Skip to main content
Molecular Therapy. Nucleic Acids logoLink to Molecular Therapy. Nucleic Acids
. 2021 Aug 8;26:417–431. doi: 10.1016/j.omtn.2021.07.024

Noncoding RNAs in pediatric brain tumors: Molecular functions and pathological implications

Shaohuai Chen 1,2, Xiangyang Deng 1,2, Hansong Sheng 1, Yuxi Rong 1, Yanhao Zheng 1, Yusong Zhang 1, Jian Lin 1,
PMCID: PMC8426460  PMID: 34552822

Abstract

Brain tumors are common solid pediatric malignancies and the main reason for cancer-related death in the pediatric setting. Recently, evidence has revealed that noncoding RNAs (ncRNAs), including microRNAs (miRNAs), long ncRNAs (lncRNAs), and circular RNAs (circRNAs), play a critical role in brain tumor development and progression. Therefore, in this review article, we describe the functions and molecular mechanisms of ncRNAs in multiple types of cancer, including medulloblastoma, pilocytic astrocytoma, ependymoma, atypical teratoid/rhabdoid tumor, glioblastoma, diffuse intrinsic pontine glioma, and craniopharyngioma. We also mention the limitations of using ncRNAs as therapeutic targets because of the nonspecificity of ncRNA targets and the delivery methods of ncRNAs. Due to the critical role of ncRNAs in brain oncogenesis, targeting aberrantly expressed ncRNAs might be an effective strategy to improve the outcomes of pediatric patients with brain tumors.

Keywords: lncRNAs, miRNAs, circRNAs, brain tumor, therapy

Graphical abstract

graphic file with name fx1.jpg


In this review article, the authors describe the functions and molecular mechanisms of ncRNAs, such as miRNAs, lncRNAs, and circRNAs, in pediatric brain malignancies, including medulloblastoma, pilocytic astrocytoma, ependymoma, atypical teratoid/rhabdoid tumor, glioblastoma, diffuse intrinsic pontine glioma, and craniopharyngioma.

Introduction

Brain tumors are the most frequent solid pediatric malignancies and the main cause of cancer-related death in the pediatric setting.1, 2, 3 The World Health Organization (WHO) classification of tumors of the central nervous system (CNS) includes diffuse astrocytic and oligodendroglial tumors, other astrocytic tumors, ependymal tumors, other gliomas, choroid plexus tumors, neuronal and mixed neuronal-glial tumors, tumors of the pineal region, embryonal tumors, and tumors of the cranial and paraspinal nerves.4 Medulloblastoma (MB), pilocytic astrocytoma (PA), ependymoma (EPN), and atypical teratoid/rhabdoid tumor (ATRT) have become important health problems with adverse medical consequences in children. Treatment of these tumors usually requires a multimodality approach that includes surgical intervention, radiotherapy, and chemotherapy. Nonetheless, since the developing nervous system is highly susceptible to damage from these conventional therapeutic strategies and resistance to therapeutic drugs can occur, the treatment of malignant pediatric brain tumors still faces difficult challenges.2,5

Great advances in molecular genetics, epigenetics, and cellular biology have provided a wealth of clinically and biologically significant insights into these deadly childhood diseases, potentially enabling the development of more effective and less toxic treatment strategies. Noncoding RNAs (ncRNAs) are emerging as essential regulators of diverse biological processes, including human oncogenesis and tumor progression. Recently, ncRNAs have attracted increasing attention because they lack the capacity to encode proteins.6, 7, 8 Based on a 200-nt cutoff in mature transcript length, ncRNAs are commonly divided into small ncRNAs (sncRNAs, 18–200 nt) and long ncRNAs (lncRNAs, >200 nt).9 To date, several kinds of sncRNAs have been well defined, including microRNAs (miRNAs), small nuclear RNAs (snRNAs), piwi-interacting RNAs (piRNAs), and small nucleolar RNAs (snoRNAs).10, 11, 12 According to their genomic localization and evolutionary lineage, lncRNAs are classified as long intergenic RNAs (lincRNAs), antisense RNAs, sense intronic RNAs, enhancer RNAs (eRNAs), and pseudogenes.13 As a new research hotspot in the field of miRNAs and lncRNAs, many circular RNAs (circRNAs) have also been observed in and associated with distinct cancers.14,15 These molecules play a pivotal role in all critical biological processes controlling various levels of gene expression via epigenetic modification, transcription, RNA splicing, and scaffold assembly.7,16,17 Moreover, their aberrant expression is confirmed to be involved in oncogenesis and disease progression, making them a new class of potential treatment targets with broad applicability.

With the number of ncRNAs steadily increasing due to the rapid development of high-throughput sequencing and bioinformatics technologies, it is necessary to summarize the current research progress on ncRNAs in pediatric brain tumors. A deeper understanding of the function and mechanism of action of ncRNAs will drive the development of new treatment strategies for pediatric neuro-oncology. Herein, we conducted a systematic literature review regarding the deregulation of ncRNAs with a focus on miRNAs, lncRNAs, and circRNAs, as well as the pathological implications for the biology of pediatric CNS tumors to provide insights into the diagnostic, prognostic, and therapeutic potential of ncRNAs (Tables 1 and 2).

Table 1.

lncRNAs in brain tumors

LncRNAs Expression Phenotype Downstream targets Refs.
Gm15577 tumorigenesis Negr1 18
CRNDE proliferation, apoptosis, migration, invasion miR-29c-3p 19,20
linc-NeD125 proliferation, migration, invasion miR-19a-3p, miR-19b-3p, miR-106a-5p, CDK6, MYCN, SNCAIP 21
Nkx2-2as tumorigenesis miR-103/107/BTG2/Tis21/PC3, miR-548 m/LATS1/2 22
UCA1 proliferation, migration PI3K/AKT pathways 23
CCAT1 proliferation, migration MAPK pathway 24
LOXL1-AS1 growth, migration PI3K/AKT 25
TP73-AS1 survival, migration, proliferation miR-494-3p, EIF5A2 26,27
HOTAIR proliferation, migration, invasion, apoptosis, EMT miR-1/miR-206-YY1, HOXA9, miR-125, miR-219 28, 29, 30, 31, 32, 33, 34, 35, 36, 37
LINC00899 invasion, migration RBL2 38
TRERNA1 EMT Snail 39
MALAT proliferation, progression miR-155, miR-199a, FBXW7, ZHX1 40, 41, 42, 43, 44
DGCR5 EMT epithelial markers 45

Table 2.

circRNAs in glioblastoma

CircRNAs Expression Phenotype Downstream targets Refs.
circ-FBXW7 proliferation, cell cycle FBXW7-185aa 46
circ-SHPRH tumorigenicity SHPRH-146aa 47
circSMARCA5 migration SRSF1/SRSF3/PTB 48,49
hsa_circ_0008344 proliferation, migration, invasion, colony formation, apoptosis miR-433-3p/miR-450b-3p 50
circNT5E proliferation, migration, invasion miR-422a 51
circPINTexon2 proliferation PINT87aa 52
circMMP9 proliferation, invasion, metastasis miR-124 53
circ-0029426 proliferation, migration, invasion, apoptosis miR-197 54
circ-0074027 growth, invasion miR-518a-5p/IL17RD 55
circ-0067934 proliferation, metastasis PI3K-AKT pathway 56
circ-0001730 proliferation, invasion miR-326/Wnt7B axis 57
circMTO1 proliferation miR-92/WWOX 58
circ-AKT3 proliferation, radioresistance AKT-Thr308 59
circ-PITX1 progression miR-379-5p/MAP3K2 60
circFOXO3 progression miR-138-5p, miR-432-5p 61
circ-0001801 proliferation, migration, invasion, EMT miR-628-5p/HMGB3 axis 62
circ-EPB41L5 progression EPB41L5/p-AKT 63
circENTPD7 proliferation, motility miR-101-3p/ROS1 64

PAs

PAs are the most frequent CNS neoplasms in childhood, accounting for approximately 20% of all pediatric brain tumors with an average annual age-adjusted incidence rate of 0.8.65, 66, 67 PAs are usually considered relatively benign (WHO grade I) tumors with a 10-year survival rate >90%.65 These tumors occur throughout the CNS, with the cerebellum being the most frequent location in the pediatric setting. Biologically, alterations in the mitogen-activated protein kinase (MAPK) signaling pathway, KIAA1549-BRAF fusions, and neurofibromatosis type 1 (NF1) syndrome have been shown to impact PA development.68, 69, 70

miRNAs and lncRNAs in PAs

Compelling evidence has identified a number of aberrantly expressed miRNAs in PAs, which has aided the discovery of novel diagnostic methods and effective treatments for this type of tumor. A survey of miRNA expression demonstrated that miR-142-5p and miR-25 were significantly upregulated in PAs compared to normal tissue, while miR-129 was strongly downregulated. Compared to those in other CNS pediatric tumors (ATRT, EPN, MB, and glioblastoma), multiple miRNAs, including miR-93 and miR-106b, were observed to be downregulated, whereas several miRNAs, such as miR-432 and miR-34a, were found to be upregulated in PSs.71 Dysregulated expression levels of a subset of miRNAs, including decreased expression of miR-129 and miR-124 and overexpression of miR-21, were also observed in PAs. In addition, miR-650 and miR-1276 levels were increased, while miR-744∗ and miR-187∗ levels were decreased, in NF1-associated tumors among the PA subgroups.72 Similarly, miR-15 and miR-24-1 levels were reported to be decreased in PAs.73

Jones et al.74 also found that the Xq26.3 cluster, miR-224, miR-146a, miR-34a, and the miR-106a∼miR-363 cluster were upregulated, while miR-124, miR-129, and miR-218 were downregulated. Predicted targets of differentially regulated miRNAs frequently include components of the extracellular signal-regulated kinase (ERK)/MAPK and nuclear factor κB (NF-κB) signaling pathways. Another study identified 88 miRNAs that were expressed to different degrees between PA and cerebral white matter samples.75 PA samples had the most downregulated miRNAs regulating classical pathways of tumorigenesis, while the most overexpressed miRNAs were associated with pathways such as focal adhesion, the p53 signaling pathway, and gliomagenesis. High expression of miR-34a-5p and miR-144-3p and low expression of miR-630 and miR-139-3p were further confirmed by qRT-PCR.75 Yuan et al.76 also demonstrated that miR-125 family members were downregulated in PAs compared to nonneoplastic brain, and overexpression of miR-125b in pediatric low-grade glioma decreased cell growth and invasion and induced apoptosis. Furthermore, one study analyzed the expression of lncRNA HOTAIR in five pediatric tumor types and found higher expression of this gene in juvenile PAs.28

MBs

MBs, as embryonal tumors of the cerebellum, account for approximately 20% of the total brain tumors in this patient population.67,77,78 Advances in treatment with neurosurgery, radiation therapy, and high-dose chemotherapy have significantly improved the survival rate of these patients. However, long-term sequelae, including neurocognitive, neuroendocrine, and psychosocial deficits caused by intensive therapies administered to the developing brain, remain challenging. Therefore, more effective molecular-targeted strategies with less toxicity are urgently needed to be developed for this disease.67,77

Considering the nature of its molecular heterogeneity, a great deal of genomic research has helped classify MB into four subgroups: Wingless (WNT), Sonic hedgehog (SHH), group 3, and group 4.79 These subgroups have different genetic alterations, clinical features, and results. WNT tumors, characterized by activated WNT signaling, occur primarily in children older than 3 years of age and exhibit a balanced sex ratio. These tumors have few metastases and have a favorable prognosis, with a 5-year survival rate of more than 95%.77,80, 81, 82 Although WNT MB has been considered a largely homogeneous cluster, the extent of heterogeneity within the subgroups was further analyzed. WNT-α and WNT-β are two molecular subtypes that have been identified, and their differences are age at diagnosis and frequency of monosomy 6.83 SHH tumors are characterized by activation of the SHH pathway and have a 5-year survival rate of 75%, which is worse than that of WNT patients. This MB subtype presents a bimodal age distribution, with most cases being diagnosed in both infants and adults. Recently, four molecular subtypes of SHH MB, that is, SHH-α, SHH-β, SHH-γ and SHH-δ, have been described based on gene expression data and DNA methylation.78,83 A defining feature of group 3 MB is the high level of MYC amplification, which accounts for approximately 25% of MBs with the worst prognosis. Group 4 MB is the least known of the MB subgroups, and its molecular profiles are not as well characterized. Many different subtypes of groups 3 and 4 MBs have also been proposed.78,84, 85, 86

Role of miRNAs in MBs

Along with increasing research on the ncRNA domain, increasing evidence supports the important roles of various miRNAs in MB. In this respect, the expression of miR-124 was reported to be significantly decreased in MB cells and tumor tissues.87 Further in vivo and in vitro experiments demonstrated that miR-124, acting as a tumor suppressor, inhibited tumor cell growth by targeting cyclin-dependent kinase 6 (CDK6), which is a member of the family of serine-threonine kinases that promotes cell cycle progression.87,88 SLC16A1 overexpression could promote cell proliferation and was found to be regulated by miR-124 in MB.89 In addition, the nuclear receptor Nur77, encoded by the NR4A1 gene, is commonly upregulated in MB and leads to a proliferative state that promotes cancer progression, and it was also reported to be another target of miR-124.90 In addition, miR-199b-5p expression was found to be downregulated in MB via epigenetic methylation.91,92 miR-199b-5p expression can cause specific damage to the cancer stem cell (CD133+) population through negative regulation of the transcription factor HES1, which is a principal Notch-responsive factor.92 Its obvious downregulation in metastatic MBs also suggests a potential silencing mechanism that acts through epigenetic or genetic alterations.92 Moreover, as a marker of MB tumor-propagating cells, CD15 is an additional direct target of miR-199b-5p.91

As a Notch signaling pathway regulator, miR-34a can regulate DLL1, Jagged1, Notch1, and Notch2. Re-expression of miR-34a in MB cell lines strongly inhibited cell cycle progression, proliferation, survival, migration, and invasion, and it caused apoptosis and downregulated the expression of miR-34a targets, including c-Met, SIRT1, and MYCN proteins.93, 94, 95 miR-34a deficiency also accelerated MB genesis in vivo.95 In addition, it has been reported that this miRNA could render MB cells more sensitive to chemotherapeutic agents through the adjustment of MAGE-A and p53.96,97 Comparatively, with regard to chemotherapeutic resistance, SPARC-mediated cisplatin resistance could be regulated by miR-let-7f-1 through the let-7f-1 miRNA/HMGB1 axis in MB cells.98 Furthermore, one study revealed the roles of miR-584-5p in the regulation of DNA repair, microtubule dynamics, and stemness in MB, the potentiation of vincristine, and the radiation response via the miR-584-5p/HDAC1/eIF4E3 axis.99 In addition, miR-31 activated the phosphatidylinositol 3-kinase (PI3K)/AKT and NF-κB pathways, contributing to cisplatin resistance, and inducing cell growth, invasion, and migration in MB cells.100

miR-9 and miR-125a promote cell growth arrest and apoptosis in MB cells through modulation of the pro-proliferative truncated TrkC (t-TrkC) isoform.101 It has been verified that miR-9 is a methylation-silenced tumor suppressor contributing to disease pathogenesis through regulation of HES1 oncogenic activity.102 A survey of miRNA expression indicated that, relative to their expression in normal brain tissue, multiple miRNAs, including miR-216 and miR-340, were upregulated, whereas several miRNAs, such as miR-146b and miR-23a, were downregulated in MB.71 A high-throughput miRNA microarray was performed, and some miRNAs were confirmed to be downregulated in MB, including miR-17, miR-100, miR-106b, and miR-218. The predicted target genes are involved in MB development.103 miR-217, miR-216, miR-183, miR-182, and miR-96 were found to be upregulated in tumor tissue through analysis of the GEO miRNA expression database, whereas miR-383, miR-206, miR-138, miR-128a/b, and miR-133b were identified to be downregulated.104 Inhibition of miR-217 was further confirmed to induce apoptosis and reduce migration and invasion in MBs.105 miR-128a was found to inhibit cell growth by targeting Bmi-1, resulting in increased steady-state levels of superoxide and cellular senescence in MB.106

Upregulation of miR-383 expression decreased PRDX3 expression, leading to cell apoptosis and inhibition of proliferation in MBs.107 miR-31 was reported to suppress MB tumorigenesis by negatively regulating DNA replication via MCM2.108 Arhgef6, which is upregulated in human MBs and involved in mediating experimental medulloblastomagenesis, was repressed by miR-135a.109 Transient overexpression of miR-367, which is upregulated by OCT4 in MB cells, conspicuously enhanced proliferation, invasion, and generation of neurosphere-like structures, which are enriched in CD133-expressing cells.110 Additionally, miRNA-10b contributed to MB tumorigenesis with Bcl-2 as a mediator of the effects on MB cell survival.111 Restoration of miR-30a, miR-221-3p, and miRNA-4521 expression inhibited the proliferation, clonogenic potential, and tumorigenicity of MB cells.112, 113, 114 Moreover, miR-378 downregulated the activity of UHRF1, leading to modulating MB cell proliferation and apoptosis.115 The miR-512-2 gene was deleted in one-third of MBs associated with overexpression of MYCC, which was significantly correlated with tumor anaplasia and poor prognosis.116

Several studies reported that increased expression of miR-21 could promote cancer cell migration in MB.117,118 Moreover, miR-21 was proven to act on PDCD4, regulating the expression of multiple invasion- and metastasis-related proteins, including MAP4K1, JNK, E-cadherin, and TIMP2.117 A link with the STAT3/miR-21/PIAS3 circuitry that could mediate MB progression and metastasis was also established.118 Similarly, miR-182 was found to help accelerate leptomeningeal metastatic dissemination in non-SHH MBs.119 One group reported that miR-192 modulated the expression of DHFR, integrins, and CD47 to modulate cell proliferation and anchoring, resulting in the suppression of leptomeningeal dissemination in MBs.120 A miRNA-1280/JAG2 network was found to be associated with MB metastatic dissemination and patient outcomes.121 Expression of miR-210 might promote metastasis, and miR-206 had a suppressive role in MB viability and migration by targeting LASP1 and OTX2.122, 123, 124 Moreover, re-expression of miR-218 decreased cell growth, cell colony formation, cell migration, invasion, and tumor sphere size in MBs by directly regulating SH3GL1, and CDK6, RICTOR, and cathepsin B (CTSB) might be additional targets.125,126 The absence of miR-219 could enhance cell proliferation, invasion, and migration through regulation of CD164.127,128 PTEN is a direct target of miR-106, and deletion of miR-106b inhibits cell proliferation, migration, invasion, and tumor sphere formation through suppression of PTEN.129

Notably, efforts were made to characterize four consensus molecular subgroups using miRNA profiles. Many miRNAs, such as miR-193a, the miR-224/miR-452 cluster, the miR-182/miR-183/miR-96 cluster, and miR-148a, were found to be overexpressed in WNT MB. Overexpression of miR-193a and miR-224, which are upregulated WNT pathway-specific miRNAs, was verified to inhibit proliferation, increase radiation sensitivity, and inhibit anchorage-independent growth of MB cells.130 Upregulation of miR-193a-3p, miR-224, miR-148a, miR-23b, and miR-365 in WNT subgroup tumors was further validated in a study using 103 MB patients. This study indicated that miR-10b was increased in WNT MBs, followed by group 3 subtypes, while miR-182, miR-135b, and miR-204 were downregulated in SHH variants. miR-376a had higher expression in group 4 MB than in group 3 subtypes, and miR-592 was upregulated in group 4 MB. In addition, miR-135b was detected at low expression levels in groups 3 and 4 MBs.131 Accordingly, miR-148a might contribute to downregulating metastatic incidence and upregulating survival of WNT MB. High expression of miR-148a was confirmed to inhibit proliferation, clonogenic potential, invasion potential, and tumorigenicity of MB cells by targeting neuropilin 1.132 miR-499a, a candidate tumor suppressor gene, was found to be a potential marker for WNT MB.133,134

A study confirmed that downregulation of miR-125b, miR-324-5p, and miR-326 could be associated with the modulation of endogenous SHH target genes, including Smo, Gli1, and Pitch, and re-expression of these miRNAs in MB cells suppressed progenitor and tumor cell growth.135,136 Among them was miR-326, which is associated with the development of tumor stem cells derived from SHH MB. Expression of miR-326 was further confirmed to inhibit the Hh/Gli signaling pathway, impairing MB cell proliferation and self-renewal by negatively regulating Smo and Gli2.137 Three miRNA clusters, i.e., miR-183∼96∼182, miR-17-92, and miR-106b∼25, functionally collaborated with the SHH signaling pathway in MB development in mice.137, 138, 139, 140, 141, 142 miR-183∼96∼182 was further identified as protumorigenic in MYC-driven MB through suppression of apoptosis, modulation of the mTOR pathway, and control of motility.143 Analysis of human MBs also showed three miR-17∼92 cluster miRNAs, and the SHH signaling pathway could be constitutively activated by miR-92, miR-19a, and miR-20 overexpression.140 Inhibition of MB cell proliferation and tumor growth in vivo by silencing miR-17 and miR-19a was observed.139 Furthermore, overexpression of miR-17∼92 was also associated with upregulation of MYC expression.138 Likewise, overexpression of miR-106b in precursor cells promoted SHH pathway activation.142 miR-106b also positively regulated Gli2 transcription to promote granule cell expansion.144 Moreover, low expression of miR-466f could affect the Vegfa/Nrp2 pathway, thus sustaining the mesenchymal phenotype of SHH MB stem cells.145 miR-218 expression was decreased in the SHH and group 3 MBs.125,126 Notably, decreased expression levels of miR-182 and miR-183 in SHH MB compared to non-SHH MB were also observed.119 A recent study identified 19 miRNAs that exhibited MB group 4-specific expression compared to the other subgroups.146

lncRNAs and circRNAs in MBs

Recent studies have revealed that the expression of many lncRNAs are dysregulated in MBs (Figure 1). lncRNA Gm15577 was found to be specifically expressed in the mouse cerebellum in a developmentally regulated manner by targeting Negr1, which had a distinct expression pattern in MB patients from normal patients. Gm15577 might be associated with tumorigenesis of MBs.18 Overexpression of lncRNA CRNDE promoted tumor growth both in vitro and in vivo by arresting cell cycle progression and inhibiting apoptosis.19 One study reported that interference with lncRNA SPRY4-IT1 expression inhibited cell proliferation, invasion, and metastasis of MB cells.147 In addition, lncRNA linc-NeD125 was observed to be significantly overexpressed in group 4 MBs. Further in vitro experiments proved that linc-NeD125 acted as a competing endogenous RNA (ceRNA) by sequestering three miRNAs, that is, miR-19a-3p, miR-19b-3p, and miR-106a-5p, which derepressed the major driver factors CDK6, MYCN, and SNCAIP in group 4 MBs.21 Thus, downregulation of linc-NeD125 expression inhibited group 4 cell proliferation. In addition, it was proven that ectopic expression of linc-NeD125 also attenuated cell proliferation, migration, and invasion in aggressive group 3 MBs.21

Figure 1.

Figure 1

Roles of lncRNAs in medulloblastoma

Downregulation of the lncRNA Nkx2-2as was found to contribute to tumorigenesis when SHH signaling in cerebellar granule cells was constitutively activated. Nkx2-2as functions as a ceRNA to sequester miR-103/107 and miR-548 m, and it downregulated the tumor suppressors BTG2/Tis21/PC3 and LATS1/2, promoting tumor growth both in vitro and in vivo.22 Elevated expression of the lncRNAs UCA1 and CCAT1 was also observed in MB specimens, and knockdown of UCA1 and CCAT1 significantly suppressed MB cell proliferation and migration.23,24 Gao et al.25 provided both in vitro and in vivo evidence that downregulation of the lncRNA LOXL1-AS1 impaired tumor cell growth and migration through the PI3K/AKT pathway, displaying a potent pro-oncogenic function in MB.

One group reported that the lncRNA TP73-AS1 was clinically relevant in MB and could promote the survival, migration, and proliferation of MB cells in vitro and tumorigenicity in vivo.26 Mechanistically, TP73-AS1 was further found to positively regulate EIF5A2 expression by sponging miR-494-3p.27 Moreover, knockdown of the lncRNA HOTAIR inhibited MB cell proliferation, tumor growth, migration, and invasion and promoted cell apoptosis via regulation of the miR-1/miR-206-YY1 axis and epithelial-to-mesenchymal transition (EMT).29 Likewise, depletion of CRNDE also suppressed MB cell proliferation, apoptosis, migration, invasion, and chemosensitivity to cisplatin by binding to miR-29c-3p.20 Based on next-generation sequencing for discovery and qRT-PCR for validation, circ-SKA3 and circ-DTL were proven to be overexpressed in MB tissues compared with normal cerebellar tissues, whereas circ-CRTAM, circ-MAP3K5, circ-RIMS1-1, and circ-FLT3-1 were significantly downregulated.148 Downregulation of circ-SKA3 and circ-DTL was further confirmed to suppress MB cell proliferation, migration, and invasion by regulating the expression of host genes.148

EPN

As the third most common pediatric brain tumor, EPN mainly occurs in children under 5 years of age.3,149 This devastating disease was thought to originate from ependymal cells located in the lining of ventricular surfaces in the brain, and it occurs most commonly at the midline or lateral compartments of the posterior fossa in children.150 The current therapeutic strategy for pediatric EPN remains maximal safe surgical resection of the tumor combined with radiotherapy; however, this treatment seriously affects the growth and development of pediatric patients. Based on the WHO classification of CNS tumors, EPNs have been traditionally subdivided into distinct entities and histological variants.151 However, the utility of the WHO grade-based risk classification is controversial and inconclusive due to its limited predictive power.149,152,153 Recent advances in the biological characterization of ependymal tumors have distinguished nine molecular subgroups that appear to reflect more precise clinicopathological and molecular features, with three occurring in each anatomic compartment, exhibiting the potential for guiding therapeutic decisions.154 Therefore, the discovery of new molecular biomarkers and potential mechanisms has a great impact on the understanding of EPN.

miRNAs in EPN

It is encouraging to note that a number of miRNAs have been identified to associate with the biology of ependymal tumors and serve as potential candidates for molecular therapeutic targets. Analysis of microarray data showed an upregulation of miR-34b, miR-34c, miR-200a, miR-200b, and miR-483 in EPN, while miR-124a, miR-137, miR-138, miR-193b, and miR-181d appeared to be downregulated in EPN samples.71 Costa et al.155 identified 28 miRNAs differentially expressed in EPNs compared to normal controls via miRNA expression profiling. miR-34a and miR-135a were further verified to be overexpressed, while miR-485-5p was downregulated. Another study also identified that the miR-135a-3p, miR-137, miR-17-5p, miR-181d, and let-7d-5p were upregulated in EPNs.156 Low expression of miR-10a and high expression of miR-10b and miR-29a in EPN were also validated by qRT-PCR.157

Specifically, miR-17-5p, miR-19a-3p, miR-106b-5p, miR-124-3p, and miR-203a were shown to be differentially expressed between grade II and III EPNs.156,158,159 These miRNAs were overexpressed in posterior fossa EPNs, including miR-106-b-5p and miR-19a-3p.158 Moreover, miR-203, miR-17-5p, miR-124-3p, miR-192-5p, miR-221-3p, miR-222-3p, miR-326, miR-371a-5p, and miR-520g-3p were significantly correlated with tumor relapse.155,158, 159, 160 Furthermore, let-7d, miR-596, miR-367, miR-203, miR-17-5p, miR-124-3p, miR-203, miR-15a, and miR-24-1 were found to be associated with overall survival.73,155,158,159,161 The relationship between miRNA expression and tumor treatment response has also been addressed. For example, miR-135a and miR-146b were found to be associated with a low-response phenotype, which could lead to recurrence of the tumor.157

Mechanistically, through a study on pediatric spine EPNs by Lourdusamy et al.,162 miR-10b and miR-10a were found to be upregulated and targeted chromatin modification genes. miR-124, a tumor suppressor, was downregulated in pediatric spine EPNs and repressed cell-cell communication and genes involved in metabolic processes. Yang et al.163 performed a miRNA-mRNA network analysis and identified six crucial miRNAs, including miR-34a-5p, miR-449a, miR-106a-5p, miR-124-3p, miR-128-3p, and miR-330-3p, that might be utilized as biomarkers and potential therapeutic targets for EPN. Based on miRNA-mRNA covariation and sequence-based target predictions, miR-29a/c was identified as a regulator of LAMA2, revealing a key mechanism for molecular pathogenesis.164 In addition, miR-495-3p and miR-299 were identified by qRT-PCR and had CD44 positively co-regulated potential targets, such as VEGFA and CSF1, which are associated with tumor progression and a worse prognosis.165 Two oncogenic molecules, miR-15a and miR-24-1, were also identified in coexpression networks to regulate expression of CYP11B1, KRT33B, RUNX1T1, SIK1, MAP3K4, MLANA, and SFRP5 via a weighted gene coexpression network approach.166

lncRNAs in EPN

The lncRNA LINC00899 was observed to be upregulated in spinal EPN samples. Further in vitro experiments verified the anti-oncogenic effects of downregulated LINC00899, which inhibited spinal EPN cell invasion and migration via the RBL2-dependent FoxO pathway.38 The lncRNA TRERNA1, which regulates the expression of the EMT master transcription factor Snail, was significantly overexpressed in intracranial subgroups compared to normal brain. TRERNA1 upregulation was found to be associated with higher proliferative indices and shorter progression-free survival.39 Using a genome-wide methylome analysis approach, Wang et al.167 identified lncRNA signatures associated with tumor histological characteristics based on the methylation status of lncRNA promoters. The lncRNA LINC00052 exhibited the highest importance value in the classification of spinal EPNs. Another study found low expression of the lncRNA HOTAIR in EPN, which is also known as metastasis-associated lncRNA.28 Taken together, lncRNAs participate in EPN progression (Figure 2).

Figure 2.

Figure 2

Roles of lncRNAs in ependymoma

Diffuse intrinsic pontine glioma (DIPG)

DIPGs are a devastating spectrum of disease with no effective cures, although a myriad of treatments have been studied in hundreds of clinical trials.168, 169, 170 As a subtype of advanced grade gliomas that originates in the pons and spreads to other parts of the brainstem, almost all confirmed patients will die of this disease within 2 years from the time of their initial diagnosis.169,171 To date, no chemotherapeutic strategy has been shown to improve the prognosis of DIPGs, and the predominant method of treatment for children with newly diagnosed DIPG remains focal radiotherapy to the pons.172,173 Due to the tumor location, great advances in neurosurgical techniques have allowed for DIPG surgical biopsies to be conducted safely.169,174,175 A combination of genomic profiling and drug efficacy testing will result in a better understanding of DIPG biology and assist in drug development.

According to the current research, DIPG has specific driver mutations that could promote invasion. A specific point mutation in one of the histone 3 genes, including H3.3 (H3F3A) or H3.1 (HIST1H3B), appears in most DIPG cases (80%) and causes lysine 27 on the amino-terminal tail to be replaced with a methionine (H3K27M).176, 177, 178 In fact, mutations in HIST1H3B resulted in a better prognosis than mutations in H3F3A.179 In addition, several genomic alterations or amplifications are observed in Tp53, activin A receptor type I (ACVR1), and platelet-derived growth factor receptor alpha (PDGFRA) in DIPGs.179

miRNAs in DIPG

Related studies have proven that ncRNAs have an important impact on the pathogenesis of DIPGs. miR-129-2 was found to be downregulated by hypermethylation of miR-129-2 promoter in DIPGs, leading to overexpression of NG2, which was demonstrated to contribute to the neoplastic transformation of glioma cells.180 A bioinformatics analysis of microarray data demonstrated 27 altered miRNAs associated with DIPG and built a miRNA-target regulatory network consisting of 141 miRNA-target gene pairs. Moreover, miR-26b, which interacts with TFAP2 to a higher degree in the transcription factor (TF)-miRNA-target gene regulatory network, might have a critical role in the tumorigenesis of DIPG.181

lncRNAs in DIPG

Using expression profile analysis, Liu et al.182 also identified some novel differentially expressed lncRNAs in DIPG, including AF086127, AF086217, AF086391, AF119852, AK021535, AK022370, AL050068, BC012548, and BC041658. These lncRNAs are significantly correlated with DIPG survival and have great potential as diagnostic or prognostic biomarkers. Continuous exploration of specific biomarkers will provide new possibilities for humans to understand the underlying mechanisms of DIPG.

Craniopharyngioma (CP)

Childhood-onset CPs are rare nonglial tumors of the sellar region originating from remnants of the craniopharyngeal duct epithelium with low-grade histological malignancy.183,184 CPs are traditionally classified into two histological subtypes, adamantinomatous CP (ACP) and papillary CP (PCP), which exhibit distinct genetic features and age distributions.185, 186, 187 Despite favorable survival outcomes, the quality of life of these pediatric patients is frequently impaired due to the severe sequelae of this disease.188, 189, 190 Thus, novel insights into the molecular pathogenesis of CPs will help develop new treatments targeting pathogenic pathways, thereby decreasing or eliminating adverse effects.

miRNAs in CP

Based on miRNA expression analysis, overexpression of miR-150 and decreased expression of let-7a, miR-16, miR-15a, miR-23b, miR-24-2, miR-141, miR-143, miR-145, and miR-449 were observed in ACPs.191 Further in silico analysis indicated that miR-150, miR-23b, miR-24-2, miR-141, and miR-449 could regulate CTNNB1 expression through the Wnt signaling pathway, suggesting important roles in ACP tumorigenesis.191 Another extensive miRNA expression analysis demonstrated that downregulation of miR-132 appeared to be an indicator of aggressiveness and might contribute to EMT.192 The role of ncRNAs in CPs is understudied and research is still needed to explore their underlying biological and pathogenic mechanisms.

miRNAs and lncRNA in ATRTs

Most ATRTs are characterized by genomic alterations in SMARCB1 or, to a less extent, SMARCA4 of the SWItch/sucrose nonfermentable chromatin remodeling complex.193,194 To identify new possible therapeutic targets of ATRT, Sredni et al.195 demonstrated dysregulated expression of miR-221/222 in ATRT, and their upregulation contributed to oncogenesis and development of ATRT by p27Kip1 downregulation. Both miRNAs regulate the expression level of the target gene SUN2, which is a tumor suppressor and accelerates cell proliferation and tumor malignancy both in vitro and in vivo.196 Birks et al.71 also found that miR-129, miR-142-5p, and miR-25 were differentially expressed in five pediatric brain tumor types, including ATRT, compared to normal tissue controls. In the same study, the upregulated miRNAs in ATRTs, including miR-520b, miR-629, miR-221, miR-448, and miR-373, and the downregulated miRNAs, including miR-140, miR-let-7b, miR-139, miR-153, and miR-376, were also revealed. Deletion of miRNAs let-7a3 and let-7b was found to partially contribute to the overexpression of the oncoprotein HMGA2 in ATRT tissues. Upregulation of let-7 miRNA or knockdown of HMGA2 could also inhibit rhabdoid tumor cell proliferation, colony formation, and invasion.197 In addition, it was demonstrated that miR-142-3p was downregulated in stem-like ATRT cells (ATRT-CD133+), and that its lower expression promoted tumor growth and invasive, radioresistant, and stem-like capacities. Notably, therapeutic delivery of miR-142-3p in ATRT cells effectively reduced its lethality and prolonged survival time in orthotropic-transplanted immunocompromised mice.198 More recently, tumor-associated mesenchymal stem cells were observed to secrete miR-155-enriched exosomes, and the abundant expression of exosomal miR-155 could mediate ATRT tumor migration through downregulation of the tumor suppressor SMARCA4.199 Moreover, transcriptome analysis also indicated significantly higher expression of the lncRNA HOTAIR and its associated protein-coding gene HOXC in ATRT tissues, although the underlying mechanism needs further investigation.28

Glioblastoma (GBM)

GBM (WHO grade IV) is the most common and malignant primary brain tumor. It occurs more frequently in adults and only accounts for approximately 8%–12% of all CNS tumors in children.200,201 This neoplasm is characterized by rapid diffuse and infiltrative growth and a high level of cellular heterogeneity leading to therapeutic resistance.202 Thus, despite the multimodal treatment procedure composed of surgical intervention, radiotherapy, and temozolomide-based chemotherapy, the overall survival of these patients is still unsatisfactory with a median survival of 15 months,203,204 Thus, it is urgent to discover novel therapeutic strategies.205

miRNAs in GBM

During recent years, large-scale research efforts have been made to unveil the roles of many ncRNAs in adult GBM onset, progression, invasiveness, and recurrence. However, because of their rarity, research on ncRNAs in pediatric GBM is still scant.67,206 As one of the most extensively investigated miRNAs, oncogenic miR-21 is significantly overexpressed in GBM and inversely correlated with GBM survival. Many specific molecules, including HNPRK, TAP63, PDCD4, p53, and transforming growth factor (TGF)-β and the mitochondrial apoptotic pathway regulated by miR-21, have also been validated to play a critically important role in different aspects of tumor pathogenesis.205,207,208 Another extensive investigation showed that miR-221 and miR-222 are among the most frequent and significantly overexpressed miRNAs in GBMs. High levels of miR-221/222 promote cell proliferation, cell cycle progression, migration, and invasion and inhibit cell apoptosis by directly targeting p27, p57, astrocytic connexin Cx43, PTPμ, TIMP3, and p53 upregulated modulator of apoptosis (PUMA).209, 210, 211, 212, 213, 214

It is known that p27, p57, Cx43, and PUMA proteins are encoded by PSMD9, CDKN1C, GJA1, and Bcl-2-binding component 3 (BBC3) genes, respectively. With respect to specific pediatric GBM patients, miR-129, miR-142-5p, and miR-25 were found to display differential expression compared to normal tissue controls.71 Another genome-wide microarray analysis comparing pediatric GBM patients with controls demonstrated differential expression of 266 miRNAs, of which 55 were upregulated and 71 were downregulated. Upregulated miRNAs, including miR-10b, miR-891a, miR-182, miR-155, miR-424, and miR-130b and downregulated miRNAs, including miR-138, miR-7, and miR-129, were further validated by qRT-PCR. In regard to the expression patterns of clustered miRNAs, all miR-17/92 and miR-106b/25 cluster miRNAs were upregulated, while most 14q32 cluster miRNAs were downregulated and associated with patient survival. H3F3A mutation-associated miRNA expression profiles showed that miR-15a, miR-424, miR-30e, and miR-378c were more highly expressed in H3F3A mutants than in the wild-type. A list of TP53 mutation-specific miRNAs in pediatric GBM patients was also identified. Further comparisons of miRNA expression profiles of pediatric and adult GBMs were conducted, and specially expressed miRNAs related to pediatric GBM might be associated with PDGFR-β, regulation of nuclear SMAD2/3 signaling, calcineurin, ErBB1 signaling, and cdc42 signaling pathways.215

In addition, new molecular characteristics of pediatric and adult high-grade gliomas were revealed to support their biological differences. For example, the miR-17-92 cluster was found to be upregulated in pediatric high-grade gliomas, where it controlled cell proliferation and targeted tumor suppressors such as PTEN.216 Giunti et al.217 also examined the miRNA expression profile of pediatric GBM and demonstrated that miR-137, miR-490, miR-876-3p, miR-876-5p, and miR-448 were downregulated and miR-501-3p was upregulated. The association of each of the identified differentially expressed miRNAs with NUCKS1 deserves further investigation. Moreover, overexpression of miR-487b in a pediatric glioma cell line (KNS42), which was established from a 16-year-old child with GBM, downregulated PROM1 and Nestin. This resulted in the inhibition of colony formation, whereas cell growth, proliferation, sensitivity to temozolomide, migration, and invasion were not affected.218 Using a predictive analysis approach for pediatric and adult high-grade glioma, Liu et al.219 also screened 12 microarrays and identified miR-10a, miR-10b, and miR-139 as having common differences in glioma.

lncRNAs in GBM

The lncRNA MALAT1 has been shown to be overexpressed in GBM and is associated with worse outcomes for GBM patients. Mechanistically, MALAT1 serves as a “molecular sponge” and can modulate the activity of multiple miRNAs, including miR-106-5p, miR-144-3p, miR-211, miR-203, miR-155, and miR-199a. The modulation of these miRNAs has an important impact on the pathogenesis and development of tumors, including GBM.40, 41, 42, 43, 44 The lncRNA HOTAIR has been found to be highly expressed in several types of pediatric brain tumors.28 HOTAIR also functions as a sponge miRNA, and its depletion inhibits the malignant biological behaviors of GBM.30, 31, 32, 33, 34, 35, 36, 37 One study indicated dysregulation of HOX genes and HOTAIR in pediatric GBMs.28 Another study confirmed that HOXA9 directly binds with the HOTAIR promoter in adult and pediatric glioma-derived cell lines.33 The lncRNA DGCR5 was also found to suppress EMT in pediatric primary GBM cells and might serve as a prognostic biomarker.45

circRNAs in GBM

As a new research hotspot, circRNAs have been increasingly valued by researchers in GBM (Table 2). circ-FBXW7 encodes a novel 21-kDa protein, FBXW7-185aa, which regulates cell proliferation and the cell cycle. Knockdown of FBXW7-185aa promoted malignant phenotypes in vitro and in vivo.46 Both circ-SHPRH and its encoded protein SHPRH-146aa were found to be downregulated in GBM. The overexpression of SHPRH-146aa reduced malignant behavior and tumorigenicity in vitro and in vivo.47 circSMARCA5 was significantly downregulated in GBM and associated with tumor progression. Overexpression of circSMARCA5 inhibited the migration of GBM cells by regulating a molecular axis that involves the splicing factors SRSF1/SRSF3/PTB.48 Moreover, circSMARCA5 was found to be an upstream regulator of the proangiogenic-to-antiangiogenic VEGFA isoform ratio.49 The circRNA hsa_circ_0008344 has been studied in vitro, and it showed the ability to regulate GBM cell proliferation, colony formation, migration, invasion, and the cell apoptotic rate.50 Another oncogenic circRNA, circNT5E, has been found to act as a sponge against miR-422a in GBM tumorigenesis, controlling many pathologic processes, such as cell proliferation, migration, and invasion.51 circPINTexon2 has been observed to produce a peptide named PINT87aa, which suppresses GBM cell proliferation in vitro and in vivo.52 Wang et al.53 also indicated that eIF4A3-induced circMMP9 could promote GBM cell proliferation, invasion, and metastasis via the miR-124 signaling pathway. Elevated circ_0029426 was observed in GBM tissues and could strongly promote cell proliferation, migration, and invasion and inhibit cell apoptosis by sponging miR-197.54 Moreover, the circ_0001946/miR-671-5p/CDR1 pathway modulates the development of GBM.220 Upregulation of circ_0074027 also promoted GBM cell growth and invasion by regulating the miR-518a-5p/IL17RD signaling pathway.55 hsa_circ_0067934 was found to be upregulated in GBM and promoted cancer cell proliferation and metastasis via upregulation of the PI3K-AKT pathway.56

circ_0001730 has been found to promote GBM cell proliferation and invasion via the miR-326/Wnt7B axis.57 circMTO1 has been reported to inhibit GBM cell proliferation via the miR-92/WWOX signaling pathway.58 Impaired expression of circ-AKT3 contributed to GBM tumorigenesis.59 Moreover, circ-PITX1 could act as a ceRNA to promote the progression of GBM by regulating the miR-379-5p/MAP3K2 axis.60 Similarly, circFOXO3 was discovered to promote GBM progression by sponging both miR-138-5p and miR-432-5p to regulate the expression of NFAT5.61 Furthermore, overexpression of circ-0001801 contributed to GBM cell proliferation, migration, invasion, and EMT by modulating the miR-628-5p and HMGB3 axes.62 circ-EPB41L5 was also shown to play a striking role in the progression of GBM via regulation of the miR-19a/EPB41L5/p-AKT regulatory axis.63 Most recently, circENTPD7 was found to promote GBM cell proliferation and motility by regulating miR-101-3p/ROS1.64 However, the expression of these circRNAs needs further confirmation in GBM in the pediatric setting.

Conclusions and future perspectives

The advancement of bioinformatics technology has greatly facilitated the identification of a great number of abnormally expressed ncRNAs in different types of tumors. Currently, there is consensus that ncRNAs play important roles in gene regulatory networks and hold great potential as therapeutic targets. As discussed above, accumulating evidence has clearly supported the involvement of ncRNAs in pathogenesis as oncogenes or tumor suppressors in pediatric neuro-oncology. First, ncRNAs can better enable researchers to discover molecular markers that help with tumor classification and patient risk stratification combined with other biological characteristics, thereby assisting in standardizing the selection and treatment plans.221 The molecules that are involved in tumor occurrence and development, as well as the regulation of response to therapy, can be further employed to assess therapeutic effects and screen potential patients who can significantly benefit from other therapeutic opportunities, such as immunotherapy and gene therapy.

In addition, treatments targeting these aberrantly expressed ncRNAs are a promising approach to improve the outcomes of pediatric patients with CNS tumors. Despite the great potential, the nonspecificity of ncRNA targets has to be taken into account, and the delivery method for ncRNAs should be optimized to be effective and nontoxic.221,222 Future studies must address these issues to drive ncRNA-based therapeutic development. Without a doubt, further intensified exploration is needed to discover additional ncRNAs with crucial biological functions and deepen the understanding of these molecules as therapeutic targets in the management of pediatric CNS tumors. In summary, ncRNA studies continue to provide new insights into pediatric neuro-oncology biology. Although this field faces many challenges and significant efforts are still required, clinical applications of ncRNAs for pediatric CNS tumors will drastically change the medical practice in the foreseeable future.

Acknowledgments

We thank our colleagues for their critical comments.

Author contributions

S.C., X.D., H.S., Y.R., Y. Zheng, and Y. Zhang searched the literature. S.C. and X.D. made the figures and tables. S.C., X.D., and J.L. wrote the manuscript. All authors read and approved the final manuscript.

Declaration of interests

The authors declare no competing interests.

References

  • 1.Pollack I.F., Jakacki R.I. Childhood brain tumors: Epidemiology, current management and future directions. Nat. Rev. Neurol. 2011;7:495–506. doi: 10.1038/nrneurol.2011.110. [DOI] [PubMed] [Google Scholar]
  • 2.Kumar R., Liu A.P.Y., Orr B.A., Northcott P.A., Robinson G.W. Advances in the classification of pediatric brain tumors through DNA methylation profiling: From research tool to frontline diagnostic. Cancer. 2018;124:4168–4180. doi: 10.1002/cncr.31583. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Ostrom Q.T., Gittleman H., Liao P., Vecchione-Koval T., Wolinsky Y., Kruchko C., Barnholtz-Sloan J.S. CBTRUS statistical report: Primary brain and other central nervous system tumors diagnosed in the United States in 2010–2014. Neuro-oncol. 2017;19(Suppl_5):v1–v88. doi: 10.1093/neuonc/nox158. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Louis D.N., Perry A., Reifenberger G., von Deimling A., Figarella-Branger D., Cavenee W.K., Ohgaki H., Wiestler O.D., Kleihues P., Ellison D.W. The 2016 World Health Organization Classification of Tumors of the Central Nervous System: A summary. Acta Neuropathol. 2016;131:803–820. doi: 10.1007/s00401-016-1545-1. [DOI] [PubMed] [Google Scholar]
  • 5.Bahmad H.F., Elajami M.K., El Zarif T., Bou-Gharios J., Abou-Antoun T., Abou-Kheir W. Drug repurposing towards targeting cancer stem cells in pediatric brain tumors. Cancer Metastasis Rev. 2020;39:127–148. doi: 10.1007/s10555-019-09840-2. [DOI] [PubMed] [Google Scholar]
  • 6.Du Z., Fei T., Verhaak R.G., Su Z., Zhang Y., Brown M., Chen Y., Liu X.S. Integrative genomic analyses reveal clinically relevant long noncoding RNAs in human cancer. Nat. Struct. Mol. Biol. 2013;20:908–913. doi: 10.1038/nsmb.2591. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Iyer M.K., Niknafs Y.S., Malik R., Singhal U., Sahu A., Hosono Y., Barrette T.R., Prensner J.R., Evans J.R., Zhao S. The landscape of long noncoding RNAs in the human transcriptome. Nat. Genet. 2015;47:199–208. doi: 10.1038/ng.3192. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Carvalho de Oliveira J., Molinari Roberto G., Baroni M., Bezerra Salomão K., Alejandra Pezuk J., Sol Brassesco M. miRNA dysregulation in childhood hematological cancer. Int. J. Mol. Sci. 2018;19:2688. doi: 10.3390/ijms19092688. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Cech T.R., Steitz J.A. The noncoding RNA revolution-trashing old rules to forge new ones. Cell. 2014;157:77–94. doi: 10.1016/j.cell.2014.03.008. [DOI] [PubMed] [Google Scholar]
  • 10.Esteller M., Pandolfi P.P. The epitranscriptome of noncoding RNAs in cancer. Cancer Discov. 2017;7:359–368. doi: 10.1158/2159-8290.CD-16-1292. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Anastasiadou E., Jacob L.S., Slack F.J. Non-coding RNA networks in cancer. Nat. Rev. Cancer. 2018;18:5–18. doi: 10.1038/nrc.2017.99. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Bartel D.P. Metazoan microRNAs. Cell. 2018;173:20–51. doi: 10.1016/j.cell.2018.03.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Huarte M. The emerging role of lncRNAs in cancer. Nat. Med. 2015;21:1253–1261. doi: 10.1038/nm.3981. [DOI] [PubMed] [Google Scholar]
  • 14.Memczak S., Jens M., Elefsinioti A., Torti F., Krueger J., Rybak A., Maier L., Mackowiak S.D., Gregersen L.H., Munschauer M. Circular RNAs are a large class of animal RNAs with regulatory potency. Nature. 2013;495:333–338. doi: 10.1038/nature11928. [DOI] [PubMed] [Google Scholar]
  • 15.Hansen T.B., Jensen T.I., Clausen B.H., Bramsen J.B., Finsen B., Damgaard C.K., Kjems J. Natural RNA circles function as efficient microRNA sponges. Nature. 2013;495:384–388. doi: 10.1038/nature11993. [DOI] [PubMed] [Google Scholar]
  • 16.Wu P., Mo Y., Peng M., Tang T., Zhong Y., Deng X., Xiong F., Guo C., Wu X., Li Y. Emerging role of tumor-related functional peptides encoded by lncRNA and circRNA. Mol. Cancer. 2020;19:22. doi: 10.1186/s12943-020-1147-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Jiang W., Xia J., Xie S., Zou R., Pan S., Wang Z.W., Assaraf Y.G., Zhu X. Long non-coding RNAs as a determinant of cancer drug resistance: Towards the overcoming of chemoresistance via modulation of lncRNAs. Drug Resist. Updat. 2020;50:100683. doi: 10.1016/j.drup.2020.100683. [DOI] [PubMed] [Google Scholar]
  • 18.Yue Y., Zhang W., Liu C., Niu Y., Tong W. Long non-coding RNA Gm15577 is involved in mouse cerebellar neurogenesis. Zhonghua Bing Li Xue Za Zhi. 2015;44:504–508. [PubMed] [Google Scholar]
  • 19.Song H., Han L.M., Gao Q., Sun Y. Long non-coding RNA CRNDE promotes tumor growth in medulloblastoma. Eur. Rev. Med. Pharmacol. Sci. 2016;20:2588–2597. [PubMed] [Google Scholar]
  • 20.Sun X.H., Fan W.J., An Z.J., Sun Y. Inhibition of long noncoding RNA CRNDE increases chemosensitivity of medulloblastoma cells by targeting miR-29c-3p. Oncol. Res. 2020;28:95–102. doi: 10.3727/096504019X15742472027401. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Laneve P., Po A., Favia A., Legnini I., Alfano V., Rea J., Di Carlo V., Bevilacqua V., Miele E., Mastronuzzi A. The long noncoding RNA linc-NeD125 controls the expression of medulloblastoma driver genes by microRNA sponge activity. Oncotarget. 2017;8:31003–31015. doi: 10.18632/oncotarget.16049. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Zhang Y., Wang T., Wang S., Xiong Y., Zhang R., Zhang X., Zhao J., Yang A.G., Wang L., Jia L. Nkx2-2as suppression contributes to the pathogenesis of Sonic Hedgehog medulloblastoma. Cancer Res. 2018;78:962–973. doi: 10.1158/0008-5472.CAN-17-1631. [DOI] [PubMed] [Google Scholar]
  • 23.Zhengyuan X., Hu X., Qiang W., Nanxiang L., Junbin C., Wangming Z. Silencing of urothelial carcinoma associated 1 inhibits the proliferation and migration of medulloblastoma cells. Med. Sci. Monit. 2017;23:4454–4461. doi: 10.12659/MSM.904675. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Gao R., Zhang R., Zhang C., Zhao L., Zhang Y. Long noncoding RNA CCAT1 promotes cell proliferation and metastasis in human medulloblastoma via MAPK pathway. Tumori. 2018;104:43–50. doi: 10.5301/tj.5000662. [DOI] [PubMed] [Google Scholar]
  • 25.Gao R., Zhang R., Zhang C., Liang Y., Tang W. lncRNA LOXL1-AS1 promotes the proliferation and metastasis of medulloblastoma by activating the PI3K/AKT pathway. Anal. Cell. Pathol. (Amst.) 2018;2018:9275685. doi: 10.1155/2018/9275685. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Varon M., Levy T., Mazor G., Ben David H., Marciano R., Krelin Y., Prasad M., Elkabets M., Pauck D., Ahmadov U. The long noncoding RNA TP73-AS1 promotes tumorigenicity of medulloblastoma cells. Int. J. Cancer. 2019;145:3402–3413. doi: 10.1002/ijc.32400. [DOI] [PubMed] [Google Scholar]
  • 27.Li B., Shen M., Yao H., Chen X., Xiao Z. Long noncoding RNA TP73-AS1 modulates medulloblastoma progression in vitro and in vivo by sponging miR-494-3p and targeting EIF5A2. OncoTargets Ther. 2019;12:9873–9885. doi: 10.2147/OTT.S228305. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Chakravadhanula M., Ozols V.V., Hampton C.N., Zhou L., Catchpoole D., Bhardwaj R.D. Expression of the HOX genes and HOTAIR in atypical teratoid rhabdoid tumors and other pediatric brain tumors. Cancer Genet. 2014;207:425–428. doi: 10.1016/j.cancergen.2014.05.014. [DOI] [PubMed] [Google Scholar]
  • 29.Zhang J., Li N., Fu J., Zhou W. Long noncoding RNA HOTAIR promotes medulloblastoma growth, migration and invasion by sponging miR-1/miR-206 and targeting YY1. Biomed. Pharmacother. 2020;124:109887. doi: 10.1016/j.biopha.2020.109887. [DOI] [PubMed] [Google Scholar]
  • 30.Zhang J., Chen G., Gao Y., Liang H. HOTAIR/miR-125 axis-mediated Hexokinase 2 expression promotes chemoresistance in human glioblastoma. J. Cell. Mol. Med. 2020;24:5707–5717. doi: 10.1111/jcmm.15233. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Li H., Guan C. HOTAIR inhibits the proliferation of glioblastoma cells by targeting miR-219. Cancer Biomark. 2020;28:41–47. doi: 10.3233/CBM-190467. [DOI] [PubMed] [Google Scholar]
  • 32.Ren Y., Wang Y.F., Zhang J., Wang Q.X., Han L., Mei M., Kang C.S. Targeted design and identification of AC1NOD4Q to block activity of HOTAIR by abrogating the scaffold interaction with EZH2. Clin. Epigenetics. 2019;11:29. doi: 10.1186/s13148-019-0624-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Xavier-Magalhães A., Gonçalves C.S., Fogli A., Lourenço T., Pojo M., Pereira B., Rocha M., Lopes M.C., Crespo I., Rebelo O. The long non-coding RNA HOTAIR is transcriptionally activated by HOXA9 and is an independent prognostic marker in patients with malignant glioma. Oncotarget. 2018;9:15740–15756. doi: 10.18632/oncotarget.24597. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Huang K., Sun J., Yang C., Wang Y., Zhou B., Kang C., Han L., Wang Q. HOTAIR upregulates an 18-gene cell cycle-related mRNA network in glioma. Int. J. Oncol. 2017;50:1271–1278. doi: 10.3892/ijo.2017.3901. [DOI] [PubMed] [Google Scholar]
  • 35.Wang G., Li Z., Tian N., Han L., Fu Y., Guo Z., Tian Y. miR-148b-3p inhibits malignant biological behaviors of human glioma cells induced by high HOTAIR expression. Oncol. Lett. 2016;12:879–886. doi: 10.3892/ol.2016.4743. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Fang K., Liu P., Dong S., Guo Y., Cui X., Zhu X., Li X., Jiang L., Liu T., Wu Y. Magnetofection based on superparamagnetic iron oxide nanoparticle-mediated low lncRNA HOTAIR expression decreases the proliferation and invasion of glioma stem cells. Int. J. Oncol. 2016;49:509–518. doi: 10.3892/ijo.2016.3571. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Pastori C., Kapranov P., Penas C., Peschansky V., Volmar C.H., Sarkaria J.N., Bregy A., Komotar R., St Laurent G., Ayad N.G., Wahlestedt C. The Bromodomain protein BRD4 controls HOTAIR, a long noncoding RNA essential for glioblastoma proliferation. Proc. Natl. Acad. Sci. USA. 2015;112:8326–8331. doi: 10.1073/pnas.1424220112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Chen Q.B., Li Z.H., Fu Y., Lv N.N., Tian N., Han L., Tian Y. Downregulated long non-coding RNA LINC00899 inhibits invasion and migration of spinal ependymoma cells via RBL2-dependent FoxO pathway. Cell Cycle. 2019;18:2566–2579. doi: 10.1080/15384101.2019.1652046. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 39.Malgulwar P.B., Nambirajan A., Singh M., Suri V., Sarkar C., Sharma M.C. Expression and clinical significance of translation regulatory long non-coding RNA 1 (TRERNA1) in ependymomas. Pathol. Oncol. Res. 2020;26:975–1981. doi: 10.1007/s12253-019-00736-8. [DOI] [PubMed] [Google Scholar]
  • 40.Liao K., Lin Y., Gao W., Xiao Z., Medina R., Dmitriev P., Cui J., Zhuang Z., Zhao X., Qiu Y. Blocking lncRNA MALAT1/miR-199a/ZHX1 axis inhibits glioblastoma proliferation and progression. Mol. Ther. Nucleic Acids. 2019;18:388–399. doi: 10.1016/j.omtn.2019.09.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Zhuang M., Zhao S., Jiang Z., Wang S., Sun P., Quan J., Yan D., Wang X. MALAT1 sponges miR-106b-5p to promote the invasion and metastasis of colorectal cancer via SLAIN2 enhanced microtubules mobility. EBioMedicine. 2019;41:286–298. doi: 10.1016/j.ebiom.2018.12.049. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Cao S., Wang Y., Li J., Lv M., Niu H., Tian Y. Tumor-suppressive function of long noncoding RNA MALAT1 in glioma cells by suppressing miR-155 expression and activating FBXW7 function. Am. J. Cancer Res. 2016;6:2561–2574. [PMC free article] [PubMed] [Google Scholar]
  • 43.Wang Y., Zhang Y., Yang T., Zhao W., Wang N., Li P., Zeng X., Zhang W. Long non-coding RNA MALAT1 for promoting metastasis and proliferation by acting as a ceRNA of miR-144-3p in osteosarcoma cells. Oncotarget. 2017;8:59417–59434. doi: 10.18632/oncotarget.19727. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Tao F., Tian X., Ruan S., Shen M., Zhang Z. miR-211 sponges lncRNA MALAT1 to suppress tumor growth and progression through inhibiting PHF19 in ovarian carcinoma. FASEB J. 2018;32:6330–6343. doi: 10.1096/fj.201800495RR. [DOI] [PubMed] [Google Scholar]
  • 45.Yang F., Huang Y.L. DGCR5 suppresses the EMT of pediatric primary glioblastoma multiforme cell and serves as a prognostic biomarker. Eur. Rev. Med. Pharmacol. Sci. 2019;23:10024–10034. doi: 10.26355/eurrev_201911_19569. [DOI] [PubMed] [Google Scholar]
  • 46.Yang Y., Gao X., Zhang M., Yan S., Sun C., Xiao F., Huang N., Yang X., Zhao K., Zhou H. Novel role of FBXW7 circular RNA in repressing glioma tumorigenesis. J. Natl. Cancer Inst. 2018;110:304–315. doi: 10.1093/jnci/djx166. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Zhang M., Huang N., Yang X., Luo J., Yan S., Xiao F., Chen W., Gao X., Zhao K., Zhou H. A novel protein encoded by the circular form of the SHPRH gene suppresses glioma tumorigenesis. Oncogene. 2018;37:1805–1814. doi: 10.1038/s41388-017-0019-9. [DOI] [PubMed] [Google Scholar]
  • 48.Barbagallo D., Caponnetto A., Cirnigliaro M., Brex D., Barbagallo C., D’Angeli F., Morrone A., Caltabiano R., Barbagallo G.M., Ragusa M. circSMARCA5 inhibits migration of glioblastoma multiforme cells by regulating a molecular axis involving splicing factors SRSF1/SRSF3/PTB. Int. J. Mol. Sci. 2018;19:480. doi: 10.3390/ijms19020480. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Barbagallo D., Caponnetto A., Brex D., Mirabella F., Barbagallo C., Lauretta G., Morrone A., Certo F., Broggi G., Caltabiano R. circSMARCA5 regulates VEGFA mRNA splicing and angiogenesis in glioblastoma multiforme through the binding of SRSF1. Cancers (Basel) 2019;11:194. doi: 10.3390/cancers11020194. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Zhou J., Wang H., Chu J., Huang Q., Li G., Yan Y., Xu T., Chen J., Wang Y. Circular RNA hsa_circ_0008344 regulates glioblastoma cell proliferation, migration, invasion, and apoptosis. J. Clin. Lab. Anal. 2018;32:e22454. doi: 10.1002/jcla.22454. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Wang R., Zhang S., Chen X., Li N., Li J., Jia R., Pan Y., Liang H. circNT5E acts as a sponge of miR-422a to promote glioblastoma tumorigenesis. Cancer Res. 2018;78:4812–4825. doi: 10.1158/0008-5472.CAN-18-0532. [DOI] [PubMed] [Google Scholar]
  • 52.Zhang M., Zhao K., Xu X., Yang Y., Yan S., Wei P., Liu H., Xu J., Xiao F., Zhou H. A peptide encoded by circular form of LINC-PINT suppresses oncogenic transcriptional elongation in glioblastoma. Nat. Commun. 2018;9:4475. doi: 10.1038/s41467-018-06862-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Wang R., Zhang S., Chen X., Li N., Li J., Jia R., Pan Y., Liang H. EIF4A3-induced circular RNA MMP9 (circMMP9) acts as a sponge of miR-124 and promotes glioblastoma multiforme cell tumorigenesis. Mol. Cancer. 2018;17:166. doi: 10.1186/s12943-018-0911-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Zhang G., Sun W., Zhu L., Feng Y., Wu L., Li T. Overexpressed circ_0029426 in glioblastoma forecasts unfavorable prognosis and promotes cell progression by sponging miR-197. J. Cell. Biochem. 2019;120:10295–10302. doi: 10.1002/jcb.28313. [DOI] [PubMed] [Google Scholar]
  • 55.Qian L., Guan J., Wu Y., Wang Q. Upregulated circular RNA circ_0074027 promotes glioblastoma cell growth and invasion by regulating miR-518a-5p/IL17RD signaling pathway. Biochem. Biophys. Res. Commun. 2019;510:515–519. doi: 10.1016/j.bbrc.2019.01.140. [DOI] [PubMed] [Google Scholar]
  • 56.Xin J., Zhang X.Y., Sun D.K., Tian L.Q., Xu P. Up-regulated circular RNA hsa_circ_0067934 contributes to glioblastoma progression through activating PI3K-AKT pathway. Eur. Rev. Med. Pharmacol. Sci. 2019;23:3447–3454. doi: 10.26355/eurrev_201904_17709. [DOI] [PubMed] [Google Scholar]
  • 57.Lu Y., Deng X., Xiao G., Zheng X., Ma L., Huang W. circ_0001730 promotes proliferation and invasion via the miR-326/Wnt7B axis in glioma cells. Epigenomics. 2019;11:1335–1352. doi: 10.2217/epi-2019-0121. [DOI] [PubMed] [Google Scholar]
  • 58.Zhang X., Zhong B., Zhang W., Wu J., Wang Y. Circular RNA circMTO1 inhibits proliferation of glioblastoma cells via miR-92/WWOX signaling pathway. Med. Sci. Monit. 2019;25:6454–6461. doi: 10.12659/MSM.918676. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Xia X., Li X., Li F., Wu X., Zhang M., Zhou H., Huang N., Yang X., Xiao F., Liu D. A novel tumor suppressor protein encoded by circular AKT3 RNA inhibits glioblastoma tumorigenicity by competing with active phosphoinositide-dependent kinase-1. Mol. Cancer. 2019;18:131. doi: 10.1186/s12943-019-1056-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Lv X., Wang M., Qiang J., Guo S. Circular RNA circ-PITX1 promotes the progression of glioblastoma by acting as a competing endogenous RNA to regulate miR-379-5p/MAP3K2 axis. Eur. J. Pharmacol. 2019;863:172643. doi: 10.1016/j.ejphar.2019.172643. [DOI] [PubMed] [Google Scholar]
  • 61.Zhang S., Liao K., Miao Z., Wang Q., Miao Y., Guo Z., Qiu Y., Chen B., Ren L., Wei Z. circFOXO3 promotes glioblastoma progression by acting as a competing endogenous RNA for NFAT5. Neuro-oncol. 2019;21:1284–1296. doi: 10.1093/neuonc/noz128. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Chen W.L., Jiang L., Wang J.S., Liao C.X. circ-0001801 contributes to cell proliferation, migration, invasion and epithelial to mesenchymal transition (EMT) in glioblastoma by regulating miR-628-5p/HMGB3 axis. Eur. Rev. Med. Pharmacol. Sci. 2019;23:10874–10885. doi: 10.26355/eurrev_201912_19791. [DOI] [PubMed] [Google Scholar]
  • 63.Lv T., Miao Y., Xu T., Sun W., Sang Y., Jia F., Zhang X. circ-EPB41L5 regulates the host gene EPB41L5 via sponging miR-19a to repress glioblastoma tumorigenesis. Aging (Albany NY) 2020;12:318–339. doi: 10.18632/aging.102617. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Zhu F., Cheng C., Qin H., Wang H., Yu H. A novel circular RNA circENTPD7 contributes to glioblastoma progression by targeting ROS1. Cancer Cell Int. 2020;20:118. doi: 10.1186/s12935-020-01208-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Collins V.P., Jones D.T., Giannini C. Pilocytic astrocytoma: Pathology, molecular mechanisms and markers. Acta Neuropathol. 2015;129:775–788. doi: 10.1007/s00401-015-1410-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Ostrom Q.T., de Blank P.M., Kruchko C., Petersen C.M., Liao P., Finlay J.L., Stearns D.S., Wolff J.E., Wolinsky Y., Letterio J.J., Barnholtz-Sloan J.S. Alex’s Lemonade Stand Foundation Infant and Childhood Primary Brain and Central Nervous System Tumors Diagnosed in the United States in 2007–2011. Neuro-oncol. 2015;16(Suppl 10):x1–x36. doi: 10.1093/neuonc/nou327. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Pezuk J.A., Salomão K.B., Baroni M., Pereira C.A., Geron L., Brassesco M.S. Aberrantly expressed microRNAs and their implications in childhood central nervous system tumors. Cancer Metastasis Rev. 2019;38:813–828. doi: 10.1007/s10555-019-09820-6. [DOI] [PubMed] [Google Scholar]
  • 68.Jones D.T., Hutter B., Jäger N., Korshunov A., Kool M., Warnatz H.J., Zichner T., Lambert S.R., Ryzhova M., Quang D.A., International Cancer Genome Consortium PedBrain Tumor Project Recurrent somatic alterations of FGFR1 and NTRK2 in pilocytic astrocytoma. Nat. Genet. 2013;45:927–932. doi: 10.1038/ng.2682. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Pfister S., Janzarik W.G., Remke M., Ernst A., Werft W., Becker N., Toedt G., Wittmann A., Kratz C., Olbrich H. BRAF gene duplication constitutes a mechanism of MAPK pathway activation in low-grade astrocytomas. J. Clin. Invest. 2008;118:1739–1749. doi: 10.1172/JCI33656. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Jones D.T., Kocialkowski S., Liu L., Pearson D.M., Bäcklund L.M., Ichimura K., Collins V.P. Tandem duplication producing a novel oncogenic BRAF fusion gene defines the majority of pilocytic astrocytomas. Cancer Res. 2008;68:8673–8677. doi: 10.1158/0008-5472.CAN-08-2097. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Birks D.K., Barton V.N., Donson A.M., Handler M.H., Vibhakar R., Foreman N.K. Survey of microRNA expression in pediatric brain tumors. Pediatr. Blood Cancer. 2011;56:211–216. doi: 10.1002/pbc.22723. [DOI] [PubMed] [Google Scholar]
  • 72.Ho C.Y., Bar E., Giannini C., Marchionni L., Karajannis M.A., Zagzag D., Gutmann D.H., Eberhart C.G., Rodriguez F.J. MicroRNA profiling in pediatric pilocytic astrocytoma reveals biologically relevant targets, including PBX3, NFIB, and METAP2. Neuro-oncol. 2013;15:69–82. doi: 10.1093/neuonc/nos269. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Braoudaki M., Lambrou G.I., Giannikou K., Papadodima S.A., Lykoudi A., Stefanaki K., Sfakianos G., Kolialexi A., Tzortzatou-Stathopoulou F., Tzetis M. miR-15a and miR-24-1 as putative prognostic microRNA signatures for pediatric pilocytic astrocytomas and ependymomas. Tumour Biol. 2016;37:9887–9897. doi: 10.1007/s13277-016-4903-7. [DOI] [PubMed] [Google Scholar]
  • 74.Jones T.A., Jeyapalan J.N., Forshew T., Tatevossian R.G., Lawson A.R., Patel S.N., Doctor G.T., Mumin M.A., Picker S.R., Phipps K.P. Molecular analysis of pediatric brain tumors identifies microRNAs in pilocytic astrocytomas that target the MAPK and NF-κB pathways. Acta Neuropathol. Commun. 2015;3:86. doi: 10.1186/s40478-015-0266-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Darrigo Júnior L.G., Lira R.C.P., Fedatto P.F., Marco Antonio D.S., Valera E.T., Aguiar S., Yunes J.A., Brandalise S.R., Neder L., Saggioro F.P. MicroRNA profile of pediatric pilocytic astrocytomas identifies two tumor-specific signatures when compared to non-neoplastic white matter. J. Neurooncol. 2019;141:373–382. doi: 10.1007/s11060-018-03042-6. [DOI] [PubMed] [Google Scholar]
  • 76.Yuan M., Da Silva A.C.A.L., Arnold A., Okeke L., Ames H., Correa-Cerro L.S., Vizcaino M.A., Ho C.Y., Eberhart C.G., Rodriguez F.J. MicroRNA (miR) 125b regulates cell growth and invasion in pediatric low grade glioma. Sci. Rep. 2018;8:12506. doi: 10.1038/s41598-018-30942-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Liu K.W., Pajtler K.W., Worst B.C., Pfister S.M., Wechsler-Reya R.J. Molecular mechanisms and therapeutic targets in pediatric brain tumors. Sci. Signal. 2017;10:eaaf7593. doi: 10.1126/scisignal.aaf7593. [DOI] [PubMed] [Google Scholar]
  • 78.Northcott P.A., Robinson G.W., Kratz C.P., Mabbott D.J., Pomeroy S.L., Clifford S.C., Rutkowski S., Ellison D.W., Malkin D., Taylor M.D. Medulloblastoma. Nat. Rev. Dis. Primers. 2019;5:11. doi: 10.1038/s41572-019-0063-6. [DOI] [PubMed] [Google Scholar]
  • 79.Taylor M.D., Northcott P.A., Korshunov A., Remke M., Cho Y.J., Clifford S.C., Eberhart C.G., Parsons D.W., Rutkowski S., Gajjar A. Molecular subgroups of medulloblastoma: The current consensus. Acta Neuropathol. 2012;123:465–472. doi: 10.1007/s00401-011-0922-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Kool M., Korshunov A., Remke M., Jones D.T., Schlanstein M., Northcott P.A., Cho Y.J., Koster J., Schouten-van Meeteren A., van Vuurden D. Molecular subgroups of medulloblastoma: An international meta-analysis of transcriptome, genetic aberrations, and clinical data of WNT, SHH, group 3, and group 4 medulloblastomas. Acta Neuropathol. 2012;123:473–484. doi: 10.1007/s00401-012-0958-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Gajjar A., Chintagumpala M., Ashley D., Kellie S., Kun L.E., Merchant T.E., Woo S., Wheeler G., Ahern V., Krasin M.J. Risk-adapted craniospinal radiotherapy followed by high-dose chemotherapy and stem-cell rescue in children with newly diagnosed medulloblastoma (St Jude Medulloblastoma-96): Long-term results from a prospective, multicentre trial. Lancet Oncol. 2006;7:813–820. doi: 10.1016/S1470-2045(06)70867-1. [DOI] [PubMed] [Google Scholar]
  • 82.Northcott P.A., Korshunov A., Witt H., Hielscher T., Eberhart C.G., Mack S., Bouffet E., Clifford S.C., Hawkins C.E., French P. Medulloblastoma comprises four distinct molecular variants. J. Clin. Oncol. 2011;29:1408–1414. doi: 10.1200/JCO.2009.27.4324. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Cavalli F.M.G., Remke M., Rampasek L., Peacock J., Shih D.J.H., Luu B., Garzia L., Torchia J., Nor C., Morrissy A.S. Intertumoral heterogeneity within medulloblastoma subgroups. Cancer Cell. 2017;31:737–754.e6. doi: 10.1016/j.ccell.2017.05.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Northcott P.A., Buchhalter I., Morrissy A.S., Hovestadt V., Weischenfeldt J., Ehrenberger T., Gröbner S., Segura-Wang M., Zichner T., Rudneva V.A. The whole-genome landscape of medulloblastoma subtypes. Nature. 2017;547:311–317. doi: 10.1038/nature22973. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Cho Y.J., Tsherniak A., Tamayo P., Santagata S., Ligon A., Greulich H., Berhoukim R., Amani V., Goumnerova L., Eberhart C.G. Integrative genomic analysis of medulloblastoma identifies a molecular subgroup that drives poor clinical outcome. J. Clin. Oncol. 2011;29:1424–1430. doi: 10.1200/JCO.2010.28.5148. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Schwalbe E.C., Lindsey J.C., Nakjang S., Crosier S., Smith A.J., Hicks D., Rafiee G., Hill R.M., Iliasova A., Stone T. Novel molecular subgroups for clinical classification and outcome prediction in childhood medulloblastoma: A cohort study. Lancet Oncol. 2017;18:958–971. doi: 10.1016/S1470-2045(17)30243-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Pierson J., Hostager B., Fan R., Vibhakar R. Regulation of cyclin dependent kinase 6 by microRNA 124 in medulloblastoma. J. Neurooncol. 2008;90:1–7. doi: 10.1007/s11060-008-9624-3. [DOI] [PubMed] [Google Scholar]
  • 88.Silber J., Hashizume R., Felix T., Hariono S., Yu M., Berger M.S., Huse J.T., VandenBerg S.R., James C.D., Hodgson J.G., Gupta N. Expression of miR-124 inhibits growth of medulloblastoma cells. Neuro-oncol. 2013;15:83–90. doi: 10.1093/neuonc/nos281. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Li K.K., Pang J.C., Ching A.K., Wong C.K., Kong X., Wang Y., Zhou L., Chen Z., Ng H.K. miR-124 is frequently down-regulated in medulloblastoma and is a negative regulator of SLC16A1. Hum. Pathol. 2009;40:1234–1243. doi: 10.1016/j.humpath.2009.02.003. [DOI] [PubMed] [Google Scholar]
  • 90.Tenga A., Beard J.A., Takwi A., Wang Y.M., Chen T. Regulation of nuclear receptor Nur77 by miR-124. PLoS ONE. 2016;11:e0148433. doi: 10.1371/journal.pone.0148433. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Andolfo I., Liguori L., De Antonellis P., Cusanelli E., Marinaro F., Pistollato F., Garzia L., De Vita G., Petrosino G., Accordi B. The micro-RNA 199b-5p regulatory circuit involves Hes1, CD15, and epigenetic modifications in medulloblastoma. Neuro-oncol. 2012;14:596–612. doi: 10.1093/neuonc/nos002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Garzia L., Andolfo I., Cusanelli E., Marino N., Petrosino G., De Martino D., Esposito V., Galeone A., Navas L., Esposito S. MicroRNA-199b-5p impairs cancer stem cells through negative regulation of HES1 in medulloblastoma. PLoS ONE. 2009;4:e4998. doi: 10.1371/journal.pone.0004998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Guessous F., Zhang Y., Kofman A., Catania A., Li Y., Schiff D., Purow B., Abounader R. MicroRNA-34a is tumor suppressive in brain tumors and glioma stem cells. Cell Cycle. 2010;9:1031–1036. doi: 10.4161/cc.9.6.10987. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Li Y., Guessous F., Zhang Y., Dipierro C., Kefas B., Johnson E., Marcinkiewicz L., Jiang J., Yang Y., Schmittgen T.D. MicroRNA-34a inhibits glioblastoma growth by targeting multiple oncogenes. Cancer Res. 2009;69:7569–7576. doi: 10.1158/0008-5472.CAN-09-0529. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Thor T., Künkele A., Pajtler K.W., Wefers A.K., Stephan H., Mestdagh P., Heukamp L., Hartmann W., Vandesompele J., Sadowski N. miR-34a deficiency accelerates medulloblastoma formation in vivo. Int. J. Cancer. 2015;136:2293–2303. doi: 10.1002/ijc.29294. [DOI] [PubMed] [Google Scholar]
  • 96.Fan Y.N., Meley D., Pizer B., Sée V. mir-34a mimics are potential therapeutic agents for p53-mutated and chemo-resistant brain tumour cells. PLoS ONE. 2014;9:e108514. doi: 10.1371/journal.pone.0108514. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Weeraratne S.D., Amani V., Neiss A., Teider N., Scott D.K., Pomeroy S.L., Cho Y.J. miR-34a confers chemosensitivity through modulation of MAGE-A and p53 in medulloblastoma. Neuro-oncol. 2011;13:165–175. doi: 10.1093/neuonc/noq179. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Pannuru P., Dontula R., Khan A.A., Herbert E., Ozer H., Chetty C., Lakka S.S. miR-let-7f-1 regulates SPARC mediated cisplatin resistance in medulloblastoma cells. Cell. Signal. 2014;26:2193–2201. doi: 10.1016/j.cellsig.2014.06.014. [DOI] [PubMed] [Google Scholar]
  • 99.Abdelfattah N., Rajamanickam S., Panneerdoss S., Timilsina S., Yadav P., Onyeagucha B.C., Garcia M., Vadlamudi R., Chen Y., Brenner A. miR-584-5p potentiates vincristine and radiation response by inducing spindle defects and DNA damage in medulloblastoma. Nat. Commun. 2018;9:4541. doi: 10.1038/s41467-018-06808-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Ma H., Cao W., Ding M. MicroRNA-31 weakens cisplatin resistance of medulloblastoma cells via NF-κB and PI3K/AKT pathways. Biofactors. 2020;46:831–838. doi: 10.1002/biof.1616. [DOI] [PubMed] [Google Scholar]
  • 101.Ferretti E., De Smaele E., Po A., Di Marcotullio L., Tosi E., Espinola M.S., Di Rocco C., Riccardi R., Giangaspero F., Farcomeni A. MicroRNA profiling in human medulloblastoma. Int. J. Cancer. 2009;124:568–577. doi: 10.1002/ijc.23948. [DOI] [PubMed] [Google Scholar]
  • 102.Fiaschetti G., Abela L., Nonoguchi N., Dubuc A.M., Remke M., Boro A., Grunder E., Siler U., Ohgaki H., Taylor M.D. Epigenetic silencing of miRNA-9 is associated with HES1 oncogenic activity and poor prognosis of medulloblastoma. Br. J. Cancer. 2014;110:636–647. doi: 10.1038/bjc.2013.764. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Liu W., Gong Y.H., Chao T.F., Peng X.Z., Yuan J.G., Ma Z.Y., Jia G., Zhao J.Z. Identification of differentially expressed microRNAs by microarray: A possible role for microRNAs gene in medulloblastomas. Chin. Med. J. (Engl.) 2009;122:2405–2411. [PubMed] [Google Scholar]
  • 104.Dai J., Li Q., Bing Z., Zhang Y., Niu L., Yin H., Yuan G., Pan Y. Comprehensive analysis of a microRNA expression profile in pediatric medulloblastoma. Mol. Med. Rep. 2017;15:4109–4115. doi: 10.3892/mmr.2017.6490. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Kumar V., Kumar V., Chaudhary A.K., Coulter D.W., McGuire T., Mahato R.I. Impact of miRNA-mRNA profiling and their correlation on medulloblastoma tumorigenesis. Mol. Ther. Nucleic Acids. 2018;12:490–503. doi: 10.1016/j.omtn.2018.06.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Venkataraman S., Alimova I., Fan R., Harris P., Foreman N., Vibhakar R. MicroRNA 128a increases intracellular ROS level by targeting Bmi-1 and inhibits medulloblastoma cancer cell growth by promoting senescence. PLoS ONE. 2010;5:e10748. doi: 10.1371/journal.pone.0010748. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Wang X.M., Zhang S.F., Cheng Z.Q., Peng Q.Z., Hu J.T., Gao L.K., Xu J., Jin H.T., Liu H.Y. MicroRNA383 regulates expression of PRDX3 in human medulloblastomas. Zhonghua Bing Li Xue Za Zhi. 2012;41:547–552. doi: 10.3760/cma.j.issn.0529-5807.2012.08.009. [DOI] [PubMed] [Google Scholar]
  • 108.Jin Y., Xiong A., Zhang Z., Li S., Huang H., Yu T.T., Cao X., Cheng S.Y. MicroRNA-31 suppresses medulloblastoma cell growth by inhibiting DNA replication through minichromosome maintenance 2. Oncotarget. 2014;5:4821–4833. doi: 10.18632/oncotarget.2043. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Hemmesi K., Squadrito M.L., Mestdagh P., Conti V., Cominelli M., Piras I.S., Sergi L.S., Piccinin S., Maestro R., Poliani P.L. miR-135a inhibits cancer stem cell-driven medulloblastoma development by directly repressing Arhgef6 expression. Stem Cells. 2015;33:1377–1389. doi: 10.1002/stem.1958. [DOI] [PubMed] [Google Scholar]
  • 110.Kaid C., Silva P.B., Cortez B.A., Rodini C.O., Semedo-Kuriki P., Okamoto O.K. miR-367 promotes proliferation and stem-like traits in medulloblastoma cells. Cancer Sci. 2015;106:1188–1195. doi: 10.1111/cas.12733. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Pal R., Greene S. microRNA-10b is overexpressed and critical for cell survival and proliferation in medulloblastoma. PLoS ONE. 2015;10:e0137845. doi: 10.1371/journal.pone.0137845. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Singh S.V., Dakhole A.N., Deogharkar A., Kazi S., Kshirsagar R., Goel A., Moiyadi A., Jalali R., Sridhar E., Gupta T. Restoration of miR-30a expression inhibits growth, tumorigenicity of medulloblastoma cells accompanied by autophagy inhibition. Biochem. Biophys. Res. Commun. 2017;491:946–952. doi: 10.1016/j.bbrc.2017.07.140. [DOI] [PubMed] [Google Scholar]
  • 113.Senfter D., Samadaei M., Mader R.M., Gojo J., Peyrl A., Krupitza G., Kool M., Sill M., Haberler C., Ricken G. High impact of miRNA-4521 on FOXM1 expression in medulloblastoma. Cell Death Dis. 2019;10:696. doi: 10.1038/s41419-019-1926-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Yang Y., Cui H., Wang X. Downregulation of EIF5A2 by miR-221-3p inhibits cell proliferation, promotes cell cycle arrest and apoptosis in medulloblastoma cells. Biosci. Biotechnol. Biochem. 2019;83:400–408. doi: 10.1080/09168451.2018.1553604. [DOI] [PubMed] [Google Scholar]
  • 115.Zhang Z.Y., Zhu B., Zhao X.W., Zhan Y.B., Bao J.J., Zhou J.Q., Zhang F.J., Yu B., Liu J., Wang Y.M. Regulation of UHRF1 by microRNA-378 modulates medulloblastoma cell proliferation and apoptosis. Oncol. Rep. 2017;38:3078–3084. doi: 10.3892/or.2017.5939. [DOI] [PubMed] [Google Scholar]
  • 116.Lv S.Q., Kim Y.H., Giulio F., Shalaby T., Nobusawa S., Yang H., Zhou Z., Grotzer M., Ohgaki H. Genetic alterations in microRNAs in medulloblastomas. Brain Pathol. 2012;22:230–239. doi: 10.1111/j.1750-3639.2011.00523.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Grunder E., D’Ambrosio R., Fiaschetti G., Abela L., Arcaro A., Zuzak T., Ohgaki H., Lv S.Q., Shalaby T., Grotzer M. MicroRNA-21 suppression impedes medulloblastoma cell migration. Eur. J. Cancer. 2011;47:2479–2490. doi: 10.1016/j.ejca.2011.06.041. [DOI] [PubMed] [Google Scholar]
  • 118.Ray S., Coulter D.W., Gray S.D., Sughroue J.A., Roychoudhury S., McIntyre E.M., Chaturvedi N.K., Bhakat K.K., Joshi S.S., McGuire T.R., Sharp J.G. Suppression of STAT3 NH2-terminal domain chemosensitizes medulloblastoma cells by activation of protein inhibitor of activated STAT3 via de-repression by microRNA-21. Mol. Carcinog. 2018;57:536–548. doi: 10.1002/mc.22778. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Bai A.H., Milde T., Remke M., Rolli C.G., Hielscher T., Cho Y.J., Kool M., Northcott P.A., Jugold M., Bazhin A.V. MicroRNA-182 promotes leptomeningeal spread of non-Sonic Hedgehog-medulloblastoma. Acta Neuropathol. 2012;123:529–538. doi: 10.1007/s00401-011-0924-x. [DOI] [PubMed] [Google Scholar]
  • 120.Yang S.Y., Choi S.A., Lee J.Y., Park A.K., Wang K.C., Phi J.H., Koh E.J., Park W.Y., Park S.H., Hwang D.W. miR-192 suppresses leptomeningeal dissemination of medulloblastoma by modulating cell proliferation and anchoring through the regulation of DHFR, integrins, and CD47. Oncotarget. 2015;6:43712–43730. doi: 10.18632/oncotarget.6227. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Wang F., Remke M., Bhat K., Wong E.T., Zhou S., Ramaswamy V., Dubuc A., Fonkem E., Salem S., Zhang H. A microRNA-1280/JAG2 network comprises a novel biological target in high-risk medulloblastoma. Oncotarget. 2015;6:2709–2724. doi: 10.18632/oncotarget.2779. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Gao Y., Li P., Liu Z., Diao X., Song C. Expression levels of vascular endothelial cell growth factor and microRNA-210 are increased in medulloblastoma and metastatic medulloblastoma. Exp. Ther. Med. 2015;10:2138–2144. doi: 10.3892/etm.2015.2810. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Pan X., Wang Z., Wan B., Zheng Z. MicroRNA-206 inhibits the viability and migration of medulloblastoma cells by targeting LIM and SH3 protein 1. Exp. Ther. Med. 2017;14:3894–3900. doi: 10.3892/etm.2017.5016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Panwalkar P., Moiyadi A., Goel A., Shetty P., Goel N., Sridhar E., Shirsat N. miR-206, a cerebellum enriched miRNA is downregulated in all medulloblastoma subgroups and its overexpression is necessary for growth inhibition of medulloblastoma cells. J. Mol. Neurosci. 2015;56:673–680. doi: 10.1007/s12031-015-0548-z. [DOI] [PubMed] [Google Scholar]
  • 125.Shi J., Yang L., Wang T., Zhang J., Guo X., Huo X., Niu H. miR-218 is downregulated and directly targets SH3GL1 in childhood medulloblastoma. Mol. Med. Rep. 2013;8:1111–1117. doi: 10.3892/mmr.2013.1639. [DOI] [PubMed] [Google Scholar]
  • 126.Venkataraman S., Birks D.K., Balakrishnan I., Alimova I., Harris P.S., Patel P.R., Handler M.H., Dubuc A., Taylor M.D., Foreman N.K., Vibhakar R. MicroRNA 218 acts as a tumor suppressor by targeting multiple cancer phenotype-associated genes in medulloblastoma. J. Biol. Chem. 2013;288:1918–1928. doi: 10.1074/jbc.M112.396762. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Shi J.A., Lu D.L., Huang X., Tan W. miR-219 inhibits the proliferation, migration and invasion of medulloblastoma cells by targeting CD164. Int. J. Mol. Med. 2014;34:237–243. doi: 10.3892/ijmm.2014.1749. [DOI] [PubMed] [Google Scholar]
  • 128.Lucon D.R., Rocha Cde.S., Craveiro R.B., Dilloo D., Cardinalli I.A., Cavalcanti D.P., Aguiar Sdos.S., Maurer-Morelli C., Yunes J.A. Downregulation of 14q32 microRNAs in primary human desmoplastic medulloblastoma. Front. Oncol. 2013;3:254. doi: 10.3389/fonc.2013.00254. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Li K.K., Xia T., Ma F.M., Zhang R., Mao Y., Wang Y., Zhou L., Lau K.M., Ng H.K. miR-106b is overexpressed in medulloblastomas and interacts directly with PTEN. Neuropathol. Appl. Neurobiol. 2015;41:145–164. doi: 10.1111/nan.12169. [DOI] [PubMed] [Google Scholar]
  • 130.Gokhale A., Kunder R., Goel A., Sarin R., Moiyadi A., Shenoy A., Mamidipally C., Noronha S., Kannan S., Shirsat N.V. Distinctive microRNA signature of medulloblastomas associated with the WNT signaling pathway. J. Cancer Res. Ther. 2010;6:521–529. doi: 10.4103/0973-1482.77072. [DOI] [PubMed] [Google Scholar]
  • 131.Kunder R., Jalali R., Sridhar E., Moiyadi A., Goel N., Goel A., Gupta T., Krishnatry R., Kannan S., Kurkure P. Real-time PCR assay based on the differential expression of microRNAs and protein-coding genes for molecular classification of formalin-fixed paraffin embedded medulloblastomas. Neuro-oncol. 2013;15:1644–1651. doi: 10.1093/neuonc/not123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Yogi K., Sridhar E., Goel N., Jalali R., Goel A., Moiyadi A., Thorat R., Panwalkar P., Khire A., Dasgupta A. miR-148a, a microRNA upregulated in the WNT subgroup tumors, inhibits invasion and tumorigenic potential of medulloblastoma cells by targeting Neuropilin 1. Oncoscience. 2015;2:334–348. doi: 10.18632/oncoscience.137. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Li Y., Jiang T., Shao L., Liu Y., Zheng C., Zhong Y., Zhang J., Chang Q. mir-449a, a potential diagnostic biomarker for WNT group of medulloblastoma. J. Neurooncol. 2016;129:423–431. doi: 10.1007/s11060-016-2213-y. [DOI] [PubMed] [Google Scholar]
  • 134.Li Y.X., Shao L.W., Jiang T., Liu Y., Chang Q. miR-449a is a potential epigenetic biomarker for WNT subtype of medulloblastoma. Zhonghua Bing Li Xue Za Zhi. 2017;46:684–689. doi: 10.3760/cma.j.issn.0529-5807.2017.10.005. [DOI] [PubMed] [Google Scholar]
  • 135.Ferretti E., De Smaele E., Miele E., Laneve P., Po A., Pelloni M., Paganelli A., Di Marcotullio L., Caffarelli E., Screpanti I. Concerted microRNA control of hedgehog signalling in cerebellar neuronal progenitor and tumour cells. EMBO J. 2008;27:2616–2627. doi: 10.1038/emboj.2008.172. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Ferretti E., De Smaele E., Di Marcotullio L., Screpanti I., Gulino A. Hedgehog checkpoints in medulloblastoma: The chromosome 17p deletion paradigm. Trends Mol. Med. 2005;11:537–545. doi: 10.1016/j.molmed.2005.10.005. [DOI] [PubMed] [Google Scholar]
  • 137.Miele E., Po A., Begalli F., Antonucci L., Mastronuzzi A., Marras C.E., Carai A., Cucchi D., Abballe L., Besharat Z.M. β-Arrestin1-mediated acetylation of Gli1 regulates hedgehog/Gli signaling and modulates self-renewal of SHH medulloblastoma cancer stem cells. BMC Cancer. 2017;17:488. doi: 10.1186/s12885-017-3477-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Northcott P.A., Fernandez-L A., Hagan J.P., Ellison D.W., Grajkowska W., Gillespie Y., Grundy R., Van Meter T., Rutka J.T., Croce C.M. The miR-17/92 polycistron is up-regulated in Sonic Hedgehog-driven medulloblastomas and induced by N-myc in Sonic Hedgehog-treated cerebellar neural precursors. Cancer Res. 2009;69:3249–3255. doi: 10.1158/0008-5472.CAN-08-4710. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Murphy B.L., Obad S., Bihannic L., Ayrault O., Zindy F., Kauppinen S., Roussel M.F. Silencing of the miR-17∼92 cluster family inhibits medulloblastoma progression. Cancer Res. 2013;73:7068–7078. doi: 10.1158/0008-5472.CAN-13-0927. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Uziel T., Karginov F.V., Xie S., Parker J.S., Wang Y.D., Gajjar A., He L., Ellison D., Gilbertson R.J., Hannon G., Roussel M.F. The miR-17∼92 cluster collaborates with the Sonic Hedgehog pathway in medulloblastoma. Proc. Natl. Acad. Sci. USA. 2009;106:2812–2817. doi: 10.1073/pnas.0809579106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Zhang Z., Li S., Cheng S.Y. The miR-183∼96∼182 cluster promotes tumorigenesis in a mouse model of medulloblastoma. J. Biomed. Res. 2013;27:486–494. doi: 10.7555/JBR.27.20130010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Zindy F., Kawauchi D., Lee Y., Ayrault O., Ben Merzoug L., McKinnon P.J., Ventura A., Roussel M.F. Role of the miR-17∼92 cluster family in cerebellar and medulloblastoma development. Biol. Open. 2014;3:597–605. doi: 10.1242/bio.20146734. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Weeraratne S.D., Amani V., Teider N., Pierre-Francois J., Winter D., Kye M.J., Sengupta S., Archer T., Remke M., Bai A.H. Pleiotropic effects of miR-183∼96∼182 converge to regulate cell survival, proliferation and migration in medulloblastoma. Acta Neuropathol. 2012;123:539–552. doi: 10.1007/s00401-012-0969-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Constantin L., Wainwright B.J. MicroRNAs promote granule cell expansion in the cerebellum through Gli2. Cerebellum. 2015;14:688–698. doi: 10.1007/s12311-015-0672-x. [DOI] [PubMed] [Google Scholar]
  • 145.Besharat Z.M., Sabato C., Po A., Gianno F., Abballe L., Napolitano M., Miele E., Giangaspero F., Vacca A., Catanzaro G., Ferretti E. Low expression of miR-466f-3p sustains epithelial to mesenchymal transition in Sonic Hedgehog medulloblastoma stem cells through Vegfa-Nrp2 signaling pathway. Front. Pharmacol. 2018;9:1281. doi: 10.3389/fphar.2018.01281. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Gershanov S., Toledano H., Michowiz S., Barinfeld O., Pinhasov A., Goldenberg-Cohen N., Salmon-Divon M. MicroRNA-mRNA expression profiles associated with medulloblastoma subgroup 4. Cancer Manag. Res. 2018;10:339–352. doi: 10.2147/CMAR.S156709. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Shi P.F., Ji H.L., Luo Y.K., Mao T.M., Chen X., Zhou K.Y. Effect of long noncoding RNA SPRY4-IT1 on proliferation and metastasis of medulloblastoma. Zhongguo Ying Yong Sheng Li Xue Za Zhi. 2017;33:78–82. doi: 10.12047/j.cjap.5448.2017.020. [DOI] [PubMed] [Google Scholar]
  • 148.Lv T., Miao Y.F., Jin K., Han S., Xu T.Q., Qiu Z.L., Zhang X.H. Dysregulated circular RNAs in medulloblastoma regulate proliferation and growth of tumor cells via host genes. Cancer Med. 2018;7:6147–6157. doi: 10.1002/cam4.1613. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Pajtler K.W., Mack S.C., Ramaswamy V., Smith C.A., Witt H., Smith A., Hansford J.R., von Hoff K., Wright K.D., Hwang E. The current consensus on the clinical management of intracranial ependymoma and its distinct molecular variants. Acta Neuropathol. 2017;133:5–12. doi: 10.1007/s00401-016-1643-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Wu J., Armstrong T.S., Gilbert M.R. Biology and management of ependymomas. Neuro-oncol. 2016;18:902–913. doi: 10.1093/neuonc/now016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Lopes M.B.S. The 2017 World Health Organization classification of tumors of the pituitary gland: A summary. Acta Neuropathol. 2017;134:521–535. doi: 10.1007/s00401-017-1769-8. [DOI] [PubMed] [Google Scholar]
  • 152.Hübner J.M., Kool M., Pfister S.M., Pajtler K.W. Epidemiology, molecular classification and WHO grading of ependymoma. J. Neurosurg. Sci. 2018;62:46–50. doi: 10.23736/S0390-5616.17.04152-2. [DOI] [PubMed] [Google Scholar]
  • 153.Rudà R., Reifenberger G., Frappaz D., Pfister S.M., Laprie A., Santarius T., Roth P., Tonn J.C., Soffietti R., Weller M., Moyal E.C. EANO guidelines for the diagnosis and treatment of ependymal tumors. Neuro-oncol. 2018;20:445–456. doi: 10.1093/neuonc/nox166. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Pajtler K.W., Witt H., Sill M., Jones D.T., Hovestadt V., Kratochwil F., Wani K., Tatevossian R., Punchihewa C., Johann P. Molecular classification of ependymal tumors across all CNS compartments, histopathological grades, and age groups. Cancer Cell. 2015;27:728–743. doi: 10.1016/j.ccell.2015.04.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Costa F.F., Bischof J.M., Vanin E.F., Lulla R.R., Wang M., Sredni S.T., Rajaram V., Bonaldo Mde.F., Wang D., Goldman S. Identification of microRNAs as potential prognostic markers in ependymoma. PLoS ONE. 2011;6:e25114. doi: 10.1371/journal.pone.0025114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Cipro Š., Belhajová M., Eckschlager T., Zámečník J. MicroRNA expression in pediatric intracranial ependymomas and their potential value for tumor grading. Oncol. Lett. 2019;17:1379–1383. doi: 10.3892/ol.2018.9685. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Tantawy M., Elzayat M.G., Yehia D., Taha H. Identification of microRNA signature in different pediatric brain tumors. Genet. Mol. Biol. 2018;41:27–34. doi: 10.1590/1678-4685-GMB-2016-0334. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Zakrzewska M., Fendler W., Zakrzewski K., Sikorska B., Grajkowska W., Dembowska-Bagińska B., Filipek I., Stefańczyk Ł., Liberski P.P. Altered microRNA expression is associated with tumor grade, molecular background and outcome in childhood infratentorial ependymoma. PLoS ONE. 2016;11:e0158464. doi: 10.1371/journal.pone.0158464. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Margolin-Miller Y., Yanichkin N., Shichrur K., Toledano H., Ohali A., Tzaridis T., Michowitz S., Fichman-Horn S., Feinmesser M., Pfister S.M. Prognostic relevance of miR-124-3p and its target TP53INP1 in pediatric ependymoma. Genes Chromosomes Cancer. 2017;56:639–650. doi: 10.1002/gcc.22467. [DOI] [PubMed] [Google Scholar]
  • 160.Ahram M., Amarin J.Z., Suradi H.H., Abdelhamid S.S., Makhamreh M.M., Bawadi R.M., Al-Hussaini M. Association of microRNAs with the clinicopathologic characteristics of ependymoma. J. Mol. Neurosci. 2018;66:307–313. doi: 10.1007/s12031-018-1178-z. [DOI] [PubMed] [Google Scholar]
  • 161.Liang Y., Yang W., Zhu Y., Yuan Y. Prognostic role of microRNA-203 in various carcinomas: Evidence from a meta-analysis involving 13 studies. Springerplus. 2016;5:1538. doi: 10.1186/s40064-016-3225-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Lourdusamy A., Luo L.Z., Storer L.C., Cohen K.J., Resar L., Grundy R.G. Transcriptomic analysis in pediatric spinal ependymoma reveals distinct molecular signatures. Oncotarget. 2017;8:115570–115581. doi: 10.18632/oncotarget.23311. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Yang B., Dai J.X., Pan Y.B., Ma Y.B., Chu S.H. Identification of biomarkers and construction of a microRNA-mRNA regulatory network for ependymoma using integrated bioinformatics analysis. Oncol. Lett. 2019;18:6079–6089. doi: 10.3892/ol.2019.10941. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Lourdusamy A., Rahman R., Smith S., Grundy R. MicroRNA network analysis identifies miR-29 cluster as key regulator of LAMA2 in ependymoma. Acta Neuropathol. Commun. 2015;3:26. doi: 10.1186/s40478-015-0206-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Shu C., Wang Q., Yan X., Wang J. Prognostic and microRNA profile analysis for CD44 positive expression pediatric posterior fossa ependymoma. Clin. Transl. Oncol. 2018;20:1439–1447. doi: 10.1007/s12094-018-1876-6. [DOI] [PubMed] [Google Scholar]
  • 166.Liu F., Dong H., Mei Z., Huang T. Investigation of miRNA and mRNA co-expression network in ependymoma. Front. Bioeng. Biotechnol. 2020;8:177. doi: 10.3389/fbioe.2020.00177. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.Wang L., Zhang C., Xie Y., Jiang W., Huang J., Guo S., Xu F., Wang J. Detecting the long non-coding RNA signature related to spinal cord ependymal tumor subtype using a genome-wide methylome analysis approach. Mol. Med. Rep. 2019;20:1531–1540. doi: 10.3892/mmr.2019.10388. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Jones C., Baker S.J. Unique genetic and epigenetic mechanisms driving paediatric diffuse high-grade glioma. Nat. Rev. Cancer. 2014;14:651–661. doi: 10.1038/nrc3811. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Cohen K.J., Jabado N., Grill J. Diffuse intrinsic pontine gliomas-current management and new biologic insights. Is there a glimmer of hope? Neuro-oncol. 2017;19:1025–1034. doi: 10.1093/neuonc/nox021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170.Kaye E.C., Baker J.N., Broniscer A. Management of diffuse intrinsic pontine glioma in children: Current and future strategies for improving prognosis. CNS Oncol. 2014;3:421–431. doi: 10.2217/cns.14.47. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171.Rashed W.M., Maher E., Adel M., Saber O., Zaghloul M.S. Pediatric diffuse intrinsic pontine glioma: Where do we stand? Cancer Metastasis Rev. 2019;38:759–770. doi: 10.1007/s10555-019-09824-2. [DOI] [PubMed] [Google Scholar]
  • 172.Gajjar A., Bowers D.C., Karajannis M.A., Leary S., Witt H., Gottardo N.G. Pediatric brain tumors: Innovative genomic information is transforming the diagnostic and clinical landscape. J. Clin. Oncol. 2015;33:2986–2998. doi: 10.1200/JCO.2014.59.9217. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Ramaswamy V., Remke M., Taylor M.D. An epigenetic therapy for diffuse intrinsic pontine gliomas. Nat. Med. 2014;20:1378–1379. doi: 10.1038/nm.3769. [DOI] [PubMed] [Google Scholar]
  • 174.Buczkowicz P., Bartels U., Bouffet E., Becher O., Hawkins C. Histopathological spectrum of paediatric diffuse intrinsic pontine glioma: Diagnostic and therapeutic implications. Acta Neuropathol. 2014;128:573–581. doi: 10.1007/s00401-014-1319-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.Puget S., Beccaria K., Blauwblomme T., Roujeau T., James S., Grill J., Zerah M., Varlet P., Sainte-Rose C. Biopsy in a series of 130 pediatric diffuse intrinsic pontine gliomas. Childs Nerv. Syst. 2015;31:1773–1780. doi: 10.1007/s00381-015-2832-1. [DOI] [PubMed] [Google Scholar]
  • 176.Buczkowicz P., Hoeman C., Rakopoulos P., Pajovic S., Letourneau L., Dzamba M., Morrison A., Lewis P., Bouffet E., Bartels U. Genomic analysis of diffuse intrinsic pontine gliomas identifies three molecular subgroups and recurrent activating ACVR1 mutations. Nat. Genet. 2014;46:451–456. doi: 10.1038/ng.2936. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Schwartzentruber J., Korshunov A., Liu X.Y., Jones D.T., Pfaff E., Jacob K., Sturm D., Fontebasso A.M., Quang D.A., Tönjes M. Driver mutations in histone H3.3 and chromatin remodelling genes in paediatric glioblastoma. Nature. 2012;482:226–231. doi: 10.1038/nature10833. [DOI] [PubMed] [Google Scholar]
  • 178.Wu G., Broniscer A., McEachron T.A., Lu C., Paugh B.S., Becksfort J., Qu C., Ding L., Huether R., Parker M., St. Jude Children’s Research Hospital–Washington University Pediatric Cancer Genome Project Somatic histone H3 alterations in pediatric diffuse intrinsic pontine gliomas and non-brainstem glioblastomas. Nat. Genet. 2012;44:251–253. doi: 10.1038/ng.1102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179.Kluiver T.A., Alieva M., van Vuurden D.G., Wehrens E.J., Rios A.C. Invaders exposed: Understanding and targeting tumor cell invasion in diffuse intrinsic pontine glioma. Front. Oncol. 2020;10:92. doi: 10.3389/fonc.2020.00092. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180.Yadavilli S., Scafidi J., Becher O.J., Saratsis A.M., Hiner R.L., Kambhampati M., Mariarita S., MacDonald T.J., Codispoti K.E., Magge S.N. The emerging role of NG2 in pediatric diffuse intrinsic pontine glioma. Oncotarget. 2015;6:12141–12155. doi: 10.18632/oncotarget.3716. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.Wei L., He F., Zhang W., Chen W., Yu B. Bioinformatics analysis of microarray data to reveal the pathogenesis of diffuse intrinsic pontine glioma. Biol. Res. 2018;51:26. doi: 10.1186/s40659-018-0175-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182.Liu Y., Liu H., Zhang D. Identification of novel long non-coding RNA in diffuse intrinsic pontine gliomas by expression profile analysis. Oncol. Lett. 2018;16:6401–6406. doi: 10.3892/ol.2018.9461. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183.Müller H.L., Merchant T.E., Warmuth-Metz M., Martinez-Barbera J.P., Puget S. Craniopharyngioma. Nat. Rev. Dis. Primers. 2019;5:75. doi: 10.1038/s41572-019-0125-9. [DOI] [PubMed] [Google Scholar]
  • 184.Müller H.L., Merchant T.E., Puget S., Martinez-Barbera J.P. New outlook on the diagnosis, treatment and follow-up of childhood-onset craniopharyngioma. Nat. Rev. Endocrinol. 2017;13:299–312. doi: 10.1038/nrendo.2016.217. [DOI] [PubMed] [Google Scholar]
  • 185.Goschzik T., Gessi M., Dreschmann V., Gebhardt U., Wang L., Yamaguchi S., Wheeler D.A., Lauriola L., Lau C.C., Müller H.L., Pietsch T. Genomic alterations of adamantinomatous and papillary craniopharyngioma. J. Neuropathol. Exp. Neurol. 2017;76:126–134. doi: 10.1093/jnen/nlw116. [DOI] [PubMed] [Google Scholar]
  • 186.Hölsken A., Sill M., Merkle J., Schweizer L., Buchfelder M., Flitsch J., Fahlbusch R., Metzler M., Kool M., Pfister S.M. Adamantinomatous and papillary craniopharyngiomas are characterized by distinct epigenomic as well as mutational and transcriptomic profiles. Acta Neuropathol. Commun. 2016;4:20. doi: 10.1186/s40478-016-0287-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187.Müller H.L. Childhood craniopharyngioma—Current concepts in diagnosis, therapy and follow-up. Nat. Rev. Endocrinol. 2010;6:609–618. doi: 10.1038/nrendo.2010.168. [DOI] [PubMed] [Google Scholar]
  • 188.Muller H.L. Childhood craniopharyngioma. Recent advances in diagnosis, treatment and follow-up. Horm. Res. 2008;69:193–202. doi: 10.1159/000113019. [DOI] [PubMed] [Google Scholar]
  • 189.Müller H.L. Consequences of craniopharyngioma surgery in children. J. Clin. Endocrinol. Metab. 2011;96:1981–1991. doi: 10.1210/jc.2011-0174. [DOI] [PubMed] [Google Scholar]
  • 190.Müller H.L. Craniopharyngioma and hypothalamic injury: Latest insights into consequent eating disorders and obesity. Curr. Opin. Endocrinol. Diabetes Obes. 2016;23:81–89. doi: 10.1097/MED.0000000000000214. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191.Campanini M.L., Colli L.M., Paixao B.M., Cabral T.P., Amaral F.C., Machado H.R., Neder L.S., Saggioro F., Moreira A.C., Antonini S.R., de Castro M. CTNNB1 gene mutations, pituitary transcription factors, and microRNA expression involvement in the pathogenesis of adamantinomatous craniopharyngiomas. Horm. Cancer. 2010;1:187–196. doi: 10.1007/s12672-010-0041-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192.Samis J., Vanin E.F., Sredni S.T., de Bonaldo Mde F., Costa F.F., Tomita T., Habiby R., Zimmerman D., Soares M.B. Extensive miRNA expression analysis in craniopharyngiomas. Childs Nerv. Syst. 2016;32:1617–1624. doi: 10.1007/s00381-016-3131-1. [DOI] [PubMed] [Google Scholar]
  • 193.Lee R.S., Stewart C., Carter S.L., Ambrogio L., Cibulskis K., Sougnez C., Lawrence M.S., Auclair D., Mora J., Golub T.R. A remarkably simple genome underlies highly malignant pediatric rhabdoid cancers. J. Clin. Invest. 2012;122:2983–2988. doi: 10.1172/JCI64400. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194.Hasselblatt M., Isken S., Linge A., Eikmeier K., Jeibmann A., Oyen F., Nagel I., Richter J., Bartelheim K., Kordes U. High-resolution genomic analysis suggests the absence of recurrent genomic alterations other than SMARCB1 aberrations in atypical teratoid/rhabdoid tumors. Genes Chromosomes Cancer. 2013;52:185–190. doi: 10.1002/gcc.22018. [DOI] [PubMed] [Google Scholar]
  • 195.Sredni S.T., Bonaldo Mde F., Costa F.F., Huang C.C., Hamm C.A., Rajaram V., Tomita T., Goldman S., Bischof J.M., Soares M.B. Upregulation of mir-221 and mir-222 in atypical teratoid/rhabdoid tumors: Potential therapeutic targets. Childs Nerv. Syst. 2010;26:279–283. doi: 10.1007/s00381-009-1028-y. [DOI] [PubMed] [Google Scholar]
  • 196.Hsieh T.H., Chien C.L., Lee Y.H., Lin C.I., Hsieh J.Y., Chao M.E., Liu D.J., Chu S.S., Chen W., Lin S.C. Downregulation of SUN2, a novel tumor suppressor, mediates miR-221/222-induced malignancy in central nervous system embryonal tumors. Carcinogenesis. 2014;35:2164–2174. doi: 10.1093/carcin/bgu105. [DOI] [PubMed] [Google Scholar]
  • 197.Zhang K., Gao H., Wu X., Wang J., Zhou W., Sun G., Wang J., Wang Y., Mu B., Kim C. Frequent overexpression of HMGA2 in human atypical teratoid/rhabdoid tumor and its correlation with let-7a3/let-7b miRNA. Clin. Cancer Res. 2014;20:1179–1189. doi: 10.1158/1078-0432.CCR-13-1452. [DOI] [PubMed] [Google Scholar]
  • 198.Lee Y.Y., Yang Y.P., Huang M.C., Wang M.L., Yen S.H., Huang P.I., Chen Y.W., Chiou S.H., Lan Y.T., Ma H.I. MicroRNA142-3p promotes tumor-initiating and radioresistant properties in malignant pediatric brain tumors. Cell Transplant. 2014;23:669–690. doi: 10.3727/096368914X678364. [DOI] [PubMed] [Google Scholar]
  • 199.Yang Y.P., Nguyen P.N.N., Ma H.I., Ho W.J., Chen Y.W., Chien Y., Yarmishyn A.A., Huang P.I., Lo W.L., Wang C.Y. Tumor mesenchymal stromal cells regulate cell migration of atypical teratoid rhabdoid tumor through exosome-mediated miR155/SMARCA4 pathway. Cancers (Basel) 2019;11:720. doi: 10.3390/cancers11050720. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 200.Bondy M.L., Scheurer M.E., Malmer B., Barnholtz-Sloan J.S., Davis F.G., Il’yasova D., Kruchko C., McCarthy B.J., Rajaraman P., Schwartzbaum J.A., Brain Tumor Epidemiology Consortium Brain tumor epidemiology: Consensus from the Brain Tumor Epidemiology Consortium. Cancer. 2008;113(7, Suppl):1953–1968. doi: 10.1002/cncr.23741. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 201.Fangusaro J. Pediatric high grade glioma: A review and update on tumor clinical characteristics and biology. Front. Oncol. 2012;2:105. doi: 10.3389/fonc.2012.00105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 202.Novakova J., Slaby O., Vyzula R., Michalek J. MicroRNA involvement in glioblastoma pathogenesis. Biochem. Biophys. Res. Commun. 2009;386:1–5. doi: 10.1016/j.bbrc.2009.06.034. [DOI] [PubMed] [Google Scholar]
  • 203.Sturm D., Bender S., Jones D.T., Lichter P., Grill J., Becher O., Hawkins C., Majewski J., Jones C., Costello J.F. Paediatric and adult glioblastoma: Multiform (epi)genomic culprits emerge. Nat. Rev. Cancer. 2014;14:92–107. doi: 10.1038/nrc3655. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 204.Stupp R., Hegi M.E., Mason W.P., van den Bent M.J., Taphoorn M.J., Janzer R.C., Ludwin S.K., Allgeier A., Fisher B., Belanger K., European Organisation for Research and Treatment of Cancer Brain Tumour and Radiation Oncology Groups. National Cancer Institute of Canada Clinical Trials Group Effects of radiotherapy with concomitant and adjuvant temozolomide versus radiotherapy alone on survival in glioblastoma in a randomised phase III study: 5-year analysis of the EORTC-NCIC trial. Lancet Oncol. 2009;10:459–466. doi: 10.1016/S1470-2045(09)70025-7. [DOI] [PubMed] [Google Scholar]
  • 205.DeOcesano-Pereira C., Machado R.A.C., Chudzinski-Tavassi A.M., Sogayar M.C. Emerging roles and potential applications of non-coding RNAs in glioblastoma. Int. J. Mol. Sci. 2020;21:2611. doi: 10.3390/ijms21072611. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 206.Luo J.W., Wang X., Yang Y., Mao Q. Role of micro-RNA (miRNA) in pathogenesis of glioblastoma. Eur. Rev. Med. Pharmacol. Sci. 2015;19:1630–1639. [PubMed] [Google Scholar]
  • 207.Papagiannakopoulos T., Shapiro A., Kosik K.S. MicroRNA-21 targets a network of key tumor-suppressive pathways in glioblastoma cells. Cancer Res. 2008;68:8164–8172. doi: 10.1158/0008-5472.CAN-08-1305. [DOI] [PubMed] [Google Scholar]
  • 208.Chao T.F., Xiong H.H., Liu W., Chen Y., Zhang J.X. miR-21 mediates the radiation resistance of glioblastoma cells by regulating PDCD4 and hMSH2. J. Huazhong Univ. Sci. Technolog. Med. Sci. 2013;33:525–529. doi: 10.1007/s11596-013-1153-4. [DOI] [PubMed] [Google Scholar]
  • 209.Zhang C., Zhang J., Hao J., Shi Z., Wang Y., Han L., Yu S., You Y., Jiang T., Wang J. High level of miR-221/222 confers increased cell invasion and poor prognosis in glioma. J. Transl. Med. 2012;10:119. doi: 10.1186/1479-5876-10-119. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 210.Hao J., Zhang C., Zhang A., Wang K., Jia Z., Wang G., Han L., Kang C., Pu P. miR-221/222 is the regulator of Cx43 expression in human glioblastoma cells. Oncol. Rep. 2012;27:1504–1510. doi: 10.3892/or.2012.1652. [DOI] [PubMed] [Google Scholar]
  • 211.Medina R., Zaidi S.K., Liu C.G., Stein J.L., van Wijnen A.J., Croce C.M., Stein G.S. MicroRNAs 221 and 222 bypass quiescence and compromise cell survival. Cancer Res. 2008;68:2773–2780. doi: 10.1158/0008-5472.CAN-07-6754. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 212.Quintavalle C., Garofalo M., Zanca C., Romano G., Iaboni M., del Basso De Caro M., Martinez-Montero J.C., Incoronato M., Nuovo G., Croce C.M., Condorelli G. miR-221/222 overexpession in human glioblastoma increases invasiveness by targeting the protein phosphate PTPμ. Oncogene. 2012;31:858–868. doi: 10.1038/onc.2011.280. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 213.Zhang C.Z., Kang C.S., Pu P.Y., Wang G.X., Jia Z.F., Zhang A.L., Han L., Xu P. [Inhibitory effect of knocking down microRNA-221 and microRNA-222 on glioma cell growth in vitro and in vivo] Zhonghua Zhong Liu Za Zhi. 2009;31:721–726. [PubMed] [Google Scholar]
  • 214.Zhang C.Z., Zhang J.X., Zhang A.L., Shi Z.D., Han L., Jia Z.F., Yang W.D., Wang G.X., Jiang T., You Y.P. miR-221 and miR-222 target PUMA to induce cell survival in glioblastoma. Mol. Cancer. 2010;9:229. doi: 10.1186/1476-4598-9-229. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 215.Jha P., Agrawal R., Pathak P., Kumar A., Purkait S., Mallik S., Suri V., Chand Sharma M., Gupta D., Suri A. Genome-wide small noncoding RNA profiling of pediatric high-grade gliomas reveals deregulation of several miRNAs, identifies downregulation of snoRNA cluster HBII-52 and delineates H3F3A and TP53 mutant-specific miRNAs and snoRNAs. Int. J. Cancer. 2015;137:2343–2353. doi: 10.1002/ijc.29610. [DOI] [PubMed] [Google Scholar]
  • 216.Miele E., Buttarelli F.R., Arcella A., Begalli F., Garg N., Silvano M., Po A., Baldi C., Carissimo G., Antonelli M. High-throughput microRNA profiling of pediatric high-grade gliomas. Neuro-oncol. 2014;16:228–240. doi: 10.1093/neuonc/not215. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 217.Giunti L., Da Ros M., De Gregorio V., Magi A., Landini S., Mazzinghi B., Buccoliero A.M., Genitori L., Giglio S., Sardi I. A microRNA profile of pediatric glioblastoma: The role of NUCKS1 upregulation. Mol. Clin. Oncol. 2019;10:331–338. doi: 10.3892/mco.2019.1795. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 218.Ames H.M., Yuan M., Vizcaíno M.A., Yu W., Rodriguez F.J. MicroRNA profiling of low-grade glial and glioneuronal tumors shows an independent role for cluster 14q32.31 member miR-487b. Mod. Pathol. 2017;30:204–216. doi: 10.1038/modpathol.2016.177. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 219.Liu A., Zhao H., Sun B., Han X., Zhou D., Cui Z., Ma X., Zhang J., Yuan L. A predictive analysis approach for paediatric and adult high-grade glioma: miRNAs and network insight. Ann. Transl. Med. 2020;8:242. doi: 10.21037/atm.2020.01.12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 220.Li X., Diao H. Circular RNA circ_0001946 acts as a competing endogenous RNA to inhibit glioblastoma progression by modulating miR-671-5p and CDR1. J. Cell. Physiol. 2019;234:13807–13819. doi: 10.1002/jcp.28061. [DOI] [PubMed] [Google Scholar]
  • 221.Huang J., Peng J., Guo L. Non-coding RNA: A new tool for the diagnosis, prognosis, and therapy of small cell lung cancer. J. Thorac. Oncol. 2015;10:28–37. doi: 10.1097/JTO.0000000000000394. [DOI] [PubMed] [Google Scholar]
  • 222.Slack F.J., Chinnaiyan A.M. The role of non-coding RNAs in oncology. Cell. 2019;179:1033–1055. doi: 10.1016/j.cell.2019.10.017. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Molecular Therapy. Nucleic Acids are provided here courtesy of The American Society of Gene & Cell Therapy

RESOURCES