Skip to main content
Erschienen in: Journal of Experimental & Clinical Cancer Research 1/2017

Open Access 01.12.2017 | Research

RETRACTED ARTICLE: TGF-β2 initiates autophagy via Smad and non-Smad pathway to promote glioma cells’ invasion

verfasst von: Chao Zhang, Xin Zhang, Ran Xu, Bin Huang, An-Jing Chen, Chao Li, Jian Wang, Xin-Gang Li

Erschienen in: Journal of Experimental & Clinical Cancer Research | Ausgabe 1/2017

Abstract

Background

Glioblastoma multiforme (GBM) is characterized by lethal aggressiveness and patients with GBM are in urgent need for new therapeutic avenues to improve quality of life. Current studies on tumor invasion focused on roles of cytokines in tumor microenvironment and numerous evidence suggests that TGF-β2 is abundant in glioma microenvironment and vital for glioma invasion. Autopagy is also emerging as a critical factor in aggressive behaviors of cancer cells; however, the relationship between TGF-β2 and autophagy in glioma has been poorly understood.

Methods

U251, T98 and U87 GBM cell lines as well as GBM cells from a primary human specimen were used in vitro and in vivo to evaluate the effect of TGF-β2 on autophagy. Western blot, qPCR, immunofluorescence and transmission-electron microscope were used to detect target molecular expression. Lentivirus and siRNA vehicle were introduced to establish cell lines, as well as mitotracker and seahorse experiment to study the metabolic process in glioma. Preclinical therapeutic efficacy was evaluated in orthotopic xenograft mouse models.

Results

Here we demonstrated that TGF-β2 activated autophagy in human glioma cell lines and knockdown of Smad2 or inhibition of c-Jun NH2-terminal kinase, attenuated TGF-β2-induced autophagy. TGF-β2-induced autophagy is important for glioma invasion due to the alteration of epithelial-mesenchymal transition and metabolism conversion, particularly influencing mitochondria trafficking and membrane potential (△Ψm). Autopaghy also initiated a feedback on TGF-β2 in glioma by keeping its autocrine loop and affecting Smad2/3/7 expression. A xenograft model provided additional confirmation on combination of TGF-β inhibitor (Galunisertib) and autophagy inhibitor (CQ) to better “turn off” tumor growth.

Conclusion

Our findings elucidated a potential mechanism of autophagy-associated glioma invasion that TGF-β2 could initiate autophagy via Smad and non-Smad pathway to promote glioma cells’ invasion.
Hinweise

Electronic supplementary material

The online version of this article (https://​doi.​org/​10.​1186/​s13046-017-0628-8) contains supplementary material, which is available to authorized users.
A Retraction Note to this article is available online at https://​doi.​org/​10.​1186/​s13046-021-02219-8.
Abkürzungen
AA
Astrocytoma
Baf
Bafilomycin A1
CQ
Chloroquine
DMSO
Dimethyl sulfoxide
EMT
Epithelial-mesenchymal transition
GBM
Glioblastoma multiforme
JNK
c-Jun NH2-terminal kinase
mTOR
Mammalian target of rapamycin
NTC
Normal tumor cell
Rapa
Rapamycin

Background

Malignant brain tumors are among the most intractable tumors and are known for their devastating proliferation, invasion and chemotherapy resistance [1]. Despite enormous efforts to target various pathways by suppressing oncogenes or angiogenesis, no improvement in survival has occurred, necessitating further study of the mechanisms of glioma oncogenesis [2]. Of all the hallmarks of glioma, the invasive ability poses a great barrier to surgeons and limits treatment options. Survival is measured in months (median survival is 14.6 months) [3]. In aggressive tumors, many kinds of autocrine and paracrine cytokines derived from astrocytes, microglia or glioma were found, which promotes tumor cell invasion, progression and metastasis. And during glioma invasion, the cytokines are reported to promote the trans-differentiation of immobile, polarized epithelial cells to motile, invasive mesenchymal cells, which is known as the epithelial-mesenchymal transition (EMT) [46]. Thus, much research has focused on the effect of cytokines on glioma invasion [7, 8].
Among the cytokines, TGF-β (transforming growth factor-β) is reported to be associated with essential molecules in glioma invasion [911]. And also in high grade tumors, increased TGF-β is related with poor clinical prognosis [8, 12, 13]. TGF-β could be divided into three isoforms in mammals: TGF-β1, TGF-β2, TGF-β3 and shared approximately 80% amino acid sequence identity and are distributed in different tissues [14].
In all the isoforms of TGF-β, TGF-β2 is specifically overexpressed in highly aggressive glioma and is involved in brain tumor development [14, 15]. Elevated TGF-β2 expression levels are usually observed in the later stages of tumor progression and in up to 95% of high grade gliomas [1517]. TGF-β2 initiates an autocrine loop to promote its own expression and enable oncogenic activity. Thus the hyperactive TGF-β pathway and its key role in glioma oncogenesis provide a new target to cure gliomas. For instance, the TGF-β pathway inhibitors AP12009 and Galunisertib have been applied in phase II/III clinical cases, yielding encouraging results [18, 19].
Besides, this cytokine also has a dual role in oncogenesis. Depending on conditions and tumor stage, TGF-β can act as either a tumor suppressor or as a tumor promoter [19, 20].The mechanism of TGF-β transformation from tumor suppressor to tumor promoter is unclear. Currently, researchers are searching for the “switch factors” causing this behavior.
Previous studies have shown that autophagy could be activated by TGF-β and potentiate TGF-β-mediated growth inhibition in human hepatocellular carcinoma cells [21]. In our experiments, we tried to elucidate the effect of TGF-β on glioma from the autophagy aspect. Autophagy is an evolutionarily conserved lysosomal degradation pathway in which the cell re-digests its own proteins and organelles, thus maintaining macromolecular synthesis and ATP production [22]. The intactness of this flux is important for tumor cell invasion which consumes high levels of ATP, involves secretion factors, and reorganizes skeleton proteins [23, 24]. The inhibition of autophagy impairs tumor cell invasion. [25] Autophagy-mediated tumor promotion may work through suppressing the p53 response, maintaining mitochondria function, sustaining metabolic homeostasis and preventing the diversion of tumor progression [26]. However, the exact mechanism underlying how autophagy impairs glioma tumor progression needs to be explored and relationship between autophagy and cytokines also needs large studies. As autophagy and TGF-β are both involved in glioma invasion and participate in similar tumor functions, whether there are innate connections between these two factors in glioma and whether these associations influence glioma oncogenesis remain unknown. Our current study reports that TGF-β2 activates autophagy in glioma, influencing glioma invasion and metabolism reprogramming.

Methods

Cell culture and reagents

The high-grade, human glioma cell lines U251, U87 MG and T98G were obtained from American Type Culture Collection (Manassas, VA, USA) and used for in vitro experiments. Tumor cells were maintained as monolayer cultures in Dulbecco’s Modified Eagle’s Medium (DMEM; GIBCO) supplemented with 5% fetal calf serum (FCS). Primary cell line (P3) was obtained from the Department of Biomedicine, University of Bergen. When indicated, the cells were treated with TGF-β2 (10 ng/ml; PeproTech) and/or the small molecule TGF-β receptor inhibitor LY2157299 (10 μM; Selleck), chloroquine (10 μM; Selleck), bafilomycin A1 (10 μM; Tocris) and rapamycin (20 μM; Selleck). The inhibitor was added 2 h before the addition of TGF-β2.

Constructs, transfection, and lentiviral/retroviral infection

The siATG5, siATG7 shRNA-expressing vector was obtained from GenePharma (Shanghai, China). GFP-LC3 (pBABEpuro, 22,405)-expressing vectors were obtained from Addgene (Cambridge, MA, USA). For the stable downregulation of ATG5, ATG7 scrambled and shRNA pGIPZ vectors were purchased from Open Biosystems (Thermo Scientific) or obtained through the Open Biosystems library. Lentiviral and retroviral supernatants were prepared following the manufacturer’s instructions and provided by GenePharma. Lentiviral infections were carried out accordingly. A validated Stealth RNAi (GenePharma) specific to Smad2 was also transfected into the U251, T98, and U87 cells using Lipofectamine 2000 according to the manufacturer’s protocol.

Migration and invasion assays

The migratory ability of cells was determined by wound healing assays. The rate of wound closure was monitored at different time points under a microscope and quantified using ImageJ software. The invasion potential was determined on collagen coated Transwell assay (Corning, 8um). DMEM containing 0.1% FBS was added to the upper wells, while DMEM containing 10% FBS (a chemoattractant) was added to the lower wells. Transwell insert membranes were fixed and then stained with 0.25% eosin. Representative pictures of the membranes with cells were acquired at 40× magnification, and the total number of cells in 10 individual fields per membrane were counted.

Western blotting

In brief, the cells were harvested, and lysed with protein extraction agent (Beyotime, Beijing, China). 25–50 μg of proteins per sample per lane were loaded for sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE). Primary antibodies were incubated overnight at 4 °C. Rabbit polyclonal primary antibodies were used against LC3B, SQSTM1, MMP2, MMP9, vimentin, N-cadherin, β-catenin, Slug2, Akt, JNK, SMAD2 (1:1000; Cell Signaling), TGF-β2 (1:500; Abcam), phospho-SMAD2, phospho-Akt, and phospho-JNK (1:500; Cell Signaling). Purified mouse anti-β-actin was used (1:1000; Santa Cruz Biotechnology, Inc.). Secondary antibodies were incubated with anti-mouse immunoglobulin G (IgG), anti-rabbit IgG (1:5000; Santa Cruz Biotechnology) for 1 h at RT. The proteins were visualized using Millipore’s enhanced chemiluminescence (ECL) and detection system analyzed (ChemiDoc Touch, BioRad).

Intracranial injection mouse model

U87 cells were prepared for intracranial injection into NOD SCID male mice obtained from Vital River Laboratories. A total of five animals per condition were used, and animals were grouped as follows: PBS group, LY2157299 (75 mg/kg/d) group, CQ (25 mg/kg/d) group and LY2157299 (75 mg/kg/d) combined with CQ (25 mg/kg/d) group, drugs were used by oral application and 10ul U87 cells (1 × 106/μl PBS) were injected in the cerebral cortex using a stereotactic frame. The mice were monitored and killed when they presented with neurological signs or after two months, during which time the tumor volumes and invading distance were monitored by MRI (General Electric, 3.0 T). Brains of three groups (PBS group, LY2157299 group and LY2157299 combined CQ group) were harvested and fixed in 4% paraformaldehyde for 48 h, embedded in paraffin and prepared for IHC. These experiments were approved by the Animal Care and Use Committee of Shandong University and conformed to the Animal Management Rule of the Chinese Ministry of Health (documentation 55, 2001).

Quantitative real-time PCR

Total RNA was isolated using the RNeasy mini kit (Qiagen) according to the manufacturer’s instructions. RNA was analyzed quantitatively using a Nanodrop (Nanodrop Technologies, Rockland, DE, USA). Total RNA (1 μg) was reverse transcribed into cDNA using a cDNA synthesis kit (Toyobo) according to the manufacturer’s instructions. RT-PCR was performed in a Roche LightCycler 2.0 detector with the Toyobo SYBR Green Supermix. The reactions were analyzed using SDS software (Version 2.4). The threshold cycles (CT) were calculated and the relative gene expression was analyzed after normalizing to glyceraldehyde 3-phosphate dehydrogenase (GAPDH). The human primers are listed in Additional file 1: Figure S1.

Immunofluorescence

GBM cells were plated on glass slides in 24-well culture plates at a concentration of 2 × 105 cells/well for 24 h and were subsequently treated with drugs for an additional 48 h in DMEM containing FBS. The cells were then fixed with a 4% formaldehyde solution in PBS, permeabilized with 0.5% Triton X-100 in PBS, stained with primary antibody overnight, and labeled with anti-mouse or anti-rabbit IgG conjugated with FITC and DyLight 594 (Santa Cruz Biotechnology). The cells were counterstained with DAPI and observed under an Olympus BX61 fluorescence microscope. Pictures were scanned using a DP71 CCD (charge-coupled device) digital camera.

Mitochondrial morphology

Live cells were fluorescently labeled with 25 nM MitoTracker Red (Invitrogen, Molecular Probes) and were then fixed and permeabilized for antibody conjugating. All treatments were performed according to the manufacturer’s instructions. Mitochondrial morphology was analyzed using an Olympus BX61 fluorescence microscope.

Measurement of △Ψm

For the △Ψm measurements, the cells were loaded with 50 nM tetramethylrhodamine methylester (JC-1) for 30 min at room temperature. The dye was present during the experiment according to the manufacturer. The mitochondrial membrane potential was analyzed using an Olympus BX61 fluorescence microscope equipped with a 40× objective and quantified by flow cytometry (Novocyte, ACEA).

Enzyme-linked Immunosorbent assay

For the quantitative determination of activated human TGF-β2, MMP2, MMP9 concentrations in cell culture supernatants, the quantitative sandwich enzyme immunoassay technique was used with commercially available, specific immunoassay kits for human TGF-β2 (R&D Systems), MMP2, and MMP9 (eBioscience). The minimum detectable dose of TGF-β2 was less than 7.0 pg/ml. The assay was performed in triplicate according to the manufacturer’s instructions.

ATP—Lactate determination

The total ATP and lactate levels were measured by plating 2 × 105 cells in 6-well plates overnight. The ells were counted and media samples were collected for ATP and lactate assays. The total ATP levels were determined using the Cell Titer-Glo Luminescent assay (Promega) according to the manufacturer’s instructions. Lactate was measured using the L-Lactate assay kit (Abcam). The data were expressed as moles of ATP and mg/dl lactate, and all values were normalized to the number of cells.

Statistical analysis

Results are presented as the mean ± SD from at least three independent experiments. The comparisons were performed using with two-tailed Student’s t test, Spearman correlation test, Pearson correlation test, Wilcoxon matched-pair test, one-way ANOVA and long-rank analyse. Significant differences: *p < 0.05; **p < 0.01;***p < 0.001.

Results

TGF-β2 and LC3B are highly expressed in glioma and correlated with each other indicating poor prognosis in glioma patients

Although large studies has been conducted [27, 28], the role of autophagy’s markers Light Chain 3B (LC3B), ATG5 and ATG7 in glioma prognosis was still controversial. By applying immunochemistry on samples with different pathologic grades obtained from patients in Qilu Hospital, China from 2012 to 2015, we found that the percentage of LC3B, ATG5, ATG7 positive cells was associated with the pathologic grade (Fig. 1a). During this IHC process, we also found abundant TGF-β2 positive cells in GBM samples accompanied with autophagy markers positive. To quantitatively analyze the immunochemistry samples, we used the IRS scoring model (IRS = SI (staining intensity) × PP (percentage of positive cells)) [29], Wilcoxon matched-pair test, and Spearman correlation test to assess the results. Then the autophagy related markers and TGF-β2 were both identified to be associated with the pathologic grades (Fig. 1b) and correlated with each other when searched in REMBRANDT database (Fig. 1c).
To further explore the relationship between autophagy and TGF-β, we concentrated on LC3B and TGF-β.The immunofluorescence images indicated that these two molecules were co-localized in glioma tissues (Fig. 1d). We verified these conclusions in primary tumor samples and qRT-PCR analysis showed similar results in normal brain tissues and glioma patients: Student’s t test, LC3B: GBM vs Normal, p = 0.0401, TGF-β2: GBM vs Normal, P = 0.0490; Pearson correlation test, r = 0.4101, p = 0.0144 (Fig. 1e).
Then based on the data we obtained from qRT-PCR, LC3B and TGF-β2 expression levels were also found to be associated with glioma grades in Chi-square test, whereas patient age and gender had no influence (Fig. 1f). We then added the survival time index and used the long-rank test to analyze. Data showed that high TGF-β2 expression indicated poor prognosis and short survival time in glioma patients (Fig. 1g). And among the samples with high TGF-β2 expression, robustly positive LC3B expression usually indicated a worse survival outcome (Fig. 1h). As the relationship between TGF-β2 and autophagy in glioma has not been explored, these results indicate that TGF-β2 and LC3B are highly expressed in glioma and correlated with each other. High expression levels of TGF-β2 with LC3B indicates poor prognoses in glioma patients.

TGF-β2 induces autophagy in human glioma cell lines in a time and dose-dependent manner

To investigate the exact relationship between TGF-β2 and LC3B in glioma, we treated the tumor cells with a TGF-β2 dose gradient ranging from 0.1 ng/ml to 10 ng/ml, and after 24 h, Western blotting was used to detect autophagy levels. As an increase in the LC3B-II/I ratio is a credible marker of autophagy enhancement [30], TGF-β2 promoted endogenous LC3B-I to LC3B-II conversion in three different cell lines and one primary cell line in a dose-dependent manner (Fig. 2a Additional file 6: Figure S6a). To determine the proper time required for TGF-β stimulation, we set a gradient time course and also tested the conversion ratio by western blot. Our results indicated that 24 h was appropriate to induce glioma autophagy (Fig. 2b and Additional file 6: Figure 6b).
To confirm this phenomenon, we applied a lentiviral system and generated stably expressing GFP-LC3 cells to monitor autophagy (TGF-β2, 10 ng/ml, 24 h) (Fig. 2c). In autophagy studies, the analysis of GFP-LC3 localization is widely performed by counting LC3-GFP positive cells (LC3-GFP dots greater than four) [30]. We found that TGF-β2 treatment increased the percentage of LC3-GFP positive cells (Fig. 2c). As transmission electron microscopy is the gold standard for identifying autophagosome double-membrane structure, we used transmission electron microscopy and found that glioma autophagosomes increased after TGF-β2 treatment (Fig. 2d, Additional file 2: Figure S2a).
Considering the disadvantage of cell lines to represent glioma’s characters, we introduced primary cells in our in vitro experiment, which were obtained from newly surgical GBM samples in Qilu hospital and grades were confirmed by pathology IHC. As shown in western blot (Fig. 2e), TGF-β2 could also initiate autophagy in primary cell under the same condition (10 ng/ml; 24 h) which indicate that the results of cell lines may represent the primary cells to some extent in our experiment.
Effects of 3-MA or Baf on autophagy flux mediated by TGF-β were also determined. Glioma cells were incubated with 3-MA (10 mmol/L) or Baf (10 nmol/L) during the treatment with TGF-β2 (Fig. 2f) and results indicated that Baf could effectively inhibit TGF-β induced autophagy except for 3-MA. According to the results of a microarray that compared the control and TGF-β2 treatment groups, the mRNA levels of the autophagy related genes BECN1, ATG5, ATG7 were also up-regulated after TGF-β2 treatment (Fig. 2g). To summarize, autophagy can be induced by TGF-β2 (10 ng/ml, 24 h) and blocked by Baf (10 nmol/L, 24 h). We next investigated the biological significance of this phenomenon.

TGF-β2 induced autophagy is related to glioma invasion and migration

To explore the biological meaning of TGF-β2-induced autophagy in oncogenesis, we first introduced a stable siRNA expression system targeting the essential autophagy gene ATG5 and ATG7. Efficiency of siRNA was examined by Western blot (Fig. 3a), and the ratio of LC3B-I to LC3B-II conversion and SQSTM1 (also well known as P62) degradation indicated that siATG5, siATG7 was an effective target for inhibiting autophagy flux. As we know from previous studies, TGF-β2 promotes glioma oncogenesis behavior [9, 31, 32]. We used methods involving CCK-8 and BrdU proliferation assays to assess cell proliferation and observed no obvious effect after TGF-β2 treatment in normal tumor cells (NTC) or autophagy-deficient tumor cells (Fig. 3b, Additional file 3: Figure S3).
Then we turned to examine the behavior of autophagy in glioma invasion and analyzed results with wound healing assays in U251, T98, and U87 cell lines. In stable siATG5 and siATG7 tumor cells, autophagy impaired groups exhibited delayed wound closure compared to scramble group tumor cells when exposed to TGF-β2 stimulation (Fig. 3c). And also we carried out a Transwell assay and found that fewer siATG5 and siATG7 cells migrated through the Transwell chamber micropores after TGF-β2 stimulation (Fig. 3d, Additional file 2: Figure S2b). Inefficient ECM degradation usually indicates potential changes in the function or expression level of matrix metalloproteinase (MMPs), which are involved in ECM degradation and the epithelial-mesenchymal transition (EMT). Summarizing the results above, TGF-β2 induced glioma autophagy participated in the invasive process but not tumor proliferation.

TGF-β2 induced EMT in glioma cells could be blocked by suppressing autophagy

To identify the mechanism of autophagy-influenced glioma invasion, we conducted qRT-PCR to determine the expression levels of MMPs in normal glioma cells and Baf-treated tumor cells with or without TGF-β2 stimulation (Fig. 4a). The Baf treatment down-regulated the mRNA expression of the mostly MMP family in cells exposed to TGF-β2, and in contrast control group with TGF-β2 showed increasing expression levels of most MMPs (Fig. 4a, Additional file 4: Figure S4). MMP2 and MMP9, which are the most well-known MMPs for their important roles in tumor cell invasion [33, 34], were also down-regulated by Baf. And this finding was confirmed by Western blot experiments used to detect the cleaved, active forms of MMP2 and MMP9 (Fig. 4b). As the proteolytic cleavage of the ECM is usually governed by cell surface and soluble MMPs, we used an ELISA assay to detect soluble MMP2 and MMP9 in the supernatant of cultured NTC- and Baf-treated cells, similar results were expressed (Fig. 4c). In addition to degrading ECM, MMP2, 9 are also markers of EMT. We used Western blotting to examine other EMT markers to assess the potential association between EMT, TGF-β2 and autophagy. N-cadherin, Vimentin, Slug2, and β-catenin increased in normal tumor cells (NTC) treated with TGF-β2 but decreased in autophagy-inhibited cells however E-cadherin showed reverse effect (Fig. 4d). Immunofluorescence again demonstrated a direct change of Vimentin and N-cadherin between NTC and Baf-treated cells after treatment with TGF-β2 (Fig. 4e, f). Taken together, these results provide a potential explanation for how autophagy impairs glioma invasion: the inhibition of autophagy may lead to an impaired EMT and a failure in the invasive behavior of glioma cells.

Autophagy induction by TGF-β2 is mediated by c-Jun NH2-terminal kinase and the Smad pathway and in turn affects TGF-β2 synthesis and secretion

We next investigated the exact mechanism of the process. Because autophagy is usually activated through mTOR-dependent and mTOR-independent pathways [35, 36], we used Western blotting to determine whether either of these pathways functioned in this process. Inhibiting the AKT-PI3K-mTOR pathway by 3-MA did not attenuate TGF-β2-induced autophagy (Fig. 5a). Thus, we assessed the mTOR-independent pathway using the c-Jun NH2-terminal kinase (JNK) pathway inhibitor SP600125. This treatment efficiently down-regulated the LC3B-II level in TGF-β2 treated glioma cells, indicating JNK pathway was one potential pathway responsible for TGF-β2 induced autophagy. TGF-β signals also went through downstream Smad and non-Smad pathways. The JNK pathway is a non-Smad pathway, and therefore we examined whether the Smad pathway also participated in autophagy induction. After transient siRNA with siSmad2, the addition of TGF-β2 did not stimulate autophagy in glioma, suggesting that the Smad pathway was also involved in autophagy induction (Fig. 5b).
Based on previous study, TGF-β2 function requires sustained autocrine signaling by tumor cells [31]. Thus, we explored whether autophagy would in turn affect TGF-β2 autocrine signaling (Fig. 5c), finding that rapamycin elevated the endogenous TGF-β2 levels in glioma cell and enhanced Smad2 phosphorylation. However, in siATG5-treated tumor cells, the Western blotting results showed reverse effect (Fig. 5c). The ELISA assay for TGF-β2 also demonstrated these phenomenon (Fig. 5d).
We next explored how autophagy influenced TGF-β2. The qRT-PCR results demonstrated that enhancing autophagy by rapamycin increased the R-Smad expression levels (Smad2, Smad3) and decreased the I-Smad expression level (Smad7). In contrast, CQ down-regulated Smad2 and Smad3 and up-regulated Smad7 (Fig. 5e). Microarray also indicated that promoting autophagy could also in turn regulate TGF-β genes’ expression (Fig. 5f). LY21457299 is a clinical applied TGF-β inhibitors, however Western blotting demonstrated that LY2157299 had little effect on the endogenous TGF-β2 levels. After blocking autophagy flux with CQ or siATG5, the TGF-β2 level decreased (Fig. 5g).
Lactate was also reported to promote TGF-β synthesis and MMP2 production in glioma cells [37]. And lactate was associated with autophagy process tightly. We hypothesis that autophagy may mediate TGF-β2 autocrine via lactate secretion. In our experiments, lactate synthesis was increased in TGF-β2-stimulated tumor cells however decreased after siATG5 or CQ-treatment (Fig. 5h). And when we added exogenous lactate in cultured tumor cells, results confirmed that lactate could stimulate TGF-β2 and MMP2 expression and EMT-related protein changes, however CQ, except for LY2157299, could inhibit this process (Fig. 5i). Therefore, we summarized that TGF-β2 could induced autophagy in glioma via both Smad and non-Smad pathway (JNK pathway), and inhibiting autophagy could also in turn mediated endogenous TGF-β2 level in an autocrine fashion which might resulted from the changing of lactate level secreted by glioma cells.

TGF-β2 and autophagy mediated energy mitochondria function of glioma

In addition to the signal pathway activation and invasion related genes variation, there might be other mechanisms leading to the failure of TGF-β2 caused by autophagy inhibition. As tumor cell invasion is a highly energy- and material-consuming process [23, 24] and autophagy is tightly associated with tumor metabolism [26, 38, 39], We thus hypothesized that TGF-β2-induced autophagy is enhanced to generate more ATP and materials for invasion. First we introduced the MitoTracker Red probe to observe the mitochondrial morphology and localization. Based on one newly issued research [40], we identified the mitochondria morphology as fragmented, intermediate and tubular according to the length of the mitochondria (Fig. 6a, Additional file 4: Figure S4a). In our experiment, tubular form was found to be a more favorable form in the cell invasion apex and underwent more trafficking. TGF-β2-treated NTCs favored the tubular mitochondria, whereas autophagy inhibited tumor cells formed more fragmented mitochondria.
Mitochondrial trafficking can also be classified as three forms [41]: perinuclear, polarized and infiltrating (Fig. 6b, Additional file 5: Figure S5a) according to the distance from nuclei to mitochondria. Then TGF-β group showed an increased percentage of the polarized form and Baf group showed more perinuclear form (one-way ANOVA, polarized form, U251 Con 40 ± 2.1% vs TGF 72 ± 6.2% vs Baf 30 ± 5.5% p = 0.019, perinuclei Con 52 ± 3.7% vs TGF 20 ± 4.8% vs Baf 65 ± 6.2% p = 0.026). That indicated Baf stopped TGF-β2 stimulation reprogramming mitochondrial trafficking to fuel tumor cell invasion at a long distance (Fig. 6b).
Apart from above two factors, mitochondrial membrane potential (△Ψm) is also a vital factor for ATP generation. We used a JC-1 probe to detect the changes of △Ψm in mitochondria. JC-1 probe is an ideal probe that forms J-aggregates with red fluorescence in high △Ψm conditions and J-monomers with green fluorescence in low △Ψm conditions [42]. We observed red fluorescence increasing after TGF-β2 treatment in NTCs and green fluorescence enhancing in Baf treatment tumor cells though also treated with TGF-β2 (Fig. 6c, Additional file 5: Figure S5b). This phenomenon was confirmed by flow cytometry (FCM): one-way ANOVA, green percentage, U251 Con 4.55 ± 3.26% vs TGF 1.82 ± 0.67% vs Baf 15.55 ± 3.28% p = 0.038 (Fig. 6d).
As we could observe the changes of mito-fussion and fission in TGF-β2 and Baf treated groups, the exact molecular signal pathway were also explored by Western blot according to related studies [40, 43]. Results indicated that the phosphorylation of Drp1 and the expression of Opa3 and Mitofusion1 was responsible for the effect (Fig. 6e).
Besides the mitochondria changing, tumor cells were also characterized for aerobic glycolysis (Warburg-like metabolism). We thus recurred to XFe24 analysis (Seahorse) and found that TGF-β2 also altered metabolism program towards to aerobic glycolysis which resulted in a higher consumption rate of oxygen (OCAR) and an increased extracellular acidification (ECAR) to sustain ATP providing and then inhibiting autophagy could block this process (Fig. 6f). ATP assay kit and L-lactate testing kit also confirmed the same results (Fig. 6g, h). These results above provided a further explanation for how inhibiting autophagy might impair TGF-β2-stimulated glioma invasion: the mitochondria relocalization, morphological changes, down regulation in membrane potential and aerobic glycolysis reprogramming.

In vivo xenograft models demonstrated that treatment with combined TGF-β2 and autophagy inhibition provides more benefits than galunisertib single application

We used tumor xenograft models to test this finding and explore the clinical significance. After planting U87 cells in severe combined immune deficiency (SCID) mice, we assigned the animals to the following treatment groups: PBS group, LY2157299 group, CQ group and LY2157299 combined with CQ group. LY2157299 combined with CQ was the most favorable treatment for the mice with tumor xenografts according to the survival time and tumor volumes results monitored by 3 Tesla MRI (Fig. 7a, b), while groups with single CQ showed limited cure effect on tumors which was also in accord with other reference [44]. After 50 days, samples were obtained from the SCID mice and stained with HE, images showed that brains of groups with PBS was infiltrated with tumor cells among the normal brain and tumors borders, while combination treatment with LY2157299 and CQ showed sharp and clear borders between normal tissues and tumor (Fig. 7c). This may indicated a more moderate invasiveness in dual-drug used groups. With IHC, we could also observed a decreased expression level of TGF-β in LY2157299 treated groups (P = 0.0476) and combination drugs groups showed the least expression level (P = 0.0056) (Fig. 7d).The in vivo results showed the potential benefits of combined treatment using TGF-β and autophagy pathway inhibitors as CQ could improve the effect of TGF-β inhibitors. This finding provides additional evidence that TGF-β may partially act via autophagy to function in glioma invasion.

Discussion

In the present study, we explored one finding that TGF-β2 was highly expressed and correlated with LC3B in clinical samples. And in vitro, TGF-β2 induced autophagy in glioma cell lines in a time- and dose-dependent fashion. As TGF-β is an abundant component of the glioma tumor microenvironment, our research may provide another explanation as to why high levels of LC3B expression are usually found in high grade gliomas. In addition to hypoxia and starvation, TGF-β2 and other cytokines (IL-2, IL-6, TNF-α, IFN-γ) may potentially promote glioma autophagy. Much research has been focused on cytokines and autophagy [45, 46], however few answered the question that what was the meaning of cytokine-initiated autophagy.
Our study demonstrated that autophagy flux is vital for TGF-β-induced glioma invasion and we attributed partial reasons to the failure of EMT, which should increase with TGF-β2 stimulation. Reasonablely illustration in our study was that autophagy blockage suppressed EMT-related protein expression and also decreased the level of L-lactate which induces TGF-β2 autocrine and promotes EMT marker expression. Moreover, other researchers have also reported that autophagy-related genes such as BECN1 and SQSTM1 are associated with EMT [47, 48]. These findings provide a new opportunity for discussing the EMT under autophagy during tumor invasion. However, the exact mechanism by which autophagy influences the EMT is mysterious, and further exploration needs to be done in the future.
In addition to EMT, autophagy and TGF-β are both broadly involved in tumor metabolism. Tumor invasion is a high-energy-consumption process, so the ATP generation failure induced by autophagy inhibition may also explain the deficiencies in tumor invasion (Fig. 6g, h). Here is our hypothesis that tumor cells evolved to allow TGF-β to activate autophagy as TGF-β boosted metabolism as well as large reactive oxygen species (ROS) production [49] and if there was no factor eliminating ROS promptly, tumor cells would soon apoptosis for mitochondria damage. Besides, there was a long debate about mitochondrial fusion and fission, including which process is more favorable for tumor invasion and metabolism. Interestingly, TGF-β promotes glioma invasion and elevates ATP generation, causing increased fusion forms of mitochondria. Although we have determined some proteins (Drp1, Opa3 and Mitofusion1) of mitochondira conversion which TGF-β targeted on, a lot of work still need to be done to enrich the outline of the process. Autophagy may have a larger role in tumor invasion that has yet not to be determined.
The two main downstream pathways of TGF-β signaling are the Smad-dependent and Smad-independent pathways [31, 50]. In our study, both were found to participate in TGF-β2-induced autophagy in glioma (Fig. 5a, c). TGF-β activates the PI3K-Akt-mTOR pathway during the TGF-β-induced EMT, however the phosphorylation of mTOR usually inhibits autophagy. TGF-β may play both stimulatory and inhibitory roles in autophagy. But at least in U251 cells, the autophagy stimulation by the Smad and JNK pathways may outweigh the antiautophagic effect of mTOR. Whether this change from an inhibitory role to a stimulatory role in autophagy indicates that the TGF-β changes from functioning as a tumor suppressor to tumor promoter also needs more exploration.
We knew that the TGF-β effects on glioma require sustained autocrine pathway activation [31]. Therefore, we explored the effect of autophagy flux in cytokines’ autocrine loop. Our results demonstrated that autophagy inhibitors mediated TGF-β expression in mRNA levels (Fig. 5e). Experiments (Fig. 5h) also proved that the increased amounts of metabolic products resulting from enhanced autophagy (lactate) leads to increased autocrine TGF-β2 levels. According to these results, TGF-β pathway might influence the invasive behaviors of tumor cells via TGF-β-autophagy-lactate-TGF-β loop. LY2157299 was a newly identified TβRI inhibitor with phase III clinical trials. However, in glioma LY2157299 showed little effect on the TGF-β2 autocrine level, which may result in keeping the tumor cells in a favorable microenvironment that tends to promote oncogenesis. Thus, we advised to administer a combined treatment of LY2157299 and CQ to mice, based on the tumor xenograft models results.
In clinical trials, it is still controversial that LC3B is associated with the grade of the glioma. High LC3B expression of LC3B sometimes is associated with an improved outcome for patients with poorer performance. For patients with normal performance, survival was better for patients with lower LC3B staining [51]. We found that TGF-β2 and LC3B dual positive patients had distinctly shorter survival times than TGF-β2 positive and LC3B negative patients. We hypothesize that the autophagy level may be a valid predictor of patient prognosis only in patients whose IHC showed both TGF-β2 and LC3B staining. Many factors can influence autophagy, and possibly only when autophagy was mainly activated by cytokines rather than other factors, autophagy was an effective predictor. Another explanation is that TGF-β2 only induces glioma autophagy when it reaches a certain level in the tumor microenvironment. Therefore, LC3B positive staining usually indicates a higher level of TGF-β2 in glioma, leading to a poor outcome. These hypotheses was verified primarily with statistical results. However, whether other cytokines are coordinated with these results and whether this could be a widely used phenomenon remain interesting, unanswered questions.
Overall, our work provides a novel role for autophagy in TGF-β2-induced glioma invasion that potentially occurs through the regulation of energy metabolism and autocrine TGF-β (mode pattern Additional file 7: Figure S7). The functional alteration of TGF-β from a tumor suppressor to a tumor promoter may be associated with autophagy. Furthermore, the combination of TGF-β and autophagy inhibitors improves disease in vivo, which may in turn guide the application of CQ in clinical cases. More generally, this study resulted in a systematic identification of cytokines and autophagy, which can also be used for similar studies.

Conclusion

Our findings elucidated a potential mechanism of autophagy-associated glioma invasion that TGF-β2 could initiate autophagy via Smad and non-Smad pathway to promote glioma cells’ invasion. During this process, the alteration of epithelial-mesenchymal transition and metabolism conversion were considered to involved in. And based on this study, we believe a new approche to cure glioma could be found and new opinion could be inspired to refresh our view about cancer.

Acknowledgements

The authors thank Yu Hou for editorial assistance and Shuo Xu for assistance in statistical analyses.

Grant support

This work was supported by Natural Science Foundation of China Grant (81,502,164, 81,402,060 and 81,572,487), the Shandong Provincial Natural Science Foundation Grant (BS2015YY004 and BS2014YY033), the Special foundation for Taishan Scholars (No.ts20110814, tshw201502056), the Fundamental Research Funds of Shandong University, the Department of Science & Technology of Shandong Province (2015GGE27101 and 2015ZDXX0801A01), the University of Bergen, the Helse Bergen, Norway and the Norwegian Centre for International Cooperation in Education (SIU)(UTF-2014/10047).

Competing interests

The authors declare that they have no competing interests.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.
Literatur
1.
Zurück zum Zitat Ricard D, Idbaih A, Ducray F, Lahutte M, Hoang-Xuan K, Delattre J-Y. Primary brain tumours in adults. Lancet. 2012;379:1984–96.PubMedCrossRef Ricard D, Idbaih A, Ducray F, Lahutte M, Hoang-Xuan K, Delattre J-Y. Primary brain tumours in adults. Lancet. 2012;379:1984–96.PubMedCrossRef
2.
Zurück zum Zitat Cloughesy TF, Cavenee WK, Mischel PS. Glioblastoma: from molecular pathology to targeted treatment. Annu Rev Pathol-Mech. 2014;9:1–25.CrossRef Cloughesy TF, Cavenee WK, Mischel PS. Glioblastoma: from molecular pathology to targeted treatment. Annu Rev Pathol-Mech. 2014;9:1–25.CrossRef
3.
Zurück zum Zitat Owonikoko TK, Arbiser J, Zelnak A, Shu H-KG, Shim H, Robin AM, Kalkanis SN, Whitsett TG, Salhia B, Tran NL. Current approaches to the treatment of metastatic brain tumours. Nat Rev Clin Oncol. 2014;11:203–22.PubMedPubMedCentralCrossRef Owonikoko TK, Arbiser J, Zelnak A, Shu H-KG, Shim H, Robin AM, Kalkanis SN, Whitsett TG, Salhia B, Tran NL. Current approaches to the treatment of metastatic brain tumours. Nat Rev Clin Oncol. 2014;11:203–22.PubMedPubMedCentralCrossRef
4.
Zurück zum Zitat Friedl P, Locker J, Sahai E, Segall JE. Classifying collective cancer cell invasion. Nat Cell Biol. 2012;14:777–83.PubMedCrossRef Friedl P, Locker J, Sahai E, Segall JE. Classifying collective cancer cell invasion. Nat Cell Biol. 2012;14:777–83.PubMedCrossRef
5.
Zurück zum Zitat Gao D, Vahdat LT, Wong S, Chang JC, Mittal V. Microenvironmental regulation of epithelial–mesenchymal transitions in cancer. Cancer Res. 2012;72:4883–9.PubMedPubMedCentralCrossRef Gao D, Vahdat LT, Wong S, Chang JC, Mittal V. Microenvironmental regulation of epithelial–mesenchymal transitions in cancer. Cancer Res. 2012;72:4883–9.PubMedPubMedCentralCrossRef
6.
Zurück zum Zitat Scheel C, Weinberg RA. Cancer stem cells and epithelial–mesenchymal transition: concepts and molecular links. Semin Cancer Biol. 2012;22:396–403.PubMedPubMedCentralCrossRef Scheel C, Weinberg RA. Cancer stem cells and epithelial–mesenchymal transition: concepts and molecular links. Semin Cancer Biol. 2012;22:396–403.PubMedPubMedCentralCrossRef
7.
Zurück zum Zitat Bhat KP, Balasubramaniyan V, Vaillant B, Ezhilarasan R, Hummelink K, Hollingsworth F, Wani K, Heathcock L, James JD, Goodman LD. Mesenchymal differentiation mediated by NF-κB promotes radiation resistance in glioblastoma. Cancer Cell. 2013;24:331–46.PubMedCrossRef Bhat KP, Balasubramaniyan V, Vaillant B, Ezhilarasan R, Hummelink K, Hollingsworth F, Wani K, Heathcock L, James JD, Goodman LD. Mesenchymal differentiation mediated by NF-κB promotes radiation resistance in glioblastoma. Cancer Cell. 2013;24:331–46.PubMedCrossRef
8.
Zurück zum Zitat Joseph JV, Balasubramaniyan V, Walenkamp A, Kruyt FA. TGF-β as a therapeutic target in high grade gliomas–promises and challenges. Biochem Pharmacol. 2013;85:478–85.PubMedCrossRef Joseph JV, Balasubramaniyan V, Walenkamp A, Kruyt FA. TGF-β as a therapeutic target in high grade gliomas–promises and challenges. Biochem Pharmacol. 2013;85:478–85.PubMedCrossRef
9.
Zurück zum Zitat Joseph J, Conroy S, Tomar T, Eggens-Meijer E, Bhat K, Copray S, Walenkamp A, Boddeke E, Balasubramanyian V, Wagemakers M. TGF-β is an inducer of ZEB1-dependent mesenchymal transdifferentiation in glioblastoma that is associated with tumor invasion. Cell Death Dis. 2014;5:e1443.PubMedPubMedCentralCrossRef Joseph J, Conroy S, Tomar T, Eggens-Meijer E, Bhat K, Copray S, Walenkamp A, Boddeke E, Balasubramanyian V, Wagemakers M. TGF-β is an inducer of ZEB1-dependent mesenchymal transdifferentiation in glioblastoma that is associated with tumor invasion. Cell Death Dis. 2014;5:e1443.PubMedPubMedCentralCrossRef
10.
Zurück zum Zitat Kahlert U, Nikkhah G, Maciaczyk J. Epithelial-to-mesenchymal (−like) transition as a relevant molecular event in malignant gliomas. Cancer Lett. 2013;331:131–8.PubMedCrossRef Kahlert U, Nikkhah G, Maciaczyk J. Epithelial-to-mesenchymal (−like) transition as a relevant molecular event in malignant gliomas. Cancer Lett. 2013;331:131–8.PubMedCrossRef
11.
Zurück zum Zitat Pala A, Karpel-Massler G, Kast RE, Wirtz CR, Halatsch M-E. Epidermal to mesenchymal transition and failure of EGFR-targeted therapy in glioblastoma. Cancers. 2012;4:523–30.PubMedPubMedCentralCrossRef Pala A, Karpel-Massler G, Kast RE, Wirtz CR, Halatsch M-E. Epidermal to mesenchymal transition and failure of EGFR-targeted therapy in glioblastoma. Cancers. 2012;4:523–30.PubMedPubMedCentralCrossRef
12.
Zurück zum Zitat de Castro JV, Gonçalves CS, Costa S, Linhares P, Vaz R, Nabiço R, Amorim J, Viana-Pereira M, Reis RM, Costa BM. Impact of TGF-β1-509C/T and 869T/C polymorphisms on glioma risk and patient prognosis. Tumor Biol. 2015;36:6525–32.CrossRef de Castro JV, Gonçalves CS, Costa S, Linhares P, Vaz R, Nabiço R, Amorim J, Viana-Pereira M, Reis RM, Costa BM. Impact of TGF-β1-509C/T and 869T/C polymorphisms on glioma risk and patient prognosis. Tumor Biol. 2015;36:6525–32.CrossRef
13.
Zurück zum Zitat Kim Y. Regulation of cell proliferation and migration in glioblastoma: new therapeutic approach. Front Oncol. 2013;3:53.PubMedPubMedCentral Kim Y. Regulation of cell proliferation and migration in glioblastoma: new therapeutic approach. Front Oncol. 2013;3:53.PubMedPubMedCentral
14.
Zurück zum Zitat Bruna A, Darken RS, Rojo F, Ocaña A, Peñuelas S, Arias A, Paris R, Tortosa A, Mora J, Baselga J. High TGFβ-Smad activity confers poor prognosis in glioma patients and promotes cell proliferation depending on the methylation of the PDGF-B gene. Cancer Cell. 2007;11:147–60.PubMedCrossRef Bruna A, Darken RS, Rojo F, Ocaña A, Peñuelas S, Arias A, Paris R, Tortosa A, Mora J, Baselga J. High TGFβ-Smad activity confers poor prognosis in glioma patients and promotes cell proliferation depending on the methylation of the PDGF-B gene. Cancer Cell. 2007;11:147–60.PubMedCrossRef
15.
Zurück zum Zitat Jachimczak P, Hessdörfer B, Fabel-Schulte K, Wismeth C, Brysch W, Schlingensiepen KH, Bauer A, Blesch A, Bogdahn U. Transforming growth factor-β-mediated autocrine growth regulation of gliomas as detected with phosphorothioate antisense oligonucleotides. Int J Cancer. 1996;65:332–7.PubMedCrossRef Jachimczak P, Hessdörfer B, Fabel-Schulte K, Wismeth C, Brysch W, Schlingensiepen KH, Bauer A, Blesch A, Bogdahn U. Transforming growth factor-β-mediated autocrine growth regulation of gliomas as detected with phosphorothioate antisense oligonucleotides. Int J Cancer. 1996;65:332–7.PubMedCrossRef
16.
Zurück zum Zitat Kjellman C, Olofsson SP, Hansson O, Von Schantz T, Lindvall M, Nilsson I, Salford LG, Sjögren HO, Widegren B. Expression of TGF-β isoforms, TGF-β receptors, and SMAD molecules at different stages of human glioma. Int J Cancer. 2000;89:251–8.PubMedCrossRef Kjellman C, Olofsson SP, Hansson O, Von Schantz T, Lindvall M, Nilsson I, Salford LG, Sjögren HO, Widegren B. Expression of TGF-β isoforms, TGF-β receptors, and SMAD molecules at different stages of human glioma. Int J Cancer. 2000;89:251–8.PubMedCrossRef
17.
Zurück zum Zitat Maxwell M, Galanopoulos T, Neville-Golden J, Antoniades HN. Effect of the expression of transforming growth factor-β2 in primary human glioblastomas on immunosuppression and loss of immune surveillance. J Neurosurg. 1992;76:799–804.PubMedCrossRef Maxwell M, Galanopoulos T, Neville-Golden J, Antoniades HN. Effect of the expression of transforming growth factor-β2 in primary human glioblastomas on immunosuppression and loss of immune surveillance. J Neurosurg. 1992;76:799–804.PubMedCrossRef
18.
Zurück zum Zitat Bogdahn U, Hau P, Stockhammer G, Venkataramana N, Mahapatra A, Suri A, Balasubramaniam A, Nair S, Oliushine V, Parfenov V. Targeted therapy for high-grade glioma with the TGF-β2 inhibitor trabedersen: results of a randomized and controlled phase IIb study. Neuro-Oncology. 2011;13:132–42.PubMedCrossRef Bogdahn U, Hau P, Stockhammer G, Venkataramana N, Mahapatra A, Suri A, Balasubramaniam A, Nair S, Oliushine V, Parfenov V. Targeted therapy for high-grade glioma with the TGF-β2 inhibitor trabedersen: results of a randomized and controlled phase IIb study. Neuro-Oncology. 2011;13:132–42.PubMedCrossRef
19.
Zurück zum Zitat Hau P, Jachimczak P, Schlaier J, Bogdahn U. TGF-β2 signaling in high-grade gliomas. Curr Pharm Biotechnol. 2011;12:2150–7.PubMedCrossRef Hau P, Jachimczak P, Schlaier J, Bogdahn U. TGF-β2 signaling in high-grade gliomas. Curr Pharm Biotechnol. 2011;12:2150–7.PubMedCrossRef
20.
Zurück zum Zitat Platten M, Wick W, Weller M. Malignant glioma biology: role for TGF-β in growth, motility, angiogenesis, and immune escape. Microsc Res Techniq. 2001;52:401–10.CrossRef Platten M, Wick W, Weller M. Malignant glioma biology: role for TGF-β in growth, motility, angiogenesis, and immune escape. Microsc Res Techniq. 2001;52:401–10.CrossRef
21.
Zurück zum Zitat Kiyono K, Suzuki HI, Matsuyama H, Morishita Y, Komuro A, Kano MR, Sugimoto K, Miyazono K. Autophagy is activated by TGF-beta and potentiates TGF-beta-mediated growth inhibition in human hepatocellular carcinoma cells. Cancer Res. 2009;69:8844–52.PubMedCrossRef Kiyono K, Suzuki HI, Matsuyama H, Morishita Y, Komuro A, Kano MR, Sugimoto K, Miyazono K. Autophagy is activated by TGF-beta and potentiates TGF-beta-mediated growth inhibition in human hepatocellular carcinoma cells. Cancer Res. 2009;69:8844–52.PubMedCrossRef
23.
Zurück zum Zitat Bettum IJ, Gorad SS, Barkovskaya A, Pettersen S, Moestue SA, Vasiliauskaite K, Tenstad E, Øyjord T, Risa Ø, Nygaard V. Metabolic reprogramming supports the invasive phenotype in malignant melanoma. Cancer Lett. 2015;366:71–83.PubMedCrossRef Bettum IJ, Gorad SS, Barkovskaya A, Pettersen S, Moestue SA, Vasiliauskaite K, Tenstad E, Øyjord T, Risa Ø, Nygaard V. Metabolic reprogramming supports the invasive phenotype in malignant melanoma. Cancer Lett. 2015;366:71–83.PubMedCrossRef
24.
Zurück zum Zitat O. Keunen, M. Johansson, A. Oudin, M. Sanzey, S.A.A. Rahim, F. Fack, F. Thorsen, T. Taxt, M. Bartos, R. Jirik, Anti-VEGF Treatment reduces blood supply and increases tumor cell invasion in glioblastoma, Proc Natl Acad Sci U S A, 108 (2011) 3749–3754. O. Keunen, M. Johansson, A. Oudin, M. Sanzey, S.A.A. Rahim, F. Fack, F. Thorsen, T. Taxt, M. Bartos, R. Jirik, Anti-VEGF Treatment reduces blood supply and increases tumor cell invasion in glioblastoma, Proc Natl Acad Sci U S A, 108 (2011) 3749–3754.
25.
Zurück zum Zitat Macintosh RL, Timpson P, Thorburn J, Anderson KI, Thorburn A, Ryan KM. Inhibition of autophagy impairs tumor cell invasion in an organotypic model. Cell Cycle. 2012;11:2022–9.PubMedPubMedCentralCrossRef Macintosh RL, Timpson P, Thorburn J, Anderson KI, Thorburn A, Ryan KM. Inhibition of autophagy impairs tumor cell invasion in an organotypic model. Cell Cycle. 2012;11:2022–9.PubMedPubMedCentralCrossRef
27.
Zurück zum Zitat Aoki H, Kondo Y, Aldape K, Yamamoto A, Iwado E, Yokoyama T, Hollingsworth EF, Kobayashi R, Hess K, Shinojima N, Shingu T, Tamada Y, Zhang L, Conrad C, Bogler O, Mills G, Sawaya R, Kondo S. Monitoring autophagy in glioblastoma with antibody against isoform B of human microtubule-associated protein 1 light chain 3. Autophagy. 2008;4:467–75.PubMedCrossRef Aoki H, Kondo Y, Aldape K, Yamamoto A, Iwado E, Yokoyama T, Hollingsworth EF, Kobayashi R, Hess K, Shinojima N, Shingu T, Tamada Y, Zhang L, Conrad C, Bogler O, Mills G, Sawaya R, Kondo S. Monitoring autophagy in glioblastoma with antibody against isoform B of human microtubule-associated protein 1 light chain 3. Autophagy. 2008;4:467–75.PubMedCrossRef
28.
Zurück zum Zitat Winardi D, Tsai HP, Chai CY, Chung CL, Loh JK, Chen YH, Hsieh CL. Correlation of altered expression of the autophagy marker LC3B with poor prognosis in astrocytoma. Biomed Res Int. 2014;2014:723176.PubMedPubMedCentral Winardi D, Tsai HP, Chai CY, Chung CL, Loh JK, Chen YH, Hsieh CL. Correlation of altered expression of the autophagy marker LC3B with poor prognosis in astrocytoma. Biomed Res Int. 2014;2014:723176.PubMedPubMedCentral
29.
Zurück zum Zitat Weber DC, Tille J-C, Combescure C, Egger J-F, Laouiti M, Hammad K, Granger P, Rubbia-Brandt L, Miralbell R. The prognostic value of expression of HIF1alpha, EGFR and VEGF-A, in localized prostate cancer for intermediate-and high-risk patients treated with radiation therapy with or without androgen deprivation therapy. Radiat Oncol. 2012;7:1748–17.CrossRef Weber DC, Tille J-C, Combescure C, Egger J-F, Laouiti M, Hammad K, Granger P, Rubbia-Brandt L, Miralbell R. The prognostic value of expression of HIF1alpha, EGFR and VEGF-A, in localized prostate cancer for intermediate-and high-risk patients treated with radiation therapy with or without androgen deprivation therapy. Radiat Oncol. 2012;7:1748–17.CrossRef
30.
Zurück zum Zitat Klionsky DJ, Abdalla FC, Abeliovich H, Abraham RT, Acevedo-Arozena A, Adeli K, Agholme L, Agnello M, Agostinis P, Aguirre-Ghiso JA. Guidelines for the use and interpretation of assays for monitoring autophagy. Autophagy. 2012;8:445–544.PubMedPubMedCentralCrossRef Klionsky DJ, Abdalla FC, Abeliovich H, Abraham RT, Acevedo-Arozena A, Adeli K, Agholme L, Agnello M, Agostinis P, Aguirre-Ghiso JA. Guidelines for the use and interpretation of assays for monitoring autophagy. Autophagy. 2012;8:445–544.PubMedPubMedCentralCrossRef
31.
Zurück zum Zitat Ikushima H, Todo T, Ino Y, Takahashi M, Miyazawa K, Miyazono K. Autocrine TGF-β signaling maintains tumorigenicity of glioma-initiating cells through Sry-related HMG-box factors. Cell Stem Cell. 2009;5:504–14.PubMedCrossRef Ikushima H, Todo T, Ino Y, Takahashi M, Miyazawa K, Miyazono K. Autocrine TGF-β signaling maintains tumorigenicity of glioma-initiating cells through Sry-related HMG-box factors. Cell Stem Cell. 2009;5:504–14.PubMedCrossRef
32.
Zurück zum Zitat Wick W, Platten M, Weller M. Glioma cell invasion: regulation of metalloproteinase activity by TGF-β. J Neuro-Oncol. 2001;53:177–85.CrossRef Wick W, Platten M, Weller M. Glioma cell invasion: regulation of metalloproteinase activity by TGF-β. J Neuro-Oncol. 2001;53:177–85.CrossRef
33.
Zurück zum Zitat Forsyth P, Wong H, Laing T, Rewcastle N, Morris D, Muzik H, Leco K, Johnston R, Brasher P, Sutherland G. Gelatinase-A (MMP-2), gelatinase-B (MMP-9) and membrane type matrix metalloproteinase-1 (MT1-MMP) are involved in different aspects of the pathophysiology of malignant gliomas. Brit J Cancer. 1999, 1828;79 Forsyth P, Wong H, Laing T, Rewcastle N, Morris D, Muzik H, Leco K, Johnston R, Brasher P, Sutherland G. Gelatinase-A (MMP-2), gelatinase-B (MMP-9) and membrane type matrix metalloproteinase-1 (MT1-MMP) are involved in different aspects of the pathophysiology of malignant gliomas. Brit J Cancer. 1999, 1828;79
34.
Zurück zum Zitat Sawaya RE, Yamamoto M, Gokaslan ZL, Wang SW, Mohanam S, Fuller GN, McCutcheon IE, Stetler-Stevenson WG, Nicolson GL, Rao JS. Expression and localization of 72 kDa type IV collagenase (MMP-2) in human malignant gliomas in vivo. Clin Exp Metastatis. 1996;14:35–42.CrossRef Sawaya RE, Yamamoto M, Gokaslan ZL, Wang SW, Mohanam S, Fuller GN, McCutcheon IE, Stetler-Stevenson WG, Nicolson GL, Rao JS. Expression and localization of 72 kDa type IV collagenase (MMP-2) in human malignant gliomas in vivo. Clin Exp Metastatis. 1996;14:35–42.CrossRef
35.
Zurück zum Zitat Wang RC, Wei Y, An Z, Zou Z, Xiao G, Bhagat G, White M, Reichelt J, Levine B. Akt-mediated regulation of autophagy and tumorigenesis through Beclin 1 phosphorylation. Science. 2012;338:956–9.PubMedPubMedCentralCrossRef Wang RC, Wei Y, An Z, Zou Z, Xiao G, Bhagat G, White M, Reichelt J, Levine B. Akt-mediated regulation of autophagy and tumorigenesis through Beclin 1 phosphorylation. Science. 2012;338:956–9.PubMedPubMedCentralCrossRef
36.
Zurück zum Zitat Williams A, Sarkar S, Cuddon P, Ttofi EK, Saiki S, Siddiqi FH, Jahreiss L, Fleming A, Pask D, Goldsmith P. Novel targets for Huntington's disease in an mTOR-independent autophagy pathway. Nat Chem Biol. 2008;4:295–305.PubMedPubMedCentralCrossRef Williams A, Sarkar S, Cuddon P, Ttofi EK, Saiki S, Siddiqi FH, Jahreiss L, Fleming A, Pask D, Goldsmith P. Novel targets for Huntington's disease in an mTOR-independent autophagy pathway. Nat Chem Biol. 2008;4:295–305.PubMedPubMedCentralCrossRef
37.
Zurück zum Zitat Baumann F, Leukel P, Doerfelt A, Beier CP, Dettmer K, Oefner PJ, Kastenberger M, Kreutz M, Nickl-Jockschat T, Bogdahn U, Bosserhoff AK, Hau P. Lactate promotes glioma migration by TGF-beta2-dependent regulation of matrix metalloproteinase-2. Neuro-Oncology. 2009;11:368–80.PubMedPubMedCentralCrossRef Baumann F, Leukel P, Doerfelt A, Beier CP, Dettmer K, Oefner PJ, Kastenberger M, Kreutz M, Nickl-Jockschat T, Bogdahn U, Bosserhoff AK, Hau P. Lactate promotes glioma migration by TGF-beta2-dependent regulation of matrix metalloproteinase-2. Neuro-Oncology. 2009;11:368–80.PubMedPubMedCentralCrossRef
38.
39.
Zurück zum Zitat van der Schaft DW, Hillen F, Pauwels P, Kirschmann DA, Castermans K, Egbrink MG, Tran MG, Sciot R, Hauben E, Hogendoorn PC, Delattre O, Maxwell PH, Hendrix MJ, Griffioen AW. Tumor cell plasticity in Ewing sarcoma, an alternative circulatory system stimulated by hypoxia. Cancer Res. 2005;65:11520–8.PubMedCrossRef van der Schaft DW, Hillen F, Pauwels P, Kirschmann DA, Castermans K, Egbrink MG, Tran MG, Sciot R, Hauben E, Hogendoorn PC, Delattre O, Maxwell PH, Hendrix MJ, Griffioen AW. Tumor cell plasticity in Ewing sarcoma, an alternative circulatory system stimulated by hypoxia. Cancer Res. 2005;65:11520–8.PubMedCrossRef
40.
Zurück zum Zitat Xie Q, Wu Q, Horbinski CM, Flavahan WA, Yang K, Zhou W, Dombrowski SM, Huang Z, Fang X, Shi Y, Ferguson AN, Kashatus DF, Bao S, Rich JN. Mitochondrial control by DRP1 in brain tumor initiating cells. Nat Neurosci. 2015;18:501–10.PubMedPubMedCentralCrossRef Xie Q, Wu Q, Horbinski CM, Flavahan WA, Yang K, Zhou W, Dombrowski SM, Huang Z, Fang X, Shi Y, Ferguson AN, Kashatus DF, Bao S, Rich JN. Mitochondrial control by DRP1 in brain tumor initiating cells. Nat Neurosci. 2015;18:501–10.PubMedPubMedCentralCrossRef
41.
Zurück zum Zitat Caino MC, Ghosh JC, Chae YC, Vaira V, Rivadeneira DB, Faversani A, Rampini P, Kossenkov AV, Aird KM, Zhang R. PI3K therapy reprograms mitochondrial trafficking to fuel tumor cell invasion. Proc Natl Acad Sci U S A. 2015;112:8638–43.PubMedPubMedCentralCrossRef Caino MC, Ghosh JC, Chae YC, Vaira V, Rivadeneira DB, Faversani A, Rampini P, Kossenkov AV, Aird KM, Zhang R. PI3K therapy reprograms mitochondrial trafficking to fuel tumor cell invasion. Proc Natl Acad Sci U S A. 2015;112:8638–43.PubMedPubMedCentralCrossRef
42.
Zurück zum Zitat Gao Y, Su Y, Qu L, Xu S, Meng L, Cai S-Q, Shou C. Mitochondrial apoptosis contributes to the anti-cancer effect of Smilax Glabra Roxb. Toxicol Lett. 2011;207:112–20.PubMedCrossRef Gao Y, Su Y, Qu L, Xu S, Meng L, Cai S-Q, Shou C. Mitochondrial apoptosis contributes to the anti-cancer effect of Smilax Glabra Roxb. Toxicol Lett. 2011;207:112–20.PubMedCrossRef
43.
Zurück zum Zitat Ryu SW, Yoon J, Yim N, Choi K, Choi C. Downregulation of OPA3 is responsible for transforming growth factor-beta-induced mitochondrial elongation and F-actin rearrangement in retinal pigment epithelial ARPE-19 cells. PLoS One. 2013;8:e63495.PubMedPubMedCentralCrossRef Ryu SW, Yoon J, Yim N, Choi K, Choi C. Downregulation of OPA3 is responsible for transforming growth factor-beta-induced mitochondrial elongation and F-actin rearrangement in retinal pigment epithelial ARPE-19 cells. PLoS One. 2013;8:e63495.PubMedPubMedCentralCrossRef
44.
Zurück zum Zitat Rosenfeld MR, Ye X, Supko JG, Desideri S, Grossman SA, Brem S, Mikkelson T, Wang D, Chang YC, Hu J, McAfee Q, Fisher J, Troxel AB, Piao S, Heitjan DF, Tan KS, Pontiggia L, O'Dwyer PJ, Davis LE, Amaravadi RK. A phase I/II trial of hydroxychloroquine in conjunction with radiation therapy and concurrent and adjuvant temozolomide in patients with newly diagnosed glioblastoma multiforme. Autophagy. 2014;10:1359–68.PubMedPubMedCentralCrossRef Rosenfeld MR, Ye X, Supko JG, Desideri S, Grossman SA, Brem S, Mikkelson T, Wang D, Chang YC, Hu J, McAfee Q, Fisher J, Troxel AB, Piao S, Heitjan DF, Tan KS, Pontiggia L, O'Dwyer PJ, Davis LE, Amaravadi RK. A phase I/II trial of hydroxychloroquine in conjunction with radiation therapy and concurrent and adjuvant temozolomide in patients with newly diagnosed glioblastoma multiforme. Autophagy. 2014;10:1359–68.PubMedPubMedCentralCrossRef
45.
Zurück zum Zitat Bhutia SK, Dash R, Das SK, Azab B, Su Z-z, Lee S-G, Grant S, Yacoub A, Dent P, Curiel DT. Mechanism of autophagy to apoptosis switch triggered in prostate cancer cells by antitumor cytokine melanoma differentiation-associated gene 7/interleukin-24. Cancer Res. 2010;70:3667–76.PubMedPubMedCentralCrossRef Bhutia SK, Dash R, Das SK, Azab B, Su Z-z, Lee S-G, Grant S, Yacoub A, Dent P, Curiel DT. Mechanism of autophagy to apoptosis switch triggered in prostate cancer cells by antitumor cytokine melanoma differentiation-associated gene 7/interleukin-24. Cancer Res. 2010;70:3667–76.PubMedPubMedCentralCrossRef
46.
Zurück zum Zitat Harris J, De Haro SA, Master SS, Keane J, Roberts EA, Delgado M, Deretic V. T helper 2 cytokines inhibit autophagic control of intracellular mycobacterium tuberculosis. Immunity. 2007;27:505–17.PubMedCrossRef Harris J, De Haro SA, Master SS, Keane J, Roberts EA, Delgado M, Deretic V. T helper 2 cytokines inhibit autophagic control of intracellular mycobacterium tuberculosis. Immunity. 2007;27:505–17.PubMedCrossRef
47.
Zurück zum Zitat Bertrand M, Petit V, Jain A, Amsellem R, Johansen T, Larue L, Codogno P, Beau I. SQSTM1/p62 regulates the expression of junctional proteins through epithelial-mesenchymal transition factors. Cell Cycle. 2015;14:364–74.PubMedPubMedCentralCrossRef Bertrand M, Petit V, Jain A, Amsellem R, Johansen T, Larue L, Codogno P, Beau I. SQSTM1/p62 regulates the expression of junctional proteins through epithelial-mesenchymal transition factors. Cell Cycle. 2015;14:364–74.PubMedPubMedCentralCrossRef
48.
Zurück zum Zitat Cicchini M, Chakrabarti R, Kongara S, Price S, Nahar R, Lozy F, Zhong H, Vazquez A, Kang Y, Karantza V. Autophagy regulator BECN1 suppresses mammary tumorigenesis driven by WNT1 activation and following parity. Autophagy. 2014;10:2036–52.PubMedPubMedCentralCrossRef Cicchini M, Chakrabarti R, Kongara S, Price S, Nahar R, Lozy F, Zhong H, Vazquez A, Kang Y, Karantza V. Autophagy regulator BECN1 suppresses mammary tumorigenesis driven by WNT1 activation and following parity. Autophagy. 2014;10:2036–52.PubMedPubMedCentralCrossRef
50.
Zurück zum Zitat Siegel PM, Massagué J. Cytostatic and apoptotic actions of TGF-β in homeostasis and cancer. Nat Rev Cancer. 2003;3:807–20.PubMedCrossRef Siegel PM, Massagué J. Cytostatic and apoptotic actions of TGF-β in homeostasis and cancer. Nat Rev Cancer. 2003;3:807–20.PubMedCrossRef
51.
Zurück zum Zitat Aoki H, Kondo Y, Aldape K, Yamamoto A, Iwado E, Yokoyama T, Hollingsworth EF, Kobayashi R, Hess K, Shinojima N. Monitoring autophagy in glioblastoma with antibody against isoform B of human microtubule-associated protein 1 light chain 3. Autophagy. 2008;4:467–75.PubMedCrossRef Aoki H, Kondo Y, Aldape K, Yamamoto A, Iwado E, Yokoyama T, Hollingsworth EF, Kobayashi R, Hess K, Shinojima N. Monitoring autophagy in glioblastoma with antibody against isoform B of human microtubule-associated protein 1 light chain 3. Autophagy. 2008;4:467–75.PubMedCrossRef
Metadaten
Titel
RETRACTED ARTICLE: TGF-β2 initiates autophagy via Smad and non-Smad pathway to promote glioma cells’ invasion
verfasst von
Chao Zhang
Xin Zhang
Ran Xu
Bin Huang
An-Jing Chen
Chao Li
Jian Wang
Xin-Gang Li
Publikationsdatum
01.12.2017
Verlag
BioMed Central
Erschienen in
Journal of Experimental & Clinical Cancer Research / Ausgabe 1/2017
Elektronische ISSN: 1756-9966
DOI
https://doi.org/10.1186/s13046-017-0628-8

Weitere Artikel der Ausgabe 1/2017

Journal of Experimental & Clinical Cancer Research 1/2017 Zur Ausgabe

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.