1,628
Views
29
CrossRef citations to date
0
Altmetric
Review

Interplay of chromatin modifications and non-coding RNAs in the heart

, , &
Pages 101-112 | Received 14 Aug 2013, Accepted 06 Sep 2013, Published online: 10 Oct 2013

Abstract

Precisely regulated patterns of gene expression are dependent on the binding of transcription factors and chromatin-associated determinants referred to as co-activators and co-repressors. These regulatory components function with the core transcriptional machinery to serve in critical activities to alter chromatin modification and regulate gene expression. While we are beginning to understand that cell-type specific patterns of gene expression are necessary to achieve selective cardiovascular developmental programs, we still do not know the molecular machineries that localize these determinants in the heart. With clear implications for the epigenetic control of gene expression signatures, the ENCODE (Encyclopedia of DNA Elements) Project Consortium determined that about 90% of the human genome is transcribed while only 1-2% of transcripts encode proteins. Emerging evidence suggests that non-coding RNA (ncRNA) serves as a signal for decoding chromatin modifications and provides a potential molecular basis for cell type-specific and promoter-specific patterns of gene expression. The discovery of the histone methyltransferase enzyme EZH2 in the regulation of gene expression patterns implicated in cardiac hypertrophy suggests a novel role for chromatin-associated ncRNAs and is the focus of this article.

Introduction

The mammalian heart is the first organ to form in the vertebrate embryo. During development, heart chambers undergo structural changes mediated by specific cellular and extracellular cues such as hormone stimulation. Heart development involves stage-specific changes that are precisely regulated by spatial and temporal events on chromatin to regulate specific gene expression patterns.Citation1 For example, genes expressed at later stages in cardiac development, such as cardiomyocyte maturation and terminal differentiation, show mono-methylation of histone H3 lysine 4 (H3K4me1) at early stages of development, whereas activation at later stages are often specified by H3K4me3 modification.Citation1 During lineage commitment there are stage-specific acetylation (H3K27ac) and methylation (H3K4me1, H3K4me3 and H3K27me3) of lysine residues on histone H3 regulating gene expression and cardiac differentiation. For example, both methylation and acetylation of histone proteins at distinct lysine positions determine specific histone modification signatures that predict gene expression patterns that serve as transcription-factor binding sites as well as the exchange of co-regulatory complexes on promoters.Citation2 Gene activation in pluripotent stem cells is associated with H3K4me1 patterns at gene promoters, which are also activated at later stages in the cardiac lineage, which is in striking contrast to H3K27me3 patterns and genes destined for suppression. Genes that code for the adult isoform cardiac contractile protein such as α-myosin heavy chain (α-MHC) and the transcription factor NKX2.5, are activated specifically at later stages of cardiac differentiation. These genes show high levels of H3K27me3 deposition at pluripotent stage, which are gradually erased and replaced by H3K4me3 modification.Citation3

Cardiomyocyte cells respond using adaptive mechanisms to changing environmental stimuli such as increased workload. Such physiological changes are marked by an increase in cardiomyocyte size and ventricular mass, which is referred to as cardiac hypertrophy. Chronic exercise training or pregnancy can increase heart muscle mass and contractile ability, often referred to as physiological hypertrophy.Citation4 However, there is a fine balance between physiological and pathological hypertrophy which are distinguished by cardiac failure. Pathophysiological surroundings such as acute and chronic myocardial stress including hypertension, valvular disease, and myocardial infarction, can dramatically increase the size of the ventricular chamber.Citation4,Citation5 This is referred to as pathological-cardiac hypertrophy and, like physiological hypertrophy, stimulates a phase of neurohumoral and biomechanical signals within the myocardium. While it is considered that physiological hypertrophy is generally advantageous as well as reversible, pathological hypertrophy causes irreversible remodeling leading to deformation of the ventricles and reduced heart contractility.Citation6

The discovery of specific activator and repressor complexes important in cardiac development has revealed several mechanistic insights into myocardial function, cardiac development as well as heart disease. Ventricular hypertrophy is associated with re-activation of fetal genes that include ANP, BNP, and β-MHC as well as the suppression of SERCA2a and α-MHC genes in the adult heart.Citation6 The recruitment of ATPase-dependent chromatin remodeling complexes that belong to the SWI/SNF familyCitation7 have been shown to contextually associate with either histone acetyltransferases (HATs) or histone deacetylases (HDACs) to regulate cardiac gene expression.Citation8 Indeed, the recruitment and binding of p300 HAT enzyme on gene promoters is closely associated with chamber-specific gene expression patterns conferred by histone acetylation under physiological states.Citation9 In addition, recent studies have expanded the complexity of regulatory determinants that participate in cardiac gene function, for example histone modifying proteins such as EZH2 and ASXL2 specify MHC gene expression in postnatal cardiac homeostasis.Citation10,Citation11 Human homologs of Drosophila genes (Enhancer of zeste homolog 2 and Additional sex combs-like protein 2) EZH2 and ASXL2 are members of the Polycomb group (PcG) protein family and implicated in maintaining gene repressive states by chromatin modification during later stages of heart development.

Mechanisms that regulate gene expression are under the direct control of specific classes of transcription factors and core machinery that serve to alter chromatin structure and function. However the precise actions of transcription factors and chromatin remodeling determinants, including histone and non-histone modifying enzymes in gene transcription are poorly characterized in the heart. Moreover, the diversity of transcription factors and chromatin modifying enzymes specifying gene expression patterns presents a major conceptual problem when attempting to predict specific interactions with target genes. In this article we explore the basis of cell type-specific and gene-specific patterns of gene regulation that integrate chromatin-interacting ncRNAs with histone modifying enzymes that functionally serve to alter gene structure and expression. Recent experimental observations show that chromatin remodeling and histone modification confer important transcriptional programs as a result of development and cardiac disease.Citation12-Citation15 The diverse interplay of histone modifying enzymes interacting with long non-coding RNAs (lncRNA) that serve to localize DNA-binding proteins as well as direct specific post-translational modifications to regulate gene expression has been described and is the focus of our discussion.Citation16-Citation18

Physiological Roles of lncRNAs in the Heart

Recent advances in nucleic acid sequencing technologies has revealed that nearly 90% of the genome is transcribed in one tissue type or another, with estimates that between 70-98% constitute ncRNAs.Citation19-Citation21 These transcripts are broadly classified in two groups according to nucleotide length: short ncRNAs (<200 nt), such as microRNA (miRNA) and long ncRNAs (>200 nt), such as the natural antisense transcripts (NATs) (). Interestingly, ncRNAs have been thought for some time to interact with DNA to regulate important nuclear functions. Indeed, Jacob and Monod explored this concept of base complementarity between RNA and DNA sequencesCitation22 which later was experimentally examined in triplex-forming sequences derived from human c-MYC.Citation23 Direct evidence of interacting ncRNA mediating gene silencing-epigenetic changes exposed recruitment of important regulatory components in RNA-dependent DNA methylation.Citation24

Table 1. Classification of functional ncRNAs. Transcriptional gene silencing functions of short (grey background) and long ncRNAs by chromatin interaction

When, in 1993 two studies published back-to-back in Cell described a putative role for short ncRNAs in C. elegans development, the importance of these critical findings was probably underappreciated in transcription biology.Citation25,Citation26 How ncRNAs recognize and interact with target sequences to regulate gene expression still remains poorly characterized. Although short ncRNAs are strongly conserved but of unknown function, the seminal discoveries by the groups led by Ambros and Ruvkun have revealed a regulatory complexity mediated by ncRNAs. The field has expanded tremendously with a better understanding of the significance in biology and disease. Recent studies now show that during development, ncRNAs are expressed in a dynamic fashion and regulated by specific cellular and environmental cues.Citation16,Citation17

The importance of short ncRNAs in heart development was elegantly demonstrated by cardiac-specific deletion of miRNA-processing enzyme, DICER.Citation27 Abundantly expressed in the heart, miR-1 and miR-133 are associated with cardiovascular development and myeloid differentiation.Citation28-Citation30 Recently, functional paradigms for several lncRNAs have also been described such as the participation in embryonic differentiation and cell-lineage development as well as transcriptional control.Citation16,Citation17,Citation31,Citation32 While lncRNAs can serve as spliceosome and ribosome components in eukaryotic RNA metabolism, recent experimental observations indicate a role in organizing chromatin conformation and shaping the genome. For example, chromatin interacting lncRNAs were recently identified as key determinants of gene imprinting (such as XIST and KCNQ1OT1 as well as AIR), whereas the recruitment of PRC2 components are implicated in gene suppression events that involve HOTAIR and TUG1.Citation33-Citation35 Recently, knockdown of lncRNAs expressed in embryonic stem cells has revealed more than one hundred functional lncRNAs associated with the maintenance of pluripotency.Citation36 In addition, several lncRNAs have been implicated in normal heart physiology. For example, in the mouse, Braveheart (Bvht) and Fendrr are thought to have critical roles in cardiac lineage specification during embryonic development.Citation16,Citation17 The silencing of Bvht in mES cells results in the loss of cardiomyocyte beating in embryoid bodies (EB) at day 11 of differentiation.Citation16 Whereas the expression of tissue-specific Fendrr is a regulator of heart and body wall development.Citation17 While these results are not fully understood, it is hypothesized that Bvht and Fendrr control gene expression by interacting with the regulatory cofactors, PRC2 and TrxG/MLL complexes. These studies highlight the importance of lncRNA transcripts defining chromatin structure and gene expression necessary for heart development. Recent studies have also identified putative roles for over expressed lncRNAs in cancer (MALAT1 and HOTAIR) and Alzheimer’s (BACE1-AS), as well as reduced expression of lncRNAs in anemia (LincRNA-EPS) and Huntington’s disease (HTT-AS).Citation37-Citation40 In addition to the general involvement of DNA-binding motifs that function in the recruitment of transcription factors, new roles for lncRNAs in mediating chromatin-protein interactions have recently been described.Citation20,Citation41 Several lncRNAs have putative sequence motifs and structural domains implicated in protein association and interacting with specific gene targets. Indeed, several chromatin-interacting proteins have recently been described to have ncRNA-binding domains such as the polycomb-group (PcG) proteins, which are involved in the suppression of gene expression mediated by chromatin modification.Citation42,Citation43

Non-Coding RNAs Connect EZH2 with Chromatin

The expression of lncRNAs and natural antisense transcripts have recently been shown to regulate gene transcription and protein translation in the heart.Citation14,Citation15 The antisense (AS) transcripts to NPPA (AS-NPPA) and β-MHC (AS-β-MHC) are examples of regulatory lncRNAs in the myocardium. These transcripts are thought to associate with chromatin and regulate the expression of sense counterparts, NPPA and β-MHC whose expressions are regulated by EZH2 in the heart. The EZH2 lysine methyltransferase has a binding domain that is thought to mediate interaction with lncRNAs.Citation42 For instance, phosphorylation of threonine (T365) of EZH2 interacts with HOTAIR and XIST.Citation33 Although well characterized in cancer, the specific interactions of ncRNAs with histone modifying determinants such as EZH2, remain poorly described in the heart.Citation44 Several lysine methyltransferase proteins have a conserved SET-domain region, which is thought to be critical to chromatin association as well as enzymatic activity. A number of methyl-writing SET-domain family members such as G9A, SET7, SMYD3, SET2, SET1, and EZH2, can bind to single-stranded DNA and RNA.Citation45-Citation48 In addition, several MLL family proteins that contain the SET-domain are known to interact with ncRNA either directly or indirectly.Citation49,Citation50 The methyl-erasing enzyme, LSD1, is thought to bind directly to the 3’ end of the HOTAIR lncRNA to regulate HOXD gene expression.Citation51

Recent data published by several groups suggest putative roles for antisense transcripts in mediating EZH2 interactions with chromatin ().Citation10,Citation52 The expression of genes encoding contractile proteins and transcription factors implicated in heart disease are altered in EZH2-knockout mouse models.Citation10 Deep sequencing of chromatin immunoprecipitated from the mouse heart using antibodies that recognize EZH2 show direct interaction with genes implicated in cardiac disease ().Citation52 Interestingly, EZH2 appears to bind novel bi-directional promoter (bdP) sequence to regulate sense and antisense RNA expression. For example, the heart displays altered expression of tumor suppressor related genes CDKN2B, CDKN2A, and ARF encoding the INK4/ARF locus at chromosome 9p21 in EZH2-null mice.Citation10,Citation52 The ANRIL antisense is thought to regulate these genes by PcG-dependent silencing.Citation53 But, whether ANRIL directly regulates EZH2 chromatin interaction at the 9p21 region in cardiomyocyte cells remains to be determined. In favor of a role in cardiac homeostasis, individuals homozygous for the SNP allele at the 9p21 region show altered ANRIL expression and increased susceptibility to atherogenic plaque development and coronary heart disease (CHD) as well as diabetes.Citation54,Citation55 While CDKN2A expression levels were reduced in 9p21 knockout hearts, there was no evidence for cardiac hypertrophy or cardiovascular pathology.Citation56 Other studies also report ANRIL interactions with PcG proteins such as CBX7 and SUZ12 to regulate CDKN2B and CDKN2A gene expression.Citation57,Citation58 Overexpression of ANRIL in cultured cells significantly altered the expression of a large number of distant genes proposing ANRIL as a trans regulatory element.Citation58 Ontology analysis has identified genes involved in the regulation of chromatin structure and function.Citation58

Table 2. Chromatin immunoprecipitation in mouse left ventricle shows specific interaction of EZH2 at genes with bi-directional transcription.

Cardiac hypertrophy and heart failure are associated with changes in the expression of α- and β-MHC mRNAs and this shift in myosin-isoform distribution serves important roles in cardiac muscle fiber shortening.Citation59 The silencing of α-MHC in the failing hearts has led renewed interest to restore expression of this gene in hypertrophic tissue.Citation59 The MHC genes are clustered on chromosome 14 in humans and mice (chromosome 15 in rat) and the α- and β- MHC genes are separated by an intergenic sequence of ~4.5 kb in length ().Citation60 The β-MHC gene is upstream of α-MHC and both transcribe mature mRNA approximately 7 kb in length.Citation60 The complexity of MHC gene regulation presents interesting conceptual problems as well as experimental challenges, with the identification of transcripts on opposing DNA strands. This complementary sequence to the canonical mRNA represents the antisense or non-coding RNA.Citation61 The intergenic region of MHC is thought to contain a bdP that transcribes both AS β-MHC and α-MHC in opposite directions.Citation61 Transcription of AS β-MHC progresses in the direction of the β-MHC gene and is thought to regulate the expression of MHC genes in response to pressure overload.Citation61,Citation62

Figure 1. Interplay of chromatin modifications and non-coding RNAs regulate MHC genes in the heart. The expression of cardiac α- and β-MHC genes is regulated in (A) healthy and (B) diseased heart. The bi-directional promoter (bdP) of the α- and β-MHC intergenic region comprises binding sequences for GATA, CTF1/NF1, RAR, T3R, MEF-2 transcription factors. Both the α- and β-MHC genes encode miRNA-208a and miRNA-208b that function in heart health and disease. The bdP is known to transcribe AS β-MHC which serves to regulate β-MHC sense (mRNA) transcription by chromatin interaction. The co-regulatory chromatin determinants BRG1, histone deacetylases (HDACs), and EZH2 are involved in the suppression of AS β-MHC and α-MHC genes in disease.

Figure 1. Interplay of chromatin modifications and non-coding RNAs regulate MHC genes in the heart. The expression of cardiac α- and β-MHC genes is regulated in (A) healthy and (B) diseased heart. The bi-directional promoter (bdP) of the α- and β-MHC intergenic region comprises binding sequences for GATA, CTF1/NF1, RAR, T3R, MEF-2 transcription factors. Both the α- and β-MHC genes encode miRNA-208a and miRNA-208b that function in heart health and disease. The bdP is known to transcribe AS β-MHC which serves to regulate β-MHC sense (mRNA) transcription by chromatin interaction. The co-regulatory chromatin determinants BRG1, histone deacetylases (HDACs), and EZH2 are involved in the suppression of AS β-MHC and α-MHC genes in disease.

The regulation of MHC isoforms involves the coordinated actions of core machinery that include DNA transcription factors, chromatin remodeling, and expression of antisense RNA transcripts. Perhaps the most interesting of recent experimental results highlights the complex regulation of the MHC genes includes both transcriptional and post-transcriptional changes. Recent experiments in EZH2 mutant mice reveal changes to MHC isoform regulation characteristic of the hypertrophic heart.Citation10 In addition to H3K27me3 modification by EZH2 is the direct involvement of histone-modifying enzymes such as HDAC9, ASXL2, and chromatin remodeling enzymes, such as BRG1 and PARP1 which interact with the bdP ().Citation11,Citation63 DNase hypersensitive sites are also associated with MHC gene expression at various developmental stages of the heart.Citation64 Whatever the role of EZH2, showing its involvement in chromatin dependent association with ncRNA is the first step in revealing how MHC isoform expression is regulated in heart disease.

Novel ncRNAs in the Heart

Long ncRNA expression recently described in the heart with regulatory roles involving chromatin modification and function is summarized in . RNA sequencing of the myocardium has revealed specific transcriptome profiles for coding and non-coding transcripts that distinguish the stages of the failing heart.Citation65 Recent studies have identified more than 1300 previously unannotated exons with altered expression levels in animal models of heart failure.Citation65 Among these, almost 682 exons displayed differential expression and the majority (81%) of unannotated RNAs expressed were non-coding RNAs. For example, the expression of H19 lncRNA was highest in heart failure tissue when compared to cardiac hypertrophy. The function of H19 in the myocardium remains poorly characterized, as for human heart explants, transcriptome profiling has shown the expression of putative ncRNAs associated with the development of cardiomyopathy.Citation66 These studies suggest that a large number of novel transcripts are dynamically expressed in the myocardium. Serial analysis of gene expression (SAGE) of different human tissue types has identified cardiac-specific expression of NCRNA00116.Citation67 Despite the tremendous advances in technology used to identify novel RNA species, the physiological function of these molecules remains largely uncharted.Citation68

Table 3. Long ncRNA expression in the heart

Analysis of ncRNA Dependent-Chromatin Interactions

Recent methodological developments in transcript analysis have seen a tremendous amount of information generated from massive parallel sequencing. While historically difficult to ascribe function to the large number of non-coding RNAs, these transcripts are readily identifiable using RNA sequencing approaches. A number of lncRNAs contain chromatin binding domains and other sequences involved in the interactions with proteins as well as regulating gene expression.Citation41,Citation43 In the next section, we discuss some of the methodological developments that have enabled the characterization long ncRNA dependent-chromatin interactions.

Methods Used in the Detection and Characterization of lncRNAs

Important protein-coding genes including those implicated in heart disease have antisense transcription and ncRNA expression.Citation69,Citation70 Conventionally, in first-strand synthesis, complementary DNA (cDNA) is generated at low temperatures (37 °C) using random/oligo-dT primers that are non-specific to gene sequences as well as lacking strand-specific (5’ to 3’ orientation) information. To distinguish sense from antisense, strand-specific oligonucleotides are used to anneal either mRNA (sense) or ncRNA (antisense) at high temperatures (50‒60 oC) followed by first-strand cDNA synthesis. For example, strand-specific primers to cardiac MHC and troponin genes have been used to quantitatively assay sense (mRNA) and antisense (ncRNA) expression in the heart.Citation71,Citation72 Recently, several novel procedures have been developed to quantify strand-specific expression of the transcriptome ().Citation73,Citation74

Table 4. Methodologies for the detection, characterization and structural analysis of lncRNA. ncRNA-chromatin interaction assays are highlighted with grey background

Almost 90% of the human transcriptome is alternatively spliced in terminally differentiated cardiomyocytes and neurons.Citation75 RNA splice variants greatly increase biodiversity of proteins.Citation76 For example, distinct alternative splicing of the cardiac steroid receptor activator (SRA) transcript can generate SRA protein-coding transcript as well as non-coding regulatory SRA transcript.Citation77 Consistent with this idea, splice variants in the heart are known to exist for ANRIL and regulate circularization of this transcript, whereby one variant type interacts with EZH2 whilst the other is masked for the EZH2 binding domain.Citation78 Alternative splicing of ncRNA is perhaps key to understanding ncRNA dependent-chromatin interactions. Several strategies, such as exon-scanning and rapid amplification of cDNA ends (RACE) have successfully identified splice variants to cardiac troponin I- and NPPA- antisense transcripts ().Citation68,Citation79 Other examples of lncRNAs identified include KCNQ1OT1 and HOTTIP.Citation50,Citation80

RNA sequencing (RNA-Seq) approaches generate millions of reads that often fail to accurately identify gene structure as well as result in missing detection of low-abundant transcripts and non-polyadenylated ncRNAs.Citation81 Transcript profiling can be studied using tiling arrays or targeted RNA CaptureSeq (RNA capture sequencing).Citation82,Citation83 For example, Mercer et al.Citation82 used this approach because rare transcripts are thought to be below the detection limits of conventional RNA-Seq. Surprisingly, the study reported complex ncRNA transcription and widespread expression of novel transcripts.Citation82 The authors characterize alternative splice junctions to the HOTAIR transcript predicted to interfere with PcG binding.Citation82 Taken together, these data suggest that post-transcriptional splicing can regulate ncRNA dependent-chromatin binding.

Protein expression may also be determined by RNA stability and recent experimental observations suggest dynamic regulation of ncRNA stability in response to specific environmental cues. Pulse labeling of RNA followed by sequencing or 5'-bromo-uridine immunoprecipitation chase–deep sequencing analysis (BRIC-Seq) has identified novel and highly stable lncRNAs.Citation32 This technique is used to study RNA decay and has revealed that some lncRNAs infact have short half-lives (t1/2 < 4 h) such as the cardiac ANRIL transcript, HOTAIR, TUG1, and GAS5. Other intriguing observations from the study highlighted that hundreds of short-lived regulatory RNAs designated as short-lived non-coding transcripts (SLiTs) have putative roles in nuclear function.Citation32 An alternative method of studying RNA stability is transcriptional inhibition by Actinomycin D (ActD).Citation84 Mouse neuroblastoma cells exposed to ActD over a 32 h period identified over 800 lncRNAs and 12 000 mRNAs that were classified highly stable with a half-life > 16 h or low stability with a half-life < 2 h.Citation85 The regulatory RNA, NEAT1 was identified as one of the least stable ncRNAs which is thought to be dynamically regulated. Similarly, global run-on sequencing (GRO-Seq) and native elongating transcript sequencing (NET-Seq) techniques have been be used to assay nascent RNA transcripts.Citation86,Citation87 These studies identified immediate transcriptional response to estrogen signaling demonstrating that lncRNAs are dynamically regulated.Citation86 The most obvious conclusion is that low stability lncRNAs are non-functional, but this argument is perhaps overly simplistic, when interpreted slightly differently, long non-coding RNAs may act immediately after transcription to mediate chromatin-dependent interactions.

Long ncRNA-Chromatin Interaction Assays

Long ncRNAs that stably interact with chromatin at specific genomic sites can be detected by fluorescent in situ hybridization (FISH) of the target RNA using antisense probes.Citation88 FISH has traditionally been the method of choice to study long ncRNA dependent-chromatin interactions.Citation89,Citation90 More recently, FISH was employed to assay changes in chromatin architecture for KCNQ1OT1 a lncRNA that regulates KCNQ1 expression in the developing heart.Citation91 Alternatively, locus-specific lncRNA interactions can be examined using formaldehyde fixation and chromatin immunoprecipitation methods (RNA-ChIP) that use antibodies that recognize RNA-binding proteins such as EZH2 and G9A.Citation80 Alternatively, native RNA-ChIP using MNase digestion have also been successfully applied to the study of chromatin associated RNAs.Citation92 In striking contrast to formaldehyde crosslinking, immunoprecipitation of native soluble chromatin allows for direct mapping of mono-, di- and tri-nucleosomal structures.Citation93

Long ncRNAs can interact in a locus-specific manner using homologous complementary sequences.Citation94 The applicability of biotinylated RNA tiling probes complementary to target lncRNA was recently used to immunoprecipitate interacting DNA sequences and proteins. Examples of these methods include ChIRP (chromatin isolation by RNA purification) and CHART (chromatin hybridization analysis of RNA targets) which have identified novel genome-wide interactions for HOTAIR and ROX2.Citation95,Citation96 Infact, with the advent of high-throughput sequencing it has been possible to identify novel RNAs using crosslinking immunoprecipitation (HITS-CLIP) and photoactivatable ribonucleoside enhanced crosslinking and immunoprecipitation (PAR-CLIP).Citation97,Citation98 These methodologies were recently used to identify the interaction of EZH2 with several ncRNAs, including ANRIL which has been associated with many diseases including coronary artery disease, diabetes and cancer.Citation99

Structural Analysis of lncRNAs

Besides sequence-based chromatin recognition, RNA folding can also influence ncRNA dependent-chromatin interactions.Citation100 For example, genes that code for DMD, P450, MLL, and ETS-1 are circular transcripts with diverse functions.Citation101-Citation104 The hypertrophy responsive NCX1 gene is thought to produce circular poly(A-) transcripts in the human heart, however the biological significance of circular RNAs has remained elusive.Citation105 Recent evidence now suggests that circular RNAs function as miRNA sponges that compete with RNA binding proteins to form a class of post-transcriptional regulators.Citation106,Citation107 Accordingly, circular antisense RNAs are targeted by RISC components for gene regulation.Citation108 The mechanism of RNA circularization is a result of non-canonical post-transcriptional exon scrambling (PTES). Non-canonical PTES appears to be a predominant event in human liver as well as the heart.Citation109 Because of their low abundance, the majority of circular transcripts are largely undetectable by conventional RNA-sequencing. To investigate the circular component of the transcriptome, protocols employ RNaseR, an enzyme that degrades linear but not circular transcripts.Citation104 Coupled with RNaseR, next generation sequencing has identified PTES mediated circular RNA transcripts to hundreds of human genes, the majority of which were not polyadenylated.Citation104 Infact, circular and linear forms of cardiac antisense RNA, ANRIL have been reported.Citation78 The expression of circular ANRIL might be associated with atherosclerotic vascular disease. Thousands of human mRNA and ncRNA transcripts are extensively methylatedCitation110 and these RNA modifications are thought to alter Argonaute binding as well as transcript folding.Citation111 Moreover, recent identification of specific ncRNA structures such as the TINCR boxes regulate the interaction of these transcripts with regulatory proteins.Citation100 FragSeq or fragmentation sequencing is a novel method that integrates RNA structure analysis with genome-wide sequencing.Citation112 The Nuclease P1 enzyme is used to cleave single-stranded nucleic acids thereby preserving the intra- and inter-molecular RNA interactions. The development of these methodologies has revolutionized genome-wide analysis of cellular RNAs , which will be critical in defining regulatory networks at the genomic scale.Citation113

Conclusions and Future Considerations

Recent experimental observations show lncRNAs regulate cardiac gene expression. This is probably best exemplified at the bidirectional promoter of the MHC genes which involves the interaction of EZH2 with the antisense β-MHC transcript to regulate MHC isoform switch (). While always considered to be integral elements in the post-transcriptional control of gene expression it is the recent technological developments that have been critical to understand the role of ncRNAs in the heart. The advent of massive parallel sequencing has brought improved understanding of the regulatory mechanisms underlying cardiac pathology and developmental growth as well as integrating functional genomics. Although the relevance of the non-coding genome to cardiac disease has mainly been studied in the context of the widespread disruption of expression, studies now show that ncRNAs are also critical determinants of gene regulation. Taken together with their emerging role with chromatin modification, the non-coding genome should provide new strategies and specific targets to prevent, restore or reverse the effects of pathological hypertrophy in the failing heart.

Abbreviations:
EZH2=

enhancer of zeste homolog 2

ncRNA=

non-coding RNA

lncRNA=

long non-coding RNA

MHC=

myosin heavy chain

HDAC=

histone deacetylase

HAT=

histone acetyltransferase

miRNA=

MicroRNA

NAT=

natural antisense transcript

PcG=

polycomb-group

AS=

antisense

SET=

Su(var)3-9 and enhancer of zeste

bdP=

bi-directional promoter

Disclosure of Potential Conflicts of Interest

No potential conflicts of interest were disclosed.

Acknowledgments

The authors acknowledge grant and fellowship support from the National Health and Medical Research Council (NHMRC) and the National Heart Foundation of Australia (NHF). Mathiyalagan P was awarded a Monash Graduate Scholarship and El-Osta A and Du X-J are Senior Research Fellows of the NHMRC. Supported in part by the Victorian Government’s Operational Infrastructure Support program.

10.4161/epi.26405

References

  • Wamstad JA, Alexander JM, Truty RM, Shrikumar A, Li F, Eilertson KE, Ding H, Wylie JN, Pico AR, Capra JA, et al. Dynamic and coordinated epigenetic regulation of developmental transitions in the cardiac lineage. Cell 2012; 151:206 - 20; http://dx.doi.org/10.1016/j.cell.2012.07.035; PMID: 22981692
  • Strahl BD, Allis CD. The language of covalent histone modifications. Nature 2000; 403:41 - 5; http://dx.doi.org/10.1038/47412; PMID: 10638745
  • Paige SL, Thomas S, Stoick-Cooper CL, Wang H, Maves L, Sandstrom R, Pabon L, Reinecke H, Pratt G, Keller G, et al. A temporal chromatin signature in human embryonic stem cells identifies regulators of cardiac development. Cell 2012; 151:221 - 32; http://dx.doi.org/10.1016/j.cell.2012.08.027; PMID: 22981225
  • Bernardo BC, Weeks KL, Pretorius L, McMullen JR. Molecular distinction between physiological and pathological cardiac hypertrophy: experimental findings and therapeutic strategies. Pharmacol Ther 2010; 128:191 - 227; http://dx.doi.org/10.1016/j.pharmthera.2010.04.005; PMID: 20438756
  • Kehat I, Molkentin JD. Molecular pathways underlying cardiac remodeling during pathophysiological stimulation. Circulation 2010; 122:2727 - 35; http://dx.doi.org/10.1161/CIRCULATIONAHA.110.942268; PMID: 21173361
  • Frey N, Olson EN. Cardiac hypertrophy: the good, the bad, and the ugly. Annu Rev Physiol 2003; 65:45 - 79; http://dx.doi.org/10.1146/annurev.physiol.65.092101.142243; PMID: 12524460
  • Chang L, Kiriazis H, Gao XM, Du XJ, El-Osta A. Cardiac genes show contextual SWI/SNF interactions with distinguishable gene activities. Epigenetics 2011; 6:760 - 8; http://dx.doi.org/10.4161/epi.6.6.16007; PMID: 21586902
  • Backs J, Olson EN. Control of cardiac growth by histone acetylation/deacetylation. Circ Res 2006; 98:15 - 24; http://dx.doi.org/10.1161/01.RES.0000197782.21444.8f; PMID: 16397154
  • Mathiyalagan P, Chang L, Du XJ, El-Osta A. Cardiac ventricular chambers are epigenetically distinguishable. Cell Cycle 2010; 9:612 - 7; http://dx.doi.org/10.4161/cc.9.3.10612; PMID: 20090419
  • Delgado-Olguín P, Huang Y, Li X, Christodoulou D, Seidman CE, Seidman JG, Tarakhovsky A, Bruneau BG. Epigenetic repression of cardiac progenitor gene expression by Ezh2 is required for postnatal cardiac homeostasis. Nat Genet 2012; 44:343 - 7; http://dx.doi.org/10.1038/ng.1068; PMID: 22267199
  • Lai HL, Grachoff M, McGinley AL, Khan FF, Warren CM, Chowdhury SA, Wolska BM, Solaro RJ, Geenen DL, Wang QT. Maintenance of adult cardiac function requires the chromatin factor Asxl2. J Mol Cell Cardiol 2012; 53:734 - 41; http://dx.doi.org/10.1016/j.yjmcc.2012.08.014; PMID: 23046516
  • Han P, Hang CT, Yang J, Chang CP. Chromatin remodeling in cardiovascular development and physiology. Circ Res 2011; 108:378 - 96; http://dx.doi.org/10.1161/CIRCRESAHA.110.224287; PMID: 21293009
  • Takeuchi JK, Lou X, Alexander JM, Sugizaki H, Delgado-Olguín P, Holloway AK, Mori AD, Wylie JN, Munson C, Zhu Y, et al. Chromatin remodelling complex dosage modulates transcription factor function in heart development. Nat Commun 2011; 2:187; http://dx.doi.org/10.1038/ncomms1187; PMID: 21304516
  • Schonrock N, Harvey RP, Mattick JS. Long noncoding RNAs in cardiac development and pathophysiology. Circ Res 2012; 111:1349 - 62; http://dx.doi.org/10.1161/CIRCRESAHA.112.268953; PMID: 23104877
  • Luther HP. Role of endogenous antisense RNA in cardiac gene regulation. J Mol Med (Berl) 2005; 83:26 - 32; http://dx.doi.org/10.1007/s00109-004-0613-5; PMID: 15592803
  • Klattenhoff CA, Scheuermann JC, Surface LE, Bradley RK, Fields PA, Steinhauser ML, Ding H, Butty VL, Torrey L, Haas S, et al. Braveheart, a long noncoding RNA required for cardiovascular lineage commitment. Cell 2013; 152:570 - 83; http://dx.doi.org/10.1016/j.cell.2013.01.003; PMID: 23352431
  • Grote P, Wittler L, Hendrix D, Koch F, Währisch S, Beisaw A, Macura K, Bläss G, Kellis M, Werber M, et al. The tissue-specific lncRNA Fendrr is an essential regulator of heart and body wall development in the mouse. Dev Cell 2013; 24:206 - 14; http://dx.doi.org/10.1016/j.devcel.2012.12.012; PMID: 23369715
  • Mattick JS. RNA as the substrate for epigenome-environment interactions: RNA guidance of epigenetic processes and the expansion of RNA editing in animals underpins development, phenotypic plasticity, learning, and cognition. Bioessays 2010; 32:548 - 52; http://dx.doi.org/10.1002/bies.201000028; PMID: 20544741
  • Mercer TR, Dinger ME, Mattick JS. Long non-coding RNAs: insights into functions. Nat Rev Genet 2009; 10:155 - 9; http://dx.doi.org/10.1038/nrg2521; PMID: 19188922
  • Mercer TR, Mattick JS. Structure and function of long noncoding RNAs in epigenetic regulation. Nat Struct Mol Biol 2013; 20:300 - 7; http://dx.doi.org/10.1038/nsmb.2480; PMID: 23463315
  • Clark MB, Amaral PP, Schlesinger FJ, Dinger ME, Taft RJ, Rinn JL, Ponting CP, Stadler PF, Morris KV, Morillon A, et al. The reality of pervasive transcription. PLoS Biol 2011; 9:e1000625 - , discussion e1001102; http://dx.doi.org/10.1371/journal.pbio.1000625; PMID: 21765801
  • Jacob F, Monod J. Genetic regulatory mechanisms in the synthesis of proteins. J Mol Biol 1961; 3:318 - 356
  • Belotserkovskii BP, De Silva E, Tornaletti S, Wang G, Vasquez KM, Hanawalt PC. A triplex-forming sequence from the human c-MYC promoter interferes with DNA transcription. J Biol Chem 2007; 282:32433 - 32441
  • Schmitz KM, Mayer C, Postepska A, Grummt I. Interaction of noncoding RNA with the rDNA promoter mediates recruitment of DNMT3b and silencing of rRNA genes. Genes Dev 2010; 24:2264 - 2269
  • Lee RC, Feinbaum RL, Ambros V. The C. elegans heterochronic gene lin-4 encodes small RNAs with antisense complementarity to lin-14. Cell 1993; 75:843 - 54; http://dx.doi.org/10.1016/0092-8674(93)90529-Y; PMID: 8252621
  • Wightman B, Ha I, Ruvkun G. Posttranscriptional regulation of the heterochronic gene lin-14 by lin-4 mediates temporal pattern formation in C. elegans. Cell 1993; 75:855 - 62; http://dx.doi.org/10.1016/0092-8674(93)90530-4; PMID: 8252622
  • Chen JF, Murchison EP, Tang R, Callis TE, Tatsuguchi M, Deng Z, Rojas M, Hammond SM, Schneider MD, Selzman CH, et al. Targeted deletion of Dicer in the heart leads to dilated cardiomyopathy and heart failure. Proc Natl Acad Sci U S A 2008; 105:2111 - 6; http://dx.doi.org/10.1073/pnas.0710228105; PMID: 18256189
  • Schlesinger J, Schueler M, Grunert M, Fischer JJ, Zhang Q, Krueger T, Lange M, Tönjes M, Dunkel I, Sperling SR. The cardiac transcription network modulated by Gata4, Mef2a, Nkx2.5, Srf, histone modifications, and microRNAs. PLoS Genet 2011; 7:e1001313; http://dx.doi.org/10.1371/journal.pgen.1001313; PMID: 21379568
  • Zhao Y, Ransom JF, Li A, Vedantham V, von Drehle M, Muth AN, Tsuchihashi T, McManus MT, Schwartz RJ, Srivastava D. Dysregulation of cardiogenesis, cardiac conduction, and cell cycle in mice lacking miRNA-1-2. Cell 2007; 129:303 - 17; http://dx.doi.org/10.1016/j.cell.2007.03.030; PMID: 17397913
  • van Rooij E, Sutherland LB, Qi X, Richardson JA, Hill J, Olson EN. Control of stress-dependent cardiac growth and gene expression by a microRNA. Science 2007; 316:575 - 9; http://dx.doi.org/10.1126/science.1139089; PMID: 17379774
  • Dinger ME, Amaral PP, Mercer TR, Pang KC, Bruce SJ, Gardiner BB, Askarian-Amiri ME, Ru K, Soldà G, Simons C, et al. Long noncoding RNAs in mouse embryonic stem cell pluripotency and differentiation. Genome Res 2008; 18:1433 - 45; http://dx.doi.org/10.1101/gr.078378.108; PMID: 18562676
  • Tani H, Mizutani R, Salam KA, Tano K, Ijiri K, Wakamatsu A, Isogai T, Suzuki Y, Akimitsu N. Genome-wide determination of RNA stability reveals hundreds of short-lived noncoding transcripts in mammals. Genome Res 2012; 22:947 - 56; http://dx.doi.org/10.1101/gr.130559.111; PMID: 22369889
  • Kaneko S, Li G, Son J, Xu CF, Margueron R, Neubert TA, Reinberg D. Phosphorylation of the PRC2 component Ezh2 is cell cycle-regulated and up-regulates its binding to ncRNA. Genes Dev 2010; 24:2615 - 20; http://dx.doi.org/10.1101/gad.1983810; PMID: 21123648
  • Han Y, Liu Y, Gui Y, Cai Z. Long intergenic non-coding RNA TUG1 is overexpressed in urothelial carcinoma of the bladder. J Surg Oncol 2013; 107:555 - 9; http://dx.doi.org/10.1002/jso.23264; PMID: 22961206
  • Khalil AM, Guttman M, Huarte M, Garber M, Raj A, Rivea Morales D, Thomas K, Presser A, Bernstein BE, van Oudenaarden A, et al. Many human large intergenic noncoding RNAs associate with chromatin-modifying complexes and affect gene expression. Proc Natl Acad Sci U S A 2009; 106:11667 - 72; http://dx.doi.org/10.1073/pnas.0904715106; PMID: 19571010
  • Guttman M, Donaghey J, Carey BW, Garber M, Grenier JK, Munson G, Young G, Lucas AB, Ach R, Bruhn L, et al. lincRNAs act in the circuitry controlling pluripotency and differentiation. Nature 2011; 477:295 - 300; http://dx.doi.org/10.1038/nature10398; PMID: 21874018
  • Schmidt LH, Spieker T, Koschmieder S, Schäffers S, Humberg J, Jungen D, Bulk E, Hascher A, Wittmer D, Marra A, et al. The long noncoding MALAT-1 RNA indicates a poor prognosis in non-small cell lung cancer and induces migration and tumor growth. [RNA indicates a poor prognosis in non-small cell lung cancer and induces migration and tumor growth.] J Thorac Oncol 2011; 6:1984 - 92; http://dx.doi.org/10.1097/JTO.0b013e3182307eac; PMID: 22088988
  • Hu W, Yuan B, Flygare J, Lodish HF. Long noncoding RNA-mediated anti-apoptotic activity in murine erythroid terminal differentiation. Genes Dev 2011; 25:2573 - 8; http://dx.doi.org/10.1101/gad.178780.111; PMID: 22155924
  • Faghihi MA, Modarresi F, Khalil AM, Wood DE, Sahagan BG, Morgan TE, Finch CE, St Laurent G 3rd, Kenny PJ, Wahlestedt C. Expression of a noncoding RNA is elevated in Alzheimer’s disease and drives rapid feed-forward regulation of beta-secretase. Nat Med 2008; 14:723 - 30; http://dx.doi.org/10.1038/nm1784; PMID: 18587408
  • Chung DW, Rudnicki DD, Yu L, Margolis RL. A natural antisense transcript at the Huntington’s disease repeat locus regulates HTT expression. Hum Mol Genet 2011; 20:3467 - 77; http://dx.doi.org/10.1093/hmg/ddr263; PMID: 21672921
  • Rinn JL, Chang HY. Genome regulation by long noncoding RNAs. Annu Rev Biochem 2012; 81:145 - 66; http://dx.doi.org/10.1146/annurev-biochem-051410-092902; PMID: 22663078
  • Margueron R, Reinberg D. The Polycomb complex PRC2 and its mark in life. Nature 2011; 469:343 - 9; http://dx.doi.org/10.1038/nature09784; PMID: 21248841
  • Kanhere A, Jenner RG. Noncoding RNA localisation mechanisms in chromatin regulation. Silence 2012; 3:2; http://dx.doi.org/10.1186/1758-907X-3-2; PMID: 22292981
  • Benetatos L, Voulgaris E, Vartholomatos G, Hatzimichael E. Non-coding RNAs and EZH2 interactions in cancer: long and short tales from the transcriptome. Int J Cancer 2013; 133:267 - 74; http://dx.doi.org/10.1002/ijc.27859; PMID: 23001607
  • Nagano T, Mitchell JA, Sanz LA, Pauler FM, Ferguson-Smith AC, Feil R, Fraser P. The Air noncoding RNA epigenetically silences transcription by targeting G9a to chromatin. Science 2008; 322:1717 - 20; http://dx.doi.org/10.1126/science.1163802; PMID: 18988810
  • Pagans S, Kauder SE, Kaehlcke K, Sakane N, Schroeder S, Dormeyer W, Trievel RC, Verdin E, Schnolzer M, Ott M. The Cellular lysine methyltransferase Set7/9-KMT7 binds HIV-1 TAR RNA, monomethylates the viral transactivator Tat, and enhances HIV transcription. Cell Host Microbe 2010; 7:234 - 44; http://dx.doi.org/10.1016/j.chom.2010.02.005; PMID: 20227666
  • Krajewski WA, Nakamura T, Mazo A, Canaani E. A motif within SET-domain proteins binds single-stranded nucleic acids and transcribed and supercoiled DNAs and can interfere with assembly of nucleosomes. Mol Cell Biol 2005; 25:1891 - 9; http://dx.doi.org/10.1128/MCB.25.5.1891-1899.2005; PMID: 15713643
  • Xu S, Wu J, Sun B, Zhong C, Ding J. Structural and biochemical studies of human lysine methyltransferase Smyd3 reveal the important functional roles of its post-SET and TPR domains and the regulation of its activity by DNA binding. Nucleic Acids Res 2011; 39:4438 - 49; http://dx.doi.org/10.1093/nar/gkr019; PMID: 21266482
  • Ruthenburg AJ, Allis CD, Wysocka J. Methylation of lysine 4 on histone H3: intricacy of writing and reading a single epigenetic mark. Mol Cell 2007; 25:15 - 30; http://dx.doi.org/10.1016/j.molcel.2006.12.014; PMID: 17218268
  • Wang KC, Yang YW, Liu B, Sanyal A, Corces-Zimmerman R, Chen Y, Lajoie BR, Protacio A, Flynn RA, Gupta RA, et al. A long noncoding RNA maintains active chromatin to coordinate homeotic gene expression. Nature 2011; 472:120 - 4; http://dx.doi.org/10.1038/nature09819; PMID: 21423168
  • Tsai MC, Manor O, Wan Y, Mosammaparast N, Wang JK, Lan F, Shi Y, Segal E, Chang HY. Long noncoding RNA as modular scaffold of histone modification complexes. Science 2010; 329:689 - 93; http://dx.doi.org/10.1126/science.1192002; PMID: 20616235
  • He A, Ma Q, Cao J, von Gise A, Zhou P, Xie H, Zhang B, Hsing M, Christodoulou DC, Cahan P, et al. Polycomb repressive complex 2 regulates normal development of the mouse heart. Circ Res 2012; 110:406 - 15; http://dx.doi.org/10.1161/CIRCRESAHA.111.252205; PMID: 22158708
  • Kotake Y, Nakagawa T, Kitagawa K, Suzuki S, Liu N, Kitagawa M, Xiong Y. Long non-coding RNA ANRIL is required for the PRC2 recruitment to and silencing of p15(INK4B) tumor suppressor gene. Oncogene 2011; 30:1956 - 62; http://dx.doi.org/10.1038/onc.2010.568; PMID: 21151178
  • Broadbent HM, Peden JF, Lorkowski S, Goel A, Ongen H, Green F, Clarke R, Collins R, Franzosi MG, Tognoni G, et al, PROCARDIS consortium. Susceptibility to coronary artery disease and diabetes is encoded by distinct, tightly linked SNPs in the ANRIL locus on chromosome 9p. Hum Mol Genet 2008; 17:806 - 14; http://dx.doi.org/10.1093/hmg/ddm352; PMID: 18048406
  • McPherson R, Pertsemlidis A, Kavaslar N, Stewart A, Roberts R, Cox DR, Hinds DA, Pennacchio LA, Tybjaerg-Hansen A, Folsom AR, et al. A common allele on chromosome 9 associated with coronary heart disease. Science 2007; 316:1488 - 91; http://dx.doi.org/10.1126/science.1142447; PMID: 17478681
  • Visel A, Zhu Y, May D, Afzal V, Gong E, Attanasio C, Blow MJ, Cohen JC, Rubin EM, Pennacchio LA. Targeted deletion of the 9p21 non-coding coronary artery disease risk interval in mice. Nature 2010; 464:409 - 12; http://dx.doi.org/10.1038/nature08801; PMID: 20173736
  • Yap KL, Li S, Muñoz-Cabello AM, Raguz S, Zeng L, Mujtaba S, Gil J, Walsh MJ, Zhou MM. Molecular interplay of the noncoding RNA ANRIL and methylated histone H3 lysine 27 by polycomb CBX7 in transcriptional silencing of INK4a. Mol Cell 2010; 38:662 - 74; http://dx.doi.org/10.1016/j.molcel.2010.03.021; PMID: 20541999
  • Sato K, Nakagawa H, Tajima A, Yoshida K, Inoue I. ANRIL is implicated in the regulation of nucleus and potential transcriptional target of E2F1. Oncol Rep 2010; 24:701 - 7; PMID: 20664976
  • Krenz M, Robbins J. Impact of beta-myosin heavy chain expression on cardiac function during stress. J Am Coll Cardiol 2004; 44:2390 - 7; http://dx.doi.org/10.1016/j.jacc.2004.09.044; PMID: 15607403
  • Mahdavi V, Chambers AP, Nadal-Ginard B. Cardiac alpha- and beta-myosin heavy chain genes are organized in tandem. Proc Natl Acad Sci U S A 1984; 81:2626 - 30; http://dx.doi.org/10.1073/pnas.81.9.2626; PMID: 6585819
  • Haddad F, Qin AX, Bodell PW, Zhang LY, Guo H, Giger JM, Baldwin KM. Regulation of antisense RNA expression during cardiac MHC gene switching in response to pressure overload. Am J Physiol Heart Circ Physiol 2006; 290:H2351 - 61; http://dx.doi.org/10.1152/ajpheart.01111.2005; PMID: 16415074
  • Haddad F, Jiang W, Bodell PW, Qin AX, Baldwin KM. Cardiac myosin heavy chain gene regulation by thyroid hormone involves altered histone modifications. Am J Physiol Heart Circ Physiol 2010; 299:H1968 - 80; http://dx.doi.org/10.1152/ajpheart.00644.2010; PMID: 20833952
  • Hang CT, Yang J, Han P, Cheng HL, Shang C, Ashley E, Zhou B, Chang CP. Chromatin regulation by Brg1 underlies heart muscle development and disease. Nature 2010; 466:62 - 7; http://dx.doi.org/10.1038/nature09130; PMID: 20596014
  • Huang WY, Liew CC. A conserved GATA motif in a tissue-specific DNase I hypersensitive site of the cardiac alpha-myosin heavy chain gene. Biochem J 1997; 325:47 - 51; PMID: 9224628
  • Lee JH, Gao C, Peng G, Greer C, Ren S, Wang Y, Xiao X. Analysis of transcriptome complexity through RNA sequencing in normal and failing murine hearts. Circ Res 2011; 109:1332 - 41; http://dx.doi.org/10.1161/CIRCRESAHA.111.249433; PMID: 22034492
  • Movassagh M, Choy MK, Knowles DA, Cordeddu L, Haider S, Down T, Siggens L, Vujic A, Simeoni I, Penkett C, et al. Distinct epigenomic features in end-stage failing human hearts. Circulation 2011; 124:2411 - 22; http://dx.doi.org/10.1161/CIRCULATIONAHA.111.040071; PMID: 22025602
  • Gibb EA, Vucic EA, Enfield KS, Stewart GL, Lonergan KM, Kennett JY, Becker-Santos DD, MacAulay CE, Lam S, Brown CJ, et al. Human cancer long non-coding RNA transcriptomes. PLoS One 2011; 6:e25915; http://dx.doi.org/10.1371/journal.pone.0025915; PMID: 21991387
  • Annilo T, Kepp K, Laan M. Natural antisense transcript of natriuretic peptide precursor A (NPPA): structural organization and modulation of NPPA expression. BMC Mol Biol 2009; 10:81; http://dx.doi.org/10.1186/1471-2199-10-81; PMID: 19671135
  • Katayama S, Tomaru Y, Kasukawa T, Waki K, Nakanishi M, Nakamura M, Nishida H, Yap CC, Suzuki M, Kawai J, et al, RIKEN Genome Exploration Research Group, Genome Science Group (Genome Network Project Core Group), FANTOM Consortium. Antisense transcription in the mammalian transcriptome. Science 2005; 309:1564 - 6; http://dx.doi.org/10.1126/science.1112009; PMID: 16141073
  • Core LJ, Waterfall JJ, Lis JT. Nascent RNA sequencing reveals widespread pausing and divergent initiation at human promoters. Science 2008; 322:1845 - 8; http://dx.doi.org/10.1126/science.1162228; PMID: 19056941
  • Haddad F, Qin AX, Giger JM, Guo H, Baldwin KM. Potential pitfalls in the accuracy of analysis of natural sense-antisense RNA pairs by reverse transcription-PCR. BMC Biotechnol 2007; 7:21; http://dx.doi.org/10.1186/1472-6750-7-21; PMID: 17480233
  • Voigtsberger S, Bartsch H, Baumann G, Luther HP. Cell type-specific expression of endogenous cardiac Troponin I antisense RNA in the neonatal rat heart. Mol Cell Biochem 2009; 324:1 - 11; http://dx.doi.org/10.1007/s11010-008-9974-3; PMID: 19184367
  • He Y, Vogelstein B, Velculescu VE, Papadopoulos N, Kinzler KW. The antisense transcriptomes of human cells. Science 2008; 322:1855 - 7; http://dx.doi.org/10.1126/science.1163853; PMID: 19056939
  • Levin JZ, Yassour M, Adiconis X, Nusbaum C, Thompson DA, Friedman N, Gnirke A, Regev A. Comprehensive comparative analysis of strand-specific RNA sequencing methods. Nat Methods 2010; 7:709 - 15; http://dx.doi.org/10.1038/nmeth.1491; PMID: 20711195
  • Wang ET, Sandberg R, Luo S, Khrebtukova I, Zhang L, Mayr C, Kingsmore SF, Schroth GP, Burge CB. Alternative isoform regulation in human tissue transcriptomes. Nature 2008; 456:470 - 6; http://dx.doi.org/10.1038/nature07509; PMID: 18978772
  • Mironov AA, Fickett JW, Gelfand MS. Frequent alternative splicing of human genes. Genome Res 1999; 9:1288 - 93; http://dx.doi.org/10.1101/gr.9.12.1288; PMID: 10613851
  • Chooniedass-Kothari S, Emberley E, Hamedani MK, Troup S, Wang X, Czosnek A, Hube F, Mutawe M, Watson PH, Leygue E. The steroid receptor RNA activator is the first functional RNA encoding a protein. FEBS Lett 2004; 566:43 - 7; http://dx.doi.org/10.1016/j.febslet.2004.03.104; PMID: 15147866
  • Burd CE, Jeck WR, Liu Y, Sanoff HK, Wang Z, Sharpless NE. Expression of linear and novel circular forms of an INK4/ARF-associated non-coding RNA correlates with atherosclerosis risk. PLoS Genet 2010; 6:e1001233; http://dx.doi.org/10.1371/journal.pgen.1001233; PMID: 21151960
  • Bartsch H, Voigtsberger S, Baumann G, Morano I, Luther HP. Detection of a novel sense-antisense RNA-hybrid structure by RACE experiments on endogenous troponin I antisense RNA. RNA 2004; 10:1215 - 24; http://dx.doi.org/10.1261/rna.5261204; PMID: 15272119
  • Pandey RR, Mondal T, Mohammad F, Enroth S, Redrup L, Komorowski J, Nagano T, Mancini-Dinardo D, Kanduri C. Kcnq1ot1 antisense noncoding RNA mediates lineage-specific transcriptional silencing through chromatin-level regulation. Mol Cell 2008; 32:232 - 46; http://dx.doi.org/10.1016/j.molcel.2008.08.022; PMID: 18951091
  • van der Brug M, Nalls MA, Cookson MR. Deep sequencing of coding and non-coding RNA in the CNS. Brain Res 2010; 1338:146 - 54; http://dx.doi.org/10.1016/j.brainres.2010.03.039; PMID: 20307502
  • Mercer TR, Gerhardt DJ, Dinger ME, Crawford J, Trapnell C, Jeddeloh JA, Mattick JS, Rinn JL. Targeted RNA sequencing reveals the deep complexity of the human transcriptome. Nat Biotechnol 2012; 30:99 - 104; http://dx.doi.org/10.1038/nbt.2024; PMID: 22081020
  • Kampa D, Cheng J, Kapranov P, Yamanaka M, Brubaker S, Cawley S, Drenkow J, Piccolboni A, Bekiranov S, Helt G, et al. Novel RNAs identified from an in-depth analysis of the transcriptome of human chromosomes 21 and 22. Genome Res 2004; 14:331 - 42; http://dx.doi.org/10.1101/gr.2094104; PMID: 14993201
  • Hurwitz J, Furth JJ, Anders M, Evans A. The role of deoxyribonucleic acid in ribonucleic acid synthesis. II. The influence of deoxyribonucleic acid on the reaction. J Biol Chem 1962; 237:3752 - 9; PMID: 13955883
  • Clark MB, Johnston RL, Inostroza-Ponta M, Fox AH, Fortini E, Moscato P, Dinger ME, Mattick JS. Genome-wide analysis of long noncoding RNA stability. Genome Res 2012; 22:885 - 98; http://dx.doi.org/10.1101/gr.131037.111; PMID: 22406755
  • Hah N, Danko CG, Core L, Waterfall JJ, Siepel A, Lis JT, Kraus WL. A rapid, extensive, and transient transcriptional response to estrogen signaling in breast cancer cells. Cell 2011; 145:622 - 34; http://dx.doi.org/10.1016/j.cell.2011.03.042; PMID: 21549415
  • Churchman LS, Weissman JS. Nascent transcript sequencing visualizes transcription at nucleotide resolution. Nature 2011; 469:368 - 73; http://dx.doi.org/10.1038/nature09652; PMID: 21248844
  • Levsky JM, Singer RH. Fluorescence in situ hybridization: past, present and future. J Cell Sci 2003; 116:2833 - 8; http://dx.doi.org/10.1242/jcs.00633; PMID: 12808017
  • Chureau C, Chantalat S, Romito A, Galvani A, Duret L, Avner P, Rougeulle C. Ftx is a non-coding RNA which affects Xist expression and chromatin structure within the X-inactivation center region. Hum Mol Genet 2011; 20:705 - 18; http://dx.doi.org/10.1093/hmg/ddq516; PMID: 21118898
  • Tripathi V, Ellis JD, Shen Z, Song DY, Pan Q, Watt AT, Freier SM, Bennett CF, Sharma A, Bubulya PA, et al. The nuclear-retained noncoding RNA MALAT1 regulates alternative splicing by modulating SR splicing factor phosphorylation. Mol Cell 2010; 39:925 - 38; http://dx.doi.org/10.1016/j.molcel.2010.08.011; PMID: 20797886
  • Korostowski L, Sedlak N, Engel N. The Kcnq1ot1 long non-coding RNA affects chromatin conformation and expression of Kcnq1, but does not regulate its imprinting in the developing heart. PLoS Genet 2012; 8:e1002956; http://dx.doi.org/10.1371/journal.pgen.1002956; PMID: 23028363
  • Mondal T, Rasmussen M, Pandey GK, Isaksson A, Kanduri C. Characterization of the RNA content of chromatin. Genome Res 2010; 20:899 - 907; http://dx.doi.org/10.1101/gr.103473.109; PMID: 20404130
  • Gregory RI, Randall TE, Johnson CA, Khosla S, Hatada I, O’Neill LP, Turner BM, Feil R. DNA methylation is linked to deacetylation of histone H3, but not H4, on the imprinted genes Snrpn and U2af1-rs1. Mol Cell Biol 2001; 21:5426 - 36; http://dx.doi.org/10.1128/MCB.21.16.5426-5436.2001; PMID: 11463825
  • Zappulla DC, Cech TR. RNA as a flexible scaffold for proteins: yeast telomerase and beyond. Cold Spring Harb Symp Quant Biol 2006; 71:217 - 24; http://dx.doi.org/10.1101/sqb.2006.71.011; PMID: 17381300
  • Chu C, Qu K, Zhong FL, Artandi SE, Chang HY. Genomic maps of long noncoding RNA occupancy reveal principles of RNA-chromatin interactions. Mol Cell 2011; 44:667 - 78; http://dx.doi.org/10.1016/j.molcel.2011.08.027; PMID: 21963238
  • Simon MD, Wang CI, Kharchenko PV, West JA, Chapman BA, Alekseyenko AA, Borowsky ML, Kuroda MI, Kingston RE. The genomic binding sites of a noncoding RNA. Proc Natl Acad Sci U S A 2011; 108:20497 - 502; http://dx.doi.org/10.1073/pnas.1113536108; PMID: 22143764
  • Ule J, Jensen KB, Ruggiu M, Mele A, Ule A, Darnell RB. CLIP identifies Nova-regulated RNA networks in the brain. Science 2003; 302:1212 - 5; http://dx.doi.org/10.1126/science.1090095; PMID: 14615540
  • Hafner M, Landthaler M, Burger L, Khorshid M, Hausser J, Berninger P, Rothballer A, Ascano M Jr., Jungkamp AC, Munschauer M, et al. Transcriptome-wide identification of RNA-binding protein and microRNA target sites by PAR-CLIP. Cell 2010; 141:129 - 41; http://dx.doi.org/10.1016/j.cell.2010.03.009; PMID: 20371350
  • Guil S, Soler M, Portela A, Carrère J, Fonalleras E, Gómez A, Villanueva A, Esteller M. Intronic RNAs mediate EZH2 regulation of epigenetic targets. Nat Struct Mol Biol 2012; 19:664 - 70; http://dx.doi.org/10.1038/nsmb.2315; PMID: 22659877
  • Kretz M, Siprashvili Z, Chu C, Webster DE, Zehnder A, Qu K, Lee CS, Flockhart RJ, Groff AF, Chow J, et al. Control of somatic tissue differentiation by the long non-coding RNA TINCR. Nature 2013; 493:231 - 5; http://dx.doi.org/10.1038/nature11661; PMID: 23201690
  • Hsu MT, Coca-Prados M. Electron microscopic evidence for the circular form of RNA in the cytoplasm of eukaryotic cells. Nature 1979; 280:339 - 40; http://dx.doi.org/10.1038/280339a0; PMID: 460409
  • Surono A, Takeshima Y, Wibawa T, Ikezawa M, Nonaka I, Matsuo M. Circular dystrophin RNAs consisting of exons that were skipped by alternative splicing. Hum Mol Genet 1999; 8:493 - 500; http://dx.doi.org/10.1093/hmg/8.3.493; PMID: 9949208
  • Zaphiropoulos PG. Exon skipping and circular RNA formation in transcripts of the human cytochrome P-450 2C18 gene in epidermis and of the rat androgen binding protein gene in testis. Mol Cell Biol 1997; 17:2985 - 93; PMID: 9154796
  • Salzman J, Gawad C, Wang PL, Lacayo N, Brown PO. Circular RNAs are the predominant transcript isoform from hundreds of human genes in diverse cell types. PLoS One 2012; 7:e30733; http://dx.doi.org/10.1371/journal.pone.0030733; PMID: 22319583
  • Li XF, Lytton J. A circularized sodium-calcium exchanger exon 2 transcript. J Biol Chem 1999; 274:8153 - 60; http://dx.doi.org/10.1074/jbc.274.12.8153; PMID: 10075718
  • Hansen TB, Jensen TI, Clausen BH, Bramsen JB, Finsen B, Damgaard CK, Kjems J. Natural RNA circles function as efficient microRNA sponges. Nature 2013; 495:384 - 8; http://dx.doi.org/10.1038/nature11993; PMID: 23446346
  • Memczak S, Jens M, Elefsinioti A, Torti F, Krueger J, Rybak A, Maier L, Mackowiak SD, Gregersen LH, Munschauer M, et al. Circular RNAs are a large class of animal RNAs with regulatory potency. Nature 2013; 495:333 - 8; http://dx.doi.org/10.1038/nature11928; PMID: 23446348
  • Hansen TB, Wiklund ED, Bramsen JB, Villadsen SB, Statham AL, Clark SJ, Kjems J. miRNA-dependent gene silencing involving Ago2-mediated cleavage of a circular antisense RNA. EMBO J 2011; 30:4414 - 22; http://dx.doi.org/10.1038/emboj.2011.359; PMID: 21964070
  • Al-Balool HH, Weber D, Liu Y, Wade M, Guleria K, Nam PL, Clayton J, Rowe W, Coxhead J, Irving J, et al. Post-transcriptional exon shuffling events in humans can be evolutionarily conserved and abundant. Genome Res 2011; 21:1788 - 99; http://dx.doi.org/10.1101/gr.116442.110; PMID: 21948523
  • Squires JE, Patel HR, Nousch M, Sibbritt T, Humphreys DT, Parker BJ, Suter CM, Preiss T. Widespread occurrence of 5-methylcytosine in human coding and non-coding RNA. Nucleic Acids Res 2012; 40:5023 - 33; http://dx.doi.org/10.1093/nar/gks144; PMID: 22344696
  • Motorin Y, Helm M. RNA nucleotide methylation. Wiley Interdiscip Rev RNA 2011; 2:611 - 31; http://dx.doi.org/10.1002/wrna.79; PMID: 21823225
  • Underwood JG, Uzilov AV, Katzman S, Onodera CS, Mainzer JE, Mathews DH, Lowe TM, Salama SR, Haussler D. FragSeq: transcriptome-wide RNA structure probing using high-throughput sequencing. Nat Methods 2010; 7:995 - 1001; http://dx.doi.org/10.1038/nmeth.1529; PMID: 21057495
  • Westhof E, Romby P. The RNA structurome: high-throughput probing. Nat Methods 2010; 7:965 - 7; http://dx.doi.org/10.1038/nmeth1210-965; PMID: 21116245
  • Benhamed M, Herbig U, Ye T, Dejean A, Bischof O. Senescence is an endogenous trigger for microRNA-directed transcriptional gene silencing in human cells. Nat Cell Biol 2012; 14:266 - 75; http://dx.doi.org/10.1038/ncb2443; PMID: 22366686
  • Morris KV, Chan SW, Jacobsen SE, Looney DJ. Small interfering RNA-induced transcriptional gene silencing in human cells. Science 2004; 305:1289 - 92; http://dx.doi.org/10.1126/science.1101372; PMID: 15297624
  • Huang XA, Yin H, Sweeney S, Raha D, Snyder M, Lin H. A major epigenetic programming mechanism guided by piRNAs. Dev Cell 2013; 24:502 - 16; http://dx.doi.org/10.1016/j.devcel.2013.01.023; PMID: 23434410
  • Schubert T, Pusch MC, Diermeier S, Benes V, Kremmer E, Imhof A, Längst G. Df31 protein and snoRNAs maintain accessible higher-order structures of chromatin. Mol Cell 2012; 48:434 - 44; http://dx.doi.org/10.1016/j.molcel.2012.08.021; PMID: 23022379
  • Han J, Kim D, Morris KV. Promoter-associated RNA is required for RNA-directed transcriptional gene silencing in human cells. Proc Natl Acad Sci U S A 2007; 104:12422 - 7; http://dx.doi.org/10.1073/pnas.0701635104; PMID: 17640892
  • Melo CA, Drost J, Wijchers PJ, van de Werken H, de Wit E, Oude Vrielink JA, Elkon R, Melo SA, Léveillé N, Kalluri R, et al. eRNAs are required for p53-dependent enhancer activity and gene transcription. Mol Cell 2013; 49:524 - 35; http://dx.doi.org/10.1016/j.molcel.2012.11.021; PMID: 23273978
  • Johnsson P, Ackley A, Vidarsdottir L, Lui WO, Corcoran M, Grandér D, Morris KV. A pseudogene long-noncoding-RNA network regulates PTEN transcription and translation in human cells. Nat Struct Mol Biol 2013; 20:440 - 6; http://dx.doi.org/10.1038/nsmb.2516; PMID: 23435381
  • Hawkins PG, Morris KV. Transcriptional regulation of Oct4 by a long non-coding RNA antisense to Oct4-pseudogene 5. Transcription 2010; 1:165 - 75; http://dx.doi.org/10.4161/trns.1.3.13332; PMID: 21151833
  • Bond AM, Vangompel MJ, Sametsky EA, Clark MF, Savage JC, Disterhoft JF, Kohtz JD. Balanced gene regulation by an embryonic brain ncRNA is critical for adult hippocampal GABA circuitry. Nat Neurosci 2009; 12:1020 - 7; http://dx.doi.org/10.1038/nn.2371; PMID: 19620975
  • Horard B, Gilson E. Telomeric RNA enters the game. Nat Cell Biol 2008; 10:113 - 5; http://dx.doi.org/10.1038/ncb0208-113; PMID: 18246034
  • Haeger P, Cuevas R, Forray MI, Rojas R, Daza C, Rivadeneira J, Gysling K. Natural expression of immature Ucn antisense RNA in the rat brain. Evidence favoring bidirectional transcription of the Ucn gene locus. Brain Res Mol Brain Res 2005; 139:115 - 28; http://dx.doi.org/10.1016/j.molbrainres.2005.05.024; PMID: 15979199
  • Ishii N, Ozaki K, Sato H, Mizuno H, Saito S, Takahashi A, Miyamoto Y, Ikegawa S, Kamatani N, Hori M, et al. Identification of a novel non-coding RNA, MIAT, that confers risk of myocardial infarction. J Hum Genet 2006; 51:1087 - 99; http://dx.doi.org/10.1007/s10038-006-0070-9; PMID: 17066261
  • Roeszler KN, Itman C, Sinclair AH, Smith CA. The long non-coding RNA, MHM, plays a role in chicken embryonic development, including gonadogenesis. Dev Biol 2012; 366:317 - 26; http://dx.doi.org/10.1016/j.ydbio.2012.03.025; PMID: 22546690
  • Tran VG, Court F, Duputié A, Antoine E, Aptel N, Milligan L, Carbonell F, Lelay-Taha MN, Piette J, Weber M, et al. H19 antisense RNA can up-regulate Igf2 transcription by activation of a novel promoter in mouse myoblasts. PLoS One 2012; 7:e37923; http://dx.doi.org/10.1371/journal.pone.0037923; PMID: 22662250
  • Khalil AM, Faghihi MA, Modarresi F, Brothers SP, Wahlestedt C. A novel RNA transcript with antiapoptotic function is silenced in fragile X syndrome. PLoS One 2008; 3:e1486; http://dx.doi.org/10.1371/journal.pone.0001486; PMID: 18213394
  • Sleutels F, Zwart R, Barlow DP. The non-coding Air RNA is required for silencing autosomal imprinted genes. Nature 2002; 415:810 - 3; http://dx.doi.org/10.1038/415810a; PMID: 11845212
  • Ritter O, Luther HP, Haase H, Baltas LG, Baumann G, Schulte HD, Morano I. Expression of atrial myosin light chains but not alpha-myosin heavy chains is correlated in vivo with increased ventricular function in patients with hypertrophic obstructive cardiomyopathy. J Mol Med (Berl) 1999; 77:677 - 85; http://dx.doi.org/10.1007/s001099900030; PMID: 10569205
  • Potts JD, Vincent EB, Runyan RB, Weeks DL. Sense and antisense TGF beta 3 mRNA levels correlate with cardiac valve induction. Dev Dyn 1992; 193:340 - 5; http://dx.doi.org/10.1002/aja.1001930407; PMID: 1511174
  • Robb GB, Carson AR, Tai SC, Fish JE, Singh S, Yamada T, Scherer SW, Nakabayashi K, Marsden PA. Post-transcriptional regulation of endothelial nitric-oxide synthase by an overlapping antisense mRNA transcript. J Biol Chem 2004; 279:37982 - 96; http://dx.doi.org/10.1074/jbc.M400271200; PMID: 15234981
  • Friedrichs F, Zugck C, Rauch GJ, Ivandic B, Weichenhan D, Müller-Bardorff M, Meder B, El Mokhtari NE, Regitz-Zagrosek V, Hetzer R, et al. HBEGF, SRA1, and IK: Three cosegregating genes as determinants of cardiomyopathy. Genome Res 2009; 19:395 - 403; http://dx.doi.org/10.1101/gr.076653.108; PMID: 19064678
  • Rodríguez-Campos A, Azorín F. RNA is an integral component of chromatin that contributes to its structural organization. PLoS One 2007; 2:e1182; http://dx.doi.org/10.1371/journal.pone.0001182; PMID: 18000552

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.