BAMBI overexpression together with β-sitosterol ameliorates NSCLC via inhibiting autophagy and inactivating TGF-β/Smad2/3 pathway

  • Authors:
    • Xingchun Wang
    • Minjie Li
    • Mengyao Hu
    • Ping Wei
    • Wei Zhu
  • View Affiliations

  • Published online on: March 15, 2017     https://doi.org/10.3892/or.2017.5508
  • Pages: 3046-3054
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Non-small cell lung cancer (NSCLC) has the highest mortality rate among all solid tumors with a poor prognosis. The BMP and activin receptor membrane bound inhibitor (BAMBI) has been identified as a hallmark of NSCLC and β-sitosterol possesses antitumor potentiality. This study explores the effect of BAMBI overexpression and β-sitosterol in the context of NSCLC. The results revealed that the transfection of pcDNA‑BAMBI and β-sitosterol treatment significantly reduced the levels of autophagy markers light chain 3 (LC3) II and Beclin 1, whereas the levels of LC3 I and p62 were promoted. The reduced punctate accumulations of GFP-LC3 were detected in pcDNA-BAMBI and β-sitosterol groups, especially in pcDNA-BAMBI + β-sitosterol group. BAMBI overexpression and β-sitosterol induced G0/G1 cell cycle arrest and inhibted cell proliferation in A549 cells. In addition, the levels of transforming growth factor-β (TGF‑β)/p-Smad2/3/c-Myc pathway proteins were decreased. The TGF-β overexpression further confirmed that BAMBI overexpression and β-sitosterol treatment suppre­ssed autohagy and viability of A549 cells was through TGF-β/Smad2/3/c-Myc pathway. Finally, the tumor growth was suppressed in NSCLC xenografts, and the inhibitory effect was stronger under treatment of pcDNA-BAMBI toge­ther with β-sitosterol. These results indicate that BAMBI overexpression and β-sitosterol may serve as novel targets for the treatment of NSCLC.

Introduction

Lung cancer is the leading cause of deaths with the most rapidly increasing incidence worldwide. Non-small cell lung cancer (NSCLC) has the highest mortality rate among all solid tumors with a poor prognosis (1). More than half of lung carcinomas are detected in a progressed or already metastasized state with a 5-year survival, for lacking of characteristic early symptoms (2). The general therapy treating the majority of patients is chemotherapy, which often induces resistance (3). It is necessary to develop novel therapeutic approaches to better understand the lung cancer progression.

Autophagy is a fundamental cellular homeostatic process that cells use to degrade and recycle cellular proteins (4). This process can be induced in response to either intracellular or extracellular factors, such as hypoxia, low cellular energy state and organelle damage (5). The microtubule-associated protein 1 light chain 3 (LC3), functions as a structural component in the formation of autophagosomes. The conversion of the cytosolic form of LC3 (LC3 I) to lipidated form (LC3 II) indicates autophagosome formation (6). Beclin 1, is required for the initiation and in the process of autophagosome formation (7). p62 acts as a receptor or adaptor for autophagic degradation of ubiquitinated proteins, the upregulation of p62 is commonly detected in human tumors and contributes directly to tumorigenesis (8). Thus, LC3, Beclin 1 and p62 were considered as autophagy markers in many studies (9,10).

Transforming growth factor-β (TGF-β) has a crucial role in homeostasis, fibrosis angiogenesis, carcinogenesis and differentiation of the cell. A report indicated that TGF-β reduced cell apoptosis via induction of autophagy (11). TGF-β also induces the transcriptional activation of several autophagy-related genes, including BECLIN 1, ATG5, ATG7 and death-associated protein kinase (DAPK) (12,13). Thus hinting that the inhibitor of TGF-β may control cell autophagy and proliferation. BMP and activin receptor membrane bound inhibitor (BAMBI) gene is evolutionally conserved in vertebrates (14). BAMBI was described as a modulator, with a putative function as a dominant negative, non-signaling, competitive pseudo-receptor for members of the TGF-β receptor type 1 (TβR1) family (14). Apart from being suggested as a competitive receptor antagonist for the TGF-β receptor family, BAMBI was indicated as a negative regulator for the subsequent Smad pathway (15), indicating BAMBI may antagonize autophagy through downregulating TGF-β/Smad pathway.

β-Sitosterol is a natural product isolated from traditional Chinese herbs, including Trifolium repens, Houttuynia cordata and Lasia spinosa (16). β-Sitosterol has been applied in treating many diseases because of the anti-inflammatory, anti-proliferative and anticancer effects (17,18). However, the effect of BAMBI together with β-sitosterol on cell autophagy and proliferation in NSCLC is rarely reported.

In this study, we aimed to explore the effect of BAMBI overexpression and β-sitosterol in the context of NSCLC. We found that BAMBI overexpression and β-sitosterol suppressed the autophagy flux of A549 cells. Besides, BAMBI overexpression and β-sitosterol restrained tumor growth in vitro and in vivo through inactivating TGF-β/Smad2/3 pathway. Taken together, our results suggest BAMBI overexpression together with β-sitosterol treatment may provide novel insight into the mechanism and treatment of NSCLC.

Materials and methods

Cell culture

Human NSCLC A549, NCI-H1975 and H1299 cells were obtained from the American Type Culture Collection (ATCC; Manassas, VA, USA), and cultivated in modified Eagle's medium (MEM), supplemented with 20% fetal bovine serum (FBS) and 1% antibiotic-antimycotic (Invitrogen Life Technologies, Carlsbad, CA, USA). The cells were incubated at 37°C in a humidified 21% O2, 5% CO2 atmosphere.

Constructs and transfection

Human BAMBI expression plasmid was constructed by inserting BAMBI cDNA purchased from Beyotime (Shanghai, China) into the pcDNA3-EGFP (Invitrogen, Shanghai, China) expressing vector. The plasmid was amplified with Top 10 bacteria (Invitrogen), extracted, and purified by Plasmid Midi kit (Qiagen, Shanghai, China). For transfection, the three NSCLC cells (A549, NCI-H1975 and H1299) were cultured in antibiotic- and serum-free α-MEM medium (at 50–60% confluence). BAMBI plasmid (2 µg/well) or the vector (2 µg/well) was transfected into NSCLC cells with Lipofectamine™ 2000 (Invitrogen) following the manufacturer's instructions. After transfection for 48 h, BAMBI expression in transfected cells was examined by western blotting.

β-Sitosterol treatment

For the treatment of β-sitosterol, 2×104 A549 cells transfected with or without pcDNA-BAMBI were added to 96-well plates. Then, the RPMI-1640 medium was added to make the volume in each well up to 200 µl. After culturing for 24 h, 10 mg/ml β-sitosterol extracts were added to each well, respectively. Control group was added with equal volume of 0.01 mol/l phosphate-buffered saline (PBS). After incubation at 37°C and 5% CO2 for another 24 h, cells were collected for the following experiments.

Western blotting

NSCLC cells were lysed in lysis buffer (Beyotime) supplemented with 1 mM phenylmethanesulfonyl fluoride (PMSF). The protein concentration was determined using the BCA protein assay (Tiangen Biotech Co., Ltd., Beijing, China). Twenty micrograms of protein in each sample was separated by 12% SDS-PAGE and electrotransferred to polyvinylidene fluoride (PVDF) membranes (Millipore, Billerica, MA, USA) for immunoblotting. The following primary antibodies were used: anti-BAMBI (1:500, ab203070), anti-LC3 (1:500, ab48394), anti-Beclin 1 (1:500, ab55878), anti-p62 (1:200, ab56416), anti-Smad2/3 (1:1,000, ab202445), anti-p-Smad2/3 (1:1,000, ab63399), anti-c-Myc (1:10,000, ab32072), anti-TGF-β (1:500, ab66043), anti-caspase-3 (1:5,000, ab32351), anti-caspase-8 (1:1,000, ab32397), anti-caspase-9 (1:2,000, ab202068), anti-mTOR (1:2,000, ab32028) and anti-GAPDH (1:2,500, ab9485) (all from Abcam, Cambridge, MA, USA), which was used as the internal reference. After incubation with the appropriate horseradish peroxidase (HRP)-conjugated secondary antibody (Santa Cruz Biotechnology, Inc., Santa Cruz, CA, USA), proteins were detected using a ChemiDoc XRS imaging system and Quantity One analysis software (Bio-Rad Laboratories, Inc., San Francisco, CA, USA).

Autophagy measurement using GFP-LC3

The autophagy measurement was conducted according to a previous study (19). Briefly, A549 cells were transfected with a GFP-LC3 expression plasmid (Sigma-Aldrich, St. Louis, MO, USA) incorporated into the lentiviral vector using Lipofectamine 2000 reagent. After selection with puromycin, expression of fluorescence was confirmed by microscopic evaluation before irradiation. Cells were then observed for the fluorescence of GFP-LC3 under a fluorescence microscope.

Autophagic flux measurement

A549 cells were treated with or without 20 µM chloroquine for 1 h prior to β-sitosterol treatment and BAMBI transfection. The recovered cell lysates were used to detect the accumulation of LC3 II, autophagic flux were detected by immunoblotting according to a previous study (20), briefly, cells were rinsed with PBS and lysed with an lysis buffer. The proteins were then separated by SDS-PAGE and transferred onto nitrocellulose membranes. The membranes were incubated with primary antibodies against LC3 and GAPDH and probed with an HRP-labeled secondary antibody and detected using an ECL reagent. Protein expression level was measured by a ChemiDoc XRS imaging system (Bio-Rad Laboratories, Inc.).

Flow cytometry analysis of the cell cycle

A549 cells at 1×106 cells/well were cultured in 6-well plates and transfected with or without pcDNA-BAMBI for 48 h. The cells were then treated with or withour 10 mg/ml β-sitosterol extracts, respectively. Cells were harvested and fixed in 70% ice-cold ethanol for 24 h, followed by staining with propidium iodide (PI). The different cell cycle phases were analyzed with the FACSCalibur instrument using CellQuest software (Becton-Dickinson, Mountain View, CA, USA).

Detection of apoptosis

For the apoptosis assay, A549 cells with β-sitosterol treatment and BAMBI transfection were washed with PBS and stained with PI and Annexin V-FITC (V13241; Invitrogen Life Technologies) on ice for 10–15 min. The stained cells were analyzed using a FACScan flow cyto-meter (Becton-Dickinson) and the number of apoptotic cells was quantified using FlowJo software (Tree Star Inc., Ashland, OR, USA).

3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay

The cell viability of A549 cells was assessed using MTT assay. Shortly afterwards, cells were transfected according to the above description and were seeded in 96-well plates at 6×103 cells/well. The surviving fractions were determined at 0, 24, 48, 72, 96 and 120 h. For the detection the cytotoxicity of autophagy inhibitors on A549 cells, cells were pretreated with or without 100 nM bafilomycin A1 for 1 h prior to β-sitosterol treatment and BAMBI transfection. The surviving fractions were determined after 24 h. Thereafter, the old medium was discarded and fresh medium containing MTT (5 mg/ml MTT in PBS; Sangon Biotech Co., Ltd., Shanghai, China) was added and incubated for an additional 4 h. Then, cell proliferation was measured with a spectrophotometer (Bio-Rad Laboratories, Inc.) at 470 nm. Each experiment was performed in triplicate.

TGF-β overexpression

The TGF-β overexpression was achieved by PCR amplification using TGF-β cDNA as a template, and the hNUDC expressing vector was constructed by inserting the TGF-β cDNA into pcDNA3.1 vector. The recombinant plasmid was transfected into 3×106 A549 cells with or without pcDNA-BAMBI transfection and β-sitosterol treatment using a nucleofector instrument. Forty-eight hours later, subsequent experiments were performed on the cells. The experiment was replicated thrice for data calculations.

NSCLC xenografts

Twenty NOD/SCID mice (Jackson Laboratory, Bar Harbor, ME, USA) (male; body weight, 20–22 g; age, 8-weeks) were purchased from the Institute of Zoology, Chinese Academy of Medical Sciences (Shanghai, China). A549 cells (5×106) were injected subcutaneously into ten NOD/SCID mice, another ten mice were injected with equal number of A549 cells transfected with pcDNA-BAMBI. Half of the mice with or without pcDNA-BAMBI transfection were then injected with β-sitosterol (1 mg/kg body weight) every other day. The tumor volumes were measured daily after the injection, and all the rats were assigned to euthanasia at the end of measurements on day 25. All animal experiments were performed according to current prescribed guidelines and under a protocol approved by the Institutional Animal Care and Use Committee.

Statistical analysis

All results are presented as mean ± SD from a minimum of three replicates. Differences between groups were evaluated by SPSS version 15.0 statistical software (SPSS, Inc., Chicago, IL, USA) by one-way analysis of variance (ANOVA) followed by Bonferroni's test. Differences were considered statistically significant at P<0.05.

Results

BAMBI is overexpressed by transfection of the plasmid of pcDNA-BAMBI into NSCLC cells

The pcDNA-BAMBI plasmid was transfected into three NSCLC cell lines (NCI-H1975, H1299 and A549). No significant difference of BAMBI level was detected between control and pcDNA groups, but the expression of BAMBI was strongly promoted in pcDNA-BAMBI group compared with pcDNA group, especially in A549 cells (P<0.05) (Fig. 1A-C). The results indicated that BAMBI was overexpressed successfully by inserting its plasmid into NSCLC cells.

BAMBI overexpression and β-sitosterol suppress autophagy of A549 cells

We next explored whether BAMBI overexpression played a functional role in autophagy. A549 cells were chosen for the following experiments. β-Sitosterol was added alone or with pcDNA-BAMBI transfection. Western blotting indicated that LC3 II and Beclin 1 expression levels declined significantly in pcDNA-BAMBI and β-sitosterol groups compared with control group (P<0.05). In comparison with β-sitosterol function alone, the levels of LC3 II and Beclin 1 were particularly decreased when adding β-sitosterol in pcDNA-BAMBI group (P<0.05). Conversely, the levels of LC3 I and autophagy substrate p62 were increased significantly under the treatment of pcDNA-BAMBI and β-sitosterol (P<0.05). The expression of LC3 I and p62 were strongly promoted under treatment of pcDNA-BAMBI together with β-sitosterol (P<0.05) (Fig. 2A-C). In addition, immunofluorescent assay displayed reduced punctate accumulations of GFP-LC3 in pcDNA-BAMBI and β-sitosterol groups, especially in pcDNA-BAMBI + β-sitosterol group compared with control group (Fig. 2D). In addition, lipidated LC3 II degradation were used to monitor autophagic flux as the LC3 II is degraded by autolysosome. Thus, LC3 II in cells with or without chloroquine was used to examine the effects of BAMBI overexpression and β-sitosterol on autophagic flux. Compared to the control group, the level of LC3 II was higher in A549 cells with BAMBI overexpression and β-sitosterol treatment (P<0.05). However, no significant difference was observed in cells pretreated with or without chloroquine and treated with β-sitosterol with BAMBI transfection (P>0.05) (Fig. 2E). These results suggested that BAMBI overexpression and β-sitosterol suppressed autophagy of A549 cells.

BAMBI overexpression and β-sitosterol inhibit cell progression and proliferation of A549 cells

Flow cytometric analysis exhibited that G0/G1 phase arrest in A549 cells was observed in pcDNA-BAMBI and β-sitosterol groups. Besides, a large accumulation of G0/G1 phase was measured in pcDNA-BAMBI + β-sitosterol group (Fig. 3A and B). These results suggested an inactivation of G0/G1 progression in A549 cells following incubation with pcDNA-BAMBI and β-sitosterol. Besides, cell apoptosis assay indicated that BAMBI transfection strengthened the apoptosis-inducing effect of β-sitosterol in A549 cells. The number of apoptotic cells was increased in pcDNA-BAMBI + β-sitosterol group compared with β-sitosterol group (P<0.05) (Fig. 3C and D). Accordingly, the expression level of caspase-3, −8 and −9 was increased, while the level of mTOR was decreased in cells treated with β-sitosterol together with BAMBI overexpression compared with β-sitosterol group (P<0.05) (Fig. 3E). Further, the cell proliferation assay was performed in A549 cells. The transfection of pcDNA-BAMBI and β-sitosterol treatment strongly decreased cell growth compared with control group (P<0.01). The inhibitory effect was stronger in pcDNA-BAMBI + β-sitosterol group compared β-sitosterol group (P<0.05) (Fig. 3F). The cytotoxicity of autophagy inhibitor bafilomycin A1 on A549 cells was measured, the results displayed that pcDNA-BAMBI and β-sitosterol decreased the viability of A549 cells, the adding of bafilomycin A1 enhanced this effect (Fig. 3G). These findings suggest that BAMBI overexpression and β-sitosterol inhibited the viabilityof A549 cells.

The TGF-β/Smad2/3 pathway is restrained by BAMBI overexpression and β-sitosterol

BAMBI is a member of TGF-β family, we further explored the effect of pcDNA-BAMBI and β-sitosterol on the expression of TGF-β pathway proteins. The results indicated that the level of Smad2/3 exhibited no significant difference, while the expression of TGF-β, p-Smad2/3 and c-Myc were decreased by pcDNA-BAMBI and β-sitosterol compared with control group (P<0.05). Moreover, the protein expression was strongly suppressed under the treatment of pcDNA-BAMBI together with β-sitosterol (Fig. 4A-E). These results suggested that BAMBI overexpression and β-sitosterol restrained the TGF-β/Smad2/3/c-Myc pathway.

The inhibitory effect of BAMBI and β-sitosterol on autophagy and viability of A549 cells is through TGF-β/Smad2/3 pathway

To confirm the TGF-β pathway was involved in the inhibition effect of BAMBI and β-sitosterol on the viability of A549. Firstly, the plasmid of pcDNA-TGF-β was constructed and transfected into A549 cells. Western blotting revealed that the level of TGF-β, p-Smad2/3 and c-Myc was promoted in pcDNA-TGF-β group in comparison with control group (P<0.01). The downregulation of their expression was partially counteracted when cells in pcDNA-BAMBI + β-sitosterol group were transfected with pcDNA-TGF-β (P<0.05) (Fig. 4F-I).

We measured the levels of autophagy markers, the results displayed that A549 cells transfected pcDNA-TGF-β exhibited enhanced expression of LC3 II and Beclin 1 with decreased LC3 I and p62 levels compared with control group (P<0.05). The expression of LC3 II and Beclin 1 was strongly restrained in pcDNA-BAMBI + β-sitosterol group in comparison with control group (P<0.05), and their levels were increased adding pcDNA-TGF-β in pcDNA-BAMBI + β-sitosterol group (P<0.05). The opposite behavior was observed in the expression of LC3 I and p62 (Fig. 5A and B). In addition, a large accumulation of GFP-LC3 was measured by pcDNA-TGF-β transfection. The GFP-LC3 II punctate accumulation was significantly reduced in pcDNA-BAMBI + β-sitosterol, but increased adding pcDNA-TGF-β in pcDNA-BAMBI + β-sitosterol groups (Fig. 5C).

In accordance with these results, the G0/G1 phase and cell proliferation were accelerated in pcDNA-TGF-β group compared with control group. In addition, the cell cycle arrest and the inhibition of cell growth were partially offset when cells in pcDNA-BAMBI + β-sitosterol group were transfected with pcDNA-TGF-β (P<0.05) (Fig. 6A-C). These results indicated that the inhibitory effect of BAMBI overexpression and β-sitosterol on the viability of A549 is through the TGF-β/Smad2/3/c-Myc pathway.

BAMBI overexpression and β-sitosterol suppress the proliferation of NSCLC in vivo

To further explore the tumor suppression effect of BAMBI overexpression and β-sitosterol, we assessed tumor growth of NSCLC xenografts under treatment of pcDNA-BAMBI and/or β-sitosterol. As shown in Fig. 7A and B, tumor growth was suppressed in pcDNA-BAMBI and β-sitosterol groups compared with control group (P<0.05), and the minimum tumor volume was detected in pcDNA-BAMBI + β-sitosterol group. The in vivo experiments were convincing that BAMBI overexpression and β-sitosterol suppressed the proliferation of NSCLC.

Discussion

Chemotherapy is common in treatment of NSCLC, but the development of chemoresistance in NSCLC is a major obstacle in treating patients (21). A study indicated that human lung cancer tissues that experienced chemotherapy showed increase of autophagy (22). Thus, a strategy that is specific to anti-autophagy with tumor-suppressive effect would be beneficial in treating NSCLC. BAMBI is a type I TGF-β receptor antagonist, the epigenetic silencing of BAMBI was identified as a hallmark of NSCLC (3). The methanol extract from β-sitosterol possesses antitumor potentiality. The mechanism of BAMBI and β-sitosterol on cell autophagy and growth was explored in this study.

The physiological function of BAMBI remains unclear, high expression of BAMBI has been detected in colorectal cancer (11) and ovarian cancer (12). On the contrary, BAMBI is epigenetically silenced in high-grade bladder cancer (14) and absent from breast cancer (15). In the context of lung diseases, BAMBI was almost completely absent from the lung cancer tissues compared with the tumor-free lung tissues. Besides, BAMBI downregulation drives the invasiveness of NSCLC (3), indicating that the upregulation of BAMBI could reduce the severity of NSCLC. In this study, BAMBI was overexpressed successfully by inserting its plasmid into three NSCLC cell lines (NCI-H1975, H1299 and A549). Recent studies revealed the paradoxical nature of autophagy in deciding cell-fate machinery. Autophagy induces cell death, suppresses inflammation and enhances genomic stability; on the contrary, autophagy also renders cells viable in stressful conditions and is considered a pro-survival mechanism (23,24). Studies indicated that TGF-β controls autophagic responses during angiogenesis, and fibrogenesis in many human cellular systems, such as atrial myofibroblasts (25), renal tubular epithelial (26) and endothelial cells (27). Research also indicated that the TGF-β-activated fibroblasts increased autophagy and greatly sustained the growth of breast cancer cells (28). These findings suggested that the inhibitor of TGF-β may prevent tumorigenesis by regulating autophagy. As the antagonist of TGF-β receptor, the transfection of pcDNA-BAMBI together with β-sitosterol treatment in this study significantly reduced the levels of LC3 II and Beclin 1 with decreased GFP-LC3 punctuate structures, whereas the levels of LC3 I and p62 were strongly promoted. Besides, no significant increase of autophagy flux was observed in cells pretreated with or without chloroquine and treated with β-sitosterol and BAMBI transfection. These results suggested that BAMBI overexpression and β-sitosterol suppressed autophagy in NSCLC cells.

Apart from the effect in cell autophagy, BAMBI gene suppression was indicated as one of the epigenetic events affecting the invasiveness or aggressiveness of bladder cancers (29) and NSCLC (3). BAMBI overexpression significantly increased the number of apoptotic cells in T24 line (29). Research also indicated that BAMBI was stimulated by fibroblast growth factor 18, which increased apoptosis in ovarian granulosa cells (30). These studies suggested a cell apoptosis-inducing role of BAMBI. As component in active fractions of plants, β-sitosterol has a protective effect against colon, prostate, stomach, ovarian and breast cancers via interfering with multiple cell signaling pathways, including cell cycle, apoptosis, proliferation, invasion, angiogenesis and carcinogenesis (3133). Animal studies have shown that β-sitosterol, extracted from Aristolochia mollissima Hance, has an inhibitory effect on the proliferation of osteosarcoma HOS cells (34). β-Sitosterol has also been reported to affect cell cycle progression by inducing sub-G1 arrest in human colon cancer cells (HT-29) (35) and U266 multiple myeloma cells (36). Consistent with these studies, pcDNA-BAMBI and β-sitosterol applied in this study suppressed cell proliferation and induced accumulation of G0/G1 arrest and cell apoptosis in A549 cells. The inhibitory effect was stronger when cells were under pcDNA-BAMBI transfection plus β-sitosterol treatment. These findings revealed an anticancer effect of BAMBI overexpression and β-sitosterol in NSCLC cells.

The TGF-β signaling pathway is believed to contribute to carcinoma development by increasing cancer cell motility, invasiveness and metastasis (37). TGF-β has immunosuppressive properties and may enable cancer cells to evade the immune responses (38). Upon binding of the ligand to TGF-β receptors type 2 (TGF-βR2), TGF-βR1 is phosphorylated and recruits SMAD2 and SMAD3, migrates to the nucleus, and stimulates target gene expression (39). In lung cancer, high TGF-β serum levels is associated with a poor prognosis, lymph node metastasis, and tumor progression (40,41). In addition, TGF-β was suggested as an independent risk factor for the occurrence of pulmonary metastasis in NSCLC (42). A study also revealed that the presence of p-SMAD2 and 3 was significantly higher in the NSCLC cancer tissues compared with the tumor-free lung tissues (3). In this study, the level of TGF-β, p-Smad2/3 and c-Myc was decreased by pcDNA-BAMBI transfection and β-sitosterol treatment. The transfection of pcDNA-TGF-β further confirmed the inhibitory effect in this pathway. In addition, the inhibition of cell autophagy and cell survival were partially offset when cells in pcDNA-BAMBI + β-sitosterol group were transfected with pcDNA-TGF-β. These results confirmed that BAMBI overexpression and β-sitosterol suppression of cell autophagy and viability of NSCLC is through the TGF-β/Smad2/3/c-Myc pathway.

In vivo experiment showed that BAMBI transduction abolished protumor effects of an orthotopic breast cancer xenograft model (43) and modulated the effects of diabetes via inhibiting TGF-β signaling (44). Our study found that BAMBI overexpression and β-sitosterol suppressed tumor growth in NSCLC xenografts, revealing the antitumor effect of BAMBI overexpression and β-sitosterol.

In conclusion, this study explored pcDNA-BAMBI transfection and β-sitosterol treatment on autophagy and progression of NSCLC cells. We found that BAMBI overexpression and β-sitosterol suppressed cell autophagy, induced G0/G1 cell cycle arrest and inhibited cell proliferation in vitro and in vivo possibly through inactivating the TGF-β/Smad2/3/c-Myc pathway. Moreover, the inhibitory effect was stronger when pcDNA-BAMBI and β-sitosterol functioned together. These results indicate that BAMBI overexpression and β-sitosterol may serve as novel targets for the treatment of NSCLC.

Acknowledgements

The authors would like to thank the members of Internal Medicine, the Cancer Hospital of Linyi, for providing technical support and helpful discussions concerning the present study.

Glossary

Abbreviations

Abbreviations:

NSCLC

non-small cell lung cancer

BAMBI

BMP and activin receptor membrane bound inhibitor

LC3

light chain 3

TGF-β

transforming growth factor-β

References

1 

Jemal A, Siegel R, Ward E, Hao Y, Xu J, Murray T and Thun MJ: Cancer statistics, 2008. CA Cancer J Clin. 58:71–96. 2008. View Article : Google Scholar : PubMed/NCBI

2 

Siegel R, Naishadham D and Jemal A: Cancer statistics, 2012. CA Cancer J Clin. 62:10–29. 2012. View Article : Google Scholar : PubMed/NCBI

3 

Marwitz S, Depner S, Dvornikov D, Merkle R, Szczygieł M, Müller-Decker K, Lucarelli P, Wäsch M, Mairbäurl H, Rabe KF, et al: Downregulation of the TGFβ pseudoreceptor BAMBI in non-small cell lung cancer enhances TGFβ signaling and invasion. Cancer Res. 76:3785–3801. 2016. View Article : Google Scholar : PubMed/NCBI

4 

Xie Z and Klionsky DJ: Autophagosome formation: core machinery and adaptations. Nat Cell Biol. 9:1102–1109. 2007. View Article : Google Scholar : PubMed/NCBI

5 

Mizushima N: Physiological functions of autophagy. Curr Top Microbiol Immunol. 335:71–84. 2009.PubMed/NCBI

6 

Suzuki K, Kirisako T, Kamada Y, Mizushima N, Noda T and Ohsumi Y: The pre-autophagosomal structure organized by concerted functions of APG genes is essential for autophagosome formation. EMBO J. 20:5971–5981. 2001. View Article : Google Scholar : PubMed/NCBI

7 

Yue Z, Jin S, Yang C, Levine AJ and Heintz N: Beclin 1, an autophagy gene essential for early embryonic development, is a haploinsufficient tumor suppressor. Proc Natl Acad Sci USA. 100:15077–15082. 2003. View Article : Google Scholar : PubMed/NCBI

8 

Son YO, Pratheeshkumar P, Roy RV, Hitron JA, Wang L, Zhang Z and Shi X: Nrf2/p62 signaling in apoptosis resistance and its role in cadmium-induced carcinogenesis. J Biol Chem. 289:28660–28675. 2014. View Article : Google Scholar : PubMed/NCBI

9 

Masuda GO, Yashiro M, Kitayama K, Miki Y, Kasashima H, Kinoshita H, Morisaki T, Fukuoka T, Hasegawa T, Sakurai K, et al: Clinicopathological correlations of autophagy-related proteins LC3, Beclin 1 and p62 in gastric cancer. Anticancer Res. 36:129–136. 2016.PubMed/NCBI

10 

Deng Q, Wang Z, Wang L, Zhang L, Xiang X, Wang Z and Chong T: Lower mRNA and protein expression levels of LC3 and Beclin1, markers of autophagy, were correlated with progression of renal clear cell carcinoma. Jpn J Clin Oncol. 43:1261–1268. 2013. View Article : Google Scholar : PubMed/NCBI

11 

Ding Y, Kim JK, Kim SI, Na HJ, Jun SY, Lee SJ and Choi ME: TGF-{beta}1 protects against mesangial cell apoptosis via induction of autophagy. J Biol Chem. 285:37909–37919. 2010. View Article : Google Scholar : PubMed/NCBI

12 

Korah J, Canaff L and Lebrun JJ: The retinoblastoma tumor suppressor protein (pRb)/E2 promoter binding factor 1 (E2F1) pathway as a novel mediator of TGFβ-induced autophagy. J Biol Chem. 291:2043–2054. 2016. View Article : Google Scholar : PubMed/NCBI

13 

Kiyono K, Suzuki HI, Matsuyama H, Morishita Y, Komuro A, Kano MR, Sugimoto K and Miyazono K: Autophagy is activated by TGF-beta and potentiates TGF-beta-mediated growth inhibition in human hepatocellular carcinoma cells. Cancer Res. 69:8844–8852. 2009. View Article : Google Scholar : PubMed/NCBI

14 

Onichtchouk D, Chen YG, Dosch R, Gawantka V, Delius H, Massagué J and Niehrs C: Silencing of TGF-beta signalling by the pseudoreceptor BAMBI. Nature. 401:480–485. 1999. View Article : Google Scholar : PubMed/NCBI

15 

Seki E, De Minicis S, Osterreicher CH, Kluwe J, Osawa Y, Brenner DA and Schwabe RF: TLR4 enhances TGF-beta signaling and hepatic fibrosis. Nat Med. 13:1324–1332. 2007. View Article : Google Scholar : PubMed/NCBI

16 

Vijaya. Yadav AK: In vitro anthelmintic assessment of selected phytochemicals against Hymenolepis diminuta, a zoonotic tapeworm. J Parasit Dis. 40:1082–1086. 2016. View Article : Google Scholar : PubMed/NCBI

17 

Dey YN, Sharma G, Wanjari MM, Kumar D, Lomash V and Jadhav AD: Beneficial effect of Amorphophallus paeoniifolius tuber on experimental ulcerative colitis in rats. Pharm Biol. 55:53–62. 2016. View Article : Google Scholar : PubMed/NCBI

18 

Cheng D, Guo Z and Zhang S: Effect of β-sitosterol on the expression of HPV E6 and p53 in cervical carcinoma cells. Contemp Oncol (Pozn). 19:36–42. 2015.PubMed/NCBI

19 

Jo GH, Bögler O, Chwae YJ, Yoo H, Lee SH, Park JB, Kim YJ, Kim JH and Gwak HS: Radiation-induced autophagy contributes to cell death and induces apoptosis partly in malignant glioma cells. Cancer Res Treat. 47:221–241. 2015. View Article : Google Scholar : PubMed/NCBI

20 

Shu CW, Chang HT, Wu CS, Chen CH, Wu S, Chang HW, Kuo SY, Fu E, Liu PF and Hsieh YD: RelA-mediated BECN1 expression is required for reactive oxygen species-induced autophagy in oral cancer cells exposed to low-power laser irradiation. PLoS One. 11:e01605862016. View Article : Google Scholar : PubMed/NCBI

21 

Toge M, Yokoyama S, Kato S, Sakurai H, Senda K, Doki Y, Hayakawa Y, Yoshimura N and Saiki I: Critical contribution of MCL-1 in EMT-associated chemo-resistance in A549 non-small cell lung cancer. Int J Oncol. 46:1844–1848. 2015.PubMed/NCBI

22 

Lee JG, Shin JH, Shim HS, Lee CY, Kim DJ, Kim YS and Chung KY: Autophagy contributes to the chemo-resistance of non-small cell lung cancer in hypoxic conditions. Respir Res. 16:1382015. View Article : Google Scholar : PubMed/NCBI

23 

Ju LL, Zhao CY, Ye KF, Yang H and Zhang J: Expression and clinical implication of Beclin1, HMGB1, p62, survivin, BRCA1 and ERCC1 in epithelial ovarian tumor tissues. Eur Rev Med Pharmacol Sci. 20:1993–2003. 2016.PubMed/NCBI

24 

Jiang K, Liu M, Lin G, Mao B, Cheng W, Liu H, Gal J, Zhu H, Yuan Z, Deng W, et al: Tumor suppressor Spred2 interaction with LC3 promotes autophagosome maturation and induces autophagy-dependent cell death. Oncotarget. 7:25652–25667. 2016.PubMed/NCBI

25 

Ghavami S, Cunnington RH, Gupta S, Yeganeh B, Filomeno KL, Freed DH, Chen S, Klonisch T, Halayko AJ, Ambrose E, et al: Autophagy is a regulator of TGF-β1-induced fibrogenesis in primary human atrial myofibroblasts. Cell Death Dis. 6:e16962015. View Article : Google Scholar : PubMed/NCBI

26 

Ding Y, Kim S, Lee SY, Koo JK, Wang Z and Choi ME: Autophagy regulates TGF-β expression and suppresses kidney fibrosis induced by unilateral ureteral obstruction. J Am Soc Nephrol. 25:2835–2846. 2014. View Article : Google Scholar : PubMed/NCBI

27 

Pan CC, Kumar S, Shah N, Bloodworth JC, Hawinkels LJ, Mythreye K, Hoyt DG and Lee NY: Endoglin regulation of Smad2 function mediates Beclin1 expression and endothelial autophagy. J Biol Chem. 290:14884–14892. 2015. View Article : Google Scholar : PubMed/NCBI

28 

Guido C, Whitaker-Menezes D, Capparelli C, Balliet R, Lin Z, Pestell RG, Howell A, Aquila S, Andò S, Martinez-Outschoorn U, et al: Metabolic reprogramming of cancer-associated fibroblasts by TGF-β drives tumor growth: connecting TGF-β signaling with ‘Warburg-like’ cancer metabolism and L-lactate production. Cell Cycle. 11:3019–3035. 2012. View Article : Google Scholar : PubMed/NCBI

29 

Khin SS, Kitazawa R, Win N, Aye TT, Mori K, Kondo T and Kitazawa S: BAMBI gene is epigenetically silenced in subset of high-grade bladder cancer. Int J Cancer. 125:328–338. 2009. View Article : Google Scholar : PubMed/NCBI

30 

Jiang Z, Guerrero-Netro HM, Juengel JL and Price CA: Divergence of intracellular signaling pathways and early response genes of two closely related fibroblast growth factors, FGF8 and FGF18, in bovine ovarian granulosa cells. Mol Cell Endocrinol. 375:97–105. 2013. View Article : Google Scholar : PubMed/NCBI

31 

Shoja MH, Reddy ND, Nayak PG, Srinivasan KK and Rao CM: Glycosmis pentaphylla (Retz.) DC arrests cell cycle and induces apoptosis via caspase-3/7 activation in breast cancer cells. J Ethnopharmacol. 168:50–60. 2015. View Article : Google Scholar : PubMed/NCBI

32 

Sayeed Bin MS and Ameen SS: Beta-sitosterol: a promising but orphan nutraceutical to fight against cancer. Nutr Cancer. 67:1214–1220. 2015.PubMed/NCBI

33 

Baskar AA, Al Numair KS, Gabriel Paulraj M, Alsaif MA, Muamar MA and Ignacimuthu S: β-Sitosterol prevents lipid peroxidation and improves antioxidant status and histoarchitecture in rats with 1,2-dimethylhydrazine-induced colon cancer. J Med Food. 15:335–343. 2012. View Article : Google Scholar : PubMed/NCBI

34 

Yu Y, Bo Z, Chao H and Minghua Z: A study on the anticancer activity of ethanol extract of Aristolochia mollissima Hance on osteosarcoma HOS cells. Afr J Tradit Complement Altern Med. 10:551–554. 2013. View Article : Google Scholar : PubMed/NCBI

35 

Li C and Wang MH: Aristolochia debilis Sieb. et Zucc. induces apoptosis and reactive oxygen species in the HT-29 human colon cancer cell line. Cancer Biother Radiopharm. 28:717–724. 2013. View Article : Google Scholar : PubMed/NCBI

36 

Sook SH, Lee HJ, Kim JH, Sohn EJ, Jung JH, Kim B, Kim JH, Jeong SJ and Kim SH: Reactive oxygen species-mediated activation of AMP-activated protein kinase and c-Jun N-terminal kinase plays a critical role in beta-sitosterol-induced apoptosis in multiple myeloma U266 cells. Phytother Res. 28:387–394. 2014. View Article : Google Scholar : PubMed/NCBI

37 

Akhurst RJ and Derynck R: TGF-beta signaling in cancer - a double-edged sword. Trends Cell Biol. 11:S44–S51. 2001. View Article : Google Scholar : PubMed/NCBI

38 

Kumai T, Oikawa K, Aoki N, Kimura S, Harabuchi Y, Celis E and Kobayashi H: Tumor-derived TGF-beta and prostaglandin E2 attenuate anti-tumor immune responses in head and neck squamous cell carcinoma treated with EGFR inhibitor. J Transl Med. 12:2652014. View Article : Google Scholar : PubMed/NCBI

39 

Moustakas A and Heldin CH: The regulation of TGFbeta signal transduction. Development. 136:3699–3714. 2009. View Article : Google Scholar : PubMed/NCBI

40 

Chen Y, Zou L, Zhang Y, Chen Y, Xing P, Yang W, Li F, Ji X, Liu F and Lu X: Transforming growth factor-β1 and α-smooth muscle actin in stromal fibroblasts are associated with a poor prognosis in patients with clinical stage I–IIIA nonsmall cell lung cancer after curative resection. Tumour Biol. 35:6707–6713. 2014. View Article : Google Scholar : PubMed/NCBI

41 

Yang H, Wang L, Zhao J, Chen Y, Lei Z, Liu X, Xia W, Guo L and Zhang HT: TGF-β-activated SMAD3/4 complex transcriptionally upregulates N-cadherin expression in non-small cell lung cancer. Lung Cancer. 87:249–257. 2015. View Article : Google Scholar : PubMed/NCBI

42 

Zu L, Xue Y, Wang J, Fu Y, Wang X, Xiao G, Hao M, Sun X, Wang Y, Fu G, et al: The feedback loop between miR-124 and TGF-β pathway plays a significant role in non-small cell lung cancer metastasis. Carcinogenesis. 37:333–343. 2016. View Article : Google Scholar : PubMed/NCBI

43 

Shangguan L, Ti X, Krause U, Hai B, Zhao Y, Yang Z and Liu F: Inhibition of TGF-β/Smad signaling by BAMBI blocks differentiation of human mesenchymal stem cells to carcinoma-associated fibroblasts and abolishes their protumor effects. Stem Cells. 30:2810–2819. 2012. View Article : Google Scholar : PubMed/NCBI

44 

Fan Y, Li X, Xiao W, Fu J, Harris RC, Lindenmeyer M, Cohen CD, Guillot N, Baron MH, Wang N, et al: BAMBI elimination enhances alternative TGF-β signaling and glomerular dysfunction in diabetic mice. Diabetes. 64:2220–2233. 2015. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

May-2017
Volume 37 Issue 5

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Wang X, Li M, Hu M, Wei P and Zhu W: BAMBI overexpression together with β-sitosterol ameliorates NSCLC via inhibiting autophagy and inactivating TGF-β/Smad2/3 pathway. Oncol Rep 37: 3046-3054, 2017
APA
Wang, X., Li, M., Hu, M., Wei, P., & Zhu, W. (2017). BAMBI overexpression together with β-sitosterol ameliorates NSCLC via inhibiting autophagy and inactivating TGF-β/Smad2/3 pathway. Oncology Reports, 37, 3046-3054. https://doi.org/10.3892/or.2017.5508
MLA
Wang, X., Li, M., Hu, M., Wei, P., Zhu, W."BAMBI overexpression together with β-sitosterol ameliorates NSCLC via inhibiting autophagy and inactivating TGF-β/Smad2/3 pathway". Oncology Reports 37.5 (2017): 3046-3054.
Chicago
Wang, X., Li, M., Hu, M., Wei, P., Zhu, W."BAMBI overexpression together with β-sitosterol ameliorates NSCLC via inhibiting autophagy and inactivating TGF-β/Smad2/3 pathway". Oncology Reports 37, no. 5 (2017): 3046-3054. https://doi.org/10.3892/or.2017.5508