Open access peer-reviewed chapter

lncRNAs: Role in Regulation of Gene Expression

Written By

Pranjal Kumar and Nikita Bhandari

Submitted: 22 February 2022 Reviewed: 12 April 2022 Published: 09 June 2022

DOI: 10.5772/intechopen.104900

From the Edited Volume

Gene Expression

Edited by Fumiaki Uchiumi

Chapter metrics overview

95 Chapter Downloads

View Full Metrics

Abstract

The long non-coding RNAs (lncRNAs) are a subclass of ncRNA which is more than 200 nucleotides long and processed similar to mRNA by RNA polymerase II with very few differences between them. In the last two decades, it has become a hot topic of research as it has been found differentially expressed in disease versus normal conditions including cancers. They regulate many biological functions including regulation of gene expression and epigenetic control. lncRNAs can control gene expression at the transcriptional level, and post-transcriptional level. Also, they can play a structural role to function as scaffolds for protein complexes. They interact with DNA, RNA, and proteins. They have been shown to possess competitive binding sites for miRNAs, which makes them a master regulator of gene expression by masking miRNAs and altering many biological functions. They are found to be associated with many cellular functions including cell proliferation, migration, and invasion. The lncRNAs can be utilized as biomarkers and can be targeted for personalized therapy.

Keywords

  • ncRNA
  • lncRNA
  • miRNA sponge
  • gene expression
  • biomarker
  • targeted therapy
  • epigenetic regulation
  • etc.

1. Introduction

Ribonucleic acid (RNA) is a biological macromolecule, which serves as genetic material as well as carries the information from deoxyribonucleic acid (DNA). RNAs are of multiple types and sub-categorized as per the functions they carry out, such as messenger RNA (mRNA), transfer RNA (tRNA), ribosomal RNA (rRNA), etc. Among these, mRNA is called coding RNA as it gets translated into protein while others are considered non-coding RNAs (ncRNAs) with no coding potential. The ncRNAs have been studied since the 1950s, which were mostly limited to tRNA and rRNA. Later, microRNAs (miRNAs) were discovered and to date, it is the most studied ncRNAs. ncRNAs were further subdivided as per their sizes. The term lncRNA stands for long non-coding RNA. It has been sub-categorized as lncRNA having more than 200 nucleotides [1, 2].

The human genome consists of more than 100,000 lncRNA genes [3, 4]. They are mainly transcribed by RNA polymerase II which leads to the 5′-capping i.e. (addition of 7-methyl guanosine at the 5′-end), and polyadenylation at the 3′-end [5]. The processing of lncRNAs transcription is similar to that of mRNAs. Earlier it was thought to be junk, but recent advancements in the understanding of ncRNAs have found it as regulatory biomolecules. Although the total number of lncRNAs is always debatable with the number of functionally characterized lncRNAs. With the recent findings, it is clear that they are part of many biological processes and their regulation.

There are an increasing number of lncRNAs that are functionally characterized, but still, there is a need to have more pieces of evidence to support the recent findings. These lncRNAs are associated with many cellular functions including gene expression and regulation. lncRNAs have been shown to control many gene expressions, some of them control only neighboring gene expressions while some function at a distant position. They have structural and functional roles in the regulation of gene expression. Recent studies over the last decade show that they are part of the regulatory roles in embryonic development [6] as well as in human diseases including cancers [7, 8], heart diseases [9], etc. The lncRNAs functions have been found to be associated at the transcriptional level, translational level, and chromatin levels [10]. In this chapter, we are going to review lncRNA biology, from their biogenesis to functions mainly in gene expression and regulation.

Advertisement

2. Genomic organization of lncRNAs

There are great numbers of noncoding regions (about 98–99% of the genome sequences in human) distributed between the coding region [11, 12]. The lncRNAs’ sequences are present throughout in the genome and can be studied as different types depending upon their location in the genome. The lncRNAs can be broadly categorized in five types based on their genomic location [6]. They are stand-alone (also called as long intergenic non-coding RNA), natural antisense lncRNAs (tend to be highly enriched near promoter or terminator regions), pseudogenes (extra copy of existing genes, which are no more capable of coding), long intronic lncRNAs (synthesized from the intronic region of already annotated genes) and divergent lncRNAs (close to the transcription start site, promoter or enhancer regions). These all types of transcripts are presented in Figure 1. They are classified as different types based on their genomic positions but it is not on the basis of their function which simply help in the organizing the diverse species of lncRNAs [6].

Figure 1.

Genomic organization of IncRNAs. IncRNAs are named as per their relative location corresponding to already annotated protein coding genes. Arrow denotes the direction of IncRNA gene sequence at their Transcription Start Site. Protein Coding Genes (exons) are shown in purple boxes, various types of IncRNAs are shown in different colors. In case of pseudogenic IncRNAs, it is premature termination of transcription (yellow color TGA).

The lncRNAs are synthesized from distinct genomic location, therefore, they have been named accordingly. The lncRNAs which come from intergenic regions are called as intergenic lncRNAs, also known as stand-alone lncRNAs. Similarly, the lncRNAs which are synthesized from intronic regions of a protein coding genes are called as intronic lncRNAs. All other types are also named as per their genomic location [6].

Advertisement

3. Biogenesis and localization of lncRNAs

The lncRNAs are synthesized similarly to mRNA i.e. they are transcribed by RNA polymerase II, capped at 5′-end (m7G), poly-A tail at the 3′-end (polyadenylation), and spliced to remove introns. Figure 2a illustrates the comparison between the biogenesis of mRNAs and lncRNAs. Figure 2b (adapted from Chun-Jie Guo et al. 2020) [13] tells that a greater number of lncRNAs are found to be localized in the nucleus. The basic difference between the lncRNAs and mRNAs includes the sequence conservation and the number of exons. The lncRNAs are comparatively less conserved than mRNAs and they are composed of fewer exons [13, 14, 15].

Figure 2.

Biogenesis and Localization of IncRNAs. (a) Diagrammatic representation of IncRNA and mRNA transcription and the localization of IncRNAs in different cellular compartments for their function. (b) Some well studied IncRNAs at their respective cellular compartments (Chun-Jie Guo et al. 2020).

The expression of lncRNAs is controlled by histone modification at their promoter regions [16, 17]. Also, the phosphorylation status of RNA polymerase II defines the expression of lncRNAs. They are also transcribed by dysregulated RNA polymerase II, which leads to the accumulation of some faulty lncRNAs on chromatin which are soon degraded by ribonuclease complex known as RNA exosomes [18]. lncRNAs are found to be localized in the nucleus as well as cytoplasm. One of the reasons for their nuclear retention is somewhat associated with weak splicing signals i.e. the length of the segment between the branch point and 3′ splice site is comparatively longer than the length in mRNA [17, 19, 20]. Other factors, including splicing inhibitors [13] and alternative poly-A signals, can also regulate the localization of some lncRNAs. Some of the lncRNAs, which localize to the cytoplasm are processed similar to mRNA and transported out of the nucleus, while others with only one or very few exons are transported through the nuclear RNA export factor 1 (NXF1) [21].

Advertisement

4. Functions of lncRNAs

Although lncRNAs are being studied for the last two decades, still sufficient information needs to be gathered as compared to other non-coding RNAs. However, recent researches show that it has a role in multiple biological processes. It has been shown to function at multiple levels including regulation at the transcriptional and post-transcriptional level, structural function, and had roles at the level of genome integrity. Here, we are going to describe the role of a few well-studied lncRNAs in little detail and tabulate different lncRNAs with their known functions. The lncRNAs functions can be understood in the following ways:

4.1 Regulation at transcriptional level

To understand the regulatory role of lncRNAs at the transcription level, it is best to use the example of well characterized lncRNA, Xist, which is most studied among others. Xist is a ~ 17 kb long lncRNA, which is synthesized and expressed from X-chromosome (inactive state) and represses the gene expression through PRC1 and PRC2 [22, 23, 24, 25]. As we all are aware that the female mammals carry a pair of X chromosome, while males carry single X chromosome, therefore one of the X chromosome is inactivated in females during early developmental events to ensure the dosage compensation between the two genders and Xist plays an important role in the process of X Chromosome Inactivation (XCI) [26]. Xist helps in maintaining the 3-D conformation of the X-chromosome such that it appears to be fully compact and maintains its inactive state (Xi) i.e. heterochromatin structure [26]. When Xist gets depleted from the chromatin, the inactive state of the X-chromosome gets its active state (Xa) by the process of X chromosome reactivation (XCR) which is not completely understood. How exactly does Xist play its role is through various chromatin factors including BRG1 and cohesin get repelled from the inactive state of the X-chromosome, leading to the disruption of the topologically associated domain (TADs) and in turn preventing the formation of chromatin super loops [27, 28]. In order to make the inactive state (the higher-order heterochromatin structure) Xist accompanies PRC1, PRC2 (Polycomb Repressive Complexes), and SMCHD1 (Structural Maintenance of Chromosomes Flexible Hinge Domain Containing 1) [29]. Altogether, it regulates gene expression by recruiting epigenetic factors or functioning as a protein complex scaffold. Figure 3 represents the entire process diagrammatically.

Figure 3.

Regulatory roles of Xist. Xist inhibits the BRG1 and Cohesin, and recruits PRC1 and PRC2 to fold in compact form. Later it tethers with SMCHD1 to further compact the chromosome to form Xi. Adapted from [30].

HOTAIR, another lincRNA, which is found to work at transcriptional level. LincRNA, stands for long intergenic non-coding RNA, a sub class of long non-coding RNA (lncRNA). LincRNAs are transcribed from the intergenic regions of protein-coding genes. HOTAIR, one among many lincRNAs, is synthesized from the genomic region of HOXC gene and it is of ~2.2 kb in length. It controls the gene expression by modulating histone modification of target gene at HOXD loci [31]. The lincRNA binds to Polycomb Repressive Complex 2 (PRC2) and silence the transcription of HOXD loci [31, 32, 33]. The large number of lncRNAs are now discovered because of technological advancement and found to function as HOTAIR [10, 34, 35, 36]. It is also found to be associated with cancer and play important role in tumorigenesis, and metastasis [34].

There are many lncRNAs are listed in Table 1 that function similarly or differently at the transcriptional level and control the gene expression and cell fate. Some of the lncRNAs AIRN [80], ANCR [81, 82], ANRIL [37, 83, 84], BCAR4 [85] are nuclearly localized. AIRN functions at transcriptional level while remaining three controls the gene expression by modulating the histone modification.

FunctionlncRNAsInteracting PartnerMechanism of FunctionPatho-physiologyReferences
Regulation of TranscriptionANRILPRC1, PRC2Recruits PRC to the promoters of CDKN2A and CDKN2BCancer and other diseases[37, 38]
LINC-PINTPRC2Suppresses the gene expressionDown-regulated in many cancers[39]
lncPRESS1Sirtuin 6As decoy to regulate gene expressionESC differentiation[40]
XistPRC2, hnRNPK, YY1Inactivate gene on X-chromosomeCancer and development[41, 42, 43, 44, 45, 46]
TARIDGADD45AForms R-loopsDemethylation[47, 48]
UMLILOWDR5-MLLCXCL chemokines transcriptionTranscription of immune genes[49]
HOTTIPWDR5-MLLHOXA genesLeukaemogenesis[50, 51]
COOLAIRPRC2Histone H3 K27-me3Regulates flowering time[52, 53]
Post-transcriptional RegulationPNCTRPTBP1Inhibits splicingUpregulated in cancers[54]
PNUTSmir-205Upregulate ZEB1 & ZEB2, promote EMTEMT in Breast Cancer[55]
TINCRSTAU1RNA stability and expressionDysregulated in many cancers[56]
FAST𝛃-TrCPInhibit 𝛃-catenin degradation, activate WNT signalingPluripotency[13]
NKILAp65Inhibits NF-𝜿BSilencing of NKILA improves immune therapy[57]
Structural FunctionsNEAT1MALAT1Scaffold lncRNABreast and Skin cancers[58, 59, 60, 61, 62, 63]
MALAT1NEAT1, U1 snRNA, SR proteinSR protein phosphorylationExpressed in many cancers[58, 63, 64, 65, 66, 67, 68, 69, 70, 71]
sno-lncRNAsRBFOX2mRNA splicingPrader-Willi Syndrome[72]
Genome IntegritylincRNA-p21hnRNPKRepress p53 induced gene expressionDysregulated in many cancers[73, 74]
PANDANF-YARepress proapoptotic geneInhibits apoptosis and senescence[75]
NORADPumilio, RBMXPromote genomic stabilityDysregulated in many cancers[76, 77, 78, 79]

Table 1.

List of lncRNAs with their functions.

4.2 Regulation at post-transcriptional level

There are lncRNAs which control the gene expression at post transcriptional levels. Here, PNUTS lncRNA serve the purpose to understand the mechanism they use. PNUTS is also known as PPP1R10, which generates mRNA, but alternative splicing leads to the synthesis of PNUTS lncRNA. This lncRNA actually functions as sponge and have binding sites for mir-205, which has been shown to bind ZEB1 and ZEB2 mRNA and causes the degradation of ZEB1 and ZEB2 mRNA. ZEB1 and ZEB2 are well known transcription factors associated with epithelial-mesenchymal transition [55]. In normal condition, the level of ZEB1 and ZEB2 is regulated by mir-205, but in cancer condition, when PNUTS lncRNA level goes up, mir-205 competitively binds to the lncRNA and becomes unavailable to their target ZEB1 and ZEB2, therefore it cannot degrade ZEB1 and ZEB2 mRNA, ultimately leading to the high level of ZEB1 and ZEB2 transcription factors. In turn, EMT proceeds and cancer progresses. Figure 4 illustrates the role of PNUTS as post-transcriptional regulator of gene expression. This lncRNA does not affect the transcription process, but it does regulation of gene expression through microRNA-sponge function [55].

Figure 4.

PNUTs as miRNA sponge. Regulation of Zeb1 and Zeb2 expression at post-transcriptional level. The level of PNUTS affect the EMT process. In cancer cells, PNUTS is expressed at high level and functions as miRNA sponge to bind miR-205 which targets ZEB1 and ZEB2, and EMT proceeds. In normal cells miR-205 binds to ZEB1 and ZEB2 and degrade them, ultimately results in No EMT progression.

MALAT1 stands for Metastasis-associated lung adenocarcinoma transcript 1, also known as Nuclear Enriched Abundant Transcript 2 (NEAT2). It is ~8 kb long and transcribed from single exon [86]. As the name suggests, MALAT1 was first identified as metastasis associated lncRNA in non-small cell lung cancer (NSCLC) and was used as prognostic marker for NSCLC patient [87]. MALAT1 plays important role in splicing of mRNA. It interacts with serine-arginine splicing factor (SR protein) and governs the distribution of various splicing factors in nuclear speckle domain. It maintains the phosphorylated SR proteins level and changes in the level of MALAT1 affect the alternate splicing of endogenous mRNAs [88].

There are many other lncRNAs (See the Table 1), which work similar to PNUTS lncRNA and control the expression of different genes and control many biological processes.

4.3 Structural roles of lncRNAs

MALAT1 is so far well characterized lncRNA. The lncRNA has been shown to function at different levels including transcriptional where it facilitates the transcription factor binding to the promoters, can be part of splicing regulation, can regulate the gene expression epigenetically by interacting with PRC2 components namely EZH2, EED and SUZ12 to block miRNA or other gene expression usually through trimethylation at lysine 27 of histone H3 [89]. The lncRNA can be used as biomarkers and this can be chosen for targeted drug therapy. MALAT1 also functions as sponge for miR-1 binding and found to be engaged in the development of bone and joint diseases [90]. MALAT1 functions as miRNA sponge and captures miR-1, which is associated with Cx43 repression and OPLL (Ossification of the posterior longitudinal ligament). Figure 5 represents the explained roles of lncRNA MALAT1.

Figure 5.

Regulatory roles of MALAT1. MALAT1 (~8.7kb IcnRNA, single exon) governs the distribution of splicing factors and play an important role in alternate splicing (a). It also control the gene expression via epigenetic machinery using PRC2 (b). It regulates the transcription (c) and can effectively trap miRNAs as sponge (d).

4.4 Other roles of lncRNAs

lncRNAs have been shown to perform various function. It interacts with all major biomolecules such as DNA, RNA, and proteins and modulate chromatin remodeling, expression of neighboring (adjacent or nearby) or distant genes. It can also affect RNA splicing, RNA stability and translation [91]. It directly interacts with DNA to form R-loop or RNA DNA triplex (RNA: DNA: DNA loop). It functions to control and regulate the gene expression at chromatin level to modulate the histone modification and activation or repression of gene. It also functions as sponge to capture multiple miRNAs and ultimately governs the expression of genes [92]. It also functions in rRNA maturation in mitochondria, a lncRNA RMRP is a part of mitochondrial RNA processing endoribonuclease (MRP) and carry out the maturation of rRNA [93].

Advertisement

5. Conclusions

lncRNAs are a comparatively new class of non-coding RNA, which has been shown to execute many biological functions. The lncRNA genes can be found anywhere in the genome e.g. intronic, overlapping, anti-sense, or stand-alone. It has been shown to perform many biological functions including a regulatory role in controlling gene expression. This sub-class of ncRNAs is tissue-specific and often shows differential expression patterns in diseases including cancers and heart diseases. These lncRNAs can be utilized as biomarkers as well as for targeted therapy. They function to regulate gene expression at various levels such as transcriptional and post-transcriptional as well as structural. It also functions as a sponge for miRNAs binding and adds another way of regulating gene expression. They may be a key player in cancer progression. It needs further investigations to find its involvement in other biological functions. This will help us to move forward from considering junk to useful biomolecules. The current understanding of lncRNA biology is somewhat limited, which will be further discussed and elaborated on in the future.

Advertisement

Acknowledgments

I would like to express my sincere thanks to the Indian Institute of Technology Dharwad for providing all the necessary facilities and access to different journals. I am thankful to Paula Gavran for her help and assistance throughout the submission of this chapter.

Advertisement

Conflict of interest

The authors declare no conflict of interest.

Advertisement

Authors’ contribution

PK drafted the chapter, edited the figures as per reviewers’ comments and suggestions. NB drew the figures. The authors read and approved the chapter for submission.

References

  1. 1. Ponting CP, Oliver PL, Reik W. Evolution and functions of long noncoding RNAs. Cell. 2009;136(4):629-641
  2. 2. Derrien T, Johnson R, Bussotti G, Tanzer A, Djebali S, Tilgner H, et al. The GENCODE v7 catalog of human long noncoding RNAs: Analysis of their gene structure, evolution, and expression. Genome Research. 2012;22(9):1775-1789
  3. 3. Uszczynska-Ratajczak B, Lagarde J, Frankish A, Guigó R, Johnson R. Towards a complete map of the human long non-coding RNA transcriptome. Nature Reviews Genetics. 2018;19(9):535-548
  4. 4. Fang S, Zhang L, Guo J, Niu Y, Wu Y, Li H, et al. NONCODEV5: A comprehensive annotation database for long non-coding RNAs. Nucleic Acids Research. 2017;46(D1):D308-D314
  5. 5. Guttman M, Amit I, Garber M, French C, Lin MF, Feldser D, et al. Chromatin signature reveals over a thousand highly conserved large non-coding RNAs in mammals. Nature. 2009;458(7235):223-227
  6. 6. Kung JTY, Colognori D, Lee JT. Long noncoding RNAs: Past, present, and future. Genetics. 2013;193(3):651-669
  7. 7. Schmitt AM, Chang HY. Long noncoding RNAs: At the intersection of cancer and chromatin biology. Cold Spring Harbor Perspectives in Medicine. 2017;7(7):a026492
  8. 8. Schmitz SU, Grote P, Herrmann BG. Mechanisms of long noncoding RNA function in development and disease. Cellular and Molecular Life Sciences. 2016;73(13):2491-2509
  9. 9. Turton N, Swan R, Mahenthiralingam T, Pitts D, Dykes IM. The functions of long non-coding RNA during embryonic cardiovascular development and its potential for diagnosis and treatment of congenital heart disease. Journal of Cardiovascular Development and Disease. 2019;6(2):21
  10. 10. Bhan A, Mandal SS. LncRNA HOTAIR: A master regulator of chromatin dynamics and cancer. Biochimica et Biophysica Acta (BBA) - Reviews on Cancer. 2015;1856(1):151-164
  11. 11. International Human Genome Sequencing Consortium. Initial sequencing and analysis of the human genome. Nature. 2001;409(6822):860-921
  12. 12. Venter JC, Adams MD, Myers EW, Li PW, Mural RJ, Sutton GG, et al. The sequence of the human genome. Science. 2001;291(5507):1304-1351
  13. 13. Guo C-J, Ma X-K, Xing Y-H, Zheng C-C, Xu Y-F, Shan L, et al. Distinct processing of lncRNAs contributes to non-conserved functions in stem cells. Cell. 2020;181(3):621-636.e22
  14. 14. Quinn JJ, Zhang QC, Georgiev P, Ilik IA, Akhtar A, Chang HY. Rapid evolutionary turnover underlies conserved lncRNA–genome interactions. Genes & Development. 2016;30(2):191-207
  15. 15. Hezroni H, Koppstein D, Schwartz Matthew G, Avrutin A, Bartel David P, Ulitsky I. Principles of long noncoding RNA evolution derived from direct comparison of transcriptomes in 17 species. Cell Reports. 2015;11(7):1110-1122
  16. 16. Lagarde J, Uszczynska-Ratajczak B, Carbonell S, Pérez-Lluch S, Abad A, Davis C, et al. High-throughput annotation of full-length long noncoding RNAs with capture long-read sequencing. Nature Genetics. 2017;49(12):1731-1740
  17. 17. Melé M, Mattioli K, Mallard W, Shechner DM, Gerhardinger C, Rinn JL. Chromatin environment, transcriptional regulation, and splicing distinguish lincRNAs and mRNAs. Genome Research. 2016;27(1):27-37
  18. 18. Schlackow M, Nojima T, Gomes T, Dhir A, Carmo-Fonseca M, Proudfoot NJ. Distinctive patterns of transcription and RNA processing for human lincRNAs. Molecular Cell. 2017;65(1):25-38
  19. 19. Rosenberg Alexander B, Patwardhan Rupali P, Shendure J, Seelig G. Learning the sequence determinants of alternative splicing from millions of random sequences. Cell. 2015;163(3):698-711
  20. 20. Zuckerman B, Ulitsky I. Predictive models of subcellular localization of long RNAs. RNA. 2019;25(5):557-572
  21. 21. Zuckerman B, Ron M, Mikl M, Segal E, Ulitsky I. Gene architecture and sequence composition underpin selective dependency of nuclear export of long RNAs on NXF1 and the TREX complex. Molecular Cell. 2020;79(2):251-267.e6
  22. 22. Brown CJ, Hendrich BD, Rupert JL, Lafrenière RG, Xing Y, Lawrence J, et al. The human XIST gene: Analysis of a 17 kb inactive X-specific RNA that contains conserved repeats and is highly localized within the nucleus. Cell. 1992;71(3):527-542
  23. 23. Wang J, Mager J, Chen Y, Schneider E, Cross JC, Nagy A, et al. Imprinted X inactivation maintained by a mouse Polycomb group gene. Nature Genetics. 2001;28(4):371-375
  24. 24. Plath K, Talbot D, Hamer KM, Otte AP, Yang TP, Jaenisch R, et al. Developmentally regulated alterations in Polycomb repressive complex 1 proteins on the inactive X chromosome. Journal of Cell Biology. 2004;167(6):1025-1035
  25. 25. Zhao J, Sun BK, Erwin JA, Song J-J, Lee JT. Polycomb proteins targeted by a short repeat RNA to the mouse X chromosome. Science. 2008;322(5902):750-756
  26. 26. Dixon-McDougall T, Brown C. The making of a Barr body: The mosaic of factors that eXIST on the mammalian inactive X chromosome. Biochemistry and Cell Biology. 2016;94(1):56-70
  27. 27. Minajigi A, Froberg JE, Wei C, Sunwoo H, Kesner B, Colognori D, et al. A comprehensive Xist interactome reveals cohesin repulsion and an RNA-directed chromosome conformation. Science. 2015;349(6245)
  28. 28. Jégu T, Blum R, Cochrane JC, Yang L, Wang C-Y, Gilles M-E, et al. Xist RNA antagonizes the SWI/SNF chromatin remodeler BRG1 on the inactive X chromosome. Nature Structural & Molecular Biology. 2019;26(2):96-109
  29. 29. Wang C-Y, Jégu T, Chu H-P, Oh HJ, Lee JT. SMCHD1 merges chromosome compartments and assists formation of super-structures on the inactive X. Cell. 2018;174(2):406-421.e25
  30. 30. Guh C-Y, Hsieh Y-H, Chu H-P. Functions and properties of nuclear lncRNAs—From systematically mapping the interactomes of lncRNAs. Journal of Biomedical Science. 2020;27(1):44
  31. 31. Rinn JL, Kertesz M, Wang JK, Squazzo SL, Xu X, Brugmann SA, et al. Functional demarcation of active and silent chromatin domains in human HOX loci by noncoding RNAs. Cell. 2007;129(7):1311-1323
  32. 32. Kopp F, Mendell JT. Functional classification and experimental dissection of long noncoding RNAs. Cell. 2018;172(3):393-407
  33. 33. Portoso M, Ragazzini R, Brenčič Ž, Moiani A, Michaud A, Vassilev I, et al. PRC2 is dispensable for HOTAIR-mediated transcriptional repression. The EMBO Journal. 2017;36(8):981-994
  34. 34. Tang Q , Hann S. HOTAIR: An oncogenic long non-coding RNA in human cancer. Cellular Physiology and Biochemistry. 2018;47(3):893-913
  35. 35. Zhou X, Chen J, Tang W. The molecular mechanism of HOTAIR in tumorigenesis, metastasis, and drug resistance. Acta Biochimica et Biophysica Sinica. 2014;46(12):1011-1015
  36. 36. Wu Y, Zhang L, Wang Y, Li H, Ren X, Wei F, et al. Long noncoding RNA HOTAIR involvement in cancer. Tumor Biology. 2014;35(10):9531-9538
  37. 37. Yap KL, Li S, Muñoz-Cabello AM, Raguz S, Zeng L, Mujtaba S, et al. Molecular interplay of the noncoding RNA ANRIL and methylated histone H3 lysine 27 by Polycomb CBX7 in transcriptional silencing of INK4a. Molecular Cell. 2010;38(5):662-674
  38. 38. Holdt LM, Hoffmann S, Sass K, Langenberger D, Scholz M, Krohn K, et al. Alu elements in ANRIL non-coding RNA at chromosome 9p21 modulate Atherogenic cell functions through trans-regulation of gene networks. McCarthy MI, editor. PLoS Genetics. 2013;9(7):e1003588
  39. 39. Marín-Béjar O, Mas AM, González J, Martinez D, Athie A, Morales X, et al. The human lncRNA LINC-PINT inhibits tumor cell invasion through a highly conserved sequence element. Genome Biology. 2017;18(1):202
  40. 40. Jain AK, Xi Y, McCarthy R, Allton K, Akdemir KC, Patel LR, et al. LncPRESS1 is a p53-regulated LncRNA that safeguards pluripotency by disrupting SIRT6-mediated De-acetylation of histone H3K56. Molecular Cell. 2016;64(5):967-981
  41. 41. Schertzer MD, Braceros KCA, Starmer J, Cherney RE, Lee DM, Salazar G, et al. lncRNA-induced spread of Polycomb controlled by genome architecture, RNA abundance, and CpG Island DNA. Molecular Cell. 2019;75(3):523-537.e10
  42. 42. Lee Jeannie T, Bartolomei MS. X-inactivation, imprinting, and long noncoding RNAs in health and disease. Cell. 2013;152(6):1308-1323
  43. 43. Pintacuda G, Wei G, Roustan C, Kirmizitas BA, Solcan N, Cerase A, et al. hnRNPK recruits PCGF3/5-PRC1 to the Xist RNA B-repeat to establish Polycomb-mediated chromosomal silencing. Molecular Cell. 2017;68(5):955-969.e10
  44. 44. Colognori D, Sunwoo H, Kriz AJ, Wang C-Y, Lee JT. Xist Deletional analysis reveals an interdependency between Xist RNA and Polycomb complexes for spreading along the inactive X. Molecular Cell. 2019;74(1):101-117.e10
  45. 45. McHugh CA, Chen C-K, Chow A, Surka CF, Tran C, McDonel P, et al. The Xist lncRNA interacts directly with SHARP to silence transcription through HDAC3. Nature. 2015;521(7551):232-236
  46. 46. Jeon Y, Lee JT. YY1 tethers Xist RNA to the inactive X nucleation center. Cell. 2011;146(1):119-133
  47. 47. Arab K, Karaulanov E, Musheev M, Trnka P, Schäfer A, Grummt I, et al. GADD45A binds R-loops and recruits TET1 to CpG island promoters. Nature Genetics. 2019;51(2):217-223
  48. 48. Arab K, Park YJ, Lindroth AM, Schäfer A, Oakes C, Weichenhan D, et al. Long noncoding RNA TARID directs demethylation and activation of the tumor suppressor TCF21 via GADD45A. Molecular Cell. 2014;55(4):604-614
  49. 49. Fanucchi S, Fok ET, Dalla E, Shibayama Y, Börner K, Chang EY, et al. Immune genes are primed for robust transcription by proximal long noncoding RNAs located in nuclear compartments. Nature Genetics. 2018;51(1):138-150
  50. 50. Wang KC, Yang YW, Liu B, Sanyal A, Corces-Zimmerman R, Chen Y, et al. A long noncoding RNA maintains active chromatin to coordinate homeotic gene expression. Nature. 2011;472(7341):120-124
  51. 51. Luo H, Zhu G, Xu J, Lai Q , Yan B, Guo Y, et al. HOTTIP lncRNA promotes hematopoietic stem cell self-renewal leading to AML-like disease in mice. Cancer Cell. 2019;36(6):645-659.e8
  52. 52. Rosa S, Duncan S, Dean C. Mutually exclusive sense–antisense transcription at FLC facilitates environmentally induced gene repression. Nature Communications. 2016;7(1):13031
  53. 53. Csorba T, Questa JI, Sun Q , Dean C. Antisense COOLAIR mediates the coordinated switching of chromatin states at FLC during vernalization. Proceedings of the National Academy of Sciences. 2014;111(45):16160-16165
  54. 54. Yap K, Mukhina S, Zhang G, Tan JSC, Ong HS, Makeyev EV. A short tandem repeat-enriched RNA assembles a nuclear compartment to control alternative splicing and promote cell survival. Molecular Cell. 2018, 72;(3):525-540.e13
  55. 55. Grelet S, Link LA, Howley B, Obellianne C, Palanisamy V, Gangaraju VK, et al. A regulated PNUTS mRNA to lncRNA splice switch mediates EMT and tumour progression. Nature Cell Biology. 2017;19(9):1105-1115
  56. 56. Kretz M, Siprashvili Z, Chu C, Webster DE, Zehnder A, Qu K, et al. Control of somatic tissue differentiation by the long non-coding RNA TINCR. Nature. 2012;493(7431):231-235
  57. 57. Huang D, Chen J, Yang L, Ouyang Q , Li J, Lao L, et al. NKILA lncRNA promotes tumor immune evasion by sensitizing T cells to activation-induced cell death. Nature Immunology. 2018;19(10):1112-1125
  58. 58. Hutchinson JN, Ensminger AW, Clemson CM, Lynch CR, Lawrence JB, Chess A. A screen for nuclear transcripts identifies two linked noncoding RNAs associated with SC35 splicing domains. BMC Genomics. 2007;8:39
  59. 59. Sasaki YTF, Ideue T, Sano M, Mituyama T, Hirose T. MENε/β noncoding RNAs are essential for structural integrity of nuclear paraspeckles. Proceedings of the National Academy of Sciences. 2009;106(8):2525-2530
  60. 60. Sunwoo H, Dinger ME, Wilusz JE, Amaral PP, Mattick JS, Spector DL. MEN ε/β nuclear-retained non-coding RNAs are up-regulated upon muscle differentiation and are essential components of paraspeckles. Genome Research. 2008;19(3):347-359
  61. 61. Yamazaki T, Souquere S, Chujo T, Kobelke S, Chong YS, Fox AH, et al. Functional domains of NEAT1 architectural lncRNA induce Paraspeckle assembly through phase separation. Molecular Cell. 2018;70(6):1038-1053.e7
  62. 62. Lin Y, Schmidt BF, Bruchez MP, McManus C. Structural analyses of NEAT1 lncRNAs suggest long-range RNA interactions that may contribute to paraspeckle architecture. Nucleic Acids Research. 2018;46(7):3742-3752
  63. 63. Lu Z, Zhang Q , Lee B, Flynn Ryan A, Smith Martin A, Robinson James T, et al. RNA duplex map in living cells reveals higher-order transcriptome structure. Cell. 2016;165(5):1267-1279
  64. 64. Wilusz JE, JnBaptiste CK, Lu LY, Kuhn C-D, Joshua-Tor L, Sharp PA. A triple helix stabilizes the 3′ ends of long noncoding RNAs that lack poly(a) tails. Genes & Development. 2012;26(21):2392-2407
  65. 65. Tripathi V, Song DY, Zong X, Shevtsov SP, Hearn S, Fu X-D, et al. SRSF1 regulates the assembly of pre-mRNA processing factors in nuclear speckles. Weis K, editor. Molecular Biology of the Cell. 2012;23(18):3694-3706
  66. 66. Yang L, Lin C, Liu W, Zhang J, Ohgi Kenneth A, Grinstein Jonathan D, et al. ncRNA- and Pc2 methylation-dependent gene relocation between nuclear structures mediates gene activation programs. Cell. 2011;147(4):773-788
  67. 67. Arun G, Diermeier S, Akerman M, Chang K-C, Wilkinson JE, Hearn S, et al. Differentiation of mammary tumors and reduction in metastasis upon Malat1 lncRNA loss. Genes & Development. 2015;30(1):34-51
  68. 68. Malakar P, Shilo A, Mogilevsky A, Stein I, Pikarsky E, Nevo Y, et al. Long noncoding RNA MALAT1 promotes hepatocellular carcinoma development by SRSF1 upregulation and mTOR activation. Cancer Research. 2016;77(5):1155-1167
  69. 69. Fei J, Jadaliha M, Harmon TS, Li ITS, Hua B, Hao Q , et al. Quantitative analysis of multilayer organization of proteins and RNA in nuclear speckles at super resolution. Journal of Cell Science. 2017;130(24):4180-4192
  70. 70. Engreitz Jesse M, Sirokman K, McDonel P, Shishkin AA, Surka C, Russell P, et al. RNA-RNA interactions enable specific targeting of noncoding RNAs to nascent pre-mRNAs and chromatin sites. Cell. 2014;159(1):188-199
  71. 71. Wilusz JE, Freier SM, Spector DL. 3′ end processing of a long nuclear-retained noncoding RNA yields a tRNA-like cytoplasmic RNA. Cell. 2008;135(5):919-932
  72. 72. Yin Q-F, Yang L, Zhang Y, Xiang J-F, Wu Y-W, Carmichael Gordon G,et al. Long noncoding RNAs with snoRNA ends. Molecular Cell. 2012;48(2):219-230
  73. 73. Huarte M, Guttman M, Feldser D, Garber M, Koziol MJ, Kenzelmann-Broz D, et al. A large intergenic noncoding RNA induced by p53 mediates global gene repression in the p53 response. Cell. 2010;142(3):409-419
  74. 74. Dimitrova N, Zamudio Jesse R, Jong Robyn M, Soukup D, Resnick R, Sarma K, et al. LincRNA-p21 activates p21 In cis to promote Polycomb target gene expression and to enforce the G1/S checkpoint. Molecular Cell. 2014;54(5):777-790
  75. 75. Hung T, Wang Y, Lin MF, Koegel AK, Kotake Y, Grant GD, et al. Extensive and coordinated transcription of noncoding RNAs within cell cycle promoters. Nature Genetics. 2011;43(7):621-629
  76. 76. Lee S, Kopp F, Chang T-C, Sataluri A, Chen B, Sivakumar S, et al. Noncoding RNA NORAD regulates genomic stability by sequestering PUMILIO proteins. Cell. 2016;164(1-2):69-80
  77. 77. Tichon A, Perry RB-T, Stojic L, Ulitsky I. SAM68 is required for regulation of Pumilio by the NORAD long noncoding RNA. Genes & Development. 2018;32(1):70-78
  78. 78. Schmitt AM, Garcia JT, Hung T, Flynn RA, Shen Y, Qu K, et al. An inducible long noncoding RNA amplifies DNA damage signaling. Nature Genetics. 2016;48(11):1370-1376
  79. 79. Munschauer M, Nguyen CT, Sirokman K, Hartigan CR, Hogstrom L, Engreitz JM, et al. The NORAD lncRNA assembles a topoisomerase complex critical for genome stability. Nature. 2018;561(7721):132-136
  80. 80. Latos PA, Pauler FM, Koerner MV, Şenergin HB, Hudson QJ, Stocsits RR, et al. Airn transcriptional overlap, but not its lncRNA products, induces imprinted Igf2r silencing. Science. 2012;338(6113):1469-1472
  81. 81. Kretz M, Webster DE, Flockhart RJ, Lee CS, Zehnder A, Lopez-Pajares V, et al. Suppression of progenitor differentiation requires the long noncoding RNA ANCR. Genes & Development. 2012;26(4):338-343
  82. 82. Zhu L, Xu P-C. Downregulated LncRNA-ANCR promotes osteoblast differentiation by targeting EZH2 and regulating Runx2 expression. Biochemical and Biophysical Research Communications. 2013;432(4):612-617
  83. 83. Pasmant E, Laurendeau I, Héron D, Vidaud M, Vidaud D, Bièche I. Characterization of a germ-line deletion, including the entire INK4/ARF locus, in a melanoma-neural system tumor family: Identification of ANRIL, an antisense noncoding RNA whose expression Coclusters with ARF. Cancer Research. 2007;67(8):3963-3969
  84. 84. Visel A, Zhu Y, May D, Afzal V, Gong E, Attanasio C, et al. Targeted deletion of the 9p21 non-coding coronary artery disease risk interval in mice. Nature. 2010;464(7287):409-412
  85. 85. Xing Z, Lin A, Li C, Liang K, Wang S, Liu Y, et al. lncRNA directs cooperative epigenetic regulation downstream of chemokine signals. Cell. 2014;159(5):1110-1125
  86. 86. Zhao Y, Yu Y, You S, Zhang C, Wu L, Zhao W, et al. Long non-coding RNA MALAT1 as a detection and diagnostic molecular marker in various human cancers: A pooled analysis based on 3255 subjects. OncoTargets and Therapy. 2020;13:5807-5817
  87. 87. Sun Y, Ma L. New insights into long non-coding RNA MALAT1 in cancer and metastasis. Cancers. 2019;11(2):216
  88. 88. Tripathi V, Ellis JD, Shen Z, Song DY, Pan Q , Watt AT, et al. The nuclear-retained noncoding RNA MALAT1 regulates alternative splicing by modulating SR splicing factor phosphorylation. Molecular Cell. 2010;39(6):925-938
  89. 89. Amodio N, Raimondi L, Juli G, Stamato MA, Caracciolo D, Tagliaferri P, et al. MALAT1: A druggable long non-coding RNA for targeted anti-cancer approaches. Journal of Hematology & Oncology. 2018;11(1):63
  90. 90. Yuan X, Guo Y, Chen D, Luo Y, Chen D, Miao J, et al. Long non-coding RNA MALAT1 functions as miR-1 sponge to regulate Connexin 43-mediated ossification of the posterior longitudinal ligament. Bone. 2019;127:305-314
  91. 91. Statello L, Guo C-J, Chen L-L, Huarte M. Gene regulation by long non-coding RNAs and its biological functions. Nature Reviews Molecular Cell Biology. 2021;22(2):96-118
  92. 92. López-Urrutia E, Bustamante Montes LP. Ladrón de Guevara Cervantes D, Pérez-Plasencia C, Campos-Parra AD. Crosstalk between long non-coding RNAs, micro-RNAs and mRNAs: Deciphering molecular mechanisms of master regulators in cancer. Frontiers in. Oncology. 2019;9:669
  93. 93. Noh JH, Kim KM, Abdelmohsen K, Yoon J-H, Panda AC, Munk R, et al. HuR and GRSF1 modulate the nuclear export and mitochondrial localization of the lncRNA RMRP. Genes & Development. 2016;30(10):1224-1239

Written By

Pranjal Kumar and Nikita Bhandari

Submitted: 22 February 2022 Reviewed: 12 April 2022 Published: 09 June 2022