The molecular pathways driving mesenchymal glioblastoma (GBM) are still not well understood. We report here that truncated glioma-associated oncogene homolog 1 (tGLI1) is a tumor-specific transcription factor that facilitates GBM growth, is enriched in the mesenchymal subtype of GBM and glioma stem cells (GSC), and promotes mesenchymal GSC by upregulating transcription of CD44. In an orthotopic GBM xenograft mouse model, tGLI1-overexpressing tumors grew more aggressively with increased proliferation and angiogenesis compared with control and GLI1-overexpressing xenografts. tGLI1 was highly expressed in GBM clinical specimens but undetectable in normal brains, whereas GLI1 was expressed in both tissues. A tGLI1 activation signature (tGAS) correlated with glioma grade, tumor angiogenesis, and poor overall survival, and GBMs with high tGAS were enriched with mesenchymal GBM/GSC gene signatures. Neurospheres contained increased levels of tGLI1, but not GLI1, compared with the monolayer culture; mesenchymal GSC expressed more tGLI1 than proneural GSC. Ectopic tGLI1 expression enhanced the ability of mesenchymal GSC to yield neurospheres in vitro and to form tumors in mouse brains. Selective tGLI1 knockdown reduced neurosphere formation of GBM cells. tGLI1 bound to and transactivated the promoter of the CD44 gene, a marker and mediator for mesenchymal GSC, leading to its expression. Collectively, these findings advance our understanding of GBM biology by establishing tGLI1 as a novel transcriptional activator of CD44 and a novel mediator of mesenchymal GBM and GSC.

Significance: These findings highlight the role of a tumor-specific gain-of-function transcription factor tGLI1 in mesenchymal glioma stem cell maintenance and mesenchymal GBM growth. Cancer Res; 78(10); 2589–600. ©2018 AACR.

Glioblastoma (GBM) is the most common and most lethal brain tumor in adults, accounting for 15% of all brain tumors. GBM prognosis remains poor, with a median survival of 14 months and less than 5% of patients surviving 5 years after diagnosis (1, 2). Extensive genomic analyses have divided GBMs into three to four distinct subtypes with slight variations (3, 4). The proneural and mesenchymal subtypes of GBM are commonly delineated across two classification systems. Mesenchymal GBMs tend to respond poorly to chemotherapy and radiation, and are associated with a worse prognosis (3–5).

Recent studies of GBM identified subpopulations of tumor cells exhibiting stem cell–like properties, including the ability to self-renew, persistently proliferate, and differentiate into multiple cell lineages (6). The subpopulation of stem-like cells, or glioma stem cells (GSC), is a key driver of tumor initiation, recurrence, and chemoresistance (7). Recently, patient-derived GSCs were identified to contain two distinct and mutually exclusive subtypes termed “proneural” and “mesenchymal” (8, 9). CD44 was identified as a key marker of mesenchymal GSCs (8). High CD44 expression in GBM is predictive of poorer overall survival and is associated with increased GBM invasion, proliferation, and chemoresistance (10). Mesenchymal GSCs were found to be more aggressive and radioresistant; therefore, understanding the signaling pathways controlling the mesenchymal GSC phenotype is the key to developing targeted treatments for mesenchymal GSCs in GBM.

The Sonic Hedgehog (SHH) pathway plays an essential role in GSC function (11). The glioma-associated oncogene homolog 1 (GLI1) family of zinc-finger transcription factors function as the terminal effectors of the SHH signaling pathway (12). Our laboratory discovered truncated GLI1 (tGLI1) as a novel alternatively spliced, gain-of-function variant of GLI1, with a 41 amino acid deletion spanning the entire exon 3 and part of exon 4, which retains all of the known functional domains of GLI1 (13). We reported that tGLI1 regulates known GLI1 target genes to a similar degree as wild-type GLI1 (13) but gained the ability to transcriptionally activate genes not regulated by GLI1, including CD24, HPA1, TEM7, VEGFA, VEGFC, and VEGFR2, thus promoting cancer cell growth, migration, invasion, and angiogenesis (13–17). We also showed that tGLI1 is detectable only in cell lines, patient-derived xenografts, and primary specimens of GBM, but is undetectable in normal brain tissue or other normal human tissues (18). The tumor-specific expression pattern of tGLI1 in invasive breast cancer was reported in our previous study (14). Other groups have confirmed our findings that tGLI1 is detectable in triple-negative breast cancer (19) and malignant gliomas (20). Metastatic hepatoma cells also express tGLI1 (21).

GLI1 has been shown to drive transcription of pluripotency markers in cancer stem cells; however, the role that gain-of-function tGLI1 plays in cancer stem cell function remains unknown (11, 22). It is also unknown whether tGLI1 promotes GBM progression using orthotopic xenograft mouse models. To address these gaps of knowledge, we initiated the current study that includes two different animal models and found that tGLI1 drives the formation of larger, more proliferative, and more highly vascularized tumors than GLI1 in orthotopic GBM and GSC xenograft mouse models. Mechanistic studies provided evidence linking tGLI1 to mesenchymal GBM and GSCs. Our results also established tGLI1 as a novel transcriptional activator of the CD44 gene, a known marker and regulator of mesenchymal GSCs and also other cancer types.

Cell lines and patient specimens

U373MG cells were obtained from ATCC and cultured according to their recommendations. Luciferase-expressing G48LL2 cells were developed by Dr. Waldemar Debinski (23). BTCOE 4810/4795 cell lines were developed and validated from patient tumors at Wake Forest Brain Tumor Center of Excellence, Winston-Salem, NC (24). Patient-derived GSCs (proneural GSC-11 and GSC-23; mesenchymal GSC-20 and GSC-28) were kind gifts from Drs. Erik Sulman and Krishna Bhat at The University of Texas MD Anderson Cancer Center (Houston, TX; ref. 8). GSCs were passaged under neurosphere-forming conditions in serum-free DMEM/F12 growth medium supplemented with B27, FGF, and EGF in order to preserve stem-like properties. All cells used were tested every 6 months for Mycoplasma; only cells free of Mycoplasma were used in the studies. Normal brain tissue microarray (BNC17011) and glioma tissue microarray (GL2083) were purchased from US Biomax. Additional GBM samples were from Wake Forest Brain Tumor Center of Excellence (24). We have obtained written informed consent from the patients; the studies were approved by Wake Forest Institutional Review Board and were conducted in accordance with recognized ethical guidelines.

Generation of lentiviral GLI1- and tGLI1-expressing vectors and isogenic stable cell lines

The open reading frame for tGLI1 and GLI1 were cloned into the lentiviral expressing vector pCDH-CMV-MCS-EF1-RFP-Puro (CD516B-2) by System Biosciences. Plasmids were sequenced to confirm insertion of tGLI1 and GLI1 open reading frames. Lentiviral packaging was performed using a third-generation lentiviral pPACK packaging system from System Biosciences. The pPACK packaging plasmids along with the tGLI1 or GLI1 lentiviral expressing plasmid were cotransfected into 293TN cells for 48 to 96 hours, and the cell culture medium was subsequently collected. Viral particles were concentrated, titers measured, and used to infect G48LL2 and GSC-28 cells. Infected cells were treated with puromycin and FACS sorted for RFP-positive cells.

Animal studies

Female nude mice 6 to 8 weeks of age were used. Actively growing G48LL2 or GSC-28 cells were injected at a concentration of 1 to 5 × 105 cells in 5 μL PBS into the right frontal lobe. For the GSC-28 in vivo study, immediately prior to inoculation into mouse brain, GSC-28 neurospheres were harvested and disassociated to single-cell suspension. Mice were anesthetized with a ketamine/xylazine mixture to the coronal suture through a scalp incision according to an approved Institutional Animal Care and Use Committee protocol. For bioluminescent imaging, xenograft-bearing mice were injected with d-luciferin intraperitoneally at 100 mg/kg body weight and then imaged weekly using PerkinElmer IVIS100 imager.

tGLI1 activation signature and gene set enrichment analysis

A tGLI1 activation signature (tGAS) was generated by averaging the median-centered expression of the six tGLI1 target genes (CD24, VEGFA, VEGFC, HPA1, TEM7, VEGFR2; refs. 13–17). Gene set enrichment analysis was performed by generating the Gene MatriX file (.gmx) by using published signatures for AngioMatrix (25), The Cancer Genome Atlas (TCGA) GBM subtypes (4), the Phillips GBM subtypes (3), and the GSC subtypes (8). The Gene Cluster Text file (.gct) was generated from the TCGA GBM dataset. The Categorical Class file (.cls) was generated based on the tGAS score or the GLI1 expression of patients in the TCGA GBM dataset. The number of permutations for gene set enrichment analysis (GSEA) was set to 1,000, and we used the TCGA gene list as the chip platform. For generation of heat maps, patients were divided by a high or low tGAS score and the genes included in the map were genes within the published signatures for the indicated GBM subtypes (3, 4). Heat maps were generated using Morpheus software developed by the Broad Institute, Cambridge, MA.

tGLI1 knockdown using a tGLI1-targeting locked nucleic acid oligonucleotide

A tGLI1-targeting locked nucleic acid (LNA) oligonucleotide (oligo) and a nontargeting control oligo were custom made by Exiqon. Sequence for the control LNA oligo is C+T+G+T+C*T*T*C*A*G*T*T*C+A+A+C. BLAST analysis did not reveal binding of the control oligo to any genes. Sequence for the tGLI1-targeting LNA oligo is C+A+A+C+T*T*G*A*C*T*T*C*T+G+T+C.

Phosphorothioated bases are indicated by *, whereas LNA bases are labeled by +. Both modifications make the oligos resistant to nuclease-mediated degradation. U373MG GBM cells with high endogenous tGLI1 expression were transfected for 48 hours using Lipofectamine 2000 (Invitrogen) and then seeded for the neurosphere formation assay. tGLI1 and GLI1 mRNA expression was determined by RT-qPCR.

Statistical analyses

Data are presented as mean ± SE. The Student t test, Pearson correlation, univariate/multivariate Cox proportional hazards tests, and one-way ANOVA were performed using SigmaPlot version 11.0.

tGLI1 promotes growth of GBM in an orthotopic mouse model

Whether tGLI1 plays a role in promoting GBM intracranial growth has not been investigated. Herein, we generated isogenic cell lines from a low-passage G48LL2 human GBM cell line stably expressing empty vector, GLI1, or tGLI1 (Fig. 1A). Expression levels for GLI1 and tGLI1 in these cell lines are similar to those found in GBM specimens (Fig. 1A, bottom). The isogenic lines were implanted intracranially into female nude mice, and tumor growth was tracked by weekly bioluminescence imaging. Results showed that GBM cells expressing tGLI1 formed larger tumors (Fig. 1B). Representative images are shown in Fig. 1C and D. Analysis of brain sections using IHC indicated increased proliferation index (Ki-67 IHC) and increased microvessel density (CD31 IHC) in tGLI1-expressing tumors (Fig. 1E–H). Representative IHC images are shown in Fig. 1I. Using custom GLI1- and tGLI1-specific antibodies that we developed and validated (17), we further examined GBM specimens and normal brain tissues for GLI1 and tGLI1 expression, and found tGLI1 to be highly expressed in GBM samples but undetectable in normal brain, whereas GLI1 is expressed in both samples (Fig. 1J and K). Results in Fig. 1 demonstrate that tGLI1 promotes GBM tumor growth in the brain microenvironment and that tGLI1 is expressed in GBM, but not in normal brain.

Figure 1.

tGLI1 is expressed in a tumor-specific fashion and promotes intracranial GBM growth. A, Top, isogenic G48LL2 GBM cell lines carrying lentiviral mock, GLI1, or tGLI1 vector were subjected to immunoblotting for GLI1 and tGLI1 expression. Bottom, expression levels for GLI1 and tGLI1 in these cell lines are similar to those found in GBM specimens, as shown by IHC. B, tGLI1 rendered GBM more aggressive in growth. Isogenic luciferase-expressing G48LL2 cell lines were injected into the right frontal lobe of female nude mice (n = 5 per group), and tumor growth was assessed weekly via bioluminescent imaging. C, Representative bioluminescent images of actively growing tumors at day 56. D, Representative bioluminescent images of ex vivo mouse brains. E–I, tGLI1-overexpressing GBM xenografts were more proliferative, and more vascularized mouse brains were subjected to hematoxylin and eosin (H&E) staining and IHC with indicated antibodies. Immunostained sections were scored by a pathologist, and H-scores were calculated. I shows representative IHC images. J and K, tGLI1 is highly expressed in GBM specimens, but not in normal brain tissues. A cohort of normal healthy brain tissues (n = 80) and GBM patient samples (n = 63) were subjected to IHC using GLI1- and tGLI1-specific antibodies. Immunostained sections were scored by a pathologist to derive H-scores. K shows representative IHC images. Student t test was used to compute P values. NS, nonsignificant.

Figure 1.

tGLI1 is expressed in a tumor-specific fashion and promotes intracranial GBM growth. A, Top, isogenic G48LL2 GBM cell lines carrying lentiviral mock, GLI1, or tGLI1 vector were subjected to immunoblotting for GLI1 and tGLI1 expression. Bottom, expression levels for GLI1 and tGLI1 in these cell lines are similar to those found in GBM specimens, as shown by IHC. B, tGLI1 rendered GBM more aggressive in growth. Isogenic luciferase-expressing G48LL2 cell lines were injected into the right frontal lobe of female nude mice (n = 5 per group), and tumor growth was assessed weekly via bioluminescent imaging. C, Representative bioluminescent images of actively growing tumors at day 56. D, Representative bioluminescent images of ex vivo mouse brains. E–I, tGLI1-overexpressing GBM xenografts were more proliferative, and more vascularized mouse brains were subjected to hematoxylin and eosin (H&E) staining and IHC with indicated antibodies. Immunostained sections were scored by a pathologist, and H-scores were calculated. I shows representative IHC images. J and K, tGLI1 is highly expressed in GBM specimens, but not in normal brain tissues. A cohort of normal healthy brain tissues (n = 80) and GBM patient samples (n = 63) were subjected to IHC using GLI1- and tGLI1-specific antibodies. Immunostained sections were scored by a pathologist to derive H-scores. K shows representative IHC images. Student t test was used to compute P values. NS, nonsignificant.

Close modal

High tGLI1 activity is associated with poor overall survival of patients with GBM and increased angiogenesis of GBM

We asked whether tGLI1 activity can be used as a prognostic indicator for GBM. To indicate tGLI1 activation status, we created tGAS using expression levels of its six target genes (13–17). We then analyzed a GEO dataset (GSE4290) consisting of mRNA expression profiles of 23 normal brains, 45 grade II gliomas, 31 grade III gliomas, and 81 grade IV gliomas (GBM) for tGAS, and found GBMs to have the highest tGAS scores (Fig. 2A). In contrast, GLI1 levels were not significantly different among four groups (Fig. 2B). Next, we examined whether tGAS was associated with clinical outcome for patients with GBM using a TCGA GBM database and univariate/multivariate analyses, and observed that high tGAS resulted in high hazard ratios (HR; Fig. 2C), suggesting that tGLI1 activation is independently associated with poor overall survival of patients with GBM. Kaplan–Meier analysis further showed that patients with GBM with high tGAS were associated with worse overall survival compared to patients with low tGAS (Fig. 2D). In contrast, GLI1 mRNA was not associated with overall survival (Fig. 2E).

Figure 2.

High tGAS is associated with poor overall survival of patients with GBM and increased angiogenesis of GBM samples. A and B, tGLI1 is most activated in GBM across four grades of gliomas. tGAS scores (A) and GLI1 mRNA expression (B) were determined for patient tumors in the GSE4290 dataset across normal healthy brain and glioma samples. Student t test was used to compute P values. NS, nonsignificant. C, Increased tGLI1 activity is independently associated with poor overall survival of patients with GBM. Univariate and multivariate Cox proportional hazards were calculated using the tGAS score, GLI1 expression, patient age, patient sex, Karnofsky performance score (KPS), and tumor size as variables. CI, confidence interval; HR, hazard ratio. The TCGA GBM dataset was used, with the outcome variable being overall survival. D and E, Patients with high tGAS in their GBMs had worse overall survival compared with those with low tGAS. Kaplan–Meier survival graphs were drawn using a high or low tGAS score (D) or GLI1 expression (E) and data from the TCGA GBM dataset. Log-rank method was used to compute P values. F–I, tGAS positively correlated with GBM angiogenesis. The tGAS score (F and G) or GLI1 expression (H and I) was correlated with markers of tumor vascularity using CD31 (F and H) and VE-cadherin (G and I) using regression analysis. J and K, GBMs with high tGAS were enriched with angiogenesis gene signature. GSEA was performed using the AngioMatrix signature that is representative of GBM angiogenesis. Patients were separated by a high or low tGAS score (J) or GLI1 expression (K) using the TCGA GBM dataset. L and M, A positive correlation between tGAS and angiogenesis in GBM specimens. The AngioMatrix signature score was correlated with the tGAS score (L) or GLI1 expression (M) using the TCGA GBM dataset.

Figure 2.

High tGAS is associated with poor overall survival of patients with GBM and increased angiogenesis of GBM samples. A and B, tGLI1 is most activated in GBM across four grades of gliomas. tGAS scores (A) and GLI1 mRNA expression (B) were determined for patient tumors in the GSE4290 dataset across normal healthy brain and glioma samples. Student t test was used to compute P values. NS, nonsignificant. C, Increased tGLI1 activity is independently associated with poor overall survival of patients with GBM. Univariate and multivariate Cox proportional hazards were calculated using the tGAS score, GLI1 expression, patient age, patient sex, Karnofsky performance score (KPS), and tumor size as variables. CI, confidence interval; HR, hazard ratio. The TCGA GBM dataset was used, with the outcome variable being overall survival. D and E, Patients with high tGAS in their GBMs had worse overall survival compared with those with low tGAS. Kaplan–Meier survival graphs were drawn using a high or low tGAS score (D) or GLI1 expression (E) and data from the TCGA GBM dataset. Log-rank method was used to compute P values. F–I, tGAS positively correlated with GBM angiogenesis. The tGAS score (F and G) or GLI1 expression (H and I) was correlated with markers of tumor vascularity using CD31 (F and H) and VE-cadherin (G and I) using regression analysis. J and K, GBMs with high tGAS were enriched with angiogenesis gene signature. GSEA was performed using the AngioMatrix signature that is representative of GBM angiogenesis. Patients were separated by a high or low tGAS score (J) or GLI1 expression (K) using the TCGA GBM dataset. L and M, A positive correlation between tGAS and angiogenesis in GBM specimens. The AngioMatrix signature score was correlated with the tGAS score (L) or GLI1 expression (M) using the TCGA GBM dataset.

Close modal

Results in Fig. 1H indicated that tGLI1 promoted GBM vascularity. To confirm this observation, we correlated tGAS with vascularity markers, and found tGAS, but not GLI1, to significantly associate with CD31 and VE-cadherin (Fig. 2F–I). GSEA with the AngioMatrix signature that has been shown to be associated with tumor vascularity in GBM (25) further indicated that GBMs with high tGAS, but not high GLI1, had significant enrichment with the AngioMatrix signature (Fig. 2J–M). Results in Fig. 2 demonstrate that tGLI1, but not GLI1, is associated with poor overall survival and enhanced tumor angiogenesis in patients with GBM.

tGLI1 activation is enriched in the mesenchymal subtype of GBM

We examined whether the extent of tGLI1 activation differs among GBM subtypes. First, we divided the GBM cohort (TCGA) into two groups with high tGAS or low tGAS. We then used GSEA to determine the degrees of enrichment with established gene signatures for the four TCGA GBM subtypes, namely, mesenchymal, proneural, neural, and classical subtypes (4). As shown in Fig. 3A, GBM tumors with high tGAS were enriched for the TCGA mesenchymal gene signature, but not for the signatures for the other three subtypes. Interestingly, GBM tumors with low tGAS were significantly enriched for the TCGA classical signature, which is less aggressive than the mesenchymal subtype. Furthermore, we analyzed the same GBM cohort for the enrichment for another set of GBM subtype–specific gene signatures, namely, Phillips mesenchymal, proneural, and proliferation signatures (3). Consistent with the results of Fig. 3A, GBMs with high tGAS were enriched for the Phillips mesenchymal gene signature, but not the Phillips proneural or proliferation signatures (Fig. 3B). Mesenchymal GBMs had the highest tGAS scores among the four TCGA subtypes (Fig. 3C). In contrast, high GLI1 expression was not associated with either TCGA or Phillips mesenchymal subtype, but associated with the TCGA classical subtype (Fig. 3D). GBMs with low GLI1 were enriched with both TCGA proneural and Phillips proneural gene signatures (Fig. 3E). These findings indicate that tGLI1 activation is a hallmark of mesenchymal GBM.

Figure 3.

GBMs with high tGAS are enriched with the mesenchymal GBM gene signatures. The TCGA dataset with the gene expression profile of 165 GBMs was used. A and B, GBM tumors with high tGAS were enriched for the TCGA and Phillips mesenchymal gene signatures, but not for those for the other subtypes. Heat maps were drawn using Morpheus software, with patients separated by a high or low tGAS score using genes in the signatures for each of the four TCGA GBM subtypes (A) or the three Phillips GBM subtypes (B). Right, GSEA. C, The mesenchymal subtype of GBMs had the highest tGAS scores among the four TCGA subtypes. The tGAS score was determined for the four TCGA GBM subtypes. D and E, High GLI1 mRNA expression was not associated with either the TCGA or Phillips mesenchymal subtype, but was associated with the TCGA classical subtype. GSEA was performed using the signatures for the TCGA GBM subtypes (D) or the Phillips GBM subtypes (E), and patients were separated by high or low GLI1 mRNA expression.

Figure 3.

GBMs with high tGAS are enriched with the mesenchymal GBM gene signatures. The TCGA dataset with the gene expression profile of 165 GBMs was used. A and B, GBM tumors with high tGAS were enriched for the TCGA and Phillips mesenchymal gene signatures, but not for those for the other subtypes. Heat maps were drawn using Morpheus software, with patients separated by a high or low tGAS score using genes in the signatures for each of the four TCGA GBM subtypes (A) or the three Phillips GBM subtypes (B). Right, GSEA. C, The mesenchymal subtype of GBMs had the highest tGAS scores among the four TCGA subtypes. The tGAS score was determined for the four TCGA GBM subtypes. D and E, High GLI1 mRNA expression was not associated with either the TCGA or Phillips mesenchymal subtype, but was associated with the TCGA classical subtype. GSEA was performed using the signatures for the TCGA GBM subtypes (D) or the Phillips GBM subtypes (E), and patients were separated by high or low GLI1 mRNA expression.

Close modal

tGLI1 promotes neurosphere formation and transcriptionally activates CD44 expression

Mesenchymal GBM is associated with poor patient survival and multidrug resistance—key clinical aspects of GBM thought to be driven by the cancer stem cell subpopulation (3, 5, 26). The role of GLI1 in GSC function has been studied; however, the role that tGLI1 plays in GSCs is unknown. We first compared monolayer culture with GSC-containing neurospheres for tGLI1 and GLI1 expression, and found tGLI1, but not GLI1, to be enriched in neurospheres (Fig. 4A). We further found that tGLI1-overexpressing cells formed significantly more neurospheres (Fig. 4B and C). Furthermore, we specifically knocked down tGLI1 expression in U373MG GBM cells with high endogenous tGLI1 expression to further determine the importance of tGLI1 in GBM. As shown in Fig. 4D, we designed and used a tGLI1-targeting LNA phosphorothioated oligo that has been chemically modified to resist nuclease-mediated degradation. As shown in Fig. 4D (left), the tGLI1-targeting LNA oligo specifically knocked down tGLI1 expression but had no effect on GLI1. Importantly, GBM cells with tGLI1 knockdown showed a reduced ability to form neurospheres compared to cells with the nontargeting control LNA oligo (Fig. 4D, right). Blast analysis did not reveal binding of the control oligos to any genes. These results further support our conclusion that endogenous tGLI1 is essential for the neurosphere-forming ability of GBM cells.

Figure 4.

tGLI1 promotes neurosphere formation and transcriptionally activates CD44 expression. A, tGLI1 was significantly enriched in the neurospheres compared with the monolayer GBM cells. G48LL2 and BTCOE 4810 cell lines were collected under monolayer and neurosphere-forming conditions. Total RNA from cells was subjected to qPCR for tGLI1 and GLI1 levels. B and C, tGLI1-overexpressing cells formed significantly more neurospheres, suggesting an increase in the stem-like cell population. G48LL2 (B) and U373MG (C) cells with stable expression of vector, GLI1, or tGLI1 were subjected to the neurosphere assay. D, tGLI1 knockdown significantly inhibited neurosphere formation. U373MG cells transfected with the tGLI1-targeting or nontargeting control LNA oligos were subjected to the neurosphere assay. E, tGLI1 enhanced CD44 expression. Total RNA from G48a cells with stable expression of either vector or tGLI1 was subjected to qPCR for the indicated genes. F, tGLI1-expressing GBM cells had increased CD44+ cells. Isogenic G48LL2 cell lines with stable expression of control or tGLI1 vector were subjected to flow cytometry with CD44 or CD133 antibodies. G, Preferential binding of tGLI1, but not GLI1, to the CD44 promoter. U373MG cells with transient expression of control vector or tGLI1 were subjected to the ChIP assay, followed by PCR using primers for three regions of the CD44 gene promoter. H, tGLI1 transactivated the CD44 promoter in two GBM cell lines and HEK293 cells. Cells were transiently transfected with control or tGLI1 vector along with the CD44 promoter luciferase reporter, stimulated with SHH (100 ng/mL) for 4 hours, and subjected to the luciferase assay. I, SHH stimulation enhanced tGLI1-mediated activation of the CD44 gene promoter. BTCOE 4795 and U373MG cell lines were transiently transfected with control or tGLI1 vector together with the CD44 promoter luciferase reporter. Cells were then treated with or without SHH (100 ng/mL) for 4 hours, harvested, and subjected to the luciferase assay. Student t test was performed to calculate P values. NS, nonsignificant. All experiments were done at least three times to derive means and SDs.

Figure 4.

tGLI1 promotes neurosphere formation and transcriptionally activates CD44 expression. A, tGLI1 was significantly enriched in the neurospheres compared with the monolayer GBM cells. G48LL2 and BTCOE 4810 cell lines were collected under monolayer and neurosphere-forming conditions. Total RNA from cells was subjected to qPCR for tGLI1 and GLI1 levels. B and C, tGLI1-overexpressing cells formed significantly more neurospheres, suggesting an increase in the stem-like cell population. G48LL2 (B) and U373MG (C) cells with stable expression of vector, GLI1, or tGLI1 were subjected to the neurosphere assay. D, tGLI1 knockdown significantly inhibited neurosphere formation. U373MG cells transfected with the tGLI1-targeting or nontargeting control LNA oligos were subjected to the neurosphere assay. E, tGLI1 enhanced CD44 expression. Total RNA from G48a cells with stable expression of either vector or tGLI1 was subjected to qPCR for the indicated genes. F, tGLI1-expressing GBM cells had increased CD44+ cells. Isogenic G48LL2 cell lines with stable expression of control or tGLI1 vector were subjected to flow cytometry with CD44 or CD133 antibodies. G, Preferential binding of tGLI1, but not GLI1, to the CD44 promoter. U373MG cells with transient expression of control vector or tGLI1 were subjected to the ChIP assay, followed by PCR using primers for three regions of the CD44 gene promoter. H, tGLI1 transactivated the CD44 promoter in two GBM cell lines and HEK293 cells. Cells were transiently transfected with control or tGLI1 vector along with the CD44 promoter luciferase reporter, stimulated with SHH (100 ng/mL) for 4 hours, and subjected to the luciferase assay. I, SHH stimulation enhanced tGLI1-mediated activation of the CD44 gene promoter. BTCOE 4795 and U373MG cell lines were transiently transfected with control or tGLI1 vector together with the CD44 promoter luciferase reporter. Cells were then treated with or without SHH (100 ng/mL) for 4 hours, harvested, and subjected to the luciferase assay. Student t test was performed to calculate P values. NS, nonsignificant. All experiments were done at least three times to derive means and SDs.

Close modal

To identify the mechanism by which tGLI1 promotes a GBM stem cell phenotype, we determined the effects of tGLI1 on known stem cell–related genes and found that tGLI1-overexpressing GBM cells showed increased expression of CD44, along with a decrease in Sox2, Nanog, OCT4, and CD133 expression (Fig. 4E; Supplementary Table S1). Flow cytometry confirmed increased CD44+ cells in tGLI1-expressing GBM cells but no significant change in the CD133+ population (Fig. 4F). CD44 is regarded as the marker for the mesenchymal GSCs, whereas CD133 is the marker for the proneural GSCs (8). Using the chromatin immunopreciptation (ChIP) assay, we detected preferential binding of tGLI1, but not GLI1, to three regions of the CD44 gene promoter (Fig. 4G; Supplementary Fig. S1), which show no homology to consensus GLI1-binding sequences. Using the luciferase assay, we showed that tGLI1 transactivated the CD44 promoter in two SHH-stimulated GBM cell lines and HEK293 cells (Fig. 4H), and that SHH enhanced tGLI1-mediated activation of the CD44 gene promoter (Fig. 4I; Supplementary Table S2). Results in Fig. 4 indicate a novel, important role that tGLI1 plays in GBM stem–like cells and in the transcriptional activation of CD44.

tGLI1 is preferentially expressed and activated in the mesenchymal subtype of GSC

Recent studies classified GSCs isolated from GBM specimens into the proneural or mesenchymal subtypes based on gene expression profiles (8, 9). CD44 was defined as the marker for the mesenchymal GSCs, whereas CD133 was for the proneural GSCs. Because tGLI1 binds to and transactivates the CD44 promoter and tGLI1 is preferentially activated in mesenchymal GBM, we determined whether tGLI1 expression is associated with the mesenchymal subtype of GSC. Here, we examined patient-derived GSCs, two mesenchymal and two proneural GSC neurospheres, and found the mesenchymal GSC neurospheres to express higher levels of tGLI1 and lower levels of GLI1 compared to proneural GSC neurospheres (Fig. 5A). We also observed that mesenchymal GSC neurospheres had higher tGAS than the proneural GSC neurospheres (Fig. 5B). We further analyzed a TCGA GBM dataset for the relationship between tGAS and GSC signatures, and observed that GBMs with high tGAS are enriched for the mesenchymal GSC signature, but not the proneural GSC signature (Fig. 5C, left). In contrast, GLI1 was not enriched for either GSC signature (Fig. 5C, right).

Figure 5.

tGLI1 is preferentially expressed and activated in the mesenchymal subtype of GSCs. A, Mesenchymal (MES) GSC neurospheres expressed higher levels of tGLI1 and lower levels of GLI1 compared with proneural (PN) GSC neurospheres. Four different GSCs previously characterized as mesenchymal or proneural subtype were subjected to total RNA extraction, followed by RT-qPCR for tGLI1 and GLI1 expression levels. B, tGAS was significantly higher in mesenchymal GSC neurospheres (GSC-20 and GSC-28) compared with the proneural GSC neurospheres (GSC-11 and GSC-23). tGAS scores were calculated for proneural GSC (n = 11) and mesenchymal GSC (n = 6) lines that were isolated and profiled for expression by Bhat and colleagues (8). C, GBMs with high tGAS are enriched for the mesenchymal GSC signature, but not the proneural GSC signature. The TCGA GBM dataset was analyzed by GSEA for the extent of enrichment with the mesenchymal and proneural GSC signatures (8). Patients were divided into two groups according to tGAS (left) or GLI1 expression (right). D and E, tGLI1 overexpression increased neurosphere-forming capability of both proneural (D) and mesenchymal (E) GSCs. GSC-11 (PN) and GSC-28 (MES) neurospheres transiently transfected with control vector or tGLI1 vector were subjected to the neurosphere assay (left) and RT-qPCR for tGLI1 expression levels (right). F and G, tGLI1 increased CD44 and decreased CD133 expression at the mRNA (F) and protein (G) levels. H, High tGLI1-expressing MES GSC neurospheres expressed higher levels of CD44 and lower levels of CD133 compared with low tGLI1-expressing PN GSC neurospheres. Extracted total RNA was subjected to RT-qPCR for expression of tGLI1, CD44, and CD133. I, A positive correlation between tGAS and CD44, but not CD133, in the GBM cohort (N = 165). The tGAS score was correlated with expression levels of CD44 or CD133 in the TCGA GBM dataset using Pearson correlation. Student t test was used to calculate P values.

Figure 5.

tGLI1 is preferentially expressed and activated in the mesenchymal subtype of GSCs. A, Mesenchymal (MES) GSC neurospheres expressed higher levels of tGLI1 and lower levels of GLI1 compared with proneural (PN) GSC neurospheres. Four different GSCs previously characterized as mesenchymal or proneural subtype were subjected to total RNA extraction, followed by RT-qPCR for tGLI1 and GLI1 expression levels. B, tGAS was significantly higher in mesenchymal GSC neurospheres (GSC-20 and GSC-28) compared with the proneural GSC neurospheres (GSC-11 and GSC-23). tGAS scores were calculated for proneural GSC (n = 11) and mesenchymal GSC (n = 6) lines that were isolated and profiled for expression by Bhat and colleagues (8). C, GBMs with high tGAS are enriched for the mesenchymal GSC signature, but not the proneural GSC signature. The TCGA GBM dataset was analyzed by GSEA for the extent of enrichment with the mesenchymal and proneural GSC signatures (8). Patients were divided into two groups according to tGAS (left) or GLI1 expression (right). D and E, tGLI1 overexpression increased neurosphere-forming capability of both proneural (D) and mesenchymal (E) GSCs. GSC-11 (PN) and GSC-28 (MES) neurospheres transiently transfected with control vector or tGLI1 vector were subjected to the neurosphere assay (left) and RT-qPCR for tGLI1 expression levels (right). F and G, tGLI1 increased CD44 and decreased CD133 expression at the mRNA (F) and protein (G) levels. H, High tGLI1-expressing MES GSC neurospheres expressed higher levels of CD44 and lower levels of CD133 compared with low tGLI1-expressing PN GSC neurospheres. Extracted total RNA was subjected to RT-qPCR for expression of tGLI1, CD44, and CD133. I, A positive correlation between tGAS and CD44, but not CD133, in the GBM cohort (N = 165). The tGAS score was correlated with expression levels of CD44 or CD133 in the TCGA GBM dataset using Pearson correlation. Student t test was used to calculate P values.

Close modal

We found that tGLI1 ectopic expression enhanced the neurosphere-forming capability of both proneural and mesenchymal GSCs (Fig. 5D and E), and led to an increase in CD44 expression and a decrease in CD133 expression at both the mRNA and protein levels (Fig. 5F and G). Further analysis of the four different GSCs indicated a pattern where high tGLI1-expressing mesenchymal GSC neurospheres expressed higher levels of CD44 and lower levels of CD133 compared with low tGLI1-expressing proneural GSC neurospheres (Fig. 5H). Consistent with these observations, analysis of the TCGA GBM dataset revealed a positive correlation between tGAS and CD44, but not tGAS and CD133 (Fig. 5I). Collectively, results in Fig. 5 demonstrate that tGLI1 is preferentially expressed and activated in the mesenchymal GSC neurospheres over proneural GSC neurospheres, enhances expression of CD44, a mesenchymal GSC marker, and positively correlates with CD44 expression in a GBM cohort.

Increased tGLI1 expression enhanced the propensity of mesenchymal GSC to form xenografts

We have shown that tGLI1 ectopic expression promoted GBM growth in an orthotopic mouse model and that tGLI1 made GSCs form greater numbers of neurospheres. In light of these observations, we aimed to validate our GSC findings in vivo. We first generated three isogenic mesenchymal GSC-28 isogenic lines with stable expression of control, GLI1, or tGLI1 lentiviral vectors (Fig. 6A). Of note, GSC-28 neurospheres express high endogenous tGLI1 (see Fig. 5A). Expression levels for GLI1 and tGLI1 in these stable GSC lines are similar to those found in GBM specimens, as shown by IHC (Fig. 6A, bottom). GSC neurospheres were harvested and disassociated into single-cell suspension and were then implanted into the mouse brains; mice were imaged weekly. As shown in the growth curves and representative tumor images in Fig. 6B–D, increased tGLI1 expression rendered GSC-28 cells more aggressive in growths. Mice bearing tGLI1-expressing GSC-28 xenografts had a shortened survival time (Fig. 6E). IHC analyses further demonstrated that tGLI1-expressing GSC-28 tumors had the highest proliferative index (Ki-67 IHC) and microvessel density (CD31 IHC; Fig. 6F–J). In summary, these results indicate that tGLI1 renders GSCs more aggressive in growth and angiogenesis in the brain microenvironment.

Figure 6.

Increased tGLI1 expression enhanced the propensity of GSC to form xenografts. A, Generation of isogenic GSC-28 cells with stable expression of control, GLI1, or tGLI1 vector. GSC neurospheres were analyzed by immunoblotting for GLI1 and tGLI1 levels (top). Expression levels for GLI1 and tGLI1 in these GSC lines are similar to those found in GBM specimens as shown by IHC (bottom). All GSC lines were maintained as neurospheres. B, GSC-28 cells expressing tGLI1 formed larger tumors compared with GLI1- or vector-expressing cells. Isogenic lines were implanted into the right frontal lobe of nude mice (N = 6 per group) and tumor growth was assessed weekly via bioluminescent imaging. C, Representative images of actively growing tumors from animals. D, Representative images of ex vivo mouse brains. E, Mice bearing ectopic tGLI1-expressing GSC-28 xenografts had a shortened survival time. Kaplan–Meier survival graph is shown. Log-rank test was used to determine P values. F, Mouse brains were subjected to hematoxylin and eosin (H&E) staining and IHC with indicated antibodies. Representative images are shown. G–J, tGLI1-expressing GSC-28 tumors had the highest proliferative index and microvessel density. Immunostained mouse brains were scored to determine H-scores. Student t test was used to compute P values. NS, nonsignificant.

Figure 6.

Increased tGLI1 expression enhanced the propensity of GSC to form xenografts. A, Generation of isogenic GSC-28 cells with stable expression of control, GLI1, or tGLI1 vector. GSC neurospheres were analyzed by immunoblotting for GLI1 and tGLI1 levels (top). Expression levels for GLI1 and tGLI1 in these GSC lines are similar to those found in GBM specimens as shown by IHC (bottom). All GSC lines were maintained as neurospheres. B, GSC-28 cells expressing tGLI1 formed larger tumors compared with GLI1- or vector-expressing cells. Isogenic lines were implanted into the right frontal lobe of nude mice (N = 6 per group) and tumor growth was assessed weekly via bioluminescent imaging. C, Representative images of actively growing tumors from animals. D, Representative images of ex vivo mouse brains. E, Mice bearing ectopic tGLI1-expressing GSC-28 xenografts had a shortened survival time. Kaplan–Meier survival graph is shown. Log-rank test was used to determine P values. F, Mouse brains were subjected to hematoxylin and eosin (H&E) staining and IHC with indicated antibodies. Representative images are shown. G–J, tGLI1-expressing GSC-28 tumors had the highest proliferative index and microvessel density. Immunostained mouse brains were scored to determine H-scores. Student t test was used to compute P values. NS, nonsignificant.

Close modal

We made the following important novel observations in this study: (i) tGLI1 promotes the neurosphere-forming ability of GBM and GSCs in vitro, and their intracranial growth and angiogenesis in vivo; (ii) tGLI1 is highly expressed in GBM but undetectable in normal brain, whereas GLI1 is equally expressed in both tissues; (iii) tGLI1 is predominantly activated in the mesenchymal subtype of GBM and GSCs that are more aggressive among different subtypes (4); (iv) patients with GBM with high tGLI1 activity in their tumors had shortened overall survival and increased tumor angiogenesis compared with those with low tGLI1 activity; and (v) tGLI1 functions as a transcriptional activator of CD44. By reporting these findings, our study advances the biological understanding of GBM and GSCs and transcriptional regulation of an important stem cell marker/mediator, CD44.

Our bioinformatics analyses revealed tGLI1 activation as a hallmark of mesenchymal GBM and GSCs, which is an important finding. We used two independent mesenchymal gene signatures and found both signatures to be highly enriched in GBMs and GSCs with high tGAS, an indicator for tGLI1 transcriptional activity. Of note, the classification of GBM into distinct subtypes was first reported by Phillips and colleagues in 2006, with three distinct subtypes (3), whereas a more recently study in 2010 by Verhaak and colleagues classified GBM into four distinct subtypes (4). Both studies identified a common subtype named the mesenchymal subtype according to the expression of genes associated with a mesenchymal cell phenotype. Notably, several earlier studies identified mediators and markers for the mesenchymal subtype of GBM and GSCs. For example, RTVP-1 was found to express at a higher level in mesenchymal GBM associated with tumor recurrence and poor clinical outcome (27). RTVP-1 overexpression induced mesenchymal differentiation of human neural stem cells, whereas silencing RTVP-1 inhibited the mesenchymal transformation and stemness of GSCs. An important future task is to explore the potential cross-talk between RTVP-1 and tGLI1 in regulating mesenchymal GSCs.

It is also important future task to investigate the potential interactions between tGLI1 and TNFα/NF-κB in light of the observations that both are enriched in CD44+ mesenchymal GSCs and that the proneural GSCs can undergo differentiation to a mesenchymal state in a TNFα/NF-κB–dependent manner (8, 28). Most recently, S100A4 was reported as a novel biomarker of GSCs that is enriched in cells with tumor-initiating and sphere-forming abilities; selective ablation of S100A4-expressing cells blocked tumor growth in vitro and in vivo (29). Whether tGLI1 crosstalks with S100A4, thereby promoting mesenchymal GSCs is unknown, but this line of future research is warranted.

Mesenchymal GBM and GSCs are not only more tumorigenic but also more resistant to radiation therapy (8). Given the ability of tGLI1 to promote mesenchymal GBM and GSCs, it is possible that tGLI1-expressing cells are more resistant to radiation therapy. Although the direct role of tGLI1 in radiation resistance has not been reported, it has been shown that tumor cells with hyperactive SHH-GLI1 signaling are more resistant to radiation therapy (30–32). Because tGLI1 functions as a gain-of-function GLI1 and tGLI1 has a higher propensity than GLI1 to promote GSCs in vitro and in vivo, we speculate that tGLI1+ GSCs are more resistant to radiation therapy compared to GLI1+ GSCs, which could be tested in future studies.

CD44 is regarded as a marker for cancer stem cells for a number of cancers, including breast cancer, pancreatic cancer, and GBM (8, 33, 34), and is associated with tumor initiation and progression (33, 35–37). However, emerging evidence suggests that CD44 contributes to the stem cell phenotype via various mechanisms, such as osteopontin signaling and promotion of HIF2α activity (38). However, transcriptional regulation of the CD44 gene is still not well understood. It has been reported that the EGR1 transcription factor can induce CD44 expression in antigen-stimulated B cells (39). NF-κB has been shown to upregulate CD44 expression in GBM (8) and breast cancer (40). NF-κB cooperates with AP-1 to bind to a cis-element of the CD44 promoter, leading to CD44 expression in breast cancer in a cell type–specific manner (41). Y-box binding protein-1 transcription factor induces CD44 expression in breast cancers (42). Surprisingly, the FOXO3 tumor suppressor has been shown to induce CD44 expression in pancreatic cancer (43). Our discovery that tGLI1 directly activates CD44 gene expression sheds important new light into the molecular mechanisms contributing to high CD44 expression in GSCs and possibly cancer stem cells in other tumor types.

Our present and previous studies uncovered that tGLI1 is expressed in a tumor-specific fashion in GBM and breast cancer (13–17). Expression of tGLI1 in malignant gliomas (20) and breast cancer (19) has been confirmed by other groups. Interestingly, a recent study detected tGLI1 in metastatic hepatoma cells (21). The mechanisms for the tumor-specific expression of tGLI1 are, however, still not elucidated. Since the splicing machinery is highly dysregulated in cancers (44), it is likely that the splicing factors that synthesize tGLI1 are aberrantly overexpressed in GBM, breast cancer, and metastatic hepatomas. Identification of these splicing factors constitutes an important task that could lead to strategies that inhibit tumor progression through inhibiting tGLI1 production.

Evidence from our laboratory and those of other groups suggests that tGLI1 may be regarded as a novel therapeutic target for multiple cancer types (13–17, 19, 21, 45). This notion is supported by the observations that tGLI1 is expressed tumor specifically and that tGLI1 plays an important role in tumor growth, angiogenesis and cancer stem cell renewal. We further speculate that targeting tGLI1 rather than GLI1 could minimize normal tissue toxicity because tGLI1 is only detected in cancer tissues, whereas GLI1 is expressed in both cancerous and normal tissues. Smoothened inhibitors can inhibit both tGLI1 and GLI1; however, their efficacy is only modest in GBM and breast cancer due to their inability to suppress noncanonical smoothened-independent activation of tGLI1 and GLI1 (46). Our study provides novel insights into the biology of GBM and GSCs, particularly those belonging to the mesenchymal subtype, defines tGLI1 as a novel mediator of GBM growth and GSC self-renewal, and establishes tGLI1 as a novel transcriptional activator of CD44.

No potential conflicts of interest were disclosed.

Conception and design: R.L. Carpenter, H.-W. Lo

Development of methodology: R.L. Carpenter, D. Zhu

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): T.K. Rimkus, R.L. Carpenter, S. Sirkisoon, D. Zhu, S.B. Tatter

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): T.K. Rimkus, R.L. Carpenter, D. Zhu, M.D. Chan, G.J. Lesser, W. Debinski

Writing, review, and/or revision of the manuscript: T.K. Rimkus, R.L. Carpenter, B.C. Pasche, M.D. Chan, G.J. Lesser, S.B. Tatter, K. Watabe, W. Debinski, H.-W. Lo

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): R.L. Carpenter, D. Zhu

Study supervision: R.L. Carpenter, H.-W. Lo

The authors thank Drs. Erik Sulman and Krishna Bhat at The University of Texas MD Anderson Cancer Center (Houston, TX) for gifting us the GSC cells. The authors also acknowledge the financial support from the NIH: R01NS087169 to H.-W. Lo; T32CA079448 to R.L. Carpenter; R01NS087169-3S1 to H.-W. Lo and S.R. Sirkisoon; P30CA012197 to B.C. Pasche; and from the U.S. Department of Defense: W81XWH-17-1-0044 to H.-W. Lo.

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.
Kumthekar
PU
,
Macrie
BD
,
Singh
SK
,
Kaur
G
,
Chandler
JP
,
Sejpal
SV
. 
A review of management strategies of malignant gliomas in the elderly population
.
Am J Cancer Res
2014
;
4
:
436
44
.
2.
Ostrom
QT
,
Gittleman
H
,
Farah
P
,
Ondracek
A
,
Chen
Y
,
Wolinsky
Y
, et al
CBTRUS statistical report: primary brain and central nervous system tumors diagnosed in the United States in 2006–2010
.
Neuro Oncol
2013
;
15
:
ii1
56
.
3.
Phillips
HS
,
Kharbanda
S
,
Chen
R
,
Forrest
WF
,
Soriano
RH
,
Wu
TD
, et al
Molecular subclasses of high-grade glioma predict prognosis, delineate a pattern of disease progression, and resemble stages in neurogenesis
.
Cancer Cell
2006
;
9
:
157
73
.
4.
Verhaak
RGW
,
Hoadley
KA
,
Purdom
E
,
Wang
V
,
Qi
Y
,
Wilkerson
MD
, et al
An integrated genomic analysis identifies clinically relevant subtypes of glioblastoma characterized by abnormalities in PDGFRA, IDH1, EGFR and NF1
.
Cancer Cell
2010
;
17
:
98
110
.
5.
Colman
H
,
Zhang
L
,
Sulman
EP
,
McDonald
JM
,
Shooshtari
NL
,
Rivera
A
, et al
A multigene predictor of outcome in glioblastoma
.
Neuro Oncol
2010
;
12
:
49
57
.
6.
Lathia
JD
,
Mack
SC
,
Mulkearns-Hubert
EE
,
Valentim
CLL
,
Rich
JN
. 
Cancer stem cells in glioblastoma
.
Genes Dev
2015
;
29
:
1203
17
.
7.
Stopschinski
BE
,
Beier
CP
,
Beier
D
. 
Glioblastoma cancer stem cells–from concept to clinical application
.
Cancer Lett
2013
;
338
:
32
40
.
8.
Bhat
KP
,
Balasubramaniyan
V
,
Vaillant
B
,
Ezhilarasan
R
,
Hummelink
K
,
Hollingsworth
F
, et al
Mesenchymal differentiation mediated by NF-kappaB promotes radiation resistance in glioblastoma
.
Cancer Cell
2013
;
24
:
331
46
.
9.
Mao
P
,
Joshi
K
,
Li
J
,
Kim
SH
,
Li
P
,
Santana-Santos
L
, et al
Mesenchymal glioma stem cells are maintained by activated glycolytic metabolism involving aldehyde dehydrogenase 1A3
.
Proc Natl Acad Sci U S A
2013
;
110
:
8644
9
.
10.
Mooney
KL
,
Choy
W
,
Sidhu
S
,
Pelargos
P
,
Bui
TT
,
Voth
B
, et al
The role of CD44 in glioblastoma multiforme
.
J Clin Neurosci
2016
;
34
:
1
5
.
11.
Clement
V
,
Sanchez
P
,
de Tribolet
N
,
Radovanovic
I
,
Ruiz i Altaba
A
. 
HEDGEHOG-GLI1 signaling regulates human glioma growth, cancer stem cell self-renewal, and tumorigenicity
.
Curr Biol
2007
;
17
:
165
72
.
12.
Stecca
B
,
Ruiz
IAA
. 
Context-dependent regulation of the GLI code in cancer by HEDGEHOG and non-HEDGEHOG signals
.
J Mol Cell Biol
2010
;
2
:
84
95
.
13.
Lo
HW
,
Zhu
H
,
Cao
X
,
Aldrich
A
,
Ali-Osman
F
. 
A novel splice variant of GLI1 that promotes glioblastoma cell migration and invasion
.
Cancer Res
2009
;
69
:
6790
8
.
14.
Cao
X
,
Geradts
J
,
Dewhirst
M
,
Lo
H-W
. 
Upregulation of VEGF-A and CD24 gene expression by the tGLI1 transcription factor contributes to the aggressive behavior of breast cancer cells
.
Oncogene
2012
;
31
:
104
15
.
15.
Carpenter
RL
,
Paw
I
,
Zhu
H
,
Sirkisoon
S
,
Xing
F
,
Watabe
K
, et al
The gain-of-function GLI1 transcription factor TGLI1 enhances expression of VEGF-C and TEM7 to promote glioblastoma angiogenesis
.
Oncotarget
2015
;
6
:
22653
65
.
16.
Han
W
,
Carpenter
RL
,
Lo
H-W
. 
TGLI1 upregulates expression of VEGFR2 and VEGF-A, leading to a robust VEGF-VEGFR2 autocrine loop and cancer cell growth
.
Cancer Hallmarks
2013
;
1
:
28
37
.
17.
Zhu
H
,
Carpenter
RL
,
Han
W
,
Lo
H-W
. 
The GLI1 splice variant TGLI1 promotes glioblastoma angiogenesis and growth
.
Cancer Lett
2014
;
343
:
51
61
.
18.
Carpenter
RL
,
Lo
HW
. 
Hedgehog pathway and GLI1 isoforms in human cancer
.
Discov Med
2012
;
13
:
105
13
.
19.
Di Mauro
C
,
Rosa
R
,
D'Amato
V
,
Ciciola
P
,
Servetto
A
,
Marciano
R
, et al
Hedgehog signalling pathway orchestrates angiogenesis in triple-negative breast cancers
.
Br J Cancer
2017
;
116
:
1425
35
.
20.
Gricius
D
,
Kazlauskas
A
. 
Analysis of oncogene GLI1 protein expression levels between differing grades of astrocytoma
.
BIOLOGIJA
2014
;
60
:
142
7
.
21.
Fan
YH
,
Ding
J
,
Nguyen
S
,
Liu
XJ
,
Xu
G
,
Zhou
HY
, et al
Aberrant hedgehog signaling is responsible for the highly invasive behavior of a subpopulation of hepatoma cells
.
Oncogene
2016
;
35
:
116
24
.
22.
Ruiz i Altaba
A
. 
Hedgehog signaling and the Gli code in stem cells, cancer, and metastases
.
Sci Signal
2011
;
4
:
pt9
.
23.
Debinski
W
,
Gibo
DM
. 
Fos-related antigen 1 modulates malignant features of glioma cells
.
Mol Cancer Res
2005
;
3
:
237
49
.
24.
Ferluga
S
,
Tomé
CML
,
Herpai
DM
,
D'Agostino
R
,
Debinski
W
. 
Simultaneous targeting of Eph receptors in glioblastoma
.
Oncotarget
2016
;
7
:
59860
76
.
25.
Langlois
B
,
Saupe
F
,
Rupp
T
,
Arnold
C
,
van der Heyden
M
,
Orend
G
, et al
AngioMatrix, a signature of the tumor angiogenic switch-specific matrisome, correlates with poor prognosis for glioma and colorectal cancer patients
.
Oncotarget
2014
;
5
:
10529
45
.
26.
Pelloski
CE
,
Mahajan
A
,
Maor
M
,
Chang
EL
,
Woo
S
,
Gilbert
M
, et al
YKL-40 expression is associated with poorer response to radiation and shorter overall survival in glioblastoma
.
Clin Cancer Res
2005
;
11
:
3326
34
.
27.
Giladi
ND
,
Ziv-Av
A
,
Lee
HK
,
Finniss
S
,
Cazacu
S
,
Xiang
C
, et al
RTVP-1 promotes mesenchymal transformation of glioma via a STAT-3/IL-6-dependent positive feedback loop
.
Oncotarget
2015
;
6
:
22680
97
.
28.
Kim
SH
,
Ezhilarasan
R
,
Phillips
E
,
Gallego-Perez
D
,
Sparks
A
,
Taylor
D
, et al
Serine/threonine kinase MLK4 determines mesenchymal identity in glioma stem cells in an NF-kappaB-dependent manner
.
Cancer Cell
2016
;
29
:
201
13
.
29.
Chow
KH
,
Park
HJ
,
George
J
,
Yamamoto
K
,
Gallup
AD
,
Graber
JH
, et al
S100A4 is a biomarker and regulator of glioma stem cells that is critical for mesenchymal transition in glioblastoma
.
Cancer Res
2017
;
77
:
5360
73
.
30.
Zhou
J
,
Wu
K
,
Gao
D
,
Zhu
G
,
Wu
D
,
Wang
X
, et al
Reciprocal regulation of hypoxia-inducible factor 2alpha and GLI1 expression associated with the radioresistance of renal cell carcinoma
.
Int J Radiat Oncol Biol Phys
2014
;
90
:
942
51
.
31.
Gan
GN
,
Eagles
J
,
Keysar
SB
,
Wang
G
,
Glogowska
MJ
,
Altunbas
C
, et al
Hedgehog signaling drives radioresistance and stroma-driven tumor repopulation in head and neck squamous cancers
.
Cancer Res
2014
;
74
:
7024
36
.
32.
Chen
YJ
,
Lin
CP
,
Hsu
ML
,
Shieh
HR
,
Chao
NK
,
Chao
KS
. 
Sonic hedgehog signaling protects human hepatocellular carcinoma cells against ionizing radiation in an autocrine manner
.
Int J Radiat Oncol Biol Phys
2011
;
80
:
851
9
.
33.
Jijiwa
M
,
Demir
H
,
Gupta
S
,
Leung
C
,
Joshi
K
,
Orozco
N
, et al
CD44v6 regulates growth of brain tumor stem cells partially through the AKT-mediated pathway
.
PLoS One
2011
;
6
:
e24217
.
34.
Li
C
,
Heidt
DG
,
Dalerba
P
,
Burant
CF
,
Zhang
L
,
Adsay
V
, et al
Identification of pancreatic cancer stem cells
.
Cancer Res
2007
;
67
:
1030
7
.
35.
Bourguignon
LYW
,
Peyrollier
K
,
Xia
W
,
Gilad
E
. 
Hyaluronan-CD44 interaction activates stem cell marker nanog, stat-3-mediated MDR1 gene expression, and ankyrin-regulated multidrug efflux in breast and ovarian tumor cells
.
J Biol Chem
2008
;
283
:
17635
51
.
36.
Breyer
R
,
Hussein
S
,
Radu
DL
,
Putz
KM
,
Gunia
S
,
Hecker
H
, et al
Disruption of intracerebral progression of C6 rat glioblastoma by in vivo treatment with anti-CD44 monoclonal antibody
.
J Neurosurg
2000
;
92
:
140
9
.
37.
Prince
ME
,
Sivanandan
R
,
Kaczorowski
A
,
Wolf
GT
,
Kaplan
MJ
,
Dalerba
P
, et al
Identification of a subpopulation of cells with cancer stem cell properties in head and neck squamous cell carcinoma
.
Proc Natl Acad Sci U S A
2007
;
104
:
973
8
.
38.
Pietras
A
,
Katz
AM
,
Ekstrom
EJ
,
Wee
B
,
Halliday
JJ
,
Pitter
KL
, et al
Osteopontin-CD44 signaling in the glioma perivascular niche enhances cancer stem cell phenotypes and promotes aggressive tumor growth
.
Cell Stem Cell
2014
;
14
:
357
69
.
39.
Maltzman
JS
,
Carman
JA
,
Monroe
JG
. 
Role of EGR1 in regulation of stimulus-dependent CD44 transcription in B lymphocytes
.
Mol Cell Biol
1996
;
16
:
2283
94
.
40.
Smith
SM
,
Lyu
YL
,
Cai
L
. 
NF-kappaB affects proliferation and invasiveness of breast cancer cells by regulating CD44 expression
.
PLoS One
2014
;
9
:
e106966
.
41.
Smith
SM
,
Cai
L
. 
Cell specific CD44 expression in breast cancer requires the interaction of AP-1 and NFkappaB with a novel cis-element
.
PLoS One
2012
;
7
:
e50867
.
42.
To
K
,
Fotovati
A
,
Reipas
KM
,
Law
JH
,
Hu
K
,
Wang
J
, et al
Y-box binding protein-1 induces the expression of CD44 and CD49f leading to enhanced self-renewal, mammosphere growth, and drug resistance
.
Cancer Res
2010
;
70
:
2840
51
.
43.
Kumazoe
M
,
Takai
M
,
Bae
J
,
Hiroi
S
,
Huang
Y
,
Takamatsu
K
, et al
FOXO3 is essential for CD44 expression in pancreatic cancer cells
.
Oncogene
2017
;
36
:
2643
54
.
44.
Biamonti
G
,
Catillo
M
,
Pignataro
D
,
Montecucco
A
,
Ghigna
C
. 
The alternative splicing side of cancer
.
Semin Cell Dev Biol
2014
;
32
:
30
6
.
45.
Gricius
D
,
Kazlauskas
A
. 
Analysis of oncogene GLI1 protein expression levels between differing grades of astrocytoma
.
BIOLOGIJA
2014
;
60
:
142
7
.
46.
Rimkus
TK
,
Carpenter
RL
,
Qasem
S
,
Chan
M
,
Lo
HW
. 
Targeting the sonic hedgehog signaling pathway: review of smoothened and GLI inhibitors
.
Cancers (Basel)
2016
;
8. doi: 10.3390/cancers8020022
.