Open Access

Inhibition of the CyclinD1 promoter in response to sonic hedgehog signaling pathway transduction is mediated by Gli1

  • Authors:
    • Zhongxiao Lin
    • Hansong Sheng
    • Chaoguo You
    • Ming Cai
    • Yiping Zhang
    • Li Sheng Yu
    • Xiaoming Yu
    • Jian Lin
    • Nu Zhang
  • View Affiliations

  • Published online on: December 13, 2016     https://doi.org/10.3892/etm.2016.3969
  • Pages: 307-314
  • Copyright: © Lin et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Medulloblastoma (MB) is the most common malignant tumor of the central nervous system in children. Accumulating evidence suggests a major role for the activation of the sonic hedgehog (SHH) signaling pathway in the development of MB cells; however, the mechanisms underlying the effect of this pathway on tumor survival and growth remain poorly understood. The Gli family zinc finger 1 (Gli1) transcription factor is considered as a mediator of the SHH signaling pathway in MB cells. Therefore, the present study investigated whether the SHH signaling pathway promotes the apoptosis of MB cells via downregulation of Gli1. GANT61, a novel Gli1 inhibitor, is known to have an in vitro activity against tumors. In the current study, Daoy cells were treated with different concentrations of GANT61 for 24 h, and the effect on cell proliferation was assayed by cell counting kit‑8 assay. In addition, the cell cycle progression and apoptosis were assayed by flow cytometry analysis and hematoxylin-eosin (HE) staining. The effects of GANT61 treatment on SHH signaling pathway at the mRNA level were assayed by polymerase chain reaction (PCR). To further elucidate the inhibitory effects of GANT61 on the expression of Gli1 and CyclinD1, their protein levels were examined by western blot and immunofluorescence. The results indicated that GANT61 significantly inhibited the proliferation of Daoy cells in a dose‑dependent manner, compared with the control group (P<0.05). HE staining revealed that cells had increasingly abnormal protuberance with increasing GANT61 concentration. Flow cytometry analysis also demonstrated that GANT61 induced G1/S arrest and apoptosis of Daoy cells in a dose‑dependent manner (P<0.05). Gli1 and CyclinD1 mRNA expression levels were downregulated by GANT61 treatment (P<0.05); similarly, their protein levels were downregulated by GANT61 treatment in a dose‑dependent manner (P<0.05). In conclusion, Gli1 expression was significantly associated with CyclinD1 expression in MB. These data demonstrated that Gli1 is an important mediator of the SHH pathway activity in MB, and may be a novel agent for use in combined chemotherapeutic regimens.

Introduction

Medulloblastoma (MB) is the most common pediatric malignant brain tumor, and has a poor clinical outcome (1,2). With currently available multimodality therapies, including surgery, radiotherapy and chemotherapy, numerous children have a favorable prognosis; however, the majority of patients suffer from considerable long-term disabilities and morbidity following aggressive multimodal therapy (35). Attempts to further improve the outcomes have been restricted by the cytotoxicity of conventional medication and the nature of the disease. Therefore, an increased understanding of the mechanisms underlying MB is crucial in the development of novel therapeutic approaches.

Aberrant activation of the sonic hedgehog (SHH) signaling pathway has been implicated in the development of MB (68). The Gli family zinc finger 1 (Gli1) transcription factor is considered to be a mediator of the SHH signaling pathway in MB, although its tumorigenic nature and its relative contribution to tumorigenesis remain poorly understood (9).

CyclinD1 is a key protein in the cyclin family that regulates the G1/S transition and is highly expressed in multiple types of tumors (10,11). This protein is regulated by a complex system of signal transduction pathways (12,13). CyclinD1 expression is known to be regulated by Gli1 in MB. Furthermore, GANT61 is a specific Gli1 inhibitor, which has been shown to inhibit the DNA binding activity of Gli1 by binding to the zinc-finger domain (1416).

In order to examine the role of Gli1 in MB, our previous studies screened for genes preferentially regulated by Gli1 in MB cells (17,18). CyclinD1 plays important role in tumor proliferation, and thus the expression of CyclinD1 was investigated in MB cells.

Materials and methods

Reagents and antibodies

GANT61 (Sigma-Aldrich; Merck KGaA, Darmstadt, Germany) was dissolved in dimethyl sulfoxide (DMSO) and stored at −20°C until required for use. The final DMSO concentration in all cultures, including the vehicle control groups, was 0.1% in RPMI 1640 medium (Gibco; Thermo Fisher Scientific, Inc., Grand Island, NY, USA). Fetal bovine serum (FBS) and 0.25% trypsin/EDTA were purchased from Gibco (Thermo Fisher Scientific, Inc.). The hematoxylin and eosin (HE) staining kit (G1060) was purchased from SuoLaibao Technology Co., Ltd. (Beijing, China), and the FITC-Annexin V kit from Abcam (ab14150; Cambridge, MA, USA). The cell counting kit-8 (CCK-8) assay for cell proliferation analysis was purchased from Dojindo Chemical Research Institute (Tokyo, Japan), while the PrimeScript RT Master Mix and reverse transcription (RT) kit (RR014A) was obtained from Takara Bio, Inc. (Shiga, Japan; PrimeScript RT Master Mix). In addition, SYBR Green I was purchased from Beijing Noble Ryder Technology Co., Ltd. (Beijing, China). Antibodies against Gli1 (ab49314) and CyclinD1 (ab187364) were acquired from Abcam, while β-actin antibody (AP0060) was purchased from Bioworld Technology, Inc. (Louis Park, MN, USA). The secondary antibody of Gli1 (BL003A) and CyclinD1 (BL001A) were acquired from Biosharp (Wuhan, China) (19).

Cell culture

Daoy, an MB cell line, was purchased from ATCC (Manassas, VA, USA). The Daoy cells were maintained in RPMI 1640 medium supplemented with 10% fetal bovine serum (500 ml; Gibco), 100 µg/ml penicillin and 100 µg/ml streptomycin (Invitrogen; Thermo Fisher Scientific, Inc., Carlsbad, CA, USA) at 37°C with 5% CO2. Prior to each experiment, trypan blue staining (Sigma-Aldrich) was used to define the cell vitality. The cell activity was determined to be >98%.

Cell proliferation analysis

CCK-8 assay was performed to investigate the cell proliferation, according to the manufacturer's instructions of the kit. Briefly, Daoy cells in exponential growth phase were pipetted into single cells following trypsin digestion. Cells were seeded in a 96-well plate at a density of 8×103 cells/well. RPMI 1640 medium containing 10% FBS was used to culture the cells for 24 h prior to replacing with serum-free medium. Next, the cells were starved for 6 h and then incubated in RPMI 1640 medium supplemented with 1% FBS. The cell culture groups included three groups treated with different concentrations of GANT61 (10, 20 and 40 µM) and a negative untreated control group with normal growing cells, while wells with no cells acted as the blank control. A total of six replicates per group were investigated. The cells were continually cultured in the incubator for a further 24 h before the culture medium was discarded. Subsequently, 100 µl fresh RPMI 1640 medium and 10 µl CCK-8 solution were added into each well. The cells were placed in the incubator to avoid light exposure, and the absorbance at 450 nm (A450) was measured at 0.5, 1, 2 and 4 h, with a Bio-Rad 680 microplate reader (Bio-Rad Laboratories, Inc., Hercules, CA, USA).. The proliferation inhibition rate was calculated as follows: Proliferation inhibition (%) = [A450 (negative control group) - A450 (GANT61-treated group)] / A450 (negative control group) ×100%.

HE staining

Daoy cells in the exponential growth phase were digested into a concentration of 1×106 cells/ml, added to glass coverslips and cultured for 24 h in an incubator. The medium was replaced, followed by addition of different concentrations of GANT61 (10, 20 and 40 µM), while the group without GANT61 treatment served as the control. Subsequently, the cells were extracted after culturing for 24 h, washed with phosphate-buffered saline (PBS) for three times and fixed in 4% paraformaldehyde for 60 min, followed by washing three times with PBS. The cells were then stained with hematoxylin for 5 min and washed by tap water. Following incubation in differentiation buffer for a few seconds and washing with water, eosin was added for 10 min. After washing with tap water, the stained sample was dehydrated, sealed and prepared for microscopic observation.

Flow cytometry

In order to investigate the cell cycle progression, flow cytometry analysis was performed using the FITC-Annexin V kit, according to the manufacturer's instructions. Briefly, 2×104 cells were transferred into 10-ml centrifuge tubes, and centrifuged for 5 min at 250–500 × g at 4°C. After the culture medium was discarded, cells were washed once with the binding buffer and centrifuged for 5 min at at 250–500 × g at 4°C. The final concentration of 1 µg/ml propidium iodide (PI) with FITC-Annexin V (included in the kit) was dissolved in incubation buffer. Resuspended cells were labeled in the dark for 10–15 min with 100 µl solution buffer at room temperature. Cells were then precipitated by centrifugation at at 250–500 × g at 4°C for 5 min and washed with incubation buffer. The sample was then incubated at the 4°C for 20 min in the dark without vibration. Detection and quantification of apoptotic cells was obtained by flow cytometry. This test was performed according to the manufacturer's instructions

RT-polymerase chain reaction (PCR) array analysis

Daoy cells were seeded in RPMI 1640 medium supplemented with 10% FBS, followed by exposure to different concentrations of GANT61 for 24 h, while the control was not treated with any GANT61. Total RNA was extracted from the cells with TRIzol reagent (Invitrogen; Thermo Fisher Scientific, Inc.) after 24 h, according to the manufacturer's instructions. The total RNA extracted was then treated with the PrimeScript RT Master Mix for removal of contaminating DNA and for reverse transcription into cDNA. Briefly, Primers specific for each of the signaling molecules were designed using NCBI/Primer-BLAST and used to generate the PCR products. The following primers were used: GLI1-Forward: 5′-GGGAGGAAAGCAGACTGACT-3′; GLI1-Reverse: 5′-TGGAGAGGTCTTCAGTGCTG-3′; CyclinD1-Forward: 5′-GCATGTTCGTGGCCTCTAAG-3′; CyclinD1-Reverse: 5′-CGTGTTTGCGGATGATCTGT-3′; GAPDH-Forward: 5′-CTCTCTGCTCCTCCCTGTTC-3′; GAPDH-Reverse: 5′-CAATCTCCACTTTGCCACTGC-3′. Target sequences were amplified at 95°C for 1 min, followed by 40 cycles of 95°C for 5 sec and 60°C for 30 sec. GAPDH was used as endogenous normalization control. Subsequently, the samples were investigated by PCR array. Data were analyzed by the ΔΔCq method to determine the mRNA expression levels, as previously described (20,21). The experiment was performed in triplicate and repeated three times.

Western blot analysis

Daoy cells were synchronized in RPMI 1640 medium with 10% FBS, followed by exposure to different concentrations of GANT61 for 24 h, while the control was not treated with any GANT61. The protein profile in the samples was examined by western blot analysis. Briefly, cells were collected and washed three times with PBS. Next, the cells were lysed in fresh radioimmunoprecipitation assay protein lysis buffer containing phenylmethylsulfonyl fluoride (ratio, 100:1) on ice. The total protein concentration was determined by the BCA method (ab102536; Abcam). Following separation by 10% SDS-PAGE, the samples were transferred to polyvinylidene difluoride films. Protein blots were visualized by Ponceau S staining. The films were subsequently blocked with 5% non-fat milk for 2 h at room temperature. Anti-Gli1 (1:500) and anti-CyclinD1 (1:1,000) protein antibodies were added and incubated overnight at 4°C. The films were then incubated with the secondary antibody (1:10,000) at room temperature for 1 h and washed three times with Tris-buffered saline/Tween 20 buffer. An enhanced chemiluminescence reagent (WBKLS0500; Merck Millipore, Billerica, MA, USA) was used to detect the protein levels, which were scanned using a Bio-Rad exposure system, and Image Lab 3.0 software used for quantification (Bio-Rad Laboratories, Inc.).

Immunofluorescence analysis

Daoy cells (5×103) were seeded on glass coverslips and treated with different concentrations of GANT61. At 24 h after incubation, the cells were fixed with 4% paraformaldehyde for 10 min and permeabilized with 1% Triton X-100 in PBS for 10 min. Next, the cells were incubated with rabbit anti-Gli1 and mouse anti-CyclinD1 antibodies at 37°C for 1 h and washed with PBS. Subsequently, incubation for 1 h with DyLight594-conjugated goat anti-rabbit and FITC conjugated goat anti-mouse secondary antibodies (111-165-003 and 111-025-003; 1:10,000; Jackson ImmunoResearch Laboratories, Inc., West Grove, PA, USA) was performed, followed by DAPI staining. The cells were then mounted and observed under a fluorescence microscope.

Statistical analysis

SPSS version 19.0 (IBM Corp., Armonk, NY, USA) software was used for statistical analysis. Data were statistically analyzed by one-way analysis of variance. All experimental data are expressed as the mean ± standard deviation. P<0.05 indicated a statistically significant difference.

Results

Morphological changes of Daoy cells following GANT61 treatment

Daoy cells were cultured for 24 h, and then different concentrations of GANT61 (10, 20 or 40 µM in 0.1% DMSO) were added to examine the effects of GANT61 on the cell morphology. The cells were cultured for a further 24 h and then subjected to inverted microscopic observation. As shown in Fig. 1, the normal, non-adherent Daoy cells in the untreated control group were spherical in shape. Similarly, normal adherent cells were intercellular tight, follow flaky aggregational growth and morphological rules, and their shapes were rectangular or triangular. Notably, groups treated with increasing concentrations of GANT61 demonstrated an evident decreased in cell number, as well as changes in morphology and diversity, which the cells presented with shrinkage and abnormal form. (Fig. 1).

GANT61 inhibits the proliferation and induces cell cycle arrest of Daoy cells

Marked morphological changes and decreased cell number was observed following GANT61 treatment (Fig. 1), indicating reduced cell proliferation or induced cell apoptosis. To elucidate whether cell proliferation was decreased following treatment with different concentrations of GANT61 for 24 h, the cell proliferation was detected by a CCK-8 assay. As shown in Fig. 2A, GANT61 significantly inhibited the proliferation of Daoy cells. The inhibition of proliferation in GANT61-treated groups compared with the control group was dose-dependent (P<0.05; Fig. 2A). Furthermore, to examine whether the growth inhibition of the cells was a result of cell cycle arrest, Daoy cells were stained with FITC-Annexin V and PI, and then subjected to flow cytometry. As displayed in Fig. 2B and C, the percentage of cells in G1 phase increased (P<0.05) with increasing concentration of GANT61 treatment, whereas cells in S phase decreased in a dose-dependent manner (P<0.05). This indicated that GANT61 resulted in cell cycle arrest of Daoy cells at the G1/S transition.

GANT61 promotes cell apoptosis of Daoy cells

To determine whether GANT61 treatment induced cells apoptosis, normal growing Daoy cells were treated with various concentrations of GANT61. After 24 h, the cells were subjected to HE staining and flow cytometry analysis. As shown in Fig. 3A, the HE staining results demonstrated that normal cells had a regular morphology. However, clearly visible abnormal morphologies were observed in Daoy cells treated with GANT61, with abnormal protuberance observed. The abnormal protuberance, chromatin condensation and fragmentation features were more evident at increased concentrations of GANT61, thus indicating a dose-dependent effect. HE staining also demonstrated decreased in cell number, increased cell shrinkage and nuclear fragmentation. As shown in Fig. 3B, the percentage of apoptotic cells increased significantly in the GANT61-treated cells, compared with the untreated group (P<0.05). These results verified the prediction that GANT61 induced cell apoptosis in Daoy cells (19).

GANT61 inhibits the expression of Gli1 and CyclinD1 in the mRNA and protein level

To examine the underlying mechanism of reduced cell apoptosis and cell cycle arrest, the total RNA of the cells were extracted by TRIzol reagent, reverse transcribed into cDNA and then subjected to PCR. Gli1 is an important transcription factor in the SHH signaling pathway, regulating the transcription of multiple downstream target genes, including CyclinD1, the oncogene controlling cell cycle entry (22,23). As shown in Fig. 4A, the results revealed that GANT61 was able to significantly inhibit the gene expression of Gli1 (P<0.05). Along with the decreased expression of the Gli1 gene, CyclinD1 mRNA appeared to be downregulated synchronously (P<0.05). In addition, protein levels were assayed by immunofluorescence analysis. As indicated in Fig. 4B and C, CyclinD1 was mainly localized in the cytosol of Daoy cells, whereas Gli1, as a transcription factor, was located in both the cell cytosol and nucleus. Following treatment with GANT61 for 24 h, Daoy cells showed decreased levels of Gli1 protein compared with that in untreated cells (P<0.05). Subsequently, CyclinD1 was also decreased, as one of the Gli1 transcriptional targets (P<0.05). The inhibition by GANT61 on Gli1 and CyclinD1 was dose-dependent. To further elucidate the inhibitory effects of GANT61 on the expression of Gli1 and CyclinD1, their protein levels were examined by western blot analysis. Daoy cells treated with GANT61 for 24 h were lysed and separated by SDS-PAGE, and the protein expression levels of Gli1 and CyclinD1 were detected using the corresponding antibodies. The results demonstrated that GANT61 was able to decrease the level of Gli1 protein (Fig. 5). In line with the decreased expression of Gli1 protein, CyclinD1 protein also appeared to be downregulated (P<0.05). The inhibition of Gli1 and CyclinD1 protein levels by GANT61 was in a dose-dependent manner (P<0.05). These results were consistent with the data obtained by qPCR and immunofluorescence analyses, indicating that GANT61 can significantly inhibit Gli1 and CyclinD1 expression at the mRNA and protein levels.

Discussion

Aberrant activation of the SHH signaling pathway is implicated in various types of human cancer (24). The SHH signaling pathway is important in regulating cell proliferation and differentiation in the embryonic development of the cerebellum (25). MB is characterized by constitutive activation of the SHH signaling pathway, and is genetically characterized by mutations in patched homolog 1 (PTCH1), which blocks the function of smoothened (SMO), or other downstream pathway mutations (26). Gli1 expression is inhibited by suppressor-of-fused, preventing it from activating gene transcription. The binding of SHH to PTCH1 or other mutations releases a basal repression on SMO, which is then activated (27). Subsequently, Gli1 is released and activates a series of gene transcriptions (28,29).

Inhibitors of the SHH signaling pathway are currently being developed to mainly target SMO or its upstream sites (30). Numerous studies using such inhibitors in MB have demonstrated the efficacy of this treatment, and these findings have been translated into Phase I and II clinical trials (3134). While these therapies have shown promising results, various significant challenges remain, including the possible long-term bone marrow suppression and drug toxicity (35,36). As the majority of targeted therapies for MB have focused on SMO, it is concerning that only a single mechanism has been identified and targeted, making resistance a frequently encountered complication (37). SMO mutation is not the only mechanism of acquired drug resistance, as the development of other downstream hedgehog pathway component mutations have since been implicated in SHH inhibitor resistance. Kool et al (38) indicated that amplifications of Gli may result in inability to respond to current SMO inhibitors. Such aberrations include the amplification of Gli and the upregulation of PI3K/AKT signaling, manifesting in vivo as tumor regrowth in the same mouse model (39,40).

Gli1 serves a crucial role in the transformation and proliferation of malignant cells (41). It is also important for preventing apoptosis and maintaining the malignant proliferation of tumor cells, and is involved in tumor cell protection against chemotherapy (42). Berman et al (43) indicated that the expression level of Gli1 may reflect the degree of activation of the SHH signaling pathway. Inhibition of abnormal activation of this signaling pathway by inhibiting the expression of Gli1 can inhibit the growth of tumor cells. Gli1, as the main transcription factor downstream of the SHH signaling pathway, may be able to inhibit tumor cell proliferation and differentiation through downregulation of downstream target genes. On the basis of the pivotal role of Gli1 in malignant cells, it has become increasingly evident that Gli1 is a promising target for anticancer therapy. A direct strategy to interfere with Gli1 activity is to induce selective inhibition of its DNA transcription.

GANT61, an agent that exerts an inhibitory activity of the SHH signaling pathway, functions by selectively binding to Gli1 and has been found to suppress proliferation in various tumors (44,45). In the present study, GANT61 had in vitro activity against tumor proliferation, and induced cell cycle arrest and apoptosis. Furthermore, GANT61 was found to inhibit the Gli1 mRNA and protein expression levels. Dysregulation of cell cycle progression is considered to serve an important role in cancer; thus, the current study investigated whether Gli1 is associated with the typical oncogene CyclinD1 in the cell cycle. CyclinD1 is a key protein regulating the G1/S transition in the cell cycle and is highly expressed in multiple types of tumors (46). CyclinD1 is frequently deregulated in various cancer types, and is a biomarker of cancer phenotype and disease progression (46,47). Overexpressed CyclinD1 accelerates the cell cycle transition, leading to uncontrolled cell proliferation and the development of cancer. The present study identified that the mRNA expression of Gli1 was significantly associated with CyclinD1 expression in MB, and a similar observation was identified regarding the protein levels. Suppressing the expression of Gli1 may inhibit the overexpression of CyclinD1 and the proliferation of tumor cells, and synchronously promote cell apoptosis. Therefore, blocking the expression of Gli1 may be an attractive therapeutic strategy for MB.

In conclusion, SHH signaling pathway can regulate tumor cell cycle and apoptosis in different molecular levels. Increased expression of Gli1 induced the upregulation of CyclinD1 expression, thus promoting cell proliferation, which may be one of the growth patterns of tumor cells. Therefore, Gli1 may be an important target for MB treatment. Therapies using Gli1-targeted inhibitors alone or combined with other cytotoxic chemotherapeutics may become an effective targeted treatment of MB. However, the association of the SHH signaling pathway and other pathways in MB cells with the specific mechanism of apoptosis induced by targeted therapy requires further investigation.

Acknowledgements

The present study was supported by grants from the Natural Science Foundation of Zhejiang Province (no. LY13H160033), the Zhejiang Medical and Health Science and Technology Plan Project (no. 2012RCA043) and the Foundation of Wenzhou Scientific and Technological Bureau Protect (no. Y20140717).

References

1 

Gerber NU, Mynarek M, von Hoff K, Friedrich C, Resch A and Rutkowski S: Recent developments and current concepts in medulloblastoma. Cancer Treat Rev. 40:356–365. 2014. View Article : Google Scholar : PubMed/NCBI

2 

Gilbertson RJ: Medulloblastoma: Signalling a change in treatment. Lancet Oncol. 5:209–218. 2004. View Article : Google Scholar : PubMed/NCBI

3 

Packer RJ and Vezina G: Management of and prognosis with medulloblastoma: Therapy at a crossroads. Arch Neurol. 65:1419–1424. 2008. View Article : Google Scholar : PubMed/NCBI

4 

Rutkowski S, von Hoff K, Emser A, Zwiener I, Pietsch T, Figarella-Branger D, Giangaspero F, Ellison DW, Garre ML, Biassoni V, et al: Survival and prognostic factors of early childhood medulloblastoma: An international meta-analysis. J Clin Oncol. 28:4961–4968. 2010. View Article : Google Scholar : PubMed/NCBI

5 

Moxon-Emre I, Bouffet E, Taylor MD, Laperriere N, Scantlebury N, Law N, Spiegler BJ, Malkin D, Janzen L and Mabbott D: Impact of craniospinal dose, boost volume, and neurologic complications on intellectual outcome in patients with medulloblastoma. J Clin Oncol. 32:1760–1768. 2014. View Article : Google Scholar : PubMed/NCBI

6 

Archer TC, Weeraratne SD and Pomeroy SL: Hedgehog-GLI pathway in medulloblastoma. J Clin Oncol. 30:2154–2156. 2012. View Article : Google Scholar : PubMed/NCBI

7 

Gotschel F, Berg D, Gruber W, Bender C, Eberl M, Friedel M, Sonntag J, Rüngeler E, Hache H, Wierling C, et al: Synergism between Hedgehog-GLI and EGFR signaling in Hedgehog-responsive human medulloblastoma cells induces downregulation of canonical Hedgehog-target genes and stabilized expression of GLI1. PLoS One. 8:e654032013. View Article : Google Scholar : PubMed/NCBI

8 

Cho YJ, Tsherniak A, Tamayo P, Santagata S, Ligon A, Greulich H, Berhoukim R, Amani V, Goumnerova L, Eberhart CG, et al: Integrative genomic analysis of medulloblastoma identifies a molecular subgroup that drives poor clinical outcome. J Clin Oncol. 29:1424–1430. 2011. View Article : Google Scholar : PubMed/NCBI

9 

Mcmillan R and Matsui W: Molecular pathways: The hedgehog signaling pathway in cancer. Clin Cancer Res. 18:4883–4888. 2012. View Article : Google Scholar : PubMed/NCBI

10 

Shahi MH, Afzal M, Sinha S, Eberhart CG, Rey JA, Fan X and Castresana JS: Regulation of sonic hedgehog-GLI1 downstream target genes PTCH1, Cyclin D2, Plakoglobin, PAX6 and NKX2.2 and their epigenetic status in medulloblastoma and astrocytoma. BMC Cancer. 10:6142010. View Article : Google Scholar : PubMed/NCBI

11 

Kenney AM, Cole MD and Rowitch DH: Nmyc upregulation by sonic hedgehog signaling promotes proliferation in developing cerebellar granule neuron precursors. Development. 130:15–28. 2003. View Article : Google Scholar : PubMed/NCBI

12 

Musgrove EA, Caldon CE, Barraclough J, Stone A and Sutherland RL: Cyclin D as a therapeutic target in cancer. Nat Rev Cancer. 11:558–572. 2011. View Article : Google Scholar : PubMed/NCBI

13 

Barbash O, Zamfirova P, Lin DI, Chen X, Yang K, Nakagawa H, Lu F, Rustgi AK and Diehl JA: Mutations in Fbx4 inhibit dimerization of the SCF (Fbx4) ligase and contribute to cyclin D1 overexpression in human cancer. Cancer Cell. 14:68–78. 2008. View Article : Google Scholar : PubMed/NCBI

14 

Fu M, Wang C, Li Z, Sakamaki T and Pestell RG: Minireview: Cyclin D1: Normal and abnormal functions. Endocrinology. 145:5439–5447. 2004. View Article : Google Scholar : PubMed/NCBI

15 

Musgrove EA: Cyclins: Roles in mitogenic signaling and oncogenic transformation. Growth Factors. 24:13–19. 2006. View Article : Google Scholar : PubMed/NCBI

16 

Fu J, Rodova M, Roy SK, Sharma J, Singh KP, Srivastava RK and Shankar S: GANT-61 inhibits pancreatic cancer stem cell growth in vitro and in NOD/SCID/IL2R gamma null mice xenograft. Cancer Lett. 330:22–32. 2013. View Article : Google Scholar : PubMed/NCBI

17 

Peukert S and Miller-Moslin K: Small-molecule inhibitors of the hedgehog signaling pathway as cancer therapeutics. ChemMedChem. 5:500–512. 2010. View Article : Google Scholar : PubMed/NCBI

18 

Mazumdar T, Devecchio J, Shi T, Jones J, Agyeman A and Houghton JA: Hedgehog signaling drives cellular survival in human colon carcinoma cells. Cancer Res. 71:1092–1102. 2011. View Article : Google Scholar : PubMed/NCBI

19 

Lin Z, Li S, Sheng H, Cai M, Ma LY, Hu L, Xu S, Yu LS and Zhang N: Suppression of GLI sensitizes medulloblastoma cells to mitochondria-mediated apoptosis. J Cancer Res Clin Oncol. 142:2469–2478. 2016. View Article : Google Scholar : PubMed/NCBI

20 

Rafiee M, Keramati MR, Ayatollahi H, Sadeghian MH, Barzegar M, Asgharzadeh A and Alinejad M: Down-Regulation of Ribosomal S6 kinase RPS6KA6 in Acute Myeloid Leukemia Patients. Cell J. 18:159–164. 2016.PubMed/NCBI

21 

Floris I, Billard H, Boquien CY, Joram-Gauvard E, Simon L, Legrand A, Boscher C, Rozé JC, Bolaños-Jiménez F and Kaeffer B: MiRNA Analysis by Quantitative PCR in Preterm Human Breast Milk Reveals Daily Fluctuations of hsa-miR-16-5p. PLoS One. 10:e1404882015. View Article : Google Scholar

22 

Huang XB, Shi Y, Wang CS, Wang XD, Cheng J and Che FF: Synergistic Inhibitory Effect of Arsenic Trioxide Combined with Itraconazole on Hedgehog Pathway of Multiple Myeloma NCI-H929 Cells. Zhongguo Shi Yan Xue Ye Xue Za Zhi. 24:1459–1465. 2016.(In Chinese). PubMed/NCBI

23 

Du WZ, Feng Y, Wang XF, Piao XY, Cui YQ, Chen LC, Lei XH, Sun X, Liu X, Wang HB, et al: Curcumin suppresses malignant glioma cells growth and induces apoptosis by inhibition of SHH/GLI1 signaling pathway in vitro and vivo. CNS Neurosci Ther. 19:926–936. 2013. View Article : Google Scholar : PubMed/NCBI

24 

Nakamura M and Katano M: Hedgehog signaling pathway and its impact on development of cancer therapy. Fukuoka Igaku Zasshi. 99:102–106. 2008.(In Japanese). PubMed/NCBI

25 

Malek R, Matta J, Taylor N, Perry ME and Mendrysa SM: The p53 inhibitor MDM2 facilitates Sonic Hedgehog-mediated tumorigenesis and influences cerebellar foliation. PLoS One. 6:e178842011. View Article : Google Scholar : PubMed/NCBI

26 

Sahebjam S, Siu LL and Razak AA: The utility of hedgehog signaling pathway inhibition for cancer. Oncologist. 17:1090–1099. 2012. View Article : Google Scholar : PubMed/NCBI

27 

Wang X, Venugopal C, Manoranjan B, McFarlane N, O'Farrell E, Nolte S, Gunnarsson T, Hollenberg R, Kwiecien J, Northcott P, et al: Sonic hedgehog regulates Bmi1 in human medulloblastoma brain tumor-initiating cells. Oncogene. 31:187–199. 2012. View Article : Google Scholar : PubMed/NCBI

28 

Jiang J and Hui CC: Hedgehog signaling in development and cancer. Dev Cell. 15:801–812. 2008. View Article : Google Scholar : PubMed/NCBI

29 

Ruiz i Altaba A, Palma V and Dahmane N: Hedgehog-Gli signalling and the growth of the brain. Nat Rev Neurosci. 3:24–33. 2002. View Article : Google Scholar : PubMed/NCBI

30 

Tang Y, Gholamin S, Schubert S, Willardson MI, Lee A, Bandopadhayay P, Bergthold G, Masoud S, Nguyen B, Vue N, et al: Epigenetic targeting of Hedgehog pathway transcriptional output through BET bromodomain inhibition. Nat Med. 20:732–740. 2014. View Article : Google Scholar : PubMed/NCBI

31 

Jimeno A, Weiss GJ, Miller WH Jr, Gettinger S, Eigl BJ, Chang AL, Dunbar J, Devens S, Faia K, Skliris G, et al: Phase I study of the Hedgehog pathway inhibitor IPI-926 in adult patients with solid tumors. Clin Cancer Res. 19:2766–2774. 2013. View Article : Google Scholar : PubMed/NCBI

32 

Berlin J, Bendell JC, Hart LL, Firdaus I, Gore I, Hermann RC, Mulcahy MF, Zalupski MM, Mackey HM, Yauch RL, et al: A randomized phase II trial of vismodegib versus placebo with FOLFOX or FOLFIRI and bevacizumab in patients with previously untreated metastatic colorectal cancer. Clin Cancer Res. 19:258–267. 2013. View Article : Google Scholar : PubMed/NCBI

33 

Kim EJ, Sahai V, Abel EV, Griffith KA, Greenson JK, Takebe N, Khan GN, Blau JL, Craig R, Balis UG, et al: Pilot clinical trial of hedgehog pathway inhibitor GDC-0449 (vismodegib) in combination with gemcitabine in patients with metastatic pancreatic adenocarcinoma. Clin Cancer Res. 20:5937–5945. 2014. View Article : Google Scholar : PubMed/NCBI

34 

D'Amato C, Rosa R, Marciano R, D'Amato V, Formisano L, Nappi L, Raimondo L, Di Mauro C, Servetto A, Fulciniti F, et al: Inhibition of Hedgehog signalling by NVP-LDE225 (Erismodegib) interferes with growth and invasion of human renal cell carcinoma cells. Br J Cancer. 111:1168–1179. 2014. View Article : Google Scholar : PubMed/NCBI

35 

Rudin CM, Hann CL, Laterra J, Yauch RL, Callahan CA, Fu L, Holcomb T, Stinson J, Gould SE, Coleman B, et al: Treatment of medulloblastoma with hedgehog pathway inhibitor GDC-0449. N Engl J Med. 361:1173–1178. 2009. View Article : Google Scholar : PubMed/NCBI

36 

Kimura H, Ng JM and Curran T: Transient inhibition of the Hedgehog pathway in young mice causes permanent defects in bone structure. Cancer Cell. 13:249–260. 2008. View Article : Google Scholar : PubMed/NCBI

37 

Meani RE, Lim SW, Chang AL and Kelly JW: Emergence of chemoresistance in a metastatic basal cell carcinoma patient after complete response to hedgehog pathway inhibitor vismodegib (GDC-0449). Australas J Dermatol. 55:218–221. 2014. View Article : Google Scholar : PubMed/NCBI

38 

Kool M, Jones DT, Jäger N, Northcott PA, Pugh TJ, Hovestadt V, Piro RM, Esparza LA, Markant SL, Remke M, et al: Genome sequencing of SHH medulloblastoma predicts genotype-related response to smoothened inhibition. Cancer Cell. 25:393–405. 2014. View Article : Google Scholar : PubMed/NCBI

39 

Samkari A, White J and Packer R: SHH inhibitors for the treatment of medulloblastoma. Expert Rev Neurother. 15:763–770. 2015. View Article : Google Scholar : PubMed/NCBI

40 

Pan S, Wu X, Jiang J, Gao W, Wan Y, Cheng D, Han D, Liu J, Englund NP, Wang Y, et al: Discovery of NVP-LDE225, a potent and selective smoothened antagonist. ACS Med Chem Lett. 1:130–134. 2010. View Article : Google Scholar : PubMed/NCBI

41 

Ruiz i Altaba A, Mas C and Stecca B: The Gli code: An information nexus regulating cell fate, stemness and cancer. Trends Cell Biol. 17:438–447. 2007. View Article : Google Scholar : PubMed/NCBI

42 

Katoh Y and Katoh M: Hedgehog target genes: Mechanisms of carcinogenesis induced by aberrant hedgehog signaling activation. Curr Mol Med. 9:873–886. 2009. View Article : Google Scholar : PubMed/NCBI

43 

Berman DM, Karhadkar SS, Hallahan AR, Pritchard JI, Eberhart CG, Watkins DN, Chen JK, Cooper MK, Taipale J, Olson JM and Beachy PA: Medulloblastoma growth inhibition by hedgehog pathway blockade. Science. 297:1559–1561. 2002. View Article : Google Scholar : PubMed/NCBI

44 

Miyazaki Y, Matsubara S, Ding Q, Tsukasa K, Yoshimitsu M, Kosai K and Takao S: Efficient elimination of pancreatic cancer stem cells by hedgehog/GLI inhibitor GANT61 in combination with mTOR inhibition. Mol Cancer. 15:492016. View Article : Google Scholar : PubMed/NCBI

45 

Benvenuto M, Masuelli L, De Smaele E, Fantini M, Mattera R, Cucchi D, Bonanno E, Di Stefano E, Frajese GV, Orlandi A, et al: In vitro and in vivo inhibition of breast cancer cell growth by targeting the Hedgehog/GLI pathway with SMO (GDC-0449) or GLI (GANT-61) inhibitors. Oncotarget. 7:9250–9270. 2016.PubMed/NCBI

46 

Li X, Hao Z, Fan R, Zou X, Jin H, Pan Y, He L, Du R, Gao L, Liu D and Fan D: CIAPIN1 inhibits gastric cancer cell proliferation and cell cycle progression by downregulating CyclinD1 and upregulating P27. Cancer Biol Ther. 6:1539–1545. 2007. View Article : Google Scholar : PubMed/NCBI

47 

Seiler R, Thalmann GN, Rotzer D, Perren A and Fleischmann A: CCND1/CyclinD1 status in metastasizing bladder cancer: A prognosticator and predictor of chemotherapeutic response. Mod Pathol. 27:87–95. 2014. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

January-2017
Volume 13 Issue 1

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Lin Z, Sheng H, You C, Cai M, Zhang Y, Yu LS, Yu X, Lin J and Zhang N: Inhibition of the CyclinD1 promoter in response to sonic hedgehog signaling pathway transduction is mediated by Gli1. Exp Ther Med 13: 307-314, 2017
APA
Lin, Z., Sheng, H., You, C., Cai, M., Zhang, Y., Yu, L.S. ... Zhang, N. (2017). Inhibition of the CyclinD1 promoter in response to sonic hedgehog signaling pathway transduction is mediated by Gli1. Experimental and Therapeutic Medicine, 13, 307-314. https://doi.org/10.3892/etm.2016.3969
MLA
Lin, Z., Sheng, H., You, C., Cai, M., Zhang, Y., Yu, L. S., Yu, X., Lin, J., Zhang, N."Inhibition of the CyclinD1 promoter in response to sonic hedgehog signaling pathway transduction is mediated by Gli1". Experimental and Therapeutic Medicine 13.1 (2017): 307-314.
Chicago
Lin, Z., Sheng, H., You, C., Cai, M., Zhang, Y., Yu, L. S., Yu, X., Lin, J., Zhang, N."Inhibition of the CyclinD1 promoter in response to sonic hedgehog signaling pathway transduction is mediated by Gli1". Experimental and Therapeutic Medicine 13, no. 1 (2017): 307-314. https://doi.org/10.3892/etm.2016.3969