AdipoR1-mediated miR-3908 inhibits glioblastoma tumorigenicity through downregulation of STAT2 associated with the AMPK/SIRT1 pathway

Retraction in: /10.3892/or.2024.8697

  • Authors:
    • Xiangming Liu
    • Jinglong Chen
    • Jinqian Zhang
  • View Affiliations

  • Published online on: April 20, 2017     https://doi.org/10.3892/or.2017.5589
  • Pages: 3387-3396
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

A prospective method of treatment for cancer is to inhibit oncogene signaling pathways with microRNA (miRNA or miR). In the present study, whether the expression of STAT2, AdipoR1/AMPK/SIRT1 pathway of glioma is regulated by miR-3908 was explored. To confirm whether the predicted miR-3908 is matched with STAT2 and AdipoR1, 3'UTR luciferase activity of STAT2 and AdipoR1 was assessed. In the presence of the mimics or inhibitors of miR-3908, cell function of glioma cells, such as proliferation, growth, migration, invasion and apoptosis were analyzed. The expression of AdipoR1 and its downstream AMPK/SIRT1 pathway proteins or STAT2 were examined. Luciferase reporter analysis showed that miR-3908 directly target STAT2 and AdipoR1. miR-3908 suppressed expression of STAT2 or AdipoR1 and downregulated AdipoR1 pathway genes, including AMPK, p-AMPK and SIRT1. miR-3908 inhibited tumorigenicity, migration, growth and invasion in glioma cell lines U251 and U87 as well as increased apoptosis of these cells. The pathways related to tumorigenicity and tumor progression, STAT2 and AdipoR1/AMPK/SIRT1 could be restrained by miR-3908. In conclusion, restoration of miR-3908 expression induced suppression of cancer progression and glioblastoma tumorigenicity. The present study discovered novel tumorigenesis associated with miR-3908, which may represent a new target in treatment for glioblastoma.

Introduction

The primary malignant tumors in CNS (central nervous system) contain 80% of glioma (1). Based on the diagnostic criteria of WHO (World Health Organization) in 2007, four grades are classified: AGI (pilocytic astrocytomas) or grade I, AGII (diffuse astrocytomas) or grade II, AGIII (anaplastic astrocytomas) or grade III and GBM (glioblastomas) or AGIV (grade IV) (2).

Because invasive malignant tumor cells surround normal brain tissue and GBM is diffusely infiltrating, it could not be cured by resection (25). Moreover, the prognosis of GBM after treatment with conventional therapeutic methods is not satisfactory (5,6), the 5-year survival rate is lower than 5%, especially in elderly patients (7). It is a challenge to identify the potential targets for treatment on GBM. The mechanistic insight associated with establishment and progression of tumor was provided according to novel overexpressed or mutated molecules, which might be the new research aims to explore anticancer drugs. In addition, there are specific mutations in cancer cells providing both mechanistic insight associated with tumor establishment and progression, as well as potential targets on the development of new anticancer drugs (8).

The miRNAs (MicroRNAs) are a group of tiny molecules and non-coding RNA, 22–25 nucleotides in length that function on regulation of gene expression at post-transcriptional level. MicroRNAs (miRNAs) control gene expression by pairing with incompletely matching aim sites of the 3 untranslated regions (UTRs) of mRNA, and cause translational repression and/or mRNA destabilization, thereby downregulating the expression of the targeted gene (9). Growing literature indicates the vital function of miRNAs on expression regulation at post-transcriptional level. Moreover, the regulated expression of genes involve in numerous biological processes, especially for different pathogenic disorders, including variety of tumors, especially in GBM (10).

STAT2 belongs to the protein family of STAT (11,12), which play various roles of growth factors and cytokines. The transcription activator could be phosphorylated by kinases related to receptor, and then translocated to nucleus of cells based on heterodimers or homodimers (12).

Based on epidemiologic studies, it is well known that the development of multiple cancers or advanced progression of disease was inversely related to the expression levels of adiponectin (13). Two adiponectin single nucleotide polymorphisms have been shown to increase prostate, colon and breast cancer risk (1416). Adiponectin deficiency through the use of knockout mice has shown accelerated hepatic tumor formation (17) and increased colon formation (18), yet, it delayed tumor growth in a mammary MMTV-PyV mT model due to decreased vascularization and increased apoptosis in early stages of disease (19,20). Tumor promoting effects are likely secondary to initiation, but no clear studies have implicated adiponectin as an initiator of cancer development.

The adiponectin receptors have been detected in gastric, colon, prostate, breast, pancreatic and many other cancers (2125). Adiponectin receptor detection in gastric cancers was associated with longer overall survival (26). Two single nucleotide polymorphisms of adiponectin receptor 1 associate with prostate and breast cancer risk (15,16). Six genetic associations in the AdipoR1 and AdipoR2 genes have been detected in diabetic patients (27). Deletion of the AdipoR1, but not AdipoR2, resulted in promotion of epithelial cell proliferation and increased number of aberrant crypt foci in a murine model (18). Future studies addressing the functional role of each adiponectin receptor in cancer initiation and progression will add a substantial contribution to our understanding and the importance of adiponectin signaling in these diseases.

To assess the role of miR-3908 in glioma cells, we first identified whether miR-3908 sequences exist in the STAT2 and AdipoR1 mRNA, and then evaluated the levels of miR-3908 expression in glioma cells. The present study demonstrated that STAT2 and AdipoR1 are the targets of miR-3908 in glioma cells. STAT2 was associated with AdipoR1/AMPK/SIRT1 pathway signaling. AdipoR1 has also been revealed to associate with cancer risk and to control survival of cancer cells by regulating activation of AMPK and mediating the expression of SIRT1. We, therefore, studied the regulation effect of miR-3908 on regulating activation of AMPK in glioma cells. In particular, the cell behavior effects of miR-3908 are associated with the regulation of AdipoR1/AMPK/SIRT1 signaling pathways.

Materials and methods

Cells

U251 and U87 of Homo sapiens glioma cell lines were purchased from the American Type Culture Collection (ATCC; Manassas, VA, USA). These cells were cultured with Dulbeccos modified Eagles medium (DMEM) contained fetal bovine serum (FBS) and PSN in a humidified environment of 5% CO2.

Agents

TaqMan miRNA assay, Human miRNA precursors, inhibitors of miRNA, Lipofectamine 2000 and mirVana™miRNA isolation kit were all obtained from Invitrogen (Carlsbad, CA, USA). The mitochondrial MTS and apoptosis assay was performed with CellTiter 96® AQueous One Solution Cell Proliferation assay and Caspase-GloH® 3/7 assay (Promega, Madison, WI, USA), respectively. The constructs of luciferase reporter gene contained 3UTR of STAT2 and AdipoR1 was purchased from GeneCopoeia (Rockville, MD, USA).

miRNA transfection

The manufacturers protocol was followed, and Lipofectamine 3000 was performed for transient transfection of miRNA inhibitors or precursors. The mimics of hsa-miR-3908 (HMI1345) (Sigma-Aldrich), a negative control (miR-NC; AM17110) and inhibitors of hsa-miR-3908 (HLTUD1345) (Sigma-Aldrich) were used for the experiments.

QRT-PCR

following the manufacturers instructions, total RNA of cells was extracted with TRIzol reagent (Invitrogen), and then reverse transcription was performed. The mirVana™ miRNA isolation kit (Life Technologies) was used to determine intrinsic expression of miR-3908 from cells with TaqMan miRNA assay (normalized with RNU6B values) based on its manufacturers instructions.

Western blotting

Cells were lysed, and then total proteins were extracted with RIPA lysis buffer. Total proteins were analyzed with electrophoresis method using SDS-PAGE (sodium dodecyl sulfate-polyacrylamide) gel, and then transferred to a PVDF (polyvinylidene fluoride) membrane with 0.45 µm pore size (Roche, Branchburg, NJ, USA). The membranes were blocked with 5% skim milk at room temperature and then washed three times with TBST (28). The membranes were probed with primary antibodies: STAT2 (1:2,000 dilutions; Santa Cruz Biotechnology, Santa Cruz, CA, USA); AdipoR1, AMPK, p-AMPK and SIRT1 (1:3,000 dilution; Cell Signaling Technology, Danvers, MA, USA); or β-actin (1:3000 dilution; Cell Signaling Technology), at 4°C, overnight. Then, they were incubated at room temperature with appropriate secondary antibodies (1:5,000 dilutions; Santa Cruz Biotechnology).

Analysis of proliferation and clonogenicity in glioma cells

The ability of cell proliferation was detected with MTS assay. For clonogenicity assay, cells colonies were stained with crystal violet and fixed with formalin. The different amounts of developed colonies were evaluated.

Cell migration assay

The ability of mitosis in glioma cells were measured with wound scratch assay. Cells were scratched, and then the movement of cells was observed and measured at 48 h. The migration cells were counted and the total number was quantified.

Cell invasion assay

For invasion assay, Matrigel invasion chambers with 4 µm pores were used. Cells were seeded in the upper chambers (coated in Matrigel) at 2×105 concentration in serum-free medium. The chemo-attractant was added to the culture media, and then incubated for 48 h at 37°C. In the top Transwell, non-invaded cells were removed with a cotton swab. After fixing with formalin, those translocated successfully were stained with crystal violet, and then numbered under a microscope.

Apoptosis assays

According to protocol (29), the staining with Annexin V was implemented to determine cell apoptosis. Cells (3×105) were cultured for 48 h, subsequently washed with PBS, and then incubated with Annexin V solution. Cells were washed again and fixed with 1% paraformaldehyde. 7-AAD was used for dual staining of cells. The total number of 7-AAD+Annexin-V double-positive cells was tested with a flow cytometer (29).

Statistical analysis

Data are displayed as mean ± SD. The significant differences between data were estimated with two-tailed Students t-test or one-way ANOVA analysis. For comparison of quantitative data, t-tests or ANOVA (analysis of variance) were conducted between groups. Medians were compared between groups through Kruskal-Wallis ANOVA. SPSS software (version 18.0; SPSS, Inc., Chicago, IL, USA) was used to perform the analyses. P<0.05 was considered statistically significant.

Results

Expression of STAT2 and AdipoR1 was suppressed by miR-3908 in glioma cells

To study if microRNA modulates STAT2 and AdipoR1, or downstream pathway of AdipoR1 in glioma cells, the online software DIANA Tools (microT v4.0) was used to predict miRNAs that target STAT2 and AdipoR1. Of these miRNAs, miR-3908 potentially co-targeted mRNAs of STAT2 and AdipoR1 3UTRs (Fig. 1). Further experiments were carried out with luciferase reporter gene assay to identify whether 3UTRs of STAT2 and AdipoR1 was bonded straight with miR-3908. The results showed that the relative luciferase activities of STAT2 and AdipoR1 3UTR were obviously downregulated in glioma cell line U251 and U87 transfected with miR-3908 (Fig. 2A and B), respectively. However, it was identified that miR-3908 could not directly bond with AMPK and SIRT1 3UTRs (Fig. 2C and D). In glioma cell line U251 and U87, which were only cultured after 48 h, the expression levels of endogenous miR-3908 in these cell lines were detected and are shown in Fig. 2E. The results confirmed that mRNAs of STAT2 and AdipoR1 are straight goals of miR-3908.

Figure 2.

STAT2 and AdipoR1 mRNAs are direct targets of miR-3908 in glioma cell lines U251 and U87. To study if microRNA modulates STAT2 and AdipoR1, or downstream pathway of AdipoR1 in glioma cells, firstly we used DIANA microT v4.0 online software (http://diana.cslab.ece.ntua.gr/) to predict if one or more miRNAs target STAT2 and AdipoR1, the key factor that regulates homeostasis and fatty acid, cholesterol and lipid biosynthesis. Among the miRNAs, miR-3908 was retrieved because miR-3908 potentially co-targeted the 3UTRs of STAT2 and AdipoR1 mRNAs. To further prove whether miR-3908 directly binds with the 3UTRs of STAT2 and AdipoR1, we carried out a 3UTR luciferase reporter assay in miR-3908 transfected glioma cell line U251 and U87, respectively. (A) The relative 3UTR luciferase activities of STAT2 and AdipoR1 significantly decreased in miR-3908 transfected glioma cell line U251 compared to control (P<0.01). (B) The relative 3UTR luciferase activities of STAT2 and AdipoR1 significantly decreased in miR-3908 transfected glioma cell line U87 compared to control (P<0.01).**P<0.01. (C) The relative 3UTR luciferase activities of AMPK and SIRT1 decreased hardly at all in miR-3908 transfected U251 compared to control (P>0.05). (D) The relative 3UTR luciferase activities of AMPK and SIRT1 decreased hardly at all in miR-3908 transfected U87 compared to control (P>0.05). (E) In glioma cell line U251 and U87, which was only cultured after 48 h, the expression levels of endogenous levels of miR-3908 in these cell lines were detected. These results confirmed that STAT2 and AdipoR1 mRNAs are direct targets of miR-3908 in glioma cell lines U251 and U87. The data are presented as means ± SD from three independent experiments. *P<0.05, **P<0.01.

To verify if miR-3908 affects the mRNAs and protein expression of STAT2, AdipoR1, AMPK and SIRT1 in glioma cells, we accomplished relative quantification analyses with qRT-PCR or western blot analysis, respectively (Fig. 3). The results indicated that miR-3908 inhibited the mRNA expression of STAT2 or AdipoR1 in U251 or U87 glioma cell lines, but not AMPK and SIRT1 (Fig. 3A), as well as the proteins expression of STAT2 and AdipoR1 (Fig. 3B). Yet, the protein expression of AMPK and SIRT1 also decreased (Fig. 3B). Moreover, the inhibitors of miR-3908 promoted mRNA expression of STAT2 or AdipoR1 in U251 or U87 glioma cell lines, but not AMPK and SIRT1 (Fig. 3C). Altogether, our results demonstrated that miR-3908 directly regulates the protein expression of STAT2 and AdipoR1 by interacting with its 3UTR.

miR-3908 suppresses cell growth and clonogenicity in glioma cells

For evaluating the possible biotic outcomes induced by miR-3908, we overexpressed or downregulated miR-3908, and then conducted several functional experiments associated with cancer progression and tumorigenicity in U251 and U87 cells. Compared with miR-NC cells, proliferation of U251 (Fig. 4A) and U87 (Fig. 4B) cells were inhibited when they were transfected with miR-3908, respectively. Moreover, the inhibitors of miR-3908 accentuated the process of these glioma cells (Fig. 4).

Furthermore, the colony formation assay revealed that miR-3908 exhibited significantly decreased ability for colony formation compared with the control group in U251 (Fig. 5A and C) and U87 cell lines (Fig. 5B and C) (P<0.01). But, miR-3908 downregulation facilitated significantly the ability of colony formation in both cell lines (Fig. 5). These results suggested that miR-3908 also lowered clonogenicity of U251 and U87 cells (Fig. 5).

miR-3908 suppresses migration and invasion of glioma cells

It was demonstrated that the migration ability was obviously inhibited by miR-3908 compared to control in both cell lines U251 (Fig. 6A and C) and U87 (Fig. 6B and C) (P<0.01). Moreover, downregulation of miR-3908 promoted significantly the ability of migration in both cell lines (Fig. 6). Furthermore, the present study suggested that miR-3908 also inhibited the invasion ability of U251 (Fig. 7A and C) and U87 cells (Fig. 7B and C), respectively. Besides, the inhibitors of miR-3908 enhanced obviously the invasion of the two cell lines (Fig. 7). The capability of invading surrounding tissues and migrating efficiently is a sign of progressive and metastatic cells.

In vitro, the expression of miR-3908 significantly restrained migration (Fig. 6) and invasion (Fig. 7) in U251 and U87 cells, respectively. In turn, as miR-3908 inhibitors block miR-3908 in glioma cells, the ability of cell migration and invasion was enhanced. Based on these data, we could suggest that miR-3908 constrains pathways related to tumorigenicity and tumor progression of glioma.

Apoptosis of glioma cells

To verify whether apoptosis of glioma cells could be induced by miR-3908, dual-staining measurement with 7-AAD and Annexin V-FITC was conducted, as well as microplate assays for caspase activity. Our results showed the increased fractions of apoptotic glioma cells induced by miR-3908 (P<0.01) in U251 (Fig. 8A) and U87 (Fig. 8B). Obviously, miR-3908 also induced caspase-3/-7 activities in U251 (Fig. 8C) and U87 cells (Fig. 8D). Conversely, inhibitors of miR-3908 suppressed these activities (Fig. 8). As these results show, miR-3908 induces glioma cell death.

miR-3908 suppresses cancer progression and tumorigenicity of glioma through STAT2 and AdipoR1/AMPK/SIRT1 pathway

The expression of STAT2, AdipoR1, AMPK, p-AMPK and SIRT1 proteins was detected by western blot analysis. Upregulation of miR-3908 in glioma cell lines U251 (Fig. 9A) and U87 (Fig. 9B) significantly inhibited the expression of STAT2, AdipoR1, AMPK, p-AMPK and SIRT1. Furthermore, treatment with inhibitors of miR-3908 in glioma cells significantly increased the expression of STAT2, AdipoR1, AMPK, p-AMPK and SIRT1 (Fig. 8). It showed that miR-3908 suppressed tumorigenicity and cancer progression of glioma through restraining STAT2 and AdipoR1/AMPK/SIRT1 pathways.

Discussion

GBM is the primary and deadliest malignancy of the brain and extremely common (3032) with near highest fatality in two years after diagnosis (33,34). The cellular capacity, which reconstitute the tumor entirely, and tumorigenicity of cell subpopulations are the major reasons of therapeutic failure (31). Growing evidence indicates cell subpopulations of GBM induce its lethality based on the properties of its tumorigenicity and self-renewal (35). It demonstrated that the primary tumor could generate phenocopies (36,37). Key transcription factors could be regulated with epigenetic modifications and then control transformation between therapeutic developments and tumorigenic states of GBM (31).

MicroRNAs suppress translation of messenger RNA or promote degradation of it (38,39) and are vital regulators for oncogenesis. Moreover, their regulations on signaling in tumor cells are sophisticated. Most expression of microRNAs is inhibited in tumors (4043). However, some amplified expression of microRNAs act as oncogenic in variety of tumors (4346). The essential signaling activation of STATs and NF-κB pathways induce regulation of Notch pathway genes in glioma, which was identified as the novel targets for treatment on GBM (47). STAT2 plays a vital role on carcinogenesis of skin and colon, and could activate the signaling pathway of oncogenic STAT3 (48).

In the present study, we studied if microRNA modulates STAT2 and AdipoR1, or downstream pathway of AdipoR1 in glioma cells. The online software DIANA Tools (microT v4.0) were used to predict miRNAs that target STAT2 and AdipoR1. Within these miRNAs, miR-3908 potentially co-targeted mRNAs of STAT2 and AdipoR1 3UTRs. Then, the 3UTR luciferase reporter assays were carried out. It identified that miR-3908 could directly bind with 3UTRs of STAT2 and AdipoR1, but not AMPK and SIRT1. Moreover, miR-3908 directly regulated the protein expression of STAT2 and AdipoR1 by interacting with its 3UTR. The protein expression of AMPK and SIRT1 was regulated indirectly by STAT2 and AdipoR1 in glioma cells. Why the protein expression levels of AMPK and SIRT1 are decreased, but their mRNA levels are not been affected? It may be due to the change in the translation of these mRNAs induced by mir-3908. mir-3908 may act as translational modulators on mRNAs of AMPK and SIRT1. It is known to influence the evolution and stability of AMPK and SIRT1 mRNAs but they can also affect translation efficiency.

Furthermore, the possible biological outcomes induced by miR-3908 in glioma cell lines U251 and U87 were investigated with the mimics and inhibitors of miR-3908. Several functional experiments associated with cancer progression and tumorigenicity was performed. Our results demonstrated that, compared with miR-NC cells, proliferation and colony formation of U251 and U87 cells would be suppressed when they were transfected with miR-3908. Moreover, the inhibitors of miR-3908 accentuated these processes of glioma cells. These results suggested that miR-3908 inhibited the proliferation of U251 and U87 cells, also lowering their clonogenicity.

The migration experiment and wound scratch assay were also conducted. These results demonstrated that miR-3908 obviously repressed the invasion and migration ability compared to control in the cell lines U251 and U87. Moreover, downregulation of miR-3908 promoted significantly the ability of invasion and migration in U251 and U87 cells. The capability of invading surrounding tissues and migrating efficiently is a sign of progressive and metastatic cells. In vitro, the expression of miR-3908 significantly restrained migration (Fig. 6) and invasion (Fig. 7) in U251 and U87 cells, respectively. In turn, as miR-3908 inhibitors block miR-3908 in glioma cells, the ability of cell migration and invasion was enhanced. These data demonstrated that miR-3908 constrains pathways concerned with cancer progression and tumorigenicity in glioma cells.

To verify whether apoptosis of glioma cells could be induced by miR-3908, Dual-staining measurement with 7-AAD and Annexin V-FITC was conducted, as well as the microplate assays for caspase activity. Our results showed increased fractions of apoptotic glioma cells induced by miR-3908 (P<0.01) in U251 (Fig. 8A) and U87 (Fig. 8B). Obviously, miR-3908 also induced caspase-3/7 activities in U251 (Fig. 8C) and U87 cells (Fig. 8D). Conversely, inhibitors of miR-3908 suppressed these activities (Fig. 7). As the results show, miR-3908 induces glioma cell apoptosis.

AMPK is the vital energy sensor responsible for the homeostasis of cellular energy (49). While cellular energy is exhausted by hypoxia, starvation, stress or other reasons, intracellular AMP increase, and then activates AMPK through α-subunit phosphorylation caused by upstream kinases (50). Activated AMPK stimulate pathways of energy-producing catabolics (such as glucose transport and fatty acid oxidation) and suppress anabolic pathways of energy-consuming, and then homeostasis of cellular energy is restored. Sirtuins are deacetylases dependent on NAD+, and a response to shift of stress or energy metabolism. SIRT1 (sirtuin 1) is almost characterized in this family. AMPK and SIRT1 could emphatically manage the activities of each other (51). In liver or skeletal muscle, adiponectin modulates energy metabolism by the AMPK/SIRT1 pathway (52). Osmotin regulates AdipoR1, and then activates SIRT1 and AMPK to block production of Aβ (53).

The expression of STAT2, AdipoR1, AMPK, p-AMPK and SIRT1 proteins was detected by western blot analysis. Upregulation of miR-3908 in glioma cell lines U251 (Fig. 9A) and U87 (Fig. 9B) significantly inhibited the expression of STAT2, AdipoR1, AMPK, p-AMPK and SIRT1. Furthermore, treatment with inhibitors of miR-3908 in glioma cells significantly increased the expression of STAT2, AdipoR1, AMPK, p-AMPK and SIRT1 (Fig. 8). It showed that miR-3908 suppressed tumorigenicity and cancer progression of glioma through restraining STAT2 and AdipoR1/AMPK/SIRT1 pathways.

In conclusion, STAT2 and AdipoR1 are direct targets of miR-3908. miR-3908 could inhibit tumorigenicity and tumor progression of glioma through suppressing STAT2 and AdipoR1/AMPK/SIRT1 pathways. When miR-3908 is restored it will induce suppression of cancer progression and glioblastoma tumorigenicity. miR-3908 is a novel discovery of a research target for treatment in glioblastoma.

Acknowledgements

The present study was supported by grants from the National Natural Science Foundation of China (nos. 30600524 and 81341067).

References

1 

Dolecek TA, Propp JM, Stroup NE and Kruchko C: CBTRUS statistical report: Primary brain and central nervous system tumors diagnosed in the United States in 2005–2009. Neuro Oncol. 14 Suppl 5:v1–v49. 2012. View Article : Google Scholar : PubMed/NCBI

2 

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

3 

Reifenberger G and Collins VP: Pathology and molecular genetics of astrocytic gliomas. J Mol Med (Berl). 82:656–670. 2004. View Article : Google Scholar : PubMed/NCBI

4 

Ohgaki H and Kleihues P: Genetic alterations and signaling pathways in the evolution of gliomas. Cancer Sci. 100:2235–2241. 2009. View Article : Google Scholar : PubMed/NCBI

5 

Ohgaki H and Kleihues P: Genetic profile of astrocytic and oligodendroglial gliomas. Brain Tumor Pathol. 28:177–183. 2011. View Article : Google Scholar : PubMed/NCBI

6 

Ohgaki H and Kleihues P: Genetic pathways to primary and secondary glioblastoma. Am J Pathol. 170:1445–1453. 2007. View Article : Google Scholar : PubMed/NCBI

7 

Ostrom QT, Gittleman H, Farah P, Ondracek A, Chen Y, Wolinsky Y, Stroup NE, Kruchko C and Barnholtz-Sloan JS: CBTRUS statistical report: Primary brain and central nervous system tumors diagnosed in the United States in 2006–2010. Neuro-oncol. 15 Suppl 2:ii1–ii56. 2013. View Article : Google Scholar : PubMed/NCBI

8 

Chung S and Nakamura Y: MELK inhibitor, novel molecular targeted therapeutics for human cancer stem cells. Cell Cycle. 12:1655–1656. 2013. View Article : Google Scholar : PubMed/NCBI

9 

Au Yeung CL, Co NN, Tsuruga T, Yeung TL, Kwan SY, Leung CS, Li Y, Lu ES, Kwan K, Wong KK, et al: Exosomal transfer of stroma-derived miR21 confers paclitaxel resistance in ovarian cancer cells through targeting APAF1. Nat Commun. 7:111502016. View Article : Google Scholar : PubMed/NCBI

10 

Skog J, Würdinger T, van Rijn S, Meijer DH, Gainche L, Sena-Esteves M, Curry WT Jr, Carter BS, Krichevsky AM and Breakefield XO: Glioblastoma microvesicles transport RNA and proteins that promote tumour growth and provide diagnostic biomarkers. Nat Cell Biol. 10:1470–1476. 2008. View Article : Google Scholar : PubMed/NCBI

11 

Yan R, Qureshi S, Zhong Z, Wen Z and Darnell JE Jr: The genomic structure of the STAT genes: Multiple exons in coincident sites in Stat1 and Stat2. Nucleic Acids Res. 23:459–463. 1995. View Article : Google Scholar : PubMed/NCBI

12 

Wang J, Pham-Mitchell N, Schindler C and Campbell IL: Dysregulated Sonic hedgehog signaling and medulloblastoma consequent to IFN-alpha-stimulated STAT2-independent production of IFN-gamma in the brain. J Clin Invest. 112:535–543. 2003. View Article : Google Scholar : PubMed/NCBI

13 

Barb D, Williams CJ, Neuwirth AK and Mantzoros CS: Adiponectin in relation to malignancies: A review of existing basic research and clinical evidence. Am J Clin Nutr. 86:s858–s866. 2007.PubMed/NCBI

14 

Kaklamani VG, Wisinski KB, Sadim M, Gulden C, Do A, Offit K, Baron JA, Ahsan H, Mantzoros C and Pasche B: Variants of the adiponectin (ADIPOQ) and adiponectin receptor 1 (ADIPOR1) genes and colorectal cancer risk. JAMA. 300:1523–1531. 2008. View Article : Google Scholar : PubMed/NCBI

15 

Kaklamani VG, Sadim M, Hsi A, Offit K, Oddoux C, Ostrer H, Ahsan H, Pasche B and Mantzoros C: Variants of the adiponectin and adiponectin receptor 1 genes and breast cancer risk. Cancer Res. 68:3178–3184. 2008. View Article : Google Scholar : PubMed/NCBI

16 

Kaklamani V, Yi N, Zhang K, Sadim M, Offit K, Oddoux C, Ostrer H, Mantzoros C and Pasche B: Polymorphisms of ADIPOQ and ADIPOR1 and prostate cancer risk. Metabolism. 60:1234–1243. 2011. View Article : Google Scholar : PubMed/NCBI

17 

Kamada Y, Matsumoto H, Tamura S, Fukushima J, Kiso S, Fukui K, Igura T, Maeda N, Kihara S, Funahashi T, et al: Hypoadiponectinemia accelerates hepatic tumor formation in a nonalcoholic steatohepatitis mouse model. J Hepatol. 47:556–564. 2007. View Article : Google Scholar : PubMed/NCBI

18 

Fujisawa T, Endo H, Tomimoto A, Sugiyama M, Takahashi H, Saito S, Inamori M, Nakajima N, Watanabe M, Kubota N, et al: Adiponectin suppresses colorectal carcinogenesis under the high-fat diet condition. Gut. 57:1531–1538. 2008. View Article : Google Scholar : PubMed/NCBI

19 

Landskroner-Eiger S, Qian B, Muise ES, Nawrocki AR, Berger JP, Fine EJ, Koba W, Deng Y, Pollard JW and Scherer PE: Proangiogenic contribution of adiponectin toward mammary tumor growth in vivo. Clin Cancer Res. 15:3265–3276. 2009. View Article : Google Scholar : PubMed/NCBI

20 

Denzel MS, Hebbard LW, Shostak G, Shapiro L, Cardiff RD and Ranscht B: Adiponectin deficiency limits tumor vascularization in the MMTV-PyV-mT mouse model of mammary cancer. Clin Cancer Res. 15:3256–3264. 2009. View Article : Google Scholar : PubMed/NCBI

21 

Ishikawa M, Kitayama J, Yamauchi T, Kadowaki T, Maki T, Miyato H, Yamashita H and Nagawa H: Adiponectin inhibits the growth and peritoneal metastasis of gastric cancer through its specific membrane receptors AdipoR1 and AdipoR2. Cancer Sci. 98:1120–1127. 2007. View Article : Google Scholar : PubMed/NCBI

22 

Dalamaga M, Migdalis I, Fargnoli JL, Papadavid E, Bloom E, Mitsiades N, Karmaniolas K, Pelecanos N, Tseleni-Balafouta S, Dionyssiou-Asteriou A, et al: Pancreatic cancer expresses adiponectin receptors and is associated with hypoleptinemia and hyperadiponectinemia: A case-control study. Cancer Causes Control. 20:625–633. 2009. View Article : Google Scholar : PubMed/NCBI

23 

Drew JE, Farquharson AJ, Padidar S, Duthie GG, Mercer JG, Arthur JR, Morrice PC and Barrera LN: Insulin, leptin, and adiponectin receptors in colon: Regulation relative to differing body adiposity independent of diet and in response to dimethylhydrazine. Am J Physiol Gastrointest Liver Physiol. 293:G682–G691. 2007. View Article : Google Scholar : PubMed/NCBI

24 

Mistry T, Digby JE, Chen J, Desai KM and Randeva HS: The regulation of adiponectin receptors in human prostate cancer cell lines. Biochem Biophys Res Commun. 348:832–838. 2006. View Article : Google Scholar : PubMed/NCBI

25 

Takahata C, Miyoshi Y, Irahara N, Taguchi T, Tamaki Y and Noguchi S: Demonstration of adiponectin receptors 1 and 2 mRNA expression in human breast cancer cells. Cancer Lett. 250:229–236. 2007. View Article : Google Scholar : PubMed/NCBI

26 

Barresi V, Grosso M, Giuffrè G, Tuccari G and Barresi G: The expression of adiponectin receptors Adipo-R1 and Adipo-R2 is associated with an intestinal histotype and longer survival in gastric carcinoma. J Clin Pathol. 62:705–709. 2009. View Article : Google Scholar : PubMed/NCBI

27 

Crimmins NA and Martin LJ: Polymorphisms in adiponectin receptor genes ADIPOR1 and ADIPOR2 and insulin resistance. Obes Rev. 8:419–423. 2007. View Article : Google Scholar : PubMed/NCBI

28 

Hosoi T, Hyoda K, Okuma Y, Nomura Y and Ozawa K: Akt up- and down-regulation in response to endoplasmic reticulum stress. Brain Res. 1152:27–31. 2007. View Article : Google Scholar : PubMed/NCBI

29 

Zhang J, Sun Q, Bo J, Huang R, Zhang M, Xia Z, Ju L and Xiang G: Single-walled carbon nanohorn (SWNH) aggregates inhibited proliferation of human liver cell lines and promoted apoptosis, especially for hepatoma cell lines. Int J Nanomed. 9:759–773. 2014. View Article : Google Scholar

30 

Atkins C, Liu Q, Minthorn E, Zhang SY, Figueroa DJ, Moss K, Stanley TB, Sanders B, Goetz A, Gaul N, et al: Characterization of a novel PERK kinase inhibitor with antitumor and antiangiogenic activity. Cancer Res. 73:1993–2002. 2013. View Article : Google Scholar : PubMed/NCBI

31 

Kozono D, Li J, Nitta M, Sampetrean O, Gonda D, Kushwaha DS, Merzon D, Ramakrishnan V, Zhu S, Zhu K, et al: Dynamic epigenetic regulation of glioblastoma tumorigenicity through LSD1 modulation of MYC expression. Proc Natl Acad Sci USA. 112:pp. E4055–E4064. 2015; View Article : Google Scholar : PubMed/NCBI

32 

Ng K, Kim R, Kesari S, Carter B and Chen CC: Genomic profiling of glioblastoma: Convergence of fundamental biologic tenets and novel insights. J Neurooncol. 107:1–12. 2012. View Article : Google Scholar : PubMed/NCBI

33 

Wen PY and Kesari S: Malignant gliomas in adults. N Engl J Med. 359:492–507. 2008. View Article : Google Scholar : PubMed/NCBI

34 

Stupp R, Mason WP, van den Bent MJ, Weller M, Fisher B, Taphoorn MJ, Belanger K, Brandes AA, Marosi C, Bogdahn U, et al: European Organisation for Research and Treatment of Cancer Brain Tumor and Radiotherapy Groups; National Cancer Institute of Canada Clinical Trials Group: Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med. 352:987–996. 2005. View Article : Google Scholar : PubMed/NCBI

35 

Bartek J Jr, Ng K, Bartek J, Fischer W, Carter B and Chen CC: Key concepts in glioblastoma therapy. J Neurol Neurosurg Psychiatry. 83:753–760. 2012. View Article : Google Scholar : PubMed/NCBI

36 

Singh SK, Hawkins C, Clarke ID, Squire JA, Bayani J, Hide T, Henkelman RM, Cusimano MD and Dirks PB: Identification of human brain tumour initiating cells. Nature. 432:396–401. 2004. View Article : Google Scholar : PubMed/NCBI

37 

Zhou BB, Zhang H, Damelin M, Geles KG, Grindley JC and Dirks PB: Tumour-initiating cells: Challenges and opportunities for anticancer drug discovery. Nat Rev Drug Discov. 8:806–823. 2009. View Article : Google Scholar : PubMed/NCBI

38 

Mukherji S, Ebert MS, Zheng GX, Tsang JS, Sharp PA and van Oudenaarden A: MicroRNAs can generate thresholds in target gene expression. Nat Genet. 43:854–859. 2011. View Article : Google Scholar : PubMed/NCBI

39 

Guo H, Ingolia NT, Weissman JS and Bartel DP: Mammalian microRNAs predominantly act to decrease target mRNA levels. Nature. 466:835–840. 2010. View Article : Google Scholar : PubMed/NCBI

40 

Lu J, Getz G, Miska EA, Alvarez-Saavedra E, Lamb J, Peck D, Sweet-Cordero A, Ebert BL, Mak RH, Ferrando AA, et al: MicroRNA expression profiles classify human cancers. Nature. 435:834–838. 2005. View Article : Google Scholar : PubMed/NCBI

41 

Martello G, Rosato A, Ferrari F, Manfrin A, Cordenonsi M, Dupont S, Enzo E, Guzzardo V, Rondina M, Spruce T, et al: A MicroRNA targeting dicer for metastasis control. Cell. 141:1195–1207. 2010. View Article : Google Scholar : PubMed/NCBI

42 

Kumar MS, Lu J, Mercer KL, Golub TR and Jacks T: Impaired microRNA processing enhances cellular transformation and tumorigenesis. Nat Genet. 39:673–677. 2007. View Article : Google Scholar : PubMed/NCBI

43 

Volinia S, Galasso M, Costinean S, Tagliavini L, Gamberoni G, Drusco A, Marchesini J, Mascellani N, Sana ME, Abu Jarour R, et al: Reprogramming of miRNA networks in cancer and leukemia. Genome Res. 20:589–599. 2010. View Article : Google Scholar : PubMed/NCBI

44 

Darido C, Georgy SR, Wilanowski T, Dworkin S, Auden A, Zhao Q, Rank G, Srivastava S, Finlay MJ, Papenfuss AT, et al: Targeting of the tumor suppressor GRHL3 by a miR-21-dependent proto-oncogenic network results in PTEN loss and tumorigenesis. Cancer Cell. 20:635–648. 2011. View Article : Google Scholar : PubMed/NCBI

45 

Olive V, Bennett MJ, Walker JC, Ma C, Jiang I, Cordon-Cardo C, Li QJ, Lowe SW, Hannon GJ and He L: miR-19 is a key oncogenic component of mir-17-92. Genes Dev. 23:2839–2849. 2009. View Article : Google Scholar : PubMed/NCBI

46 

Conkrite K, Sundby M, Mukai S, Thomson JM, Mu D, Hammond SM and MacPherson D: miR-17~92 cooperates with RB pathway mutations to promote retinoblastoma. Genes Dev. 25:1734–1745. 2011. View Article : Google Scholar : PubMed/NCBI

47 

Garner JM, Fan M, Yang CH, Du Z, Sims M, Davidoff AM and Pfeffer LM: Constitutive activation of signal transducer and activator of transcription 3 (STAT3) and nuclear factor κB signaling in glioblastoma cancer stem cells regulates the Notch pathway. J Biol Chem. 288:26167–26176. 2013. View Article : Google Scholar : PubMed/NCBI

48 

Gamero AM, Young MR, Mentor-Marcel R, Bobe G, Scarzello AJ, Wise J and Colburn NH: STAT2 contributes to promotion of colorectal and skin carcinogenesis. Cancer Prev Res (Phila). 3:495–504. 2010. View Article : Google Scholar : PubMed/NCBI

49 

Hardie DG: AMP-activated protein kinase: An energy sensor that regulates all aspects of cell function. Genes Dev. 25:1895–1908. 2011. View Article : Google Scholar : PubMed/NCBI

50 

Viollet B, Lantier L, Devin-Leclerc J, Hebrard S, Amouyal C, Mounier R, Foretz M and Andreelli F: Targeting the AMPK pathway for the treatment of Type 2 diabetes. Front Biosci (Landmark Ed). 14:3380–3400. 2009. View Article : Google Scholar : PubMed/NCBI

51 

Ruderman NB, Xu XJ, Nelson L, Cacicedo JM, Saha AK, Lan F and Ido Y: AMPK and SIRT1: A long-standing partnership? Am J Physiol Endocrinol Metab. 298:E751–E760. 2010. View Article : Google Scholar : PubMed/NCBI

52 

Shen Z, Liang X, Rogers CQ, Rideout D and You M: Involvement of adiponectin-SIRT1-AMPK signaling in the protective action of rosiglitazone against alcoholic fatty liver in mice. Am J Physiol Gastrointest Liver Physiol. 298:G364–G374. 2010. View Article : Google Scholar : PubMed/NCBI

53 

Shah SA, Yoon GH, Chung SS, Abid MN, Kim TH, Lee HY and Kim MO: Novel osmotin inhibits SREBP2 via the AdipoR1/AMPK/SIRT1 pathway to improve Alzheimer's disease neuropathological deficits. Mol Psychiatry. 22:407–416. 2016. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

June-2017
Volume 37 Issue 6

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
Liu X, Chen J and Zhang J: AdipoR1-mediated miR-3908 inhibits glioblastoma tumorigenicity through downregulation of STAT2 associated with the AMPK/SIRT1 pathway Retraction in /10.3892/or.2024.8697. Oncol Rep 37: 3387-3396, 2017
APA
Liu, X., Chen, J., & Zhang, J. (2017). AdipoR1-mediated miR-3908 inhibits glioblastoma tumorigenicity through downregulation of STAT2 associated with the AMPK/SIRT1 pathway Retraction in /10.3892/or.2024.8697. Oncology Reports, 37, 3387-3396. https://doi.org/10.3892/or.2017.5589
MLA
Liu, X., Chen, J., Zhang, J."AdipoR1-mediated miR-3908 inhibits glioblastoma tumorigenicity through downregulation of STAT2 associated with the AMPK/SIRT1 pathway Retraction in /10.3892/or.2024.8697". Oncology Reports 37.6 (2017): 3387-3396.
Chicago
Liu, X., Chen, J., Zhang, J."AdipoR1-mediated miR-3908 inhibits glioblastoma tumorigenicity through downregulation of STAT2 associated with the AMPK/SIRT1 pathway Retraction in /10.3892/or.2024.8697". Oncology Reports 37, no. 6 (2017): 3387-3396. https://doi.org/10.3892/or.2017.5589