Skip to main content

A review on the role of MEG8 lncRNA in human disorders

Abstract

Maternally expressed 8 (MEG8) is a long non-coding RNA which is expressed in the nucleus. It is highly expressed in adrenal, placenta and brain. Recent studies have shown contribution of MEG8 in different disorders ranging from neoplastic ones to diabetic nephropathy, atherosclerosis, ischemic stroke, trophoblast dysfunction and abortion, Henoch-Schonlein purpura and osteoarthritis. It has an oncogenic role in the development of lung, pancreatic and liver cancer. In the current review, we summarize the role of this lncRNA in mentioned disorders, based on the evidence obtained from in vitro, in vivo and human studies.

Introduction

Long non-coding RNAs (lncRNAs) have been largely investigated for their contribution in human disorders, particularly cancer [1]. These transcripts have sizes more than 200 nucleotides, do not possess considerable open reading frames and regulate gene expression through diverse epigenetic mechanisms. They participate in transcriptional and post-transcriptional regulation via interacting with DNA, RNA or proteins [2]. Moreover, they are involved in the regulation of mRNA splicing and can serve as precursors for microRNAs (miRNAs) [3]. Thus, lncRNAs regulate gene expression at almost all levels.

Maternally expressed 8 (MEG8), alternatively named as RNA Imprinted and Accumulated in Nucleus (Rian), is an example of which is expressed in the nucleus [4, 5]. In human, MEG8 gene resides in a cluster of imprinted genes on chromosome 14q32.3. The encoded transcript is has a preferential expression from the maternal allele in skeletal muscle, and seems to be regulated in a coordinate manner with other imprinted genes in this genomic area (https://www.ncbi.nlm.nih.gov/gene/79104). It is highly expressed in adrenal, placenta and brain [6]. This small nucleolar RNA host gene has 52 known splice variants (https://asia.ensembl.org/Homo_sapiens/Gene/Splice?db=core;g=ENSG00000225746;r=14:100894770-101038859).

Recent studies have shown contribution of MEG8 in different disorders ranging from neoplastic ones to diabetic nephropathy, atherosclerosis, ischemic stroke, trophoblast dysfunction and abortion, Henoch-Schonlein purpura and osteoarthritis. In the current review, we summarize the role of this lncRNA in mentioned disorders, based on the evidence obtained from in vitro, in vivo and human studies.

In vitro studies

Liu et al. have investigated function of MEG8 in lung cancer. For this purpose, they have transfected lung epithelial BEAS-2B cells with MEG8 overexpressing vector. Moreover, they have transfected lung cancer A549 and H1299 cells with MEG8 or miR-107 overexpressing vectors as well as knockdown plasmids. Up-regulation of MEG8 has increased proliferation, migration and invasion of lung epithelial cells. On the other hand, MEG8 knockdown or miR-107 up-regulation has blocked cell progression of lung cancer cells. Their functional studies have confirmed competitive binding of MEG8 and CDK6 with miR-107 and their function in regulation of progression of lung cancer. In addition, MEG8 knockdown or miR-107 overexpression could suppress Rb and E2F3 phosphorylation. Taken together, MEG8 could enhance progression of lung cancer through regulation of miR-107/CDK6 axis and activation of Rb/E2F3 pathway [7]. Another study in lung cancer has shown up-regulation of MEG8, parallel with down-regulation of miR-15a-5p and miR-15b-5p in cancer cell lines. MEG8 silencing has suppressed proliferation, migration, and invasion of lung cancer cells through targeting miR-15a-5p/miR-15b-5p [8].

Terashima et al. have shown induction of expression of MEG8 in the course of TGF-β-mediated epithelial-mesenchymal transition (EMT) in both lung and pancreatic cancer cells. Up-regulation of MEG8 could suppress expression of miR-34a and miR-203, leading to over-expression of SNAI1 and SNAI2 transcription factors and subsequent repression of cadherin 1/E-cadherin. Mechanistically, MEG8 interacts with EZH2 protein and increases recruitment of EZH2 to the regulatory sections of miR-34a and miR-203. EZH2 enhances histone H3 methylation in these regions and suppress transcription of these miRNA genes. Concurrent expression of MEG8 and MEG3 can increase EMT-associated alterations in cell morphology and increase motility of cells in the absence of TGF-β. Taken together, MEG8 participates in the induction of EMT through epigenetic mechanisms [9].

The effects of MEG8 silencing have also been investigated in human hemangioma endothelial cells. Notably, MEG8 silencing has suppressed proliferation of these cells and increased their apoptosis through modulation of the effects of miR-203 on JAG1 and Notch expressions [10]. Figure 1 summarizes the effect of MEG8 in the pathogenesis of lung cancer and hamangioma.

Fig. 1
figure 1

Oncogenic roles of MEG8 in lung cancer and hemangioma

Expression of MEG8 has been found to be elevated in hepatocellular carcinoma (HCC) cells. MEG8 silencing has significantly suppressed the proliferative and invasive abilities of these cells. Furthermore, MEG8 has been shown to sponge miR-367-3p to increase 14-3-3ζ levels, suppress degradation of TGFβR1, and promote TGF-β signaling [11].

Moreover, MEG8 has been shown to participate in the progression of bone-invasive pituitary adenoma through sponging miR-454-3p and increasing TNF-α expression [12]. Similarly, expression of MEG8 has been found to be elevated in Wilms tumor cells, parallel with up-regulation of CRK and down-regulation of miR-23a-3p. MEG8 silencing or miR-23a-3p up-regulation has blocked viability, migration potential and invasive properties of these cells. Mechanistically, MEG8 binds with miR-23a-3p to release CRK from inhibitory effects of this miRNA. Taken together, MEG8 regulates pathogenesis of Wilms tumor through miR-23a-3p/CRK axis [13]. Figure 2 shows the oncogenic roles of MEG8 in hepatocellular carcinoma, bone invasive pituitary adenoma and Wilms tumor.

Fig. 2
figure 2

Oncogenic roles of MEG8 in hepatocellular carcinoma, bone invasive pituitary adenoma and Wilms tumor

Exposure of podocyte cells with high-glucose conditions has led to over-expression of MEG8 and miR-770-5p. In fact, up-regulation of MEG8 increases miR-770-5p levels through decreasing methylation of the miR-770-5p gene. Up-regulation of MEG8 and miR-770-5p can increase cell apoptosis under high-glucose conditions. Taken together, MEG8 can increase miR-770-5p levels via epigenetic mechanism to induce diabetic nephropathy through enhancing cell apoptosis [14].

MEG8 can also contribute in the pathogenesis of other non-neoplastic conditions. For instance, it participate in the pathoetiology of atherosclerosis through regulation of proliferation, migration and apoptosis of vascular smooth muscle cells via affecting expression of PPARα [15]. This lncRNA can attenuate cerebral ischemia following ischemic stroke via influencing miR-130a-5p/VEGFA axis [16].

Over-expression of MEG8 in trophoblast cells has reduced proliferation and invasion of these cells, while its silencing has exerted the opposite effects. This imprinted lncRNA participates in the modulation of function of trophoblast cells during early stages [17].

MEG8 can also contribute in the pathogenesis of Henoch Schonlein purpura through sponging miR-181a-5p, influencing levels of SHP2 expression and increasing M1 macrophage polarization [18]. Finally, this lncRNA can regulate proliferation and apoptosis of chondrocytes, thus participating in the pathogenesis of osteoarthritis [19].

MEG8 has also been shown to contribute to the pathoetiology of cardiovascular diseases through epigenetic mechanisms. In vitro studies have demonstrated that MEG8 knock-down impairs angiogenic sprouting and reduces proliferation of HUVEC cells. RNA sequencing experiments have shown up-regulation of the inhibitor of angiogenesis TFPI2 after MEG8 silencing. From a mechanistical point of view, MEG8 silencing can lead to a decrease in H3K27me3 marks at the TFPI2 promoter [20]. Table 1 shows summary of in vitro studies about the role of MEG8 in human disorders.

Table 1 Summary of in vitro studies about the role of MEG8 in human disorders (∆: knock-down or deletion, VSMC: vascular smooth muscle cell, OGD: oxygen-glucose deprivation)

Animal studies

Studies in xenograft models of lung cancer have confirmed that MEG8 enhances tumor growth through modulation of miR-15a/b-5p/PSAT1. MEG8 silencing has considerably decreased tumor growth and burden in animal models of lung cancer [8]. This lncRNA has a similar effect bone-invasive pituitary adenomas [12].

Contribution of MEG8 in the pathogenesis of trophoblast dysfunction and abortion has been investigated in an animal study. Sheng et al. have obtained placental samples from pregnant female mice at three important developmental stages and assessed lncRNA signature using microarray technique. They have shown that Meg8 might have a crucial role in this process [17]. Table 2 shows summary of in vivo studies about the role of MEG8 in diverse disorders.

Table 2 Summary of in vivo studies about the role of MEG8 in diverse disorders (∆: knock-down or deletion)

Studies in clinical samples

A comprehensive assessment of transcriptome profile and clinical features has led to identification of a ceRNA network that has prognostic impact in uterine corpus endometrial carcinoma. MEG8 has been among 10 lncRNAs that have been related with prognosis of this type of cancer [22].

Expression of MEG8 has been found to be increased in tissue samples obtained from lung cancer patients compared to corresponding normal tissues. Notably, expression of this lncRNA has been negatively correlated with expression levels of miR-15a-5p and miR-15b-5p in these cancerous samples [8]. In HCC patients, over-expression of MEG8 has been correlated with the poor prognosis, edmondson Steiner grading, venous infiltration, and the number of tumor nodules [11]. In Wilms tumor samples, expression of MEG8 has been correlated with histological subtype, lymphatic invasion, and National Wilms Tumor Study (NWTS) stage [13]. On the other hand, an in silico approach in ovarian cancer has shown that the expression of MEG8 is associated with to better overall survival in Kaplan-Meier analysis. This approach has led to identification of the MEG8/miR-378d/SOBP axis as a functional axis in progression and prognosis of ovarian cancer [23]. Similar to ovarian cancer, expression of MEG8 has been found to be down-regulated in colorectal cancer samples compared with controls. This trend has also been demonstrated in the precancerous colonic lesions [24]. Moreover, Meg8 has been shown to have lower expression in neoplastic stromal cell population of giant cell tumors compared with mesenchymal stem cells [25].

Expression of MEG8 and miR-770-5p has been shown to be increased in plasma of diabetic patients, particularly in those with diabetic [14] (Fig. 3). Moreover, this lncRNA has a possible role in trophoblast dysfunction, since it is over-expressed in human spontaneous abortion villi. Moreover, methylation of its promoter region has been shown to be elevated in spontaneous abortion villus samples [17]. Table 3 shows the results of studies that reported dysregulation of MEG8 in clinical samples.

Table 3 Results of studies that reported dysregulation of MEG8 in clinical samples

Discussion

MEG8 participates in the pathoetiology of different disorders ranging from neoplastic ones to diabetic nephropathy, atherosclerosis, ischemic stroke, trophoblast dysfunction and abortion, Henoch-Schonlein purpura and osteoarthritis. It has an oncogenic role in the development of lung, pancreatic and liver cancer. However, in ovarian and colorectal cancers its expression has opposite trend.

Functional effects of MEG8 up-regulation/silencing have investigated in different contexts. This lncRNA has interactions with a variety of miRNAs such as miR-107, miR-15a-5p, miR-15b-5p, miR-34a and miR-203, miR-497-5p, miR-367-3p, miR-454-3p, miR-23a-3p, miR-770-5p, miR-181a, miR-130a-5p and miR-181a-5p. In addition to serving as molecular sponge for a number of miRNAs, MEG8 can enhance recruitment of EZH2 to the regulatory regions of miRNAs, thus regulating their expression. In fact, the regulatory role of MEG8 on expression of miR-34a and miR-203 is exerted through recruitment of EZH2 [9]. Moreover, MEG8 has been shown to increase miR-770-5p levels through decreasing methylation of the miR-770-5p gene [14]. Thus, the regulatory effects of MEG8 on miRNAs can be exerted through different mechanisms.

Although dysregulation of MEG8 has been reported in different neoplastic and non-neoplastic conditions, the diagnostic value of this lncRNA has not been investigated. Moreover, since it has different expression trends in different malignancies, future studies are needed to find the underlying mechanism of its contribution in different tumors and assess the presence of tissue-specific mechanisms.

Conclusion

A single study in patients with Temple syndrome has shown that DNA-methylation of the MEG3- and MEG8- differentially methylated region depends on the DNA-methylation pattern of the IG-differentially methylated region [27]. However, the functional link between these differentially methylated regions has not been investigated in other contexts. Thus, future studies are needed to find the mechanism of dysregulation of MEG8 in different pathological contexts.

Fig. 3
figure 3

Summarizes the effect of MEG8 lncRNA in the pathogenesis of different types of human diseases

Availability of data and materials

The analyzed data sets generated during the study are available from the corresponding author on reasonable request.

References

  1. Qian Y, Shi L, Luo Z. Long non-coding RNAs in cancer: implications for diagnosis, prognosis, and therapy. Front Med (Lausanne). 2020;7:612393.

    Article  Google Scholar 

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

    Article  CAS  Google Scholar 

  3. Jarroux J, Morillon A, Pinskaya M. History, discovery, and classification of lncRNAs. Long Non Coding RNA Biol 2017;1–46.

  4. Charlier C, Segers K, Wagenaar D, Karim L, Berghmans S, Jaillon O, et al. Human-ovine comparative sequencing of a 250-kb imprinted domain encompassing the callipyge (clpg) locus and identification of six imprinted transcripts: DLK1, DAT, GTL2, PEG11, antiPEG11, and MEG8. Genome Res. 2001;11(5):850–62 Epub 2001/05/05. eng.

    Article  CAS  Google Scholar 

  5. Hatada I, Morita S, Obata Y, Sotomaru Y, Shimoda M, Kono T. Identification of a new imprinted gene, Rian, on mouse chromosome 12 by fluorescent differential display screening. J BioChem. 2001;130(2):187–90 Epub 2001/08/02. eng.

    Article  CAS  Google Scholar 

  6. Fagerberg L, Hallström BM, Oksvold P, Kampf C, Djureinovic D, Odeberg J, et al. Analysis of the human tissue-specific expression by genome-wide integration of transcriptomics and antibody-based proteomics. Mol Cell Proteom MCP. 2014;13(2):397–406 Epub 2013/12/07. eng.

    Article  CAS  Google Scholar 

  7. Liu Y, Li L, Shang P, Song X. LncRNA MEG8 promotes tumor progression of non-small cell lung cancer via regulating miR-107/CDK6 axis. Anticancer Drugs. 2020;31(10):1065–73.

    Article  Google Scholar 

  8. Guo K, Qi D, Huang B. LncRNA MEG8 promotes NSCLC progression by modulating the miR-15a-5p-miR-15b-5p/PSAT1 axis. Cancer Cell Int. 2021;21(1):1–16.

    Article  Google Scholar 

  9. Terashima M, Ishimura A, Wanna-Udom S, Suzuki T. MEG8 long noncoding RNA contributes to epigenetic progression of the epithelial-mesenchymal transition of lung and pancreatic cancer cells. J Biol Chem. 2018;293(47):18016–30.

    Article  CAS  Google Scholar 

  10. Hu Z, Liu X, Guo J, Zhuo L, Chen Y, Yuan H. Knockdown of lncRNA MEG8 inhibits cell proliferation and invasion, but promotes cell apoptosis in hemangioma, via miR–203–induced mediation of the Notch signaling pathway. Mol Med Rep. 2021;24(6):1–8.

    Article  Google Scholar 

  11. Lou J, Yan W, Li Q-Y, Zhu A-K, Tan B-Q, Dong R, et al. LncRNA MEG8 plays an oncogenic role in hepatocellular carcinoma progression through miR-367-3p/14-3-3ζ/TGFβR1 axis. Neoplasma. 2020.

  12. Zhu H-B, Li B, Guo J, Miao Y-Z, Shen Y-T, Zhang Y-Z, et al. LncRNA MEG8 promotes TNF-α expression by sponging miR-454-3p in bone-invasive pituitary adenomas. Aging. 2021;13(10):14342.

    Article  CAS  Google Scholar 

  13. Shen J, Shu Q. Silencing of lncRNA MEG8 represses the viability, migration, and invasion of Wilms’ tumor cells through mediating miR-23a-3p/CRK axis. Urol Int 2021:1–13.

  14. Zhang J, Song L, Ma Y, Yin Y, Liu X, Luo X, et al. lncRNA MEG8 upregulates miR-770-5p through methylation and promotes cell apoptosis in diabetic nephropathy. Diabetes Metab Syndr Obes Targets Ther. 2020;13:2477.

    Article  CAS  Google Scholar 

  15. Zhang B, Dong Y, Zhao Z. LncRNA MEG8 regulates vascular smooth muscle cell proliferation, migration and apoptosis by targeting PPARα. Biochem Biophys Res Commun. 2019;510(1):171–6.

    Article  CAS  Google Scholar 

  16. Sui S, Sun L, Zhang W, Li J, Han J, Zheng J, et al. LncRNA MEG8 attenuates cerebral ischemia after ischemic stroke through targeting miR-130a-5p/VEGFA signaling. Cell Mol Neurobiol. 2021;41(6):1311–24.

    Article  CAS  Google Scholar 

  17. Sheng F, Sun N, Ji Y, Ma Y, Ding H, Zhang Q, et al. Aberrant expression of imprinted lncRNA MEG8 causes trophoblast dysfunction and abortion. J Cell Biochem. 2019;120(10):17378–90.

    Article  CAS  Google Scholar 

  18. Dai J-C, Yin M-Y, Ren M-Y. IncRNA MEG8 sponging miR-181a-5p contributes to M1 macrophage polarization by regulating SHP2 expression in Henoch Schonlein purpura rats. Ann Med. 2021;53(1):1576–88.

    Article  Google Scholar 

  19. Xie W, Jiang L, Huang X, Shang H, Gao M, You W, et al. lncRNA MEG8 is downregulated in osteoarthritis and regulates chondrocyte cell proliferation, apoptosis and inflammation. Exp Ther Med. 2021;22(4):1–8.

    Article  Google Scholar 

  20. Kremer V, Bink DI, Stanicek L, van Ingen E, Gimbel T, Hilderink S, et al. MEG8 regulates Tissue Factor Pathway Inhibitor 2 (TFPI2) expression in the endothelium. Sci Rep. 2022;12(1):843 Epub 2022/01/19. eng.

    Article  CAS  Google Scholar 

  21. Ma Q, Dai X, Lu W, Qu X, Liu N, Zhu C. Silencing long non-coding RNA MEG8 inhibits the proliferation and induces the ferroptosis of hemangioma endothelial cells by regulating miR-497-5p/NOTCH2 axis. Biochem Biophys Res Commun. 2021;556:72–8.

    Article  CAS  Google Scholar 

  22. Cai Y, Cui J, Wang Z, Wu H. Comprehensive bioinformatic analyses of lncRNA-mediated ceRNA network for uterine corpus endometrial carcinoma. Transl Cancer Res. 2022; 11(7):1994–2012 Epub 2022/08/16. eng.

    Article  CAS  Google Scholar 

  23. Lei J, He ZY, Wang J, Hu M, Zhou P, Lian CL, et al. Identification of MEG8/miR-378d/SOBP axis as a novel regulatory network and associated with immune infiltrates in ovarian carcinoma by integrated bioinformatics analysis. Cancer Med. 2021;10(8):2924–39.

    Article  CAS  Google Scholar 

  24. Kalmár A, Nagy ZB, Galamb O, Csabai I, Bodor A, Wichmann B, et al. Genome-wide expression profiling in colorectal cancer focusing on lncRNAs in the adenoma-carcinoma transition. BMC Cancer. 2019;19(1):1–16.

    Article  Google Scholar 

  25. Lehner B, Kunz P, Saehr H, Fellenberg J. Epigenetic silencing of genes and microRNAs within the imprinted Dlk1-Dio3 region at human chromosome 14.32 in giant cell tumor of bone. BMC Cancer. 2014;14(1):1–10.

    Article  Google Scholar 

  26. Zhang W, Cao D, Wang Y, Ren W. LncRNA MEG8 is upregulated in gestational diabetes mellitus (GDM) and predicted kidney injury. J Diabetes Complicat. 2021;35(1):107749.

    Article  CAS  Google Scholar 

  27. Bens S, Kolarova J, Gillessen-Kaesbach G, Buiting K, Beygo J, Caliebe A, et al. The differentially methylated region of MEG8 is hypermethylated in patients with Temple syndrome. Epigenomics. 2015;7(7):1089–97.

    Article  CAS  Google Scholar 

  28. Beygo J, Küchler A, Gillessen-Kaesbach G, Albrecht B, Eckle J, Eggermann T, et al. New insights into the imprinted MEG8-DMR in 14q32 and clinical and molecular description of novel patients with Temple syndrome. Eur J Hum Genet. 2017;25(8):935–45.

    Article  CAS  Google Scholar 

  29. He W, Sun Υ, Zhang S, Feng X, Xu M, Dai J, et al. Profiling the DNA methylation patterns of imprinted genes in abnormal semen samples by next-generation bisulfite sequencing. J Assist Reprod Genet. 2020;37(9):2211–21.

    Article  Google Scholar 

Download references

Acknowledgements

This study was financially supported by Grant from Medical School of Shahid Beheshti University of Medical Sciences.

Funding

Not applicable.

Author information

Authors and Affiliations

Authors

Contributions

SGF wrote the manuscript and revised it. MT supervised and designed the study. TK, SS and BMH collected the data and designed the figures and tables. All authors read and approved the submitted version.

Corresponding authors

Correspondence to Mohammad Taheri or Seyedpouzhia Shojaei.

Ethics declarations

Ethics approval and consent to participant

Not applicable.

Consent of publication

Not applicable.

Competing interests

The authors declare they have no conflict of interest.

Additional information

Publisher’s Note

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

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

Ghafouri-Fard, S., Khoshbakht, T., Hussen, B.M. et al. A review on the role of MEG8 lncRNA in human disorders. Cancer Cell Int 22, 285 (2022). https://doi.org/10.1186/s12935-022-02705-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12935-022-02705-9

Keywords