scholarly journals Serum lncRNA-ANRIL and SOX9 expression levels in glioma patients and their relationship with poor prognosis

2021 ◽  
Vol 19 (1) ◽  
Author(s):  
Youlu Sun ◽  
Yuesong Jing ◽  
Yuxin Zhang

Abstract Background lncRNA-CDKN2B antisense RNA 1 (ANRIL) and SRY-box transcription factor 9 (SOX9) has abnormal expression in many tumors including glioma, but the underlying molecular mechanism is unclear. This study set out to investigate the serum lncRNA-ANRIL and SOX9 levels in glioma patients and their effects on prognosis. Methods We enrolled 142 glioma patients admitted to our hospital from May 2014 to May 2016 into the research group (RG) and 120 healthy subjects receiving concurrent physical examinations into the control group (CG). Fasting peripheral blood (4 mL each) was sampled from subjects from the two groups. Using the quantitative real-time polymerase chain reaction (qRT-PCR), lncRNA-ANRIL and SOX9 were measured to explore their values in the early diagnosis of glioma. Patients from RG were followed up for 3 years to analyze the influence of lncRNA-ANRIL and SOX9 on patient prognosis. We purchased glioma cell lines U251 and U87 and grouped them according to the transfection of different plasmids. We conducted CCK8 assay to test cell proliferation, Transwell assay to test cell invasion, the flow cytometry to test cell apoptosis, and Western Blot assay to measure bcl-2 and bax protein levels. Results ANRIL and SOX9 were evidently higher in RG than in CG (P<0.01). The receiver operating characteristic (ROC) curve revealed that the diagnostic sensitivity of ANRIL combined with SOX9 for glioma was 81.62%, and the specificity was 90.83% (P<0.01). ANRIL and SOX9 were closely related to tumor grade, tumor diameter, distant metastasis, and family history of glioma (P<0.01). In total, 135 patients were successfully followed up (95.07%). Patients with high levels of ANRIL and SOX9 had a markedly poorer prognosis than those with low levels (P<0.05). ANRIL and SOX9 were markedly higher in glioma cell lines (U251 and U87) than in normal brain cells (P<0.01). The proliferation and invasion of U251 cells were notably reduced after the transfection of ANRIL and SOX9 inhibitory sequences (P<0.01), but the apoptosis was notably increased (P<0.01). Bcl-2 expression was markedly increased in lncRNA-ANRIL-inhibitor and SOX9-inhibitor (P<0.01), while bax expression was markedly reduced in lncRNA-ANRIL-inhibitor and SOX9-inhibitor (P<0.01). Conclusion lncRNA-ANRIL and SOX9 levels were higher in glioma patients than in healthy people. High-lncRNA-ANRIL and SOX9 levels were strongly associated with unfavorable prognosis of patients. The testing of biological behaviors revealed that lncRNA-ANRIL and SOX9 worked as tumor-promoting genes in glioma.

2019 ◽  
Vol 21 (Supplement_6) ◽  
pp. vi36-vi37
Author(s):  
Mitsutoshi Nakada ◽  
Jiakang Zhang ◽  
Taskuya Furuta ◽  
Shabierjiang Jiapaer ◽  
Sho Tamai ◽  
...  

Abstract BACKGROUND Among potential glioblastoma (GBM) blood biomarkers that we identified recently, we focused on gelsolin (GSN), a key regulator of actin filament disassembly. GSN was significantly lower in the blood of patients with GBM than in that of healthy controls. In this study, we analyzed the function of GSN and identified microRNAs (miRs) involved in GSN expression in GBM. METHODS QRT-PCR and western blot were introduced to evaluate the expression level of GSN in normal brain and GBM tissue. The localization of GSN was examined by immunohistochemistry and immunocytochemistry using human samples. The association between the expression level of GSN and progression free survival (PFS) /overall survival (OS) in GBM was assessed by Kaplan-Meier analysis. The function of GSN and its signal transduction in glioma cell lines were analyzed using small interfering RNA (siRNA) knockdown of GSN. Additionally, miRs controlling GSN expression were retrieved from databases of miRs, and miRs related to GSN expression were identified in GBM tissues. RESULTS The expression level of GSN was significantly lower in GBM tissues compared to normal brains. Normal astrocytes mainly expressed GSN. High expressor of GSN showed longer PFS and OS than low expressor. Proliferation and invasion in glioma cell lines were significantly promoted by siRNA for GSN accompanied with the activation of glycogen synthase kinase 3β. In GBM tissues, the expression levels of GSN and miR-654-5p, 450b-5p showed an inverse correlation. The inhibitor for miR-654-5p and miR-450b-5p accelerated GSN expression resulting reduction of proliferation. CONCLUSION GSN plays a role as suppressor of proliferation and invasion in GBM. miR-654-5p and miR-450b-5p which control GSN expression can be targets against GBM.


2014 ◽  
Vol 2014 ◽  
pp. 1-7 ◽  
Author(s):  
Suning Huang ◽  
Gang Chen ◽  
Yiwu Dang ◽  
Long-Hua Chen

Background.Overexpression of decoy receptor 3 (DcR3) have been reported in various classes of malignancies. However, its expression and clinicopathological contribution in gliomas has not been fully elucidated.Objective.To explore the expression and clinical significance of DcR3 protein in relation to tumor cell differentiation and proliferation in glioma cell lines and tissues.Methods.One hundred and twenty-five samples of glioma patients and 18 cases of normal brain tissues were recruited. The expression of DcR3 protein was detected using immunohistochemistry. Tumor differentiation was assessed by histologic characters and the status of glial fibrillary acidic protein (GFAP). Tumor cell labeling indexes (LIs) of Ki-67 and PCNA were also obtained. The relationship between the DcR3 level and clinicopathological features was investigated, including tumor differentiation, LIs, and survival. Meanwhile, the expression of DcR3 protein was also measured in the supernatants of 8 glioma cell lines and glioma cells freshly prepared from 8 human glioblastoma specimens by using western blot.Results.The level of DcR3 protein in gliomas was significantly higher than that in normal brain tissues (P<0.01). DcR3 expression showed positive correlations with tumor pathological grade(r=0.621,P<0.01)and negative with GFAP expression (r=-0.489,P<0.01). Furthermore, there were positive correlations between DcR3 expression and Ki-67, PCNA LIs (r=0.529,P<0.01;r=0.556,P<0.01). The survival in the DcR3 negative group was 50 ± 1.79 months, longer than that of the DcR3 positive group (48.36 ± 2.90), however, without significance(P=0.149). Different levels of DcR3 could also be detected in the culturing supernatants of all the 8 glioma cell lines and glioma cells freshly obtained from 8 human glioblastoma specimens.Conclusions.The overexpression of DcR3 might play a crucial role in the tumorigenesis, differentiation, and proliferation of glioma.


2021 ◽  
Author(s):  
Wenjin Qiu ◽  
Xiaomin Cai ◽  
Kaya Xu ◽  
Shibin Song ◽  
Zumu Xiao ◽  
...  

Abstract Background Regenerating liver phosphatase 1 (PRL1) is an established oncogene in various cancers, although its biological functions and the underlying mechanisms in glioblastoma multiforme (GBM) remain unclear. Methods PRL1 expression levels were analyzed in glioma tissues and cell lines. Multiple glioma cell lines were transfected with PRL1-overexpressing and shRNA constructs. In vitro proliferation, migration and invasion assays were conducted. Western blot and ubiquitylation assays were performed for molecular and mechanistic analyses. PRL1 expression levels were correlated with downstream ubiquitin pathway and clinical parameters using archival GBM samples. Results PRL1 was significantly upregulated in glioma tissues and cell lines, and positively correlated with the tumor grade. Ectopic expression of PRL1 in glioma cell lines significantly enhanced their tumorgenicity and invasion both in vitro and in vivo by promoting EMT. Conversely, knocking down PRL1 blocked EMT in the GBM cells, and inhibited their invasion, migration and tumorigenic growth. PRL1 also stabilized Snail2 through deubiquitination by activating USP36. Snail2 was identified as a crucial mediator of the oncogenic effects of PRL1 in GBM. Finally, PRL1 protein levels were positively correlated with that of Snail2 and predicted poor outcome of GBMs. Conclusions PRL1 promotes GBM progression by activating USP36-mediated Snail2 deubiquitination. This novel PRL1/USP36/Snail2 axis may be a promising therapeutic target for glioblastoma.


2018 ◽  
Vol 19 (11) ◽  
pp. 3353 ◽  
Author(s):  
Yi-Hsuan Chen ◽  
Dueng-Yuan Hueng ◽  
Wen-Chiuan Tsai

Proteolipid protein 2 (PLP2), a membrane protein of the endoplasmic reticulum, is related to tumor proliferation and metastasis in some human cancers, but not in gliomas. First, we performed western-blot analysis, real-time quantitative PCR and immunohistochemical stains to detect PLP2 expression in 4 glioma cell lines and human glioma tissues. In addition, we used small interfering RNA (SiPLP2) and short hairpin RNA (shPLP2) to knockdown PLP2 expression in GBM8401 and LN229 glioma cell lines. After then, the alteration of PLP2 suppressed glioma cells behavior were examined by cell proliferation, wound healing, cell invasion, and colonies formation assays. Finally, the possible mechanism of PLP2 was analyzed by detecting the expression of the proteins related to cell-cycle checkpoints, cell-proliferative signaling factors, and cell-matrix interaction. Compared with normal brain cell lysates and mRNA, all glioma cell lines displayed PLP2 protein and mRNA overexpression. Besides, higher PLP2 IHC staining significantly correlated with more advanced tumor grades and poorer prognosis in human gliomas. Both siPLP2 transfected gliomas showed a clear inhibition of glioma cell proliferation, migration, and invasion as well as down-regulating p-p38, p-ERK, MMP-2, and MMP-9 expression. In conclusion, we successfully demonstrated that PLP2 overexpression played an oncogenic role in glioma development and aggressive tumor behavior.


2016 ◽  
Vol 70 (2) ◽  
pp. 162-167 ◽  
Author(s):  
Wen-Chiuan Tsai ◽  
Dueng-Yuan Hueng ◽  
Shin Nieh ◽  
Hong-Wei Gao

AimsAlthough ARID4B is known to promote tumour metastasis in breast cancer and inhibit transformation and progression in leukaemia, the possible effect of ARID4B on primary brain tumours (PBTs) is not well characterised. We tested the hypothesis that expression of ARID4B correlates with WHO grade and survival in patients with PBTs.MethodsWestern blot analysis was performed on protein lysates prepared from normal brain tissue and glioma cell lines (U87MG, LN229, GBM8401 and U118MG). Subsequently, immunohistochemical analysis of ARID4B was performed on 2 tissue microarrays, including 12 normal brain tissues, 63 meningiomas with different subtypes, 232 gliomas of various grades and degrees of differentiation, 8 central neurocytomas and 4 chordomas. The ARID4B immunostaining score was calculated by multiplying the intensity score by the percentage of tumour cells expressing ARID4B.ResultsIn vitro, ARID4B protein expression was increased in some glioma cell lines. In addition, the average ARID4B immunostaining score was 38.03, 79.09, 129.76 and 119.32, respectively, in gliomas of WHO grade I, II, III and IV. Higher ARID4B immunostaining score was significantly correlated with more advanced WHO grade of gliomas (p=7.4×10–6) and meningiomas. Finally, higher ARID4B expression tended to the shorter survival rates, but did not reach statistical significance.ConclusionsARID4B overexpression presented in most of PBTs, rather than non-neoplastic brain tissue, and correlated with WHO grades in meningiomas and gliomas. Therefore, ARID4B is a satisfactory biomarker to highlight tumour component and predict tumour behaviour in primary brain neoplasms.


2021 ◽  
Author(s):  
Zijin Zhao ◽  
Miaomiao Liu ◽  
Wenyong Long ◽  
Jian Yuan ◽  
Haoyu Li ◽  
...  

Abstract Background Long non-coding RNA (lncRNA) CRNDE is highly expressed in glioma acting as an oncogene in gliomas. However, the regulatory roles of CRNDE in Temozolomide (TMZ) chemoresistance to glioblastoma multiforme (GBM) still are poorly understood. Therefore, we intended to explore the role and possible mechanism of CRNDE in TMZ-induced chemoresistance in GBM. Methods Firstly, we tested the expression level of CRNDE in glioma cell lines and in 30 cases of normal brain tissues and 58 cases of glioma tissue specimens by qRT-PCR. Meanwhile, the correlation between CRNDE level and the clinicopathological characteristics and survival time of patients were analyzed. Then, the CRNDE expression in various glioma cell lines was detected, and CRNDE knockdown cell models were constructed. Subsequently, to explore the effect of CRNDE on chemosensitivity to TMZ, cell viability was detected by the CCK-8 assay and IC50 values, and cell proliferation was detected by cell clone assay and EdU assay, as well as cell survival was detected by apoptosis with flow cytometry under TMZ treatment. Further, the expression of drug-resistance protein ABCG2, autophagic related proteins and PI3K/AKT pathway related proteins were determined by western blot or qRT-PCR in TMZ-treated glioma cells. Finally, the tumor volume and weight were measured and ABCG2 expression was detected by immunohistochemistry assay in mouse tumor xenograft model. Results Our integrated approach demonstrated lncRNA CRNDE was a poor prognosis factor for GBM patient, which was up-regulated in patients who are resistant to TMZ, and closely associated with chemotherapeutic response status to TMZ treatment. Further, functional assays revealed that knockdown of CRNDE could notably reduce glioma cell viability and proliferation, and elevate apoptosis to enhance the chemosensitivity to TMZ in vitro and in vivo. Mechanistically, the depression of CRNDE down-regulated LC3 II/I, Beclin1 and Atg5 expression levels and increased expression of p62 to inhibit autophagy due to the activation of PI3K/Akt/mTOR pathway as well as highly correlated with ABCG2 expression. Conclusions Overall, the study provided a preclinical basis in developing LncRNA CRNDE as a reliable clinical predictor of outcome and prognosis of GBM patient, a potential biomarker for predicting TMZ treatment response and a new therapeutic target for enhancing TMZ sensitivity in GBM by modulating the autophagy through PI3K/Akt/mTOR pathway.


2020 ◽  
Vol 10 (7) ◽  
pp. 1022-1028
Author(s):  
Bo Shu ◽  
Dan Liang ◽  
Hui Tong ◽  
Chunan Cao ◽  
Hongchuan Gan ◽  
...  

Objectives: Neuregulin 4 (NRG4) is a novel signaling protein involved in many physiological activities. This study aims to explore role of NRG4 in glioma. Study design: Serum samples were obtained from patients with glioma, RT-qPCR assay was performed to detect the levels of NRG4 and ErbB4 in the patient's serum and glioma cell lines U251, SHG44, LN229 and T98G. The results suggested that the expression of NRG4 and ErbB4 were markedly higher in patients and glioma cell lines, especially in U251. Further, Small interfering RNAs (si-RNAs) that targeting NRG4 (si-NRG4) and the overexpressed plasmid of ErbB4 (pcDNA-ErbB4) were transfected into U251 cells. Cell proliferation was measured by CCK8 assay, migration and invasion was assessed by wound healing and transwell assays respectively. And the expression of PI3 K-p110 , PI3 K-p110 , pAKT-ser473, pAKT-thr308 and AKT were measured by western blot assay. Results: We found that downregulation of NRG4 significantly inhibited proliferation, migration and invasion of U251 cells. Moreover, NRG4 inhibition markedly reduced the expression of PI3 K-p110 , pAKT-ser473 and pAKT-thr308. And overexpression of ErbB4 alleviated the impact caused by NRG4 inhibition. Conclusion: NRG4 inhibition suppresses proliferation, migration and invasion of U251 via blocking the PI3 K/AKT pathway. NRG4 could serve as a potential target of glioma treatment.


Author(s):  
Victor Ruiz-Rodado ◽  
Adrian Lita ◽  
Tyrone Dowdy ◽  
Orieta Celiku ◽  
Alejandra Cavazos-Saldana ◽  
...  

2000 ◽  
Vol 10 (3) ◽  
pp. 159-168 ◽  
Author(s):  
Friederike Schmidt ◽  
Peter Groscurth ◽  
Johannes Dichgans ◽  
Michael Weller

Sign in / Sign up

Export Citation Format

Share Document