Skip to main content
Erschienen in: Cancer Cell International 1/2018

Open Access 01.12.2018 | Primary research

ADAR3 expression is an independent prognostic factor in lower-grade diffuse gliomas and positively correlated with the editing level of GRIA2Q607R

verfasst von: Ying Zhang, Kuanyu Wang, Zheng Zhao, Si Sun, Kenan Zhang, Ruoyu Huang, Fan Zeng, Huimin Hu

Erschienen in: Cancer Cell International | Ausgabe 1/2018

Abstract

Background

RNA editing by adenosine deaminases acting on RNA (ADARs) converts adenosines to inosines (A-to-I) in RNA, that alters gene expression and generates protein diversity. Dysregulation of A-to-I editing has been found associated with a number of nervous system diseases. However, the role of ADAR3, a brain specific high expression adenosine deaminase, in gliomas has rarely been investigated. In this study we illuminated the clinical significance and molecular features of ADAR3 in patients with glioma.

Methods

309 glioma samples from Chinese Glioma Genome Atlas were enrolled into this study. In validation sets, 601 glioma samples in TCGA, 410 glioma samples in REMBRANDT and 258 glioma samples in GSE16011 were obtained. Relationships between ADAR3 expression and prognosis-related genomic alteration, outcome and gene ontology analysis were investigated. Moreover, the characteristic of GRIA2Q607R editing in gliomas has been investigated. Graphpad Prism 5.0, SPSS 16.0 and R language were used to perform statistical analysis and graphical work.

Results

ADAR3 expression was down regulated along with glioma grade progression in CGGA dataset. ADAR3 was characteristically highly expressed in neural subtype and IDH1/2 mutant preference. Moreover, high expression of ADAR3 predicted a better prognosis in lower-grade glioma (LGG) patients and multivariate analysis suggested ADAR3 expression was an independent prognostic indicator. The results of the three other validation datasets showed similar findings. Bioinformatics analyses suggested that ADAR3 may play a role in the malignant transformation of glioma cells by affecting cell proliferation, angiogenesis or cell adhesion. Furthermore, the editing level of GRIA2Q607R was significantly correlated with ADAR3 expression.

Conclusions

Our study demonstrated the clinical and molecular characterization of ADAR3 in glioma development and progression. ADAR3 expression was negatively associated with tumor malignant in the overall glioma patients. And it was a favorable independent prognostic indicator of LGG patients. ADAR3 appeared to act as a tumor suppressor in glioma cells. Therefore, ADAR3 represented a potential therapeutic target and useful prognostic factor for glioma patients.
Hinweise

Electronic supplementary material

The online version of this article (https://​doi.​org/​10.​1186/​s12935-018-0695-8) contains supplementary material, which is available to authorized users.
Ying Zhang and Kuanyu Wang contributed equally to this work
Abkürzungen
LGG
lower-grade glioma
ADARs
adenosine deaminases acting on RNA
WHO
World Health Organization
A-to-I
adenosine to inosine
GBM
glioblastoma
CGGA
Chinese Glioma Genome Atlas
RNAseq
RNA sequencing
TCGA
The Cancer Genome Atlas
OS
survival time
DAVID
Database for Annotation, Visualization and Integrated Discovery
GSEA
gene set enrichment analysis
ROC
receiver operating characteristic

Background

Gliomas are the most common primary intracranial tumor, representing 81% of malignant brain tumors [1]. According to 2007 World Health Organization (WHO) classification criteria, gliomas can be divided into four grades based on the degree of malignancy (WHO Grade I-IV). WHO Grade I apply to lesions with low proliferative potential and the possibility of cure following surgical resection alone. WHO Grade II and III are called lower-grade gliomas (LGG) [2]. Because of they are generally infiltrative in nature, surgical resection is not enough, and patients may need adjuvant radiation and/or chemotherapy [3]. WHO grade IV (reserved for glioblastoma) is the most malignant form of glioma, which has a 5-year relative survival of ~ 5% [1]. Studies over the past two decades have clarified several genetic alterations in gliomas, such as mutations in IDH1/2, TP53 and ATRX, TERT promoter mutation, MGMT promoter methylation and 1p/19q co-deletion, etc. Some of them contribute to glioma classification, prognosis or guidance in therapeutic decisions. In the 2016 WHO classification of central nervous system (CNS) tumors, classification of diffuse gliomas (WHO Grade II-IV) has fundamentally changed: for the first time, a large subset of these tumors is now defined based on IDH1 or IDH2 mutation and co-deletion of chromosomal arms 1p and 19q [4]. This breaks with the principle of diagnosis based entirely on phenotypic by incorporating genotypic parameters into the classification of CNS tumor entities [4]. The exploit of novel and reliable biomarkers for the prediction of gliomas may further help to elucidate the molecular mechanism of glioma development and progression.
RNA editing is one of the posttranscriptional mechanisms that precisely alters RNA sequences, thus regulates gene expression and generates structurally or functionally different isoforms of proteins. The most predominant pattern of RNA editing converts adenosine to inosine (A-to-I) in coding and non-coding RNA sequences, which is mediated by ADAR enzymes [5]. A-to-I editing is most abundant in the CNS and is critical for maintaining proper neuronal function [6]. Targets of this type of RNA editing are transcripts encoding proteins involved in neurotransmitter receptors and voltage-gated ion channels, including α-amino-3-hydroxy-5-methyl-4-isoxazole-propionic acid (AMPA) [6], potassium channel Kv1.1 (KCNA1) [7], G protein-coupled serotonin receptor 5-HT2CR (5-hydroxytryptamine receptor subtype 2C) [8] and the α3 subunit of GABAA (γ-aminobutyric acid type A) receptor (GABRA3) [9]. Changes in the A-to-I editing have been associated with a number of human diseases, such as amyotrophic lateral sclerosis (ALS), transient forebrain ischemia, epilepsy, metastatic melanoma, glioblastoma (GBM, WHO grade IV) and hepatocellular carcinoma (HCC) [6, 10, 11].
In human, three ADAR family members (ADAR1, ADAR2 and ADAR3) and two ADAD (adenosine deaminase domain-containing) proteins (ADAD1 and ADAD2) have been identified. They all contain at least one dsRNA binding domain (dsRBD) and a conserved C-terminal deaminase domain, whereas ADAR3 contains a unique Arg-rich ssRNA binding domain (R domain) at its N-terminus [12]. Unlike the wide expression of ADAR1 and ADAR2, ADAR3 expression is restricted to the brain [13]. Moreover, ADAR3 is not catalytically active and is thought to act as a competitive inhibitor of ADAR1 and ADAR2 in the brain [13]. Recently, ADAR3 was proved to directly compete with ADAR2 for binding to GRIA2 pre-mRNA, inhibiting RNA editing at the Q607R editing site of GRIA2 in GBM cell line [14]. This editing site is almost 100% edited in mammalian brain and controls the calcium permeability of AMPA receptor channels, which are involved in fast excitatory synaptic transmission [15, 16]. These studies suggest that ADAR3 may be associated with the tumorigenesis and progression of glioma. However, the clinical significance and molecular features of glioma with ADAR3 expression remain elusive.
In this study, we evaluated the expression pattern, prognostic significance and potential biological association of ADAR3 in patients with glioma. We collected ADAR3 mRNA expression and clinical information in 1578 glioma samples from four independent datasets. Meanwhile, the expression pattern of ADAR3 in different types of gliomas was evaluated by t test and one-way ANOVA test. In addition, the overall survival of glioma patients was assessed based on ADAR3 expression level and the prognostic value of ADAR3 in glioma was tested using Cox regression analysis. Furthermore, the bioinformatics analyses were applied to predict the biological process of ADAR3 in gliomas. Finally, the relationship of GRIA2Q607R editing and ADAR3 expression has been analyzed based on CGGA RNAseq dataset. These results suggested that ADAR3 was a novel independent prognostic indicator of LGG patients and appear to act as a tumor suppressor in glioma cells.

Methods

Patients and samples

309 specimens of WHO grade II–IV from Chinese Glioma Genome Atlas (CGGA) were included in our study. This study was approved by the Institutional Review Boards of Beijing Tiantan Hospital (Beijing, China). The written informed consents were obtained from all the participants enrolled in the study. All experiment methods were performed in accordance with the relevant guidelines and regulations. The establishment and management of our CGGA databank have been reported previously [17].

ADAR3 expression analysis in datasets

The CGGA RNA sequencing (RNAseq) database (http://​www.​cgga.​org.​cn) with 309 glioma samples (104 grade II, 67 grade III, and 138 grade IV samples) were obtained as the discovery set. In validation sets, 601 glioma samples (211 grade II, 236 grade III, and 154 grade IV samples) in The Cancer Genome Atlas (TCGA) RNAseq database (http://​cancergenome.​nih.​gov), 410 glioma samples (99 grade II, 84 grade III, and 227 grade IV samples) in The Repository for Molecular Brain Neoplasia Data (REMBRANDT, http://​caintegrator-info.​nci.​nih.​gov/​REMBRANDT), and 258 glioma samples (23 grade II, 84 grade III, and 151 grade IV samples) in GSE16011 microarray database (https://​www.​ncbi.​nlm.​nih.​gov/​geo/​) were obtained. The raw data were centralized and standardized through the scale function in R language before analysis. In the four datasets, only samples classified WHO grade II-IV were included for survival and grade expression pattern analysis.

Characterization of A-to-I RNA editing of GRIA2Q607R

RNAseq library preparation, sequencing and RNAseq data analysis were processed as our previous research [18]. Then we obtained the RNAseq BAM files of 309 glioma samples in the CGGA RNAseq database. On the basis of the RNAseq reads mapped to the human reference genome (hg19), the editing level at Q607R site of GRIA2 in a given sample was calculated as the number of edited reads G divided by the total number of reads A + G (G/A + G), and the total number of reads A + G less than 30 were excluded.

Statistical analysis

Student’s t-test and one-way ANOVA test were used to test the significance of differences between patients with different grades of gliomas by R language. Overall survival time (OS) was calculated from the date of histological diagnosis until death or the last follow-up. Kaplan–Meier survival analysis and the log-rank test were used to assess the statistical significance between stratified survival groups. Half of the patients who had relatively higher ADAR3 expression were defined as high expression group, while the other half part of patients were considered as lower expression group. Univariate and multivariate Cox regression analysis including gender, age at diagnosis, WHO grade, MGMT promoter methylation, IDH1/2 mutation status, radiotherapy and chemotherapy were used to assess prognostic value of ADAR3 in glioma by using SPSS 16.0 (Armonk, NY, USA). A P value less than 0.05 was considered to be statistical significant.
The Pearson correlation analysis was performed by R language to calculate the correlation between ADAR3 and other genes in CGGA RNAseq datasets. GO and KEGG pathway analysis were performed using the online Database for Annotation, Visualization and Integrated Discovery (DAVID, http://​david.​abcc.​ncifcrf.​gov).
The mRNA expression profile of glioma samples from the CGGA mRNA sequencing was analyzed by gene set enrichment analysis (GSEA, http://​www.​broad.​mit.​edu/​gsea). For GSEA, the expression level of ADAR3 was divided into low- or high-expression group by a criterion of whether the expression level was greater than the median value.

Results

ADAR3 expression is down regulated along with glioma grade progression and shows a subtype preference

To get an overview of adenosine deaminase domain containing proteins status in gliomas, we firstly investigated the mRNA expression of these proteins in patients. ADAD1 and ADAD2 were discarded for their rarely expression in CGGA mRNA sequencing database (CGGA RNAseq). This may be due to their specifically expression in the testes and necessary for spermatogenesis [19]. Then we compared the mRNA expression of ADAR1, ADAR2 and ADAR3 in patients with different WHO grades (II, III and IV) based CGGA RNAseq. Among them, only ADAR3 expression showed a significant negative correlation with tumor grade (P < 0.0001, Fig. 1c). And the expression of ADAR3 also decreased along with grade progression in TCGA RNA sequencing (TCGA RNAseq), GSE16011 and REMBRANDT microarray databases (P < 0.0001, Additional file 1: Figure S1C, F and I). Although the expression of ADAR1 and ADAR2 was lowest in glioblastoma (GBM, WHO grade IV) of TCGA RNAseq (Additional file 1: Figure S1A, B), the same results did not validated in GSE16011 and REMBRANDT microarray databases (Additional file 1: Figure S1D, E, G, H). These results indicated that ADAR3 expression associated with malignant progression of glioma.
To analyze the molecular relevance of ADAR3 in glioma, we investigated the characteristic of ADAR3 expression in different molecular subtypes established by TCGA network [20]. The neural subtype had the highest ADAR3 expression, followed by proneural subtype in CGGA, TCGA, GSE16011 and REMBRANDT datasets. Although the differences between classical subtype and mesenchymal subtype is inconsistent in four datasets, the expression of ADAR3 was still the lowest in these two subtypes (Fig. 2a, Additional file 2: Figure S2A–C). For the further investigation, the gliomas were divided into two groups, neural subtype and non-neural subtype. Receiver operating characteristic (ROC) analysis for ADAR3 mRNA in prediction of neural were performed in four datasets, we found that ADAR3 had a high efficiency to predict neural subtype (Fig. 2b, Additional file 2: Figure S2D–F). This results indicated that ADAR3 could serve as a biomarker for neural subtype.
Then we investigated the correlation between ADAR3 mRNA expression level and IDH1 or/and IDH2 (IDH) mutation, which is a canonical indicator of glioma [21]. The patients with mutant IDH (IDH-mut) showed much stronger expression of ADAR3 than those with wildtype IDH (IDH-wt) in CGGA and TCGA datasets (Fig. 2c, Additional file 2: Figure S2G). We further detected its expression in diffuse gliomas patients based on grade, IDH and 1p/19q status. ADAR3 expression was highest in LGG IDH-mut and 1p/19q codeleted stratified patients (Fig. 2d, Additional file 2: Figure S2H). The difference between GBM IDH-mut and GBM IDH-wt is not significant, but the mean value of ADAR3 in GBM IDH-mut still showed an upward trend compared with that of GBM IDH-wt (Fig. 2e, Additional file 2: Figure S2I). These results indicated that ADAR3 had a IDH mutant preference.
Moreover, we conducted an overview of ADAR3 expression with several genetic alterations which are related with progression of glioma (Fig. 4a, Table 1). Consistent with the above results, lower WHO grade, neural subtype and IDH mutants are enriched in higher ADAR3 expression (P < 0.0001). Meanwhile, deletion of 1p or/and 19q, confirming favorable prognostic indicators [22], aggregated in gliomas with higher ADAR3 expression (P < 0.0001). These results indicated that higher ADAR3 expression may be an optimistic factor for patients with glioma.
Table 1
Characteristics of glioma patients in low and high ADAR3 expression group in CGGA dataset
Variable
Total (n = 309)
High ADAR3 expression (n = 154)
Low ADAR3 expression (n = 155)
P value
Age
 < 45
178
104
74
0.0005
 ≥ 45
131
50
81
 
Gender
 Male
115
57
58
1
 Female
194
97
97
 
Grade
 II
104
86
18
< 0.0001
 III
67
35
32
 
 IV
138
33
105
 
KPS
 < 80
58
42
16
0.0006
 ≥ 80
108
46
62
 
 NA
143
67
76
 
Subtype
 Neural
76
71
5
< 0.0001
 Proneural
99
61
38
 
 Classical
69
14
55
 
 Mesenchymal
65
8
57
 
IDH mutation
 Mutant
155
108
47
< 0.0001
 Wildtype
154
46
108
 
MGMT
 Unmethylated
111
48
63
0.0462
 Methylated
136
67
69
 
 NA
62
39
23
 
1p/19q deletion
 No
220
102
118
< 0.0001
 Yes
36
31
5
 
 NA
53
21
32
 
TERT
 No
158
79
79
0.6426
 Yes
91
43
48
 
 NA
60
33
27
 

ADAR3 is an independent prognostic indicator for LGG patients

To evaluate the prognostic value of ADAR3 expression, Kaplan–Meier survival analyses were performed in CGGA RNAseq, TCGA RNAseq, GSE16011 and REMBRANDT datasets, using the median ADAR3 expression as a cut-off. When taking all grades of patients into account, half of the patients who had relatively higher ADAR3 expression had a significantly longer survival time than those had lower ADAR3 expression in four datasets (Log-rank test, all P < 0.0001, Fig. 3a, Additional file 3: Figure S3A). Moreover, the prognostic value of ADAR3 expression was also analyzed in patients with lower-grade glioma (LGG, WHO grade II-III) and GBM, respectively. There was a significant correlation between low expression of ADAR3 and decreased overall survival rate in LGG patients (Log-rank test, all P < 0.05, Fig. 3b, Additional file 3: Figure S3B). Except to CGGA RNAseq dataset (Log-rank test, P = 0.0318, Fig. 3c), we failed to identify such significant differences in GBM patients (Log-rank test, all P > 0.05, Additional file 3: Figure S3C). Moreover, the similar trend was observed in LGG IDH-mut and 1p/19q non-codeleted and LGG IDH-wt patients (P = 6e − 04, P = 0.0043, Fig. 3e, f). CDKN2A loss is associated with shorter overall survival in LGG IDH-mut and 1p/19q non-codeleted patients [23]. We further compared ADAR3 expression in this group, the patients with CDKN2A loss showed much lower expression of ADAR3 than those with intact CDKN2A in TCGA dataset (P = 0.0247, Additional file 4: Figure S4). Although no significant difference was found in LGG IDH-mut and 1p/19q codeleted patients, it probably due to the small sample size (P = 0.2482, Fig. 3d). There was no significant difference in GBM IDH-mut and GBM IDH-wt patients (P = 0.953, P = 0.2619, Fig. 3g, h). Our results suggest that ADAR3 expression was at least a prognostic indicator in patients with lower-grade diffuse gliomas.
Furthermore, univariate and multivariate analyses were utilized to evaluate the independent value of ADAR3 expression and other clinic pathological variables predicting overall survival (OS) in glioma patients. As shown in Table 2, factors including age at diagnosis, WHO Grade, ADAR3 expression, MGMT promoter methylation, IDH1/2 mutation status and radiotherapy were significantly associated with the OS of glioma patients. Multivariate Cox regression analysis indicated that ADAR3 expression was an independent prognostic indicator for the OS of glioma patients in CGGA dataset (Table 2, HR, 0.571; 95% CI 0.430–0.760; P < 0.001). Moreover, we performed univariate and multivariate analyses in patients with LGG or GBM separately. As shown in Table 2, ADAR3 expression was an independent prognostic indicator for the OS of LGG patients in CGGA dataset (Table 3, HR, 0.419; 95% CI, 0.289–0.608; P < 0.001), but not for the OS of GBM patients (Table 3, HR, 0.804; 95% CI, 0.556–1.163; P = 0.247). These results indicate that ADAR3 expression is an independent prognostic factor in patients with LGG.
Table 2
Clinic pathologic factors associated with OS in the Cox hazard regression analysis for glioma patients from the CGGA dataset
Variable
Univariate Cox Regression
Multivariate Cox Regression
HR
95% CI
P value
HR
95% CI
P value
Gender
1.187
0.841–1.675
0.330
   
Age at diagnosis
1.038
1.022–1.053
0.001
0.998
0.979–1.018
0.875
WHO Grade
3.469
2.709–4.443
0.001
1.926
1.370–2.709
< 0.001
ADAR3 expression
0.356
0.284–0.447
< 0.001
0.571
0.430–0.760
< 0.001
MGMT promoter methylation
0.529
0.374–0.750
< 0.001
0.665
0.444–0.997
0.048
IDH1/2 Mutation status
0.229
0.159–0.331
0.001
0.637
0.370–1.097
0.104
Radiotherapy
0.421
0.291–0.611
< 0.001
0.409
0.276–0.606
< 0.001
Chemotherapy
1.386
0.969–1.983
0.074
   
P value < 0.05 was considered statistically significant
CI, confidence interval; HR, hazard ratio; IDH1/2, isocitrate dehydrogenase 1/2; MGMT, O6-methylguanine-DNA methyltransferase
Table 3
Clinic pathologic factors associated with OS in the Cox hazard regression analysis for LGG and GBM patients from the CGGA dataset
Variable
Lower-grade glioma
Glioblastoma
Univariate
Multivariate
Univariate
Multivariate
P value
HR
95% CI
P value
P value
HR
95% CI
P value
Gender
0.783
   
0.355
   
Age at diagnosis
< 0.001
1.023
0.992–1.054
0.143
0.569
   
ADAR3 expression
< 0.001
0.419
0.289–0.608
< 0.001
0.035
0.804
0.556–1.163
0.247
MGMT promoter methylation
0.225
   
0.010
0.551
0.342–0.886
0.014
IDH1/2 mutation status
< 0.001
0.595
0.290–1.221
0.157
0.089
   
Radiotherapy
0.056
   
< 0.001
0.447
0.273–0.733
0.001
Chemotherapy
0.002
1.397
0.705–2.770
0.338
< 0.001
0.441
0.271–0.717
< 0.001
P value < 0.05 was considered statistically significant
CI, confidence interval; HR, hazard ratio; IDH1/2, isocitrate dehydrogenase 1/2; MGMT, O6-methylguanine-DNA methyltransferase
To illuminate the biological feature of glioma with different ADAR3 expression, we performed Pearson correlation analysis to evaluate the correlation of ADAR3 expression and other genes in CGGA sequencing dataset. Totally, the positively or negatively correlated genes with ADAR3 mRNA expression in CGGA RNAseq dataset were 844 and 891, respectively (|R| ≥ 0.5, Fig. 4a, Additional file 5: Table S1). The correlated genes were used for functional annotation analysis with DAVID and ranked by P value in increasing order. We found that ADAR3 positive related genes were mainly involved in normal biological process, such as chemical synaptic transmission, positive regulation of GTPase activity, intracellular signal transduction, nervous system development and several kinds of ion transport (Fig. 4b). While ADAR3 negative related genes were frequently involved in the processes of cell adhesion, cell division, extracellular matrix organization, angiogenesis, cell proliferation and response to drug (Fig. 4b). Then, GSEA was applied between the high- and low-expression level of ADAR3. Based on CGGA mRNA sequencing gene expression profile, we observed that GO terms including “transmission of nerve impulse”, “modulation of synaptic transmission”, “neurotransmitter transport” and “synaptic signaling” were significantly up-regulated in the high-expression group compared to the low-expression group (Fig. 4c). Moreover, “epithelial mesenchymal transition”, “apoptosis”, “tumor necrosis factor mediated signaling pathway” and “angiogenesis” were enriched in the low-expression group (Fig. 4d). These analyzes suggested that down-regulation of ADAR3 may potentiate the malignant transformation activity in glioma cells by affecting cell proliferation, angiogenesis or cell adhesion.

ADAR3 expression is positively correlated with A-to-I editing of GRIA2Q607R

The Q607R editing in the second transmembrane domain is nearly 100% throughout the mammalian brain, which is essential for normal receptor function [24]. Underediting of this site has been identified in malignant gliomas [25]. It is specifically edited by ADAR2, while ADAR3 directly competes with ADAR2 for binding to GRIA2 transcript, inhibiting RNA editing in U87 [14]. For analysis relationship between the Q607R editing level and ADAR family expression level in glioma samples, we first calculated the editing level of Q607R site in GRIA2 based on CGGA RNAseq database. Compared to Grade II gliomas, the editing level of GRIA2Q607R is lower in Grade IV (P < 0.05, Fig. 5a). Meanwhile, the Q607R editing is related with different molecular subtype of glioma. Differential editing level at Q607R in GRIA2 has been identified in different TCGA subtypes (Anova, P = 5e-6, Fig. 5b). And the Q607R editing level is lower in IDH-mut glioma (P < 0.05, Fig. 5c). Moreover, the low Q607R editing level indicted a shorter overall survival time (Log-rank test, P = 0.0425, Fig. 5d). These results indicated that underediting of Q607R site in GRIA2 is a malignant marker for glioma based on a large cohort analysis.
Then, we calculated the relation of ADAR1, ADAR2 and ADAR3 expression with GRIA2Q607R editing level in glioma by Pearson analysis. The ADAR2 and ADAR3 expression are positively correlated with the GRIA2Q607R editing level (R = 0.3623, P = 3.94E−08; R = 0.3092, P = 3.43E−06; Fig. 5e), while ADAR1 expression is negatively correlated (R = − 0.1778, P = 8.67E−03, Fig. 5e). Collectively, these results indicated that the low expression of ADAR3 may induce unedited GRIA2 transcripts level which can promote cell migration and tumor invasion.

Discussion

A-to-I editing is the most prevalent transcriptional modification in human cells, which is an endogenous process causing genetic diversity [5]. In vertebrates, ADAR1, ADAR2 and ADAR3 are main members that catalyzed A-to-I editing [26]. It has been demonstrated that ADAR enzymes are essential proteins by perturbing expression levels and functions in animal model. ADAR1 deficient mice die during embryonic development, owing to defective hematopoiesis, widespread apoptosis, liver disintegration and an increasing activity of interferon signaling [2729]. ADAR2/ mice became prone to seizures and died within 3 weeks after birth [30]. The catalytic activity of ADAR3 has not been demonstrated and ADAR3/ mice are viable and appear to be normal [5]. However, the catalytically inactive ADAR3, which localizes exclusively high in the brain, predominantly acts as an inhibitor of editing in the brain through competitive binds to dsRNA substrates [13, 14, 31].
Furthermore, a majority of transcripts encoding proteins involved in neurotransmission are often targets of A-to-I editing, resulting in changes in the amino acid sequence of protein and physiological function of these ion channels and receptors [11]. Currently, mutations or changes in expression induced disorder of ADAR activity have been linked to a variety of human diseases, ranging from neurological and neurodegenerative diseases, metabolic diseases, viral infections, autoimmune disorders to cancers [10]. Previously, ADAR2 has been reported to relate with glioblastoma in both children and adults. ADAR2 promotes CDC14B editing and overexpression in astrocytoma cells, leading to Skp2 degradation and upregulation of p21 and p27 proteins, consequently causing cells to accumulate in the G1 phase of the cell cycle [32]. ADAR2 deaminase activity is essential to inhibit glioblastoma proliferation and tumor growth [32]. Meanwhile, the impaired ADAR2 activity in GBM inhibits a subset of onco-miRNAs (miR221, miR222, miR-21, miR-376a and miR-589–3p) editing, leading to tumor cells proliferation, migration and invasion [3335]. Recently, Oakes et al. have demonstrated that ADAR3 directly competed with ADAR2 for binding to GRIA transcript and inhibited RNA editing at the Q/R site of GRIA2 in glioblastoma [14], and this editing position of GRIA2 was substantially underedited in malignant human brain tumors compared with control tissues [25]. These suggested that ADAR3 may play a critical role in oncogenesis and development of glioblastoma. However, the clinical and molecular characterizations of ADAR3 in glioma still required further studies.
In this study, we analyzed the expression level of ADAR3 in four independent datasets including 1578 glioma patients. Most notably, ADAR3 mRNA expression decreased along with WHO grade progression, suggesting that the expression of ADAR3 gradually attenuated with the malignant increase of pathological glioma. Moreover, the ADAR3 expression level was significantly highest in the phenotype of known favorable molecular, such as neural subtype and LGG IDH-mut and 1p/19q codeleted stratified patients. The association of ADAR3 with glioma progress indicates a tumor suppressor role of ADAR3 in the tumorigenesis and progression of glioma. However, an increase in ADAR3 protein expression in the tumor tissue compared to adjacent tissue was observed in 5 out of 6 glioblastoma patient samples in previous study [14]. Thus, inconsistencies between different studies suggest the possibility that ADAR3 protein expression or activity may be modulated by a post-transcriptional way. At the same time, further study will be needed to investigate ADAR3 protein expression based on a large cohort.
From the overall survival curve, the low expression of ADAR3 indicated shorter overall survival time and lower survival rate in glioma patients. After divided the patients into LGG and GBM subgroup, the high expression of ADAR3 is also a favorable indicator in LGG group, but not in GBM group. Furthermore, the high expression of ADAR3 was associated with longer overall survival time in LGG IDH-mut and 1p/19q non-codeleted and LGG IDH-wt patients. Combined with univariate and multivariate Cox analysis, ADAR3 expression is an independent prognostic factor in patients with diffuse glioma, especially in LGG group. Recently, several genes have been reported to be prognostic factors in gliomas, such as IDH1 [36], FGFR3 [37], Notch1 [38]. Our research illuminated the clinical features of ADAR3 in diffuse glioma and confirmed that ADAR3 expression had a guiding significance for the prognosis of patients with LGG.
Through an analysis of the biological process of ADAR3 in glioma, we found that ADAR3 play an important role in the normal biological process, such as signal transduction, chemical synaptic transmission, neurotransmitter transport and intracellular signal transduction. This is consistent with A-to-I editing targets, which are primarily transcripts of neurotransmitter receptors and ion channels proteins [11]. Meanwhile, downregulation of ADAR3 will promote cell proliferation, angiogenesis, cell adhesion and migration. This indicated that inhibition the expression of ADAR3 in brain cells would promote the cell malignant transition. Collectively, these data strongly support the tumor suppressor role of ADAR3 in glioma progression.
The reduced GRIA2 editing at Q607R site has been observed in malignant gliomas [25], and the unedited GRIA2 protein promotes cell migration and invasion in these cell lines [24]. Oakes et al. reported that overexpression of ADAR3 inhibited RNA editing at the Q607R site of GRIA2 in astrocyte and astrocytoma cell lines [14], which indicted the competitive inhibition of ADAR2 with ADAR3 on this site. Based on our clinical data, we found the editing level of GRIA2Q607R is positively related with ADAR2 and ADAR3 mRNA expression, which is inconsistent with previous in vitro assay. These results indicated that the regulation of GRIA2 editing in gliomas is a more complex model than previous studies, and the tumor suppressor role of ADAR3 may partly related with the underedited level of Q607R.

Conclusions

Our preliminary study demonstrates the clinical and molecular characterization of ADAR3 in glioma development and progression. Unlike other adenosine deaminases, ADAR3 is a favorable independent prognostic indicator of LGG patients. And it appeared to act as a tumor suppressor in glioma cells. Therefore, further studies are necessary to confirm its underlying molecular mechanisms in glioma.

Authors’ contributions

YZ and KYW provided equal contributions to the design of the study, data analysis. ZZ, SS, KNZ, RYH and FZ participated in data downloading and preliminary analysis. YZ planned and wrote the manuscript. HMH critically revised the manuscript. All authors have read and approved the final manuscript.

Acknowledgements

We appreciate the patients who have participated in CGGA and this study.

Competing interests

The authors declare that they have no competing interests.

Availability of data and materials

All data generated and analyzed during this study are included in this published article and its additional files.
Not applicable.
The study was approved by the Institutional Review Boards of Beijing Tiantan Hospital (Beijing, China).

Funding

This study was supported by funds from the National Nature Science Foundation of China (81761168038); The National Key Research and Development Plan (2016YFC0902500); Capital Medical Development Research Fund (2016-1-1072).

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.
Literatur
1.
Zurück zum Zitat Ostrom QT, Bauchet L, Davis FG, Deltour I, Fisher JL, Langer CE, Pekmezci M, Schwartzbaum JA, Turner MC, Walsh KM, et al. The epidemiology of glioma in adults: a “state of the science” review. Neuro Oncol. 2014;16(7):896–913.CrossRefPubMedPubMedCentral Ostrom QT, Bauchet L, Davis FG, Deltour I, Fisher JL, Langer CE, Pekmezci M, Schwartzbaum JA, Turner MC, Walsh KM, et al. The epidemiology of glioma in adults: a “state of the science” review. Neuro Oncol. 2014;16(7):896–913.CrossRefPubMedPubMedCentral
2.
Zurück zum Zitat Brat DJ, Verhaak RG, Aldape KD, Yung WK, Salama SR, Cooper LA, Rheinbay E, Miller CR, Vitucci M, Morozova O, et al. Comprehensive, integrative genomic analysis of diffuse lower-grade gliomas. N Engl J Med. 2015;372(26):2481–98.CrossRefPubMed Brat DJ, Verhaak RG, Aldape KD, Yung WK, Salama SR, Cooper LA, Rheinbay E, Miller CR, Vitucci M, Morozova O, et al. Comprehensive, integrative genomic analysis of diffuse lower-grade gliomas. N Engl J Med. 2015;372(26):2481–98.CrossRefPubMed
3.
Zurück zum Zitat Louis DN, Ohgaki H, Wiestler OD, Cavenee WK, Burger PC, Jouvet A, Scheithauer BW, Kleihues P. The 2007 WHO classification of tumours of the central nervous system. Acta Neuropathol. 2007;114(2):97–109.CrossRefPubMedPubMedCentral Louis DN, Ohgaki H, Wiestler OD, Cavenee WK, Burger PC, Jouvet A, Scheithauer BW, Kleihues P. The 2007 WHO classification of tumours of the central nervous system. Acta Neuropathol. 2007;114(2):97–109.CrossRefPubMedPubMedCentral
4.
Zurück zum Zitat Louis DN, Perry A, Reifenberger G, von Deimling A, Figarella-Branger D, Cavenee WK, Ohgaki H, Wiestler OD, Kleihues P, Ellison DW. The 2016 World Health Organization Classification of Tumors of the Central Nervous System: a summary. Acta Neuropathol. 2016;131(6):803–20.CrossRefPubMed Louis DN, Perry A, Reifenberger G, von Deimling A, Figarella-Branger D, Cavenee WK, Ohgaki H, Wiestler OD, Kleihues P, Ellison DW. The 2016 World Health Organization Classification of Tumors of the Central Nervous System: a summary. Acta Neuropathol. 2016;131(6):803–20.CrossRefPubMed
6.
Zurück zum Zitat Tariq A, Jantsch MF. Transcript diversification in the nervous system: a to I RNA editing in CNS function and disease development. Front Neurosci. 2012;6:99.CrossRefPubMedPubMedCentral Tariq A, Jantsch MF. Transcript diversification in the nervous system: a to I RNA editing in CNS function and disease development. Front Neurosci. 2012;6:99.CrossRefPubMedPubMedCentral
7.
Zurück zum Zitat Bhalla T, Rosenthal JJ, Holmgren M, Reenan R. Control of human potassium channel inactivation by editing of a small mRNA hairpin. Nat Struct Mol Biol. 2004;11(10):950–6.CrossRefPubMed Bhalla T, Rosenthal JJ, Holmgren M, Reenan R. Control of human potassium channel inactivation by editing of a small mRNA hairpin. Nat Struct Mol Biol. 2004;11(10):950–6.CrossRefPubMed
8.
Zurück zum Zitat Burns CM, Chu H, Rueter SM, Hutchinson LK, Canton H, Sanders-Bush E, Emeson RB. Regulation of serotonin-2C receptor G-protein coupling by RNA editing. Nature. 1997;387(6630):303–8.CrossRefPubMed Burns CM, Chu H, Rueter SM, Hutchinson LK, Canton H, Sanders-Bush E, Emeson RB. Regulation of serotonin-2C receptor G-protein coupling by RNA editing. Nature. 1997;387(6630):303–8.CrossRefPubMed
11.
Zurück zum Zitat Behm M, Ohman M. RNA editing: a contributor to neuronal dynamics in the mammalian brain. Trends Genet. 2016;32(3):165–75.CrossRefPubMed Behm M, Ohman M. RNA editing: a contributor to neuronal dynamics in the mammalian brain. Trends Genet. 2016;32(3):165–75.CrossRefPubMed
12.
Zurück zum Zitat Nishikura K. A-to-I editing of coding and non-coding RNAs by ADARs. Nat Rev Mol Cell Biol. 2016;17(2):83–96.CrossRefPubMed Nishikura K. A-to-I editing of coding and non-coding RNAs by ADARs. Nat Rev Mol Cell Biol. 2016;17(2):83–96.CrossRefPubMed
13.
Zurück zum Zitat Chen CX, Cho DS, Wang Q, Lai F, Carter KC, Nishikura K. A third member of the RNA-specific adenosine deaminase gene family, ADAR3, contains both single- and double-stranded RNA binding domains. RNA. 2000;6(5):755–67.CrossRefPubMedPubMedCentral Chen CX, Cho DS, Wang Q, Lai F, Carter KC, Nishikura K. A third member of the RNA-specific adenosine deaminase gene family, ADAR3, contains both single- and double-stranded RNA binding domains. RNA. 2000;6(5):755–67.CrossRefPubMedPubMedCentral
14.
Zurück zum Zitat Oakes E, Anderson A, Cohen-Gadol A, Hundley HA. Adenosine deaminase that acts on RNA 3 (ADAR3) binding to glutamate receptor subunit B Pre-mRNA inhibits RNA editing in glioblastoma. J Biol Chem. 2017;292(10):4326–35.CrossRefPubMedPubMedCentral Oakes E, Anderson A, Cohen-Gadol A, Hundley HA. Adenosine deaminase that acts on RNA 3 (ADAR3) binding to glutamate receptor subunit B Pre-mRNA inhibits RNA editing in glioblastoma. J Biol Chem. 2017;292(10):4326–35.CrossRefPubMedPubMedCentral
15.
Zurück zum Zitat Lomeli H, Mosbacher J, Melcher T, Hoger T, Geiger JR, Kuner T, Monyer H, Higuchi M, Bach A, Seeburg PH. Control of kinetic properties of AMPA receptor channels by nuclear RNA editing. Science. 1994;266(5191):1709–13.CrossRefPubMed Lomeli H, Mosbacher J, Melcher T, Hoger T, Geiger JR, Kuner T, Monyer H, Higuchi M, Bach A, Seeburg PH. Control of kinetic properties of AMPA receptor channels by nuclear RNA editing. Science. 1994;266(5191):1709–13.CrossRefPubMed
16.
Zurück zum Zitat Wen W, Lin CY, Niu L. R/G editing in GluA2Rflop modulates the functional difference between GluA1 flip and flop variants in GluA1/2R heteromeric channels. Sci Rep. 2017;7(1):13654.CrossRefPubMedPubMedCentral Wen W, Lin CY, Niu L. R/G editing in GluA2Rflop modulates the functional difference between GluA1 flip and flop variants in GluA1/2R heteromeric channels. Sci Rep. 2017;7(1):13654.CrossRefPubMedPubMedCentral
17.
Zurück zum Zitat Bao ZS, Chen HM, Yang MY, Zhang CB, Yu K, Ye WL, Hu BQ, Yan W, Zhang W, Akers J, et al. RNA-seq of 272 gliomas revealed a novel, recurrent PTPRZ1-MET fusion transcript in secondary glioblastomas. Genome Res. 2014;24(11):1765–73.CrossRefPubMedPubMedCentral Bao ZS, Chen HM, Yang MY, Zhang CB, Yu K, Ye WL, Hu BQ, Yan W, Zhang W, Akers J, et al. RNA-seq of 272 gliomas revealed a novel, recurrent PTPRZ1-MET fusion transcript in secondary glioblastomas. Genome Res. 2014;24(11):1765–73.CrossRefPubMedPubMedCentral
18.
Zurück zum Zitat Zhao Z, Meng F, Wang W, Wang Z, Zhang C, Jiang T. Comprehensive RNA-seq transcriptomic profiling in the malignant progression of gliomas. Sci Data. 2017;4:170024.CrossRefPubMedPubMedCentral Zhao Z, Meng F, Wang W, Wang Z, Zhang C, Jiang T. Comprehensive RNA-seq transcriptomic profiling in the malignant progression of gliomas. Sci Data. 2017;4:170024.CrossRefPubMedPubMedCentral
19.
Zurück zum Zitat Schumacher JM, Lee K, Edelhoff S, Braun RE. Distribution of Tenr, an RNA-binding protein, in a lattice-like network within the spermatid nucleus in the mouse. Biol Reprod. 1995;52(6):1274–83.CrossRefPubMed Schumacher JM, Lee K, Edelhoff S, Braun RE. Distribution of Tenr, an RNA-binding protein, in a lattice-like network within the spermatid nucleus in the mouse. Biol Reprod. 1995;52(6):1274–83.CrossRefPubMed
20.
Zurück zum Zitat Cancer Genome Atlas Research Network. Comprehensive genomic characterization defines human glioblastoma genes and core pathways. Nature. 2008;455(7216):1061–8.CrossRef Cancer Genome Atlas Research Network. Comprehensive genomic characterization defines human glioblastoma genes and core pathways. Nature. 2008;455(7216):1061–8.CrossRef
21.
Zurück zum Zitat Yan H, Parsons DW, Jin G, McLendon R, Rasheed BA, Yuan W, Kos I, Batinic-Haberle I, Jones S, Riggins GJ, et al. IDH1 and IDH2 mutations in gliomas. N Engl J Med. 2009;360(8):765–73.CrossRefPubMedPubMedCentral Yan H, Parsons DW, Jin G, McLendon R, Rasheed BA, Yuan W, Kos I, Batinic-Haberle I, Jones S, Riggins GJ, et al. IDH1 and IDH2 mutations in gliomas. N Engl J Med. 2009;360(8):765–73.CrossRefPubMedPubMedCentral
22.
Zurück zum Zitat Smith JS, Perry A, Borell TJ, Lee HK, O’Fallon J, Hosek SM, Kimmel D, Yates A, Burger PC, Scheithauer BW, et al. Alterations of chromosome arms 1p and 19q as predictors of survival in oligodendrogliomas, astrocytomas, and mixed oligoastrocytomas. J Clin Oncol. 2000;18(3):636–45.CrossRefPubMed Smith JS, Perry A, Borell TJ, Lee HK, O’Fallon J, Hosek SM, Kimmel D, Yates A, Burger PC, Scheithauer BW, et al. Alterations of chromosome arms 1p and 19q as predictors of survival in oligodendrogliomas, astrocytomas, and mixed oligoastrocytomas. J Clin Oncol. 2000;18(3):636–45.CrossRefPubMed
23.
Zurück zum Zitat Reis GF, Pekmezci M, Hansen HM, Rice T, Marshall RE, Molinaro AM, Phillips JJ, Vogel H, Wiencke JK, Wrensch MR, et al. CDKN2A loss is associated with shortened overall survival in lower-grade (World Health Organization Grades II-III) astrocytomas. J Neuropathol Exp Neurol. 2015;74(5):442–52.CrossRefPubMed Reis GF, Pekmezci M, Hansen HM, Rice T, Marshall RE, Molinaro AM, Phillips JJ, Vogel H, Wiencke JK, Wrensch MR, et al. CDKN2A loss is associated with shortened overall survival in lower-grade (World Health Organization Grades II-III) astrocytomas. J Neuropathol Exp Neurol. 2015;74(5):442–52.CrossRefPubMed
24.
Zurück zum Zitat Ishiuchi S, Tsuzuki K, Yoshida Y, Yamada N, Hagimura N, Okado H, Miwa A, Kurihara H, Nakazato Y, Tamura M, et al. Blockage of Ca(2+)-permeable AMPA receptors suppresses migration and induces apoptosis in human glioblastoma cells. Nat Med. 2002;8(9):971–8.CrossRefPubMed Ishiuchi S, Tsuzuki K, Yoshida Y, Yamada N, Hagimura N, Okado H, Miwa A, Kurihara H, Nakazato Y, Tamura M, et al. Blockage of Ca(2+)-permeable AMPA receptors suppresses migration and induces apoptosis in human glioblastoma cells. Nat Med. 2002;8(9):971–8.CrossRefPubMed
25.
Zurück zum Zitat Maas S, Patt S, Schrey M, Rich A. Underediting of glutamate receptor GluR-B mRNA in malignant gliomas. Proc Natl Acad Sci USA. 2001;98(25):14687–92.CrossRefPubMedPubMedCentral Maas S, Patt S, Schrey M, Rich A. Underediting of glutamate receptor GluR-B mRNA in malignant gliomas. Proc Natl Acad Sci USA. 2001;98(25):14687–92.CrossRefPubMedPubMedCentral
26.
Zurück zum Zitat Slavov D, Clark M, Gardiner K. Comparative analysis of the RED1 and RED2 A-to-I RNA editing genes from mammals, pufferfish and zebrafish. Gene. 2000;250(1–2):41–51.CrossRefPubMed Slavov D, Clark M, Gardiner K. Comparative analysis of the RED1 and RED2 A-to-I RNA editing genes from mammals, pufferfish and zebrafish. Gene. 2000;250(1–2):41–51.CrossRefPubMed
27.
Zurück zum Zitat Hartner JC, Schmittwolf C, Kispert A, Muller AM, Higuchi M, Seeburg PH. Liver disintegration in the mouse embryo caused by deficiency in the RNA-editing enzyme ADAR1. J Biol Chem. 2004;279(6):4894–902.CrossRefPubMed Hartner JC, Schmittwolf C, Kispert A, Muller AM, Higuchi M, Seeburg PH. Liver disintegration in the mouse embryo caused by deficiency in the RNA-editing enzyme ADAR1. J Biol Chem. 2004;279(6):4894–902.CrossRefPubMed
28.
Zurück zum Zitat Wang Q, Miyakoda M, Yang W, Khillan J, Stachura DL, Weiss MJ, Nishikura K. Stress-induced apoptosis associated with null mutation of ADAR1 RNA editing deaminase gene. J Biol Chem. 2004;279(6):4952–61.CrossRefPubMed Wang Q, Miyakoda M, Yang W, Khillan J, Stachura DL, Weiss MJ, Nishikura K. Stress-induced apoptosis associated with null mutation of ADAR1 RNA editing deaminase gene. J Biol Chem. 2004;279(6):4952–61.CrossRefPubMed
29.
Zurück zum Zitat Hartner JC, Walkley CR, Lu J, Orkin SH. ADAR1 is essential for the maintenance of hematopoiesis and suppression of interferon signaling. Nat Immunol. 2009;10(1):109–15.CrossRefPubMed Hartner JC, Walkley CR, Lu J, Orkin SH. ADAR1 is essential for the maintenance of hematopoiesis and suppression of interferon signaling. Nat Immunol. 2009;10(1):109–15.CrossRefPubMed
30.
Zurück zum Zitat Higuchi M, Maas S, Single FN, Hartner J, Rozov A, Burnashev N, Feldmeyer D, Sprengel R, Seeburg PH. Point mutation in an AMPA receptor gene rescues lethality in mice deficient in the RNA-editing enzyme ADAR2. Nature. 2000;406(6791):78–81.CrossRefPubMed Higuchi M, Maas S, Single FN, Hartner J, Rozov A, Burnashev N, Feldmeyer D, Sprengel R, Seeburg PH. Point mutation in an AMPA receptor gene rescues lethality in mice deficient in the RNA-editing enzyme ADAR2. Nature. 2000;406(6791):78–81.CrossRefPubMed
31.
Zurück zum Zitat Tan MH, Li Q, Shanmugam R, Piskol R, Kohler J, Young AN, Liu KI, Zhang R, Ramaswami G, Ariyoshi K, et al. Dynamic landscape and regulation of RNA editing in mammals. Nature. 2017;550(7675):249–54.CrossRefPubMedPubMedCentral Tan MH, Li Q, Shanmugam R, Piskol R, Kohler J, Young AN, Liu KI, Zhang R, Ramaswami G, Ariyoshi K, et al. Dynamic landscape and regulation of RNA editing in mammals. Nature. 2017;550(7675):249–54.CrossRefPubMedPubMedCentral
32.
Zurück zum Zitat Galeano F, Rossetti C, Tomaselli S, Cifaldi L, Lezzerini M, Pezzullo M, Boldrini R, Massimi L, Di Rocco CM, Locatelli F, et al. ADAR2-editing activity inhibits glioblastoma growth through the modulation of the CDC14B/Skp2/p21/p27 axis. Oncogene. 2013;32(8):998–1009.CrossRefPubMed Galeano F, Rossetti C, Tomaselli S, Cifaldi L, Lezzerini M, Pezzullo M, Boldrini R, Massimi L, Di Rocco CM, Locatelli F, et al. ADAR2-editing activity inhibits glioblastoma growth through the modulation of the CDC14B/Skp2/p21/p27 axis. Oncogene. 2013;32(8):998–1009.CrossRefPubMed
33.
Zurück zum Zitat Tomaselli S, Galeano F, Alon S, Raho S, Galardi S, Polito VA, Presutti C, Vincenti S, Eisenberg E, Locatelli F, et al. Modulation of microRNA editing, expression and processing by ADAR2 deaminase in glioblastoma. Genome Biol. 2015;16:5.CrossRefPubMedPubMedCentral Tomaselli S, Galeano F, Alon S, Raho S, Galardi S, Polito VA, Presutti C, Vincenti S, Eisenberg E, Locatelli F, et al. Modulation of microRNA editing, expression and processing by ADAR2 deaminase in glioblastoma. Genome Biol. 2015;16:5.CrossRefPubMedPubMedCentral
34.
Zurück zum Zitat Choudhury Y, Tay FC, Lam DH, Sandanaraj E, Tang C, Ang BT, Wang S. Attenuated adenosine-to-inosine editing of microRNA-376a* promotes invasiveness of glioblastoma cells. J Clin Invest. 2012;122(11):4059–76.CrossRefPubMedPubMedCentral Choudhury Y, Tay FC, Lam DH, Sandanaraj E, Tang C, Ang BT, Wang S. Attenuated adenosine-to-inosine editing of microRNA-376a* promotes invasiveness of glioblastoma cells. J Clin Invest. 2012;122(11):4059–76.CrossRefPubMedPubMedCentral
35.
Zurück zum Zitat Cesarini V, Silvestris DA, Tassinari V, Tomaselli S, Alon S, Eisenberg E, Locatelli F, Gallo A. ADAR2/miR-589-3p axis controls glioblastoma cell migration/invasion. Nucleic Acids Res. 2018;46(4):2045–59.CrossRefPubMed Cesarini V, Silvestris DA, Tassinari V, Tomaselli S, Alon S, Eisenberg E, Locatelli F, Gallo A. ADAR2/miR-589-3p axis controls glioblastoma cell migration/invasion. Nucleic Acids Res. 2018;46(4):2045–59.CrossRefPubMed
36.
Zurück zum Zitat Li MY, Wang YY, Cai JQ, Zhang CB, Wang KY, Cheng W, Liu YW, Zhang W, Jiang T. Isocitrate dehydrogenase 1 gene mutation is associated with prognosis in clinical low-grade gliomas. PLoS ONE. 2015;10(6):e0130872.CrossRefPubMedPubMedCentral Li MY, Wang YY, Cai JQ, Zhang CB, Wang KY, Cheng W, Liu YW, Zhang W, Jiang T. Isocitrate dehydrogenase 1 gene mutation is associated with prognosis in clinical low-grade gliomas. PLoS ONE. 2015;10(6):e0130872.CrossRefPubMedPubMedCentral
37.
Zurück zum Zitat Wang Z, Zhang C, Sun L, Liang J, Liu X, Li G, Yao K, Zhang W, Jiang T. FGFR3, as a receptor tyrosine kinase, is associated with differentiated biological functions and improved survival of glioma patients. Oncotarget. 2016;7(51):84587–93.PubMedPubMedCentral Wang Z, Zhang C, Sun L, Liang J, Liu X, Li G, Yao K, Zhang W, Jiang T. FGFR3, as a receptor tyrosine kinase, is associated with differentiated biological functions and improved survival of glioma patients. Oncotarget. 2016;7(51):84587–93.PubMedPubMedCentral
38.
Zurück zum Zitat Hai L, Zhang C, Li T, Zhou X, Liu B, Li S, Zhu M, Lin Y, Yu S, Zhang K, et al. Notch1 is a prognostic factor that is distinctly activated in the classical and proneural subtype of glioblastoma and that promotes glioma cell survival via the NF-kappaB(p65) pathway. Cell Death Dis. 2018;9(2):158.CrossRefPubMedPubMedCentral Hai L, Zhang C, Li T, Zhou X, Liu B, Li S, Zhu M, Lin Y, Yu S, Zhang K, et al. Notch1 is a prognostic factor that is distinctly activated in the classical and proneural subtype of glioblastoma and that promotes glioma cell survival via the NF-kappaB(p65) pathway. Cell Death Dis. 2018;9(2):158.CrossRefPubMedPubMedCentral
Metadaten
Titel
ADAR3 expression is an independent prognostic factor in lower-grade diffuse gliomas and positively correlated with the editing level of GRIA2Q607R
verfasst von
Ying Zhang
Kuanyu Wang
Zheng Zhao
Si Sun
Kenan Zhang
Ruoyu Huang
Fan Zeng
Huimin Hu
Publikationsdatum
01.12.2018
Verlag
BioMed Central
Erschienen in
Cancer Cell International / Ausgabe 1/2018
Elektronische ISSN: 1475-2867
DOI
https://doi.org/10.1186/s12935-018-0695-8

Weitere Artikel der Ausgabe 1/2018

Cancer Cell International 1/2018 Zur Ausgabe

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.