Open Access

Corilagin induces human glioblastoma U251 cell apoptosis by impeding activity of (immuno)proteasome

  • Authors:
    • Xianyun Qin
    • Jilan Liu
    • Dongfeng Pan
    • Wenyuan Ma
    • Panpan Cheng
    • Feng Jin
  • View Affiliations

  • Published online on: February 19, 2021     https://doi.org/10.3892/or.2021.7985
  • Article Number: 34
  • Copyright: © Qin et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Glioma is a type of common primary intracranial tumor, which is difficult to treat. It has been confirmed by research that corilagin (the primary active constituent of the matsumura leafflower herb) has significant antitumor effect. In particular, our previous research demonstrated that corilagin effectively promotes apoptosis of glioma U251 cells and has a synergistic effect when used with temozolomide. However, the mechanism by which corilagin causes apoptosis in U251 cells has yet to be investigated. Proteasomes are catalytic centers of the ubiquitin‑proteasome system, which is the major protein degradation pathway in eukaryotic cells; they are primarily responsible for the degradation of signal molecules, tumor suppressors, cyclins and apoptosis inhibitors and serve an important role in tumor cell proliferation and apoptosis. The present study investigated the pro‑apoptotic effect of corilagin on glioma U251 cells and confirmed that decreased proteasome activity and expression levels serve an important role in corilagin‑induced U251 cell apoptosis.

Introduction

Glioma the most common type of primary brain tumor, accounts for >70% of intracranial malignant tumors. According to histopathological characteristics, World Health Organization classifies glioma into four grades (1). Glioblastoma multiforme (GBM) belongs to grade IV and is the most malignant glioma. The prognosis of GBM is poor: In Switzerland and the USA, the 5-year survival rate of patients with glioblastoma is only 2.9% (2). GBM is known for its resistance to treatments such as temozolomide (TMZ), the most commonly used chemotherapy drug to treat glioma (3,4). This makes the development of novel drugs for treating glioma essential.

Corilagin is the primary active constituent of the matsumura leafflower herb (a Phyllanthus plant from the Euphorbiaceae family), which has potential for various biological activities, including antitumor, anti-inflammatory, and hepatoprotective activity (5). Corilagin inhibits malignant tumor cell proliferation and promotes tumor cell apoptosis (6). Both in vitro and in vivo tests have demonstrated that corilagin exerts a significant inhibitory effect on ovarian and colon cancer, hepatocellular (7) and esophageal squamous cell carcinoma (8) and other types of tumor cell. Corilagin inhibits breast cancer growth via reactive oxygen species (ROS)-dependent apoptosis and autophagy (9). Moreover, it induces apoptosis, autophagy and ROS generation in gastric cancer cells in vitro. (10). Our previous research (11) demonstrated that corilagin effectively inhibits the proliferation of U251 and stem-like cells. It is reported that corilagin also affects TMZ-resistant T98G glioma cells (12). Taken together, these findings suggest the potential use of corilagin in glioma treatment. However, the precise mechanism by which corilagin promotes U251 cell apoptosis warrants further research.

A key feature of glioma, similar to that of other advanced tumors, is increased protein synthesis and degradation (13). This increase is due to the role of proteasomes in the degradation of signaling molecules, tumor suppressors, cyclin and apoptosis inhibitors; proteasomes are thus associated with glioma occurrence and development. The 26S proteasome is the primary proteolytic mechanism that regulates protein degradation in eukaryotic cells. The 26S proteasome comprises 20S catalytic and 19S regulatory particles. The 20S proteasome is a cylindrical structure comprising α and β subunits arranged in four stacked heteroheptamer rings (14). The two β-rings face three active sites of the β1, β2, and β5 subunits, which face the core particle (15). When stimulated by inflammatory cytokines, such as IFN-γ, immune cells, express large multifunctional peptidase (LMP)2 (β1i), multicatalytic endopeptidase complex-like-1 (MECL-1; β2i) and LMP7 (β5i). These three inducible subunits, β1i, β2i,and β5i, replace the constitutive subunits β1, β2, and β5 to form immunoproteasomes (16). Moreover, β1/β1i, β2/β2i, and β5/β5i are responsible for caspase-, trypsin- and chymotrypsin-like activity, respectively (17). Studies (1820) have shown that β1i also exhibits a certain chymotrypsin-like activity. However, the inhibitory effect of corilagin-induced U251 cell apoptosis mediated by proteasome has yet to be investigated. The present study investigated the role of proteasomes in corilagin-induced U251 cell apoptosis.

Materials and methods

Chemicals and reagents

Corilagin analytical standard (purity, >99%) was purchased from Sigma-Aldrich (Merck KGaA) and TMZ from Selleck Chemicals. Anti-β1, anti-β2, anti-β5, anti-β1i, anti-β2i and anti-β5i antibodies were procured from Abcam; anti-β-actin and horseradish peroxidase (HRP)-conjugated goat anti-mouse and anti-rabbit IgG secondary antibodies were acquired from OriGene Technologies, Inc.; MG-132, bortezomib, carfizomib and PR-957 were procured from Selleck Chemicals. DMEM, streptomycin penicillin, and fetal calf serum were obtained from HyClone (GE Healthcare Life Sciences); trypsin was obtained from Gibco (Thermo Fisher Scientific, Inc.). BCA protein assay kit was purchased from Beyotime Institute of Biotechnology, an Annexin V/PI apoptosis detection kit from BD Biosciences and proteasome activity detection kit from Promega Corporation.

U251 cell line culture

U251 cells ware cultured at 37°C in 5% CO2 in high glucose DMEM and 10% fetal bovine serum supplemented with penicillin (100 U/ml) and streptomycin (100 µg/ml). In order to evaluate proteasome involvement in corilagin-induced cell differentiation, cells were treated with proteasome inhibitors MG-132 (0, 200 nM, 1, 5 µM) and bortezomib (0, 5, 40, 100 nM) for 72 h and carfizomib (0, 100, 200 nM, 1 µM) and PR-957 (0, 100 nM, 1, 10 µM) for 24 h at 37°C. In order to prove whether corilagin increases TMZ-induced apoptosis of U251 cells, flow cytometry was performed using U251 cells treated with increasing concentrations of corilagin (0, 25, 50, 100 and 200 µg/ml) and/or TMZ (0, 25, 50, 100 and 200 µg/ml) for 72 h at 37°C. In order to demonstrate that proteasome inhibition mediated corilagin-induced apoptosis, flow cytometry was performed using U251 cells treated with corilagin + MG-132 (200 nM) and corilagin + bortezomib (5 nM) for 72 h at 37°C.

Cell Counting Kit (CCK)-8 assay

A total of 100 µl cell suspension (5,000 cells/well) was inoculated in a 96-well plate, which was placed in a 37°C cell incubator for 24 h. Next, 10 µl corilagin at various concentrations (0, 25, 50 and 100 µg/ml) was added to the wells. After 24, 48 and 72 h of incubation at 37°C, 10 µl CCK-8 reagent (Beyotime Institute of Biotechnology) was added to each well, followed by incubation at 37°C for 2 h. The developed color was measured by determining the absorbance at 450 nm wavelength (A). Finally, the cell survival rate (%) was calculated as [(Aexperimental well-Ablank well)/(Acontrol well-Ablank well)] × 100%.

Clonogenic survival assay

U251 cells were treated with corilagin (0, 25, 50 and 100 µg/ml) for 48 h at 37°C. The cells in the logarithmic growth phase were inoculated at a density of 500–1,000 cells/well in a 6-well plate in 10% fetal bovine serum complete DMEM and incubated for 7–14 days at 37°C. For each concentration, three auxiliary wells were used. Every 3 days, cell growth was monitored and the medium was changed. When a single clone grew to a visible size, the medium was removed and cells were washed twice with phosphate-buffered saline and fixed with methanol for 20 min at room temperature. After the methanol was removed, the cells were stained with 0.2% crystal violet for 30 min at room temperature and images were captured with an inverted light microscope (Olympus IX71) at ×100 magnification.

Apoptosis by flow cytometry

U251 cells were incubated with different concentrations (0, 25, 50, 100 and 200 µg/ml) of corilagin for 24, 48, 72 and 96 h at 37°C. cells were collected and washed with phosphate-buffered saline three times. For the detection of apoptosis, cells were stained using a Annexin V/PI Apoptosis Detection kit according to the manufacturer's instructions. Samples were analyzed by flow cytometry (BD FACSAria; BD Biosciences). The data were analyzed by FlowJo 7.6.1 software (FlowJo LLC).

Western blot analysis

Corilagin (0, 25, 50 and 100 µg/ml) was added to U251 cells (30–40% density) at 37°C which were inoculated in 6-well plates. After 48 h, proteins were extracted. Protein concentration was determined using BCA protein assay kit. U251 cells were collected and lysed using RIPA cell lysis buffer (Invitrogen; Thermo Fisher Scientific, Inc.). Following two washes with PBS, cells were lysed with lysis buffer [20 mM Tris-HCl (pH, 7.5), 150 mM NaCl, 1 mM EDTA, 1% NP-40] containing Complete Protease Inhibitor Cocktail (Roche Applied Science). The cell lysates (40 µg) were separated by 12% SDS-PAGE and transferred onto a PVDF membrane. Membranes were blocked with 5% skimmed milk for 1 h at room temperature and incubated with primary antibodies at a dilution of 1:1,000 overnight at 4°C. Antibodies against the following proteins were used: β1, β2, β5, β1i, β2i, β5i and β-actin. Luminata Forte Western HRP Substrate (EMD Millipore) was used for the development of positive signals. The relative protein expression of genes was quantified by ImageJ v.1.46r software (National Institutes of Health) using β-actin as a control.

Assay for proteasome activity

The 26S proteasome activity was analyzed based on a previously published method of Promega Corporation proteasome activity assay kit (21). Cells were harvested and 10,000 cells in high glucose complete DMEM were incubated with detection reagents containing fluorogenic substrates of caspase-, trypsin- and chymotrypsin-like activity at 37°C for 1 h. The caspase-like activity was determined with Z-LLEC (45 µM), trypsin-like activity with Ac-RLR-AMC (40 µM) and chymotrypsin-like activity with Suc-LLVY-AMC (18 µM) at 37°C for 1 h. The fluorescence intensity was measured at the following wavelengths: Excitation, 380 and emission, 460 nm.

Statistical analysis

Data were analyzed by one-way ANOVA followed by Tukey's multiple comparisons test by GraphPad Prism 8.0.2 (GraphPad Software, Inc.). Data are presented as the mean ± SEM (≥3). P<0.05 was considered to indicate a statistically significant difference.

Results

Corilagin inhibits U251 proliferation

In order to determine the role of corilagin in regulating U251 growth, CCK-8 assay was performed to detect U251 cell survival following exposure to increasing corilagin concentrations (0, 25, 50, 100 and 200 µg/ml) for 24, 48 and 72 h. The results indicated that all concentrations of corilagin significantly decreased survival over 72 h, and 200 µg/ml corilagin at 24 and 48 h also significantly decreased the survival (Fig. 1A). Clone formation experiments on U251 cells treated with varying concentrations of corilagin for 48 h also demonstrated that the number of cell clones decreased in a dose-dependent manner (0, 25, 50, 100 and 200 µg/ml; Fig. 1B). Taken together, these data suggested that corilagin inhibited U251 cell proliferation.

Corilagin promotes U251 apoptosis

In order to determine the role of corilagin in regulating U251 apoptosis, apoptosis of U251 cells was assessed using an Annexin V/PI staining kit. The results revealed that U251 cell apoptosis rates significantly increased following corilagin treatment (0, 25, 50, 100 and 200 µg/ml) for 24, 48 and 72 h (Fig. 2A). Following treatment with 200 µg/ml corilagin treatment for 24, 48, and 72 h, the ratio of apoptotic cells significantly increased by 1.8-, 2.7- and 4.4-fold, respectively (Fig. 2B-D). Additionally, 50 µg/ml corilagin at 24 h and 100 µg/ml at all three time points significantly increased the ratio of apoptotic cells. Taken together, these results suggested that corilagin effectively promoted U251 apoptosis.

Corilagin combined with TMZ increases apoptosis of U251 cells

In order to determine the role of corilagin + TMZ in regulating U251 apoptosis, flow cytometry was performed. Images of Annexin V/PI double staining of U251 cells treated with increasing concentrations (0, 25, 50, 100 and 200 µg/ml) of corilagin, TMZ and corilagin + TMZ for 72 h are shown in Fig. 3A-C. The percentage of apoptotic cells (Annexin V+/PI+ and Annexin V+/PI) treated with corilagin, TMZ and corilagin + TMZ was calculated (Fig. 3D-F). At 100 µg/ml corilagin + TMZ increased the apoptosis rate from 58% in the TMZ-alone group to 94% Furthermore, 200 µg/ml corilagin + TMZ increased the apoptosis rate from 62% in the TMZ-alone group to 90% and 50 µg/ml corilagin + TMZ increased the apoptosis rate from 17 to 40%. However, 50 µg/ml corilagin group did not significantly increase the apoptosis rate compared with 0 µg/ml corilagin (Fig. 3E and F). Taken together, these results suggested that corilagin + TMZ increased apoptosis of U251 cells.

Corilagin decreases proteasome activity

In order to determine the role of proteasome in corilagin-induced U251 cell apoptosis, the effect of corilagin on proteasome activity was investigated. U251 cells were treated with corilagin at different concentrations (0, 25, 50 and 100 µg/ml) and durations (4, 8, 24 and 48 h). The proteasome activity, including caspase-, trypsin- and chymotrypsin-like activity, was measured using fluorescent-labeled peptides. At 4 h, corilagin treatment significantly increased caspase-, trypsin- and chymotrypsin-like activity in a dose-dependent manner (Fig. 4A). At 8 h, chymotrypsin-like activity was significantly increased by 100 µg/ml corilagin (Fig. 4B). At 24 and 48 h, caspase-, trypsin- and chymotrypsin-like activities significantly decreased in a dose-dependent manner (Fig. 4C and D). These results suggested that corilagin-induced apoptosis was associated with proteasome activity.

Corilagin decreases expression levels of proteasome subunits

In order to determine the effect of corilagin on proteasome catalytic subunit expression levels, U251 cells were treated with corilagin (100 µg/ml) for different durations (4, 8 and 24 h). The protein expression levels of the constitutive (i.e., β1, β2, and β5; Fig. 5A) and inducible proteasome catalytic subunits (i.e., β1i, β2i and β5i; Fig. 5B) were detected by western blotting. Corilagin treatment significantly decreased the protein levels of constitutive subunits (β1 and β2) and immunosubunits (β1i, β2i and β5i) but increased those of the constitutive subunit β5 (Fig. 5C and D), indicating that the corilagin-induced decrease in proteasome activity in U251 cells was primarily mediated by decreased expression levels of proteasome subunits β1/β1i, β2i and β5i.

Proteasome inhibitors promote U251 apoptosis

In order to determine the role of proteasome in corilagin-mediated regulation of U251 apoptosis, the apoptotic rate of U251 cells treated with proteasome inhibitors MG-132 (0, 200 nM, 1, 5 µM; Fig. 6B), bortezomib (0, 5, 40, 100 nM; Fig. 6E) for 72 h and carfizomib (0, 100, 200 nM, 1 µM; Fig. 6H) and PR-957 (0, 100, 1, 10 µM; Fig. 6K) for 24 h was assessed using an Annexin V/PI staining kit. Before detecting apoptosis, the effects of proteasome inhibitors on the activity of three proteasome enzymes was assessed. MG-132 and bortezomib significantly inhibited caspase- and chymotrypsin-like activity; lower concentrations of bortezomib were needed to achieve this inhibitory effect (Fig. 6A and D). Carfilzomib and PR-957 significantly inhibited chymotrypsin-like enzyme activity and carfilzomib also significantly inhibited caspase-like enzyme activity at a concentration of 1 µM (Fig. 6G and J). The apoptosis assay results showed that all inhibitors increased the percentage of apoptotic cells in a dose-dependent manner (Fig. 6C, F, I and L).

Proteasome inhibitors increase corilagin-induced apoptosis of U251 cells

In order to demonstrate that proteasome inhibition mediated corilagin-induced apoptosis, flow cytometry was performed using U251 cells treated with corilagin + MG-132 (200 nM) and corilagin + bortezomib (5 nM). Images of Annexin V/PI double staining of U251 cells treated with increasing concentrations (0, 25, 50, 100 and 200 µg/ml) of corilagin, corilagin + MG-132 (200 nM), and corilagin + bortezomib (5 nM) for 72 h are shown in Fig. 7A-C. The percentages of apoptotic cells (Annexin V+/PI+ and Annexin V+/PI) of cells treated with corilagin, corilagin + MG-132 and corilagin + bortezomib were calculated (Fig. 7D-F). The results showed that corilagin + MG-132 (200 nM) increased the percentage of apoptotic cells compared with corilagin-alone (50, 100 and 200 ug/ml) from 13, 16 and 27 to 17, 23 and 34%, respectively (Fig. 7E). Corilagin + bortezomib (5 nM) increased the percentage of apoptotic cells to 12, 34 and 40% (Fig. 7F). These results indicated that proteasome inhibitors increased corilagin-induced apoptosis.

Discussion

The present study investigated the role of proteasome activation in corilagin-induced U251 cell apoptosis. Corilagin primarily stimulated U251 cell apoptosis and decreased the activity and expression levels of proteasome subunits, including immunosubunits. U251 cells were treated with proteasome inhibitors, which were shown to promote apoptosis in these cells. In order to demonstrate that proteasome inhibition mediates corilagin-induced apoptosis, U251 cells were treated with corilagin in combination with the proteasome inhibitors MG-132 and bortezomib. The results of Annexin V/PI staining analysis for corilagin, corilagin + MG-132 and corilagin + bortezomib indicated that proteasome inhibitors increased corilagin-induced apoptosis. The present study therefore identified a novel potential mechanism for proteasomes in corilagin-induced U251 cell apoptosis.

The present study demonstrated that corilagin effectively promoted the apoptosis of glioma U251 cells. Research has indicated that corilagin inhibits the growth of U251 and TMZ-resistant T98G glioma cells (12). Qiu et al (8) determined that corilagin downregulates the E3 ubiquitin ligase RING finger protein 8 in the ubiquitin-proteasome pathway, which disrupts the DNA damage repair response and promotes cell death in esophageal squamous cell cancer cells.

During corilagin-induced U251 cell apoptosis, the proteasome catalytic subunit expression levels were all downregulated in the present study, except for those of β5. However, proteasome activity first increased and then decreased, particularly at 4 h, at which time proteasome activity was notably increased. This appears to contradict the decreased expression levels. The proteasome is a biological complex composed of multiple molecules, the assembly of which is complex and strictly regulated by various mechanisms, such as the assembly of proteasome activator 200 (22) and Adc17, the absence of which aggravates proteasome defects (23). The levels of proteasome are affected by proteasome subunits and assembly chaperones (17). Proteasome activity is also regulated by phosphorylation of 26S subunits (24) and by protein kinases that have been reported to stimulate proteasomal activity (25). For example, increased cAMP levels and protein kinase A activation cause phosphorylation of the 19S subunit Rpn6 (26), which increases peptide, ATP and Ubiquitin conjugate hydrolysis rates (27). The inconsistency between changes in proteasome activity and catalytic subunit expression levels require further analysis. It was hypothesized that in the early stage (4 h) of corilagin intervention in cells, proteasome subunit expression levels were affected and proteasome assembly was altered, but the proteasome activity was stimulated in response to external stress. After 8 h, proteasome function became increasingly damaged, causing proteasome activity to continue to decline, resulting in decreased physiological cell activity, finally leading to apoptosis.

In glioma cells, the expression levels of immunosubunits are high (28), but their mechanism of involvement in glioma occurrence and development remains unclear. Studies have revealed that immunoproteasomes regulate T helper cell differentiation (29,30). Several factors, including IFN-γ and TNF-α, promote catalytic subunit expression to produce immunoproteasomes and thereby affect the enzymatic activity of their substrates (17) and alter cell function (31). Notably, in the present study, β5 expression was upregulated and β5i expression was downregulated during corilagin intervention. It was hypothesized that corilagin changed the proteasome structure. The assembly of proteasome subunits is complex and strictly regulated; it is achieved by the controlled expression of proteasome subunits by a common transcription factor (such as Rpn4) in yeast (32). Cells adjust proteasome-mediated degradation by regulating proteasome levels via coordinated expression of proteasome subunits and assembly chaperones (17).

In order to demonstrate that corilagin promoted apoptosis by interfering with proteasome activity, U251 cells were treated with four proteasome inhibitors. Proteasome inhibitors inhibited proteasome activity and promoted apoptosis of U251 cells. Proteasome inhibitors are widely used to study the function of proteasomes and are employed as anticancer drugs to treat various types of cancer (18,33,34). The following proteasome inhibitors were used in the present study: MG-132, a reversible inhibitor of β1, β2, and β5; bortezomib, a reversible inhibitor of β5, β5i and β1i; carfilzomib, an irreversible inhibitor of β5 and β5i and PR-957, a specific irreversible inhibitor of β5i (35,36). Yoo et al (37) reported that glioma stem cells are sensitive to proteasome inhibitors. MG-132 has also been used in glioma research (38,39). Both bortezomib and carfilzomib have been approved for the treatment of multiple myeloma (40) and their use has also been reported in glioma research (40,41). The International Journal of Radiation Oncology, Biology, Physics published a phase-II clinical trial in 2018 (42) that evaluated the efficacy and safety of bortezomib in combination with TMZ and local radiotherapy for GBM. Bortezomib inhibits cell adhesion, angiogenesis and cytokine-mediated intercellular communication, thereby affecting the tumor microenvironment (33). Bortezomib and TMZ in combination with local radiotherapy is particularly beneficial for people with O-6-methylguanine-DNA methyltransferase methylation and its side effects (such as lymphopenia, neutropenia and thrombocytopenia) are generally mild (42). PR-957, which specifically inhibits β5i activity (35), is used to study autoimmune (43) and inflammatory disease (36). To the best of our knowledge, however, it has not been used in the study of glioma treatment. The present study demonstrated that PR-957 effectively promoted U251 cell apoptosis and its mechanism of action warrants further investigation. Given the inhibitory effect of proteasome inhibitors on glioma, future studies should investigate whether corilagin can be combined with proteasome inhibitors to improve its therapeutic effects on glioma.

Acknowledgements

Not applicable.

Funding

The present study was supported by Project of Scientific Developmental Program of Shandong Provincial Administration of Traditional Chinese Medicine (grant no. 2019-0479), Project of Health and Family Planning Commission of Shandong Province (grant nos. 2019WS362 and 2019WS361), Teacher Support Fund of Jining Medical University (grant no. JYFC2018FKJ107), General Project of Jining Science and Technology Bureau (grant no. 2016-56-60), Scientific Research Project of Jining Medical University (grant no. JY2015KJ022) and Nursery Research Program of Affiliated Hospital of Jining Medical University (grant nos. MP-2015-003 and MP-2018-012).

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

XQ and FJ designed the experiments. JL and WM performed western blot analysis, activity assays and cell experiments. PC performed flow cytometry. XQ and DP analyzed data and wrote the manuscript. XQ and FJ authenticated all the raw data. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Gusyatiner O and Hegi ME: Glioma epigenetics: From subclassification to novel treatment options. Semin Cancer Biol. 51:50–58. 2018. View Article : Google Scholar : PubMed/NCBI

2 

Ohgaki H and Kleihues P: Epidemiology and etiology of gliomas. Acta Neuropathol. 109:93–108. 2005. View Article : Google Scholar : PubMed/NCBI

3 

Stupp R, Mason WP, van den Bent MJ, Weller M, Fisher B, Taphoorn MJ, Belanger K, Brandes AA, Marosi C, Bogdahn U, et al: Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med. 352:987–996. 2005. View Article : Google Scholar : PubMed/NCBI

4 

Yang Q, Zhou Y, Chen J, Huang N, Wang Z and Cheng Y: Gene therapy for drug-resistant glioblastoma via lipid-polymer hybrid nanoparticles combined with focused ultrasound. Int J Nanomedicine. 16:185–199. 2021. View Article : Google Scholar : PubMed/NCBI

5 

Li X, Deng Y, Zheng Z, Huang W, Chen L, Tong Q and Ming Y: Corilagin, a promising medicinal herbal agent. Biomed Pharmacother. 99:43–50. 2018. View Article : Google Scholar : PubMed/NCBI

6 

Wu C, Huang H, Choi HY, Ma Y, Zhou T, Peng Y, Pang K, Shu G and Yang X: Anti-esophageal cancer effect of corilagin extracted from phmllanthi fructus via the mitochondrial and endoplasmic reticulum stress pathways. J Ethnopharmacol. 269:1137002021. View Article : Google Scholar : PubMed/NCBI

7 

Wan LF, Shen JJ, Wang YH, Zhao W, Fang NY, Yuan X and Xue BY: Extracts of Qizhu decoction inhibit hepatitis and hepatocellular carcinoma in vitro and in C57BL/6 mice by suppressing NF-κB signaling. Sci Rep. 9:14152019. View Article : Google Scholar : PubMed/NCBI

8 

Qiu F, Liu L, Lin Y, Yang Z and Qiu F: Corilagin inhibits esophageal squamous cell carcinoma by inducing DNA damage and down-regulation of RNF8. Anticancer Agents Med Chem. 19:1021–1028. 2019. View Article : Google Scholar : PubMed/NCBI

9 

Tong Y, Zhang G, Li Y, Xu J, Yuan J, Zhang B, Hu T and Song G: Corilagin inhibits breast cancer growth via reactive oxygen species-dependent apoptosis and autophagy. J Cell Mol Med. 22:3795–3807. 2018. View Article : Google Scholar

10 

Iweala EEJ, Xu J, Zhang G, Tong Y, Yuan J, Li Y and Song G: Corilagin induces apoptosis, autophagy and ROS generation in gastric cancer cells in vitro. Int J Mol Med. 43:967–979. 2019.PubMed/NCBI

11 

Yang WT, Li GH, Li ZY, Feng S, Liu XQ, Han GK, Zhang H, Qin XY, Zhang R, Nie QM and Jin F: Effect of corilagin on the proliferation and NF-κB in U251 glioblastoma cells and U251 glioblastoma stem-like Cells. Evid Based Complement Alternat Med. 2016:14183092016. View Article : Google Scholar : PubMed/NCBI

12 

Milani R, Brognara E, Fabbri E, Finotti A, Borgatti M, Lampronti I, Marzaro G, Chilin A, Lee KK, Kok SH, et al: Corilagin induces high levels of apoptosis in the temozolomide-resistant T98G glioma cell line. Oncol Res. 26:1307–1315. 2018. View Article : Google Scholar : PubMed/NCBI

13 

Lee J, Kim J, Kim EM, Kim U, Kang AR, Park JK and Um HD: p21WAF1/CIP1 promotes p53 protein degradation by facilitating p53-Wip1 and p53-Mdm2 interaction. Biochem Biophys Res Commun. 543:23–28. 2021. View Article : Google Scholar : PubMed/NCBI

14 

Sakata E, Eisele MR and Baumeister W: Molecular and cellular dynamics of the 26S proteasome. Biochim Biophys Acta Proteins Proteom. 1869:1405832021. View Article : Google Scholar : PubMed/NCBI

15 

Bard JAM, Goodall EA, Greene ER, Jonsson E, Dong KC and Martin A: Structure and function of the 26S proteasome. Annu Rev Biochem. 87:697–724. 2018. View Article : Google Scholar : PubMed/NCBI

16 

Murata S, Takahama Y, Kasahara M and Tanaka K: The immunoproteasome and thymoproteasome: Functions, evolution and human disease. Nat Immunol. 19:923–931. 2018. View Article : Google Scholar : PubMed/NCBI

17 

Rousseau A and Bertolotti A: Regulation of proteasome assembly and activity in health and disease. Nat Rev Mol Cell Biol. 19:697–712. 2018. View Article : Google Scholar : PubMed/NCBI

18 

Ettari R, Pallio G, Pizzino G, Irrera N, Zappalà M, Maiorana S, Di Chio C, Altavilla D, Squadrito F and Bitto A: Non-covalent immunoproteasome inhibitors induce cell cycle arrest in multiple myeloma MM.1R cells. J Enzyme Inhib Med Chem. 34:1307–1313. 2019. View Article : Google Scholar : PubMed/NCBI

19 

Kuzina ES, Kudriaeva AA, Maltseva DV and Belogurov AA Jr: Peptidyl aldehyde specifically interacts with immunosubunit β1i proteasome: In vitro and in vivo effects. Bull Exp Biol Med. 161:69–71. 2016. View Article : Google Scholar : PubMed/NCBI

20 

Sun C, Mo M, Wang Y, Yu W, Song C, Wang X, Chen S and Liu Y: Activation of the immunoproteasome protects SH-SY5Y cells from the toxicity of rotenone. Neurotoxicology. 73:112–119. 2019. View Article : Google Scholar : PubMed/NCBI

21 

Vilchez D, Boyer L, Morantte I, Lutz M, Merkwirth C, Joyce D, Spencer B, Page L, Masliah E, Berggren WT, et al: Increased proteasome activity in human embryonic stem cells is regulated by PSMD11. Nature. 489:304–308. 2012. View Article : Google Scholar : PubMed/NCBI

22 

Toste Rêgo A and da Fonseca PCA: Characterization of fully recombinant human 20S and 20S-PA200 proteasome complexes. Mol Cell. 76:138–147.e5. 2019. View Article : Google Scholar : PubMed/NCBI

23 

Hanssum A, Zhong Z, Rousseau A, Krzyzosiak A, Sigurdardottir A and Bertolotti A: An inducible chaperone adapts proteasome assembly to stress. Mol Cell. 55:566–577. 2014. View Article : Google Scholar : PubMed/NCBI

24 

Collins GA and Goldberg AL: The logic of the 26S proteasome. Cell. 169:792–806. 2017. View Article : Google Scholar : PubMed/NCBI

25 

Kuo CL and Goldberg AL: Ubiquitinated proteins promote the association of proteasomes with the deubiquitinating enzyme Usp14 and the ubiquitin ligase Ube3c. Proc Natl Acad Sci USA. 114:E3404–E3413. 2017. View Article : Google Scholar : PubMed/NCBI

26 

Lokireddy S, Kukushkin NV and Goldberg AL: cAMP-induced phosphorylation of 26S proteasomes on Rpn6/PSMD11 enhances their activity and the degradation of misfolded proteins. Proc Natl Acad Sci USA. 112:E7176–E7185. 2015. View Article : Google Scholar : PubMed/NCBI

27 

Myeku N, Clelland CL, Emrani S, Kukushkin NV, Yu WH, Goldberg AL and Duff KE: Tau-driven 26S proteasome impairment and cognitive dysfunction can be prevented early in disease by activating cAMP-PKA signaling. Nat Med. 22:46–53. 2016. View Article : Google Scholar : PubMed/NCBI

28 

Min L, Zeng X, Li B, Tao B, Shi J, Zhang W, Sun Q, Jing C and Wang X: Overexpression of immunoproteasome low-molecular-mass polypeptide 7 and inhibiting role of next-generation proteasome inhibitor ONX 0912 on cell growth in glioma. Neuroreport. 30:1031–1038. 2019. View Article : Google Scholar : PubMed/NCBI

29 

Qin XY, Zhang YL, Chi YF, Yan B, Zeng XJ, Li HH and Liu Y: Angiotensin II regulates Th1 T cell differentiation through angiotensin II type 1 receptor-PKA-mediated activation of proteasome. Cell Physiol Biochem. 45:1366–1376. 2018. View Article : Google Scholar : PubMed/NCBI

30 

Ettari R, Previti S, Bitto A, Grasso S and Zappalà M: Immunoproteasome-selective inhibitors: A promising strategy to treat hematologic malignancies, autoimmune and inflammatory diseases. Curr Med Chem. 23:1217–1238. 2016. View Article : Google Scholar : PubMed/NCBI

31 

Welk V, Coux O, Kleene V, Abeza C, Trümbach D, Eickelberg O and Meiners S: Inhibition of proteasome activity induces formation of alternative proteasome complexes. J Biol Chem. 291:13147–13159. 2016. View Article : Google Scholar : PubMed/NCBI

32 

Mannhaupt G, Schnall R, Karpov V, Vetter I and Feldmann H: Rpn4p acts as a transcription factor by binding to PACE, a nonamer box found upstream of 26S proteasomal and other genes in yeast. FEBS Lett. 450:27–34. 1999. View Article : Google Scholar : PubMed/NCBI

33 

Zarfati M, Avivi I, Brenner B, Katz T and Aharon A: Extracellular vesicles of multiple myeloma cells utilize the proteasome inhibitor mechanism to moderate endothelial angiogenesis. Angiogenesis. 22:185–196. 2019. View Article : Google Scholar : PubMed/NCBI

34 

Park JE, Miller Z, Jun Y, Lee W and Kim KB: Next-generation proteasome inhibitors for cancer therapy. Transl Res. 198:1–16. 2018. View Article : Google Scholar : PubMed/NCBI

35 

Huber EM, Basler M, Schwab R, Heinemeyer W, Kirk CJ, Groettrup M and Groll M: Immuno- and constitutive proteasome crystal structures reveal differences in substrate and inhibitor specificity. Cell. 148:727–738. 2012. View Article : Google Scholar : PubMed/NCBI

36 

Muchamuel T, Basler M, Aujay MA, Suzuki E, Kalim KW, Lauer C, Sylvain C, Ring ER, Shields J, Jiang J, et al: A selective inhibitor of the immunoproteasome subunit LMP7 blocks cytokine production and attenuates progression of experimental arthritis. Nat Med. 15:781–787. 2009. View Article : Google Scholar : PubMed/NCBI

37 

Yoo YD, Lee DH, Cha-Molstad H, Kim H, Mun SR, Ji C, Park SH, Sung KS, Choi SA, Hwang J, et al: Glioma-derived cancer stem cells are hypersensitive to proteasomal inhibition. EMBO Rep. 18:150–168. 2017. View Article : Google Scholar : PubMed/NCBI

38 

Su L, Guo W, Lou L, Nie S, Zhang Q, Liu Y, Chang Y, Zhang X, Li Y and Shen H: EGFR-ERK pathway regulates CSN6 to contribute to PD-L1 expression in glioblastoma. Mol Carcinog. 59:520–532. 2020. View Article : Google Scholar : PubMed/NCBI

39 

Yang XF, Zhao ZJ, Liu JJ, Yang XH, Gao Y, Zhao S, Shi S, Huang KQ and Zheng HC: SAHA and/or MG132 reverse the aggressive phenotypes of glioma cells: An in vitro and vivo study. Oncotarget. 8:3156–3169. 2017. View Article : Google Scholar : PubMed/NCBI

40 

Tang JH, Yang L, Chen JX, Li QR, Zhu LR, Xu QF, Huang GH, Zhang ZX, Xiang Y, Du L, et al: Bortezomib inhibits growth and sensitizes glioma to temozolomide (TMZ) via down-regulating the FOXM1-Survivin axis. Cancer Commun (Lond). 39:812019. View Article : Google Scholar : PubMed/NCBI

41 

Zhang M, Lu L, Ying M, Ruan H, Wang X, Wang H, Chai Z, Wang S, Zhan C, Pan J and Lu W: Enhanced Glioblastoma targeting ability of carfilzomib enabled by a DA7R-modified lipid nanodisk. Mol Pharm. 15:2437–2447. 2018. View Article : Google Scholar : PubMed/NCBI

42 

Kong XT, Nguyen NT, Choi YJ, Zhang G, Nguyen HN, Filka E, Green S, Yong WH, Liau LM, Green RM, et al: Phase 2 study of bortezomib combined with temozolomide and regional radiation therapy for upfront treatment of patients with newly diagnosed glioblastoma multiforme: Safety and efficacy assessment. Int J Radiat Oncol Biol Phys. 100:1195–1203. 2018. View Article : Google Scholar : PubMed/NCBI

43 

Liu H, Wan C, Ding Y, Han R, He Y, Xiao J and Hao J: PR-957, a selective inhibitor of immunoproteasome subunit low-MW polypeptide 7, attenuates experimental autoimmune neuritis by suppressing Th17-cell differentiation and regulating cytokine production. FASEB J. 31:1756–1766. 2017. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

April-2021
Volume 45 Issue 4

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
Qin X, Liu J, Pan D, Ma W, Cheng P and Jin F: Corilagin induces human glioblastoma U251 cell apoptosis by impeding activity of (immuno)proteasome. Oncol Rep 45: 34, 2021
APA
Qin, X., Liu, J., Pan, D., Ma, W., Cheng, P., & Jin, F. (2021). Corilagin induces human glioblastoma U251 cell apoptosis by impeding activity of (immuno)proteasome. Oncology Reports, 45, 34. https://doi.org/10.3892/or.2021.7985
MLA
Qin, X., Liu, J., Pan, D., Ma, W., Cheng, P., Jin, F."Corilagin induces human glioblastoma U251 cell apoptosis by impeding activity of (immuno)proteasome". Oncology Reports 45.4 (2021): 34.
Chicago
Qin, X., Liu, J., Pan, D., Ma, W., Cheng, P., Jin, F."Corilagin induces human glioblastoma U251 cell apoptosis by impeding activity of (immuno)proteasome". Oncology Reports 45, no. 4 (2021): 34. https://doi.org/10.3892/or.2021.7985