Abstract
N6-methyladenosine (m6A) is the most abundant modification in eukaryotic cells, and it regulates RNA transcription, processing, splicing, degradation, and translation. Long non-coding RNAs (lncRNAs), as transcriptional products with no or limited protein coding ability more than 200 nt in length, play an important role in epigenetic modification, mRNA transcription, splicing, stability, translation, and other biological functions. Extensive studies have shown that both m6A modification and lncRNAs are involved in the pathogenesis of various diseases, such as kinds of cancers, heart failure, Alzheimer’s disease, periodontitis, human abdominal aortic aneurysm, and obesity. To date, m6A modification has been identified as an important biological function in enrichment and regulation of lncRNAs. In this review, we summarize the role of m6A modification in the regulation and function of tumor-related lncRNAs. Moreover, we discuss the potential applications and possible future directions in the field.
Keywords: lncRNA, N6-methyladenosine, methylation modification, tumor, biological function
Graphical abstract

In this review, we summarize the role of m6A modification in the regulation and function of tumor-related lncRNAs. Moreover, we discuss the potential applications and possible future directions in the field.
Background
Long non-coding RNAs (lncRNAs) are novel ncRNAs longer than 200 nt and generally transcribed in the human genome.1 It is now confirmed that lncRNAs play a significant role in biological functions such as epigenetic modification, mRNA transcription, splicing, stability, and translation.2 Specifically, it has been demonstrated that lncRNA is involved in the occurrence, development, and prognosis of various diseases, including carcinomas,3, 4, 5 neuropsychiatric disorders,6,7 immune molecular mechanism,8, 9, 10 and cardiac gene programs.11 lncRNAs can interact with proteins, RNA, DNA, and can mediate their function.10 However, there are limited studies to show how lncRNAs are regulated.
Thus far, numerous studies on methylation modification in eukaryotic cells have shown that many methylation modifications can regulate RNA in eukaryotic cells, including 7-methylguanine (m7G), 5-methylcytosine (m5C), N6,2′-O-dimethyladenosine (m6Am), N1-methyladenosine (m1A), 5-hydroxymethylcytosine (5hmC), and N6-methyladenosine, among others.12 One of the most abundant modification methods in RNAs has been shown to be m6A. Studies have demonstrated that the m6A modification is dynamic and reversible, and it can influence the metabolism of mRNA and regulate RNA transcription, export, splicing, degradation, and translation.13 The m6A methylation level is related to the occurrence and progression of many diseases, such as tumors,14, 15, 16, 17 heart failure,18 Alzheimer’s disease,19 periodontitis,20 human abdominal aortic aneurysm,21 and obesity.22 However, the mechanism of how cell-specific m6A methylomes are established is poorly described.23
There are many mechanisms of both m6A modification and lncRNA that have not been elucidated. In this review, we summarize the role of m6A modification in the regulation of tumor-related lncRNAs. We also discuss the potential applications and possible future directions in this field.
m6A writers, readers, and erasers
m6A has modifications and effects on RNA through the dynamic interaction between three homologous factors, writers (methyltransferases), readers (binding proteins), and erasers (demethylases)24 (Figure 1).
Figure 1.
m6A writer complex is conducted by METTL3, METTL14, WTAP, RBM15, ZC3H13, VIRMA, and CBLL1
Additionally, METTL16 is another novel independent RNA methyltransferase. Readers such as YTHDF1–3, YTHDC1–2, IGF2BPs, HNRNPs, eIF3, FMRP, Prrc2a, METTL3, and LRPPRC could recognize the m6A writer and then have different functions on mRNA. Erasers are proteins that can regulate the process of demethylation, including FTO, ALKBH5, ALKBH9, and ALKBH10B.
Writers
An m6A writer is a protein complex with a high molecular weight of around 1 MDa,25 which consists of the following core subunits: methyltransferase-like 3 (METTL3), methyltransferase-like 14 (METTL14), and their cofactors; Wilms’ tumor 1-associating protein (WTAP); Vir like m6A methyltransferase associated (VIRMA); zinc finger CCCH-type containing 13 (ZC3H13); CBLL1 (also known as HAKAI); RNA-binding motif protein 15/15B (RBM15/15B); and the complex methylate mRNAs in a sequence context of RRACH (R = A or G; H = A, C, or U),26 often in 30 UTRs.27 Methyltransferase-like 16 (METTL16) is a novel m6A writer protein when we exclude the above-mentioned complex, m6A methyltransferase complex (MTC).28 METTL3, as a significant catalytic subunit of MTC that belongs to the class I MTase family,29 is the earliest known and key enzyme of m6A methylation modification.30 Previous studies have shown that knockdown of METTL3 directly leads to m6A decreases in mammalian embryonic stem cells (ESCs), HeLa cells, and HepG2 cells.31 Additionally, METTL3 can be detected in both the nucleus and cytoplasm,32 which means that mRNA methylation could occur in both the nucleus and cytoplasm.31 Hence, we can conclude that METTL3 plays an important role in m6A regulation. As a great adaptor needed to assist METTL3 activity, METTL14 has homology to methyltransferases.33 METTL14 could form a stable heterodimer core complex with METTL3.34 At the same time, METTL14 plays a key role in β cell survival, insulin secretion, and glucose homeostasis.35 Knockdown of METTL14 could downregulate the m6A level of X-inactive specific transcript (XIST) and augment XIST expression.36 We can regard METTL14 as the second methyltransferase enzyme. WTAP is also a pivotal component of MTC, associated with the tumor suppressor gene Wilms’ tumor 1, recruiting METTL3 and METTL14 to be localized in mRNA targets to catalyze m6A’s formation together.37 Depletion of WTAP could induce a loss of nuclear speckle localization for METTL3 and METTL14.38 VIRMA was originally known as KIAA1429, a recently identified component of MTC, which has been proven to mediate the deposition of preferential m6A in the 3′ UTR and near stop codon and is associated with selective polyadenylation (APA) in HeLa cells.39 ZC3H13 is also a novel component of MTC that stabilizes the interaction between WTAP and RBM15.40 CBLLI (HAKAI) was confirmed as an E3 ubiquitin-ligase that interacts with the tyrosine phosphorylated substrates to induce their ubiquitination and degradation.41 Just as for the other components of MTC, the downregulation of HAKAI will decrease the level of m6A and fault in embryonic development.42 RBM15 and RBM15B are paralogs and bind the MTC and recruit it to specific loci in RNAs.43 In addition to MTC, METTL16 is an independent methyltransferase that targets U6 small nuclear RNA (snRNA) and MAT2A mRNA.27
Readers
m6A readers are a type of binding proteins that can specifically decode the m6A mark and affect methylated mRNAs. In addition, different m6A readers have different functions on mRNA.44 One class of abundant m6A readers belongs to the YT521-B homology (YTH) domain family, including YTH domain family 1–3 (YTHDF1–3) and YTH domain containing 1–2 (YTHDC1–2).26 YTHDF1 was reported to enhance translational efficiency via interacting with initiation factors,45 while Liu et al.46 validated the vital oncogenic roles of YTHDF1 in tumor progression. YTHDF2 has the function to modulate mRNA instability by identifying and distributing m6A-modified mRNA as well as degrading both tumor promoter and suppressor gene mRNAs.33,47 YTHDF3 could accelerate protein synthesis with YTHDF1 and mediate the decay of methylated mRNA through YTHDF2.48 Recent research elucidated that YTHDF3 proteins could limit HIV infection of new target cells at the period of reverse transcription.49 YTHDC1 localizes to the nucleus in cultured mammalian somatic cells, while YTHDC2 is meiotic spermatocytes’ cytoplasmic.50 YTHDC1 was confirmed to modulate mRNA splice site selection in a concentration-dependent manner through minigene reporter assays.51 As the largest member of the YTH family, YTHDC2 also binds m6A preferentially within the consensus motif, and it can enhance the translation efficiency while decreasing the abundance of its target mRNAs.24 Huang et al.52 reported that insulin-like growth factor 2 mRNA-binding proteins (IGF2BPs; including IGF2BP1/2/3) are another species of m6A readers in post-transcriptional gene regulation and cancer biology. IGF2BPs have the function of stabilizing target mRNA and promoting their translation level in an m6A-dependent manner and can then affect gene expression.53 Several heterogeneous nuclear ribonucleoproteins (HNRNPs), including HNRNPC, HNRNPG, and HNRNPA2/B1, regulate alternative splicing (AS) or processing of target transcripts.26 HNRNPC could affect precursor (pre-)mRNA stability, splicing, export, and translation via binding to nascent RNA transcripts.54 Zhou et al.55 showed that HNRNPG could regulate AS through interacting with m6A-modified embryonic pre-mRNA and the phosphorylated C-terminal domain of RNA polymerase II. Moreover, through bioinformatics analysis, Wu et al.56 suggested that HNRNPA2/B1 may mediate the effects of m6A via an “m6A switch” mechanism, rather than acting as a direct m6A reader. Eukaryotic initiation factor 3 (eIF3) also serves as a reader of m6A and binds a single 5′ UTR m6A directly rather than 5′ cap to promote translation under cellular stresses.57 Fragile X mental retardation protein (FMRP) could directly bind YTHDF2 and indirectly maintain m6A-containing mRNAs instead of recruiting RNA-binding proteins (RBPs).58 Hsu et al.59 concluded that FMRP might affect the nuclear export of m6A-modified RNA targets. Recently, Wu et al.60 identified proline rich coiled-coil 2A (Prrc2a) as a novel m6A reader regulating oligodendrocytes expression. Apart from the above readers, METTL3 can act as an m6A reader as well, enhancing translation by binding a small part of cytoplasmic m6A-modified mRNA.58,61 In addition, researchers have identified leucine-rich pentatricopeptide repeat-containing protein (LRPPRC) as a potential reader.62
Erasers
m6A regulation depends on the erasers, including fat mass and obesity-associated protein (FTO) and AlkB homolog 5 (ALKBH5), to catalyze the demethylation of m6A. FTO, also known as AlkB homolog 9 (ALKBH9), was discovered as the first m6A eraser that established the dynastic and reversible m6A modification.63 FTO localizes largely in the nucleus and mediates 5%–10% of total mRNA m6A demethylation; however, FTO is also highly abundant in the cytoplasm of certain leukemia cells and can mediate up to 40% of all mRNA m6A.26 Yang et al.64 suggested that knockdown of FTO increases m6A methylation in the key pro-tumorigenic melanoma cell-intrinsic genes, which resulted in the increase of RNA decay through the m6A reader YTHDF2. Nevertheless, whether FTO recognizes and removes m6A from the internal m6A motifs is still an essential problem.65 Unlike FTO, ALKBH5 catalyzes m6A progression directly by removing the methyl group from m6A-methylated adenosine instead of oxidative demethylation.66 ALKBH5 inhibits the binding of reader protein YTHDF2 in plasmacytoma variant translocation 1 (PVT1) by downregulating the m6A modification of PVT1.67 Additionally, ALKBH5 deficiency boosts PDAC cell proliferation, migration, and invasion both in vivo and in vitro.68 ALKBH5 was also involved in glioblastoma, and it plays a key role in spermatogenesis and male fertility.69 ALKBH5 is an FTO homolog that ensures the equilibrium of m6A modification in the transcriptome.70 In addition to the two erasers, Arabidopsis is an m6A demethylase of mRNA, which affects the transformation of Arabidopsis thaliana from vegetative growth to reproductive growth and its mRNA stability.71
Characteristics, regulatory mechanisms, and biological functions of lncRNAs
lncRNAs are defined as a class of non-coding RNAs greater than 200 nt in length, which lack an open reading frame (ORF) and are unable to encode protein. lncRNAs have been detected in all species at the genomic level, including animals, plants, fungi, prokaryotes, and even viruses.72 Interestingly, only less than 2% of the genomic sequence is transcribed into mRNA,73 which suggests that the precise function of ncRNAs needs greater in-depth exploration. Many lncRNAs undergo the same RNA processing steps as mRNAs, including splicing and polyadenylation,74 while they are also transcribed by RNA polymerase II (RNA Pol II).75 Compared with protein-coding genes, the proportion of conserved specificity is apparently lower in lncRNAs.76 Similar to microRNAs (miRNAs), a type of small ncRNAs, lncRNAs have been calculated to have a high degree of tissue specificity in humans.77 Testis differs from other tissues in that it has great tissue-specific lncRNAs. Different levels of expression in different tissues suggest that specific lncRNAs can serve as biomarkers for diseases, especially cancer (Table 1).
Table 1.
lncRNA biomarkers in cancers
| Cancer | lncRNA biomarker | Expression | References |
|---|---|---|---|
| Breast cancer | HOTAIR | up | 78 |
| H19 | up | 79 | |
| LSINCT5 | up | 79 | |
| NEAT1 | up | 79 | |
| Lung cancer | SOX2-OT | up | 80 |
| ENSG0000245648 | up | 80 | |
| MALAT1 | up | 80 | |
| HOTAIR | up | 81 | |
| TUG1 | down | 81 | |
| GAS5 | down | 82 | |
| PVT1 | up | 83 | |
| Hepatocellular carcinoma | HOTAIR | up | 84 |
| UCA1 | up | 85 | |
| lncRNA-URHC | up | 86 | |
| Gastric cancer | LINC00470 | up | 87 |
| UCA1 | up | 88 | |
| H19 | up | 89 | |
| LINC00659 | up | 90 | |
| Pancreatic cancer | H19 | up | 91 |
| BX111 | up | 92 | |
| GLS-AS | down | 93 | |
| Colorectal cancer | UCA1 | up | 94 |
| MALAT1 | up | 95 | |
| PVT1 | up | 96 | |
| CRNDE | up | 97 | |
| Prostate cancer | PCA3 | up | 98 |
| HOTAIR | up | 98 | |
| DRAIC | down | 98 | |
| MALAT-1 | up | 99 | |
| Bladder cancer | UCA1 | up | 100 |
| HOTAIR | up | 101 | |
| GAS5 | up | 101 | |
| Glioma | HOXA11-AS | up | 102 |
| FOXD1-AS1 | up | 103 | |
| NEAT1 | up | 104 | |
| ANCR | up | 105 | |
| PCED1B-AS1 | up | 106 | |
| PCED1B | up | 106 |
The growing ranks of lncRNAs have attracted adequate attention on the regulatory mechanisms. Regulation of chromatin structure by histone modification and chromatin remodeling is the most classic mode. Second chromosome locus associated with prostate-1 (SChLAP1) can repress the modulation of the SWI/SNF chromatin remodeling complex to promote invasiveness in human prostate cancer.107 Additionally, lncRNAs are associated with imprinted gene clusters that can mediate the transcriptional silencing.108 The writers give an example of how Kcnq1ot1 accumulates on the chromatin of the promoter of the silenced alleles. At the same time, it also plays an inhibitory role in histone modification in mouse placenta, similar to XIST, which is representative of the field in which m6A functions.109 Zhang et al.110 concluded that UPK1A antisense RNA 1 (UPK1A-AS1) accelerated the cell cycle process and promoted the development of hepatocellular carcinoma (HCC) by interacting with EZH2 and sponging miR-138-5p. lncRNAs are also related to the activation of transcription factors to facilitate gene expression, such as lncKdm2b, and can promote ESC self-renewal via activating Zbtb3.111 Activation of Zpf292 to maintain the intestinal group 3 innate lymphoid cells (ILC3s) was also a positive effort.112 Furthermore, studies have shown that enhancer lncRNAs (eRNAs) can upregulate gene expression by interacting with the proximal promoter.113 lncRNAs also perform specific physiological functions as “miRNA sponges” to bind with miRNAs to protect mRNAs from degradation, suggesting that this posttranscriptional regulation could be a popular therapeutic target for many diseases in the future.114 For instance, muscle anabolic regulator 1 (MAR1), as a miRNA sponge, decoys miR-487b to form a complementary sequence and release inhibitory effects on Wnt5a.115 Given that m6A is the most abundant modification in mammalian RNA, it has become a research hotspot recently.
Moving forward with research on regulatory mechanisms, several scientists are focusing on the cellular physiologic functions of lncRNAs, including cellular differentiation and development.35,115,116 lncRNAs play critical roles in the disease process, such as different cancers, heart diseases, diabetes, muscle disease, and Alzheimer’s disease, among others. Since lncRNAs are involved in almost all of our lives, more attention to modulation, especially m6A, is reasonable.
Role of m6A methylation in the regulation of tumor-related lncRNAs
To date, especially in the last 5 years, studies on m6A methylation modification keep emerging, and great progress has been made. Although recent studies have shown that abnormal regulation of m6A can lead to various diseases, especially in some tumors, the role of m6A modification in tumorigenesis and tumor suppression is being gradually explored by scientists. m6A methylation could regulate almost all RNA metabolism steps by certain factors and proteins, including writers, readers, and erasers. Despite the advances made, m6A-modified ncRNAs remain to be further investigated. Our research group mainly focuses on m6A methylation modification and lncRNAs; hence, we summarize the role of m6A modification in the regulation and function of lncRNAs (Figure 2).
Figure 2.
Role of lncRNA and m6A modification in different diseases
Three major biological functions of lncRNAs are shown on the left, and three essential components of m6A modification are listed on the right.
m6A modification regulates lncRNAs
m6A modification may affect lncRNA function through a variety of regulatory mechanisms (Figure 3).
Figure 3.
m6A modification regulates lncRNA through various regulatory mechanisms, and it plays different roles in the nucleus, cytoplasm, and extracellular cells
On the one hand, m6A modification acts on the RNA-DNA triple helix structure to regulate the relationship between lncRNAs and specific DNA sites. On the other hand, m6A modification provides binding sites for readers or regulates the structure of local RNA, and then induces the binding of RBPs to regulate the function of lncRNAs.117
m6A writers could modulate lncRNAs; for instance, XIST is a target of RBM15/15B-directed methylation,72 which interacts with the m6A machinery directly via ZC3H13.118 Proteomic analysis showed that WTAP is an XIST-related protein, and RBM15/RBM15B also bind to METTL3 in a WTAP-dependent manner to act on XIST as an m6A methylase complex, thereby affecting its function.37 Both METTL16 and METTL3 are also recruited to lncRNAs, including the well-studied MALAT1 and XIST.119 Deleting the m6A domain in XIST and analysis of these genes will immediately have affects downstream, suggesting that m6A’s role in XIST is far less significant than when the cell-scope system is destroyed and the entire cellular transcription level used to analyze gene expression is analyzed.109 Xue et al.120 identified that m6A modification was installed on METTL3 to enhance the stability of the ABHD11-AS1 transcript, thereby increasing its expression, which emphasizes the function and mechanism of METTL3-induced ABHD11-AS1 in non-small cell lung cancer (NSCLC). As a critical factor sustaining m6A levels in prostate cancer cells, VIRMA downregulation reduces the stability and abundance of oncogenic lncRNAs and the invasive phenotype of prostate cancer by attenuating the overall level of m6A.121
m6A readers also have such a functional role in lncRNAs. YTHDC2 is located in the nucleus and cytoplasm and has been shown to bind to the selective m6A sites in lncRNAs.72 HNRNPC and HNRNPA2/B1 are proteins that bind to m6A sites after local and secondary structural changes of lncRNA.122 A novel lncRNA (DMDRMR) that regulates DNA methylation and cooperates with an RNA m6A reader promotes tumor growth and metastasis in clear cell renal cell carcinoma (ccRCC). Gao et al.123 elucidated that DMDRMR interacted with IGF2BP3 to regulate target genes in an m6A-dependent manner and might be a potential diagnostic, prognostic, and therapeutic target for ccRCC. As a nuclear m6A reader, YTHDC1 could regulate RNA splicing, and in gynecologic tumor cell lines, hypoxia leads to a decrease in YTHDC1 protein levels by altering splicing to produce meaninglessly mediated decay of targeted mRNA subtypes.124 At the same time, Hu et al.125 indicated that IGF2BP2 could promote pancreatic cancer cell proliferation and stemness-like properties as a m6A reader through regulating its novel target lncRNA DANCR stability. Yoneda et al.126 suggested that under the m6A modification, promoter-associated ncRNA (pncRNA)-D, an irradiation-induced 602-nt lncRNA transcribed from the promoter region of the cyclin D1 (CCND1) gene, influences the regulation of CCND1 gene expression and cell cycle progression. As an additional regulatory post-transcriptional process for gene expression, AS could be regulated by m6A modification and subsequently generate lncRNAs from one primary transcript.127 Olfr29-ps1, a lncRNA pseudogene expressed in myeloid-derived suppressor cells (MDSCs), can be regulated by the inflammatory factor interleukin-6 (IL-6), and it relies mainly on the m6A-modified Olfr29-ps1/mir-214-3-p/MyD88 regulatory pathway to regulate MDSC immune inhibition and differentiation.128 Zhu et al.129 discovered that LINC00266-1, a previously annotated lncRNA, could encode an uncharacterized peptide, named RNA-binding regulatory peptide (RBRP), which has the ability to combine IGF2BP1 and enhance its ability to recognize m6A on RNAs, and subsequently plays a carcinogenic role in tumorigenesis. The combination of METTL3/14 and WATP as a writer, ALKBH5 as an eraser, and YTHDF1 as a reader all influenced the translation of LINC00278 into YY1BM.130 Zhang et al.131 found that AKLBH5 could regulate the expression of H19, which is a lncRNA, and dexmedetomidine posttreatment could reduce the hypoxia-reoxygenation-induced senile myocardial cell injury by ALKBH5. Highly enriched m6A could enhance the RNA stability of LINC00857, and m6A-modified LINC00857 upregulation promotes tumorigenesis in pancreatic cancer by regulating the miR-150-5p/E2F3 axis.132
m6A-modified lncRNAs in tumors
The phenotype observed on teratoma assays indicated that lack of m6A could also play an important role in tumor progression; undeniably, many studies have elucidated that both m6A writers and erasers are related with cancer.124 However, most studies have focused on mRNA, and little is known about the functional correlation of RNA modification in lncRNAs. Much evidence suggests that both m6A and lncRNA play a certain role in developing and evolving tumors (Figure 4).
Figure 4.
m6A-modified lncRNAs and m6A elements that play a role in tumors in different parts of the body
Therefore, we can combine the two to explore the role and function of m6A-modified lncRNA in tumors. Next, we briefly review the relevant research results in this field in recent years.
Analysis of datasets from The Cancer Genome Atlas (TCGA) Research Network acute myeloid leukemia (AML) study showed a solid association between mutations and copy number variation of m6A regulatory genes and TP53 in AML patients. Additionally, changes in the m6A regulatory gene lead to poor survival in AML patients.124 Importantly, the treated MALAT1 transcript contains a 3′-triple helical RNA stabilization element consisting of a U-rich inner ring associated with a downstream A-rich channel to protect the MALAT1 transcript from degradation. METTL16, one novel m6A writer, recognizes and combines this triple helix, which increases the possibility that the m6A modification exists in this triple helix.133 Only a small part of MALAT1 molecules (about 2%–3%) carried the m6A modification at the other predicted sites (A2674/2684/2698) in two out of four cell lines.134 MALAT1 could also regulate the output of chimeric mRNA in an m6A-dependent manner, including YTHDC1 and METTL14, thereby controlling the differentiation of hematopoietic cells.135 Wang et al.136 further revealed that YTHDC1 recognition of MALAT1-m6A plays a key role in maintaining the composition of nuclear spots and genomic binding sites, thereby regulating the expression of several key oncogenes, and artificial tethering of YTHDC1 to m6A-deficient MALAT1 largely saved the metastatic potential of cancer cells. The Hox transcript antisense intergenic RNA (HOTAIR) is a lncRNA of about 2.2 kb that is transcribed from the antisense strand of the developmental the HOXC gene cluster on chromosome 12, and Meyer et al.137 used methylated RNA immunoprecipitation followed by sequencing (MeRIP-seq) in HEK293T cells and found a single m6A peak region (126nt) in the front half of HOTAIR, which did not overlap with m5C. Dominissini et al.138 mapped m6A to the lncRNAs, for instance, PVT1 and NEAT1 and uncharacterized lncRNA transcripts. Only a few studies have carefully mapped the locations of modified residues in a single transcript, such as m6A in lncRNA taurine upregulation 1 (TUG1).134
In addition, m6A-modified lncRNA can induce the proliferation, migration, and apoptosis of tumor cells, including pancreatic carcinoma, oophoroma, and hepatoma.139 Wu et al.140 characterized a Y-linked lncRNA, LINC00278, which is downregulated in male ESCC, and smoking could downregulate m6A modification of LINC00278 translation. Moreover, their data suggested that METTL3, METTL14, and WTAP work as writers, ALKBH5 works as an eraser, and YTHDF1 work as a reader for LINC00278 m6A modification. Sun et al.141 revealed a novel LNC942-METTL14-CXCR4/CYP1B1 signaling axis, which provides a novel target for the prevention and treatment of breast cancer (BRCA) and a mechanism of crosstalk m6A epigenetic modification. Clinically, Ni et al.16 suggested that the expression of lncRNA GAS5 in the tumor of colorectal cancer (CRC) patients is negatively correlated with the protein levels of YAP and YTHDF3. Yang et al.36 identified a “METTL14-YTHDF2-lncRNA” regulating axis in CRC cells that could mediate the degradation of XIST, significantly enhancing the proliferation and invasion ability of CRC cells in vitro and promoting the occurrence and metastasis of tumor in vivo. In clinical research, lncRNA RP11-139J23.1 is highly expressed in CRC, which is controlled by m6A methylation, Wu et al.142 elucidated that m6A-induced lncRNA RP11 could trigger the metastasis and proliferation of CRC cells by upregulation of Zeb1. Zhu et al.143 demonstrated that lncRNA KB-1980E6.3 maintains the stemness of breast cancer stem cells through the lncRNA KB-1980E6.3/IGF2BP1/c-Myc axis and suggested that disruption of this axis may provide a new therapeutic target for refractory hypoxic tumors. In addition, Hou et al.144 suggested that LINC00460 interacts with IGF2BP2 and DHX9 to promote mRNA stability of the high mobility group A1 (HMGA1), a member of non-histone chromatin protein, leading to a biological response to malignant proliferation and widespread metastasis of CRC, and HMGA1 is enhanced by METTL3, while LINC00460 relies on METTL3 to regulate HMGA1 expression. In HCC, METTL3-mediated m6A modification results in the upregulation of LINC00958 by stabilizing its mRNA, and LINC00958 sponges miR-3619-5p to increase the expression of hepatocellular carcinoma-derived growth factor, so as to promote HCC progression and lipogenesis.145 By regulating lncRNA DANCR expression, IGF2BP2 could work together with DANCR on pancreatic cancer cell proliferation and tumorigenesis.125 He et al.146 revealed that ALKBH5 could inhibit pancreatic cancer by demethylation of lncRNA KCNK15-AS1 and regulating its expression, and this new mechanism identified a potential pancreatic cancer therapeutic target. Furthermore, Chen et al.67 found that ALKBH5 can decrease the m6A modification of PVT1. Thus, the binding of YTHDF2 in PVT1 is inhibited, which promotes the proliferation of osteosarcoma cells and tumor growth. FAM225A is one of the most highly upregulated lncRNAs in nasopharyngeal carcinogenesis (NPC); Zheng et al.147 found that m6A modification in FAM225A improves its transcripts stability, which may be partly responsible for the significant upregulation of FAM225A in NPC. Ban et al.148 indicated that LNCAROD, which is overexpressed in head and neck squamous cell carcinoma (HNSCC), is stabilized by m6A methylation and promotes cancer progression via forming a ternary complex with HSPA1A and YBX1 in HNSCC. Wen et al.149 found that m6A on ncRNA NEAT1-1 plays a key role in regulating RNA Pol II Ser2 phosphorylation and may be a new specific target for the treatment and diagnosis of bone metastatic cancer, and they think that the novel complex cyclin L1/CDK19/NEAT1-1 may provide new insights into the underlying mechanism of the pathogenesis and progression of bone metastatic prostate cancer. Wang et al.150 investigated that m6A enhances the stability of RHPN1-AS1 methylated transcript by reducing RNA degradation, leading to upregulation of RHPN1-AS1 in epithelial ovarian cancer. Shen et al.151 had a conclusion that YTHDF2-mediated degradation of lncRNA FENDRR promotes cell proliferation by increasing SOX4 expression in endometrioid endometrial carcinoma.
Applications and future directions
Detailed studies of the distribution and function of chemical modifications in lncRNAs, as well as their association with related proteins, will contribute to a comprehensive understanding of the multi-layer gene expression control mechanisms active in mammalian cells.
Recent technological breakthroughs have rekindled enthusiasm for the study of m6A methylation with some ground-breaking discoveries. These allowed for scientists to designate in vivo m6A modification events and their association with disease status as functional associations. Recent investigations have emphasized that m6A modification has strong and fine-grained control over cell development programs and can facilitate appropriate, rapid, and complex responses to developmental cues.152
Many lncRNAs are gradually recognized as key factors of virus-host interaction mainly through antiviral response-dependent and antiviral response-independent approaches. In contrast, the role of lncRNA in viral infection and innate antiviral response is still unclear.19 Li and Meng9 concluded that the identification of immune-related lncRNAs may provide a new direction for the study of the molecular mechanism and treatment of low-grade glioma. Just because of the role of lncRNAs, we can deduce that m6A-modified lncRNAs can act on immunity. Nevertheless, the biological functions of m6A modification in immune-related lncRNAs are still unknown, and thus there is a need for future studies addressing lncRNAs and immunity.
Although we have made great progress in understanding the function and regulation of m6A modification, there is still much more research to be done, such as how m6A precisely regulates gene expression. The potential role of m6A and lncRNA modifications in chromatin state formation may provide additional mechanisms to explain how these modifications are involved in gene regulation during development.44 By continuously improving m6A’s detection methods, identifying more readers, writers, and erasers, and discovering the potential functions of m6A, with the joint efforts of many researchers, we will undoubtedly expand our understanding of m6A’s biological characteristics and of human health and disease. Exploring the role of m6A-modified lncRNAs in tumors will provide a broad prospect for early detection, prevention, and tumor treatment. For a long time, scholars have devoted more efforts to elucidate the downstream mechanisms of lncRNAs differentially expressed in tumors, while their upstream regulation mechanisms have not attracted much attention. For lncRNAs regulated by m6A modification, m6A modification may be one of the upstream regulatory mechanisms. For the different expressed lncRNAs regulated by m6A in tumors, we may try to intervene in the expression level of lncRNAs by using m6A inhibitors or activators except small interering RNA (siRNA) or CRISPR-CAS9 RNA editing, which may be a novel way to regulate different expressed lncRNAs to even treat tumors.
Further studies on m6A methylated lncRNAs will help us to better understand their roles in hepatic diseases or other diseases, especially in tumors.139 m6A methylated lncRNAs serve as potent prognostic biomarkers and provide useful individual treatments.
Conclusions
Two novel developed high-throughput deep-sequencing techniques, MeRIP-seq and m6A sequencing, played a key role in revealing the functional significance of m6A modification.152 With scientific technological development, increasing numbers of m6A-modified lncRNAs will be found and tested.
Additionally, the study of m6A-related proteins and their inhibitors provides new opportunities for early diagnosis, effective treatment, and even disease prognosis of cancer, especially when applied in combination with rising immunotherapies. Therefore, further molecular interactions and mechanisms need to be explored. By continuously improving detection methods, identifying more m6A readers, writers, and erasers, and discovering m6A-modified lncRNA potential functions, we will undoubtedly expand our knowledge of the contribution of m6A to human health and disease and its role of in the regulation of lncRNAs, including molecular pathways and tumorigenesis.
Acknowledgments
This work was supported by the Natural Science Foundation of Guangdong Province (2017A030308001), the National Natural Science Foundation of China (81874079 and 82073193), and by the Guangdong Provincial Clinical Medical Centre for Neurosurgery (2013B020400005).
Author contributions
Y.L. wrote the manuscript and created the figures. B.L. and H.G. reviewed and made significant revisions to the manuscript. Y.L. collected and prepared the related papers. All authors read and approved the final manuscript.
Declaration of interests
The authors declare no competing interests.
References
- 1.Xu J., Bai J., Zhang X., Lv Y., Gong Y., Liu L., Zhao H., Yu F., Ping Y., Zhang G. A comprehensive overview of lncRNA annotation resources. Brief. Bioinform. 2017;18:236–249. doi: 10.1093/bib/bbw015. [DOI] [PubMed] [Google Scholar]
- 2.Pacholewska A., Sung M.H. lncRNA expression predicts mRNA abundance. Epigenomics. 2019;11:1121–1128. doi: 10.2217/epi-2019-0003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Chen F., Li Z., Deng C., Yan H. Integration analysis for novel lncRNA markers predicting tumor recurrence in human colon adenocarcinoma. J. Transl. Med. 2019;17:299. doi: 10.1186/s12967-019-2049-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Lan T., Li H., Zhang D., Xu L., Liu H., Hao X., Yan X., Liao H., Chen X., Xie K. KIAA1429 contributes to liver cancer progression through N6-methyladenosine-dependent post-transcriptional modification of GATA3. Mol. Cancer. 2019;18:186. doi: 10.1186/s12943-019-1106-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Li R., Yang Y.E., Jin J., Zhang M.Y., Liu X., Liu X.X., Yin Y.H., Qu Y.Q. Identification of lncRNA biomarkers in lung squamous cell carcinoma using comprehensive analysis of lncRNA mediated ceRNA network. Artif. Cells Nanomed. Biotechnol. 2019;47:3246–3258. doi: 10.1080/21691401.2019.1647225. [DOI] [PubMed] [Google Scholar]
- 6.Song L., Zhang S., Duan C., Ma S., Hussain S., Wei L., Chu M. Genome-wide identification of lncRNAs as novel prognosis biomarkers of glioma. J. Cell. Biochem. 2019;120:19518–19528. doi: 10.1002/jcb.29259. [DOI] [PubMed] [Google Scholar]
- 7.Zimmer-Bensch G. Emerging roles of long non-coding RNAs as drivers of brain evolution. Cells. 2019;8:1399. doi: 10.3390/cells8111399. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Elling R., Chan J., Fitzgerald K.A. Emerging role of long noncoding RNAs as regulators of innate immune cell development and inflammatory gene expression. Eur. J. Immunol. 2016;46:504–512. doi: 10.1002/eji.201444558. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Li X., Meng Y. Survival analysis of immune-related lncRNA in low-grade glioma. BMC Cancer. 2019;19:813. doi: 10.1186/s12885-019-6032-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Robinson E.K., Covarrubias S., Carpenter S. The how and why of lncRNA function: An innate immune perspective. Biochim. Biophys. Acta Gene Regul. Mech. 2020;1863:194419. doi: 10.1016/j.bbagrm.2019.194419. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Ritter N., Ali T., Kopitchinski N., Schuster P., Beisaw A., Hendrix D.A., Schulz M.H., Müller-McNicoll M., Dimmeler S., Grote P. The lncRNA locus Handsdown regulates cardiac gene programs and is essential for early mouse development. Dev. Cell. 2019;50:644–657.e8. doi: 10.1016/j.devcel.2019.07.013. [DOI] [PubMed] [Google Scholar]
- 12.Li J., Yang X., Qi Z., Sang Y., Liu Y., Xu B., Liu W., Xu Z., Deng Y. The role of mRNA m6A methylation in the nervous system. Cell Biosci. 2019;9:66. doi: 10.1186/s13578-019-0330-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Zhang L., Hou C., Chen C., Guo Y., Yuan W., Yin D., Liu J., Sun Z. The role of N6-methyladenosine (m6A) modification in the regulation of circRNAs. Mol. Cancer. 2020;19:105. doi: 10.1186/s12943-020-01224-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Geng Y., Guan R., Hong W., Huang B., Liu P., Guo X., Hu S., Yu M., Hou B. Identification of m6A-related genes and m6A RNA methylation regulators in pancreatic cancer and their association with survival. Ann. Transl. Med. 2020;8:387. doi: 10.21037/atm.2020.03.98. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Miao W., Chen J., Jia L., Ma J., Song D. The m6A methyltransferase METTL3 promotes osteosarcoma progression by regulating the m6A level of LEF1. Biochem. Biophys. Res. Commun. 2019;516:719–725. doi: 10.1016/j.bbrc.2019.06.128. [DOI] [PubMed] [Google Scholar]
- 16.Ni W., Yao S., Zhou Y., Liu Y., Huang P., Zhou A., Liu J., Che L., Li J. Long noncoding RNA GAS5 inhibits progression of colorectal cancer by interacting with and triggering YAP phosphorylation and degradation and is negatively regulated by the m6A reader YTHDF3. Mol. Cancer. 2019;18:143. doi: 10.1186/s12943-019-1079-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Xia T., Wu X., Cao M., Zhang P., Shi G., Zhang J., Lu Z., Wu P., Cai B., Miao Y., Jiang K. The RNA m6A methyltransferase METTL3 promotes pancreatic cancer cell proliferation and invasion. Pathol. Res. Pract. 2019;215:152666. doi: 10.1016/j.prp.2019.152666. [DOI] [PubMed] [Google Scholar]
- 18.Berulava T., Buchholz E., Elerdashvili V., Pena T., Islam M.R., Lbik D., Mohamed B.A., Renner A., von Lewinski D., Sacherer M. Changes in m6A RNA methylation contribute to heart failure progression by modulating translation. Eur. J. Heart Fail. 2020;22:54–66. doi: 10.1002/ejhf.1672. [DOI] [PubMed] [Google Scholar]
- 19.Han M., Liu Z., Xu Y., Liu X., Wang D., Li F., Wang Y., Bi J. Abnormality of m6A mRNA methylation is involved in Alzheimer’s disease. Front. Neurosci. 2020;14:98. doi: 10.3389/fnins.2020.00098. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Lin W., Xu H., Wu Y., Wang J., Yuan Q. In silico genome-wide identification of m6A-associated SNPs as potential functional variants for periodontitis. J. Cell. Physiol. 2020;235:900–908. doi: 10.1002/jcp.29005. [DOI] [PubMed] [Google Scholar]
- 21.He Y., Xing J., Wang S., Xin S., Han Y., Zhang J. Increased m6A methylation level is associated with the progression of human abdominal aortic aneurysm. Ann. Transl. Med. 2019;7:797. doi: 10.21037/atm.2019.12.65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Chen J., Du B. Novel positioning from obesity to cancer: FTO, an m6A RNA demethylase, regulates tumour progression. J. Cancer Res. Clin. Oncol. 2019;145:19–29. doi: 10.1007/s00432-018-2796-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.An S., Huang W., Huang X., Cun Y., Cheng W., Sun X., Ren Z., Chen Y., Chen W., Wang J. Integrative network analysis identifies cell-specific trans regulators of m6A. Nucleic Acids Res. 2020;48:1715–1729. doi: 10.1093/nar/gkz1206. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Yang Y., Hsu P.J., Chen Y.S., Yang Y.G. Dynamic transcriptomic m6A decoration: Writers, erasers, readers and functions in RNA metabolism. Cell Res. 2018;28:616–624. doi: 10.1038/s41422-018-0040-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Reichel M., Köster T., Staiger D. Marking RNA: m6A writers, readers, and functions in Arabidopsis. J. Mol. Cell Biol. 2019;11:899–910. doi: 10.1093/jmcb/mjz085. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Shi H., Wei J., He C. Where, when, and how: Context-dependent functions of RNA methylation writers, readers, and erasers. Mol. Cell. 2019;74:640–650. doi: 10.1016/j.molcel.2019.04.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Pendleton K.E., Chen B., Liu K., Hunter O.V., Xie Y., Tu B.P., Conrad N.K. The U6 snRNA m6A methyltransferase METTL16 regulates SAM synthetase intron retention. Cell. 2017;169:824–835.e14. doi: 10.1016/j.cell.2017.05.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Warda A.S., Kretschmer J., Hackert P., Lenz C., Urlaub H., Höbartner C., Sloan K.E., Bohnsack M.T. Human METTL16 is a N6-methyladenosine (m6A) methyltransferase that targets pre-mRNAs and various non-coding RNAs. EMBO Rep. 2017;18:2004–2014. doi: 10.15252/embr.201744940. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Zheng W., Dong X., Zhao Y., Wang S., Jiang H., Zhang M., Zheng X., Gu M. Multiple functions and mechanisms underlying the role of METTL3 in human cancers. Front. Oncol. 2019;9:1403. doi: 10.3389/fonc.2019.01403. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Wang J., Yan S., Lu H., Wang S., Xu D. METTL3 attenuates LPS-induced inflammatory response in macrophages via NF-κB signaling pathway. Mediators Inflamm. 2019;2019:3120391. doi: 10.1155/2019/3120391. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Cao G., Li H.B., Yin Z., Flavell R.A. Recent advances in dynamic m6A RNA modification. Open Biol. 2016;6:160003. doi: 10.1098/rsob.160003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Chen T., Hao Y.J., Zhang Y., Li M.M., Wang M., Han W., Wu Y., Lv Y., Hao J., Wang L. m6A RNA methylation is regulated by microRNAs and promotes reprogramming to pluripotency. Cell Stem Cell. 2015;16:289–301. doi: 10.1016/j.stem.2015.01.016. [DOI] [PubMed] [Google Scholar]
- 33.Meyer K.D., Jaffrey S.R. Rethinking m6A readers, writers, and erasers. Annu. Rev. Cell Dev. Biol. 2017;33:319–342. doi: 10.1146/annurev-cellbio-100616-060758. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Liu J., Yue Y., Han D., Wang X., Fu Y., Zhang L., Jia G., Yu M., Lu Z., Deng X. A METTL3-METTL14 complex mediates mammalian nuclear RNA N6-adenosine methylation. Nat. Chem. Biol. 2014;10:93–95. doi: 10.1038/nchembio.1432. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Zhang L., Xue Z., Yan J., Wang J., Liu Q., Jiang H. lncRNA Riken-201 and Riken-203 modulates neural development by regulating the Sox6 through sequestering miRNAs. Cell Prolif. 2019;52:e12573. doi: 10.1111/cpr.12573. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Yang X., Zhang S., He C., Xue P., Zhang L., He Z., Zang L., Feng B., Sun J., Zheng M. METTL14 suppresses proliferation and metastasis of colorectal cancer by down-regulating oncogenic long non-coding RNA XIST. Mol. Cancer. 2020;19:46. doi: 10.1186/s12943-020-1146-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Chen Y., Lin Y., Shu Y., He J., Gao W. Interaction between N6-methyladenosine (m6A) modification and noncoding RNAs in cancer. Mol. Cancer. 2020;19:94. doi: 10.1186/s12943-020-01207-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Ping X.L., Sun B.F., Wang L., Xiao W., Yang X., Wang W.J., Adhikari S., Shi Y., Lv Y., Chen Y.S. Mammalian WTAP is a regulatory subunit of the RNA N6-methyladenosine methyltransferase. Cell Res. 2014;24:177–189. doi: 10.1038/cr.2014.3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Hu Y., Ouyang Z., Sui X., Qi M., Li M., He Y., Cao Y., Cao Q., Lu Q., Zhou S. Oocyte competence is maintained by m6A methyltransferase KIAA1429-mediated RNA metabolism during mouse follicular development. Cell Death Differ. 2020;27:2468–2483. doi: 10.1038/s41418-020-0516-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Knuckles P., Lence T., Haussmann I.U., Jacob D., Kreim N., Carl S.H., Masiello I., Hares T., Villaseñor R., Hess D. Zc3h13/Flacc is required for adenosine methylation by bridging the mRNA-binding factor Rbm15/Spenito to the m6A machinery component Wtap/Fl(2)d. Genes Dev. 2018;32:415–429. doi: 10.1101/gad.309146.117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Díaz-Díaz A., Roca-Lema D., Casas-Pais A., Romay G., Colombo G., Concha Á., Graña B., Figueroa A. Heat shock protein 90 chaperone regulates the E3 ubiquitin-ligase Hakai protein stability. Cancers (Basel) 2020;12:215. doi: 10.3390/cancers12010215. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Růžička K., Zhang M., Campilho A., Bodi Z., Kashif M., Saleh M., Eeckhout D., El-Showk S., Li H., Zhong S. Identification of factors required for m6 A mRNA methylation in Arabidopsis reveals a role for the conserved E3 ubiquitin ligase HAKAI. New Phytol. 2017;215:157–172. doi: 10.1111/nph.14586. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Patil D.P., Chen C.K., Pickering B.F., Chow A., Jackson C., Guttman M., Jaffrey S.R. m6A RNA methylation promotes XIST-mediated transcriptional repression. Nature. 2016;537:369–373. doi: 10.1038/nature19342. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Frye M., Harada B.T., Behm M., He C. RNA modifications modulate gene expression during development. Science. 2018;361:1346–1349. doi: 10.1126/science.aau1646. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Wang X., Zhao B.S., Roundtree I.A., Lu Z., Han D., Ma H., Weng X., Chen K., Shi H., He C. N6-methyladenosine modulates messenger RNA translation efficiency. Cell. 2015;161:1388–1399. doi: 10.1016/j.cell.2015.05.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Liu T., Wei Q., Jin J., Luo Q., Liu Y., Yang Y., Cheng C., Li L., Pi J., Si Y. The m6A reader YTHDF1 promotes ovarian cancer progression via augmenting EIF3C translation. Nucleic Acids Res. 2020;48:3816–3831. doi: 10.1093/nar/gkaa048. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Hou J., Zhang H., Liu J., Zhao Z., Wang J., Lu Z., Hu B., Zhou J., Zhao Z., Feng M. YTHDF2 reduction fuels inflammation and vascular abnormalization in hepatocellular carcinoma. Mol. Cancer. 2019;18:163. doi: 10.1186/s12943-019-1082-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Shi H., Wang X., Lu Z., Zhao B.S., Ma H., Hsu P.J., Liu C., He C. YTHDF3 facilitates translation and decay of N6-methyladenosine-modified RNA. Cell Res. 2017;27:315–328. doi: 10.1038/cr.2017.15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Jurczyszak D., Zhang W., Terry S.N., Kehrer T., Bermúdez González M.C., McGregor E., Mulder L.C.F., Eckwahl M.J., Pan T., Simon V. HIV protease cleaves the antiviral m6A reader protein YTHDF3 in the viral particle. PLoS Pathog. 2020;16:e1008305. doi: 10.1371/journal.ppat.1008305. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Kasowitz S.D., Ma J., Anderson S.J., Leu N.A., Xu Y., Gregory B.D., Schultz R.M., Wang P.J. Nuclear m6A reader YTHDC1 regulates alternative polyadenylation and splicing during mouse oocyte development. PLoS Genet. 2018;14:e1007412. doi: 10.1371/journal.pgen.1007412. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Luxton H.J., Simpson B.S., Mills I.G., Brindle N.R., Ahmed Z., Stavrinides V., Heavey S., Stamm S., Whitaker H.C. The oncogene metadherin interacts with the known splicing proteins YTHDC1, Sam68 and T-STAR and plays a novel role in alternative mRNA splicing. Cancers (Basel) 2019;11:1233. doi: 10.3390/cancers11091233. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Huang H., Weng H., Sun W., Qin X., Shi H., Wu H., Zhao B.S., Mesquita A., Liu C., Yuan C.L. Recognition of RNA N6-methyladenosine by IGF2BP proteins enhances mRNA stability and translation. Nat. Cell Biol. 2018;20:285–295. doi: 10.1038/s41556-018-0045-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Melstrom L., Chen J. RNA N6-methyladenosine modification in solid tumors: New therapeutic frontiers. Cancer Gene Ther. 2020;27:625–633. doi: 10.1038/s41417-020-0160-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Shen Y., Liu S., Fan J., Jin Y., Tian B., Zheng X., Fu H. Nuclear retention of the lncRNA SNHG1 by doxorubicin attenuates hnRNPC-p53 protein interactions. EMBO Rep. 2017;18:536–548. doi: 10.15252/embr.201643139. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Zhou K.I., Shi H., Lyu R., Wylder A.C., Matuszek Ż., Pan J.N., He C., Parisien M., Pan T. Regulation of co-transcriptional pre-mRNA splicing by m6A through the low-complexity protein hnRNPG. Mol. Cell. 2019;76:70–81.e9. doi: 10.1016/j.molcel.2019.07.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Wu B., Su S., Patil D.P., Liu H., Gan J., Jaffrey S.R., Ma J. Molecular basis for the specific and multivariant recognitions of RNA substrates by human hnRNP A2/B1. Nat. Commun. 2018;9:420. doi: 10.1038/s41467-017-02770-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Meyer K.D., Patil D.P., Zhou J., Zinoviev A., Skabkin M.A., Elemento O., Pestova T.V., Qian S.-B., Jaffrey S.R. 5′ UTR m6A promotes cap-independent translation. Cell. 2015;163:999–1010. doi: 10.1016/j.cell.2015.10.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Zaccara S., Ries R.J., Jaffrey S.R. Reading, writing and erasing mRNA methylation. Nat. Rev. Mol. Cell Biol. 2019;20:608–624. doi: 10.1038/s41580-019-0168-5. [DOI] [PubMed] [Google Scholar]
- 59.Hsu P.J., Shi H., Zhu A.C., Lu Z., Miller N., Edens B.M., Ma Y.C., He C. The RNA-binding protein FMRP facilitates the nuclear export of N6-methyladenosine-containing mRNAs. J. Biol. Chem. 2019;294:19889–19895. doi: 10.1074/jbc.AC119.010078. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Wu R., Li A., Sun B., Sun J.G., Zhang J., Zhang T., Chen Y., Xiao Y., Gao Y., Zhang Q. A novel m6A reader Prrc2a controls oligodendroglial specification and myelination. Cell Res. 2019;29:23–41. doi: 10.1038/s41422-018-0113-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Lin S., Choe J., Du P., Triboulet R., Gregory R.I. The m6A methyltransferase METTL3 promotes translation in human cancer cells. Mol. Cell. 2016;62:335–345. doi: 10.1016/j.molcel.2016.03.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Arguello A.E., DeLiberto A.N., Kleiner R.E. RNA chemical proteomics reveals the N6-methyladenosine (m6A)-regulated protein-RNA interactome. J. Am. Chem. Soc. 2017;139:17249–17252. doi: 10.1021/jacs.7b09213. [DOI] [PubMed] [Google Scholar]
- 63.Jia G., Fu Y., Zhao X., Dai Q., Zheng G., Yang Y., Yi C., Lindahl T., Pan T., Yang Y.G., He C. N6-methyladenosine in nuclear RNA is a major substrate of the obesity-associated FTO. Nat. Chem. Biol. 2011;7:885–887. doi: 10.1038/nchembio.687. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Yang S., Wei J., Cui Y.H., Park G., Shah P., Deng Y., Aplin A.E., Lu Z., Hwang S., He C., He Y.Y. m6A mRNA demethylase FTO regulates melanoma tumorigenicity and response to anti-PD-1 blockade. Nat. Commun. 2019;10:2782. doi: 10.1038/s41467-019-10669-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Li Y., Wu K., Quan W., Yu L., Chen S., Cheng C., Wu Q., Zhao S., Zhang Y., Zhou L. The dynamics of FTO binding and demethylation from the m6A motifs. RNA Biol. 2019;16:1179–1189. doi: 10.1080/15476286.2019.1621120. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Zheng G., Dahl J.A., Niu Y., Fedorcsak P., Huang C.M., Li C.J., Vågbø C.B., Shi Y., Wang W.L., Song S.H. ALKBH5 is a mammalian RNA demethylase that impacts RNA metabolism and mouse fertility. Mol. Cell. 2013;49:18–29. doi: 10.1016/j.molcel.2012.10.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Chen S., Zhou L., Wang Y. ALKBH5-mediated m6A demethylation of lncRNA PVT1 plays an oncogenic role in osteosarcoma. Cancer Cell Int. 2020;20:34. doi: 10.1186/s12935-020-1105-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Tang B., Yang Y., Kang M., Wang Y., Wang Y., Bi Y., He S., Shimamoto F. m(6)A demethylase ALKBH5 inhibits pancreatic cancer tumorigenesis by decreasing WIF-1 RNA methylation and mediating Wnt signaling. Mol. Cancer. 2020;19:3. doi: 10.1186/s12943-019-1128-6. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 69.Tang C., Klukovich R., Peng H., Wang Z., Yu T., Zhang Y., Zheng H., Klungland A., Yan W. ALKBH5-dependent m6A demethylation controls splicing and stability of long 3′-UTR mRNAs in male germ cells. Proc. Natl. Acad. Sci. USA. 2018;115:E325–E333. doi: 10.1073/pnas.1717794115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Hu B.B., Wang X.Y., Gu X.-Y., Zou C., Gao Z.J., Zhang H., Fan Y. N6-methyladenosine (m6A) RNA modification in gastrointestinal tract cancers: roles, mechanisms, and applications. Mol. Cancer. 2019;18:178. doi: 10.1186/s12943-019-1099-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Duan H.C., Wei L.H., Zhang C., Wang Y., Chen L., Lu Z., Chen P.R., He C., Jia G. ALKBH10B is an RNA N6-methyladenosine demethylase affecting arabidopsis floral transition. Plant Cell. 2017;29:2995–3011. doi: 10.1105/tpc.16.00912. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Jarroux J., Morillon A., Pinskaya M. History, discovery, and classification of lncRNAs. Adv. Exp. Med. Biol. 2017;1008:1–46. doi: 10.1007/978-981-10-5203-3_1. [DOI] [PubMed] [Google Scholar]
- 73.Wilusz J.E., Sunwoo H., Spector D.L. Long noncoding RNAs: Functional surprises from the RNA world. Genes Dev. 2009;23:1494–1504. doi: 10.1101/gad.1800909. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Nitsche A., Stadler P.F. Evolutionary clues in lncRNAs. Wiley Interdiscip. Rev. RNA. 2017;8:e1376. doi: 10.1002/wrna.1376. [DOI] [PubMed] [Google Scholar]
- 75.Guttman M., Amit I., Garber M., French C., Lin M.F., Feldser D., Huarte M., Zuk O., Carey B.W., Cassady J.P. Chromatin signature reveals over a thousand highly conserved large non-coding RNAs in mammals. Nature. 2009;458:223–227. doi: 10.1038/nature07672. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Necsulea A., Soumillon M., Warnefors M., Liechti A., Daish T., Zeller U., Baker J.C., Grützner F., Kaessmann H. The evolution of lncRNA repertoires and expression patterns in tetrapods. Nature. 2014;505:635–640. doi: 10.1038/nature12943. [DOI] [PubMed] [Google Scholar]
- 77.Cava C., Bertoli G., Castiglioni I. Portrait of tissue-specific coexpression networks of noncoding RNAs (miRNA and lncRNA) and mRNAs in normal tissues. Comput. Math. Methods Med. 2019;2019:9029351. doi: 10.1155/2019/9029351. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Xiao J., Lai H., Wei S.H., Ye Z.S., Gong F.S., Chen L.C. lncRNA HOTAIR promotes gastric cancer proliferation and metastasis via targeting miR-126 to active CXCR4 and RhoA signaling pathway. Cancer Med. 2019;8:6768–6779. doi: 10.1002/cam4.1302. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 79.Bin X., Hongjian Y., Xiping Z., Bo C., Shifeng Y., Binbin T. Research progresses in roles of lncRNA and its relationships with breast cancer. Cancer Cell Int. 2018;18:179. doi: 10.1186/s12935-018-0674-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Li Y., Yin Z., Fan J., Zhang S., Yang W. The roles of exosomal miRNAs and lncRNAs in lung diseases. Signal Transduct. Target. Ther. 2019;4:47. doi: 10.1038/s41392-019-0080-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Poulet C., Njock M.S., Moermans C., Louis E., Louis R., Malaise M., Guiot J. Exosomal long non-coding RNAs in lung diseases. Int. J. Mol. Sci. 2020;21:3580. doi: 10.3390/ijms21103580. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Li C., Lv Y., Shao C., Chen C., Zhang T., Wei Y., Fan H., Lv T., Liu H., Song Y. Tumor-derived exosomal lncRNA GAS5 as a biomarker for early-stage non-small-cell lung cancer diagnosis. J. Cell. Physiol. 2019;234:20721–20727. doi: 10.1002/jcp.28678. [DOI] [PubMed] [Google Scholar]
- 83.Huang T., Wang G., Yang L., Peng B., Wen Y., Ding G., Wang Z. Transcription factor YY1 modulates lung cancer progression by activating lncRNA-PVT1. DNA Cell Biol. 2017;36:947–958. doi: 10.1089/dna.2017.3857. [DOI] [PubMed] [Google Scholar]
- 84.Gong X., Zhu Z. Long noncoding RNA HOTAIR contributes to progression in hepatocellular carcinoma by sponging miR-217-5p. Cancer Biother. Radiopharm. 2020;35:387–396. doi: 10.1089/cbr.2019.3070. [DOI] [PubMed] [Google Scholar]
- 85.Zheng Z.K., Pang C., Yang Y., Duan Q., Zhang J., Liu W.C. Serum long noncoding RNA urothelial carcinoma-associated 1: A novel biomarker for diagnosis and prognosis of hepatocellular carcinoma. J. Int. Med. Res. 2018;46:348–356. doi: 10.1177/0300060517726441. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Li C., Yang J., Liu C., Wang X., Zhang L. Long non-coding RNAs in hepatocellular carcinoma: Ordering of the complicated lncRNA regulatory network and novel strategies for HCC clinical diagnosis and treatment. Pharmacol. Res. 2020;158:104848. doi: 10.1016/j.phrs.2020.104848. [DOI] [PubMed] [Google Scholar]
- 87.Yan J., Huang X., Zhang X., Chen Z., Ye C., Xiang W., Huang Z. lncRNA LINC00470 promotes the degradation of PTEN mRNA to facilitate malignant behavior in gastric cancer cells. Biochem. Biophys. Res. Commun. 2020;521:887–893. doi: 10.1016/j.bbrc.2019.11.016. [DOI] [PubMed] [Google Scholar]
- 88.Wang C.J., Zhu C.C., Xu J., Wang M., Zhao W.Y., Liu Q., Zhao G., Zhang Z.Z. The lncRNA UCA1 promotes proliferation, migration, immune escape and inhibits apoptosis in gastric cancer by sponging anti-tumor miRNAs. Mol. Cancer. 2019;18:115. doi: 10.1186/s12943-019-1032-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Zhou X., Yin C., Dang Y., Ye F., Zhang G. Identification of the long non-coding RNA H19 in plasma as a novel biomarker for diagnosis of gastric cancer. Sci. Rep. 2015;5:11516. doi: 10.1038/srep11516. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Sheng Y., Han C., Yang Y., Wang J., Gu Y., Li W., Guo L. Correlation between lncRNA-LINC00659 and clinical prognosis in gastric cancer and study on its biological mechanism. J. Cell. Mol. Med. 2020;24:14467–14480. doi: 10.1111/jcmm.16069. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Wang J., Zhao L., Shang K., Liu F., Che J., Li H., Cao B. Long non-coding RNA H19, a novel therapeutic target for pancreatic cancer. Mol. Med. 2020;26:30. doi: 10.1186/s10020-020-00156-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Deng S.J., Chen H.Y., Ye Z., Deng S.C., Zhu S., Zeng Z., He C., Liu M.L., Huang K., Zhong J.X. Hypoxia-induced lncRNA-BX111 promotes metastasis and progression of pancreatic cancer through regulating ZEB1 transcription. Oncogene. 2018;37:5811–5828. doi: 10.1038/s41388-018-0382-1. [DOI] [PubMed] [Google Scholar]
- 93.Deng S.J., Chen H.Y., Zeng Z., Deng S., Zhu S., Ye Z., He C., Liu M.L., Huang K., Zhong J.X. Nutrient stress-dysregulated antisense lncRNA GLS-AS impairs GLS-mediated metabolism and represses pancreatic cancer progression. Cancer Res. 2019;79:1398–1412. doi: 10.1158/0008-5472.CAN-18-0419. [DOI] [PubMed] [Google Scholar]
- 94.Luan Y., Li X., Luan Y., Zhao R., Li Y., Liu L., Hao Y., Oleg Vladimir B., Jia L. Circulating lncRNA UCA1 promotes malignancy of colorectal cancer via the miR-143/MYO6 axis. Mol. Ther. Nucleic Acids. 2020;19:790–803. doi: 10.1016/j.omtn.2019.12.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Wu C., Zhu X., Tao K., Liu W., Ruan T., Wan W., Zhang C., Zhang W. MALAT1 promotes the colorectal cancer malignancy by increasing DCP1A expression and miR203 downregulation. Mol. Carcinog. 2018;57:1421–1431. doi: 10.1002/mc.22868. [DOI] [PubMed] [Google Scholar]
- 96.Zhou D.D., Liu X.F., Lu C.W., Pant O.P., Liu X.D. Long non-coding RNA PVT1: Emerging biomarker in digestive system cancer. Cell Prolif. 2017;50:e12398. doi: 10.1111/cpr.12398. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Han P., Li J.W., Zhang B.M., Lv J.C., Li Y.M., Gu X.Y., Yu Z.W., Jia Y.H., Bai X.F., Li L. The lncRNA CRNDE promotes colorectal cancer cell proliferation and chemoresistance via miR-181a-5p-mediated regulation of Wnt/β-catenin signaling. Mol. Cancer. 2017;16:9. doi: 10.1186/s12943-017-0583-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Smolle M.A., Bauernhofer T., Pummer K., Calin G.A., Pichler M. Current insights into long non-coding RNAs (lncRNAs) in prostate cancer. Int. J. Mol. Sci. 2017;18:473. doi: 10.3390/ijms18020473. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Ren S., Liu Y., Xu W., Sun Y., Lu J., Wang F., Wei M., Shen J., Hou J., Gao X. Long noncoding RNA MALAT-1 is a new potential therapeutic target for castration resistant prostate cancer. J. Urol. 2013;190:2278–2287. doi: 10.1016/j.juro.2013.07.001. [DOI] [PubMed] [Google Scholar]
- 100.Wieczorek E., Reszka E. mRNA, microRNA and lncRNA as novel bladder tumor markers. Clin. Chim. Acta. 2018;477:141–153. doi: 10.1016/j.cca.2017.12.009. [DOI] [PubMed] [Google Scholar]
- 101.Quan J., Pan X., Zhao L., Li Z., Dai K., Yan F., Liu S., Ma H., Lai Y. lncRNA as a diagnostic and prognostic biomarker in bladder cancer: A systematic review and meta-analysis. OncoTargets Ther. 2018;11:6415–6424. doi: 10.2147/OTT.S167853. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Wang Q., Zhang J., Liu Y., Zhang W., Zhou J., Duan R., Pu P., Kang C., Han L. A novel cell cycle-associated lncRNA, HOXA11-AS, is transcribed from the 5-prime end of the HOXA transcript and is a biomarker of progression in glioma. Cancer Lett. 2016;373:251–259. doi: 10.1016/j.canlet.2016.01.039. [DOI] [PubMed] [Google Scholar]
- 103.Gao Y.F., Liu J.Y., Mao X.Y., He Z.W., Zhu T., Wang Z.B., Li X., Yin J.Y., Zhang W., Zhou H.H., Liu Z.Q. lncRNA FOXD1-AS1 acts as a potential oncogenic biomarker in glioma. CNS Neurosci. Ther. 2020;26:66–75. doi: 10.1111/cns.13152. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Wang C., Chen Y., Wang Y., Liu X., Liu Y., Li Y., Chen H., Fan C., Wu D., Yang J. Inhibition of COX-2, mPGES-1 and CYP4A by isoliquiritigenin blocks the angiogenic Akt signaling in glioma through ceRNA effect of miR-194-5p and lncRNA NEAT1. J. Exp. Clin. Cancer Res. 2019;38:371. doi: 10.1186/s13046-019-1361-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Cheng C., Dong Y., Ru X., Xia Y., Ji Y. lncRNA ANCR promotes glioma cells invasion, migration, proliferation and inhibits apoptosis via interacting with EZH2 and repressing PTEN expression. Cancer Gene Ther. 2020 doi: 10.1038/s41417-020-00263-8. Published online December 8, 2020. 10.1038/s41417-020-00263-8. [DOI] [PubMed] [Google Scholar]
- 106.Yang J., Yu D., Liu X., Changyong E., Yu S. lncRNA PCED1B-AS1 activates the proliferation and restricts the apoptosis of glioma through cooperating with miR-194-5p/PCED1B axis. J. Cell. Biochem. 2020;121:1823–1833. doi: 10.1002/jcb.29417. [DOI] [PubMed] [Google Scholar]
- 107.Atala A. Re: The long noncoding RNA SChLAP1 promotes aggressive prostate cancer and antagonizes the SWI/SNF complex. J. Urol. 2014;192:613. doi: 10.1016/j.juro.2014.05.031. [DOI] [PubMed] [Google Scholar]
- 108.Barlow D.P., Bartolomei M.S. Genomic imprinting in mammals. Cold Spring Harb. Perspect. Biol. 2014;6:a018382. doi: 10.1101/cshperspect.a018382. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Coker H., Wei G., Brockdorff N. m6A modification of non-coding RNA and the control of mammalian gene expression. Biochim. Biophys. Acta. Gene Regul. Mech. 2019;1862:310–318. doi: 10.1016/j.bbagrm.2018.12.002. [DOI] [PubMed] [Google Scholar]
- 110.Zhang D.Y., Sun Q.C., Zou X.J., Song Y., Li W.W., Guo Z.Q., Liu S.S., Liu L., Wu D.H. Long noncoding RNA UPK1A-AS1 indicates poor prognosis of hepatocellular carcinoma and promotes cell proliferation through interaction with EZH2. J. Exp. Clin. Cancer Res. 2020;39:229. doi: 10.1186/s13046-020-01748-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Ye B., Liu B., Yang L., Zhu X., Zhang D., Wu W., Zhu P., Wang Y., Wang S., Xia P. LncKdm2b controls self-renewal of embryonic stem cells via activating expression of transcription factor Zbtb3. EMBO J. 2018;37:e97174. doi: 10.15252/embj.201797174. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Liu B., Ye B., Yang L., Zhu X., Huang G., Zhu P., Du Y., Wu J., Qin X., Chen R. Long noncoding RNA lncKdm2b is required for ILC3 maintenance by initiation of Zfp292 expression. Nat. Immunol. 2017;18:499–508. doi: 10.1038/ni.3712. [DOI] [PubMed] [Google Scholar]
- 113.Ørom U.A., Derrien T., Beringer M., Gumireddy K., Gardini A., Bussotti G., Lai F., Zytnicki M., Notredame C., Huang Q. Long noncoding RNAs with enhancer-like function in human cells. Cell. 2010;143:46–58. doi: 10.1016/j.cell.2010.09.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Boon R.A., Jaé N., Holdt L., Dimmeler S. Long Noncoding RNAs: From Clinical Genetics to Therapeutic Targets? J. Am. Coll. Cardiol. 2016;67:1214–1226. doi: 10.1016/j.jacc.2015.12.051. [DOI] [PubMed] [Google Scholar]
- 115.Zhang Z.K., Li J., Guan D., Liang C., Zhuo Z., Liu J., Lu A., Zhang G., Zhang B.T. A newly identified lncRNA MAR1 acts as a miR-487b sponge to promote skeletal muscle differentiation and regeneration. J. Cachexia Sarcopenia Muscle. 2018;9:613–626. doi: 10.1002/jcsm.12281. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Ounzain S., Micheletti R., Arnan C., Plaisance I., Cecchi D., Schroen B., Reverter F., Alexanian M., Gonzales C., Ng S.Y. CARMEN, a human super enhancer-associated long noncoding RNA controlling cardiac specification, differentiation and homeostasis. J. Mol. Cell. Cardiol. 2015;89(Pt A):98–112. doi: 10.1016/j.yjmcc.2015.09.016. [DOI] [PubMed] [Google Scholar]
- 117.Ma S., Chen C., Ji X., Liu J., Zhou Q., Wang G., Yuan W., Kan Q., Sun Z. The interplay between m6A RNA methylation and noncoding RNA in cancer. J. Hematol. Oncol. 2019;12:121. doi: 10.1186/s13045-019-0805-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Dossin F., Pinheiro I., Żylicz J.J., Roensch J., Collombet S., Le Saux A., Chelmicki T., Attia M., Kapoor V., Zhan Y. SPEN integrates transcriptional and epigenetic control of X-inactivation. Nature. 2020;578:455–460. doi: 10.1038/s41586-020-1974-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Lence T., Paolantoni C., Worpenberg L., Roignant J.Y. Mechanistic insights into m6A RNA enzymes. Biochim. Biophys. Acta. Gene Regul. Mech. 2019;1862:222–229. doi: 10.1016/j.bbagrm.2018.10.014. [DOI] [PubMed] [Google Scholar]
- 120.Xue L., Li J., Lin Y., Liu D., Yang Q., Jian J., Peng J. m6A transferase METTL3-induced lncRNA ABHD11-AS1 promotes the Warburg effect of non-small-cell lung cancer. J Cell. Physiol. 2021;236:2649–2658. doi: 10.1002/jcp.30023. [DOI] [PubMed] [Google Scholar]
- 121.Barros-Silva D., Lobo J., Guimarães-Teixeira C., Carneiro I., Oliveira J., Martens-Uzunova E.S., Henrique R., Jerónimo C. VIRMA-dependent N6-methyladenosine modifications regulate the expression of long non-coding RNAs CCAT1 and CCAT2 in prostate cancer. Cancers (Basel) 2020;12:771. doi: 10.3390/cancers12040771. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Liu N., Dai Q., Zheng G., He C., Parisien M., Pan T. N6-methyladenosine-dependent RNA structural switches regulate RNA-protein interactions. Nature. 2015;518:560–564. doi: 10.1038/nature14234. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Gu Y., Niu S., Wang Y., Duan L., Pan Y., Tong Z., Zhang X., Yang Z., Peng B., Wang X. DMDRMR-mediated regulation of m6A-modified CDK4 by m6A reader IGF2BP3 drives ccRCC progression. Cancer Res. 2021;81:923–934. doi: 10.1158/0008-5472.CAN-20-1619. [DOI] [PubMed] [Google Scholar]
- 124.Batista P.J. The RNA modification N6-methyladenosine and its implications in human disease. Genomics Proteomics Bioinformatics. 2017;15:154–163. doi: 10.1016/j.gpb.2017.03.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Hu X., Peng W.X., Zhou H., Jiang J., Zhou X., Huang D., Mo Y.Y., Yang L. IGF2BP2 regulates DANCR by serving as an N6-methyladenosine reader. Cell Death Differ. 2020;27:1782–1794. doi: 10.1038/s41418-019-0461-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Yoneda R., Ueda N., Uranishi K., Hirasaki M., Kurokawa R. Long noncoding RNA pncRNA-D reduces cyclin D1 gene expression and arrests cell cycle through RNA m6A modification. J. Biol. Chem. 2020;295:5626–5639. doi: 10.1074/jbc.RA119.011556. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Zhu L.-Y., Zhu Y.R., Dai D.J., Wang X., Jin H.C. Epigenetic regulation of alternative splicing. Am. J. Cancer Res. 2018;8:2346–2358. [PMC free article] [PubMed] [Google Scholar]
- 128.Shang W., Gao Y., Tang Z., Zhang Y., Yang R. The pseudogene Olfr29-ps1 promotes the suppressive function and differentiation of monocytic MDSCs. Cancer Immunol. Res. 2019;7:813–827. doi: 10.1158/2326-6066.CIR-18-0443. [DOI] [PubMed] [Google Scholar]
- 129.Zhu S., Wang J.Z., Chen D., He Y.T., Meng N., Chen M., Lu R.X., Chen X.H., Zhang X.L., Yan G.R. An oncopeptide regulates m6A recognition by the m6A reader IGF2BP1 and tumorigenesis. Nat. Commun. 2020;11:1685. doi: 10.1038/s41467-020-15403-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Banday A.R., Papenberg B.W., Prokunina-Olsson L. When the smoke clears m6A from a Y chromosome-linked lncRNA, men get an increased risk of cancer. Cancer Res. 2020;80:2718–2719. doi: 10.1158/0008-5472.CAN-20-0961. [DOI] [PubMed] [Google Scholar]
- 131.Zhang X., Fu Q., Xu L., Yang Y., Zhao W., Zhang Y., Li H., Mi W. Dexmedetomidine postconditioning alleviates hypoxia/reoxygenation injury in senescent myocardial cells by regulating lncRNA H19 and m6A modification. Oxid. Med. Cell. Longev. 2020;2020:9250512. doi: 10.1155/2020/7059751. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Meng X., Deng Y., He S., Niu L., Zhu H. m6A-mediated upregulation of LINC00857 promotes pancreatic cancer tumorigenesis by regulating the miR-150-5p/E2F3 axis. Front. Oncol. 2021;11:629947. doi: 10.3389/fonc.2021.629947. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Jacob R., Zander S., Gutschner T. The dark side of the epitranscriptome: Chemical modifications in long non-coding RNAs. Int. J. Mol. Sci. 2017;18:2387. doi: 10.3390/ijms18112387. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Liu N., Parisien M., Dai Q., Zheng G., He C., Pan T. Probing N6-methyladenosine RNA modification status at single nucleotide resolution in mRNA and long noncoding RNA. RNA. 2013;19:1848–1856. doi: 10.1261/rna.041178.113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Chen Z.H., Chen T.Q., Zeng Z.C., Wang D., Han C., Sun Y.M., Huang W., Sun L.Y., Fang K., Chen Y.Q. Nuclear export of chimeric mRNAs depends on an lncRNA-triggered autoregulatory loop in blood malignancies. Cell Death Dis. 2020;11:566. doi: 10.1038/s41419-020-02795-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Wang X., Liu C., Zhang S., Yan H., Zhang L., Jiang A., Liu Y., Feng Y., Li D., Guo Y. N6-methyladenosine modification of MALAT1 promotes metastasis via reshaping nuclear speckles. Dev. Cell. 2021;56:702–715.e8. doi: 10.1016/j.devcel.2021.01.015. [DOI] [PubMed] [Google Scholar]
- 137.Meyer K.D., Saletore Y., Zumbo P., Elemento O., Mason C.E., Jaffrey S.R. Comprehensive analysis of mRNA methylation reveals enrichment in 3′ UTRs and near stop codons. Cell. 2012;149:1635–1646. doi: 10.1016/j.cell.2012.05.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Dominissini D., Moshitch-Moshkovitz S., Schwartz S., Salmon-Divon M., Ungar L., Osenberg S., Cesarkas K., Jacob-Hirsch J., Amariglio N., Kupiec M. Topology of the human and mouse m6A RNA methylomes revealed by m6A-seq. Nature. 2012;485:201–206. doi: 10.1038/nature11112. [DOI] [PubMed] [Google Scholar]
- 139.Xu K., Sun Y., Sheng B., Zheng Y., Wu X., Xu K. Role of identified RNA N6-methyladenosine methylation in liver. Anal. Biochem. 2019;578:45–50. doi: 10.1016/j.ab.2019.05.005. [DOI] [PubMed] [Google Scholar]
- 140.Wu S., Zhang L., Deng J., Guo B., Li F., Wang Y., Wu R., Zhang S., Lu J., Zhou Y. A novel micropeptide encoded by Y-linked LINC00278 links cigarette smoking and AR signaling in male esophageal squamous cell carcinoma. Cancer Res. 2020;80:2790–2803. doi: 10.1158/0008-5472.CAN-19-3440. [DOI] [PubMed] [Google Scholar]
- 141.Sun T., Wu Z., Wang X., Wang Y., Hu X., Qin W., Lu S., Xu D., Wu Y., Chen Q. LNC942 promoting METTL14-mediated m6A methylation in breast cancer cell proliferation and progression. Oncogene. 2020;39:5358–5372. doi: 10.1038/s41388-020-1338-9. [DOI] [PubMed] [Google Scholar]
- 142.Wu Y., Yang X., Chen Z., Tian L., Jiang G., Chen F., Li J., An P., Lu L., Luo N. m6A-induced lncRNA RP11 triggers the dissemination of colorectal cancer cells via upregulation of Zeb1. Mol. Cancer. 2019;18:87. doi: 10.1186/s12943-019-1014-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Zhu P., He F., Hou Y., Tu G., Li Q., Jin T., Zeng H., Qin Y., Wan X., Qiao Y. A novel hypoxic long noncoding RNA KB-1980E6.3 maintains breast cancer stem cell stemness via interacting with IGF2BP1 to facilitate c-Myc mRNA stability. Oncogene. 2021;40:1609–1627. doi: 10.1038/s41388-020-01638-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Hou P., Meng S., Li M., Lin T., Chu S., Li Z., Zheng J., Gu Y., Bai J. LINC00460/DHX9/IGF2BP2 complex promotes colorectal cancer proliferation and metastasis by mediating HMGA1 mRNA stability depending on m6A modification. J. Exp. Clin. Cancer Res. 2021;40:52. doi: 10.1186/s13046-021-01857-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Rong D., Dong Q., Qu H., Deng X., Gao F., Li Q., Sun P. m6A-induced LINC00958 promotes breast cancer tumorigenesis via the miR-378a-3p/YY1 axis. Cell Death Discov. 2021;7:27. doi: 10.1038/s41420-020-00382-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.He Y., Hu H., Wang Y., Yuan H., Lu Z., Wu P., Liu D., Tian L., Yin J., Jiang K., Miao Y. ALKBH5 inhibits pancreatic cancer motility by decreasing long non-coding RNA KCNK15-AS1 methylation. Cell. Physiol. Biochem. 2018;48:838–846. doi: 10.1159/000491915. [DOI] [PubMed] [Google Scholar]
- 147.Zheng Z.Q., Li Z.X., Zhou G.Q., Lin L., Zhang L.L., Lv J.W., Huang X.D., Liu R.Q., Chen F., He X.J. Long noncoding RNA FAM225A promotes nasopharyngeal carcinoma tumorigenesis and metastasis by acting as ceRNA to sponge miR-590-3p/miR-1275 and upregulate ITGB3. Cancer Res. 2019;79:4612–4626. doi: 10.1158/0008-5472.CAN-19-0799. [DOI] [PubMed] [Google Scholar]
- 148.Ban Y., Tan P., Cai J., Li J., Hu M., Zhou Y., Mei Y., Tan Y., Li X., Zeng Z. LNCAROD is stabilized by m6A methylation and promotes cancer progression via forming a ternary complex with HSPA1A and YBX1 in head and neck squamous cell carcinoma. Mol. Oncol. 2020;14:1282–1296. doi: 10.1002/1878-0261.12676. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Wen S., Wei Y., Zen C., Xiong W., Niu Y., Zhao Y. Long non-coding RNA NEAT1 promotes bone metastasis of prostate cancer through N6-methyladenosine. Mol. Cancer. 2020;19:171. doi: 10.1186/s12943-020-01293-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Wang J., Ding W., Xu Y., Tao E., Mo M., Xu W., Cai X., Chen X., Yuan J., Wu X. Long non-coding RNA RHPN1-AS1 promotes tumorigenesis and metastasis of ovarian cancer by acting as a ceRNA against miR-596 and upregulating LETM1. Aging (Albany NY) 2020;12:4558–4572. doi: 10.18632/aging.102911. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Shen J., Feng X.P., Hu R.B., Wang H., Wang Y.L., Qian J.H., Zhou Y.X. ). N-methyladenosine reader YTHDF2-mediated long noncoding RNA FENDRR degradation promotes cell proliferation in endometrioid endometrial carcinoma. Lab. Invest. 2021 doi: 10.1038/s41374-021-00543-3. Published online March 10, 2021. [DOI] [PubMed] [Google Scholar]
- 152.Maity A., Das B. N6-methyladenosine modification in mRNA: Machinery, function and implications for health and diseases. FEBS J. 2016;283:1607–1630. doi: 10.1111/febs.13614. [DOI] [PubMed] [Google Scholar]




