Adiponectin facilitates the cell cycle, inhibits cell apoptosis and induces temozolomide resistance in glioblastoma via the Akt/mTOR pathway
- Authors:
- Published online on: January 7, 2025 https://doi.org/10.3892/ol.2025.14875
- Article Number: 127
-
Copyright: © Sun et al. This is an open access article distributed under the terms of Creative Commons Attribution License.
Abstract
Introduction
Gliomas are the most common type of primary brain tumor with a global prevalence of 42.8% among primary central nervous system tumors (1,2). According to the World Health Organization (WHO), gliomas can be classified into four grades and a higher grade reflects a higher degree of malignancy (3). Glioblastoma, a WHO grade IV glioma, is the most aggressive type of glioma and accounts for ~49% of malignant brain tumors (4,5). It is estimated that the annual incidence of glioblastoma is ~3.23 per 100,000 individuals (6). Temozolomide (TMZ) is the first-line chemotherapy for glioblastoma and acts by inducing DNA nucleotide mismatch, leading to glioblastoma cell damage, apoptosis and cell cycle arrest in the G2/M phase (7–10). However, TMZ resistance develops in nearly 50% of patients with glioblastoma and is a contributor to poor prognosis (10–12). Therefore, exploring potential mechanisms that lead to TMZ resistance is worthwhile in improving the management of patients with glioblastoma.
TMZ resistance can be induced by the overexpression of O6-methylguanine-DNA methyltransferase and/or deficiency in the DNA repair pathway (11). Recently, other mechanisms for TMZ resistance have been proposed (13). The Akt/mTOR pathway plays a fundamental role in glioblastoma pathology and progression, which regulates multiple cellular processes, such as cell survival, proliferation, angiogenesis, invasion and metastasis (14). Of note, this pathway also played a fundamental role in TMZ resistance in glioblastoma according to previous studies (15–17).
Adiponectin (ADN) is a protein consisting of 244 amino acids with a molecular weight of 28 kDa, which is secreted by adipocytes and serves as a pivotal mediator of insulin sensitivity, lipid metabolism and inflammation (18). Previous studies report that ADN was closely involved in the pathology and progression of various types of cancer, including glioblastoma (19–22). For instance, one study indicated that ADN regulates pancreatic cancer cell growth through the β-catenin pathway (23). Regarding glioblastoma, another study illustrated that ADN regulates DNA synthesis, cell proliferation and cell cycle arrest to modulate glioblastoma progression (22). ADN also exerts fundamental effects on drug resistance in cancer cells (24) and ADN regulates the sensitivity of prostate cancer cells to doxorubicin (25). Another study proposed that ADN modulated sunitinib sensitivity by abrogating the PI3K/Akt/NF-κB pathway in renal cell carcinoma (26). Considering the involvement of ADN in glioblastoma progression and its ability to modulate drug resistance, it was hypothesized that ADN may also participate in TMZ resistance in glioblastoma.
The current study intended to explore the engagement of ADN in TMZ resistance in glioblastoma and the involvement of the Akt/mTOR pathway during this process.
Materials and methods
Cell culture and ADN treatment
U251 (cat. no. SCSP-559; National Collection of Authenticated Cell Cultures) and U87-MG (cat. no. TCHu138; National Collection of Authenticated Cell Cultures) cells were maintained in high-glucose Dulbecco's Modified Eagle's Medium (cat. no. SH30022.01; Hyclone; Cytiva) containing 10% fetal bovine serum (cat. no. SH30084.04; Hyclone; Cytiva) at 37°C in the presence of 5% CO2. They were subsequently treated with ADN (cat. no. 1065-AP-050; R&D Systems, Inc.) at concentrations of 0.1, 0.5, 1.0 3.0 and 10.0 µg/ml. Subsequently, the cell growth rate was determined by the Cell Counting Kit-8 (CCK-8) assay and the phosphorylated (p)-Akt (Thr308), p-Akt (Ser473), Akt and p-mTOR expression was analyzed by western blotting. The U87-MG cell line used in the present study was the American Type Culture Collection (ATCC) version, which is most probably a glioblastoma cell line of unknown origin. The authenticity of the U87-MG cell line was verified by Short Tandem Repeat (STR) Profiling. The STR Profiling results of the U87-MG cell line were as follows: Amelogenin: X; D5S818: 11, 12; D13S317: 8, 11; D7S820: 8, 9; D16S539: 12; vWA: 15, 17; TH01: 9.3; TPOX: 8; CSF1PO: 10, 11; D19S433: 15, 15.2; D21S11: 28, 32.2; D18S51: 13, 14, which were from the website of the supplier (https://www.cellbank.org.cn/search-detail.php?id=211).
LY294002 treatment
LY294002 (a PI3K inhibitor; cat. no. ab120243; Abcam) was dissolved in dimethyl sulfoxide (cat. no. PHR1309; MilliporeSigma) for further treatment. Subsequently, U251 and U87-MG cells were treated with 10 µM LY294002 in combination with ADN for 24 h at 37°C. Furthermore, the apoptotic rate was assessed by Annexin V/propidium iodide (AV/PI). The expression levels of cleaved caspase 3 (c-caspase 3), caspase 3 and Bax were evaluated by western blotting.
TMZ treatment
TMZ (cat. no. ab141055; Abcam) at concentrations of 0.1 and 1.0 mM were cultured with U251 and U87-MG cells, respectively. ADN with concentrations of 1, 2 and 3 µg/ml were cultured with cells in combination with TMZ at 0.1 and 1.0 mM at 37°C for 24 h. The mixture of 1.0 mM TMZ, 3 µg/ml ADN and 10 µM LY294002 was also added to the cells. Finally, 1 mM TMZ or LY294002 was cultured with the cells in the presence of 3 µg/ml ADN at 37°C for 24 h. Subsequently, the cell growth rate was assessed using CCK-8, the apoptotic rate and cell cycle were evaluated using AV/PI assay and cell cycle assay; the expression levels of c-caspase 3, caspase 3, Bax, cyclin B1 and cyclin D1 were determined by western blotting.
CCK-8 assay
The CCK-8 regent (cat. no. C0037; Beyotime Institute of Biotechnology) with an amount of 50 µl was added and cultivated with the cells for 1 h at 37°C. Subsequently, the optical density value at 450 nm was measured with a microplate reader (Bio-Rad Laboratories, Inc.). The cell growth rate was calculated.
AV/PI and cell cycle assays
An AV/PI kit (cat. no. C1062S; Beyotime Institute of Biotechnology) was used to detect cell apoptosis. The cells were digested with trypsin (cat. no. SH30042.01; Hyclone; Cytiva) for 1 min at room temperature and re-suspended. Following rinsing with PBS, the cells were incubated with 5 µl AV and 5 µl PI at room temperature in the dark for 15 min at room temperature. Finally, the cells were measured with a CytoFLEX flow cytometer (Beckman Coulter, Inc.). The data were analyzed with FlowJo X (FlowJo LLC). The cells in quadrant 2 (Q2) and quadrant 4 (Q4) were considered as apoptotic cells.
For the cell cycle assay, the cells were harvested and re-suspended. Subsequently, they were fixed in 70% ethanol at 4°C overnight. The cells were centrifuged (800 × g at 4°C for 3 min) and collected and stained with PI solution (cat. no. ST1569; Beyotime Institute of Biotechnology) for 30 min at room temperature. A CytoFLEX flow cytometer (Beckman Coulter, Inc.) was applied to detect the cells.
Western blotting
The cells were lysed with radio immunoprecipitation assay buffer (cat. no. V900854; MilliporeSigma) and the protein solution was quantified with the bicinchoninic acid kit (cat. no. 23225; Thermo Fisher Scientific, Inc.). The protein was denatured at 100°C for 10 min following mixing with a loading buffer. Subsequently, 10-µg protein samples were loaded into a 4–20% sodium dodecyl sulfate polyacrylamide gel electrophoresis gel for separation and transferred to the nitrocellulose membrane (cat. no. HATF00010; MilliporeSigma). The membrane was blocked with skimmed milk for 2 h at 37°C, followed by incubation with primary and secondary antibodies overnight at 4°C and secondary antibodies at 37°C for 90 min. The primary antibodies used were for the following proteins: Adiponectin receptor (ADIPOR) 1 (cat. no. APR06109G; 1:1,000; Epitomics, Inc.), ADIPOR2 (cat. no. APG01582G; 1:500; Epitomics, Inc.), Akt (cat. no. 9272S; 1:1,000; Cell Signaling Technology, Inc.), p-Akt (Thr308) (cat. no. 9275S; 1:500; Cell Signaling Technology, Inc.), p-Akt (Ser473) (cat. no. 9271S; 1:500; Cell Signaling Technology, Inc.), c-caspase 3 (cat. no. 9661S; 1:500; Cell Signaling Technology, Inc.), caspase 3 (cat. no. 9662S; 1:1,000; Cell Signaling Technology, Inc.), Bax (cat. no. 2772S; 1:1,000; Cell Signaling Technology, Inc.), p-mTOR (cat. no. 2983S; 1:500; Cell Signaling Technology, Inc.), cyclin B1 (cat. no. ab32053; 1:1,000; Abcam), cyclin D1 (cat. no. ab16663; 1:1,000; Abcam), β-actin (cat. no. 20536-1-AP; 1:4,000; Proteintech Group, Inc.) and glyceraldehyde-3-phosphate dehydrogenase (GAPDH) antibody (cat. no. 10494-1-AP; 1:4,000; Proteintech Group, Inc.). Finally, the horseradish peroxidase-conjugated secondary antibody (cat. no. ab6721; 1:10,000; Abcam) was incubated with the membrane. The protein bands were visualized with an enhanced chemiluminescence kit (cat. no. 32209; Thermo Fisher Scientific, Inc.) and X-ray film. The gray value was quantified by Gene Tools 3.7 (SynGene).
Statistical analysis
The SPSS software (v.26.0; IBM Corp.) was used for statistical analysis. One-way ANOVA was applied for the comparison among groups and Tukey's test was employed for post-hoc comparison. P<0.05 was considered to indicate a statistically significant difference.
Results
ADN increases growth and activates the Akt/mTOR pathway in U251 and U87-MG cells
Considering that ADN binds to its receptors to exert its functions, western blotting was applied to detect ADIPOR1 and ADIPOR2 in U251 and U87-MG cells. It was found that ADIPOR1 and ADIPOR2 were expressed in U251 and U87-MG cells, suggesting that these two types of cells possessed the basis for ADN to exert its functions (Fig. S1).
U251 cell growth rate was increased following treatment with 1.0, 3.0 and 10.0 µg/ml ADN compared with that noted in the control (all P<0.05; Fig. 1A). Subsequently, U251 cells were treated with 3.0 µg/ml ADN at different time points. It was found that following ADN treatment for 24 and 48 h, U251 cell growth rate was increased compared with that of the control (both P<0.05; Fig. 1B). Similarly, the effect of ADN on the U87-MG cell growth rate further indicated a concentration-dependent (Fig. 1C) and time-dependent (Fig. 1D) mode of action. It was noted that when the treatment duration was 24 h, ADN exhibited an optimal effect in promoting U251 and U87-MG cell growth. In addition, compared with the treatment duration of 24 h, the effect of ADN on U251 and U87-MG cell growth was not further promoted after 48 h (both P>0.05). Therefore, the treatment duration of ADN for 24 h was selected as the experimental condition for subsequent experiments.
According to western blotting analysis (Fig. 2A), p-Akt (Thr308)/Akt was elevated following 1.0 and 3.0 µg/ml treatment with ADN compared with that of the control in U251 cells (both P<0.05; Fig. 2B). However, p-Akt (Ser473)/Akt was not affected by ADN treatment at any concentration used in U251 cells (all P>0.05; Fig. 2C). p-mTOR/GAPDH was increased following treatment with 1.0 and 3.0 µg/ml ADN compared with those of the control in U251 cells (both P<0.05; Fig. 2D). The same trends were also noted in U87-MG cells (Fig. 2E-H).
Notably, when the concentration was 3.0 µg/ml, ADN indicated outstanding ability to activate the Akt/mTOR pathway and facilitate U251 and U87-MG cell growth. Therefore, 3.0 µg/ml ADN was selected as the experimental condition for subsequent experiments.
ADN activates the Akt/mTOR pathway to inhibit apoptosis in U251 and U87-MG cells
According to the AV/PI assay (Fig. 3A), the apoptotic rate was decreased by ADN compared with that of the vehicle (P<0.05); however, it was increased by ADN + LY294002 compared with ADN in U251 cells (P<0.05; Fig. 3B). western blotting (Fig. 3C) indicated that c-caspase 3/caspase3 (Fig. 3D) and Bax/GAPDH (Fig. 3E) were decreased by ADN compared with the vehicle (both P<0.05); however, they were elevated by ADN + LY294002 compared with ADN in U251 cells (both P<0.05). The same trends were noted in U87-MG cells (Fig. 3F-J).
ADN activates the Akt/mTOR pathway to increase U251 and U87-MG cells in the S phase
According to cell cycle assay results (Fig. 4A), the proportion of U251 cells in the S phase was increased by ADN treatment compared with the vehicle (P<0.05); however, it was reduced by ADN + LY294002 compared with ADN (P<0.05; Fig. 4B). Western blotting (Fig. 4C) indicated that cyclin B1/β-actin (Fig. 4D) and cyclin D1/β-actin (Fig. 4E) were increased following treatment with ADN compared with the vehicle (P<0.05); however, they were decreased by ADN + LY294002 compared with ADN in U251 cells (P<0.05). Similar trends were noted in U87-MG cells (Fig. 4F-J).
ADN facilitates TMZ resistance in U251 and U87-MG cells
Different concentrations of TMZ were applied to treat the glioblastoma cell lines. It was noted that treatment with 1.0 mM TMZ indicated an optimal effect on inhibiting glioblastoma cell line growth; therefore, this concentration was selected as the optimal treatment condition for the subsequent step. In addition, it was found that only 3.0 µg/ml ADN+1.0 mM TMZ increased glioblastoma cell line growth compared with 1.0 mM TMZ (both P<0.05; Fig. S2A and B). Therefore, 3.0 µg/ml ADN and 1.0 mM TMZ were selected as the conditions for subsequent experiments.
The AV/PI assay (Fig. 5A) suggested that the apoptotic rate was enhanced by TMZ compared with vehicle (P<0.05); however, it was reduced by ADN+TMZ compared with TMZ in U251 cells (P<0.05; Fig. 5B). Western blotting analysis (Fig. 5C) revealed that the ratio of c-caspase 3/caspase 3 (Fig. 5D) and the ratio of Bax/GAPDH (Fig. 5E) were increased by TMZ compared with vehicle (all P<0.05); however, they were reduced by ADN+TMZ compared with TMZ in U251 cells (both P<0.05). The same trends were noted in U87-MG cells (Fig. 5F-J).
The cell cycle assay (Fig. 6A) suggested that the proportion of U251 cells in the G2 phase was increased by TMZ compared with that of the vehicle group (P<0.05), whereas it was decreased in the ADN+TMZ group compared with that noted in the TMZ group (P<0.05; Fig. 6B). The same trends were noted in U87-MG cells (Fig. 6C and D).
ADN activates the Akt/mTOR pathway to facilitate TMZ resistance in U251 and U87-MG cells
LY294002 was added to further explore the underlying mechanism of ADN involved in TMZ resistance. It was found that the U251 cell growth rate was reduced by TMZ + ADN + LY294002 compared with TMZ+ADN (P<0.05; Fig. S2A). The same trends were discovered in U87-MG cells (Fig. S2B).
Discussion
The actions of ADN are mediated via classical receptors, which belong to the seven transmembrane domains receptor family, such as ADIPOR1 and ADIPOR2 (27–29). Notably, ADIPOR1 and ADIPOR2 are closely involved in the pathology and progression of glioblastoma (30,31) and several studies have reported that they are expressed in various tumor tissues, including glioma (22,32–34). According to a previous study, ADIPOR1 and ADIPOR2 are expressed in 83% (25/30) of human glioma tissues; in addition, 70% of glioma tissues indicated co-expression of both receptors, which were also expressed in glioblastoma cell lines (22). In line with this previous study (22), the present study also discovered that ADIPOR1 and ADIPOR2 were expressed in U251 and U87-MG cells, suggesting that these two glioblastoma cell lines possessed the basis for ADN to exert its functions. Subsequently, it was noted that ADN could facilitate glioblastoma cell line growth and cells in the S phase, while inhibiting apoptosis. A possible reason might be that ADN can bind to ADIPOR1 and ADIPOR2 to regulate several downstream pathways, such as the adenosine monophosphate-activated protein kinase (AMPK)/sirtuin 1, AMPK/mTOR and extracellular signal-regulated kinase (ERK)1/2 pathways to accelerate glioblastoma cell line proliferation, while inhibiting apoptosis (22,30,31).
The Akt/mTOR pathway, a key regulator of multiple cellular processes, is involved in the pathology and progression of various types of cancer (14,35–37). Regarding glioblastoma, previous studies have indicated that the Akt/mTOR pathway regulates cell proliferation, apoptosis, metastasis, autophagy and cell cycle arrest (38–40). Notably, a previous study discovered that Akt phosphorylation can be regulated by ADN, which is responsible for glioblastoma progression (22). The current study showed that ADN activated the Akt/mTOR pathway in glioblastoma cell lines, which was consistent with a previous study (22). It should be clarified that Ser473 and Thr308 are two phosphorylation sites of Akt (41). However, it was found that ADN could only lead to Akt phosphorylation at Thr308, while it could not induce its phosphorylation at Ser473 in glioblastoma cell lines. It is to notable that the addition of LY294002 reversed the effect of ADN on glioblastoma cell line apoptosis and cells in the S phase. The findings suggested that ADN might activate the Akt/mTOR pathway to facilitate glioblastoma progression.
TMZ is the first-line chemotherapy for patients with glioblastoma, whereas drug resistance to TMZ remains a challenging problem and is a cause of poor prognosis (42,43). U251 and U87-MG cell lines are widely used cell lines to establish TMZ-resistant glioblastoma cell models according to previous studies (44–48). Guided by these previous studies (44–48), U251 and U87-MG cell lines we also applied to explore the effect of ADN on TMZ resistance in glioblastoma. It was found that ADN reversed the effect of TMZ on apoptosis, cell cycle arrest and growth in glioblastoma cell lines, which indicated that it contributed to TMZ resistance in glioblastoma. However, the underlying mechanisms responsible for ADN-induced TMZ resistance have not been reported by previous studies. The Akt/mTOR pathway was found to be aberrantly activated in primary glioblastoma samples and its activation could impair the efficacy of TMZ treatment (16,49–51). Therefore, it was further explored whether this pathway is involved in the regulation of ADN on TMZ resistance in glioblastoma. Notably, it was discovered that the addition of LY294002 reversed the effect of ADN on TMZ resistance in glioblastoma cell lines. Therefore, ADN might induce TMZ resistance by activating the Akt/mTOR pathway in glioblastoma.
The current study observed that ADN facilitated glioblastoma progression and induced TMZ resistance by activating the Akt/mTOR pathway. However, the underlying mechanisms of ADN's regulation of the Akt/mTOR pathway were unclear. According to a previous study, it was hypothesized that ADN might bind to its receptors (ADIPOR1 and ADIPOR2) to initiate the activation of PI3K, which further activated the Akt/mTOR pathway, thereby promoting glioblastoma progression and TMZ resistance (22). However, this hypothesis should be validated by further experiments.
It should be clarified that this study applied 0.1, 0.5, 1.0, 3.0 and 10.0 µg/ml as the concentration gradient of ADN to treat U251 and U87-MG cell lines. The setting of these concentrations of ADN was exploratory in nature. Initially, the present study tried to follow the experience of a relevant previous study, which used 0.0025, 0.025 and 0.25 µg/ml as the concentration gradient of ADN to treat U251 and U87-MG cell lines (22). However, after applying similar concentrations of ADN (0.1 and 0.5 µg/ml), it was found that U251 and U87-MG cell line growth was unaffected. Consequently, it was decided to use higher concentrations of ADN (1.0, 3.0 and 10.0 µg/ml) to treat these cell lines. It is hoped that the concentration gradient of ADN set in the present study can provide a reference for future researchers embarking on similar investigations.
Recently, several studies have revealed the potential mechanisms regarding glioblastoma progression and relapse, which are conducive to improving the management of glioblastoma patients (52–54). Nevertheless, the prognosis of glioblastoma patients is unsatisfactory and one of the contributors is TMZ resistance, which is developed in ~50% of glioblastoma patients (10). Therefore, investigating potential mechanisms contributing to TMZ resistance is crucial to improving the prognosis of patients with glioblastoma. The current study found that ADN was responsible for glioblastoma progression and TMZ resistance. The findings provided perspectives that ADN and its downstream Akt/mTOR pathway might be potential therapeutic targets to reverse TMZ resistance in patients with glioblastoma. Meanwhile, considering the involvement of ADN in glioblastoma progression and TMZ resistance, ADN may serve as a potential marker for patients with glioblastoma receiving TMZ. Therefore, the detection of ADN by enzyme-linked immunosorbent assay and reverse transcription PCR might assist in predicting the prognosis of patients with glioblastoma receiving TMZ. Overall, the findings might be conducive to enhancing the management of patients with glioblastoma. To achieve this goal, further in vivo experiments are required to validate the effect of ADN on glioblastoma progression and TMZ resistance. Further clinical studies are warranted to verify the potential of ADN to serve as a biomarker for patients with glioblastoma receiving TMZ.
Several limitations of this study should be noted. i) The underlying mechanism of the regulation of ADN on the Akt/mTOR pathway should be investigated by further experiments. ii) The glioblastoma cell lines used in this study included U251 and U87-MG cell lines, which could not fully represent the glioblastoma. Further studies could consider applying primary glioblastoma cells or other glioblastoma cell lines, such as A172, LN229 and LN18 cell lines, to validate the findings of this study. iii) Further in vivo experiments were required to confirm the engagement of ADN in glioblastoma progression and TMZ resistance.
In conclusion, the present study demonstrated that ADN activated the Akt/mTOR pathway to facilitate cell cycle, inhibited cell apoptosis and induced TMZ resistance in glioblastoma.
Supplementary Material
Supporting Data
Acknowledgements
Not applicable.
Funding
The present study was supported by The Role of collagen receptor DDR2 in cerebral vasospasm: A Youth project of Shaanxi Natural Science Basic Research Program (grant no. 2016SF-033).
Availability of data and materials
The data generated in the present study may be requested from the corresponding author.
Authors' contributions
JY and JH contributed to the conception and design of the study. PS and FL were responsible for the acquisition of data. KH, JW and YC were responsible for analysis and interpretation of the data. SS contributed to the methodology of the overal study design and wrote the first draft of the manuscript. WM contributed to the interpretation of the data and manuscript revision.. JY and JH confirm the authenticity of 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
van den Bent MJ, Geurts M, French PJ, Smits M, Capper D, Bromberg JEC and Chang SM: Primary brain tumours in adults. Lancet. 402:1564–1579. 2023. View Article : Google Scholar : PubMed/NCBI | |
Salari N, Ghasemi H, Fatahian R, Mansouri K, Dokaneheifard S, Shiri MH, Hemmati M and Mohammadi M: The global prevalence of primary central nervous system tumors: A systematic review and meta-analysis. Eur J Med Res. 28:392023. View Article : Google Scholar : PubMed/NCBI | |
Weller M, Wen PY, Chang SM, Dirven L, Lim M, Monje M and Reifenberger G: Glioma. Nat Rev Dis Primers. 10:332024. View Article : Google Scholar : PubMed/NCBI | |
Grochans S, Cybulska AM, Simińska D, Korbecki J, Kojder K, Chlubek D and Baranowska-Bosiacka I: Epidemiology of glioblastoma multiforme-literature review. Cancers (Basel). 14:24122022. View Article : Google Scholar : PubMed/NCBI | |
Schaff LR and Mellinghoff IK: Glioblastoma and other primary brain malignancies in adults: A review. JAMA. 329:574–587. 2023. View Article : Google Scholar : PubMed/NCBI | |
Miller KD, Ostrom QT, Kruchko C, Patil N, Tihan T, Cioffi G, Fuchs HE, Waite KA, Jemal A, Siegel RL and Barnholtz-Sloan JS: Brain and other central nervous system tumor statistics, 2021. CA Cancer J Clin. 71:381–406. 2021. View Article : Google Scholar : PubMed/NCBI | |
Rodríguez-Camacho A, Flores-Vázquez JG, Moscardini Martelli J, Torres-Ríos JA, Olmos-Guzmán A, Ortiz-Arce CS, Cid-Sánchez DR, Pérez SR, Macías-González MDS, Hernández-Sánchez LC, et al: Glioblastoma Treatment: State-of-the-Art and Future Perspectives. Int J Mol Sci. 23:72022022. View Article : Google Scholar | |
Czarnywojtek A, Borowska M, Dyrka K, Van Gool S, Sawicka-Gutaj N, Moskal J, Kościński J, Graczyk P, Hałas T, Lewandowska AM, et al: Glioblastoma Multiforme: The latest diagnostics and treatment techniques. Pharmacology. 108:423–431. 2023. View Article : Google Scholar : PubMed/NCBI | |
Aldoghachi AF, Aldoghachi AF, Breyne K, Ling KH and Cheah PS: Recent Advances in the therapeutic strategies of glioblastoma multiforme. Neuroscience. 491:240–270. 2022. View Article : Google Scholar : PubMed/NCBI | |
Karachi A, Dastmalchi F, Mitchell DA and Rahman M: Temozolomide for immunomodulation in the treatment of glioblastoma. Neuro Oncology. 20:1566–1572. 2018. View Article : Google Scholar : PubMed/NCBI | |
Tomar MS, Kumar A, Srivastava C and Shrivastava A: Elucidating the mechanisms of Temozolomide resistance in gliomas and the strategies to overcome the resistance. Biochim Biophys Acta Rev Cancer. 1876:1886162021. View Article : Google Scholar : PubMed/NCBI | |
Teraiya M, Perreault H and Chen VC: An overview of glioblastoma multiforme and temozolomide resistance: Can LC-MS-based proteomics reveal the fundamental mechanism of temozolomide resistance? Front Oncol. 13:11662072023. View Article : Google Scholar : PubMed/NCBI | |
Daisy Precilla S, Biswas I, Kuduvalli SS and Anitha TS: Crosstalk between PI3K/AKT/mTOR and WNT/β-Catenin signaling in GBM-Could combination therapy checkmate the collusion? Cell Signal. 95:1103502022. View Article : Google Scholar : PubMed/NCBI | |
Khabibov M, Garifullin A, Boumber Y, Khaddour K, Fernandez M, Khamitov F, Khalikova L, Kuznetsova N, Kit O and Kharin L: Signaling pathways and therapeutic approaches in glioblastoma multiforme (Review). Int J Oncol. 60:692022. View Article : Google Scholar : PubMed/NCBI | |
Guo L and Wu Z: FOXM1-mediated NUF2 expression confers temozolomide resistance to human glioma cells by regulating autophagy via the PI3K/AKT/mTOR signaling pathway. Neuropathology. 42:430–446. 2022. View Article : Google Scholar : PubMed/NCBI | |
Zając A, Sumorek-Wiadro J, Langner E, Wertel I, Maciejczyk A, Pawlikowska-Pawlęga B, Pawelec J, Wasiak M, Hułas-Stasiak M, Bądziul D, et al: Involvement of PI3K pathway in glioma cell resistance to temozolomide treatment. Int J Mol Sci. 22:51552021. View Article : Google Scholar : PubMed/NCBI | |
Xiong J, Guo G, Guo L, Wang Z, Chen Z, Nan Y, Cao Y, Li R, Yang X, Dong J, et al: Amlexanox enhances temozolomide-induced antitumor effects in human glioblastoma cells by inhibiting IKBKE and the Akt-mTOR signaling pathway. ACS Omega. 6:4289–4299. 2021. View Article : Google Scholar : PubMed/NCBI | |
Khoramipour K, Chamari K, Hekmatikar AA, Ziyaiyan A, Taherkhani S, Elguindy NM and Bragazzi NL: Adiponectin: Structure, physiological functions, role in diseases, and effects of nutrition. Nutrients. 13:11802021. View Article : Google Scholar : PubMed/NCBI | |
Naimo GD, Forestiero M, Paolì A, Malivindi R, Gelsomino L, Győrffy B, Leonetti AE, Giordano F, Panza S, Conforti FL, et al: ERα/LKB1 complex upregulates E-cadherin expression and stimulates breast cancer growth and progression upon adiponectin exposure. Int J Cancer. 153:1257–1272. 2023. View Article : Google Scholar : PubMed/NCBI | |
Illiano M, Nigro E, Sapio L, Caiafa I, Spina A, Scudiero O, Bianco A, Esposito S, Mazzeo F, Pedone PV, et al: Adiponectin down-regulates CREB and inhibits proliferation of A549 lung cancer cells. Pulm Pharmacol Ther. 45:114–120. 2017. View Article : Google Scholar : PubMed/NCBI | |
Nigro E, Orlandella FM, Polito R, Mariniello RM, Monaco ML, Mallardo M, De Stefano AE, Iervolino PLC, Salvatore G and Daniele A: Adiponectin and leptin exert antagonizing effects on proliferation and motility of papillary thyroid cancer cell lines. J Physiol Biochem. 77:237–248. 2021. View Article : Google Scholar : PubMed/NCBI | |
Porcile C, Di Zazzo E, Monaco ML, D'Angelo G, Passarella D, Russo C, Di Costanzo A, Pattarozzi A, Gatti M, Bajetto A, et al: Adiponectin as novel regulator of cell proliferation in human glioblastoma. J Cell Physiol. 229:1444–1454. 2014. View Article : Google Scholar : PubMed/NCBI | |
Jiang J, Fan Y, Zhang W, Shen Y, Liu T, Yao M, Gu J, Tu H and Gan Y: Adiponectin suppresses human pancreatic cancer growth through attenuating the β-Catenin signaling pathway. Int J Biol Sci. 15:253–264. 2019. View Article : Google Scholar : PubMed/NCBI | |
Li Y, Yu C and Deng W: Roles and mechanisms of adipokines in drug resistance of tumor cells. Eur J Pharmacol. 899:1740192021. View Article : Google Scholar : PubMed/NCBI | |
Bub JD, Miyazaki T and Iwamoto Y: Adiponectin as a growth inhibitor in prostate cancer cells. Biochem Biophys Res Commun. 340:1158–1166. 2006. View Article : Google Scholar : PubMed/NCBI | |
Sun G, Zhang X, Liu Z, Zhu S, Shen P, Zhang H, Zhang M, Chen N, Zhao J, Chen J, et al: The Adiponectin-AdipoR1 axis mediates tumor progression and tyrosine kinase inhibitor resistance in metastatic renal cell carcinoma. Neoplasia. 21:921–931. 2019. View Article : Google Scholar : PubMed/NCBI | |
Bocian-Jastrzębska A, Malczewska-Herman A and Kos-Kudła B: Role of leptin and adiponectin in carcinogenesis. Cancers (Basel). 15:42502023. View Article : Google Scholar : PubMed/NCBI | |
Fang H and Judd RL: Adiponectin regulation and function. Compr Physiol. 8:1031–1063. 2018. View Article : Google Scholar : PubMed/NCBI | |
Nigro E, Daniele A, Salzillo A, Ragone A, Naviglio S and Sapio L: AdipoRon and other adiponectin receptor agonists as potential candidates in cancer treatments. Int J Mol Sci. 22:55692021. View Article : Google Scholar : PubMed/NCBI | |
Liu X, Chen J and Zhang J: AdipoR1-mediated miR-3908 inhibits glioblastoma tumorigenicity through downregulation of STAT2 associated with the AMPK/SIRT1 pathway. Oncol Rep. 37:3387–3396. 2017. View Article : Google Scholar : PubMed/NCBI | |
Jie C, Xuan W, Feng HD, Hua DM, Bo W, Fei S and Hao Z: AdipoR2 inhibits human glioblastoma cell growth through the AMPK/mTOR pathway. Eur J Med Res. 26:282021. View Article : Google Scholar : PubMed/NCBI | |
Byeon JS, Jeong JY, Kim MJ, Lee SM, Nam WH, Myung SJ, Kim JG, Yang SK, Kim JH and Suh DJ: Adiponectin and adiponectin receptor in relation to colorectal cancer progression. Int J Cancer. 127:2758–2767. 2010. View Article : Google Scholar : PubMed/NCBI | |
Jardé T, Caldefie-Chézet F, Goncalves-Mendes N, Mishellany F, Buechler C, Penault-Llorca F and Vasson MP: Involvement of adiponectin and leptin in breast cancer: Clinical and in vitro studies. Endocr Relat Cancer. 16:1197–1210. 2009. View Article : Google Scholar : PubMed/NCBI | |
Williams CJ, Mitsiades N, Sozopoulos E, His A, Wolk A, Nifli AP, Tseleni-Balafouta S and Mantzoros CS: Adiponectin receptor expression is elevated in colorectal carcinomas but not in gastrointestinal stromal tumors. Endocr Relat Cancer. 15:289–299. 2008. View Article : Google Scholar : PubMed/NCBI | |
Barzegar Behrooz A, Talaie Z, Jusheghani F, Łos MJ, Klonisch T and Ghavami S: Wnt and PI3K/Akt/mTOR survival pathways as therapeutic targets in glioblastoma. Int J Mol Sci. 23:13532022. View Article : Google Scholar : PubMed/NCBI | |
Fattahi S, Amjadi-Moheb F, Tabaripour R, Ashrafi GH and Akhavan-Niaki H: PI3K/AKT/mTOR signaling in gastric cancer: Epigenetics and beyond. Life Sci. 262:1185132020. View Article : Google Scholar : PubMed/NCBI | |
Miricescu D, Totan A, Stanescu S II, Badoiu SC, Stefani C and Greabu M: PI3K/AKT/mTOR signaling pathway in breast cancer: From molecular landscape to clinical aspects. Int J Mol Sci. 22:1732020. View Article : Google Scholar : PubMed/NCBI | |
Zheng X, Li W, Xu H, Liu J, Ren L, Yang Y, Li S, Wang J, Ji T and Du G: Sinomenine ester derivative inhibits glioblastoma by inducing mitochondria-dependent apoptosis and autophagy by PI3K/AKT/mTOR and AMPK/mTOR pathway. Acta Pharm Sin B. 11:3465–3480. 2021. View Article : Google Scholar : PubMed/NCBI | |
Yang R, Wang M, Zhang G, Bao Y, Wu Y, Li X, Yang W and Cui H: E2F7-EZH2 axis regulates PTEN/AKT/mTOR signalling and glioblastoma progression. Br J Cancer. 123:1445–1455. 2020. View Article : Google Scholar : PubMed/NCBI | |
Jiang Y, Liu J, Hong W, Fei X and Liu R: Arctigenin inhibits glioblastoma proliferation through the AKT/mTOR pathway and induces autophagy. Biomed Res Int. 2020:35426132020. View Article : Google Scholar : PubMed/NCBI | |
Manning BD and Toker A: AKT/PKB Signaling: Navigating the Network. Cell. 169:381–405. 2017. View Article : Google Scholar : PubMed/NCBI | |
Lee SY: Temozolomide resistance in glioblastoma multiforme. Genes Dis. 3:198–210. 2016. View Article : Google Scholar : PubMed/NCBI | |
Chien CH, Hsueh WT, Chuang JY and Chang KY: Dissecting the mechanism of temozolomide resistance and its association with the regulatory roles of intracellular reactive oxygen species in glioblastoma. J Biomed Sci. 28:182021. View Article : Google Scholar : PubMed/NCBI | |
Thanasupawat T, Glogowska A, Burg M, Krcek J, Beiko J, Pitz M, Zhang GJ, Hombach-Klonisch S and Klonisch T: C1q/TNF-related peptide 8 (CTRP8) promotes temozolomide resistance in human glioblastoma. Mol Oncol. 12:1464–1479. 2018. View Article : Google Scholar : PubMed/NCBI | |
Sun Q, Xu J, Yuan F, Liu Y, Chen Q, Guo L, Dong H and Liu B: RND1 inhibits epithelial-mesenchymal transition and temozolomide resistance of glioblastoma via AKT/GSK3-β pathway. Cancer Biol Ther. 25:23217702024. View Article : Google Scholar : PubMed/NCBI | |
Wang X, Jia L, Jin X, Liu Q, Cao W, Gao X, Yang M and Sun B: NF-ĸB inhibitor reverses temozolomide resistance in human glioma TR/U251 cells. Oncol Lett. 9:2586–2590. 2015. View Article : Google Scholar : PubMed/NCBI | |
Li H, Chen L, Li JJ, Zhou Q, Huang A, Liu WW, Wang K, Gao L, Qi ST and Lu YT: miR-519a enhances chemosensitivity and promotes autophagy in glioblastoma by targeting STAT3/Bcl2 signaling pathway. J Hematol Oncol. 11:702018. View Article : Google Scholar : PubMed/NCBI | |
Yang YN, Zhang XH, Wang YM, Zhang X and Gu Z: miR-204 reverses temozolomide resistance and inhibits cancer initiating cells phenotypes by degrading FAP-α in glioblastoma. Oncol Lett. 15:7563–7570. 2018.PubMed/NCBI | |
Rao V, Kumar G, Vibhavari RJA, Nandakumar K, Thorat ND, Chamallamudi MR and Kumar N: Temozolomide Resistance: A Multifarious Review on Mechanisms Beyond O-6-Methylguanine-DNA Methyltransferase. CNS Neurol Disord Drug Targets. 22:817–831. 2023. View Article : Google Scholar : PubMed/NCBI | |
Singh N, Miner A, Hennis L and Mittal S: Mechanisms of temozolomide resistance in glioblastoma-a comprehensive review. Cancer Drug Resist. 4:17–43. 2021.PubMed/NCBI | |
Chen L, Han L, Shi Z, Zhang K, Liu Y, Zheng Y, Jiang T, Pu P, Jiang C and Kang C: LY294002 enhances cytotoxicity of temozolomide in glioma by down-regulation of the PI3K/Akt pathway. Mol Med Rep. 5:575–579. 2012.PubMed/NCBI | |
Dai X, Ye L, Li H, Dong X, Tian H, Gao P, Dong J and Cheng H: Crosstalk between microglia and neural stem cells influences the relapse of glioblastoma in GBM immunological microenvironment. Clin Immunol. 251:1093332023. View Article : Google Scholar : PubMed/NCBI | |
Gao P, Wang H, Li H, Shu L, Han Z, Li S, Cheng H and Dai X: miR-21-5p Inhibits the Proliferation, Migration, and Invasion of Glioma by Targeting S100A10. J Cancer. 14:1781–1793. 2023. View Article : Google Scholar : PubMed/NCBI | |
Zhou L, Li H, Yao H, Dai X, Gao P and Cheng H: TMED family genes and their roles in human diseases. Int J Med Sci. 20:1732–1743. 2023. View Article : Google Scholar : PubMed/NCBI |