Introduction

Medulloblastoma is recognized as the most prevalent form of brain tumor among children, accounting for nearly 20% of all brain tumors found in pediatric patients [1]. Medulloblastoma is most commonly diagnosed before age 15 and has two incidence peaks between the ages of 3–4 and 8–9 [2]. The occurrence of this tumor in older patients is rare, comprising less than 1% of all primary CNS tumors in adults [3]. According to the latest classification scheme, medulloblastoma is divided into two distinct categories, i.e., histologically defined and genetically defined. Histologically, medulloblastoma can be categorized into different types, including classic, desmoplastic/nodular (DN), and large cell/anaplastic (LCA). Genetic classification divides it into four molecular subtypes, i.e., wingless (WNT), sonic hedgehog (SHH), group 3, and group 4. Each of these subtypes has distinct clinical and molecular characteristics [4]. The primary therapeutic strategies for medulloblastoma include a combination of surgical resection, radiotherapy, and chemotherapy. However, clinical outcomes have been not desirable, and 5-year survival rates range between 60 and 80% [5, 6]. Hence, an in-depth investigation of the molecular mechanisms involved in the pathogenesis of medulloblastoma is essential to improve patients' prognoses and clinical outcomes.

Non-coding RNAs (ncRNAs) are a class of RNAs that lack the ability to encode functional proteins [7]. Since their discovery, the biological significance of ncRNAs has become increasingly evident, which has led to a shift in the perspective of RNA from being a simple intermediary of protein synthesis to being a functional molecule with essential roles in regulating gene expression and genome organization [8]. In recent years, studies have indicated that these ncRNAs play vital regulatory roles in the initiation and progression of various types of cancer [7]. In line with this, it has been demonstrated that the expression levels of various ncRNAs are notably different between medulloblastoma and normal cerebellar cells [9]. NcRNAs can be classified into various classes based on size and function. The three primary classes of regulatory ncRNAs are microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and circular RNAs (circRNAs). Owing to their intrinsic characteristics, they may exhibit tissue or disease specificity and can be detected in all bodily fluids, which makes them potentially desirable to be used as biomarkers [10]. LncRNAs are a class of RNA molecules that are longer than 200 nucleotides in length [11]. Through multiple mechanisms, they play a crucial role in regulating gene expression at various levels, including epigenetic, transcriptional, and post-transcriptional regulation. This regulatory role holds particular significance in the central nervous system (CNS) [12]. Most lncRNAs are presumed to be transcribed and processed similarly to mRNAs. They are primarily transcribed by RNA polymerase II and often possess 5′-end m7G caps and 3′-end poly(A) tails [13]. Regarding the chromosomal position, lncRNAs are classified into promoter-associated lncRNAs, antisense, intronic, enhancer RNAs, divergent, intergenic, and transcription start site-associated lncRNAs [14]. LncRNAs function as competing endogenous RNAs (ceRNAs) within a regulatory network by serving as a "sponge" for target miRNAs [15]. Circular RNAs (circRNAs) are single-stranded RNA transcripts with a covalently closed circular structure. They are produced through an alternative splicing process and lack the 5′ caps and 3′ poly(A) tails; this structural feature in circRNAs renders them resistant to degradation by ribonucleases [16]. Furthermore, most circRNAs exhibit evolutionary conservation across various species [17]. CircRNAs are generated through the transcription of precursor mRNA (pre-mRNA) by RNA polymerase II. They can function as molecular sponges for miRNAs, thus regulating their biological activities [18]. Recent evidence has indicated that aberrant expression of circRNAs occurs in various types of cancer, including breast cancer [19], pancreatic ductal adenocarcinoma [20], bladder carcinoma [21], glioblastoma [

Method

Sco** review protocol

The guidelines for preferred reporting items for systematic reviews and meta-analyses extension for sco** reviews (PRISMA-ScR) are followed by the present sco** review [39] (Supplementary data). The five steps of the present sco** review include formulating the research question, identifying the relevant publications, selecting studies, charting the data, and summarizing and disclosing the findings.

Research question

Given the significant role of the ceRNA networks in the regulation of gene expression, the present study aimed to comprehensively review the current knowledge on the circRNA and lncRNA-associated ceRNA networks in medulloblastoma development.

Relevant publication identification

The Web of Science, PubMed, Scopus, and Embase were systematically searched to find the relevant studies published before 16 September 2023; the systematic searches did not have any restriction on language, country, or time. LncRNA, circRNA, miRNA, ceRNA, medulloblastoma, and their different versions, along with the Emtree and medical subject headings (MeSh) terms, were used for the systematic search.

Study selection

After retrieving the publications from the above-mentioned databases and removing duplicated records, the papers were reviewed in two phases. In the first phase, the title and abstract of the obtained studies were reviewed. In the second phase, the full texts of the remaining papers were thoroughly reviewed. The criteria for inclusion were the following. First, the included study must be an original paper published in English. Second, the included study must study the interaction between lncRNA with miRNA or circRNA with miRNAs in medulloblastoma. Third, the experimental study must contain at least one of the human medulloblastoma cell lines.

Data charting

The studied ncRNAs and the related axis, medulloblastoma cell line, and the effect of the studied axis on medulloblastoma formation were all extracted from the included studies.

Summarizing and reporting the obtained results

The present sco** review summarizes the results of the studies that were included and also investigates the effect of the identified miRNAs, circRNAs, and lncRNAs on the development of medulloblastoma that were not present in the included studies.

In silico study

To extend the understanding of the impact of ceRNA on cellular singling pathways, miRPathDB v2.0 was used to access the Reactome database. A minimum of two significant miRNAs per pathway and strong experimental evidence were the criteria for the related analysis.

Results

Systematic search results

The systematic search on Web of Science, PubMed, Scopus, and Embase identified 149 papers published before 16 September 2023. After removing duplicated studies, 57 papers were also excluded based on reviewing their title and abstracts. Ultimately, seven papers were excluded from the present sco** review because they did not meet the above-mentioned inclusion criteria in the full-text assessment phase. Figure 1 shows the study's flowchart.

Fig. 1
figure 1

The flowchart of the study

The characteristics of the included papers

The included studies were published between 2017 and 2023. Daoy was the most studied medulloblastoma human cell line. According to the ATCC, this cell line was obtained from a 4-year-old white male with desmoplastic cerebellar type. HOTAIR, NEAT1, linc-NeD125, HHIP-AS1, CRNDE, and TP73-AS1 are the identified oncogenic lncRNA in medulloblastoma, and Nkx2-2as is a tumor-suppressive lncRNA in medulloblastoma. Based on the current experimental evidence, circSKA3, which sponges miR-326, miR-520 h, and miR-383-5p, and circRNA_103128, which sponges miR-129-5p, are upregulated oncogenic circRNA in medulloblastoma (Table 1).

Table 1 The characteristics of the included studies

The enrichment analysis

Based on the Reactome database, the identified miRNAs regulate various cellular pathways, like cell cycle, apoptosis, MAPK1/ERK2 pathway, etc. For instance, miR-106a-5p, miR-23a-3p, and miR-129-5p are enriched for apoptosis (Fig. 2).

Fig. 2
figure 2

The enrichment analysis of studied microRNAs

Discussion

Despite the recent advances in our understanding of medulloblastoma biology, the clinical outcome of affected patients is still unfavorable. A better understanding of ncRNAs and ceRNAs might provide valuable insights regarding treating medulloblastoma [40]. The following discusses the current evidence on the significance of circRNA and lncRNA-associated ceRNA networks in medulloblastoma.

HOTAIR-mediated ceRNA

As a located lncRNA on chromosome 12, homeobox transcript antisense intergenic RNA (HOTAIR) can interact with PRC2, LSD1, and miRNAs, leading to gene expression regulation [41]. Zhang et al. have reported that HOTAIR expression level is substantially upregulated in medulloblastoma tissues and cell lines compared with non-tumoral ones. HOTAIR knockdown improves apoptosis rate, decreases the cell viability, clonogenicity, migration, and invasion, and reduces tumor volume in animal models; this oncogenic effect is medicated via the HOTAIR/miR-1-3p and miR-206/ YY1 axes in medulloblastoma. Also, the ectopic expression of miR-1-3p and miR-206 has been associated with decreased tumor growth in animal models [42]. In line with this, it has been shown that miR-206 is downregulated in medulloblastoma tissues, and its increased expression decreased the cell viability and migration of medulloblastoma cells via the miR-206/LASP1 axis [43].

NEAT1-mediated ceRNA

As a component of nuclear paraspeckles, nuclear-enriched abundant transcript 1 (NEAT1) is located on chromosome 11q13.1; this lncRNA is dysregulated in various cancers, like glioma and medulloblastoma [44, 45]. Ge et al. have shown that NEAT1 knockdown increases the chemosensivity of medulloblastoma cells and potentiates cisplatin-mediated apoptosis activation. This chemoresistance of medulloblastoma cells is mediated via the NEAT1/miR-23a-3p/GLS axis [44]. In line with this, the in-silico results have shown that miR-23a-3p is enriched for apoptosis. Apoptosis is a type of regulated cell death that has two pathways, i.e., intrinsic and extrinsic pathways [46]. In the intrinsic pathway, the insertion of BAX and BAK into the mitochondrial membrane releases cytochrome c, leading to the activation of caspase-9 and executioner caspases. In the extrinsic pathway, the death-inducing signaling complex results in the activation of caspase-8, caspase-10, caspases-3, caspases-6, and caspases-7; these events stimulate apoptosis activation [47, 48]. In addition, the in-silico results have shown that miR-23a-3p is enriched for the MAPK pathway as well. The MAPK/ERK pathway is among the activated oncogenic pathways in medulloblastoma and its blockade is associated with decreased proliferation, stemness, and invasion in medulloblastoma cells [49, 50]. Also, it has been reported that NEAT1 knockdown increases the chemosensitivity of glioblastoma cells to temozolomide [51].

Linc-NeD125-mediated ceRNA

Linc-NeD125 is a long intergenic ncRNA that is located on chromosome 11. Laneve et al. have reported that linc-NeD125 expression level is substantially increased in G4 medulloblastoma, and its knockdown decreases the proliferation of G4 medulloblastoma cells and downregulates the protein expression of CDK6, MYCN, SNCAIP, and KDM6A via the linc-NeD125/miR-19a-3p, miR-19b-3p, miR-106a-5p/CDK6, MYCN, SNCAIP, and KDM6A axes [52]. Consistent with this, the in-silico results have shown that miR-106a-5p is enriched for the G1 phase, cyclin D-associated events at the G1 phase, cyclin E-associated events during G1-S transition, cyclin A-CDK2 associated events at S phase entry as well as apoptosis. The complex of cyclin D with CDK4/CDK6 causes the RB-phosphorization and RB-phosphorization-mediated E2F release, leading to the transition from the G1 phase to the S phase. Cyclin E1 and E2 bind and activate CDK2, leading to RB and p27KIP1 phosphorylation. The cyclin E/CDK2 active complex leads to the S phase initiation and cyclin A/CDK2 is formed near the end of the S phase and leads to cell cycle progression from the S phase to the G2 phase. The cyclin A/CDK1 complex results in M phase entry [

Availability of data and materials

The datasets analyzed during the current study for enrichment analyses are available in the miRPathDB V 2.0 (https://mpd.bioinf.uni-sb.de/overview.html).

Abbreviations

CNS:

Central nervous system

circRNA:

Circular RNA

lncRNA:

Long non-coding RNA

miRNA:

MicroRNA

ceRNA:

Competing endogenous RNA

DN:

Desmoplastic/nodular

LCA:

Large cell/anaplastic

WNT:

Wingless

SHH:

Sonic hedgehog

pri-miRNAs:

Primary miRNA

RISC:

RNA-induced silencing complex

MREs:

MiRNA recognition elements

PRISMA-ScR:

Preferred reporting items for systematic reviews and meta-analyses extension for sco** reviews

HOTAIR:

Homeobox transcript antisense intergenic RNA

HHIP-AS1:

Hedgehog interacting protein-antisense 1

CRNDE:

Colorectal neoplasia differentially expressed

MAPK:

Mitogen-activated protein kinase

ERK:

Extracellular signal-regulated kinase

References

  1. Udaka YT, Packer RJ. Pediatric brain tumors. Neurol Clin. 2018;36(3):533–56.

    Article  PubMed  Google Scholar 

  2. Crawford JR, MacDonald TJ, Packer RJ. Medulloblastoma in childhood: new biological advances. The Lancet Neurology. 2007;6(12):1073–85.

    Article  CAS  PubMed  Google Scholar 

  3. Koeller KK, Rushing EJ. From the archives of the AFIP: medulloblastoma: a comprehensive review with radiologic-pathologic correlation. Radiographics. 2003;23(6):1613–37.

    Article  PubMed  Google Scholar 

  4. Louis DN, Perry A, Reifenberger G, von Deimling A, Figarella-Branger D, Cavenee WK, et al. The 2016 World Health Organization classification of tumors of the central nervous system: a summary. Acta Neuropathol. 2016;131(6):803–20.

    Article  PubMed  Google Scholar 

  5. Thompson EM, Hielscher T, Bouffet E, Remke M, Luu B, Gururangan S, et al. Prognostic value of medulloblastoma extent of resection after accounting for molecular subgroup: a retrospective integrated clinical and molecular analysis. Lancet Oncol. 2016;17(4):484–95.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Packer RJ, Zhou T, Holmes E, Vezina G, Gajjar A. Survival and secondary tumors in children with medulloblastoma receiving radiotherapy and adjuvant chemotherapy: results of Children’s Oncology Group trial A9961. Neuro Oncol. 2013;15(1):97–103.

    Article  CAS  PubMed  Google Scholar 

  7. Toden S, Zumwalt TJ, Goel A. Non-coding RNAs and potential therapeutic targeting in cancer. Biochim Biophys Acta. 2021;1875(1):188491.

    CAS  Google Scholar 

  8. Nemeth K, Bayraktar R, Ferracin M, Calin GA. Non-coding RNAs in disease: from mechanisms to therapeutics. Nat Rev Genet. 2024;25(3):211–32.

    Article  CAS  PubMed  Google Scholar 

  9. Joshi P, Katsushima K, Zhou R, Meoded A, Stapleton S, Jallo G, et al. The therapeutic and diagnostic potential of regulatory noncoding RNAs in medulloblastoma. Neuro-oncol Adv. 2019;1(1):vdz023.

    Article  Google Scholar 

  10. Kim T, Reitmair A. Non-coding RNAs: functional aspects and diagnostic utility in oncology. Int J Mol Sci. 2013;14(3):4934–68.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Quinn JJ, Chang HY. Unique features of long non-coding RNA biogenesis and function. Nat Rev Genet. 2016;17(1):47–62.

    Article  CAS  PubMed  Google Scholar 

  12. Knauss JL, Sun T. Regulatory mechanisms of long noncoding RNAs in vertebrate central nervous system development and function. Neuroscience. 2013;235:200–14.

    Article  CAS  PubMed  Google Scholar 

  13. Statello L, Guo CJ, Chen LL, Huarte M. Gene regulation by long non-coding RNAs and its biological functions. Nat Rev Mol Cell Biol. 2021;22(2):96–118.

    Article  CAS  PubMed  Google Scholar 

  14. Gao N, Li Y, Li J, Gao Z, Yang Z, Li Y, et al. Long non-coding RNAs: the regulatory mechanisms, research strategies, and future directions in cancers. Front Oncol. 2020;10:598817.

    Article  PubMed  PubMed Central  Google Scholar 

  15. López-Urrutia E, Bustamante Montes LP, de Guevara Cervantes DL, Pérez-Plasencia C, Campos-Parra AD. Crosstalk between long non-coding RNAs, Micro-RNAs and mRNAs: deciphering molecular mechanisms of master regulators in cancer. Front Oncol. 2019;9:669.

    Article  PubMed  PubMed Central  Google Scholar 

  16. Shang Q, Yang Z, Jia R, Ge S. The novel roles of circRNAs in human cancer. Mol Cancer. 2019;18(1):6.

    Article  PubMed  PubMed Central  Google Scholar 

  17. Pamudurti NR, Bartok O, Jens M, Ashwal-Fluss R, Stottmeister C, Ruhe L, et al. Translation of CircRNAs. Mol Cell. 2017;66(1):9-21.e7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Kristensen LS, Hansen TB, Venø MT, Kjems J. Circular RNAs in cancer: opportunities and challenges in the field. Oncogene. 2018;37(5):555–65.

    Article  CAS  PubMed  Google Scholar 

  19. Lü L, Sun J, Shi P, Kong W, Xu K, He B, et al. Identification of circular RNAs as a promising new class of diagnostic biomarkers for human breast cancer. Oncotarget. 2017;8(27):44096–107.

    Article  PubMed  PubMed Central  Google Scholar 

  20. Li H, Hao X, Wang H, Liu Z, He Y, Pu M, et al. Circular RNA expression profile of pancreatic ductal adenocarcinoma revealed by microarray. Cell Physiol Biochem. 2016;40(6):1334–44.

    Article  CAS  PubMed  Google Scholar 

  21. Huang M, Zhong Z, Lv M, Shu J, Tian Q, Chen J. Comprehensive analysis of differentially expressed profiles of lncRNAs and circRNAs with associated co-expression and ceRNA networks in bladder carcinoma. Oncotarget. 2016;7(30):47186–200.

    Article  PubMed  PubMed Central  Google Scholar 

  22. Zhu J, Ye J, Zhang L, **a L, Hu H, Jiang H, et al. Differential expression of circular RNAs in glioblastoma multiforme and its correlation with prognosis. Transl Oncol. 2017;10(2):271–9.

    Article  PubMed  PubMed Central  Google Scholar 

  23. Lv T, Miao YF, ** K, Han S, Xu TQ, Qiu ZL, Zhang XH. Dysregulated circular RNAs in medulloblastoma regulate proliferation and growth of tumor cells via host genes. Cancer Med. 2018;7(12):6147–57.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Gulyaeva LF, Kushlinskiy NE. Regulatory mechanisms of microRNA expression. J Transl Med. 2016;14(1):143.

    Article  PubMed  PubMed Central  Google Scholar 

  25. Wang N, Zhou Z, Liao X, Zhang T. Role of microRNAs in cardiac hypertrophy and heart failure. IUBMB Life. 2009;61(6):566–71.

    Article  CAS  PubMed  Google Scholar 

  26. Hu YB, Li CB, Song N, Zou Y, Chen SD, Ren RJ, Wang G. Diagnostic value of microRNA for Alzheimer’s disease: a systematic review and meta-analysis. Front Aging Neurosci. 2016;8:13.

    Article  PubMed  PubMed Central  Google Scholar 

  27. O’Reilly S. MicroRNAs in fibrosis: opportunities and challenges. Arthritis Res Ther. 2016;18:11.

    Article  PubMed  PubMed Central  Google Scholar 

  28. Hata A, Kashima R. Dysregulation of microRNA biogenesis machinery in cancer. Crit Rev Biochem Mol Biol. 2016;51(3):121–34.

    Article  CAS  PubMed  Google Scholar 

  29. Menon A, Abd-Aziz N, Khalid K, Poh CL, Naidu R. miRNA: a promising therapeutic target in cancer. Int J Mol Sci. 2022;23(19):11502.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Macfarlane LA, Murphy PR. MicroRNA: biogenesis, function and role in cancer. Curr Genomics. 2010;11(7):537–61.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Lee Y, Kim M, Han J, Yeom KH, Lee S, Baek SH, Kim VN. MicroRNA genes are transcribed by RNA polymerase II. EMBO J. 2004;23(20):4051–60.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Denli AM, Tops BB, Plasterk RH, Ketting RF, Hannon GJ. Processing of primary microRNAs by the Microprocessor complex. Nature. 2004;432(7014):231–5.

    Article  CAS  PubMed  Google Scholar 

  33. Bohnsack MT, Czaplinski K, Gorlich D. Exportin 5 is a RanGTP-dependent dsRNA-binding protein that mediates nuclear export of pre-miRNAs. RNA. 2004;10(2):185–91.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Park JE, Heo I, Tian Y, Simanshu DK, Chang H, Jee D, et al. Dicer recognizes the 5’ end of RNA for efficient and accurate processing. Nature. 2011;475(7355):201–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Salmena L, Poliseno L, Tay Y, Kats L, Pandolfi PP. A ceRNA hypothesis: the Rosetta Stone of a hidden RNA language? Cell. 2011;146(3):353–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Wang T, Zhang Y, He Y, Liu Y, Qi P. Screening and bioinformatics analysis of competitive endogenous RNA regulatory network –related to circular RNA in breast cancer. Biomed Res Int. 2021;2021:5575286.

    PubMed  PubMed Central  Google Scholar 

  37. Ding Y, Li M, Tayier T, Zhang M, Chen L, Feng S. Bioinformatics analysis of lncRNA-associated ceRNA network in melanoma. J Cancer. 2021;12(10):2921–32.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Guo L, Yang G, Kang Y, Li S, Duan R, Shen L, et al. Construction and analysis of a ceRNA network reveals potential prognostic markers in colorectal cancer. Front Genet. 2020;11:418.

    Article  PubMed  PubMed Central  Google Scholar 

  39. Tricco AC, Lillie E, Zarin W, O’Brien KK, Colquhoun H, Levac D, et al. PRISMA extension for sco** reviews (PRISMA-ScR): checklist and explanation. Ann Intern Med. 2018;169(7):467–73.

    Article  PubMed  Google Scholar 

  40. Zhao YN, Li K, Han XS, Pan YW. The mechanism of non-coding RNAs in medulloblastoma. Oncol Lett. 2021;22(5):758.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Zhu C, Wang X, Wang Y, Wang K. Functions and underlying mechanisms of lncRNA HOTAIR in cancer chemotherapy resistance. Cell Death Discovery. 2022;8(1):383.

    Article  PubMed  PubMed Central  Google Scholar 

  42. 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.

    Article  CAS  PubMed  Google Scholar 

  43. 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(4):3894–900.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Ge J, Wang B, Zhao S, Xu J. Inhibition of lncRNA NEAT1 sensitizes medulloblastoma cells to cisplatin through modulating the miR-23a-3p-glutaminase (GLS) axis. Bioengineered. 2022;13(3):7670–82.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Li Y, Wang X, Zhao Z, Shang J, Li G, Zhang R. LncRNA NEAT1 promotes glioma cancer progression via regulation of miR-98–5p/BZW1. 2021. Biosci Rep. https://doi.org/10.1042/BSR20200767.

  46. Wong RS. Apoptosis in cancer: from pathogenesis to treatment. J Exp Clin Cancer Res. 2011;30(1):87.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Pfeffer CM, Singh ATK. Apoptosis: a target for anticancer therapy. Int J Mol Sci. 2018;19(2):448.

    Article  PubMed  PubMed Central  Google Scholar 

  48. Jan R, Chaudhry GE. Understanding apoptosis and apoptotic pathways targeted cancer therapeutics. Adv Pharm Bull. 2019;9(2):205–18.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Schönholzer MT, Migliavacca J, Alvarez E, Santhana Kumar K, Neve A, Gries A, et al. Real-time sensing of MAPK signaling in medulloblastoma cells reveals cellular evasion mechanism counteracting dasatinib blockade of ERK activation during invasion. Neoplasia. 2020;22(10):470–83.

    Article  PubMed  PubMed Central  Google Scholar 

  50. da Cunha JM, Ghisleni EC, Cardoso PS, Siniglaglia M, Falcon T, Brunetto AT, et al. HDAC and MAPK/ERK inhibitors cooperate to reduce viability and stemness in medulloblastoma. J Mol Neurosci. 2020;70(6):981–92.

    Article  Google Scholar 

  51. Bi CL, Liu JF, Zhang MY, Lan S, Yang ZY, Fang JS. LncRNA NEAT1 promotes malignant phenotypes and TMZ resistance in glioblastoma stem cells by regulating let-7g-5p/MAP3K1 axis. 2020. Biosci Rep. https://doi.org/10.1042/BSR20201111.

  52. Laneve P, Po A, Favia A, Legnini I, Alfano V, Rea J, et al. The long noncoding RNA linc-NeD125 controls the expression of medulloblastoma driver genes by microRNA sponge activity. Oncotarget. 2017;8(19):31003–15.

    Article  PubMed  PubMed Central  Google Scholar 

  53. Ding L, Cao J, Lin W, Chen H, **ong X, Ao H, et al. The roles of cyclin-dependent kinases in cell-cycle progression and therapeutic strategies in human breast cancer. Int J Mol Sci. 2020;21(6):1960.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Bevilacqua V, Gioia U, Di Carlo V, Tortorelli AF, Colombo T, Bozzoni I, et al. Identification of linc-NeD125, a novel long non coding RNA that hosts miR-125b-1 and negatively controls proliferation of human neuroblastoma cells. RNA Biol. 2015;12(12):1323–37.

    Article  PubMed  PubMed Central  Google Scholar 

  55. Bartl J, Zanini M, Bernardi F, Forget A, Blümel L, Talbot J, et al. The HHIP-AS1 lncRNA promotes tumorigenicity through stabilization of dynein complex 1 in human SHH-driven tumors. Nat Commun. 2022;13(1):4061.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Song H, Han LM, Gao Q, Sun Y. Long non-coding RNA CRNDE promotes tumor growth in medulloblastoma. Eur Rev Med Pharmacol Sci. 2016;20(12):2588–97.

    CAS  PubMed  Google Scholar 

  57. Sun XH, Fan WJ, An ZJ, Sun Y. Inhibition of long noncoding RNA CRNDE increases chemosensitivity of medulloblastoma cells by targeting miR-29c-3p. Oncol Res. 2020;28(1):95–102.

    Article  PubMed  PubMed Central  Google Scholar 

  58. Liu C, Ren L, Deng J, Wang S. LncRNA TP73-AS1 promoted the progression of lung adenocarcinoma via PI3K/AKT pathway. 2019. Biosci Rep. https://doi.org/10.1042/BSR20180999.

  59. Varon M, Levy T, Mazor G, Ben David H, Marciano R, Krelin Y, et al. The long noncoding RNA TP73-AS1 promotes tumorigenicity of medulloblastoma cells. Int J Cancer. 2019;145(12):3402–13.

    Article  CAS  PubMed  Google Scholar 

  60. Li B, Shen M, Yao H, Chen X, **ao Z. Long noncoding RNA TP73-AS1 modulates medulloblastoma progression in vitro and in vivo by sponging miR-494-3p and targeting EIF5A2. Onco Targets Ther. 2019;12:9873–85.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Zhang Y, Wang T, Wang S, **ong Y, Zhang R, Zhang X, et al. Nkx2-2as suppression contributes to the pathogenesis of sonic hedgehog medulloblastoma. Can Res. 2018;78(4):962–73.

    Article  CAS  Google Scholar 

  62. Du WW, Yang W, Li X, Fang L, Wu N, Li F, et al. The circular RNA circSKA3 binds integrin β1 to induce invadopodium formation enhancing breast cancer invasion. Mol Ther. 2020;28(5):1287–98.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Wang X, Xu D, Pei X, Zhang Y, Zhang Y, Gu Y, Li Y. CircSKA3 modulates FOXM1 to facilitate cell proliferation, migration, and invasion while confine apoptosis in medulloblastoma via miR-383-5p. Cancer Manag Res. 2020;12:13415–26.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Zhao X, Guan J, Luo M. Circ-SKA3 upregulates ID3 expression by decoying miR-326 to accelerate the development of medulloblastoma. J Clin Neurosci. 2021;86:87–96.

    Article  CAS  PubMed  Google Scholar 

  65. Liu XC, Wang FC, Wang JH, Zhao JY, Ye SY. The circular RNA circSKA3 facilitates the malignant biological behaviors of medulloblastoma via miR-520 h/CDK6 pathway. Mol Biotechnol. 2022;64(9):1022–33.

    Article  CAS  PubMed  Google Scholar 

  66. Yin H, Zhao Y, Han X, Li Q, Dong Q, Liu Y, et al. Circ_103128 is associated with the tumorigenesis of medulloblastoma. J Cancer Res Clin Oncol. 2023;149(13):11339–49.

    Article  CAS  PubMed  Google Scholar 

  67. Gu X, Gong H, Shen L, Gu Q. MicroRNA-129-5p inhibits human glioma cell proliferation and induces cell cycle arrest by directly targeting DNMT3A. Am J Transl Res. 2018;10(9):2834–47.

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The authors appreciate all the cancer researchers working in the field of ncRNAs to develop novel treatments for patients.

Funding

This research received no specific grant from any funding agency, commercial or not-for-profit sectors.

Author information

Authors and Affiliations

Authors

Contributions

FNO: conceptualization, methodology, software, investigation, writing—original draft, MAS: conceptualization, methodology, writing—review & editing, supervision. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Mahdi Abdoli Shadbad.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Nejadi Orang, F., Abdoli Shadbad, M. CircRNA and lncRNA-associated competing endogenous RNA networks in medulloblastoma: a sco** review. Cancer Cell Int 24, 248 (2024). https://doi.org/10.1186/s12935-024-03427-w

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12935-024-03427-w

Keywords