MAP30 promotes apoptosis of U251 and U87 cells by suppressing the LGR5 and Wnt/β-catenin signaling pathway, and enhancing Smac expression

  • Authors:
    • Yilin Jiang
    • Junjie Miao
    • Dongliang Wang
    • Jingru Zhou
    • Bo Liu
    • Feng Jiao
    • Jiangfeng Liang
    • Yangshuo Wang
    • Cungang Fan
    • Qingjun Zhang
  • View Affiliations

  • Published online on: February 16, 2018     https://doi.org/10.3892/ol.2018.8073
  • Pages: 5833-5840
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Significant antitumor activity of Momordica anti-human immunodeficiency virus protein of 30 kDa (MAP30) purified from Momordica charantia has been the subject of previous research. However, the effective mechanism of MAP30 on malignant glioma cells has not yet been clarified. The aim of the present study was to investigate the effects and mechanism of MAP30 on U87 and U251 cell lines. A Cell Counting Kit‑8 assay, wound healing assay and Transwell assay were used to detect the effects on U87 and U251 cells treated with different concentrations of MAP30 (0.5, 1, 2, 4, 8 and 16 µM) over different periods of time. Proliferation, migration and invasion of each cell line were markedly inhibited by MAP30 in a dose‑ and time‑dependent manner. Flow cytometry and fluorescence staining demonstrated that apoptosis increased and the cell cycle was arrested in S‑phase in the two investigated cell lines following MAP30 treatment. Western blot analysis demonstrated that leucine‑rich‑repeat‑containing G‑protein‑coupled receptor 5 (LGR5) expression and key proteins in the Wnt/β‑catenin signaling pathway were apparently decreased, whereas second mitochondria‑derived activator of caspase (Smac) protein expression significantly increased with MAP30 treatment in the same manner. These results suggest that MAP30 markedly induces apoptosis in U87 and U251 cell lines by suppressing LGR5 and the Wnt/β‑catenin signaling pathway, and enhancing Smac expression in a dose‑ and time‑dependent manner.

Introduction

Gliomas are the most common primary brain tumor in adults and are categorized into four World Health Organization grades on the basis of histopathological characteristics (1). Despite current multidisciplinary treatments including surgery, radiotherapy and chemotherapy, no significant prognostic improvement has been obtained in patients with gliomas over the last decade, because of the aggressive nature of high-grade gliomas, the survival rates of patients with high-grade gliomas are <10% at 5 years (2). Previously, efforts have been made to identify new effective therapeutic agents.

Momordica anti-human immunodeficiency virus protein of 30 kDa (MAP30), first extracted and purified from Momordica charantia in 1990, is a type I ribosome-inactivating protein (RIP) (3). Previous studies indicated that MAP30 exhibits a variety of anti-infection, anti-diabetic, antiviral and antitumor bioactive effects (410). The antitumor ability of MAP30 has been the subject of previous studies (1115). However, certain researchers have already noted the effects of MAP30 on the inhibition of U87 cells in screening for anticancer effects (3,7,16,17). However, the mechanism of MAP30 on glioma cells has not been elucidated in detail. The aim of the present study was to investigate the effects and mechanism of MAP30 on U87 and U251 glioblastoma cell lines in vitro. Although some researchers identified that the DNA profile of U87MG was different from that of the original cells in 2016, it is likely to be a bona fide human glioblastoma cell line of unknown origin; therefore, in the present study the U87 and U251 cell lines were used.

Materials and methods

Materials

High-glucose Dulbecco's modified Eagle's medium (DMEM), fetal bovine serum (FBS), penicillin G and streptomycin were all purchased from Gibco; Thermo Fisher Scientific, Inc. (Waltham, MA, USA). The U87 and U251 human malignant glioblastoma cell lines were purchased from China Infrastructure of Cell Line Resources (Beijing, China). MAP30 was kindly provided by Professor Y.F. Meng (College of Life Sciences, Sichuan University, Chengdu, China) (18). The primary antibodies against leucine-rich-repeat-containing G-protein-coupled receptor 5 (LGR5; cat. no. AB75850), β-catenin (cat. no. AB32572), glycogen synthase kinase 3β phosphorylated at Ser9 (pGSK-3βSer9; cat. no. AB75814), c-Myc (cat. no. AB32072), cyclin D1 (cat. no. AB134175) and second mitochondria-derived activator of caspase (Smac; cat. no. AB32023) used for western blot analysis were purchased from Abcam (Cambridge, UK).

Cell culture

U87 and U251 cells were cultured in DMEM supplemented with 10% FBS, 100 U/ml penicillin and 100 U/ml streptomycin at 37°C in a humidified atmosphere of 5% CO2 in air. The medium was changed every 3–4 days, and cultures were split using 0.25% trypsin. All experiments were carried out on cells with viability >95%.

Cell viability assay

When U87 and U251 cells reached the exponential growth phase, they were treated with MAP30 (2.7 µM) or PBS (0.01 M, pH 7.2–7.4; Gibco; Thermo Fisher Scientific, Inc.), and images were captured under an inverted light microscope (×400 magnification; Olympus CKX41; Olympus Corporation, Tokyo, Japan) at 0 and 24 h.

A Cell Counting Kit-8 (CCK-8; Dojindo Molecular Technologies, Inc., Kumamoto, Japan) assay was used to evaluate cell viability. U87 and U251 cells were resuspended in DMEM with 10% FBS at a density of 5×103 cells/well and cultured in 96-well plates. The cells were treated with various concentrations (0.5, 1, 2, 4, 8 and 16 µM) of MAP30 for 24, 48 and 72 h, prior to medium being replaced with 10 µl CCK-8 and 100 µl FBS-free DMEM. Cells were incubated for 1 h at 37°C and the absorbance of each well was measured at 450 nm using a microplate reader.

Cell migration and invasion assays

The effect of MAP30 on glioma cell migration was assessed using wound-healing assays. U87 and U251 cells were seeded in multiwell plates and cultured until reaching confluence. A 10-µl pipette tip was used to make a straight scratch. The cells were washed with PBS, and treated for 24 h with MAP30 (2.7 µM) or PBS (0.01 M, pH 7.2–7.4) as a control. Images of the wounds were captured under an inverted light microscope (×400 magnification) at 0 and 24 h, and measured.

A cell invasion assay was performed using 24-well Transwell chambers with an 8 µm aperture (Costar; Corning Incorporated, Corning, New York, NY, USA). The inserts were coated with Matrigel (BD Biosciences, Franklin Lakes, NJ, USA). U87 and U251 cells were trypsinized and transferred to the upper Matrigel chamber with FBS-free DMEM at a density of 1×105 cells/well. The lower chamber was filled with DMEM containing 20% FBS as a chemoattractant. After 24 h of incubation at 37°C, the non-invasive cells on the upper membrane surface were gently removed with a cotton tip, and the cells that passed through the filter were stained with hematoxylin at room temperature for 1 h and counted under an inverted light microscope (×400 magnification).

Plate colony formation assays

Colony formation ability of the glioblastoma cell lines was determined using plate colony formation assays. Briefly, U87 and U251 cells were cultured in DMEM containing 10% FBS in 6-well plates at a concentration of 1,000 cells/well. The cells were treated with MAP30 0.51 and 0.60 µM at 1/4 of half-maximal inhibitory concentration (IC50) for U87 and U251 cells at 48 h. or PBS. After 7 days, the colonies were washed gently with PBS, fixed with 4% paraformaldehyde at room temperature for 5 min and stained with Giemsa (Sigma; Merck KGaA, Darmstadt, Germany) at room temperature for 30 min; images were captured under an inverted light microscope (×400 magnification) and quantified.

Hoechst 33342/propidium iodide (PI) staining

U87 cells were treated for 24 h with MAP30 (1.4 and 2.7 µM) or PBS (0.01 M, pH 7.2–7.4), fixed with ice-cold 70% ethanol (4°C), washed three times with PBS, stained with Hoechst 33342/PI solution (50 µg/ml, Sigma-Aldrich; Merck KGaA) for 10 min and washed three times with PBS. The nuclear morphological changes were observed and images were captured using fluorescence microscopy (×400 magnification; EVOS-FL; Thermo Fisher Scientific, Inc.).

Cell cycle and apoptosis assays

U87 cells were cultured in 25 ml flasks for 24 h with MAP30 (1.4 and 2.7 µM) or PBS (0.01 M, pH 7.2–7.4). For cell cycle analysis, the cells were harvested, washed, fixed with ice-cold 70% ethanol at −20°C overnight, prior to washing with PBS and suspended in 300 µl staining buffer (BD Biosciences) containing 10 µl propidium iodide (PI) and 5 µl RNase (BD Biosciences) for 15 min at room temperature in the dark.

An annexin V-fluorescein isothiocyanate (FITC)/PI Apoptosis Detection kit (BD Biosciences) was used to determine the effects of MAP30 on apoptosis of U87 cells. The cells were harvested, washed twice with ice-cold PBS, and resuspended in binding buffer at a concentration of 1×106 cells/ml. A 500 µl volume of the solution was transferred into a 5-ml culture tube, and 5 µl annexin V-FITC and 5 µl PI were added to the solution with slight shaking, prior to incubation for 15 min at room temperature in the dark.

Samples prepared using the aforementioned methods were analyzed using flow cytometry (FC500 instrument; Beckman Coulter, Inc., Brea, CA, USA) within 1 h of staining.

Western blotting

U87 cells were incubated with MAP30 at 1.4 and 2.7 µM for 24 and 48 h, and total cell proteins were extracted with ice-cold lysis buffer (1 mM EDTA, pH 8.0, 50 mM Tris/HCl, pH 7.4, 150 mM NaCl, 1% NP-40, 0.1% SDS and 0.5% sodium deoxycholate, pH 7.4). After 30 min on ice, cell debris was removed by centrifugation at 14,000 × g for 20 min at 4°C. A basic protein quantification kit (BioVision, Inc., CA, USA) was used to determine total protein concentrations. Equal amounts (20 µg) of whole cell protein extracts from each sample were size-fractionated by 8% SDS-PAGE and transferred onto a polyvinyl difluoride membrane (BioVision, Inc., Milpitas, CA, USA). Membranes were first blocked in 5% non-fat milk powder (suspended in Tris-buffered saline with Tween-20; cat. no. AB64204) at room temperature for 2 h, and incubated with a specific primary antibody (using GAPDH as a control; cat no. AB181602; dilution 1:2,000; Abcam) overnight at 4°C. Following washing with PBS containing Tween-20, membranes were incubated with a horseradish peroxidase-conjugated secondary antibody (cat. no. AB6721; Abcam; dilution, 1:3,000) at room temperature for 2 h, and protein bands were developed with Amersham ECL Western Blotting Detection reagent (GE Healthcare Life Sciences, Little Chalfont, UK) and visualized using a gel imaging analysis system (Bio-Rad Laboratories, Inc., Hercules, CA, USA).

Statistical analysis

Results are expressed as the mean ± standard deviation of multiple independent experiments. Statistical analyses were performed using two-way analysis of variance with Student-Newman-Keuls post hoc test or Student's t-test using SPSS software (version 20.0; IBM Corp., Armonk, NY, USA). The IC50 value was estimated from CCK-8 dose-response data via linear regression and log transformation. P<0.05 was considered to indicate a statistically significant difference.

Results

MAP30 affects the viability of glioblastoma cells

After treat with MAP30 at IC50 concentration for 24 h, the morphological changes in the glioma cells were observed (Fig. 1A). Following treatment of U87 and U251 cells with different concentrations of MAP30 (0.5, 1, 2, 4, 8 and 16 µM) for 24–72 h, cell viability was measured by CCK-8. It was identified that MAP30 significantly inhibited the viability of U87 and U251 cells (Fig. 1B). The IC50 values for MAP30 were identified to be 2.7±0.22 and 3.9±0.18 µM at 24 h, 2.03±0.03 and 2.4±0.03 µM at 48 h, and 1.6±0.15 and 1.9±0.16 µM at 72 h for U87 and U251 cells, respectively (Fig. 1C and D). These results reveal a time- and dose-dependent effect of the action of MAP30.

Effects of MAP30 on cell migration and invasion

Wound healing assays and Transwell assays were used to determine the effects of MAP30 on the migration and invasion of glioblastoma cells. Fig. 2A presents the wound healing of U87 cells treated with MAP30 in comparison with the control. The wound healing rate was 37.3% for MAP30-treated U87 cells compared with 60% for the control cells, representing a significant decrease (P<0.05; Fig. 2B). Fig. 2C presents the wound healing of U251 cells treated with MAP30 in comparison with the control. The wound healing rate was 43.5% for MAP30-treated U251 cells compared with 70.5% for the control cells, representing a significant decrease (P<0.05; Fig. 2D). Fig. 2E presents images of the invading control and MAP30-treated U87 and U251 cells on the bottom of the membrane. The proportions of invading MAP30-treated U87 and U251 cells were respectively decreased to 34.7 and 41.3% compared with their control counterparts (P<0.05; Fig. 2F).

Plate colony formation assays

Compared with cells treated with PBS, application of MAP30 at 0.51 and 0.60 µM (1/4 of the IC50 for U87 and U251 cells, respectively, at 48 h) significantly suppressed colony formation of U87 and U251 cells. The number of colonies formed was 5 and 12 for MAP30-treated U87 and U251 cells, respectively, compared with 78 and 113 for control U87 and U251 cells, respectively, representing a significant decrease (P<0.05; Fig. 2G).

Cell apoptosis and cycle assays

Following Hoechst 33342/PI double staining, U87 and U251 cells treated with MAP30 exhibited typical apoptotic morphological changes including chromatic agglutination, karyopyknosis and nuclear fragmentation (Fig. 3A and B).

After 24 h of treatment with MAP30 at a dose of 1.4 µM, early apoptosis was increased to 13.87 and 16.77% in the U87 and U251 cell lines, respectively. The early apoptosis ratio increased to 49.61 and 51.17% in these two cell lines when the concentration of MAP30 was increased to 2.7 µM (Fig. 3C and D). The baseline early apoptotic proportions were 0.08 and 0.25%, respectively, for U87 and U251 cells treated with PBS (P<0.05; Fig. 3F).

Cell cycle assays indicated that MAP30 induced S-phase arrest in U87 cells. The proportions of S-phase U87 and U251 cells treated with MAP30 (1.4 and 2.7 µM) for 24 h significantly increased from 21.73% (PBS-treated) to 30.79 and 41.33%, respectively (P<0.05; Fig. 3E and G).

Western blot analysis

The western blotting results demonstrated that the expression of LGR5, pGSK-3βSer9, β-catenin, c-Myc and cyclin D1 decreased, and the expression of Smac markedly increased, as the dose (1.4–2.7 µM) and treated time (24–48 h) of MAP30 increased (Fig. 4; P<0.05).

Discussion

Previously, a promising anti-angiogenic therapy (bevacizumab) failed to improve overall survival rates in two independent Phase III trials (AVAglio and RTOG 0825), suggesting that gliomas are refractory to treatment (19,20). Therefore, development of novel and effective therapeutic agents is highly desirable. Although previous studies have identified that MAP30 possesses antitumor effects in various tumor cells, including glioblastoma cells, the underlying molecular mechanism of MAP30 on glioblastoma cells has not yet been elucidated (4,6,1214,16,21,22). The results of the present study demonstrated that MAP30 displayed significant inhibition of proliferation, invasion and migration on U87 and U251 cell lines in a dose- and time-dependent manner. The IC50 values for U87 cells were lower compared with those for U251 cells, indicating that U87 cells might be more sensitive to MAP30. Therefore, the U87 cell line was selected for additional experiments. The U87 cell line is the most representative glioma cell line. Although it was identified that the DNA profile of U87MG cells was different from that of the original cells in 2016, it is likely to be a bona fide human glioblastoma cell line of unknown origin (23). These cells are thought to exhibit the characteristics of human glioblastoma (2427), including proliferation, invasion and migration. The U87 cell line remains one of the most representative glioma cell lines. In addition, the U251 cell line was investigated to support the results from the U87 cells.

The flow cytometric and fluorescence staining results demonstrated that MAP30 induced apoptosis and arrested cell cycle in S-phase. The apoptosis-promoting and cell cycle-arresting abilities of MAP30 were identified in previous studies (14,2830). MAP30 was considered to cleave adenine-ribose glycosidic bonds by targeting the conserved host protein synthesis machinery, and prevent the elongation of DNA/RNA, resulting in DNA replication arrest in S-phase (3,16,17).

The apoptotic mechanism of MAP30 in glioma cells has not been clarified. LGR5, a downstream target gene of the Wnt signaling pathway, serves a key function in tumorigenesis, and the Wnt signaling pathway is vital in proliferation, differentiation and morphogenesis of gliomas (3136). Our previous studies revealed that LGR5 expression increased with the malignant degree of human gliomas, and knockdown of LGR5 was identified to lead to significant inhibition of the proliferation of glioma cells in vitro and in vivo (37). Additionally, Hsu et al (38) demonstrated that depletion of LGR5 induced apoptosis through the loss of mitochondrial membrane potential, and inhibited the activity of Wnt/β-catenin signaling by suppressing the expression of c-Myc and cyclin D in CRC cells (38). The western blotting results of the present study indicated that the expression of LGR5, pGSK-3βSer9, β-catenin, c-Myc and cyclin D1 were markedly decreased in U87 cells following MAP30 treatment in a dose- and time-dependent manner. Thus, we hypothesize that suppression of expression of LGR5 and the Wnt/β-catenin signaling pathway may be part of the mechanism by which MAP30 inhibits proliferation and induces apoptosis in glioma cells.

On the other hand, our results indicate that the expression of Smac is significantly enhanced in U87 cells in a dose- and time-dependent manner. Evasion of programmed cell death is a hallmark of human cancer (39). Smac is considered to be an important anticancer factor in the mitochondrial apoptosis pathway, which binds directly to and neutralizes inhibitor of apoptosis (IAP) proteins, thus leading to activation of the intrinsic pathway of apoptosis (3942). In addition, Smac mimetic-mediated depletion of IAP proteins may lead to ubiquitination, proteasomal degradation of substrates and activation of nuclear factor κB (NF-κB), a transcription factor with important functions in cell death and survival signaling, and all the functions of Smac promote apoptosis (39,41,43).

In addition, previous studies fused MAP30 with human-derived cell penetrating peptide or poly (ethylene glycol) to increase the uptake efficiency, cytotoxic activity and in vivo half-life and decrease immunogenicity of MAP30 (12,44). Even more importantly, RIPs may exhibit little or no detectable adverse effects on normal cells as they may recognize features on membranes dominant to tumor cells (4,17,4547). All these characteristics would broaden the number of applications of MAP30.

On the basis of the results of the present study, it is concluded that the mechanisms of MAP30 against malignant glioma cell lines may be associated with potent suppression of LGR5 and the Wnt/β-catenin signaling pathway, and enhancement of Smac expression in a dose- and time-dependent manner, indicating that MAP30 may be a novel and effective therapeutic natural plant agent for the treatment of gliomas. However, further study using in vivo models and elucidation of the underlying molecular mechanism of action of Smac are required to confirm these results.

Acknowledgements

The present study was supported by the Peking University People's Hospital Research and Development Funds (grant nos. 2104000004, RDB2011-14 and RDC2015-05) and by the National Natural Science Foundation of China (grant no. 81001009). The authors would like to thank Professor Meng (College of Life Sciences, Sichuan University, Chengdu, China) for providing the highly purified MAP30. The authors also thank Professor Xiangjun He, Dr Junjie Miao, Dr Dongliang Wang (Peking University People's Hospital, Beijing, China) and Dr Jiangfeng Liang (Peking University International Hospital, Beijing, China) for providing the laboratory and technical advice.

References

1 

Louis DN, Ohgaki H, Wiestler OD, Cavenee WK, Burger P, Jouvet A, Scheithauer BW and Kleihues P: WHO classification of tumours of the central nervous system. Acta Neuropathol. 114:97–109. 2007. View Article : Google Scholar : PubMed/NCBI

2 

Tchoghandjian A, Soubéran A, Tabouret E, Colin C, Denicolaï E, Jiguet-Jiglaire C, El-Battari A, Villard C, Baeza-Kallee N and Figarella-Branger D: Inhibitor of apoptosis protein expression in glioblastomas and their in vitro and in vivo targeting by SMAC mimetic GDC-0152. Cell Death Dis. 7:e23252016. View Article : Google Scholar : PubMed/NCBI

3 

Lee-Huang S and Huang PL, Nara PL, Chen HC, Kung HF, Huang P, Huang HI and Huang PL: MAP 30: A new inhibitor of HIV-1 infection and replication. FEBS Lett. 272:12–18. 1990. View Article : Google Scholar : PubMed/NCBI

4 

Fang EF, Zhang CZ, Ng TB, Wong JH, Pan WL, Ye XJ, Chan YS and Fong WP: Momordica Charantia lectin, a type II ribosome inactivating protein, exhibits antitumor activity toward human nasopharyngeal carcinoma cells in vitro and in vivo. Cancer Prev Res (Phila). 5:109–121. 2012. View Article : Google Scholar : PubMed/NCBI

5 

Fang EF and Ng TB: Can bitter gourd (Momordica Charantia) be a novel therapy for human cancers? Cancer Prev Res. 1–2. 2012.

6 

Fang EF and Ng TB: Bitter gourd (Momordica charantia) is a cornucopia of health: A review of its credited antidiabetic, anti-HIV and antitumor properties. Curr Mol Med. 11:417–436. 2011. View Article : Google Scholar : PubMed/NCBI

7 

Wang X, Sun W, Cao J, Qu H, Bi X and Zhao Y: Structures of new triterpenoids and cytotoxicity activities of the isolated major compounds from the fruit of Momordica charantia L. J Agric Food Chem. 60:3927–3933. 2012. View Article : Google Scholar : PubMed/NCBI

8 

Miao J, Jiang Y, Wang D, Zhou J, Fan C, Jiao F, Liu B, Zhang J, Wang Y and Zhang Q: Trichosanthin suppresses the proliferation of glioma cells by inhibiting LGR5 expression and the Wnt/β-catenin signaling pathway. Oncol Rep. 34:2845–2852. 2015. View Article : Google Scholar : PubMed/NCBI

9 

Grover JK and Yadav SP: Pharmacological actions and potential uses of Momordica charantia: A review. J Ethnopharmacol. 93:123–132. 2004. View Article : Google Scholar : PubMed/NCBI

10 

Dandawate PR, Subramaniam D, Padhye SB and Anant S: Bitter melon: A panacea for inflammation and cancer. Chin J Nat Med. 14:81–100. 2016.PubMed/NCBI

11 

Raina K, Kumar D and Agarwal R: Promise of bitter melon (Momordica charantia) bioactives in cancer prevention and therapy. Semin Cancer Biol 40–41. 1–129. 2016.

12 

Lv Q, Yang XZ, Fu LY, Lu YT, Lu YH, Zhao J and Wang FJ: Recombinant expression and purification of a MAP30-cell penetrating peptide fusion protein with higher anti-tumor bioactivity. Protein Expr Purif. 111:9–17. 2015. View Article : Google Scholar : PubMed/NCBI

13 

Fan X, He L and Meng Y, Li G, Li L and Meng Y: A-MMC and MAP30, two ribosome-inactivating proteins extracted from Momordica charantia, induce cell cycle arrest and apoptosis in A549 human lung carcinoma cells. Mol Med Rep. 11:3553–3558. 2015. View Article : Google Scholar : PubMed/NCBI

14 

Fang EF, Zhang CZ, Wong JH, Shen JY, Li CH and Ng TB: The MAP30 protein from bitter gourd (Momordica charantia) seeds promotes apoptosis in liver cancer cells in vitro and in vivo. Cancer Lett. 324:66–74. 2012. View Article : Google Scholar : PubMed/NCBI

15 

Fan JM, Luo J, Xu J, Zhu S, Zhang Q, Gao DF, Xu YB and Zhang GP: Effects of recombinant MAP30 on cell proliferation and apoptosis of human colorectal carcinoma LoVo cells. Mol Biotechnol. 39:79–86. 2008. View Article : Google Scholar : PubMed/NCBI

16 

Fang EF, Ng TB, Shaw PC and Wong RN: Recent progress in medicinal investigations on trichosanthin and other ribosome inactivating proteins from the plant genus Trichosanthes. Curr Med Chem. 18:4410–4417. 2011. View Article : Google Scholar : PubMed/NCBI

17 

Wang YX, Neamati N, Jacob J, Palmer I, Stahl SJ, Kaufman JD, Huang PL, Huang PL, Winslow HE, Pommier Y, et al: Solution structure of anti-HIV-1 and anti-tumor protein MAP30: Structural insights into its multiple functions. Cell. 99:433–442. 1999. View Article : Google Scholar : PubMed/NCBI

18 

Meng Y, Lin S, Liu S, Fan X, Li G and Meng Y: A novel method for simultaneous production of two ribosome-inactivating proteins, α-MMC and MAP30, from Momordica charantia L. PLoS One. 9:e1019982014. View Article : Google Scholar : PubMed/NCBI

19 

Chinot OL, Wick W, Mason W, Henriksson R, Saran F, Nishikawa R, Carpentier AF, Hoang-Xuan K, Kavan P, Cernea D, et al: Bevacizumab plus radiotherapy-temozolomide for newly diagnosed glioblastoma. N Engl J Med. 370:709–722. 2014. View Article : Google Scholar : PubMed/NCBI

20 

Wick W, Chinot OL, Bendszus M, Mason W, Henriksson R, Saran F, Nishikawa R, Revil C, Kerloeguen Y and Cloughesy T: Evaluation of pseudoprogression rates and tumor progression patterns in a phase III trial of bevacizumab plus radiotherapy/temozolomide for newly diagnosed glioblastoma. Neuro Oncol. 18:1434–1441. 2016. View Article : Google Scholar : PubMed/NCBI

21 

Hlin H, Zhi-Guo Z, Cong-Hui H, Yan Z, Qing L, Bo J, Hou-Guang H, Jun-Jie Z and Pei-Ying Z: Expression of Momordica charantia MAP30 and its anti-tumor effect on bladder cancer cells. Minerva Urol Nefrol. 68:275–281. 2014.PubMed/NCBI

22 

Meng Y, Liu S, Li J, Meng Y and Zhao X: Preparation of an antitumor and antivirus agent: chemical modification of α-MMC and MAP30 from Momordica Charantia L. with covalent conjugation of polyethyelene glycol. Int J Nanomedicine. 7:3133–3142. 2012.PubMed/NCBI

23 

Allen M, Bjerke M, Edlund H, Nelander S and Westermark B: Origin of the U87MG glioma cell line: Good news and bad news. Sci Transl Med. 8:354re32016. View Article : Google Scholar : PubMed/NCBI

24 

Bedini A, Baiula M, Vincelli G, Formaggio F, Lombardi S, Caprini M and Spampinato S: Nociceptin/orphanin FQ antagonizes lipopolysaccharide-stimulated proliferation, migration and inflammatory signaling in human glioblastoma U87 cells. Biochem Pharmacol. 140:89–104. 2017. View Article : Google Scholar : PubMed/NCBI

25 

Heckler M, Osterberg N, Guenzle J, Thiede-Stan NK, Reichardt W, Weidensteiner C, Saavedra JE and Weyerbrock A: The nitric oxide donor JS-K sensitizes U87 glioma cells to repetitive irradiation. Tumour Biol. 39:10104283177039222017. View Article : Google Scholar : PubMed/NCBI

26 

Alphandéry E, Idbaih A, Adam C, Delattre JY, Schmitt C, Guyot F and Chebbi I: Development of non-pyrogenic magnetosome minerals coated with poly-l-lysine leading to full disappearance of intracranial U87-Luc glioblastoma in 100% of treated mice using magnetic hyperthermia. Biomaterials. 141:210–222. 2017. View Article : Google Scholar : PubMed/NCBI

27 

Alphandéry E, Idbaih A, Adam C, Delattre JY, Schmitt C, Guyot F and Chebbi I: Chains of magnetosomes with controlled endotoxin release and partial tumor occupation induce full destruction of intracranial U87-Luc glioma in mice under the application of an alternating magnetic field. J Control Release. 262:259–272. 2017. View Article : Google Scholar : PubMed/NCBI

28 

Ru P, Steele R, Nerurkar PV, Phillips N and Ray RB: Bitter melon extract impairs prostate cancer cell-cycle progression and delays prostatic intraepithelial neoplasia in TRAMP model. Cancer Prev Res (Phila). 4:2122–2130. 2011. View Article : Google Scholar : PubMed/NCBI

29 

Brennan VC, Wang CM and Yang WH: Bitter melon (Momordica charantia) extract suppresses adrenocortical cancer cell proliferation through modulation of the apoptotic pathway, steroidogenesis and insulin-like growth factor type 1 receptor/RAC-alpha serine/threonine-protein kinase signaling. J Med Food. 15:325–334. 2012. View Article : Google Scholar : PubMed/NCBI

30 

Ray RB, Raychoudhuri A, Steele R and Nerurkar P: Bitter melon (Momordica charantia) extract inhibits breast cancer cell proliferation by modulating cell cycle regulatory genes and promotes apoptosis. Cancer Res. 70:1925–1931. 2010. View Article : Google Scholar : PubMed/NCBI

31 

Rot S, Taubert H, Bache M, Greither T, Würl P, Eckert AW, Schubert J, Vordermark D and Kappler M: A novel splice variant of the stem cell marker LGR5/GPR49 is correlated with the risk of tumor-related death in soft-tissue sarcoma patients. BMC Cancer. 11:4292011. View Article : Google Scholar : PubMed/NCBI

32 

Yi GZ, Liu YW, Xiang W, Wang H, Chen ZY, Xie SD and Qi ST: Akt and β-catenin contribute to TMZ resistance and EMT of MGMT negative malignant glioma cell line. J Neurol Sci. 367:101–106. 2016. View Article : Google Scholar : PubMed/NCBI

33 

Morris SL and Huang S: Crosstalk of the Wnt/β-catenin pathway with other pathways in cancer cells. Genes Dis. 3:41–47. 2016. View Article : Google Scholar : PubMed/NCBI

34 

Barker N and Clevers H: Leucine-rich repeat-containing G-protein-coupled receptors as markers of adult stem cells. Gastroenterology. 138:1681–1696. 2010. View Article : Google Scholar : PubMed/NCBI

35 

Barker N, van Es JH, Kuipers J, Kujala P, van den Born M, Cozijnsen M, Haegebarth A, Korving J, Begthel H, Peters PJ and Clevers H: Identification of stem cells in small intestine and colon by marker gene Lgr5. Nature. 449:1003–1007. 2007. View Article : Google Scholar : PubMed/NCBI

36 

Hsu SY, Kudo M, Chen T, Nakabayashi K, Bhalla A, van der Spek PJ, van Duin M and Hsueh AJ: The three subfamilies of leucine-rich repeat-containing G protein-coupled receptors (LGR): Identification of LGR6 and LGR7 and the signaling mechanism for LGR7. Mol Endocrinol. 14:1257–1271. 2000. View Article : Google Scholar : PubMed/NCBI

37 

Wang D, Zhou J, Fan C, Jiao F, Liu B, Sun P, Miao J and Zhang Q: Knockdown of LGR5 suppresses the proliferation of glioma cells in vitro and in vivo. Oncol Rep. 31:41–49. 2014. View Article : Google Scholar : PubMed/NCBI

38 

Hsu HC, Liu YS, Tseng KC, Tan BC, Chen SJ and Chen HC: LGR5 regulates survival through mitochondria-mediated apoptosis and by targeting the Wnt/β-catenin signaling pathway in colorectal cancer cells. Cell Signal. 26:2333–2342. 2014. View Article : Google Scholar : PubMed/NCBI

39 

Fulda S: Promises and challenges of smac mimetics as cancer therapeutics. Clin Cancer Res. 21:5030–5036. 2015. View Article : Google Scholar : PubMed/NCBI

40 

Marschall V and Fulda S: Smac mimetic-induced upregulation of interferon-β sensitizes glioblastoma to temozolomide-induced cell death. Cell Death Dis. 6:e18882015. View Article : Google Scholar : PubMed/NCBI

41 

Mizukawa K, Kawamura A, Sasayama T, Tanaka K, Kamei M, Sasaki M and Kohmura E: Synthetic Smac peptide enhances the effect of etoposide-induced apoptosis in human glioblastoma cell lines. J Neurooncol. 77:247–255. 2006. View Article : Google Scholar : PubMed/NCBI

42 

Amendola D, De Salvo M, Marchese R, Verga Falzacappa C, Stigliano A, Carico E, Brunetti E, Moscarini M and Bucci B: Myc down-regulation affects cyclin D1/cdk4 activity and induces apoptosis via Smac/Diablo pathway in an astrocytoma cell line. Cell Prolif. 42:94–109. 2009. View Article : Google Scholar : PubMed/NCBI

43 

Scheidereit C: IkappaB kinase complexes: Gateways to NF-kappaB activation and transcription. Oncogene. 25:6685–6705. 2006. View Article : Google Scholar : PubMed/NCBI

44 

Sun Y, Sun F, Li J, Wu M, Fan X and Meng Y and Meng Y: Mono-PEGylation of Alpha-MMC and MAP30 from Momordica charantia L: Production, identification and anti-tumor activity. Molecules. 21:E14572016. View Article : Google Scholar : PubMed/NCBI

45 

Huang PL, Sun Y, Chen HC, Kung HF and Lee-Huang S: Proteolytic fragments of anti-HIV and anti-tumor proteins MAP30 and GAP31 are biologically active. Biochem Biophys Res Commun. 262:615–623. 1999. View Article : Google Scholar : PubMed/NCBI

46 

Schreiber CA, Wan L, Sun Y, Lu L, Krey LC and Lee-Huang S: The antiviral agents, MAP30 and GAP31, are not toxic to human spermatozoa and may be useful in preventing the sexual transmission of human immunodeficiency virus type 1. Fertil Steril. 72:686–690. 1999. View Article : Google Scholar : PubMed/NCBI

47 

Putnam CD and Tainer JA: The food of sweet and bitter fancy. Nat Struct Biol. 7:17–18. 2000. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

April-2018
Volume 15 Issue 4

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Jiang Y, Miao J, Wang D, Zhou J, Liu B, Jiao F, Liang J, Wang Y, Fan C, Zhang Q, Zhang Q, et al: MAP30 promotes apoptosis of U251 and U87 cells by suppressing the LGR5 and Wnt/β-catenin signaling pathway, and enhancing Smac expression. Oncol Lett 15: 5833-5840, 2018
APA
Jiang, Y., Miao, J., Wang, D., Zhou, J., Liu, B., Jiao, F. ... Zhang, Q. (2018). MAP30 promotes apoptosis of U251 and U87 cells by suppressing the LGR5 and Wnt/β-catenin signaling pathway, and enhancing Smac expression. Oncology Letters, 15, 5833-5840. https://doi.org/10.3892/ol.2018.8073
MLA
Jiang, Y., Miao, J., Wang, D., Zhou, J., Liu, B., Jiao, F., Liang, J., Wang, Y., Fan, C., Zhang, Q."MAP30 promotes apoptosis of U251 and U87 cells by suppressing the LGR5 and Wnt/β-catenin signaling pathway, and enhancing Smac expression". Oncology Letters 15.4 (2018): 5833-5840.
Chicago
Jiang, Y., Miao, J., Wang, D., Zhou, J., Liu, B., Jiao, F., Liang, J., Wang, Y., Fan, C., Zhang, Q."MAP30 promotes apoptosis of U251 and U87 cells by suppressing the LGR5 and Wnt/β-catenin signaling pathway, and enhancing Smac expression". Oncology Letters 15, no. 4 (2018): 5833-5840. https://doi.org/10.3892/ol.2018.8073