We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine
Research ArticleOpen Accesscc iconby iconnc iconnd icon

PABPC1 relevant bioinformatic profiling and prognostic value in gliomas

    Qiangwei Wang

    Beijing Neurosurgical Institute, Capital Medical University, Beijing, PR China

    ‡Authors contributed equally

    Search for more papers by this author

    ,
    Zhiliang Wang

    Beijing Neurosurgical Institute, Capital Medical University, Beijing, PR China

    ‡Authors contributed equally

    Search for more papers by this author

    ,
    Zhaoshi Bao

    Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, PR China

    ,
    Chuanbao Zhang

    Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, PR China

    ,
    Zheng Wang

    *Author for correspondence: Tel. & Fax: +86 106 709 8431;

    E-mail Address: wangzheng@ccmu.edu.cn

    Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, PR China

    &
    Tao Jiang

    **Author for correspondence: Tel. & Fax: +86 106 709 8431;

    E-mail Address: taojiang1964@163.com

    Beijing Neurosurgical Institute, Capital Medical University, Beijing, PR China

    Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, PR China

    China National Clinical Research Center for Neurological Diseases, Beijing, PR China

    Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, PR China

    Published Online:https://doi.org/10.2217/fon-2019-0268

    Abstract

    Aim: We aimed at investigating molecular features and potential clinical value of PABPC1 in gliomas. Materials & methods: We assembled totally 1000 glioma samples with mRNA expression data from Chinese Glioma Genome Atlas and The Cancer Genome Atlas. We utilized R language as the main analysis tool. Gene Ontology was performed for functional analysis. Results: PABPC1 was downregulated in gliomas with higher malignance and PABPC1 may contribute as potential predictor of proneural subtype in gliomas. Higher expression of PABPC1 was significantly related to better prognosis and related to biological process of translation. Conclusion: Our finding improves the understanding of PABPC1 as a novel biomarker with potential therapeutic connotations.

    Gliomas represent the most prevalent and fatal brain tumor in adults [1–3]. According to the updated 2016 WHO classification system of CNS, with histology and molecular parameters, gliomas could be categorized as followed: lower grade gliomas (LGG) with isocitrate dehydrogenase (IDH) mutation with or without 1p/19q-codel, LGG with IDH-wildtype subtype, glioblastoma multiforme (GBM) with IDH mutation or not [4]. In order to improve diagnostic accuracy and treatment efficacy for the glioma patients, it is crucial to identify novel biomarkers and therapeutic targets.

    RNA-binding proteins are versatile and can interact with many molecules. The poly adenosine tail of RNA can be bound by RNA poly(A) binding proteins (PABPs) specifically. PABPs are essential for initiation of translation and stabilization of mRNA [5–7]. In humans, PABPs include at least three functional proteins: PABP1, induced PABP (iPABP) and PABP3. Among them, PABP1 protein is encoded by the PABPC1 gene [8]. In many cancers, overexpression of PABPC1 promotes tumor progression and metastasis, such as esophageal cancer [9], gastric carcinoma [10], follicular thyroid cancer [11], bladder cancer [12] and so on. However, little is known about the genetic and epigenetic features of PABPC1 in gliomas.

    To explore the clinical and molecular characteristics of PABPC1 in gliomas, we utilized the gene expression data from Chinese Glioma Genome Atlas (CGGA) dataset as training set and validated in The Cancer Genome Atlas (TCGA) dataset. We detected PABPC1 expression was downregulated in gliomas with higher malignance and high expression of PABPC1 in proneural subtype and showed a beneficial effect in patients with gliomas. This integrative study of PABPC1 might provide a potential therapeutic target for glioma patients in future.

    Materials & methods

    Samples collected in this study

    In CGGA dataset (http://www.cgga.org.cn), we collected 301 whole grade glioma samples with mRNA microarray data as training set. In TCGA dataset, we downloaded mRNA sequencing data of 699 glioma samples online (https://cancergenome.nih.gov/) for validation. Thus, we obtained totally 1000 samples with mRNA expression data. Meanwhile, clinical follow-up and molecular information of these patients were collated.

    IDH1/2 mutations

    It was common to employ pyrosequencing technique to detect IDH mutation in CGGA [13], and IDH mutant status of TCGA patients downloaded online were mainly tested by pyrosequencing technique or whole exon sequencing technique.

    TCGA subtype annotation

    As for the TCGA subtype annotation, patients were divided into four subtypes (proneural–neural–classical–mesenchymal) using the published signatures and single-sample gene set enrichment analysis algorithm [14].

    Immunohistochemical

    Formalin-fixed, paraffin-embedded glioma specimens were cut (5 μm section), deparaffinized, rehydrated before antigen repair in citrate buffer. After blocking endogenous peroxidase activity with ethanol containing 3% hydrogen peroxidase, we incubated sections in rabbit anti-PABP1 antibody (number 4992, dilution:1:50, Cell Signaling Technology) over night at 4°C. Diaminobenzidine was used as chromogens and slides were counterstained with hematoxylin before mounting. Each stained slide was scored by two independent neuropathologists. All experiments were performed with negative and positive control to ensure the quality of staining. Staining was scored using a four-point scale from 0–3: 0 = no staining, 1 < 10% neoplastic cells positive staining, 2 = 10–30% neoplastic cells positive staining, 3 > 30% neoplastic cells positive staining. We selected specimens from 6 glioma patients (3 of LGG, 3 of GBM) for immunohistochemical analysis. Five fields at 400 × magnification were analyzed per specimen. The PABPC1 expression difference between two groups was tested by χ2 test.

    Gene ontology analysis

    We filtered PABPC1 significantly correlated genes (Pearson |R| >0.5; p < 0.05) by the method of Pearson correlation analysis in TCGA dataset. Then PABPC1 positively associated genes were chosen to perform gene ontology (GO) analysis in DAVID (https://david.ncifcrf.gov/) [15].

    Statistical analysis

    R language (version 3.5.1, http://www.r-project.org) was the main data analysis and mapping tool. Student’s t-test was performed for PABPC1 expression difference. For the comparison between categorical variables, we applied a two-tailed χ2 test. The time interval from diagnosis to death or the last follow-up was defined as overall survival (OS) time. Based on median PABPC1 expression, we divided patients into two groups, high- and low-PABPC1 groups. Kaplan–Meier estimate was used in survival analysis with two-sided log-rank test. There was significant difference when p-value ≤ 0.05.

    Results

    Patients

    We selected 301 glioma patients of whole grade from CGGA as discovery cohort and 699 glioma patients of whole grades from TCGA for validation in our study (Table 1). Median age of patients in CGGA was 42 (from 13 to 70) and 47 (from 14 to 89) in TCGA. Male accounted for 60% in CGGA and 52% in TCGA. CGGA contained 122 grade II, 51 WHO grade III and 128 grade IV gliomas (GBMs). TCGA dataset included 223 grade II, 245 grade III and 168 grade IV gliomas. Molecular subtype of two datasets were shown in the table as well. We also collected data on genetic alteration. In CGGA, 135 (45%) tumors were detected with IDH mutation and 43 (14%) tumors harbored 1p/19q codeletion. In TCGA, 441(63%) tumors exhibited IDH mutation and 172 (25%) showed 1p19q codeletion. Meanwhile, MGMT promoter methylation status of tumors was also available in two datasets. According to the updated 2016 WHO classification system of CNS [4], glioma patients in two databases were finally classified based on histologic and molecular data (Supplementary Table 1).

    Table 1. Baseline patient characteristics.
      CGGA (n = 301, %)TCGA (n = 699, %)
    AgeMedian (range)42(13–70)47(14–89)
    GenderMale180 (60)366 (52)
     Female121 (40)268 (38)
     NA065 (10)
    KPS scorePreoperative KPS ≥ 80173 (57)324 (46)
     Preoperative KPS < 8092 (31)70 (10)
     NA36 (12)305 (44)
    Pathological typeA80 (27)170 (24)
     O32 (11)180 (26)
     OA61 (20)118 (17)
     GBM128 (43)166 (24)
     NA065 (9)
    Molecular subtypeNeural81 (27)115 (16)
     Proneural86 (29)250 (36)
     Mesenchymal111 (37)103 (15)
     Classical23 (8)92 (13)
     NA0139 (20)
    Grade2122 (41)223 (32)
     351 (17)245 (35)
     4128 (43)166 (24)
     NA065 (9)
    IDH mutationMutant135 (45)441 (63)
     Wild162 (54)246 (35)
     NA4 (1)12 (2)
    1p/19q statusCodeletion43 (14)172 (25)
     Noncodeletion258 (86)520 (74)
     NA0 (0)7 (1)
    MGMT promoter methylationMethylated91 (30)492 (70)
     Not methylated90 (30)168 (24)
     NA120 (40)37 (5)

    CGGA: Chinese Glioma Genome Atlas; GBM: Glioblastoma multiforme; IDH: Isocitrate dehydrogenase; TCGA: The Cancer Genome Atlas.

    PABPC1 expression was downregulated in gliomas with higher malignance

    In order to investigate the distribution characteristic of PABPC1 expression, we compared the expression of PABPC1 in gliomas of different grades, histopathology and IDH mutation status. In CGGA dataset, PABPC1 showed the lowest expression in glioblastoma (GBM; Figure 1A). The result was validated in TCGA (Figure 1C). Next, we divided patients into IDH-mutant groups and IDH-wildtype and found that PABPC1 was poorly expressed in IDH-wildtype groups compared with IDH-mutant groups across different grades (Figure 1B & D). We also divided patients into groups according to 2016 WHO classification and found that PABPC1 was poorly expressed in malignant molecular phenotypes (IDH-wildtype or 1p/19q-intact, Supplementary Figure 1A& B). To further clarify the relationship between PABPC1 and glioma grade at the protein expression level, we selected specimens from six glioma patients (3 of LGG, 3 of GBM, 5 fields at 400 × magnification were analyzed per specimen) for immunohistochemical analysis. Two representative stained sections were showed in Supplementary Figure 1C&D. Consistent with the results above, the expression of PABPC1 was lower in GBM (p = 0.0028l χ2 test). Thus, both in RNA and protein expression level, PABPC1 expression was downregulated in gliomas with higher malignance.

    Figure 1. The expression level of PABPC1 in different grades and IDH status.

    (A & C) The expression difference of PABPC1 in grade II, grade III and GBM of CGGA and TCGA datasets. (B & D) The expression difference of PABPC1 in IDH-mutant and IDH-wildtype across all grades of CGGA and TCGA datasets.

    *p < 0.05; **p < 0.01; ***p < 0.001.

    CGGA: Chinese Glioma Genome Atlas; GBM: Glioblastoma multiforme; IDH: Isocitrate dehydrogenase; ns: Non-signgificant; OS: Overall survival; TCGA: The Cancer Genome Atlas.

    PABPC1 was a potential marker for proneural molecular subtype

    To depict the correlation between PABPC1 and molecular subtype, we further explored PABPC1 expression in different TCGA molecular subtype. In Figure 2A & B, our result showed that PABPC1 was highly expressed in proneural subtype in both datasets. We were inspired that PABPC1 might be a potential biomarker for proneural subtype. Hence, we drew the receiver operating characteristic curve for PABPC1 expression and proneural subtype. In CGGA, the area under the receiver operating characteristic curve was 0.786, and at the optimal cutoff point (0.268), the sensitivity and specificity were 65.1 and 82.8%, respectively (Figure 2C). In TCGA, the area under the receiver operating characteristic curve was 0.875, and at the optimal cutoff point (13.281), the sensitivity and specificity were 84.4 and 77.1%, respectively (Figure 2D). It revealed that PABPC1 was a potential marker for proneural subtype.

    Figure 2. PABPC1 expression in molecular subtypes and predictive value for proneural subtype.

    (A & B)PABPC1 was significantly increased in proneural subtype. (C & D) ROC curves exhibited highly sensitivity in predicting proneural subtype.

    ****p < 0.0001.

    ROC: Receiver operating characteristic; TCGA: The Cancer Genome Atlas.

    PABPC1 showed significant prognostic power

    To explore the prognostic value of PABPC1, we performed the Kaplan–Meier survival analysis. The patients were divided into two groups, high and low PAPBC1 groups, based on the median expression level. It showed that glioma patients of high PAPBC1 expression group had a significantly longer OS than PABPC1 low expression group in training dataset (Figure 3A; p = 0.0022). Next, we explored the prognostic value of PABPC1 in different grade gliomas and found that the OS difference was significant in WHO grade III (p = 0.029; Figure 3C) and GBM (p = 0.045; Figure 3D), except for grade II (p = 0.43; Figure 3B).

    Figure 3. The PABPC1 survival curves of all gliomas in Chinese Glioma Genome Atlas cohorts.

    Kaplan–Meier survival analysis showed that high expression of PABPC1 conferred a significantly better prognosis in all grade gliomas (A), grade III (C) and GBM (D) patients, except for grade II (B). Kaplan–Meier survival curves for LGG patients with IDH mutant and 1p/19q codeletion (E), IDH-mutant but not 1p/19q codeletion (F), IDH-wildtype (G). Kaplan–Meier survival curves also show the prognostic value of GBM patients with IDH-wildtype (H) and IDH-mutant (I) in the CGGA cohort. p-value is the result of a log-rank test between the two groups shown in each panel.

    CGGA: Chinese Glioma Genome Atlas; Codel: Codeletion; GBM: Glioblastoma multiforme; IDH: Isocitrate dehydrogenase; LGG: Lower-grade glioma; OS: Overall survival.

    The 2016 WHO guideline classified gliomas into five subtypes based on histopathology and IDH status and 1p19q codeletion [4]. Since five glioma subtypes had different characteristics and prognosis [16], we further evaluated the prognostic value of PABPC1 in five subgroups of patients. For LGG, survival time of high PABPC1 group was obviously longer than that of low PABPC1 group in LGG-IDHmut-codel (p = 0.041; Figure 3E), LGG-IDHmut-noncodel (p = 0.028; Figure 3F) and LGG-IDH-wild (p = 0.041; Figure 3G). There were similar differences in prognosis between two groups for GBM-IDHwt (p = 0.046; Figure 3H) and GBM-IDHmut (p = 0.046; Figure 3I).

    We also performed the survival analysis in TCGA dataset and obtained similar results though no statistical significance was detected in LGG-IDH-wild and GBM-IDHmut (Supplementary Figure 2). Therefore, PABPC1 was a positively prognosis predictor in gliomas, and higher expression of the PABPC1 indicated excellent survival.

    Functional annotation of PABPC1

    To better understand the underlying biological function of PABPC1, we filtered genes that were significantly correlated with PABPC1 in TCGA dataset (correlation |R| >0.5; p < 0.05) utilizing Pearson correlation analysis. As a result, 187 positively correlated genes and 116 negatively correlated genes met the criteria, respectively (Supplementary Table 2). We chose the positive genes (187 genes) for GO Analysis and detected enrichment in biological function of translation (False Discovery Rate [FDR] = 1.48E-23, Benjamini = 6.45E-24), cytoplasmic translation (FDR = 9.91E-05, Benjamini = 2.16E-05), formation of translation preinitiation complex (FDR = 0.001539, Benjamini = 2.24E-04) (Figure 4A & B & Supplementary Figure 3), which was consistent with previous research [17–20].

    Figure 4. PABPC1 was closely related to translation in gliomas.

    GO analysis showed PABPC1 was significantly associated with biological function of translation, cytoplasmic translation, formation of translation preinitiation complex in two datasets.

    GO: Gene Ontology; IDH: Isocitrate dehydrogenase; MGMT: O6-methylguanine-DNA methyltransferase promoter; TCGA: The Cancer Genome Atlas.

    Discussion

    Gliomas are associated with high disability and mortality rate, which seriously endanger human health and quality of life. Standards surgical resection with radiochemotherapy fail to improve survival of glioma patients substantially [2,21]. Hence, new diagnostic and treatment strategies are urgently needed. New biomarkers are currently being developed for identifying cancer risk, determining prognosis, detecting recurrence and predicting responses to specific drugs [22,23]. It is in urgent need to identify more biomarkers and establish new molecular classification systems for further precision medicine.

    In glioma patients, many prognosis biomarkers have been reported, such as IDH mutation, MGMTp methylation, 1p/19q codeletion, EGFR and so on [24]. And PD-L1 [25], TIM-3 [26], DCTD [27], pHH3 [28] and FGFR3 [29] have been demonstrated as biomarker in our previous study.

    We have noticed that control of mRNA stability and translation plays a crucial role in various biology processes, such as cell growth and neuronal plasticity [30,31]. mRNA-binding proteins (mRBPs) play a key role in maintaining mRNA stability and controlling transport and translation [32]. Hundreds of such mRBPs have been discovered and studied over the years [33]. Previous studies both in vivo and in vitro have confirmed that PABPC1 control cell proliferation [34–37], promotes cell migration and spreading [38], which is closely related to the malignant phenotype of cancer.

    At present, there are few reports on the characteristics and significance of PABPC1 in gliomas. We enrolled totally 1000 glioma patients with transcriptome data from CGGA and TCGA dataset in this study. We found that PABPC1 was upregulated in beneficial pathological type such as low grade, IDH1/2 mutation and 1p/19q co-deletion. Moreover, PABPC1 was preferentially expressed in proneural subtype. These results indicated that PABPC1 expression was related to beneficial biological process. These beneficial biological processes might contribute to tumor stability and therapy sensitivity.

    GO analysis of positively related genes gave us a real insight into the biological functions of PABPC1. We found that PABPC1 was significantly related to translation. Translation refers to the cellular metabolic process of protein formation, using mRNA to form specific polypeptide chain. Changes in translation can seriously affect gene expression and dysregulation of translation have been documented in several disorders, including cancer [39,40]. Meanwhile, the relationship between translation and many well-established cancer genes (e.g., PIK3CA, MYC, PTEN) has been deeply revealed [39,41]. For example, MYC hyperactivation increased translation of m7GpppN cap (eIF4F)-dependent mRNAs, resulting in chromosomal instability and cancer development [42]. Therefore, the role of translation function in cancer cannot be underestimated and the mechanism of PABPC1-regulating translation needs deeper investigation in gliomas.

    For prognosis analysis, survival curve showed that high-PABPC1 expression predicted significantly better OS in glioma either in whole grade gliomas, grade II, grade III or GBM. These results indicated that PABPC1 was a beneficial molecular prognosis marker with potential important therapeutic implications in gliomas. Due to the important role of PABPC1 in translation and gene stabilization, the mechanism of the downregulation of this gene in glioma patients with poor prognosis needs to be clarified.

    Nowadays, traditional cytotoxic agents no longer meet the requirements of modern precision medicine in glioma and an amount of selective, mechanism-based agents come forth. Our understanding of the gliomagenesis has improved, resulting in new treatment options like targeted therapy. This study will provide new insights for targeted therapy of gliomas.

    Conclusion

    We first summarized the molecular features and potential clinical value of PABPC1 in gliomas. PABPC1 is a favorable biomarker for gliomas and predicts better survival for patients of glioma. Our finding improves the understanding of PABPC1 as a novel therapeutic target for gliomas.

    Summary points
    • Gliomas represent the most prevalent and fatal brain tumor in adults. In order to improve diagnostic accuracy and treatment efficacy for the glioma patients, it is crucial to identify novel biomarkers and therapeutic targets.

    • Little is known about the genetic and epigenetic features of PABPC1 in gliomas.

    • To explore the clinical and molecular characteristics of PABPC1 in gliomas, we utilized the gene expression data from Chinese Glioma Genome Atlas dataset as training set and validated in The Cancer Genome Atlas dataset.

    • We detected PABPC1 expression was downregulated in gliomas with higher malignance.

    • PABPC1 was a potential marker for proneural molecular subtype.

    • PABPC1 was a positively prognosis predictor in gliomas, and higher expression of the PABPC1 indicated excellent survival.

    • Functional annotation revealed that PABPC1 was related to biological process of translation.

    • Our finding improves the understanding of PABPC1 as a novel biomarker with potential therapeutic connotations.

    Supplementary data

    To view the supplementary data that accompany this paper please visit the journal website at: www.futuremedicine.com/doi/suppl/10.2217/fon-2019-0268

    Financial & competing interests disclosure

    This study was funded by The National Key Research and Development Plan (grant number 2016YFC0902500); Beijing Science and Technology Plan (grant number Z141100000214009); Capital Medical Development Research Fund (grant number 2016-1-1072); The National Natural Science Foundation of China (grant number 8190101770); The National Natural Science Foundation of China (grant number 81903078). The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.

    No writing assistance was utilized in the production of this manuscript.

    Ethical conduct of research

    The authors state that they have obtained appropriate institutional review board approval or have followed the principles outlined in the Declaration of Helsinki for all human or animal experimental investigations. In addition, for investigations involving human subjects, informed consent has been obtained from the participants involved.

    Open access

    This work is licensed under the Attribution-NonCommercial-NoDerivatives 4.0 Unported License. To view a copy of this license, visit http://creativecommons.org/licenses/by-nc-nd/4.0/

    Papers of special note have been highlighted as: •• of considerable interest

    References

    • 1. Lapointe S, Perry A, Butowski NA. Primary brain tumours in adults. Lancet 392(10145), 432–446 (2018).
    • 2. Jiang T, Mao Y, Ma W et al. CGCG clinical practice guidelines for the management of adult diffuse gliomas. Cancer Lett. 375(2), 263–273 (2016).
    • 3. Omuro A, Deangelis LM. Glioblastoma and other malignant gliomas: a clinical review. JAMA 310(17), 1842–1850 (2013).
    • 4. Louis DN, Perry A, Reifenberger G et al. The 2016 World Health Organization classification of tumors of the central nervous system: a summary. Acta Neuropathol. 131(6), 803–820 (2016). •• This 2016 WHO classification of CNS tumors uses molecular parameters in addition to histology to define many tumor entities, thus formulating a concept for how CNS tumor diagnoses should be structured in the molecular era.
    • 5. Dai LX, Taylor MS, O’Donnell KA, Boeke JD. Poly(A) binding protein C1 is essential for efficient L1 retrotransposition and affects L1 RNP formation. Mol. Cell. Biol. 32(21), 4323–4336 (2012).
    • 6. Eliseeva IA, Lyabin DN, Ovchinnikov LP. Poly(A)-binding proteins: structure, domain organization, and activity regulation. Biochemistry (Mosc.) 78(13), 1377–1391 (2013).
    • 7. Gray NK, Hrabalkova L, Scanlon JP, Smith RW. Poly(A)-binding proteins and mRNA localization: who rules the roost? Biochem. Soc. Trans. 43(6), 1277–1284 (2015).
    • 8. Grange T, de Sa CM, Oddos J, Pictet R. Human mRNA polyadenylate binding protein: evolutionary conservation of a nucleic acid binding motif. Nucleic Acids Res. 15(12), 4771–4787 (1987).
    • 9. Takashima N, Ishiguro H, Kuwabara Y et al. Expression and prognostic roles of PABPC1 in esophageal cancer: correlation with tumor progression and postoperative survival. Oncol. Rep. 15(3), 667–671 (2006).
    • 10. Zhu J, Ding H, Wang X, Lu Q. PAB PC1 exerts carcinogenesis in gastric carcinoma by targeting miR-34c. Int. J. Clin. Exp. Pathol. 8(4), 3794–3802 (2015).
    • 11. Erinjeri NJ, Nicolson NG, Deyholos C, Korah R, Carling T. Whole-exome sequencing identifies two discrete druggable signaling pathways in follicular thyroid cancer. J. Am. Coll. Surg. 266 (6), 950 959 (2018).
    • 12. Chen R, Feng C, Xu Y. Cyclin-dependent Kinase-associated protein Cks2 is associated with bladder cancer progression. J. Int. Med. Res. 39(2), 533–540 (2011).
    • 13. Yan W, Zhang W, You G et al. Correlation of IDH1 mutation with clinicopathologic factors and prognosis in primary glioblastoma: a report of 118 patients from China. PLoS ONE 7(1), e30339 (2012).
    • 14. Brennan CW, Verhaak RG, Mckenna A et al. The somatic genomic landscape of glioblastoma. Cell 155(2), 462–477 (2013).
    • 15. Huang DW, Sherman BT, Lempicki RA. Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat. Protoc. 4(1), 44–57 (2009).
    • 16. Brat DJ, Verhaak RG, Aldape KD et al. Comprehensive, integrative genomic analysis of diffuse lower-grade gliomas. New Engl. J. Med. 372(26), 2481–2498 (2015).
    • 17. Sawazaki R, Imai S, Yokogawa M et al. Characterization of the multimeric structure of poly(A)-binding protein on a poly(A) tail. Sci. Rep. 8(1), 1455 (2018).
    • 18. Behm-Ansmant I, Gatfield D, Rehwinkel J, Hilgers V, Izaurralde E. A conserved role for cytoplasmic poly(A)-binding protein 1 (PABPC1) in nonsense-mediated mRNA decay. Embo. J. 26(6), 1591–1601 (2007).
    • 19. Peixeiro I, Inacio A, Barbosa C, Silva AL, Liebhaber SA, Romao L. Interaction of PABPC1 with the translation initiation complex is critical to the NMD resistance of AUG-proximal nonsense mutations. Nucleic Acids Res. 40(3), 1160–1173 (2012).
    • 20. Wydro M, Bobrowicz A, Temperley RJ, Lightowlers RN, Chrzanowska-Lightowlers ZM. Targeting of the cytosolic poly(A) binding protein PABPC1 to mitochondria causes mitochondrial translation inhibition. Nucleic Acids Res. 38(11), 3732–3742 (2010).
    • 21. Morgan LL. The epidemiology of glioma in adults: a “state of the science” review. Neuro Oncol. 17(4), 623–624 (2015).
    • 22. Ilyin SE, Belkowski SM, Plata-Salaman CR. Biomarker discovery and validation: technologies and integrative approaches. Trends Biotechnol. 22(8), 411–416 (2004).
    • 23. Bensalah K, Montorsi F, Shariat SF. Challenges of cancer biomarker profiling. Eur. Urol. 52(6), 1601–1609 (2007).
    • 24. Siegal T. Clinical relevance of prognostic and predictive molecular markers in gliomas. Adv. Tech. Stand. Neurosurg (43), 91–108 (2016). •• Summarizes the molecular markers that carry both diagnostic and prognostic information and add useful tools to traditional classification by redefining tumor subtypes within each WHO category.
    • 25. Wang Z, Zhang C, Liu X et al. Molecular and clinical characterization of PD-L1 expression at transcriptional level via 976 samples of brain glioma. Oncoimmunology 5(11), e1196310 (2016).
    • 26. Li G, Wang Z, Zhang C et al. Molecular and clinical characterization of TIM-3 in glioma through 1,024 samples. Oncoimmunology 6(8), e1328339 (2017).
    • 27. Hu H, Wang Z, Li M et al. Gene expression and methylation analyses suggest DCTD as a prognostic factor in malignant glioma. Sci. Rep. 7(1), 11568 (2017).
    • 28. Zhu P, Zhang CB, Yang P et al. Phosphohistone H3 (pHH3) is a prognostic and epithelial to mesenchymal transition marker in diffuse gliomas. Oncotarget 7(29), 45005–45014 (2016).
    • 29. Wang Z, Zhang C, Sun L et al. FGFR3, as a receptor tyrosine kinase, is associated with differentiated biological functions and improved survival of glioma patients. Oncotarget 7(51), 84587–84593 (2016).
    • 30. Costa-Mattioli M, Sossin WS, Klann E, Sonenberg N. Translational control of long-lasting synaptic plasticity and memory. Neuron 61(1), 10–26 (2009).
    • 31. Sonenberg N, Hinnebusch AG. New modes of translational control in development, behavior, and disease. Mol. Cell 28(5), 721–729 (2007).
    • 32. Dreyfuss G, Kim VN, Kataoka N. Messenger-RNA-binding proteins and the messages they carry. Nat. Rev. Mol. Cell Biol. 3(3), 195–205 (2002).
    • 33. Hentze MW, Castello A, Schwarzl T, Preiss T. A brave new world of RNA-binding proteins. Nat. Rev. Mol. Cell Biol. 19(5), 327–341 (2018).
    • 34. Stupfler B, Birck C, Seraphin B, Mauxion F. BTG2 bridges PABPC1 RNA-binding domains and CAF1 deadenylase to control cell proliferation. Nat. Commun. 7, 10811 (2016). •• Indicates that PABPC1 RRM and BTG2 APRO domains were sufficient to stimulate CAF1 deadenylase activity in vitro and interaction of BTG2 with the first RRM domain of PABPC1 was required for BTG2 to control cell proliferation.
    • 35. Katzenellenbogen RA, Vliet-Gregg P, Xu M, Galloway DA. Cytoplasmic poly(A) binding proteins regulate telomerase activity and cell growth in human papillomavirus type 16 E6-expressing keratinocytes. J. Virol. 84(24), 12934–12944 (2010).
    • 36. Zhang H, Sheng C, Yin Y et al. PABPC1 interacts with AGO2 and is responsible for the microRNA mediated gene silencing in high grade hepatocellular carcinoma. Cancer Lett. 367(1), 49–57 (2015).
    • 37. Eisermann K, Dar JA, Dong J, Wang D, Masoodi KZ, Wang Z. Poly (A) binding protein cytoplasmic 1 is a novel co-regulator of the androgen receptor. PLoS ONE 10(7), e0128495 (2015).
    • 38. Copsey AC, Cooper S, Parker R et al. The helicase, DDX3X, interacts with poly(A)-binding protein 1 (PABP1) and caprin-1 at the leading edge of migrating fibroblasts and is required for efficient cell spreading. Biochem. J. 474(18), 3109–3120 (2017). •• Indicates that DDX3X could physically interact and co-localize with poly(A)-binding protein 1 and caprin-1 at the leading edge of spreading cells to promote cell migration and spreading.
    • 39. Ruggero D. Translational control in cancer etiology. Cold Spring Harb. Perspect. Biol. 5(2), a012336 (2013).
    • 40. Bhat M, Robichaud N, Hulea L, Sonenberg N, Pelletier J, Topisirovic I. Targeting the translation machinery in cancer. Nat. Rev. Drug Discov. 14(4), 261–278 (2015).
    • 41. Silvera D, Formenti SC, Schneider RJ. Translational control in cancer. Nat. Rev. Cancer 10(4), 254–266 (2010).
    • 42. Barna M, Pusic A, Zollo O et al. Suppression of Myc oncogenic activity by ribosomal protein haploinsufficiency. Nature 456(7224), 971–975 (2008).