Glioblastoma is the most common and aggressive intrinsic brain tumor in adults. Self-renewing, highly tumorigenic glioma-initiating cells (GIC) have been linked to glioma invasive properties, immunomodulation, and increased angiogenesis, leading to resistance to therapy. TGF-β signaling has been associated with the tumorigenic activity of GIC. TGF-β is synthesized as a precursor molecule and proteolytically processed to the mature form by members of the family of the proprotein convertases subtilisin/kexin. In this study we report that furin is unique among the proprotein convertases subtilisin/kexin in being highly expressed in human GIC. Furin cleaves and promotes activation of pro–TGF-β1 and pro–TGF-β2, and TGF-β2 in turn increases furin levels. Notably, TGF-β2 controls furin activity in an ALK-5–dependent manner involving the ERK/MAPK pathway. We thus uncover a role of ERK1 in the regulation of furin activity by supporting a self-sustaining loop for high TGF-β activity in GIC.

Glioblastoma is the most common malignant primary brain tumor and is largely refractory to current therapies (1). TGF-β has long been attributed a major role in the malignant phenotype of glioblastoma (2, 3). Cytostatic responses to TGF-β are selectively inhibited in glioblastoma cells (4) with TGF-β sustaining the migratory and invasive potential as well as suppressing antitumor immune surveillance. Glioma-initiating cells (GIC) have been proposed as the cell population responsible for tumor initiation and recurrence of gliomas (5), and have been attributed immunosuppressive properties (6, 7). TGF-β may maintain the stem cell–like properties and tumorigenic activity of GIC (811). TGF-β binding to the TGF-β receptor complex leads to the activation of both SMAD transcription factors (canonical/SMAD-dependent pathway) (12) and various SMAD-independent pathways including the PI3K/AKT pathway (13), the MEK (MAPK)/ERK, and the p38 and JNK pathways (14). Persistent activation of the TGF-β pathway in glioblastoma has been described as an autocrine loop involving TGF-β2, CREB1, SMAD3, and the PI3K/AKT pathway (2). Processing of pro–TGF-β1 and pro–TGF-β2 in the cytoplasm or in the extracellular matrix in glioma cells involves proprotein convertases subtilisin/kexin (PCSK) (15). PCSK are a class of nine enzymes with a fundamental role in the processing of diverse protein precursors (16). PCSK3, generally known as furin or paired basic amino acid-cleaving enzyme, is widely expressed and responsible for most of the processing events in the constitutive secretory pathway. Furin is located in a variety of cellular compartments including the endoplasmatic reticulum, trans-Golgi network, and cell surface (17). It is also considered the major endoprotease responsible for processing TGF-β at its (R/K)-2nX-R↓ site (where n = 0–3 aa) (18). TGF-β has been reported to induce expression of its converting enzyme furin in fibroblasts (19), epithelial cells (20), and hepatocellular carcinoma cells (21) involving SMAD, MAPK ERK1/p44, and ERK2/p42. This led us to explore the regulation of furin by these pathways in human GIC and specifically the existence and regulation of a potential autocrine TGF-β–furin loop as a potential therapeutic target for anti–TGF-β treatment in glioblastoma.

LN-308 and LN-229 kindly provided by N. de Tribolet (Lausanne, Switzerland) were cultured in DMEM (Life Technologies/Thermo Fisher, Madison, WI) supplemented with 1% l-glutamine (Life Technologies) and 10% FBS (Life Technologies). The human GIC T-325, T-269, ZH-161, S-24, and ZH-305 (22) were cultured in phenol red-free Neurobasal medium (NBM) (Life Technologies), supplemented with 2% B-27 without vitamin A (Life Technologies) (20 μl/ml), 1% l-glutamine (Life Technologies), and fibroblast growth factor and epidermal growth factor (FGF/EGF) (20 ng/ml each) (PeproTech, London, U.K.). Treatment of cells was performed in NBM containing 1% l-glutamine and FGF/EGF and the following reagents: PCSK inhibitor decanoyl-RVKR-CMK (3501; Tocris Bioscience, Bristol, U.K.), recombinant human TGF-β2 (R&D Systems, Minneapolis, MN), SD-208 (Scios, Fremont, CA), and MEK1/2 inhibitor U0126 (9903; Cell Signaling Technology, Danvers, MA).

Lentivirally-mediated gene silencing of furin was performed as described (23), using a furin specific short hairpin RNA (V3LHS_310002) cloned in a pGIPZ lentiviral vector and the respective pGIPZ vector carrying a non-targeting control (RHS4349) (Dharmacon, Lafayette, CO).

Transient gene silencing was performed by electroporation (Neon Transfection System, Invitrogen/Thermo Fisher) using 100 nM of the following ON-TARGET plus, small interfering RNA (siRNA) SMART pools from Dharmacon: TGF-β1 (L-012562-00), SMAD2 (L-003561-00), SMAD3 (L-020067-00), SMAD4 (L-003902-00), ERK1 (L-003592-00), ERK2 (L-003555-00), and non-targeting control (D-001810-10).

Human PBMC were prepared from healthy donor blood and cultured in RPMI 1640 (Life Technologies) supplemented with 10% FBS (Life Technologies), 20 mM l-glutamine, 50 μM β-mercaptoethanol (Sigma, St. Louis, MO), 10 U/ml penicillin (Sigma), 10 μg/ml streptomycin (Sigma), 10 mM sodium-pyruvate (Sigma), and 1 × MEM nonessential amino acids (Life Technologies). Media from GIC seeded at 107 in 10 ml NBM for 72 h were concentrated 2.5-fold prior to addition to the PBMC. On day 0 PBMC were treated with 3 μg/ml concanavalin A (Sigma), 20 U/ml IL-2 (PeproTech), and GIC conditioned media. At day 3, treatment with IL-2 and exposure to GIC media were repeated, and at day 6 addition of IL-2 was repeated. On day 7 cells were analyzed by flow cytometry.

Immunoblot analysis was performed as previously described (23). Primary Abs were anti-furin (sc-20801; Santa Cruz Biotechnology, Dallas, TX), anti–β-actin (sc-1616; Santa Cruz), anti-human LAP/TGF-β1 (AF-246-NA; R&D Systems), anti–TGF-β1 (G1221; Promega, Madison, WI), anti–TGF-β2 (ab36495; Abcam, Cambridge, U.K.) and Abs from Cell Signaling Technology specific for SMAD1 (9743), pSMAD1/5 (Ser463/465) (9516), SMAD2 (3122), pSMAD2 (Ser456/467) (3108), SMAD3 (9513), pSMAD3 (Ser423/425) (9520), SMAD4 (9515), p44/42 MAPK (ERK1/2) (9102), pp44/42 MAPK (ERK1/2) (Thr202/Tyr204) (9106), AKT (9272), pAKT (Ser473) (9271), p38 MAPK (9212), and pp38 MAPK (Thr180/Tyr182) (9211). Secondary Abs were HRP-coupled goat anti-rabbit (sc-2004), donkey anti-goat (sc-2033) (Santa Cruz Biotechnology) or sheep anti-mouse Abs (NA931V; GE Healthcare, Amersham, U.K.).

RT-PCR was performed by using the Δ threshold cycle method with ARF1 as a housekeeping gene (see Supplemental Table I for primers).

Cellular and extracellular furin-specific activity (FSA) was measured as described (24), using the anti-furin Ab MAB15032 (R&D Systems) as capturing Ab in lysates and conditioned media of cells seeded at a density of 5 × 106 cells in 7 ml NBM supplemented with l-glutamine and FGF/EGF for 48 h. FSA is expressed as the fluorescence intensity (λexc = 380 nm, λem = 460 nm) normalized to protein concentration as determined by Bradford assay (Bio-Rad, Hercules, CA). FSA was measured in concentrated conditioned media mixed with the appropriate amount of reaction buffer and analyzed in the same manner as previously described for the lysates (24).

APC-CD25 (17-0259) and PE-FOXP3 (12-4776) Abs were from eBioscience (San Diego, CA), FITC-CD4 (55346) from BD Biosciences (San Jose, CA). A FOXP3 fixation/permeabilization kit (eBioscience) was used for FOXP3 staining. Cells were analyzed with FACSVerse (BD Biosciences) and data analyzed with FlowJo software. Fluorescence minus one controls using fluorochrome-conjugated isotype control Abs from BD Biosciences (FITC-mouse IgG1, κ [555748], APC-mouse IgG1, κ [555751] and PE-rat IgG2A, κ [553930]) were used.

Seven of the nine PCSK, i.e., PCSK1, PCSK2, PCSK4, PCSK5, PCSK6, PCSK7, and furin, process their substrate at the consensus recognition sequence (R/K)Xn(R/K)↓, which is also the recognition site for processing TGF-β. We first investigated mRNA expression levels of these enzymes in a panel of five human GIC: T-325, T-269, ZH-161, S-24, and ZH-305. Furin was expressed in all cell lines and was one of the most abundant PCSK (Fig. 1A). Furin was detectable intracellularly in cell lysates as a double band of around 100 kDa, corresponding to the two glycosylated/sialylated forms of mature furin (25) except for T-269, where only the higher molecular mass isoform was detected. In the conditioned media, furin was detectable as a major band with an apparent molecular mass of around 90 kDa (Fig. 1B).

FIGURE 1.

Furin processes TGF-β in GIC. (A) RT-PCR analysis of PCSK1, PCSK2, PCSK4, PCSK5, PCSK6, PCSK7, and furin. (B) Analysis of furin levels by immunoblot in the lysates and conditioned media. (C) Protein levels of furin and precursor forms of TGF-β1/2 were determined in cell lysates and conditioned media of furin-silenced or control cells or cells treated with 25 μM PCSK inhibitor decanoyl-RVKR-CMK for 48 h. Precursor and mature forms of TGF-β1/2 were determined in the conditioned media of control, furin-depleted, and PCSK inhibitor-treated cells. (D) Cellular and extracellular FSA was measured in lysates and conditioned media of control or furin-depleted cells. (E) Total SMAD1,2,3 and pSMAD1,2,3,5 levels and total and phosphorylated levels of AKT, ERK1/2, and p38 were determined in control or furin-depleted cell lysates by immunoblot. The predicted molecular mass is indicated in parentheses. (F) Human PBMC were treated with 2 ng/ml TGF-β2 or conditioned media from untreated ZH-161 cells or ZH-161 cells treated with 25 μM PCSK inhibitor decanoyl-RVKR-CMK, in the presence of IL-2, for 7 d. Cells were stained for CD4 (FITC), CD25 (APC), and FOXP3 (PE). Scatter plots are gated on CD4+. All experiments were performed at least twice, and data are the mean of triplicates ± SD. p = 0.0185 for PBMC treated with media from PSCK inhibitor-treated versus PSCK inhibitor-untreated ZH-161, unpaired Student t test.

FIGURE 1.

Furin processes TGF-β in GIC. (A) RT-PCR analysis of PCSK1, PCSK2, PCSK4, PCSK5, PCSK6, PCSK7, and furin. (B) Analysis of furin levels by immunoblot in the lysates and conditioned media. (C) Protein levels of furin and precursor forms of TGF-β1/2 were determined in cell lysates and conditioned media of furin-silenced or control cells or cells treated with 25 μM PCSK inhibitor decanoyl-RVKR-CMK for 48 h. Precursor and mature forms of TGF-β1/2 were determined in the conditioned media of control, furin-depleted, and PCSK inhibitor-treated cells. (D) Cellular and extracellular FSA was measured in lysates and conditioned media of control or furin-depleted cells. (E) Total SMAD1,2,3 and pSMAD1,2,3,5 levels and total and phosphorylated levels of AKT, ERK1/2, and p38 were determined in control or furin-depleted cell lysates by immunoblot. The predicted molecular mass is indicated in parentheses. (F) Human PBMC were treated with 2 ng/ml TGF-β2 or conditioned media from untreated ZH-161 cells or ZH-161 cells treated with 25 μM PCSK inhibitor decanoyl-RVKR-CMK, in the presence of IL-2, for 7 d. Cells were stained for CD4 (FITC), CD25 (APC), and FOXP3 (PE). Scatter plots are gated on CD4+. All experiments were performed at least twice, and data are the mean of triplicates ± SD. p = 0.0185 for PBMC treated with media from PSCK inhibitor-treated versus PSCK inhibitor-untreated ZH-161, unpaired Student t test.

Close modal

To investigate the processing of TGF-β1/2 by furin, lentiviral gene silencing of furin was performed in T-325, ZH-161, and ZH-305 cells, further referred to as shfurin cells (Fig. 1C–E). Furin gene silencing did not affect any other PCSK with the exception of PCSK6 in ZH-305 shfurin cells (data not shown). Decreased protein levels of both cellular and secreted furin (Fig. 1C) and a reduction in cellular and extracellular FSA (Fig. 1D) were confirmed in all furin knockdown cell lines. We then analyzed TGF-β1/2 processing in shfurin cells. For TGF-β1 we used an Ab to the N terminus of TGF-β1 detecting both pro–TGF-β1 (55 kDa) and the latency associated peptide of TGF-β1 (37 kDa), and an Ab to the C terminus detecting mature TGF-β1 (12.5 kDa) (see ZH-161/siTGF-β1 as a control, Fig. 1C). For TGF-β2 we used an Ab reacting with the C terminus of TGF-β2 detecting both pro–TGF-β2 (55 kDa) and mature TGF-β2 (12.5 kDa). Furin gene silencing resulted in increased levels of pro–TGF-β1 and correspondingly decreased levels of the two respective processing products latency associated peptide/TGF-β1 and mature TGF-β1 in ZH-161 and ZH-305, with TGF-β1 not being detectable in T-325 (Fig. 1C). In T-325 and ZH-305, showing detectable TGF-β2 protein levels, furin gene silencing increased levels of pro–TGF-β2 and decreased levels of mature TGF-β2. The effects of furin gene silencing on pro–TGF-β1/2 processing were similar to those obtained with the pan-proprotein convertase inhibitor decanoyl-RVKR-CMK (PCSK inhibitor), suggesting that furin is the main PCSK involved in pro–TGF-β1/2 processing here. Notably, in T-325 and ZH-161, pro–TGF-β processing takes place to a remarkable extent extracellularly as pro–TGF-β levels increased in shfurin cells and upon addition of the PCSK inhibitor in the conditioned media, but remained unchanged in the lysates. In ZH-305, the levels of pro–TGF-β2 increased both in cell lysates and in conditioned media following furin gene silencing, suggesting that pro–TGF-β2 processing takes place in part in the intracellular compartment in ZH-305 cells, too (Fig. 1C).

To confirm that reduced TGF-β levels affect downstream signaling activities, we analyzed the levels of pSMAD2, pSMAD3, and pSMAD1/5 as readouts for the activation of canonical TGF-β signal transduction, and of pAKT (Ser473), pERK1/2, and pp38 reflecting non-canonical TGF-β signaling (Fig. 1E). All shfurin cells showed reduced levels of pSMAD2 and pSMAD3. In ZH-161 and ZH-305, pSMAD1/5 was also reduced, pointing toward a reduction in basal bone morphogenetic protein signaling upon furin gene silencing, too. Indeed, inhibiting TGF-β by the TGF-β RI/activin receptor–like kinase (ALK)-5–specific inhibitor SD-208 did not decrease endogenous pSMAD1/5 levels in GIC, although the increase in pSMAD1/5 by exogenous TGF-β was blocked (26). The phosphorylation of AKT (Ser467) and p38 was reduced in T-325 shfurin cells, and ZH-305 shfurin cells showed a reduction in the phosphorylation of ERK and p38 as well.

TGF-β exerts several immunosuppressive functions, including the promotion of FOXP3+ regulatory T (Treg) cells. Previous studies have demonstrated that GIC induce Treg generation in vitro (6). Accordingly, we treated human PBMC with GIC-conditioned media and confirmed the induction of CD4+CD25+FOXP3+ cells: conditioned media of ZH-161 cells increased the fraction of CD4+CD25+FOXP3+ cells from 8.9 to 14.8 ± 0.1%. TGF-β2 treatment was used as a positive control. To test whether the inhibition of TGF-β processing affected the ability of GIC to induce Treg, we exposed PBMC to media derived from GIC treated with the PCSK inhibitor. This led to a decrease in the induction of the CD4+CD25+FOXP3+ population from 8.9 to 11.1 ± 0.5% (Fig. 1F), indicating that indeed the inhibition of TGF-β processing in GIC may reduce their immunosuppressive properties. Similar results were obtained for ZH-305 (data not shown).

Autocrine production of TGF-β may be necessary to maintain GIC stemness and TGF-β signaling in the tumor microenvironment (27); however, the mechanisms of how GIC maintain high TGF-β activity are not well understood (2). We therefore evaluated the effect of TGF-β2 on furin levels. Indeed, TGF-β2 induced the expression of furin and FSA in all cell lines except T-269 (Fig. 2A–C). To investigate whether the expression of PCSK other than furin was affected by TGF-β2, we analyzed the mRNA levels of the other six PCSK in ZH-161 (Fig. 2D), T-325, and ZH-305 cells (data not shown) treated with TGF-β2. Indeed, none of them were changed, pointing toward a specific effect of TGF-β2 on furin expression. Concentration and time dependency of furin induction was investigated in ZH-161 cells (Fig. 2E–G). Because we observed an induction on protein levels after 24 h (Fig. 2F) and the maximum effect was achieved with 2.5 ng/ml TGF-β2 (Fig. 2G), we selected these conditions for future experiments. We proceeded to study the signal transduction mechanisms involved in the regulation of furin by TGF-β2. Exposure of T-325, ZH-161, and ZH-305 to the ALK-5 inhibitor SD-208 had no effect on constitutive furin protein levels, but abolished TGF-β2–mediated furin induction (Fig. 2H). The transient gene silencing of SMAD2, SMAD3, or SMAD4 did not abrogate the induction of furin expression by TGF-β2, indicating involvement of SMAD-independent signal transduction (Fig. 2I).

FIGURE 2.

TGF-β2 induces furin in GIC. (A and B). Furin expression was analyzed on an mRNA level by RT-PCR (A) and on a protein level in cell lysates by immunoblot (B) in GIC treated with 2.5 ng/ml TGF-β2 for 8 (A) or 24 h (B). (C) FSA was measured in the cell lysates and in conditioned media of T-325 and ZH-161 treated with 2.5 ng/ml TGF-β2 for 48 h. (D) PCSK1, PCSK2, PCSK4, PCSK5, PCSK6, PCSK7, and furin mRNA levels in ZH-161 treated with 2.5 ng/ml TGF-β2 for 24 h. (E) Furin mRNA levels were measured by RT-PCR in ZH-161 treated with TGF-β2 for the indicated time points. (F) Furin protein levels were analyzed in cell lysates and conditioned media of ZH-161 treated with 2.5 ng/ml TGF-β2 for the indicated time points by immunoblot. (G) ZH-161 were treated with the indicated concentrations of TGF-β2 for 24 h. Furin protein levels were analyzed in cell lysates by immunoblot. (H) Furin levels were analyzed in the lysates of T-325, ZH-161, and ZH-305 treated with 2.5 ng/ml TGF-β2, 1 μM SD-208 or the combination of TGF-β2 and SD-208 for 24 h. DMSO was used as solvent control. (I) T-325, ZH-161, or ZH-305 were transfected with siRNA targeting SMAD2 (left panel), SMAD3 (central panel), SMAD4 (right panel) or non-targeting control and 24 h later treated with 2.5 ng/ml TGF-β2 for 24 h. Furin was analyzed in the cell lysates by immunoblot. All experiments were performed at least twice, and data are mean of triplicates ± SD.

FIGURE 2.

TGF-β2 induces furin in GIC. (A and B). Furin expression was analyzed on an mRNA level by RT-PCR (A) and on a protein level in cell lysates by immunoblot (B) in GIC treated with 2.5 ng/ml TGF-β2 for 8 (A) or 24 h (B). (C) FSA was measured in the cell lysates and in conditioned media of T-325 and ZH-161 treated with 2.5 ng/ml TGF-β2 for 48 h. (D) PCSK1, PCSK2, PCSK4, PCSK5, PCSK6, PCSK7, and furin mRNA levels in ZH-161 treated with 2.5 ng/ml TGF-β2 for 24 h. (E) Furin mRNA levels were measured by RT-PCR in ZH-161 treated with TGF-β2 for the indicated time points. (F) Furin protein levels were analyzed in cell lysates and conditioned media of ZH-161 treated with 2.5 ng/ml TGF-β2 for the indicated time points by immunoblot. (G) ZH-161 were treated with the indicated concentrations of TGF-β2 for 24 h. Furin protein levels were analyzed in cell lysates by immunoblot. (H) Furin levels were analyzed in the lysates of T-325, ZH-161, and ZH-305 treated with 2.5 ng/ml TGF-β2, 1 μM SD-208 or the combination of TGF-β2 and SD-208 for 24 h. DMSO was used as solvent control. (I) T-325, ZH-161, or ZH-305 were transfected with siRNA targeting SMAD2 (left panel), SMAD3 (central panel), SMAD4 (right panel) or non-targeting control and 24 h later treated with 2.5 ng/ml TGF-β2 for 24 h. Furin was analyzed in the cell lysates by immunoblot. All experiments were performed at least twice, and data are mean of triplicates ± SD.

Close modal

Indeed, blocking the MEK1/2 branch of the non-canonical TGF-β pathway by U0126 decreased constitutive furin levels and attenuated the increase in furin levels in response to TGF-β2. U0126 also decreased constitutive furin levels in T-269 cells that are the TGF-β2/furin–non-responsive model (Fig. 3A). The inhibitory effect of U0126 on the induction of furin expression by TGF-β2 translated into a reduction in FSA (Supplemental Fig. 1A). Similarly, the selective gene silencing of ERK1, but not of ERK2, reduced furin basal levels in all cell lines and attenuated the induction of furin by TGF-β2 (Fig. 3B, 3C). Gene silencing of ERK1, but not of ERK2, reduced furin on mRNA levels in both basal conditions and upon TGF-β2 treatment (Supplemental Fig. 1B). To address whether the control of furin by ERK1 and TGF-β2 is associated with a glioma stem cell–like phenotype, we examined the same GIC cultured in differentiating conditions (Supplemental Fig. 1C) and the long-term glioma cell lines, LN-308 and LN-229 (Supplemental Fig. 1D). In both cases ERK1 gene silencing reduced basal furin levels and attenuated the induction of furin by TGF-β2, suggesting that this molecular pattern is shared between cells with stemness properties and more differentiated tumor cells. The reduction in furin levels following ERK1 gene silencing was associated with a reduction in pro–TGF-β1/2 processing, with increased pro–TGF-β levels and decreased mature TGF-β levels (Supplemental Fig. 1E). MEK1/2 inhibition did not significantly affect the number of spheres, but the spheres formed by GIC (T-325, ZH-161, and ZH-305) treated with U0126 were smaller, indicating an inhibitory effect on cell proliferation as confirmed by reduced MTT metabolism (Fig. 3D). This rather implies a role for the TGF-β–ERK1–furin loop in sustaining the expansion of more differentiated, i.e., progenitor-like glioma cells, rather than in directly promoting GIC sphere formation. Targeting ERK1/2 also attenuated the immunosuppressive properties of GIC because treatment of ZH-161 and ZH-305 cells with U0126 decreased the induction of CD4+CD25+FOXP3+ cells by conditioned media of ZH-161 (Supplemental Fig. 1F) and ZH-305 (data not shown). Thus, our findings reveal that the MEK1/2 branch of the non-canonical signaling pathway, often deregulated in glioma stem cells (28), plays a pivotal role in the control of furin levels in GIC via ERK1. Altogether, we report that TGF-β induces furin activity in GIC and we specifically identify the ERK1 pathway as an essential to maintain furin activity. Our findings indicate that the disruption of ERK1 signaling would be a therapeutic strategy combating TGF-β activity in GIC.

FIGURE 3.

ERK1 controls furin expression in GIC. (A) Furin levels were determined in GIC preincubated with the MEK1/2 inhibitor U0126 (10 μM) for 1 h and then treated with 2.5 ng/ml TGF-β2 for 24 h. DMSO was used as solvent control. (B and C) Cells were transfected with siRNA pools targeting ERK1, ERK2, a combination of ERK1 and ERK2, or non-targeting control. Then 48 h later T-325, ZH-161, S-24, and ZH-305 were treated with TGF-β2 for 24 h. Furin was analyzed in the cell lysates by immunoblot. (D) Sphere formation assay in T-325, ZH-161, and ZH-305 cells treated with 10 μM U0126 or solvent control for 15 d. Data are absorbance values after MTT addition. All experiments were performed at least twice, and data are the mean of triplicates ± SD. *p < 0.05, **p < 0.01, unpaired Student t test.

FIGURE 3.

ERK1 controls furin expression in GIC. (A) Furin levels were determined in GIC preincubated with the MEK1/2 inhibitor U0126 (10 μM) for 1 h and then treated with 2.5 ng/ml TGF-β2 for 24 h. DMSO was used as solvent control. (B and C) Cells were transfected with siRNA pools targeting ERK1, ERK2, a combination of ERK1 and ERK2, or non-targeting control. Then 48 h later T-325, ZH-161, S-24, and ZH-305 were treated with TGF-β2 for 24 h. Furin was analyzed in the cell lysates by immunoblot. (D) Sphere formation assay in T-325, ZH-161, and ZH-305 cells treated with 10 μM U0126 or solvent control for 15 d. Data are absorbance values after MTT addition. All experiments were performed at least twice, and data are the mean of triplicates ± SD. *p < 0.05, **p < 0.01, unpaired Student t test.

Close modal

This work was supported by the Swiss Cancer League/Oncosuisse (Project KFS-3305-08-2013 to I.B. and M.W.) and by the Highly Specialized Medicine-2 program of the Canton of Zurich.

The online version of this article contains supplemental material.

Abbreviations used in this article:

FGF/EGF

fibroblast growth factor and epidermal growth factor

FSA

furin-specific activity

GIC

glioma-initiating cell

NBM

neurobasal medium

PCSK

proprotein convertases subtilisin/kexin

siRNA

small interfering RNA

Treg

regulatory T.

1
Weller
M.
,
van den Bent
M.
,
Hopkins
K.
,
Tonn
J. C.
,
Stupp
R.
,
Falini
A.
,
Cohen-Jonathan-Moyal
E.
,
Frappaz
D.
,
Henriksson
R.
,
Balana
C.
, et al
European Association for Neuro-Oncology (EANO) Task Force on Malignant Glioma
.
2014
.
EANO guideline for the diagnosis and treatment of anaplastic gliomas and glioblastoma.
Lancet Oncol.
15
:
e395
e403
.
2
Rodón
L.
,
Gonzàlez-Juncà
A.
,
Inda
Mdel. M.
,
Sala-Hojman
A.
,
Martínez-Sáez
E.
,
Seoane
J.
.
2014
.
Active CREB1 promotes a malignant TGFβ2 autocrine loop in glioblastoma.
Cancer Discov.
4
:
1230
1241
.
3
Weller
M.
,
Fontana
A.
.
1995
.
The failure of current immunotherapy for malignant glioma. Tumor-derived TGF-beta, T-cell apoptosis, and the immune privilege of the brain.
Brain Res. Brain Res. Rev.
21
:
128
151
.
4
Massagué
J.
2008
.
TGFbeta in cancer.
Cell
134
:
215
230
.
5
Lathia
J. D.
,
Mack
S. C.
,
Mulkearns-Hubert
E. E.
,
Valentim
C. L.
,
Rich
J. N.
.
2015
.
Cancer stem cells in glioblastoma.
Genes Dev.
29
:
1203
1217
.
6
Wei
J.
,
Barr
J.
,
Kong
L. Y.
,
Wang
Y.
,
Wu
A.
,
Sharma
A. K.
,
Gumin
J.
,
Henry
V.
,
Colman
H.
,
Priebe
W.
, et al
.
2010
.
Glioblastoma cancer-initiating cells inhibit T-cell proliferation and effector responses by the signal transducers and activators of transcription 3 pathway.
Mol. Cancer Ther.
9
:
67
78
.
7
Di Tomaso
T.
,
Mazzoleni
S.
,
Wang
E.
,
Sovena
G.
,
Clavenna
D.
,
Franzin
A.
,
Mortini
P.
,
Ferrone
S.
,
Doglioni
C.
,
Marincola
F. M.
, et al
.
2010
.
Immunobiological characterization of cancer stem cells isolated from glioblastoma patients.
Clin. Cancer Res.
16
:
800
813
.
8
Ikushima
H.
,
Todo
T.
,
Ino
Y.
,
Takahashi
M.
,
Miyazawa
K.
,
Miyazono
K.
.
2009
.
Autocrine TGF-beta signaling maintains tumorigenicity of glioma-initiating cells through Sry-related HMG-box factors.
Cell Stem Cell
5
:
504
514
.
9
Gargiulo
G.
,
Cesaroni
M.
,
Serresi
M.
,
de Vries
N.
,
Hulsman
D.
,
Bruggeman
S. W.
,
Lancini
C.
,
van Lohuizen
M.
.
2013
.
In vivo RNAi screen for BMI1 targets identifies TGF-β/BMP-ER stress pathways as key regulators of neural- and malignant glioma-stem cell homeostasis.
Cancer Cell
23
:
660
676
.
10
Anido
J.
,
Sáez-Borderías
A.
,
Gonzàlez-Juncà
A.
,
Rodón
L.
,
Folch
G.
,
Carmona
M. A.
,
Prieto-Sánchez
R. M.
,
Barba
I.
,
Martínez-Sáez
E.
,
Prudkin
L.
, et al
.
2010
.
TGF-β receptor inhibitors target the CD44(high)/Id1(high) glioma-initiating cell population in human glioblastoma.
Cancer Cell
18
:
655
668
.
11
Peñuelas
S.
,
Anido
J.
,
Prieto-Sánchez
R. M.
,
Folch
G.
,
Barba
I.
,
Cuartas
I.
,
García-Dorado
D.
,
Poca
M. A.
,
Sahuquillo
J.
,
Baselga
J.
,
Seoane
J.
.
2009
.
TGF-beta increases glioma-initiating cell self-renewal through the induction of LIF in human glioblastoma.
Cancer Cell
15
:
315
327
.
12
Siegel
P. M.
,
Massagué
J.
.
2003
.
Cytostatic and apoptotic actions of TGF-beta in homeostasis and cancer.
Nat. Rev. Cancer
3
:
807
821
.
13
Zhang
L.
,
Zhou
F.
,
ten Dijke
P.
.
2013
.
Signaling interplay between transforming growth factor-β receptor and PI3K/AKT pathways in cancer.
Trends Biochem. Sci.
38
:
612
620
.
14
Moustakas
A.
,
Heldin
C. H.
.
2005
.
Non-Smad TGF-beta signals.
J. Cell Sci.
118
:
3573
3584
.
15
Leitlein
J.
,
Aulwurm
S.
,
Waltereit
R.
,
Naumann
U.
,
Wagenknecht
B.
,
Garten
W.
,
Weller
M.
,
Platten
M.
.
2001
.
Processing of immunosuppressive pro-TGF-beta 1,2 by human glioblastoma cells involves cytoplasmic and secreted furin-like proteases.
J. Immunol.
166
:
7238
7243
.
16
Seidah
N. G.
,
Sadr
M. S.
,
Chrétien
M.
,
Mbikay
M.
.
2013
.
The multifaceted proprotein convertases: their unique, redundant, complementary, and opposite functions.
J. Biol. Chem.
288
:
21473
21481
.
17
Artenstein
A. W.
,
Opal
S. M.
.
2011
.
Proprotein convertases in health and disease.
N. Engl. J. Med.
365
:
2507
2518
.
18
Dubois
C. M.
,
Blanchette
F.
,
Laprise
M. H.
,
Leduc
R.
,
Grondin
F.
,
Seidah
N. G.
.
2001
.
Evidence that furin is an authentic transforming growth factor-beta1-converting enzyme.
Am. J. Pathol.
158
:
305
316
.
19
Blanchette
F.
,
Day
R.
,
Dong
W.
,
Laprise
M. H.
,
Dubois
C. M.
.
1997
.
TGFbeta1 regulates gene expression of its own converting enzyme furin.
J. Clin. Invest.
99
:
1974
1983
.
20
Blanchette
F.
,
Rudd
P.
,
Grondin
F.
,
Attisano
L.
,
Dubois
C. M.
.
2001
.
Involvement of Smads in TGFbeta1-induced furin (fur) transcription.
J. Cell. Physiol.
188
:
264
273
.
21
Blanchette
F.
,
Rivard
N.
,
Rudd
P.
,
Grondin
F.
,
Attisano
L.
,
Dubois
C. M.
.
2001
.
Cross-talk between the p42/p44 MAP kinase and Smad pathways in transforming growth factor beta 1-induced furin gene transactivation.
J. Biol. Chem.
276
:
33986
33994
.
22
Lemke
D.
,
Weiler
M.
,
Blaes
J.
,
Wiestler
B.
,
Jestaedt
L.
,
Klein
A. C.
,
Löw
S.
,
Eisele
G.
,
Radlwimmer
B.
,
Capper
D.
, et al
.
2014
.
Primary glioblastoma cultures: can profiling of stem cell markers predict radiotherapy sensitivity?
J. Neurochem.
131
:
251
264
.
23
Szabo
E.
,
Schneider
H.
,
Seystahl
K.
,
Rushing
E. J.
,
Herting
F.
,
Weidner
K. M.
,
Weller
M.
.
2016
.
Autocrine VEGFR1 and VEGFR2 signaling promotes survival in human glioblastoma models in vitro and in vivo.
Neuro-oncol.
18
:
1242
1252
.
24
Bourne
G. L.
,
Grainger
D. J.
.
2011
.
Development and characterisation of an assay for furin activity.
J. Immunol. Methods
364
:
101
108
.
25
Denault
J.
,
Bissonnette
L.
,
Longpré
J.
,
Charest
G.
,
Lavigne
P.
,
Leduc
R.
.
2002
.
Ectodomain shedding of furin: kinetics and role of the cysteine-rich region.
FEBS Lett.
527
:
309
314
.
26
Seystahl
K.
,
Tritschler
I.
,
Szabo
E.
,
Tabatabai
G.
,
Weller
M.
.
2015
.
Differential regulation of TGF-β-induced, ALK-5-mediated VEGF release by SMAD2/3 versus SMAD1/5/8 signaling in glioblastoma.
Neuro-oncol.
17
:
254
265
.
27
Pickup
M.
,
Novitskiy
S.
,
Moses
H. L.
.
2013
.
The roles of TGFβ in the tumour microenvironment.
Nat. Rev. Cancer
13
:
788
799
.
28
Sunayama
J.
,
Matsuda
K.
,
Sato
A.
,
Tachibana
K.
,
Suzuki
K.
,
Narita
Y.
,
Shibui
S.
,
Sakurada
K.
,
Kayama
T.
,
Tomiyama
A.
,
Kitanaka
C.
.
2010
.
Crosstalk between the PI3K/mTOR and MEK/ERK pathways involved in the maintenance of self-renewal and tumorigenicity of glioblastoma stem-like cells.
Stem Cells
28
:
1930
1939
.

M.W. has received research grants from Acceleron, Actelion, Alpinia Institute, Bayer, Isarna, Merck Sharp & Dohme, Merck & Co., Novocure, Piqur Therapeutics, and Roche. He has received honoraria for lectures or advisory board participation from Celldex, Immunocellular Therapeutics, Isarna, Magforce, Merck Sharp & Dohme, Merck & Co., Northwest Biotherapeutics, Novocure, Pfizer, Roche, and Teva. The other authors have no financial conflicts of interest.

Supplementary data