M
,
Kandel
R
,
Wunder
JS
,
Alman
BA
. 
Primary cilia attenuate hedgehog signalling in neoplastic chondrocytes
.
Oncogene
2013
;
32
:
5388
96
.
73.
Haycraft
CJ
,
Banizs
B
,
Aydin-Son
Y
,
Zhang
Q
,
Michaud
EJ
,
Yoder
BK
. 
Gli2 and Gli3 localize to cilia and require the intraflagellar transport protein polaris for processing and function
.
PLoS Genet
2005
;
1
:
e53
.
74.
Wong
SY
,
Seol
AD
,
So
PL
,
Ermilov
AN
,
Bichakjian
CK
,
Epstein
EH
 Jr
, et al
Primary cilia can both mediate and suppress Hedgehog pathway-dependent tumorigenesis
.
Nat Med
2009
;
15
:
1055
61
.
75.
Jia
J
,
Kolterud
A
,
Zeng
H
,
Hoover
A
,
Teglund
S
,
Toftgard
R
, et al
Suppressor of Fused inhibits mammalian Hedgehog signaling in the absence of cilia
.
Dev Biol
2009
;
330
:
452
60
.
76.
Chen
MH
,
Wilson
CW
,
Li
YJ
,
Law
KK
,
Lu
CS
,
Gacayan
R
, et al
Cilium-independent regulation of Gli protein function by Sufu in Hedgehog signaling is evolutionarily conserved
.
Genes Dev
2009
;
23
:
1910
28
.
77.
de la Roche
M
,
Ritter
AT
,
Angus
KL
,
Dinsmore
C
,
Earnshaw
CH
,
Reiter
JF
, et al
Hedgehog signaling controls T cell killing at the immunological synapse
.
Science
2013
;
342
:
1247
50
.
78.
Serrador
JM
,
Cabrero
JR
,
Sancho
D
,
Mittelbrunn
M
,
Urzainqui
A
,
Sanchez-Madrid
F
. 
HDAC6 deacetylase activity links the tubulin cytoskeleton with immune synapse organization
.
Immunity
2004
;
20
:
417
28
.
79.
Aldana-Masangkay
GI
,
Rodriguez-Gonzalez
A
,
Lin
T
,
Ikeda
AK
,
Hsieh
YT
,
Kim
YM
, et al
Tubacin suppresses proliferation and induces apoptosis of acute lymphoblastic leukemia cells
.
Leuk Lymphoma
2011
;
52
:
1544
55
.
80.
Gradilone
SA
,
Radtke
BN
,
Bogert
PS
,
Huang
BQ
,
Gajdos
GB
,
LaRusso
NF
. 
HDAC6 inhibition restores ciliary expression and decreases tumor growth
.
Cancer Res
2013
;
73
:
2259
70
.