ArticlePDF AvailableLiterature Review

Multimodal Long Noncoding RNA Interaction Networks: Control Panels for Cell Fate Specification

Authors:

Abstract and Figures

Lineage specification in early development is the basis for the exquisitely precise body plan of multicellular organisms. It is therefore critical to understand cell fate decisions in early development. Moreover, for regenerative medicine, the accurate specification of cell types to replace damaged/diseased tissue is strongly dependent on identifying determinants of cell identity. Long noncoding RNAs (lncRNAs) have been shown to regulate cellular plasticity, including pluripotency establishment and maintenance, differentiation and development, yet broad phenotypic analysis and the mechanistic basis of their function remains lacking. As components of molecular condensates, lncRNAs interact with almost all classes of cellular biomolecules, including proteins, DNA, mRNAs, and microRNAs. With functions ranging from controlling alternative splicing of mRNAs, to providing scaffolding upon which chromatin modifiers are assembled, it is clear that at least a subset of lncRNAs are far from the transcriptional noise they were once deemed. This review highlights the diversity of lncRNA interactions in the context of cell fate specification, and provides examples of each type of interaction in relevant developmental contexts. Also highlighted are experimental and computational approaches to study lncRNAs.
Content may be subject to copyright.
|REVIEW
Multimodal Long Noncoding RNA Interaction
Networks: Control Panels for Cell Fate Specication
Keriayn N. Smith,*
,1,2
Sarah C. Miller,*
,1,3
Gabriele Varani,
J. Mauro Calabrese,
and Terry Magnuson*
*Department of Genetics and Department of Pharmacology, University of North Carolina, Chapel Hill, North Carolina 27599, and
Department of Chemistry, University of Washington, Seattle, Washington 98195
ORCID IDs: 0000-0002-4351-2765 (K.N.S.); 0000-0003-3470-8798 (S.C.M.); 0000-0001-6642-7144 (G.V.); 0000-0002-1213-2540 (J.M.C.);
0000-0002-0792-835X (T.M.)
ABSTRACT Lineage specication in early development is the basis for the exquisitely precise body plan of multicellular organisms. It is
therefore critical to understand cell fate decisions in early development. Moreover, for regenerative medicine, the accurate specication
of cell types to replace damaged/diseased tissue is strongly dependent on identifying determinants of cell identity. Long noncoding
RNAs (lncRNAs) have been shown to regulate cellular plasticity, including pluripotency establishment and maintenance, differentiation
and development, yet broad phenotypic analysis and the mechanistic basis of their function remains lacking. As components of
molecular condensates, lncRNAs interact with almost all classes of cellular biomolecules, including proteins, DNA, mRNAs, and
microRNAs. With functions ranging from controlling alternative splicing of mRNAs, to providing scaffolding upon which chromatin
modiers are assembled, it is clear that at least a subset of lncRNAs are far from the transcriptional noise they were once deemed. This
review highlights the diversity of lncRNA interactions in the context of cell fate specication, and provides examples of each type of
interaction in relevant developmental contexts. Also highlighted are experimental and computational approaches to study lncRNAs.
KEYWORDS long noncoding RNAs; miRNAs; competing endogenous RNAs; k-mers; cell fate specication
LINEAGE specication decisions in early development pro-
vide a blueprint of the body plan in multicellular organ-
isms. Model systems such as embryonic stem (ES) cells are
often employed in the study of early cell fate decisions. Un-
derstanding cell fate is also critical for regenerative medicine,
as cell-based approaches pose signicant therapeutic promise.
Toward this end, induced pluripotent stem (iPS) cells, which
display characteristics of ES cells, and can be patient-derived,
have the potential to be differentiated into a myriad of
different cell types.
Understanding determinants of cell fate is critical both for
understanding early development, and to guide lineage com-
mitment of pluripotent stem cells to enable the replacement
of diseased cell types in patients. While central transcrip-
tional regulators of pluripotency including OCT4, SOX2,
and NANOG, which maintain the pluripotent state, and spec-
ication factors such as SOX1, MEOX1, and SOX17 (Kan et al.
2004; Shimoda et al. 2007; Wang et al. 2013) are relatively
well understood, many key cell-fate determinants remain
functionally undened. Importantly, recent developments
in transcriptomics have demonstrated that, although the ma-
jority of the mammalian genome is transcribed, protein coding
sequences amount to ,2% of transcribed genomic sequence
(Dinger et al. 2008; Alexander et al. 2010; Harrow et al. 2012),
with the number of noncoding RNA (ncRNA) genes equaling,
or possibly even outnumbering, protein-coding genes based on
estimates from GENCODE and FANTOM (Hon et al. 2017;
Frankish et al. 2019). Already, certain noncoding transcripts,
including microRNAs (miRNAs) and long noncoding RNAs
(lncRNAs; designated as transcripts .200 nt) have been im-
plicated in cell fate decisions for unspecialized cells, including
pluripotent stem cells; however, the vast majority of ncRNAs
remain understudied.
Using pluripotent cells and their derivatives for illustra-
tions, this review centers on lncRNAs, with a focus on the
Copyright © 2019 by the Genetics Society of America
doi: https://doi.org/10.1534/genetics.119.302661
Manuscript received September 11, 2019; accepted for publication October 3, 2019;
published Early Online October 16, 2019.
1
These authors contributed equally to this work.
2
Corresponding author: Department of Genetics, University of North Carolina, Room
5028 Genetic Medicine Bldg. CB#7264, 120 Mason Farm Road, Chapel Hill, NC
27599. E-mail: kns@email.unc.edu
3
Present address: Johns Hopkins University School of Medicine, Baltimore, MD
21205.
Genetics, Vol. 213, 10931110 December 2019 1093
Downloaded from https://academic.oup.com/genetics/article/213/4/1093/5930620 by guest on 11 May 2023
multimodal interactions through which they regulate cell fate
specication. These interactions identify lncRNAs as impor-
tant factors in early developmental processes and suggest that
they should be considered in the design of regenerative
medicine strategies.
LncRNAs: Interactions as Functional Determinants
The presence and number of lncRNAs appear to correlate
with organismal complexity, and their expression patterns
show subcellular, cellular, and tissue specicity, which sug-
gests context-dependent roles, particularly in the determina-
tion of cell fate (Mattick 2001). Different classes of lncRNAs
are dened based on transcription direction and location (for
example: sense, bidirectional, antisense, intronic, intergenic;
Figure 1) relative to other genes (Mattick and Rinn 2015).
While this location classication might be suggestive of
mechanism, lncRNA genomic location does not always
strictly dictate function (Mattick and Rinn 2015; Quinn and
Chang 2016). On the other hand, intermolecular interactions
with other RNAs, proteins and chromatin have revealed
emerging functional themes, and demonstrated the far-
reaching regulatory potential of lncRNAs. Here, we examine
the implication of these interactions in cell fate determina-
tion and early developmental processes.
Much of the function of lncRNAs depend on their ability
to base pair to other RNAs or DNA through conventional
or Hoogsteen base pairing, to form complex intramolecular
and intermolecular secondary and higher order structures
(Mercer and Mattick 2013). The structures formed by
lncRNAs regulate and direct interaction with RNA-binding
proteins (RBPs) to regulate, negatively or positively, their
cellular targets. These proteins are central to lncRNA mech-
anisms of action and regulation of their function (Rinn and
Ule 2014).
Localization of a lncRNA transcript can be suggestive of
its functional role and contribution to gene regulation. While
cytoplasmic lncRNAs tend to function post-transcriptionally,
many nuclear lncRNAs regulate gene expression at the tran-
scriptional level (Rinn and Chang 2012; Mercer and Mattick
2013). Within the nucleus, expression of a gene requires
chromatin decompaction, particularly in heterochromatic re-
gions. The compaction state is determined by chemical mod-
ications of nucleosomal histone proteins, controlled by
histone-modifying enzymes. LncRNAs have been shown to
interact with chromatin modiers (readers, writers, erasers)
and remodelers to facilitate changes in the chromatins bio-
chemical and accessibility landscape at specic gene loci
through both cis- and trans-acting mechanisms (Figure 2)
(Rinn and Chang 2012).
Additionally, lncRNAs have also been shown to have a
number of post-transcriptional and cytoplasmic functions in
many developmental processes. These include functioning
in mRNA stability and translation regulation through pro-
tein, miRNA, and mRNA interactions (Figure 2) (Batista and
Chang 2013; Yoon et al. 2013; Quinn and Chang 2016).
These interactions provide a basis for lncRNA functional clas-
sication, but can also be targeted to direct cell fate.
Interactions with proteins: chromatin regulation
The plasticity of pluripotent stem cells is related to the high
ratio of euchromatin to heterochromatin (Gaspar-Maia et al.
2011), making more chromatin accessible to transcription
factors, RNA polymerase, and other proteins necessary for
transcription. Pluripotent stem cells also have a high propor-
tion of poised chromatin (Fisher and Fisher 2011), which
facilitates the rapid gene derepression required for lineage
commitment. Extensive binding of lncRNAs to epigenetic reg-
ulators that control chromatin accessibility denes one cate-
gory of lncRNA function (Mercer and Mattick 2013). Studies
using ES cells and other cell types have shown that 30% of
intergenic lncRNAs were bound by at least one epigenetic
regulator (Khalil et al. 2009), indicating widespread impact
of lncRNAs on cell identity at the transcriptional level.
LncRNAs can act in cis by binding to neighboring genes and
facilitating recruitment of chromatin modier/remodelers
to the target locus (Bassett et al. 2014). The act of lncRNA
transcription can also have a cis-regulatory function in gene
expression, and inuence genome organization (Bassett et al.
2014; Engreitz et al. 2016; Melé and Rinn 2016). LncRNAs
can function in trans as well, either by serving as a recruit-
ment or scaffolding factor on which chromatin modifying
proteins assemble, or by modulating the stability of the chro-
matin regulatory protein complex (Rinn and Chang 2012;
Bassett et al. 2014).
Cis-regulatory lncRNAs control expression of neighboring
genes. In the context of cell fate specication, these lncRNA
genes are often located adjacent to key developmental regu-
lators that determine cell fate and organismal development
(Bassett et al. 2014; Engreitz et al. 2016; Melé and Rinn
2016). This mechanism is commonly used for antisense and
divergent lncRNAs that are typically ,5 kb from, and tran-
scribed in the opposite direction relative to, a transcribed
gene. For example, Evx1as and its neighboring protein-
coding gene, Evx1, demonstrate highly correlated expression
in murine ES cells (Luo et al. 2016). Depletion of Evx1as
indicated unidirectional regulation of the protein-coding
neighbor by the lncRNA where Evx1as bound to its own pro-
moter and facilitated binding of Mediator to activate tran-
scription at the locus (Luo et al. 2016). This example
highlights how lncRNAs can act in cis, tethered to their pro-
moter, to modify gene expression near their transcription site.
Another illustration of cis inuence of a lncRNA is exem-
plied by Chaserrs regulation of Chd2a chromatin remod-
eler with roles in cell differentiation in mice (Rom et al.
2019). Chaserrs transcript is produced upstream of the tran-
scription start site of Chd2, where it collaborates with CHD2
protein to repress Chd2s expression in a negative feedback
loop to maintain cellular levels of CHD2 (Rom et al. 2019).
yylncTa member of the divergent subclass of lncRNAs
known as yin yang (yy) lncRNAssupports expression of its
gene neighbor, Brachyury (T) by localizing to its locus during
1094 K. N. Smith et al.
Downloaded from https://academic.oup.com/genetics/article/213/4/1093/5930620 by guest on 11 May 2023
mesoderm commitment in human ES cells (Frank et al.
2019). yylncRNAs are primarily encoded from genomic loci
of key cell-fate regulators, thus mirroring their developmen-
tal expression patterns, and, as a class, they illustrate a broad
mechanism through which lncRNAs safeguard cell-fate deci-
sions (Frank et al. 2019).
A handful of cis-acting lncRNAs are also known to repress
gene expression over long genomic distances. In the most
extreme example, the lncRNA Xist silences gene expression
over the entire 165 million base pair X chromosome early
during the development of female mammals, as part of the
dosage compensation process called X-chromosome Inactiva-
tion (XCI) (recently reviewed by Sahakyan et al. 2018).
Xist induces stable gene silencing through two parallel
pathways. In the rst, Xist silences actively transcribed genes
through an incompletely dened mechanism that involves
the protein SPEN and the RNA element Repeat A, which is
a tandem repeat located at the 59end of Xist. In parallel, and
subsequent to Repeat-A-mediated silencing, Xist induces the
spread of Polycomb Repressive Complexes (PRCs) over tran-
scriptionally inactive chromatin (Nesterova et al. 2019; ˙
Zylicz
et al. 2019). In a mechanistic sense, this spread of PRCs over
the X is likely a major means by which Xist orchestrates stable
silencing that is inherited through subsequent cell divisions
(Wang et al. 2001; Kalantry et al. 2006; Sahakyan et al.
2018). Other cis-repressive lncRNAs that depend on PRCs
for their silencing functions, such as Kcnq1ot1,Airn,
Morrbid, and Haunt, may utilize similar mechanisms to bring
PRCs to chromatin (Regha et al. 2007; Terranova et al. 2008;
Yin et al. 2015; Kotzin et al. 2016; Schertzer et al. 2019).
Indeed, both Kcnq1ot1 and Airn were recently shown to re-
quire the Xist cofactor HNRNPK to induce the spread of PRCs
in mouse trophoblast stem cells (Schertzer et al. 2019).
In addition, the PRCs, particularly PRC2, have been shown
to interact with many RNAs, and the functional consequence
of this interaction has not always been clear. For example, the
PRC2 component SUZ12 has been shown to interact with a
lncRNA to repress a differentiation-inducing transcriptional
program in human ES cells. Here, the lncRNA tsRMST uses
multiple mechanisms, including coregulation with SUZ12
and NANOG, to block expression of lineage specication
genes and impede WNT5A-induced epithelial-mesenchymal
transition (Yu and Kuo 2016). SUZ12 and the central pluri-
potency regulator SOX2 also interacts with lncRNA_ES1 and
lncRNA_ES2 to contribute to pluripotency maintenance (Ng
et al. 2012) through unclear mechanisms.
Other lncRNAs, at least partially through their interaction
with PRC2, have been implicated in processes supporting
lineage commitment. For example, the lncRNA Braveheart
interacts with PRC2, and, perhaps in part due to a conse-
quence of this interaction, Braveheart directs murine plurip-
otent cells to a cardiac fate by moderating a mesoderm and
cardiac-specic transcription factor network (Klattenhoff
et al. 2013). Nevertheless, through a specic structured ele-
ment, Braveheart interacts with the CNBP/ZNF9 nucleic acid
binding protein, and at least a portion of Braveheart function
can be ascribed to the CNBP/ZNF9 interaction (Xue et al.
2016). Moreover, and surprisingly, even though depletion
of Braveheart results in myogenic defects through its control
of central cardiomyogenic regulators including Mesp1,
Hand1,Nkx2.5, and Tbx20, and general loss of sarcomere
gene expression, Braveheart null mice were grossly pheno-
typically normal (Han et al. 2018). Conversely, genetic abla-
tion of the lncRNA Fendrr, which has also been shown to
interact with PRC2, results in mouse embryonic lethality at
around E13.75 due to myocardial defects (Grote et al. 2013).
Here, it has been proposed that Fendrrsinteraction with both
PRC2 and Trithorax Group/MLL complexes modulates chro-
matin signatures in control of lateral mesoderm differentia-
tion (Grote et al. 2013). PRC2 function has also been
reported to be regulated in trans, in mouse ES cells and hu-
man iPS cells, by the relatively abundant lncRNAs Rian,Mirg,
and Meg3/Gtl2, which are produced from an imprinted clus-
ter (Kaneko et al. 2014). Examples of these lncRNAs with
clear roles for repressing transcription in cis also exist
(Sanli et al. 2018). Additionally, many lncRNAs produced
from the developmentally important Hox gene clusters also
bind PRC2, the most notable of which may be the lncRNA
HOTAIR. The extent to which lncRNA/PRC2 interactions in
the Hox clusters contribute to gene regulation in mammalian
development, whether the regulation occurs in cis or in trans,
and what the mechanisms are, however, remain unclear (Li
et al. 2013, 2016; Tsai et al. 2010; Amândio et al. 2016;
Selleri et al. 2016; Portoso et al. 2017).
Figure 1 General principles illus-
trating lncRNA subtypes and ge-
nomic origin. LncRNAs may
originate from various regions in
the genome, including proximal,
distal, and overlapping, with re-
spect to protein coding genes.
Sense and antisense lncRNAs
may, or may not, fully overlap
with protein coding genes. Diver-
gent and intergenic lncRNAs are
arbitrarily distinguished based on
distance from the nearest protein
coding gene.
Multimodal lncRNA Interactions 1095
Downloaded from https://academic.oup.com/genetics/article/213/4/1093/5930620 by guest on 11 May 2023
Importantly, both crosslinking immunoprecipitation (CLIP)
studies of PRC2 as well as those that have studied the RNA-
binding properties of PRC2 in vitro have found that PRC2 binds
RNA with little sequence specicity and nanomolar afnity
(Kaneko et al. 2013, 2014; Davidovich et al. 2015; Wang
et al. 2017). Collectively, these studies suggest that one func-
tion of lncRNAs, and perhaps chromatin-bound RNAs in gen-
eral, is to tether PRC2 to transcriptionally active regions of
chromatin. This tethering may keep PRC2 in a poised state,
in close proximity to future target genes, where it can initiate
stable gene silencing upon receipt of the appropriate cues
(Kaneko et al. 2013, 2014; Davidovich et al. 2015; Wang
et al. 2017). High-afnity and nonspecic interactions with
RNA may also govern PRC1 function in an analogous fashion
(Bernstein et al. 2006; Bonasio et al. 2014).
LncRNAshave beenshown tointeract with a wide variety of
chromatin modulatory factors. In addition to PRCs, these
include histone methylases (Hendrickson et al. 2016). The
H3K4 methylase MLL family is necessary for activating the
expression of certain genes (Yang et al. 2014). WDR5 is a
Figure 2 Schematic illustration of the different modes of action for lncRNAs. Localization of lncRNAs to the nucleus or cytoplasm can dictate different
mechanisms of action. Based on their ability to bind to DNA and interact with proteins, lncRNAs can guide transcription regulators and epigenetic
modulators (A); act as scaffolds to assemble chromatin regulatory factors (B); titrate away regulators of transcription by acting as decoys (C); regulate
domain- or chromosome-wide chromatin state to regulate transcriptional output (D); act as ceRNAs to capture regulatory factors such as miRNAs away
from target genes (E); contribute to the stabilization of protein complexes, proteins, and mRNAs (F); and inuence alternative splicing (G).
1096 K. N. Smith et al.
Downloaded from https://academic.oup.com/genetics/article/213/4/1093/5930620 by guest on 11 May 2023
protein-subunit of MLL, recruiting the complex to target sites
for activation (Yang et al. 2014). WDR5 engages with several
lncRNAs that have been implicated in the self-renewal of ES
cells (Yang et al. 2014). When the RNA-binding site of WDR5
was mutated in mouse ES cells, rendering it unable to bind
lncRNAs, WDR5 was signicantly less stable (Yang et al.
2014). This loss of stability resulted in a severe decrease in
H3K4me3 marks on the promoters of pluripotency-related
genes, and a loss of the ES cell state in 50% of colonies
(Yang et al. 2014).
Interactions between lncRNAs and chromatin readers are
exemplied by the interaction between DIGIT and BRD3 in
the regulation of endoderm differentiation (Daneshvar et al.
2016, 2019). Here, DIGIT supports BRD3 recruitment to
H3K18ac at regions in the genome enriched during endo-
derm differentiation in human ES cells (Daneshvar et al.
2019).
In addition to chromatin remodelers and modiers, anal-
ysis of lncRNA interactomes has also identied transcription
factors as key players in lncRNA function. Panct1a nuclear-
functioning sense lncRNA transcribed from an intron of the
gene coding for its protein interacting partner, TOBF1
exemplies this type of trans interaction in mouse ES cells
(Chakraborty et al. 2017). Panct1 was shown to facilitate the
binding of TOBF1 to pluripotency marker promoters by way
of sequence-directed binding to Oct-Sox motifs, thus recruit-
ing transcription factors such as Oct4 to promote target ex-
pression (Chakraborty et al. 2017). This interaction illustrates
the role of a lncRNA in efcient binding of pluripotency-
associated transcription factors to their target sites without
direct interaction (Chakraborty et al. 2017).
The aforementioned examples demonstrate the breadth of
lncRNAprotein interactions that inuence transcription in
developmental processes, and on which cell specication is
at least partly dependent. Many of the described lncRNA
interactions involve key transcription factors and epigenetic
regulators that affect developmental progression. Modula-
tion of specic lncRNAs could therefore be a viable avenue
for specic regulation of target expression in developmental
contexts.
Interactions with proteins: stability and sequestration
LncRNAs enhance or repress protein function through a va-
riety of mechanisms including sequestration, binding support,
and degradation, as in the aforementioned case of WDR5 and
its reliance on lncRNAs for stability (Yang et al. 2014). Con-
versely, in other contexts, lncRNAs have been shown to be
dependent on their protein partners for stability to carry out
their functions, and regulate their half-life. This is the case for
lncR492a noncoding transcript that inhibits neural differ-
entiation in mouse ES cells (Winzi et al. 2018). Knockdown of
lncR492 resulted in increased expression of neural markers
such as Pax6 and Nestin during differentiation. Proteomic
analysis indicated that lncR492 directly interacts with HuR
a mRNA binding protein withfunctions inthe (de)stabilization
of mRNA transcripts. Overexpression and knockdown of HuR
moderated the expression of lncR492, increasing and reducing
the prevalence of the lncRNA, respectivelya pattern that
suggests HuR supports the stability of the lncR492 transcript.
Finally, both lncR492 and HuR positively inuence WNT sig-
naling, which has a known inhibitory effect on neural differ-
entiation (Haegele et al. 2003), outlining the axis by which
lncR492 and HUR function (Winzi et al. 2018).
LncRNAs can impede protein function through binding and
sequestration. In the context of pluripotency, the chromatin
mark H3K56 acetylation activates core pluripotency-related
genes and is required for the maintenance of the undifferen-
tiated ES cell state. SIRT6 is a chromatin-binding protein that
removes this chromatin modication and functionally re-
presses pluripotency-related genes to promote exit from the
stem cell state (Etchegaray et al. 2015). LncPRESS1 functions
as a molecular decoy for SIRT6, sequestering the protein,
which binds to the 39-end of the lncRNA in human ES cells.
This interaction in the nucleus prevents SIRT6 from binding
promoters of pluripotency-related genes and repressing tran-
scription via deacetylation (Jain et al. 2016). Another regu-
latory interaction in this network is illustrated by P53, which
antagonizes lncPRESS1, freeing SIRT6 to further repress plu-
ripotency markers in human ES cells (Jain et al. 2016).
Emerging data indicate that regulatory lncRNAprotein
interactions occur in specialized microenvironments that dis-
play characteristics of phase-separated particles (Hnisz et al.
2017; Daneshvar et al. 2019). These liquid-like condensates
are able to exchange molecules dynamically with their sur-
roundings (Bergeron-Sandoval et al. 2016; Boeynaems et al.
2018; Lu et al. 2018). The preceding interactions demon-
strate the role lncRNA-protein interactions play in integral
processes throughout differentiation and development. Such
interactions can be investigated as points of manipulation for
control of differentiation processes, especially when they lo-
calize to distinct microdroplets, and when the mechanisms
for the interactions have been clearly dened.
Interactions with RNA
Similar to the prevalence of lncRNAprotein interactions,
lncRNARNA interactions are prolic and have widespread
effects on cell identity. Interestingly, lncRNAs interplay with
multiple other RNA types, from mRNA to other ncRNA, in-
cluding miRNAs and circular RNAs. Through different mech-
anisms, these lncRNA-RNA interactions can affect lncRNA
function through repressing or supporting downstream
targets.
Canonical functions of mRNAs are determined by their
availability and potential to be translated. In addition to long-
characterized protein factors, the half-life of a mRNA is de-
termined by various co- and post-transcriptional regulatory
factors, including lncRNAs. LncRNA interaction with mRNAs
or mRNA-regulatory factors can stabilize or facilitate the
degradation of the mRNA molecules, increasing or decreasing
translational output (Faghihi et al. 2008, 2010; Gong and
Maquat 2011). For example, Sirt1-AS interacts with Sirt1
mRNA to promote its stability, thereby inhibiting myogenic
Multimodal lncRNA Interactions 1097
Downloaded from https://academic.oup.com/genetics/article/213/4/1093/5930620 by guest on 11 May 2023
differentiation in favor of myoblast proliferation in mice
(Wang et al. 2016).
LncRNAs have also been shown to alter the translation
output of mRNAs by affecting alternative splicing. A prime
example of this mechanism of action is the interaction of
Zeb2-NATan antisense lncRNAwith its sense transcript,
protein-coding Zeb2.Zeb2-NAT was demonstrated to bind
Zeb2s59UTR, which contains an intron where the internal
ribosome entry site for the ZEB2 protein resides (Beltran et al.
2008). Without protection of the rst intron by lncRNA bind-
ing, ZEB2 protein levels are signicantly diminished. In mice,
Zeb2-NAT and ZEB2 prevent broblasts from being effectively
reprogrammed to the pluripotent state, possibly by support-
ing senescence due to E-cadherin downregulation (Beltran
et al. 2008; Bernardes de Jesus et al. 2018). Conversely, un-
der conditions of decreased Zeb2-NAT expression, the mouse
broblasts readily transitioned to ES cell-like cells in media
conditions that support the pluripotent state (Bernardes de
Jesus et al. 2018). Further, mouse ES cells in Zeb2-NAT knock-
down conditions were able to maintain the pluripotent state
in differentiation-inducing contexts (Bernardes de Jesus et al.
2018).
Perhaps even more impactful, based on their numerous
targets, is the inuence of lncRNAs on miRNAs. These small
ncRNAs (22 nt long) are key post-transcriptional regula-
tory factors that inuence target transcript translational re-
pression and/or degradation (Heinrich and Dimmeler 2012).
miRNA function has been implicated in the establishment
and maintenance of ES cell pluripotency and differentiation
(Heinrich and Dimmeler 2012). Generally, miRNAs and as-
sociated proteins assemble to form the RNA-induced silenc-
ing complex (RISC), in which the miRNA serves as a guide to
target specic mRNAs, which are degraded in proportion to
the degree of complementarity with the miRNA (Gregory
et al. 2005). LncRNAs can affect the efciency of these
processes as well, since they can dictate the abundance
of individual miRNAs by supporting miRNA stability or by
causing their degradation, for example through template-
mediated degradation (Fuchs Wightman et al. 2018). Addi-
tionally, lncRNAs can alter miRNA function by behaving as a
sponge or competing endogenous RNA (ceRNA) that seques-
ters the miRNA, thus preventing degradation of the miRNA
target genes to support or promote exit from the stem cell
state (Liu et al. 2014, see Table 1).
Linc-RoR (Regulator of Reprogramming) exemplies the
ceRNA mechanism in the context of reprogramming and the
maintenance of pluripotency. Deviation from precise linc-RoR
levels results in the differentiation of human ES cells to me-
soderm and/or endoderm if its levels are depleted, or the
inability of cells to properly differentiate if linc-RoR levels
are elevated. Linc-ROR was shown to be a ceRNA for miR-
145-5p, suppressing the miRNAs negative regulation of stem
cell regulatory factors such as OCT4 and SOX2 (Loewer et al.
2010; Wang et al. 2013).
While linc-ROR guides reprogramming, lncRNA-1064 was
shown to support neural differentiation (Weng et al. 2018).
Knockdown of lncRNA-1064 led to a decrease in neural line-
age markers and reduced neural differentiation of mouse ES
cells in vitro and in vivo in a murine teratoma model (Weng
et al. 2018). Mechanistic analysis revealed lncRNA-1064 con-
tains multiple miRNA target sites, with the most favorable
being for mir-200c (Weng et al. 2018). lncRNA-1064-medi-
ated sequestration of miR-200c transcripts enables ZEB1/2 to
reach their gene targets to signal for neural differentiation
(Weng et al. 2018). This interaction is similar to that of
lncRNA AK048794, which functions as a ceRNA with miR-
592 (Zhou et al. 2016). In mouse ES cells, miR-592 was found
to bind the 39UTR of FAM91A1, reducing its protein levels
and those of pluripotency regulators Oct4,Sox2, and Nanog,
although the downstream mechanism is less clear. Alto-
gether, these studies describe instances of lncRNAs function-
ing as ceRNAs, sequestering miRNAs, and preventing
degradation of miRNA targets to either support or undermine
pluripotency. Often, lncRNAs acting as ceRNAs can be a part
of a multi-component network, such as the case of AK048794
(Zhou et al. 2016).
Because of their prevalence and well-delineated mecha-
nism, lncRNA control of miRNA levels and availability could
be harnessed to modulate miRNA levels in therapeutic con-
texts,particularly forcases wherea wholesale loss of the target
miRNA would be phenotypically disadvantageous (Kleaveland
et al. 2018). Effective ceRNA activity would require a minimum
threshold level for the lncRNA. Therefore, one important con-
sideration pertaining to ceRNA/sponge function may be the
relative abundance of the lncRNA, miRNA, and mRNA target.
This is because the number of miRNA and target molecules
would typically outnumber that of a lncRNA (Palazzo and Lee
2015). Another key consideration is whether, and how, RNA
structure changes in response to the binding of proteins, other
RNAs, or even small molecule metabolites to regulate the activ-
ity of ceRNA, as this could inuence access to specicsites/
sequences on the RNA. This is an aspect of lncRNA regulation
that has escaped sustained attention so far, but is likely to be
an important aspect of ceRNA, and, more generally, lncRNA
regulation.
Multimodal interactions
Since lncRNA functions can vary widely, it could be expected
that their functions are multifaceted (Figure 3) and not nec-
essarily dependent on a single mechanism even in a single
cell type. Previously characterized as a lncRNA, Tug1, is pro-
duced from a highly conserved locus (Young et al. 2005), and
is involved in many developmental processes including pho-
toreceptor specication, axonal differentiation, and osteo-
genesis regulation, where some functions have been found
to be mediated by protein interactions with key cell-fate reg-
ulators such as LIN28 (Young et al. 2005; Guo et al. 2018; He
et al. 2018). While knockout mice are viable, they display
sterility with complete penetrance due to defects in sper-
matogenesis (Lewandowski et al. 2019). Intriguingly, in ad-
dition to having two distinct noncoding functions, one of
Tug1s functions is dependent on an encoded micropeptide.
1098 K. N. Smith et al.
Downloaded from https://academic.oup.com/genetics/article/213/4/1093/5930620 by guest on 11 May 2023
Table 1 Paradigmatic modes of lncRNA interaction and functional output using ES cells, differentiation and developmental processes as
models for cell fate specication
lncRNA Mode of action Interactor(s) Result Reference
Airn Transcriptional Igf2r Silences Igf2r cluster to guide development Santoro et al. (2013)
AK028326 Transcriptional Oct4 Positively regulates Oct-4 to promote self-renewal Sheik Mohamed et al.
(2010)
Apela RNA Transcriptional p53, hnRNPL Interacts with hnRNPL to repress p53-induced apoptosis M. Li et al. (2015)
Braveheart Transcriptional PRC2 (Suz12), MesP1 Regulates cardiac lineage commitment in ES cells Klattenhoff et al. (2013)
Chaserr Transcriptional CHD2 Inuences cell differentiation through regulation of Chd2 Rom et al. (2019)
Deanr1 Transcriptional SMAD2/3, FoxA2 Recruits SMAD2/3 to the FOXA2 promoter to promote
endoderm differentiation
Jiang et al. (2015)
DIGIT Transcriptional GSC, BRD3 Supports endoderm differentiation Daneshvar et al. (2016)
Evx1as Transcriptional Evx1 Promotes Evx1 expression to regulate mesendodermal dif-
ferentiation
Luo et al. (2016)
Fendrr Transcriptional PRC2, Trithorax
group
Targets promoters for proper heart and body wall formation Grote et al. (2013)
FIRRE Transcriptional CTCF Preservation of silencing of inactive X chromosome Yang et al. (2015)
Haunt Transcriptional HOXA Inhibits HOXA expression and ES cell differentiation, whereas
the Haunt locus is an enhancer for HOXA
Yin et al. (2015)
HERVH Transcriptional Oct4 Recruits Oct4 to maintain pluripotency Lu et al. (2014)
HOTTIP Transcriptional WDR5 Binds WDR5 to activate developmental regulators Wang et al. (2011)
LncPress1 Transcriptional SIRT6 Binds SIRT6 to promote expression of pluripotency-related
genes
Jain et al. (2016)
lncR492 Transcriptional HuR Associates with HuR to promote pluripotency Winzi et al. (2018)
lncRNA_ES1 Transcriptional SUZ12, SOX2 Interacts with SUZ12 and SOX2 to prevent differentiation Ng et al. (2012)
lncRNA_ES2 Transcriptional SUZ12, SOX2 Interacts with SUZ12 and SOX2 to prevent differentiation Ng et al. (2012)
Meg3/Gtl2 Transcriptional PRC2 (JARID2) Regulates recruitment of PCR2 to chromatin in iPS cells Kaneko et al. (2014)
Panct1 Transcriptional TOBF1 Regulates recruitment of TOBF1 to Oct-Sox Motifs to support
the pluripotent state
Chakraborty et al. (2017)
pRNA Transcriptional TIP5, TTF1 Interaction with TIP5, TTF1 contributes to heterochromatin
formation required for differentiation
Savi´
cet al. (2014)
RMST Transcriptional SOX2 Associates with SOX2 to regulate neural differentiation Ng et al. (2013)
tsRMST Transcriptional PRC2, NANOG, WNT Associates with PRC2, NANOG, WNT to repress differentia-
tion
Yu and Kuo (2016)
TUNA
(megamind)
Transcriptional NCL, PTBP1,
hnRNP-K
Associates with specied RBPs to
activate pluripotency genes and
neural differentiation genes
Lin et al. (2014)
yyT Transcriptional Brachyury Regulates Brachyury (T) in mesoderm specication Frank et al. (2019)
AK048794 ceRNA miR-592 Sponges miR-592 to support pluripotency Zhou et al. (2016)
Cyrano ceRNA mir-7 Inhibit mir-7 to support self-renewal Smith et al. (2017)
H19 ceRNA let-7 microRNAs Modulates let-7 to impede muscle differentiation Kallen et al. (2013)
HPAT5 ceRNA let-7 microRNAs Modulates let-7 to promote pluripotency Durruthy-Durruthy et al.
(2016)
linc-ROR ceRNA miR-145 Inhibits miR-145 suppression of self-renewal genes Wang et al. (2013)
lncRNA-1064 ceRNA miR-200c Inhibits miR-200c to regulate neural differentiation Weng et al. (2018)
MD1 ceRNA miR-133, miR-135 Supports differentiation by inhibiting miR-133, miR-135 Cesana et al. (2011)
T-UCstem1 ceRNA miR-9, PRC2 Maintains self-renewal by modulating miR-9 and PRC2 Fiorenzano et al. (2018)
HOTAIR Scaffold PRC2, LSD1 Originally proposed to coordinate PRC2 and LSD1 complexes
for proper embryonic development, although contested in
the more recent literature
Tsai et al. (2010), Li et al.
(2013), Amândio et al.
(2016), Li et al. (2016),
Selleri et al. (2016),
Portoso et al. (2017)
XIST Scaffold X chromosome,
PRC2
Inactivates X chromosome for dosage control (Brown et al. 1991)
Cyrano Multimodal/Other Stat3, signaling net-
work
Supports ES cell maintenance Smith et al. (2018)
Tug1 Multimodal/Other Lin28A, Fragile X
mental retardation
protein
Regulates various differentiation processes including osteo-
genesis, neuronal differentiation and spermatogenesis
Young et al. (2005), Guo
et al. (2018), 1; He
et al. (2018), 1;
Lewandowski et al.
(2019)
Zeb2-NAT Other Zeb2 Facilitates Zeb2 processing to regulate EMT and pluripotency Beltran et al. (2008);
Bernardes de Jesus
et al. (2018)
Multimodal lncRNA Interactions 1099
Downloaded from https://academic.oup.com/genetics/article/213/4/1093/5930620 by guest on 11 May 2023
Specically, the Tug1 locus represses downstream genes in a
cis-manner, while Tug1 RNA itself regulates genes that then
are dysregulated upon knockout. Additionally, the 59con-
served region of the Tug1 gene encodes a peptide, TUG1-
BOAT that inuences mitochondrial membrane potential
(Lewandowski et al. 2019).
The lncRNA Cyrano (OIP5-AS1,1700020I14Rik,linc-oip5,
Oip5os1) is another illustration of multimodal function.
While lncRNAs generally display limited sequence conserva-
tion even between closely related species, a 300 nt region
shows very high conservation in tetrapods, with 100 nucle-
otides conserved between zebrash and humans (Ulitsky
et al. 2011). Uniquely, there is a sequence stretch that has
nearly perfectly complementarity to miR-7 in all tetrapods
examined, which regulates miR-7 degradation (Kleaveland
et al. 2018). The conserved sequence folds into a conserved
secondary structure within which embeds the mir-7 binding
sequence and partially masks it by base pairing (G. Varani
unpublished results). How the RNA structure affects protein
recruitment and degradation remains unclear, however, and
it might be that the structure itself is incidental to miR-7
degradation. Still, it provides tantalizing suggestions that
secondary and higher order structure might be essential com-
ponents of lncRNA regulatory activities.
Cyrano shows rare maternal and zygotic expression during
early development (Karlic et al. 2017), and in various mam-
malian cells it is a proliferation regulator (Smith et al. 2017;
Deng et al. 2018; X. Liu et al. 2018; Naemura et al. 2018).
Using proteomic analyses, Cyrano was found to interact with
a developmental/signaling protein network, through which
it partially supports mouse ES cell characteristics (Smith et al.
2018). Further, Cyrano inhibition of mir-7, which targets the
pluripotency regulator Nanog, as well as Itga9, contributes to
regulation of stemness and cellular adhesion (Smith et al.
2017).
Cyrano has also been shown to exist in a multi-component
RNA network with the circular RNA Cdr1as and several
miRNAs, where it maintains appropriate Cdr1as levels in
Figure 3 By engaging in diverse
interaction patterns, a single
lncRNA can impact multiple cellu-
lar processes. LncRNAs harness
different mechanisms and access
multiple networks of interacting
partners in a context-specic
manner. (A)TUG1 can (1) regulate
neighboring genes in cis, (2) func-
tion in trans to regulate target
genes, (3) be translated into a
micropeptide that regulates mito-
chondrial membrane potential,
and (4) interact with proteins such
as Lin28A to regulate various cell
fate decisions. (B) Cyranos multi-
faceted functions are illustrated
by (1) its ability to inhibit miR-7-
mediated repression of Itga9 and
Nanog, (2) interact with proteins
to support maintenance of the
self-renewing pluripotent state,
and (3) function in a multi-RNA
regulatory network to impact
Cdr1as expression, and, ultimately,
neuronal activity and neuropsy-
chiatric behavior in mice.
1100 K. N. Smith et al.
Downloaded from https://academic.oup.com/genetics/article/213/4/1093/5930620 by guest on 11 May 2023
the brain to control neuronal activity and neuropsychiatric
behavior in mice (Piwecka et al. 2017; Kleaveland et al.
2018). However, despite displaying strong expression and
signicant molecular and cellular phenotypes, Cyrano is an
example of a lncRNA that does not show an overt develop-
mental phenotype with differing knockout strategies (Han
et al. 2018; Kleaveland et al. 2018), which raises the question
of whether there could be compensatory mechanisms for its
function.
LncRNA Properties that Underlie Their Intermolecular
Interactions
Sequence
A challenge in the lncRNA eld is identifying the function of a
lncRNA based on analysis of its sequence content alone, which
is most easily accessible. This challenge stems from the fact
that most proteins interact with RNA through sequence motifs
that are degenerate and have hidden structural preferences
(Dominguez et al. 2018). Compounding the difculty is that
the order of protein-binding modules within a lncRNA is
likely to be less important than the mere presence of the
binding modules themselves. Thus, two lncRNAs may encode
identical functions through different sequence solutions.
An additional obstacle lies in the fact that existing se-
quence alignment algorithms, which, in large part, have been
designed to detect linear sequence relationships between
evolutionarily related nucleic acid or protein species, often
fail to detect signicant homology between lncRNAs (Altschul
et al. 1990; Rice et al. 2000; Edgar and Batzoglou 2006;
Wheeler and Eddy 2013). In order to address this problem,
Kirk and colleagues recently developed a method called
SEEKR (Sequence evaluation through k-mer representation)
to quantify nonlinear sequence similarity between lncRNAs
(Kirk et al. 2018). Rather than evaluating similarity between
lncRNAs based on the extent of linear sequence homology,
SEEKR functions by counting the abundance of all possible
combinations of sequence substrings at a given length, k,
within a lncRNA, and then scaling these abundances by the
extent to which they differ from the mean abundance of each
k-mer in the group of lncRNAs being analyzed. The extent of
nonlinear similarity between lncRNAs as dened by SEEKR
was found to correlate signicantly with lncRNA subcellular
localization and with protein binding, although the ability to
predict either of these two properties from k-mer content
alone was minimal. Using a transgenic assay to monitor the
ability of a lncRNA to induce Xist-like repression, it was
found that k-mer content, but not linear sequence homology,
strongly correlated with the ability of lncRNAs to induce this
type of repression. In a subsequent study, Sprague and col-
leagues found substantial levels of nonlinear sequence simi-
larity between functional domains in Xist, and domains in the
lncRNA Rsx, a marsupial lncRNA that has been proposed to be
a functional analog of Xist that arose through convergent evo-
lution (Grant et al. 2012; Sprague et al. 2019). Collectively,
these data support the notions that different lncRNAs can en-
code similar function through different spatial arrangements
of related, but not necessarily identical, sequence motifs, and
that k-mer based classication provides an approach to detect
such similarities. The weak-to-modest predictive power of
k-mer content in most scenarios hints at ubiquitous and dif-
cult-to-model roles for RNA structure in lncRNA function (Kirk
et al. 2018). Nevertheless, k-mer based classication schemes,
which have been broadly used in other biological contexts
(Blaisdell 1989; Burge et al. 1992; Kari et al. 2015; Lees
et al. 2016; Pandey et al. 2018), represent promising avenues
that may ultimately aid in the functional classication of
lncRNAs from sequence content alone, much in the way that
functional domains can now be routinely identied in proteins
(UniProt Consortium 2015).
Structure
LncRNAs function through sequence-specic interactions
with proteins that recognize stretches of sequence, as well
as with other RNAs or DNAs by base pairing through Watson
Crick or Hoogsteen structures or by forming triple helices.
However, intermolecular recognition is often dependent on,
or regulated by, specic secondary and tertiary structural
features of the lncRNA. Intriguingly, when chemical modi-
cation techniques have been used to probe lncRNA structure,
it has generally revealed high levels of base pairing, more than
in mRNAs and comparable to the ribosome or self-splicing
intronscontexts in which secondary structure is complex
and essential (Somarowthu et al. 2015; Hawkes et al.
2016). Conversely, in cells, RBPs such as hnRNPs (Dreyfuss
et al. 1993) might keep lncRNAs less tightly folded. Further-
more, the observation of secondary structure does not neces-
sarily imply function, especially in the absence of clear
evolutionary conservation through covariation, as observed
for the ribosome. Similarities shared with the ribosome and
RNA enzymes could indicate a lncRNA architecture com-
posed of structured domains, possibly exibly connected,
that establish interactions with other RNAs, chromatin, or
specic protein complexes to bring them within functional
proximity. While this modular structure hypothesis is appeal-
ing because it would provide for intricate functional specic-
ities even in the absence of sequence conservation, it remains
to be investigated at the molecular level.
It also remains to be investigated the extent to which
secondary structures are functional, and whether they co-
alesce to form tertiary and higher order structures and inter-
actions. Thus far, relatively few lncRNAs have been
characterized at the secondary structure level, including
H19 (Hurst and Smith 1999; Juan 2000), Xist (Wutz et al.
2002; Fang et al. 2015; Lu et al. 2016; Smola et al. 2016;
F. Liu et al. 2017), Braveheart (Xue et al. 2016), HOTAIR
(Somarowthu et al. 2015), COOLAIR (Hawkes et al. 2016),
or lincRNA-p21 (Chillón and Pyle 2016) and several others.
A few studies indicate nevertheless that lncRNA structure is
important for regulation, and the principles learned through
these examples might be more broadly applicable to other
Multimodal lncRNA Interactions 1101
Downloaded from https://academic.oup.com/genetics/article/213/4/1093/5930620 by guest on 11 May 2023
lncRNAs. One study showed that regulation of transcription of
the E-cadherin gene is regulated by a sense lncRNA in epithe-
lial cells that is independently transcribed upstream of the
promoter (Pisignano et al. 2017). The structure of this RNA is
controlled by a SNP that modies local RNA secondary struc-
ture and affects loading of epigenetic enzymes that then reg-
ulate the downstream promoter. How common regulation
through conformational switching is remains unclear, but
RNA is structurally malleable and physically well-suited for
this mechanism of gene regulation. Riboswitches, for exam-
ple, are RNA structures that toggle between distinct confor-
mational states upon binding of small molecules and are
widespread in bacterial gene regulation (Mandal and
Breaker 2004). Riboswitch-like mechanisms of regulation
might be present in lncRNAs as well, but their prevalence
and function remain to be investigated. A study indicates
regulation by conformational switching is provided by the
roX lncRNA, which targets the MSL complex to the
Drosophila melanogaster X chromosome as part of the dosage
compensation process that occurs in male ies. MSL is
recruited to roX lncRNA by a conserved stem-loop structure;
once bound by the MSL-component MLE, this stem-loop un-
folds to form an alternate RNA structure that appears to trap
MSL on roX (Ilik et al. 2013, 2017; Quinn et al. 2016). In the
case of Xist, structured regions, but also regions notable for
their absence of structure, likely serve to recruit different
subsets of proteins along the length of the lncRNA (Wutz
et al. 2002; Fang et al. 2015; Smola et al. 2016; F. Liu et al.
2017). Here, toggling between different conformational
states might dene the subset of proteins that associate with
the lncRNA under distinct cellular conditions.
Resource Toolkit for LncRNA Interaction Proling
RNAs are largely dependent on proteins for their produc-
tion, processing, transport, and localization. Based on reagent
availability and ease-of-study, approaches to investigate RNA
protein interactions have historically been protein-centric.
As the diversity and functionality of lncRNAs emerged and
expanded, these tools, including RNA-immunoprecipitation
(RIP), CLIP and its variations, including HITS-CLIP, PAR-
CLIP, iCLIP, and Fast-iCLIP, have begun to reveal the magni-
tude of proteinlncRNA interactions (Ule et al. 2005; Hafner
et al. 2010; König et al. 2010; Flynn et al. 2015; J.-H. Li et al.
2015; Zarnegar et al. 2016). Similarly, efforts are ongoing to
dene the complete repertoire of RNA-binding proteins using
proteomics-based methods such as OOPS, R-DeeP, XRNAX,
and DIF-FRAC, which often incorporate RNA dependency in
their analysis (Mallam et al. 2018; Caudron-Herger et al.
2019; Queiroz et al. 2019; Trendel et al. 2019), and reveal
surprisingly widespread RNA-dependent protein function-
ality, even for well characterized proteins such as CTCF
(Caudron-Herger et al. 2019).
The expanding RNA functionalities highlight the need for
RNA-centered methods to empirically determine binding
partners of lncRNAs (Table 2). New computational resources
that enable queries on previously identied interactors, and
those that allow for prediction of new interacting candidates
have therefore seen remarkable growth in just the last few
years.
Experimental methods
Capture hybridization analysis of RNA targets: Capture
hybridization analysis of RNA targets (CHART) methodology
allows for the identication of chromatin and protein inter-
actors of lncRNAs. First, regions that are accessible for probe-
based isolation are mapped using RNase H-dependent
digestion. DNA oligonucleotides can then be used to isolate
the RNA in an afnity purication step, followed by high-
throughput sequencing to determine DNA segments con-
tacted by the lncRNA, or mass spectrometry, to determine
the protein interactome of the candidate lncRNA (Simon et al.
2011).
Chromatin isolation by RNA purication: Similar to
CHART, chromatin isolation by RNA purication (ChIRP)
or dChIRP (Chu et al. 2011; Quinn et al. 2014) uses a
probe-based afnity approach built on biotinylated oligonu-
cleotides that tile the lncRNA. Isolated interactors that bind
the lncRNA (ChIRP), or bind to a specic domain (dChIRP),
can be analyzed by high-throughput sequencing, or by meth-
ods that detect proteins, such as mass spectrometry or West-
ern blotting.
RNA antisense purication: The RNA antisense purication
(RAP) method (McHugh et al. 2015), instead of using short
probes (20 nt) as in CHART or ChIRP, utilizes longer probes
of 60 nt to increase the stability of the interaction in afnity
pulldowns.
RNA pulldown: The above-mentioned methods to investigate
lncRNA-partner molecules in intact cells were preceded by
probe-based isolation of RNAs in cell extracts in pulldown
experiments similar to protein coimmunoprecipitation.
Mapping RNA-genome interactions: Mapping RNA-genome
interactions (MARGI) uses proximity ligation to connect
chromatin-associated RNAs to their genomic targets, thus
revealing native RNA-chromatin interactions. Variations in
the approach were designed to differentiate between direct
interactions (diMARGI), mediated by protein or RNA-te-
thered interactions, of RNADNA chimeras, or passive inter-
actions (pxMARGI) (Sridhar et al. 2017).
Global RNA interaction with DNA by deep sequencing:
Global RNA interaction with DNA by deep sequencing (GRID-
Seq) harnesses in situ ligation to identify genome-wide con-
tacts between RNA and chromatin. The developers of this
method included mouse ES cells and found distinct cis- and
trans- chromatin interacting RNAs tied to cell-specic gene
expression patterns. GRID-Seq particularly enriches for chro-
matin interactions with nascent RNAs (Li et al. 2017).
1102 K. N. Smith et al.
Downloaded from https://academic.oup.com/genetics/article/213/4/1093/5930620 by guest on 11 May 2023
Chromatin associated RNA sequencing: Chromatin associ-
ated RNA sequencing (CHAR-Seq) is an in situ proximity
ligation approach coupled with enzymatic chromatin diges-
tion to detect RNADNA contacts genome-wide. After se-
quencing, CHAR-Seq maps the genomic interacting sites of
multiple classes of chromatin-associated RNAs including na-
scent transcripts, ncRNAs involved in regulation of dosage
compensation, and trans-interacting RNAs involved in RNA
processing (Bell et al. 2018).
RNA and DNA interacting complexes ligated and se-
quenced: RNA and DNA interacting complexes ligated and se-
quenced (RADICL-Seq) identies genome-wide RNA-chromatin
interactions in cross-linked nuclei. Thus far, it has been applied to
two cell typesmouse ES cells and mouse oligodendrocyte
progenitorsthat can differentiate toward multiple cell fates.
This approach revealed cell-type specicRNAchromatin inter-
actions, and was able to identify unique genome occupancy
patterns for different classes of transcripts (Bonetti et al. 2019).
Proling interacting RNAs on chromatin: Proling interact-
ing RNAs on chromatin (PIRCh-Seq) is an antibody-dependent
approach that proles RNAchromatin interactions, with less
enrichment of nascent RNAs cotranscriptionally tethered by
RNA polymerases to chromatin. PIRCh-Seq has been used to
identify the chromatin-associated transcriptome in both hu-
man and mouse ES cells and broblasts, as well as mouse
neuronal progenitors, where the authors found cell- and al-
lele-specic RNAchromatin interactions (Fang et al. 2019).
Microscopy: As previously discussed, lncRNA localization can
provide functional clues. The advent of single molecule im-
aging approaches facilitates the localization of RNA relative to
other interacting molecules. For example, labeled FISH probes
used in single molecule uorescence in situ microscopy
(smFISH) (Cabili et al. 2015; Dunagin et al. 2015), followed
by immunouorescence microscopy with three-dimensional
and quantitative uorescence image analysis allows for the
visualization of colocalized lncRNA and protein interactors
within subcellular domains (Lino Cardenas et al. 2018). Vari-
ations, including merFISH (Chen et al. 2015) and seqFISH
(Shah et al. 2016), depend on barcoding in sequential rounds
of hybridization to enable the detection of many transcripts
simultaneously. The resulting data complexity creates the need
for computational tools such as trendsceek (Edsgärd et al.
2018) to analyze these data. We can expect lncRNA monitor-
ing in spatial transcriptomics to increase as different function-
alities continue to emerge.
Bioinformatics
NPInter: Hao et al. (2016) is a repository of functional inter-
actions between noncoding RNAs and interacting partners
including small and large RNAs, DNA and proteins. At the
core of NPInter is a manual curation process based on pub-
lished literature, with a primary focus on experimentally
veried physical interactions, supplemented with in silico
predictions supported by high-throughput sequence data.
The latest version contains .900,000 interactions between
noncoding RNAs and other biomolecules from 22 organisms.
For RNAs, accession IDs from NONCODE, Ensembl, and
RefSeq are supported, and integration with the UCSC Ge-
nome Browser facilitates visualization of binding sites for
human, mouse, and yeast genomes.
POSTAR: Hu et al. (2017), Zhu et al. (2018) is a database that
enables exploration of post-transcriptional regulatory interac-
tions, based primarily on 1200 CLIP-Seq data sets. The aim of
POSTAR is to contribute a better understanding of how RBPs
impact post-transcriptional regulatory processes in six species.
Its integration with the UCSC Genome Browser facilitates
rapid visualization of RBP binding sites within transcripts.
RAID: The RAID database (Zhang et al. 2014; Yi et al. 2017),
formerly CLIPdb, incorporates experimentally derived and
Table 2 Methods to Investigate lncRNA function in cell fate determination
Approach Type Readout Output
Differential expression analysis Experimental Indirect Gene expression differences between cell types with
differentiation, or in a condition of interest
Expression correlation (+/2target or effector) Experimental Indirect Network generation to identify similarly expressed
gene clusters, including candidate target molecules
or possible upstream regulators
Afnity purication/proximity ligation and deep
sequencing (CHART/ChIRP/RAP/MARGI/GRID-Seq/
CHAR-Seq/Radicl-Seg/PIRCh-Seq)
Experimental Direct Chromatin targets
Afnity purication and mass spectrometry/Western
blot (CHART/ChIRP/RAP)
Experimental Direct Protein interactions
Single molecule uorescence in situ hybridization
(smFISH) +/2immunouorescence
Experimental Direct Subcellular localization and colocalization with targets
or effectors
Conservation analysis Experimental Direct Applicability of function across species
Structural analysis Experimental Direct Secondary structure and functional domain identi-
cation; 3D structure
Bioinformatics tools Computational Indirect Predict interactions, assess k-mer content, make
structural inferences
Multimodal lncRNA Interactions 1103
Downloaded from https://academic.oup.com/genetics/article/213/4/1093/5930620 by guest on 11 May 2023
computationally predicted RNA interactions from the pub-
lished literature, as well as other databases. RAID includes
data for 60 species and .1.2 million individual RNAprotein
and 4 million RNARNA interactions, respectively. A score
that is based on the evidence supporting the interaction in-
dicates the condence in each interaction.
RNAprotein interaction prediction: Using protein and RNA
sequence data, the family of RPISeq machine learning qual-
iers (Muppirala et al. 2011) provides RNAprotein interac-
tion probabilities. Different versions of the tool provide the
probability of interaction between a specic RNA and pro-
tein, a specic RNA and up to 100 proteins, or a specic pro-
tein and up to 100 RNAs. Additionally, the sequence of a
specic protein can be used to query the RPIntDB, which
contains .30,000 individual RNAprotein interactions.
RNAprotein interaction predictor: RPI-Pred (Suresh et al.
2015) is a Support Vector Machine based prediction tool that
uses RNA and protein sequence information and protein
structural fragment data. Users can also test multiple candi-
dates including assessing the potential of a single RNA to
interact with multiple proteins or multiple RNAs interacting
with a single protein.
StarBase: This RNA-centric resource (Yang et al. 2011; Li
et al. 2014) details interactions between various classes of
long and short RNAs, as well as between RNAs and proteins
as extracted from CLIP-Seq (PAR-CLIP, HITS-CLIP, iCLIP,
CLASH), degradome-seq, and RNARNA interactome data.
Users are also able to impute downstream effects of these
interactions based on accompanying gene expression data.
Combined approaches
Structure determination and analysis: The secondary struc-
ture of RNA can be predicted from thermodynamic principles,
but inaccuracy in the parameters means that experimental
input is required to generate a reliable model. This is most
often provided in the form of constraints on secondary struc-
ture generated from either evolutionary considerations (con-
servation of base pairs, ideally by covariation) and/or direct
experimental mapping of secondary structure using either
enzymatic, or, most often, chemical techniques such as SHAPE
and dimethyl sulfate (DMS) mapping (Kirk et al. 2018), psor-
alen crosslinking (Lu et al. 2016), and high throughput liga-
tion followed by deep-sequencing (Ramani et al. 2015).
Detection can be achieved efciently through deep-sequencing,
although capillary electrophoresis, and even polyacrylamide
gels, can be used at much lower cost when studying single
lncRNAs. Because folding in vitro and in cells might differ
because of kinetic constraints on cotranscriptional RNA fold-
ing and the presence of RBPs (Leamy et al. 2016), techniques
are being developed to probe RNA secondary structures in
cellular contexts as well. Here, the primary limitation is
sensitivity and the requirement to have sufcient RNA
for detection, which could require overexpression since most
lncRNAs are present at relatively low copy number. Higher
resolution methods such as SAXS (Small Angle X-ray Scatter-
ing) (Rambo and Tainer 2013) or X-ray crystallography and
NMR currently have very low throughput and can be used
only to investigate a few paradigmatic RNAs or systems of
particularly high biological interest.
Conclusion and Perspectives
This review summarizes multiple lines of evidence showing
that lncRNAs regulate cellular plasticity and cell fate deter-
mination, often through combination of multiple mecha-
nisms. By adding a further layer of complexity to gene
regulation, they broadly contribute to gene expression regu-
lation to either (i) maintain a blanket undifferentiated state,
(ii) promote exit toward a specied cell type, (iii) reprogram
cells to a pluripotent ground state, or (iv) contribute to cell
specication control in organismal development. LncRNAs
perform these complex functions in integrated networks with
a diverse set of cellular players with which they interact
physically and/or functionally.
Progress toward the phenotypic assessment of lncRNA
depletion occurs through loss-of-function and gain-of-
function approaches (Liu and Lim 2018), facilitated by the
advent of CRISPR/Cas9 technologies. Indeed, high-throughput
screens using CRISPR interference identied .300 lncRNAs
that impacted iPS cell growth, with a smaller subset inu-
encing pluripotency maintenance as determined by OCT4
expression (S. J. Liu et al. 2017). Functional ablation ap-
proaches include poly-A signal insertion proximal to the tran-
scription start site, although a drawback of this insertion is
residual background expression, as well as deletion of the
lncRNA locus, which results in total loss of lncRNA function,
but which may also affect unannotated regulatory elements.
Even the genetic manipulation of smaller sequences such
as promoters or single exons for well-annotated intergenic
lncRNAs should be carried out with caution to avoid modifying
regulatory genomic sequences. It should also be noted that DNA-
targeting approaches have resulted in differing phenotypes, as
exemplied by Fendrr. Studies using a reporter gene replacement
strategy for Fendrr found abnormalities in lung development and
lethality at a later time point (Sauvageau et al. 2013; Lai et al.
2015), compared with the heart and body wall abnormali-
ties resulting in prenatal lethality in earlier investigations
(Grote et al. 2013). Regardless of the specictechnicalap-
proach for DNA sequence manipulation, it will be important
to study the impact of lncRNA expression ablation on the
function and regulation of the interacting proteins, RNAs,
and chromatin in development. Specic molecular targeting
of the lncRNA itself using CRISPR/Cas13 (Abudayyeh et al.
2017; Cox et al. 2017) could facilitate such investigations.
Intriguingly, at least several lncRNAs displaying differ-
ing levels of conservation, such as Malat1,Neat1,Cyrano,
Braveheart,Evx1as, and Visc-2, and found to have profound
molecular or cellular functions, had no overt developmental
phenotype in knockout animals (Han et al. 2018). This
1104 K. N. Smith et al.
Downloaded from https://academic.oup.com/genetics/article/213/4/1093/5930620 by guest on 11 May 2023
suggests either a primary role for lncRNAs in ne-tuning
developmental functions, distinct roles in specic cellular
processes requiring situational study, or yet unearthed com-
pensatory functions, potentially by related/familial lncRNAs.
Another explanation for the absence of animal phenotypes
could be off-target effects of knockdown approaches using
RNA interference or antisense oligonucleotide-dependent
depletion (Matsui and Corey 2017). Increased use of gene
editing approaches such as CRISPR/Cas9 will help to clarify
lncRNA functionality in cell and animal models.
Improvements are needed to allow study of lncRNA inter-
actions at the single cell and single molecule level. While
technically feasible to a limited extent using imaging tech-
nologies, these methods remain specialized and low through-
put. These studies would allow the determination of cell fate
as single cell expression and epigenetic studies have indicated
substantial heterogeneity even within clonal cellular subpop-
ulations. Perhaps dynamic lncRNA interactions contribute to
this heterogeneity to dictate differing cell fates as well.
Related to this heterogeneity, it is still unclear whether a
classication system that would allow prediction of lncRNA
interactions will be found, but, if such a system existed, it is
unlikely to be based on broad segments of sequence conser-
vation because these are generally absent in lncRNAs.
However, it might be possible to base classication on the
identication of shorter sequence stretches (k-mers) or struc-
tural features of the lncRNA that facilitate interactions with
other biomolecules. Identication of the relevant structural
elements would provide insight into lncRNA interactions with
noncanonical RNA binding proteins as well, including those
without conserved and/or overt RNA binding domains.
There are untapped opportunities for progress in the
mechanistic analysis of lncRNA function for better under-
standing of specic developmental processes and some down-
stream applications, including personalized therapeutics. For
example, cancers typically progress through the acquisition of
stemness features (Malta et al. 2018) and undifferentiated
tumors have poor prognosis because they share immortality
and repopulation capacity characteristics with stem cells.
LncRNAs have emerged as central oncogenic and tumor sup-
pressive factors involved in misregulated cancer pathways
(Berger et al. 2018; Chiu et al. 2018), and many lncRNAs,
such as Cyrano, have been have been shown to support cel-
lular proliferation (Smith et al. 2017; Deng et al. 2018; X. Liu
et al. 2018; Naemura et al. 2018). Studies of lncRNAs in stem
cell contexts will not only enable better understanding of
mammalian development and differentiation, but may also
eventually facilitate better treatment of cancer and degener-
ative diseases.
Acknowledgments
Grant support: National Institutes of Health (NIH) R01
GM101974 to T.M., NIH R03 HD093977 to K.N.S, NIH
R01GM121806 to J.M.C., as well as R35 GM126942 and
RO1 GM 103834 to G.V.
Literature Cited
Abudayyeh, O. O., J. S. Gootenberg, P. Essletzbichler, S. Han, J.
Joung \et al., 2017 RNA targeting with CRISPRCas13. Nature
550: 280284. https://doi.org/10.1038/nature24049
Alexander,R.P.,G.Fang,J.Rozowsky,M.Snyder,andM.B.
Gerstein, 2010 Annotating non-coding regions of the genome.
Nat. Rev. Genet. 11: 559571.
Altschul, S. F., W. Gish, W. Miller, E. W. Myers, and D. J. Lipman,
1990 Basic local alignment search tool. J. Mol. Biol. 215: 403
410. https://doi.org/10.1016/S0022-2836(05)80360-2
Amândio, A. R., A. Necsulea, E. Joye, B. Mascrez, and D. Duboule,
2016 Hotair is dispensible for mouse development. PLoS Genet.
12:e1006232. https://doi.org/10.1371/journal.pgen.1006232
Bassett, A. R., A. Akhtar, D. P. Barlow, A. P. Bird, N. Brockdorff,
et al., 2014 Considerations when investigating LncRNA function
in vivo. eLife 3: e03058. https://doi.org/10.7554/eLife.03058
Batista, P. J., and H. Y. Chang, 2013 Long noncoding RNAs: cellular
address codes in development and disease. Cell 152: 12981307.
Bell, J. C., D. Jukam, N. A. Teran, V. I. Risca, O. K. Smith, et al.,
2018 Chromatin-associated RNA sequencing (ChAR-Seq) maps
genome-wide RNA-to-DNA contacts. eLife 7: pii: e27024. https://
doi.org/10.7554/eLife.27024
Beltran, M., I. Puig, C. Pena, J. M. Garcia, A. B. Alvarez et al.,
2008 A natural antisense transcript regulates Zeb2/Sip1 gene
expression during snail1-induced epithelial-mesenchymal transition.
Genes Dev. 22: 756769. https://doi.org/10.1101/gad.455708
Berger, A. C., A. Korkut, R. S. Kanchi, A. M. Hegde, W. Lenoir et al.,
2018 A comprehensive pan-cancer molecular study of gyneco-
logic and breast cancers. Cancer Cell 33: 690705.e9. https://
doi.org/10.1016/j.ccell.2018.03.014
Bergeron-Sandoval, L.-P., N. Safaee, and S. W. Michnick,
2016 Mechanisms and consequences of macromolecular phase
separation. Cell 165: 10671079. https://doi.org/10.1016/j.
cell.2016.05.026
Bernardes de Jesus, B., S. P. Marinho, S. Barros, A. Sousa-Franco, C.
Alves-Vale et al., 2018 Silencing of the LncRNA Zeb2-NAT fa-
cilitates reprogramming of aged broblasts and safeguards stem
cell pluripotency. Nat. Commun. 9: 94. https://www.nature.
com/articles/s41467-017-01921-6
Bernstein, E., E. M. Duncan, O. Masui, J. Gil, E. Heard et al.,
2006 Mouse polycomb proteins bind differentially to methyl-
ated histone H3 and RNA and are enriched in facultative het-
erochromatin. Mol. Cell. Biol. 26: 25602569. https://doi.org/
10.1128/MCB.26.7.2560-2569.2006
Blaisdell, B. E., 1989 Effectiveness of measures requiring and not
requiring prior sequence alignment for estimating the dissimi-
larity of natural sequences. J. Mol. Evol. 29: 526537. https://
doi.org/10.1007/BF02602924
Boeynaems, S., S. Alberti, N. L. Fawzi, T. Mittag, M. Polymenidou
et al., 2018 Protein phase separation: a new phase in cell bi-
ology. Trends Cell Biol. 28: 420435. https://doi.org/10.1016/
j.tcb.2018.02.004
Bonasio, R., E. Lecona, V. Narendra, P. Voigt, F. Parisi et al.,
2014 Interactions with RNA direct the polycomb group pro-
tein SCML2 to chromatin where it represses target genes. eLife
3: e02637. https://doi.org/10.7554/eLife.02637
Bonetti, A., F. Agostini, A. M. Suzuki, K. Hashimoto, G. Pascarella
et al., 2019 RADICL-Seq identies general and cell type-
specic principles of genome-wide RNA-chromatin interactions.
bioRxiv. https://doi.org/10.1101/681924
Brown, C. J., A. Ballabio, J. L. Rupert, R. G. Lafreniere, M. Grompe,
et al., 1991 A gene from the region of the human X inactiva-
tion centre is expressed exclusively from the inactive X chromo-
some. Nature 349: 3844. https://doi.org/10.1038/349038a0
Burge, C., A. M. Campbell, and S. Karlin, 1992 Over- and under-
representation of short oligonucleotides in DNA sequences.
Multimodal lncRNA Interactions 1105
Downloaded from https://academic.oup.com/genetics/article/213/4/1093/5930620 by guest on 11 May 2023
Proc. Natl. Acad. Sci. USA 89: 13581362. https://doi.org/
10.1073/pnas.89.4.1358
Cabili, M. N., M. C. Dunagin, P. D. McClanahan, A. Biaesch, O.
Padovan-Merhar et al., 2015 Localization and abundance anal-
ysis of human LncRNAs at single-cell and single-molecule reso-
lution. Genome Biol. 16: 20.
Caudron-Herger, M., S. F. Rusin, M. E. Adamo, J. Seiler, V. K.
Schmid et al., 2019 R-deeP: proteome-wide and quantitative
identication of RNA-dependent proteins by density gradient
ultracentrifugation. Mol. Cell 75: 184199.e10. https://doi.org/
10.1016/j.molcel.2019.04.018
Cesana, M., D. Cacchiarelli, I. Legnini, T. Santini, O. Sthandier
et al., 2011 A long noncoding RNA controls muscle differenti-
ation by functioning as a competing endogenous RNA. Cell 147:
358369. https://doi.org/10.1016/j.cell.2011.09.028
Chakraborty,D.,M.Paszkowski-Rogacz,N.Berger,L.Ding,J.Mircetic
et al., 2017 LncRNA Panct1 maintains mouse embryonic stem
cell identity by regulating TOBF1 recruitment to Oct-Sox se-
quences in early G1. Cell Rep. 21: 30123021. https://
doi.org/10.1016/j.celrep.2017.11.045
Chen, K. H., A. N. Boettiger, J. R. Moftt, S. Wang, and X. Zhuang,
2015 RNA imaging. Spatially resolved, highly multiplexed
RNA proling in single cells. Science 348: aaa6090. https://
doi.org/10.1126/science.aaa6090
Chillón, I., and A. M. Pyle, 2016 Inverted repeat Alu elements in
the human LincRNA-P21 adopt a conserved secondary structure
that regulates RNA function. Nucleic Acids Res. 44: 94629471.
https://doi.org/10.1093/nar/gkw599
Chiu, H. S., S. Somvanshi, E. Patel, T. W. Chen, V. P. Singh et al.;
Cancer Genome Atlas Research Network, 2018 Pan-cancer
analysis of LncRNA regulation supports their targeting of cancer
genes in each tumor context. Cell Rep. 23: 297312.e12. https://
doi.org/10.1016/j.celrep.2018.03.064
Chu, C., K. Qu, F. L. Zhong, S. E. Artandi, and H. Y. Chang,
2011 Genomic maps of long noncoding RNA occupancy reveal
principles of RNA-chromatin interactions. Mol. Cell 44: 667
678. https://doi.org/10.1016/j.molcel.2011.08.027
Cox, D. B. T., J. S. Gootenberg, O. O. Abudayyeh, B. Franklin, M. J.
Kellner et al., 2017 RNA editing with CRISPR-Cas13. Science
358: 10191027. https://doi.org/10.1126/science.aaq0180
Daneshvar, K., J. V. Pondick, B.-M. Kim, C. Zhou, S. R. York et al.,
2016 DIGIT is a conserved long noncoding RNA that regulates
GSC expression to control denitive endoderm differentiation of
embryonic stem cells. Cell Rep. 17: 353365. https://doi.org/
10.1016/j.celrep.2016.09.017
Daneshvar, K., M. Behfar Ardehali, I. A. Klein, A. J. Kratkiewicz, C.
Zhou et al., 2019 LncRNA DIGIT and BRD3 protein form
phase-separated condensates to regulate endoderm differentia-
tion. bioRxiv. https://doi.org/10.1101/547513
Das, P. P., D. A. Hendrix, E. Apostolou, A. H. Buchner, M. C. Canver
et al., 2015 PRC2 is required to maintain expression of the
maternal Gtl2-Rian-Mirg locus by preventing de novo DNA meth-
ylation in mouse embryonic stem cells. Cell Rep. 12: 14561470.
https://doi.org/10.1016/j.celrep.2015.07.053
Davidovich, C., X. Wang, C. Cifuentes-Rojas, K. J. Goodrich, A. R.
Gooding et al., 2015 Toward a consensus on the binding spec-
icity and promiscuity of PRC2 for RNA. Mol. Cell 57: 552558.
https://doi.org/10.1016/j.molcel.2014.12.017
Deng,J.,H.Deng,C.Liu,Y.Liang,andS.Wang,2018 Long
non-coding RNA OIP5AS1 functions as an oncogene in lung
adenocarcinoma through targeting MiR-448/Bcl-2. Biomed.
Pharmacother. 98: 102110. https://doi.org/10.1016/j.biopha.
2017.12.031
Dinger, M. E., P. P. Amaral, T. R. Mercer, K. C. Pang, S. J. Bruce
et al., 2008 Long noncoding RNAs in mouse embryonic stem
cell pluripotency and differentiation. Genome Res. 18: 1433
1445. https://doi.org/10.1101/gr.078378.108
Dominguez, D., P. Freese, M. S. Alexis, A. Su, M. Hochman et al.,
2018 Sequence, structure, and context preferences of human
RNA binding proteins. Mol. Cell 70: 854867.e9. https://
doi.org/10.1016/j.molcel.2018.05.001
Dreyfuss, G., M. J. Matunis, S. Pinol-Roma, and C. G. Burd,
1993 HnRNP proteins and the biogenesis of MRNA. Annu.
Rev. Biochem. 62: 289321. https://doi.org/10.1146/annurev.
bi.62.070193.001445
Dunagin, M., M. N. Cabili, J. Rinn, and A. Raj, 2015 Visualization
of LncRNA by single-molecule uorescence in situ hybridization.
Methods Mol. Biol. 1262:319. https://doi.org/10.1007/978-1-
4939-2253-6_1
Durruthy-Durruthy, J., V. Sebastiano, M. Wossidlo, D. Cepeda, and
J. Cui, 2016 The primate-specic noncoding RNA HPAT5 reg-
ulates pluripotency during human preimplantation develop-
ment and nuclear reprogramming. Nat. Genet. 48: 4452.
https://doi.org/10.1038/ng.3449
Edgar, R. C., and S. Batzoglou, 2006 Multiple sequence alignment.
Curr.Opin.Struct.Biol.16:368373. https://doi.org/10.1016/
j.sbi.2006.04.004
Edsgärd, D., P. Johnsson, and R. Sandberg, 2018 Identication of
spatial expression trends in single-cell gene expression data.
Nat. Methods 15: 339342. https://doi.org/10.1038/nmeth.4634
Engreitz, J. M., J. E. Haines, E. M. Perez, G. Munson, J. Chen
et al., 2016 Local regulation of gene expression by LncRNA
promoters, transcription and splicing. Nature 539: 452455.
https://doi.org/10.1038/nature20149
Etchegaray, J.-P., L. Chavez, Y. Huang, K. N. Ross, J. Choi et al.,
2015 The histone deacetylase SIRT6 controls embryonic stem
cell fate via TET-mediated production of 5-hydroxymethylcytosine.
Nat. Cell Biol. 17: 545557. https://doi.org/10.1038/ncb3147
Faghihi,M.A.,F.Modarresi,A.M.Khalil,D.E.Wood,B.G.Sahagan
et al., 2008 Expression of a noncoding RNA is elevated in
Alzheimers disease and drives rapid feed-forward regulation of
beta-secretase. Nat. Med. 14: 723730. https://doi.org/10.1038/
nm1784
Faghihi, M. A., M. Zhang, J. Huang, F. Modarresi, M. P. Van der
Brug, 2010 Evidence for natural antisense transcript-mediated
inhibition of microRNA function. Genome Biol. 11: R56. https://
doi.org/10.1186/gb-2010-11-5-r56
Fang, J., Q. Ma, C. Chu, B. Huang, L. Li et al., 2019 Functional
classication of noncoding RNAs associated with distinct his-
tone modications by PIRCh-seq. bioRxiv. https://doi.org/10.
1101/667881
Fang, R., W. N. Moss, M. Rutenberg-Schoenberg, and M. D. Simon,
2015 Probing Xist RNA structure in cells using targeted struc-
ture-seq. PLoS Genet. 11: e1005668. .https://doi.org/10.1371/
journal.pgen.1005668
Fiorenzano, A., E. Pascale, M. Gagliardi, S. Terreri, M. Papa et al.,
2018 An ultraconserved element containing LncRNA preserves
transcriptional dynamics and maintains ESC self-renewal. Stem Cell
Rep. 10: 11021114. https://doi.org/10.1016/j.stemcr.2018.01.014
Fisher, C. L., and A. G. Fisher. 2011 Chromatin states in pluripo-
tent, differentiated, and reprogrammed cells. Curr. Opin. Genet.
Dev. 21: 14046. https://doi.org/10.1016/j.gde.2011.01.015.
Flynn, R. A., L. Martin, R. C. Spitale, B. T. Do, S. M. Sagan et al.,
2015 Dissecting noncoding and pathogen RNA-protein interac-
tomes. RNA 21: 135143. https://doi.org/10.1261/rna.047803.114
Frank, S., G. Ahuja, D. Bartsch, N. Russ, W. Yao et al.,
2019 YylncT denes a class of divergently transcribed LncRNAs
and safeguards the T-mediated mesodermal commitment of hu-
man PSCs. Cell Stem Cell 24: 318327.e8. https://doi.org/
10.1016/j.stem.2018.11.005
Frankish, A., M. Diekhans, A.-M. Ferreira, R. Johnson, I. Jungreis
et al., 2019 GENCODE reference annotation for the human and
mouse genomes. Nucleic Acids Res. 47: D766D773. https://
doi.org/10.1093/nar/gky955
1106 K. N. Smith et al.
Downloaded from https://academic.oup.com/genetics/article/213/4/1093/5930620 by guest on 11 May 2023
Fuchs Wightman, F., L. E. Giono, J. P. Fededa, and M. de la Mata,
2018 Target RNAs strike back on microRNAs. Front. Genet. 9:
435. https://doi.org/10.3389/fgene.2018.00435
Gaspar-Maia, A., A. Alajem, E. Meshorer, and M. Ramalho-Santos,
2011 Open chromatin in pluripotency and reprogramming.
Nat. Rev. Mol. Cell Biol. 12: 3647 (erratum: Nat. Rev. Mol. Cell
Biol. 12: 273). https://doi.org/10.1038/nrm3036
Gong, C., and L. E. Maquat, 2011 LncRNAs transactivate STAU1-
mediated MRNA decay by duplexing with 39UTRs via Alu elements.
Nature 470: 284288. https://doi.org/10.1038/nature09701
Grant, J., S. K. Mahadevaiah, P. Khil, M. N. Sangrithi, H. Royo et al.,
2012 Rsx is a metatherian RNA with Xist-like properties in
Xchromosome inactivation. Nature 487: 254258. https://
doi.org/10.1038/nature11171
Gregory, R. I., T. P. Chendrimada, N. Cooch, and R. Shiekhattar,
2005 Human RISC couples microRNA biogenesis and posttran-
scriptional gene silencing. Cell 123: 631640. https://doi.org/
10.1016/j.cell.2005.10.022
Grote, P., L. Wittler, D. Hendrix, F. Koch, S. Währisch et al.,
2013 The tissue-specic LncRNA Fendrr is an essential regu-
lator of heart and body wall development in the mouse. Dev. Cell
24: 206214. https://doi.org/10.1016/j.devcel.2012.12.012
Guo, Y., X. Chen, R. Xing, M. Wang, X. Zhu et al., 2018 Interplay
between FMRP and LncRNA TUG1 regulates axonal develop-
ment through mediating SnoN-Ccd1 pathway. Hum. Mol. Genet.
27: 475485. https://doi.org/10.1093/hmg/ddx417
Haegele, L., B. Ingold, H. Naumann, G. Tabatabai, B. Ledermann
et al., 2003 Wnt signalling inhibits neural differentiation of
embryonic stem cells by controlling bone morphogenetic protein
expression. Mol. Cell. Neurosci. 24: 696708. https://doi.org/
10.1016/S1044-7431(03)00232-X
Hafner, M., M. Landthaler, L. Burger, M. Khorshid, J. Hausser et al.,
2010 Transcriptome-wide identication of RNA-binding pro-
tein and microRNA target sites by PAR-CLIP. Cell 141: 129
141. https://doi.org/10.1016/j.cell.2010.03.009
Han, X., S. Luo, G. Peng, J. Y. Lu, G. Cui, et al., 2018 Mouse
knockout models reveal largely dispensable but context-depen-
dent functions of LncRNAs during development. J. Mol. Cell Biol.
10: 175178. .https://doi.org/10.1093/jmcb/mjy003
Hao, Y., W. Wu, H. Li, J. Yuan, J. Luo et al., 2016 NPInter v3.0: an
upgraded database of noncoding RNA-associated interactions.
Database (Oxford) 2016: pii: baw057. https://doi.org/10.1093/
database/baw057
Harrow, J., A. Frankish, J. M. Gonzalez, E. Tapanari, M. Diekhans
et al., 2012 GENCODE: the reference human genome annota-
tion for the ENCODE project. Genome Res 22: 17601774.
https://doi.org/10.1101/gr.135350.111
Hawkes, E. J., S. P. Hennelly, I. V. Novikova, J. A. Irwin, C. Dean
et al., 2016 COOLAIR antisense RNAs form evolutionarily con-
served elaborate secondary structures. Cell Rep 16: 30873096.
https://doi.org/10.1016/j.celrep.2016.08.045
He, Q., S. Yang, X. Gu, M. Li, C. Wang et al., 2018 Long non-
coding RNA TUG1 facilitates osteogenic differentiation of peri-
odontal ligament stem cells via interacting with Lin28A. Cell
Death Dis. 9: 455. https://doi.org/10.1038/s41419-018-0484-2
Heinrich, E.-M., and S. Dimmeler, 2012 MicroRNAs and stem
cells: control of pluripotency, reprogramming, and lineage com-
mitment. Circ. Res. 110: 10141022. https://doi.org/10.1161/
CIRCRESAHA.111.243394
Hendrickson, D.G., D. R. Kelley, D. Tenen, B. Bernstein, and J. L.
Rinn, 2016 Widespread RNA binding by chromatin-associated
proteins. Genome Biol. 17: 28. https://doi.org/10.1186/s13059-
016-0878-3
Hnisz, D., K. Shrinivas, R. A. Young, A. K. Chakraborty, and P. A.
Sharp, 2017 A phase separation model for transcriptional
control. Cell 169: 1323. https://doi.org/10.1016/j.cell.2017.
02.007
Hon, C.-C., J. A. Ramilowski, J. Harshbarger, N. Bertin, O. J. L.
Rackham et al., 2017 An atlas of human long non-coding RNAs
with accurate 59ends. Nature 543: 199204. https://doi.org/
10.1038/nature21374
Hu, B., Y. T. Yang, Y. Huang, Y. Zhu, and Z. J. Lu, 2017 POSTAR:
a platform for exploring post-transcriptional regulation coordi-
nated by RNA-binding proteins. Nucleic Acids Res. 45: D104
D114. https://doi.org/10.1093/nar/gkw888
Hurst, L. D., and N. G. C. Smith, 1999 Molecular evolutionary
evidence that H19 MRNA is functional. Trends Genet. 15:
134135. https://doi.org/10.1016/S0168-9525(99)01696-0
Ilik, I. A., J. J. Quinn, P. Georgiev, F. Tavares-Cadete, D. Maticzka
et al., 2013 Tandem stem-loops in RoX RNAs act together to
mediate X chromosome dosage compensation in Drosophila. Mol.
Cell 51: 156173. https://doi.org/10.1016/j.molcel.2013.07.001
Ilik, I. A., D. Maticzka, P. Georgiev, N. M. Gutierrez, R. Backofen
et al., 2017 A mutually exclusive stemloop arrangement in RoX2
RNA is essential for Xchromosome regulation in Drosophila.Genes
Dev. 31: 19731987. https://doi.org/10.1101/gad.304600.117
Jain, A. K., Y. Xi, R. McCarthy, K. Allton, K. C. Akdemir et al.,
2016 LncPRESS1 is a P53-regulated LncRNA that safeguards
pluripotency by disrupting SIRT6-mediated de-acetylation of his-
tone H3K56. Mol. Cell 64: 967981. https://doi.org/10.1016/
j.molcel.2016.10.039
Jiang, W., Y. Liu, R. Liu, K. Zhang, and Y. Zhang, 2015 The
LncRNA DEANR1 facilitates human endoderm differentiation
by activating FOXA2 expression. Cell Rep. 11: 137148.
https://doi.org/10.1016/j.celrep.2015.03.008
Juan, V., 2000 Evidence for evolutionarily conserved secondary
structure in the H19 tumor suppressor RNA. Nucleic Acids Res.
28: 12211227. https://doi.org/10.1093/nar/28.5.1221
Kalantry, S., K. C. Mills, D. Yee, A. P. Otte, B. Panning et al.,
2006 The Polycomb group protein Eed protects the inactive
Xchromosome from differentiation-induced reactivation. Nat.
Cell Biol. 8: 195202. https://doi.org/10.1038/ncb1351
Kallen, A. N., X. B. Zhou, J. Xu, C. Qiao, J. Ma, et al., 2013 The
imprinted H19 LncRNA antagonizes let-7 microRNAs. Mol. Cell
52: 101112. https://doi.org/10.1016/j.molcel.2013.08.027
Kan, L., N. Israsena, Z. Zhang, M. Hu, L.-R. Zhao et al., 2004 Sox1
acts through multiple independent pathways to promote neuro-
genesis. Dev. Biol. 269: 580594. https://doi.org/10.1016/j.
ydbio.2004.02.005
Kaneko, S., J. Son, S. S. Shen, D. Reinberg, and R. Bonasio,
2013 PRC2 binds active promoters and contacts nascent RNAs
in embryonic stem cells. Nat. Struct. Mol. Biol. 20: 12581264.
https://doi.org/10.1038/nsmb.2700
Kaneko, S., R. Bonasio, R. Saldaña-Meyer, T. Yoshida, J. Son et al.,
2014 Interactions between JARID2 and noncoding RNAs reg-
ulate PRC2 recruitment to chromatin. Mol. Cell 53: 290300.
https://doi.org/10.1016/j.molcel.2013.11.012
Kari, L., K. A. Hill, A. S. Sayem, R. Karamichalis, N. Bryans et al.,
2015. Mapping the space of genomic signatures. PLoS One 10:
e0119815. .https://doi.org/10.1371/journal.pone.0119815
Karlic, R., S. Ganesh, V. Franke, E. Svobodova, J. Urbanova et al.,
2017 Long non-coding RNA exchange during the oocyte-to-
embryo transition in mice. DNA Res. 24: 129141 (erratum:
DNA Res. 24: 219220). https://doi.org/10.1093/dnares/dsw058
Khalil, A. M., M. Guttman, M. Huarte, M. Garber, A. Raj et al.,
2009 Many human large intergenic noncoding RNAs associate
with chromatin-modifying complexes and affect gene expres-
sion. Proc. Natl. Acad. Sci. USA 106: 1166711672.
Kirk, J. M., S. O. Kim, K. Inoue, M. J. Smola, D. M. Lee et al.,
2018 Functional classication of long non-coding RNAs by
k-Mer content. Nat. Genet. 50: 14741482. https://doi.org/
10.1038/s41588-018-0207-8
Klattenhoff, C. A., J. C. Scheuermann, L. E. Surface, R. K. Bradley, P.
A. Fields et al., 2013 Braveheart, a long noncoding RNA
Multimodal lncRNA Interactions 1107
Downloaded from https://academic.oup.com/genetics/article/213/4/1093/5930620 by guest on 11 May 2023
required for cardiovascular lineage commitment. Cell 152: 570
583. https://doi.org/10.1016/j.cell.2013.01.003
Kleaveland, B., C. Y. Shi, J. Stefano, and D. P. Bartel, 2018 A
network of noncoding regulatory RNAs acts in the mammalian
brain. Cell 174: 350362.e17. https://doi.org/10.1016/
j.cell.2018.05.022
König, J., K. Zarnack, G. Rot, T. Curk, M. Kayikci et al., 2010 ICLIP
reveals the function of HnRNP particles in splicing at individual
nucleotide resolution. Nat. Struct. Mol. Biol. 17: 909915.
https://doi.org/10.1038/nsmb.1838
Kotzin, J. J., S. P. Spencer, S. J. McCright, D. B. Uthaya Kumar, M.
A. Collet et al., 2016 The long non-coding RNA morrbid reg-
ulates bim and short-lived myeloid cell lifespan. Nature 537:
239243. https://doi.org/10.1038/nature19346
Lai, K. M., G. Gong, A. Atanasio, J. Rojas, J. Quispe et al.,
2015 Diverse phenotypes and specic transcription patterns
in twenty mouse lines with ablated LincRNAs. PLoS One 10:
e0125522. .https://doi.org/10.1371/journal.pone.0125522
Leamy, K. A., S. M. Assmann, D. H. Mathews, and P. C. Bevilacqua,
2016 Bridging the gap between in vitro and in vivo RNA
folding. Q. Rev. Biophys. 49: e10. https://doi.org/10.1017/
S003358351600007X
Lees, J. A., M. Vehkala, N. Välimäki, S. R. Harris, C. Chewapreecha
et al., 2016 Sequence element enrichment analysis to deter-
mine the genetic basis of bacterial phenotypes. Nat. Commun. 7:
12797. https://doi.org/10.1038/ncomms12797
Lewandowski, J. P., G. Dumbovi´
c, A. R. Watson, T. Hwang, E.
Jacobs-Palmer et al., 2019 The locus is essential for male fer-
tility. bioRxiv. https://doi.org/10.1101/562066
Li, J. H., S. Liu, H. Zhou, L. H. Qu, and J. H. Yang, 2014 StarBase
v2.0: decoding MiRNA-CeRNA, MiRNA-NcRNA and protein-RNA
interaction networks from large-scale CLIP-seq data. Nucleic
Acids Res. 42: D92D97. https://doi.org/10.1093/nar/gkt1248
Li, J. H., S. Liu, L. L. Zheng, J. Wu, W. J. Sun et al.,
2015 Discovery of protein-LncRNA interactions by integrating
large-scale CLIP-seq and RNA-seq datasets. Front Bioeng Bio-
technol. 2: 88. https://doi.org/10.3389/fbioe.2014.00088
Li, L., B. Liu, O. L. Wapinski, M. -C. Tsai, K. Qu et al.,
2013 Targeted disruption of hotair leads to homeotic trans-
formation and gene derepression. Cell Rep. 5: 312. https://
doi.org/10.1016/j.celrep.2013.09.003
Li, L., J. A. Helms, and H. Y. Chang, 2016 Comment on hotair is
dispensable for mouse development.PLoS Genet. 12:
e1006406. https://doi.org/10.1371/journal.pgen.1006406
Li, M., H. Gou, B. K. Tripathi, J. Huang, S. Jiang et al., 2015b An
apela RNA-containing negative feedback loop regulates P53-
mediated apoptosis in embryonic stem cells. Cell Stem Cell
16: 669683. https://doi.org/10.1016/j.stem.2015.04.002
Li, X., B. Zhou, L. Chen, L. T. Gou, H. Li et al., 2017 GRID-seq
reveals the global RNA-chromatin interactome. Nat. Biotechnol.
35: 940950. https://doi.org/10.1038/nbt.3968
Lin, N., K. Y. Chang, Z. Li, K. Gates, Z. A. Rana et al., 2014 An
evolutionarily conserved long noncoding RNA TUNA controls
pluripotency and neural lineage commitment. Mol. Cell 53:
10051019. https://doi.org/10.1016/j.molcel.2014.01.021
Lino Cardenas, C. L., C. W. Kessinger, Y. Cheng, C. MacDonald, T.
MacGillivray et al., 2018 An HDAC9-MALAT1-BRG1 complex
mediates smooth muscle dysfunction in thoracic aortic aneu-
rysm. Nat. Commun. 9: 1009. https://www.nature.com/articles/
s41467-018-03394-7
Liu, F., S. Somarowthu, and A. M. Pyle, 2017 Visualizing the
secondary and tertiary architectural domains of LncRNA
RepA. Nat. Chem. Biol. 13: 282289. https://doi.org/10.1038/
nchembio.2272
Liu, S. J., and D. A. Lim, 2018 Modulating the expression of long
noncoding RNAs for functional studies. EMBO Rep. 19:pii:
e46955. https://doi.org/10.15252/embr.201846955
Liu, S. J., M. A. Horlbeck, S. W. Cho, H. S. Birk, M. Malatesta et al.,
2017b CRISPRi-based genome-scale identication of func-
tional long noncoding RNA loci in human cells. Science 355:
pii: aah7111. 10.1126/science.aah7111.https://doi.org/10.1126/
science.aah7111
Liu, X. H., M. Sun, F. Q. Nie, Y. B. Ge, E. B. Zhang et al., 2014 Lnc
RNA HOTAIR functions as a competing endogenous RNA to
regulate HER2 expression by sponging MiR-331-3p in gastric
cancer. Mol. Cancer 13: 92. https://doi.org/10.1186/1476-
4598-13-92
Liu, X., J. Zheng, Y. Xue, H. Yu, W. Gong et al., 2018 PIWIL3/
OIP5AS1/MiR-3673p/CEBPA feedback loop regulates the bi-
ological behavior of glioma cells. Theranostics 8: 10841105.
https://doi.org/10.7150/thno.21740
Loewer, S., M. N. Cabili, M. Guttman, Y. H. Loh, K. Thomas et al.,
2010 Large intergenic non-coding RNA-RoR modulates re-
programming of human induced pluripotent stem cells. Nat
Genet. 42: 11131117.
Lu, H., D. Yu, A. S. Hansen, S. Ganguly, R. Liu et al., 2018 Phase-
separation mechanism for C-terminal hyperphosphorylation of
RNA polymerase II. Nature 558: 318323. https://doi.org/
10.1038/s41586-018-0174-3
Lu, X., F. Sachs, L. Ramsay, P. É. Jacques, J. Göke et al., 2014 The
retrovirus HERVH is a long noncoding RNA required for human
embryonic stem cell identity. Nat. Struct. Mol. Biol. 21: 423
425. https://doi.org/10.1038/nsmb.2799
Lu, Z., Q. C. Zhang, B. Lee, R. A. Flynn, M. A. Smith et al.,
2016 RNA duplex map in living cells reveals higher-order tran-
scriptome structure. Cell 165: 12671279. https://doi.org/
10.1016/j.cell.2016.04.028
Luo, S., J. Y. Lu, L. Liu, Y. Yin, C. Chen et al., 2016 Divergent
LncRNAs regulate gene expression and lineage differentiation in
pluripotent cells. Cell Stem Cell 18: 637652. https://doi.org/
10.1016/j.stem.2016.01.024
Mallam, A. L., W. Sae-Lee, J. M. Schaub, F. Tu, A. Battenhouse
et al., 2018 Systematic discovery of endogenous human ribonu-
cleoprotein complexes. bioRxiv. https://doi.org/10.1101/480061
Malta, T. M., A. Sokolov, A. J. Gentles, T. Burzykowski, L. Poisson
et al., 2018 Machine learning identies stemness features as-
sociated with oncogenic dedifferentiation. Cell 173: 338
354.e15. https://doi.org/10.1016/j.cell.2018.03.034
Mandal, M., and R. R. Breaker, 2004 Gene regulation by ribos-
witches. Nat. Rev. Mol. Cell Biol. 5: 451463. https://doi.org/
10.1038/nrm1403
Matsui, M., and D. R. Corey, 2017 Non-coding RNAs as drug
targets. Nat. Rev. Drug Discov. 16: 167179. https://doi.org/
10.1038/nrd.2016.117
Mattick, J. S., 2001 Non-coding RNAs: the architects of eukaryotic
complexity. EMBO Rep. 2: 986991. https://doi.org/10.1093/
embo-reports/kve230
Mattick, J. S., and J. L. Rinn, 2015 Discovery and annotation of
long noncoding RNAs. Nat. Struct. Mol. Biol. 22: 57. https://
doi.org/10.1038/nsmb.2942
McHugh, C. A., C. K. Chen, A. Chow, C. F. Surka, C. Tran et al.,
2015 The Xist LncRNA interacts directly with SHARP to si-
lence transcription through HDAC3. Nature 521: 232236.
https://doi.org/10.1038/nature14443
Melé, M., and J. L. Rinn, 2016 Cats Cradlingthe 3D genome by
the act of LncRNA transcription. Mol. Cell 62: 657664. https://
doi.org/10.1016/j.molcel.2016.05.011
Mercer, T. R., and J. S. Mattick, 2013 Structure and function of
long noncoding RNAs in epigenetic regulation. Nat. Struct. Mol.
Biol. 20: 300307. https://doi.org/10.1038/nsmb.2480
Muppirala, U. K., V. G. Honavar, and D. Dobbs, 2011 Predicting
RNA-protein interactions using only sequence information.
BMC Bioinformatics 12: 489. https://doi.org/10.1186/1471-
2105-12-489
1108 K. N. Smith et al.
Downloaded from https://academic.oup.com/genetics/article/213/4/1093/5930620 by guest on 11 May 2023
Naemura, M., M. Kuroki, T. Tsunoda, N. Arikawa, Y. Sawata et al.,
2018 The long noncoding RNA OIP5-AS1 is involved in the
regulation of cell proliferation. Anticancer Res. 38: 7781.
https://doi.org/10.21873/anticanres.12194
Nesterova, T. B., G. Wei, H. Coker, G. Pintacuda, J. S. Bowness
et al., 2019 Systematic allelic analysis denes the interplay
of key pathways in X chromosome inactivation. Nature Comm.
10: 3129. https://doi.org/10.1038/s41467-019-11171-3.
Ng, S. Y., R. Johnson, and L. W. Stanton, 2012 Human long non-
coding RNAs promote pluripotency and neuronal differentiation
by association with chromatin modiers and transcription factors.
EMBO J. 31: 522533. https://doi.org/10.1038/emboj.2011.459
Ng, S.-Y., G. K. Bogu, B. S. Soh, and L. W. Stanton, 2013 The long
noncoding RNA RMST interacts with SOX2 to regulate neurogenesis.
Mol. Cell 51: 349359. https://doi.org/10.1016/j.molcel.2013.07.017
Palazzo, A. F., and E. S. Lee, 2015 Non-coding RNA: what is func-
tional and what is junk? Front. Genet. 6: 2. https://doi.org/
10.3389/fgene.2015.00002
Pandey, P., M. A. Bender, R. Johnson, R. Patro, and B. Berger,
2018 Squeakr: an exact and approximate k-mer counting
system. Bioinformatics 34: 568575. .https://doi.org/10.1093/
bioinformatics/btx636
Pisignano, G., S. Napoli, M. Magistri, S. N. Mapelli, C. Pastori et al.,
2017 A promoter-proximal transcript targeted by genetic poly-
morphism controls E-cadherin silencing in human cancers. Nat.
Commun. 8: 15622. https://doi.org/10.1038/ncomms15622
Piwecka, M., P. Glaˇ
zar, L. R. Hernandez-Miranda, S. Memczak, S. A.
Wolf et al., 2017 Loss of a mammalian circular RNA locus
causes MiRNA deregulation and affects brain function. Science
357: pii: eaam8526. 10.1126/science.aam8526.https://doi.org/
10.1126/science.aam8526
Portoso, M., R. Ragazzini, ˇ
Z. Brenˇ
ciˇ
c, A. Moiani, A. Michaud et al.,
2017 PRC2 is dispensable for HOTAIR-mediated transcrip-
tional repression. EMBO J. 36: 981994. https://doi.org/10.
15252/embj.201695335
Queiroz, R. M. L., T. Smith, E. Villanueva, M. Marti-Solano, M.
Monti et al., 2019 Comprehensive identication of RNA-protein
interactions in any organism using orthogonal organic phase sep-
aration (OOPS). Nat. Biotechnol. 37: 169178 (erratum: Nat. Bio-
technol. 37: 692). https://doi.org/10.1038/s41587-018-0001-2
Quinn, J. J., and H. Y. Chang, 2016 Unique features of long non-
coding RNA biogenesis and function. Nat. Rev. Genet. 17: 47
62. https://doi.org/10.1038/nrg.2015.10
Quinn, J. J., I. A. Ilik, K. Qu,P. Georgiev, C. Chu et al.,
2014 Revealing long noncoding RNA architecture and func-
tions using domain-specic chromatin isolation by RNA puri-
cation. Nat. Biotechnol. 32: 933940. https://doi.org/10.1038/
nbt.2943
Quinn, J. J., Q. C. Zhang, P. Georgiev, I. A. Ilik, A. Akhtar et al.,
2016 Rapid evolutionary turnover underlies conserved
LncRNA-genome interactions. Genes Dev. 30: 191207. https://
doi.org/10.1101/gad.272187.115
Ramani, V., R. Qiu, and J. Shendure, 2015 High-throughput de-
termination of RNA structure by proximity ligation. Nat. Bio-
technol. 33: 980984. https://doi.org/10.1038/nbt.3289
Rambo, R. P., and J. A. Tainer, 2013 Super-resolution in solution
X-ray scattering and its applications to structural systems biol-
ogy. Annu. Rev. Biophys. 42: 415441. https://doi.org/10.1146/
annurev-biophys-083012-130301
Regha, K., M. A. Sloane, R. Huang, F. M. Pauler, K. E. Warczok
et al., 2007 Active and repressive chromatin are interspersed
without spreading in an imprinted gene cluster in the mamma-
lian genome. Mol. Cell 27: 353366. https://doi.org/10.1016/
j.molcel.2007.06.024
Rice, P., I. Longden, and A. Bleasby, 2000 EMBOSS: the European
molecular biology open software suite. Trends Genet. 16: 276
277. https://doi.org/10.1016/S0168-9525(00)02024-2
Rinn, J. L., and H. Y. Chang, 2012 Genome regulation by long
noncoding RNAs. Annu. Rev. Biochem. 81: 145166. https://
doi.org/10.1146/annurev-biochem-051410-092902
Rinn, J. L., and J. Ule, 2014 Oming in on RNA-protein interac-
tions. Genome Biol. 15: 401. https://doi.org/10.1186/gb4158
Rom, A., L. Melamed, M. J. Goldrich, R. Kadir, M. Golan et al.,
2019 Regulation of CHD2 expression by the long noncoding RNA
is essential for viability. bioRxiv. https://doi.org/10.1101/536771
Sahakyan, A., Y. Yang, and K. Plath, 2018 The role of Xist in
Xchromosome dosage compensation. Trends Cell Biol. 28:
9991013. https://doi.org/10.1016/j.tcb.2018.05.005
Sanli, I., S. Lalevée, M. Cammisa, A. Perrin, F. Rage et al.,
2018 Meg3 non-coding RNA expression controls imprinting
by preventing transcriptional upregulation in Cis. Cell Rep. 23:
337348. https://doi.org/10.1016/j.celrep.2018.03.044
Santoro, F., D. Mayer, R. M. Klement, K. E. Warczok, A. Stukalov
et al., 2013 Imprinted Igf2r silencing depends on continuous
airn LncRNA expression and is not restricted to a developmental
window. Development 140: 11841195. https://doi.org/10.1242/
dev.088849
Sauvageau, M., L. A. Goff, S. Lodato,B. Bonev, A. F. Groff et al.,
2013 Multiple knockout mouse models reveal LincRNAs are
required for life and brain development. eLife 2: e01749.
https://doi.org/10.7554/eLife.01749
Savi´
c, N., D. Bär, S. Leone, S. C. Frommel, F. A. Weber et al.,
2014 LncRNA maturation to initiate heterochromatin formation
in the nucleolus is required for exit from pluripotency in ESCs. Cell
Stem Cell 15: 720734. https://doi.org/10.1016/j.stem.2014.10.005
Schertzer, M. D., K. C. A. Braceros, J. Starmer, R. E. Cherney, D. M.
Lee et al., 2019 LncRNA-induced spread of polycomb con-
trolled by genome architecture, RNA abundance, and CpG
island DNA. Mol. Cell 75: 523537.e10. https://doi.org/
10.1016/j.molcel.2019.05.028
Selleri, L., M. S. Bartolomei, W. A. Bickmore, L. He, L. Stubbs et al.,
2016 A hox-embedded long noncoding RNA: is it all hot air?
PLoS Genet. 12: e1006485. https://doi.org/10.1371/journal.
pgen.1006485
Shah, S., E. Lubeck, W. Zhou, and L. Cai, 2016 In situ transcrip-
tion proling of single cells reveals spatial organization of cells
in the mouse Hippocampus. Neuron 92: 342357. https://
doi.org/10.1016/j.neuron.2016.10.001
Sheik Mohamed, J., P. M. Gaughwin, B. Lim, P. Robson, and L.
Lipovich. 2010 Conserved long noncoding RNAs transcription-
ally regulated by Oct4 and Nanog modulate pluripotency
in mouse embryonic stem cells. RNA 16: 324337. https://
doi.org/10.1261/rna.1441510
Shimoda, M., M. Kanai-Azuma, K. Hara, S. Miyazaki, Y. Kanai et al.,
2007 Sox17 plays a substantial role in late-stage differentia-
tion of the extraembryonic endoderm in vitro. J. Cell Sci. 120:
38593869. https://doi.org/10.1242/jcs.007856
Simon, M. D., C. I. Wang, P. V. Kharchenko, J. A. West, B. A.
Chapman et al., 2011 The genomic binding sites of a noncod-
ing RNA. Proc. Natl. Acad. Sci. USA 108: 2049720502. https://
doi.org/10.1073/pnas.1113536108
Smith,K.N.,J.Starmer,S.C.Miller,P.Sethupathy,andT.Magnuson,
2017 Long noncoding RNA moderates microRNA activity to
maintain self-renewal in embryonic stem cells. Stem Cell Rep.
9: 108121. https://doi.org/10.1016/j.stemcr.2017.05.005
Smith, K. N., J. Starmer, and T. Magnuson, 2018 Interactome
determination of a long noncoding RNA implicated in embry-
onic stem cell self-renewal. Sci. Rep. 8: 17568. https://doi.org/
10.1038/s41598-018-34864-z
Smola, M. J.,T. W. Christy, K. Inoue, C. O. Nicholson, M. Friedersdorf
et al., 2016 SHAPE reveals transcript-wide interactions, com-
plex structural domains, and protein interactions across the Xist
LncRNA in living cells. Proc. Natl. Acad. Sci. USA 113: 10322
10327. https://doi.org/10.1073/pnas.1600008113
Multimodal lncRNA Interactions 1109
Downloaded from https://academic.oup.com/genetics/article/213/4/1093/5930620 by guest on 11 May 2023
Somarowthu, S., M. Legiewicz, I. Chillón, M. Marcia, F. Liu et al.,
2015 HOTAIR forms an intricate and modular secondary
structure. Mol. Cell 58: 353361. https://doi.org/10.1016/
j.molcel.2015.03.006
Sprague, D., S. A. Waters, J. M. Kirk, J. R. Wang, P. B. Samollow
et al., 2019 Nonlinear sequence similarity between the Xist
and Rsx long noncoding RNAs suggests shared functions of tan-
dem repeat domains. RNA 25: 10041019. https://doi.org/
10.1261/rna.069815.118
Sridhar, B., M. Rivas-Astroza, T. C. Nguyen, W. Chen, Z. Yan et al.,
2017 Systematic mapping of RNA-chromatin interactions
in vivo. Curr. Biol. 27: 602609 (erratum: Curr. Biol. 27: 610
612). doi: 10.1016/j.cub.2017.01.011https://doi.org/10.1016/
j.cub.2017.01.011
Suresh, V., L. Liu, D. Adjeroh, and X. Zhou, 2015 RPI-pred: pre-
dicting NcRNA-protein interaction using sequence and struc-
tural information. Nucleic Acids Res. 43: 13701379. https://
doi.org/10.1093/nar/gkv020
Terranova, R., S. Yokobayashi, M. B. Stadler, A. P. Otte, M. van
Lohuizen et al., 2008 Polycomb group proteins Ezh2 and Rnf2
direct genomic contraction and imprinted repression in early
mouse embryos. Dev. Cell 15: 668679. https://doi.org/10.1016/
j.devcel.2008.08.015
Trendel, J., T. Schwarzl, R. Horos, A. Prakash, A. Bateman et al.,
2019 The human RNA-binding proteome and its dynamics
during translational arrest. Cell 176: 391403.e19. https://
doi.org/10.1016/j.cell.2018.11.004
Tsai, M. C., O. Manor, Y. Wan, N. Mosammaparast, J. K. Wang
et al., 2010 Long noncoding RNA as modular scaffold of his-
tone modication complexes. Science 329: 689693.
Ule, J., K. Jensen, A. Mele, and R. B. Darnell, 2005 CLIP: a method
for identifying proteinRNA interaction sites in living cells. Methods
37: 376386. https://doi.org/10.1016/j.ymeth.2005.07.018
Ulitsky, I., A. Shkumatava, C. H. Jan, H. Sive, and D. P. Bartel,
2011 Conserved function of LincRNAs in vertebrate embryonic
development despite rapid sequence evolution. Cell 147: 1537
1550. https://doi.org/10.1016/j.cell.2011.11.055
UniProt Consortium, 2015 UniProt: a hub for protein information.
Nucleic Acids Res. 43: D204D212. https://doi.org/10.1093/
nar/gku989
Wang, G.-Q., Y. Wang, Y. Xiong, X.-C. Chen, M.-L. Ma, 2016 Sirt1
AS LncRNA interacts with its MRNA to inhibit muscle formation
by attenuating function of MiR-34a. Scientic Rep. 6: 21865.
.https://doi.org/10.1038/srep21865
Wang, J., J. Mager, Y. Chen, E. Schneider, J. C. Cross et al.,
2001 Imprinted X inactivation maintained by a mouse polycomb
group gene. Nat. Genet. 28: 371375. https://doi.org/10.1038/
ng574
Wang, K. C., Y. W. Yang, B. Liu, A. Sanyal, R. Corces-Zimmerman
et al., 2011 A long noncoding RNA maintains active chromatin
to coordinate homeotic gene expression. Nature 472: 120124.
https://doi.org/10.1038/nature09819
Wang, X., K. J. Goodrich, A. R. Gooding, H. Naeem, S. Archer et al.,
2017 Targeting of Polycomb repressive complex 2 to RNA by
short repeats of consecutive guanines. Mol. Cell 65: 1056
1067.e5. https://doi.org/10.1016/j.molcel.2017.02.003
Wang, Y., Z. Xu, J. Jiang, C. Xu, J. Kang et al., 2013 Endogenous
MiRNA sponge LincRNA-RoR regulates Oct4, Nanog, and Sox2
in human embryonic stem cell self-renewal. Dev. Cell 25: 6980.
https://doi.org/10.1016/j.devcel.2013.03.002
Weng, R., C. Lu, X. Liu, G. Li, Y. Lan et al., 2018 Long noncoding
RNA-1604 orchestrates neural differentiation through the MiR-
200c/ZEB axis. Stem Cells 36: 325336. https://doi.org/10.
1002/stem.2749
Wheeler, T. J., and S. R. Eddy, 2013 Nhmmer: DNA homology
search with prole HMMs. Bioinformatics 29: 24872489.
https://doi.org/10.1093/bioinformatics/btt403
Winzi, M., N. Casas Vila, M. Paszkowski-Rogacz, L. Ding, S. Noack
et al., 2018 The long noncoding RNA LncR492 inhibits neural
differentiation of murine embryonic stem cells. PLoS One 13:
e0191682. https://doi.org/10.1371/journal.pone.0191682
Wutz, A., T. P. Rasmussen, and R. Jaenisch, 2002 Chromosomal
silencing and localization are mediated by different domains of Xist
RNA. Nat. Genet. 30: 167174. https://doi.org/10.1038/ng820
Xue, Z., S. Hennelly, B. Doyle, A. A. Gulati, I. V. Novikova et al.,
2016 A G-rich motif in the LncRNA Braveheart interacts with a
zinc-nger transcription factor to specify the cardiovascular lineage.
Mol. Cell 64: 3750. .https://doi.org/10.1016/j.molcel.2016.08.010
Yang, F., X. Deng, W. Ma, J. B. Berletch, N. Rabaia et al.,
2015 The LncRNA rre anchors the inactive X chromosome
to the nucleolus by binding CTCF and maintains H3K27me3
methylation. Genome Biol. 16: 52. https://doi.org/10.1186/
s13059-015-0618-0
Yang, J. H., J. H. Li, P. Shao, H. Zhou, Y. Q. Chen et al.,
2011 StarBase: a database for exploring MicroRNAMRNA in-
teraction maps from argonaute CLIP-seq and degradome-seq
data. Nucleic Acids Res. 39: D202D209. https://doi.org/
10.1093/nar/gkq1056
Yang, Y. W., R. A. Flynn, Y. Chen, K. Qu, B. Wan et al.,
2014 Essential role of LncRNA binding for WDR5 maintenance
of active chromatin and embryonic stem cell pluripotency. eLife
3: e02046. https://doi.org/10.7554/eLife.02046
Yi, Y., Y. Zhao, C. Li, L. Zhang, H. Huang et al., 2017 RAID v2.0:
an updated resource of RNA-associated interactions across or-
ganisms. Nucleic Acids Res. 45: D115D118. https://doi.org/
10.1093/nar/gkw1052
Yin, Y., P. Yan, J. Lu, G. Song, Y. Zhu et al., 2015 Opposing roles
for the LncRNA Haunt and its genomic locus in regulating HOXA
gene activation during embryonic stem cell differentiation. Cell Stem
Cell 16: 504516. https://doi.org/10.1016/j.stem.2015.03.007
Yoon, J.-H., K. Abdelmohsen, and M. Gorospe, 2013 Posttranscrip-
tional gene regulation by long noncoding RNA. J. Mol. Biol. 425:
37233730. https://doi.org/10.1016/j.jmb.2012.11.024
Young, T. L., T. Matsuda, and C. L. Cepko, 2005 The noncoding
RNA taurine upregulated gene 1 is required for differentiation
of the murine retina. Curr. Biol. 15: 501512. https://doi.org/
10.1016/j.cub.2005.02.027
Yu, C.-Y., and H.-C. Kuo, 2016 The trans-spliced long noncoding
RNA tsRMST impedes human embryonic stem cell differentia-
tion through WNT5A-mediated inhibition of the epithelial-to-
mesenchymal transition. Stem Cells 34: 20522062. https://
doi.org/10.1002/stem.2386
Zarnegar, B. J., R. A. Flynn, Y. Shen, B. T. Do, H. Y. Chang et al.,
2016 IrCLIP platform for efcient characterization of protein-
RNA interactions. Nat. Methods 13: 489492. https://doi.org/
10.1038/nmeth.3840
Zhang, X., D. Wu, L. Chen, X. Li, J. Yang et al., 2014 RAID: a
comprehensive resource for human RNA-associated (RNA-RNA/
RNA-Protein) interaction. RNA 20: 989993. https://doi.org/
10.1261/rna.044776.114
Zhou, Y., Q.-S. Dai, S.-C. Zhu, Y.-H. Han, H.-L. Han et al.,
2016 AK048794 maintains the mouse embryonic stem cell
pluripotency by functioning as an MiRNA sponge for MiR-592. Bio-
chem. J. 473: 36393654. https://doi.org/10.1042/BCJ20160540
Zhu, Y., G. Xu, Y. T. Yang, Z. Xu, X. Chen et al., 2018 POSTAR2:
deciphering the post-transcriptional regulatory logics. Nucleic
Acids Res. 47: D203D211. https://doi.org/10.1093/nar/gky830
˙
Zylicz, Jan Jakub, Aurélie Bousard, Kristina ˇ
Zumer, Francois Dossin,
Eusra Mohammad, Simão Teixeira da Rocha, Björn Schwalb,
et al. 2019 The Implication of Early Chromatin Changes in
X Chromosome Inactivation. Cell 176 (12): 182197.e23.
https://doi.org/10.1016/j.cell.2018.11.041.
Communicating editor: J. Rine
1110 K. N. Smith et al.
Downloaded from https://academic.oup.com/genetics/article/213/4/1093/5930620 by guest on 11 May 2023
... Moreover, since it has been shown that lncRNA activity changes with different levels of their expression, its specificity in various cell types and tissues should be assessed [18]. As it turned out, abnormalities in activity or biogenesis mechanisms of these molecules can appear in states of pathological conditions and indicate cancer progression by affecting a multitude of transcription factors along with the structure of the chromatin [19]. It has been demonstrated that the aberration of not only coding RNAs but also noncoding RNAs plays a crucial role in cancer biology. ...
... It has been demonstrated that the aberration of not only coding RNAs but also noncoding RNAs plays a crucial role in cancer biology. Although the function and activity of lncRNAs are still under investigation, in the future they may become crucial tools for predicting the development and possible treatment of cancer [19]. ...
Article
Full-text available
Long non-coding RNAs (lncRNAs) consist of at least 200 nucleotides. Although these mol- ecules do not code proteins, they carry many regulatory functions in normal cells, as well as in can- cer cells. For instance, many of these molecules have been previously correlated with tumorigenesis of different cancers and their reaction to various stress factors, such as radiotherapy, chemotherapy, or reactive oxygen species (ROS). The lncRNAs are associated not only with dysregulation in can- cers after applied treatment but also with beneficial effects that may be achieved by modulating their expression, often significantly enhancing the patients’ outcomes. A multitude of these mole- cules was previously considered as potential biomarkers of tumor development, progression, or cells’ response to radio- or chemotherapy. Irradiation, which is often used in treating numerous cancer types, is not always sufficient due to cells gaining resistance in multiple ways. In this review, studies considering lncRNAs and their reaction to radiotherapy were examined. These molecules were divided regarding their role in specific processes strictly related to irradiation, and their influ- ence on this type of treatment was explained, showing how vast an impact they have on IR-sup- ported combat with the disease. This review aims to shed some light on potential future lncRNA- based biomarkers and therapeutic targets.
... Importantly, the ivory lncRNA shows a conserved function in colour scale specification, while having also evolved divergent expression patterns that precisely delineate pattern information in multiple species. We discuss how these findings not only join a rich literature on the emerging roles of lncRNA in development and gene regulation (22)(23)(24), but also imply a direct role in the elaboration of adaptive phenotypic variation. ...
Preprint
Full-text available
Evolutionary variation in the wing pigmentation of butterflies and moths offers striking examples of adaptation by crypsis and mimicry. The cortex locus has been independently mapped as the locus controlling colour polymorphisms in 14 lepidopteran species, suggesting it acts as a genomic hotspot for the diversification of wing patterns, but functional validation through protein-coding knockouts has proven difficult to obtain. Our study unveils the role of a novel long non-coding RNA (lncRNA) which we name ivory , transcribed from the cortex locus, in modulating colour patterning in butterflies. Strikingly, ivory expression prefigures most melanic patterns during pupal development, suggesting an early developmental role in specifying scale identity. To test this, we generated CRISPR mosaic knock-outs in five nymphalid butterfly species and show that ivory mutagenesis yields transformations of dark pigmented scales into white or light-coloured scales. Genotyping of Vanessa cardui germline mutants associates these phenotypes to small on-target deletions at the conserved first exon of ivory . In contrast, cortex germline mutant butterflies with confirmed null alleles lack any wing phenotype, and exclude a colour patterning role for this adjacent gene. Overall, these results show that a lncRNA acts as a master switch of colour pattern specification, and played key roles in the adaptive diversification of colour patterns in butterflies. Significance statement Deciphering the genetic underpinnings of adaptive variation is fundamental for a comprehensive understanding of evolutionary processes. Long non-coding RNAs (lncRNAs) represent an emerging category of genetic modulators within the genome, yet they have been overlooked as a source of phenotypic diversity. In this study, we unveil the pivotal role of a lncRNA in orchestrating colour transitions between dark and light patterns during butterfly wing development. Remarkably, this lncRNA gene is nested within the cortex locus, a genetic region known to control multiple cases of adaptive variation in butterflies and moths, including iconic examples of natural selection. These findings highlight the significant influence of lncRNAs in developmental regulation, and also underscore their potential as key genetic players in the evolutionary process itself.
... On the RNA level it is the production of different types of non-coding RNA molecules which are involved in: D. Regulation of mRNA and ncRNA levels by sponging mechanism and influencing transcription process; and E. Modification of RNA molecules by capping, polyadenylation, alternative splicing, and editing. All of these processes cause modifications in gene transcription dysfunction of the activity or cellular mechanisms of lncRNAs may appear through pathological states including tumor progression through influence not only on the chromatin structure but also on several transcription factors [13]. According to GENCODE, NCBI Refseq, LNCipedia, and NON-CODE number of lncRNAs' genes is estimated between 56'946 to 17'952 locus which gives 27'381 to 172'216 transcripts [14][15][16][17]. ...
Article
Full-text available
Epigenetics is the changes in cellular phenotype without changes in the genotype. This term is not limited only to the modification of chromatin and DNA but also regards some RNAs like non-coding RNAs (ncRNAs), both short and long RNAs (lncRNAs) acting as molecular modifiers. Mobile RNAs, as free form or encapsulated in exosomes, can regulate neighboring cells or be placed in distant locations. It underlines the vast capacity of ncRNAs as epigenetic elements of transmission information and message of life. One of the amazing phenomenons is long non-coding microRNA-host-genes (lnc-MIRHGs) which processed transcripts function as lncRNAs and also as short RNAs named microRNAs (miRNAs). MIR31HG functions as a modulator of important biological and cellular processes including cell proliferation, apoptosis, cell cycle regulation, EMT process, metastasis, angiogenesis, hypoxia, senescence, and inflammation. However, in most cases, the role of MIR31HG is documented only by one study and there is a lack of exact description of molecular pathways implicated in these processes, and for some of, such as response to irradiation, no studies were done. In this review, MIR31HG, as an example of lnc-MIRHGs, was described in the context of its’ known function and its potential uses as a biomarker in oncology.
... 13 Many of these lncRNAs are associated with pluripotency and stemness in embryonic stem cells and iPSCs; other lncRNAs promote differentiation of pluripotent cells. [14][15][16] We have devised a chromatin RNA in situ reverse transcription sequencing (CRIST-seq) approach to profile lncRNAs that interact with the promoters of the stem cell core factor genes, such as Oct4 and Sox2. 13,17 These lncRNAs regulate the pluripotency network through various epigenetic mechanisms. ...
Article
Full-text available
Nuclear reprogramming of somatic cells into a pluripotent status has the potential to create patient-specific pluripotent stem cells (iPSCs) for regenerative medicine. Currently, however, the epigenetic mechanisms underlying this pluripotent reprogramming are poorly understood. To delineate this epigenetic regulatory network, we utilized a chromatin RNA in situ reverse transcription sequencing (CRIST-seq) approach to identify long noncoding RNAs (lncRNAs) embedded in the 3-dimensional intrachromosomal architecture of stem cell core factor genes. By combining CRIST-seq and RNA-seq, we identified Osilr9 (Oct4-Sox2 interacting lncRNA 9) as a pluripotency-associated lncRNA. Osilr9 expression was associated with the status of stem cell pluripotency in reprogramming. Using shRNA knockdown, we showed that this lncRNA was required for the optimal maintenance of stem cell pluripotency. Overexpression of Osilr9 induced robust activation of endogenous stem cell core factor genes in fibroblasts. Osilr9 participated in the formation of the intrachromosomal looping required for maintenance of pluripotency. After binding to the Oct4 promoter, Osilr9 recruited the DNA demethylase TET1, leading to promoter demethylation. These data demonstrate that Osilr9 is a critical chromatin epigenetic modulator that coordinates the promoter activity of core stem cell factor genes, highlighting the critical role of pluripotency-associated lncRNAs in stem cell pluripotency and reprogramming.
... lncRNAs appear to be important in functions related to the regulation of gene transcription in the nucleus or subsequent post-transcriptional modifications in the cytoplasm [21]. Abnormalities in the activity or biogenesis mechanisms of lncRNAs can appear in states of pathological conditions and indicate cancer progression by affecting not only the structure of the chromatin but also several transcription factors [22]. It has been demonstrated that lncRNAs play a key role in cancer biology. ...
Article
Full-text available
Long non-coding RNAs have proven to be important molecules in carcinogenesis. Due to little knowledge about them, the molecular mechanisms of tumorigenesis are still being explored. The aim of this work was to study the effect of ionizing radiation on the expression of lncRNAs in head and neck squamous cell carcinoma (HNSCC) in patients responding and non-responding to radiotherapy. The experimental model was created using a group of patients with response (RG, n = 75) and no response (NRG, n = 75) to radiotherapy based on the cancer genome atlas (TCGA) data. Using the in silico model, statistically significant lncRNAs were defined and further validated on six HNSCC cell lines irradiated at three different doses. Based on the TCGA model, C10orf55, C3orf35, C5orf38, CASC2, MEG3, MYCNOS, SFTA1P, SNHG3, and TMEM105, with the altered expression between the RG and NRG were observed. Analysis of pathways and immune profile indicated that these lncRNAs were associated with changes in processes, such as epithelial-to-mesenchymal transition, regulation of spindle division, and the p53 pathway, and differences in immune cells score and lymphocyte infiltration signature score. However, only C10orf55, CASC2, and SFTA1P presented statistically altered expression after irradiation in the in vitro model. In conclusion, the expression of lncRNAs is affected by ionization radiation in HNSCC, and these lncRNAs are associated with pathways, which are important for radiation response and immune response. Potentially presented lncRNAs could be used as biomarkers for personalized radiotherapy in the future. However, these results need to be verified based on an in vitro experimental model to show a direct net of interactions.
... lncRNAs are defined as RNA transcripts longer than 200 nucleotides [16]. lncRNAs interact with proteins, DNAs and mRNAs to form complex intramolecular and intermolecular secondary and highorder structures through diverse molecular mechanisms, thus mediating downstream gene expression, regulating protein activity, controlling alternative mRNAs splicing and providing scaffolding for chromatin modification [17]. lncRNAs can also function as ceRNAs by competitively binding to miRNAs, and thereby indirectly regulate tumor-related gene expression at transcriptional or post-transcriptional levels [18]. ...
Article
Full-text available
Hepatic, biliary and pancreatic cancers are a diverse set of malignancies with poor prognoses. It is possible that common molecular mechanisms are involved in the carcinogenesis of these cancers. Here, we identified LINC01537 and seven protein‐coding genes by integrative analysis of transcriptomes of mRNAs, miRNAs and lncRNAs from cholangiocarcinoma, hepatocellular carcinoma and pancreatic adenocarcinoma cohorts in TCGA. A predictive model constructed from seven biomarkers was established to successfully predict the survival rate of patients, which was then further verified in external cohorts. Additionally, patients with high‐risk scores in our model were prone to epithelial‐mesenchymal transition (EMT). Finally, activation of the biomarker PDE2A significantly attenuated migration and EMT in the HepG2 liver cancer cell line.
... 11,14,15 These roles are functionally dictated by their intermolecular interactions including with other RNA molecules, chromatin and proteins (thoroughly reviewed in Refs. 1,8,[16][17][18][19] ). OIP5-AS1 (human; terminology used throughout for clarity), also known as Oip5os1 (or 1700020I14Rik; mouse) or Cyrano (general), is often described as a canonical lncRNA. ...
Article
Full-text available
Long non-coding (lnc) RNAs have been implicated in a plethora of normal biological functions, and have also emerged as key molecules in various disease processes. OIP5-AS1, also commonly known by the alias Cyrano, is a lncRNA that displays broad expression across multiple tissues, with significant enrichment in particular contexts including within the nervous system and skeletal muscle. Thus far, this multifaceted lncRNA has been found to have regulatory functions in normal cellular processes including cell proliferation and survival, as well as in the development and progression of a myriad disease states. These widespread effects on normal and disease states have been found to be mediated through context-specific intermolecular interactions with dozens of miRNAs and proteins identified to date. This review explores recent studies to highlight OIP5-AS1's contextual yet pleiotropic roles in normal homeostatic functions as well as disease oetiology and progression, which may influence its utility in the generation of future theranostics.
Article
Over the last three decades, our view of RNA has changed from a simple intermediate supporting protein synthesis to a major regulator of biological processes. In the expanding area of RNA research, peptide nucleic acid (PNA) is emerging as a promising ligand for triple-helical recognition of complex RNAs. As discussed in this feature article, the key advantages of PNAs are high sequence specificity and affinity for RNA (>10 fold higher than for DNA) that are difficult to achieve with small molecule ligands. Emerging studies demonstrate that triple-helical binding of PNAs can modulate biological function and control dynamic conformational equilibria of complex folded RNAs. These results suggest that PNA has a unique potential as a research tool and therapeutic compound targeting RNA. The remaining problems hampering advances in these directions are limitations of sequences that can be recognized by Hoogsteen triplexes (typically purine rich tracts), poor cellular uptake and bioavailability of PNA, and potential off-target effects in biological systems. Recent exciting studies are discussed that illustrate how synthetic nucleic acid chemistry provides innovative solutions for these problems.
Article
Full-text available
Four new isoorotamide (Io)‐containing PNA nucleobases have been designed for A−U recognition of double helical RNA. New PNA monomers were prepared efficiently and incorporated into PNA nonamers for binding A−U in a PNA:RNA2 triplex. Isothermal titration calorimetry and UV thermal melting experiments revealed slightly improved binding affinity for singly modified PNA compared to known A‐binding nucleobases. Molecular dynamics simulations provided further insights into binding of Io bases in the triple helix. Together, the data revealed interesting insights into binding modes including the notion that three Hoogsteen hydrogen bonds are unnecessary for strong selective binding of an extended nucleobase. Cationic monomer Io8 additionally gave the highest affinity observed for an A‐binding nucleobase to date. These results will help inform future nucleobase design toward the goal of recognizing any sequence of double helical RNA.
Article
Full-text available
A DNA structure, known as triple-stranded DNA, is made up of three oligonucleotide chains that wind around one another to form a triple helix (TFO). Hoogsteen base pairing describes how triple-stranded DNA may be built at certain conditions by the attachment of the third strand to an RNA, PNA, or DNA, which might all be employed as oligonucleotide chains. In each of these situations, the oligonucleotides can be employed as an anchor, in conjunction with a specific bioactive chemical, or as a messenger that enables switching between transcription and replication through the triplex-forming zone. These data are also considered since various illnesses have been linked to the expansion of triplex-prone sequences. In light of metabolic acidosis and associated symptoms, some consideration is given to the impact of several low-molecular-weight compounds, including pH on triplex production in vivo. The review is focused on the development of biomedical oligonucleotides with triplexes.
Article
Full-text available
Mammalian genomes encode tens of thousands of noncoding RNAs. Most noncoding transcripts exhibit nuclear localization and several have been shown to play a role in the regulation of gene expression and chromatin remodeling. To investigate the function of such RNAs, methods to massively map the genomic interacting sites of multiple transcripts have been developed; however, these methods have some limitations. Here, we introduce RNA And DNA Interacting Complexes Ligated and sequenced (RADICL-seq), a technology that maps genome-wide RNA–chromatin interactions in intact nuclei. RADICL-seq is a proximity ligation-based methodology that reduces the bias for nascent transcription, while increasing genomic coverage and unique mapping rate efficiency compared with existing methods. RADICL-seq identifies distinct patterns of genome occupancy for different classes of transcripts as well as cell type–specific RNA-chromatin interactions, and highlights the role of transcription in the establishment of chromatin structure.
Article
Full-text available
Abstract We develop PIRCh-seq, a method which enables a comprehensive survey of chromatin-associated RNAs in a histone modification-specific manner. We identify hundreds of chromatin-associated RNAs in several cell types with substantially less contamination by nascent transcripts. Non-coding RNAs are found enriched on chromatin and are classified into functional groups based on the patterns of their association with specific histone modifications. We find single-stranded RNA bases are more chromatin-associated, and we discover hundreds of allele-specific RNA-chromatin interactions. These results provide a unique resource to globally study the functions of chromatin-associated lncRNAs and elucidate the basic mechanisms of chromatin-RNA interactions.
Article
Full-text available
Chromodomain helicase DNA binding protein 2 (Chd2) is a chromatin remodeller implicated in neurological disease. Here we show that Chaserr, a highly conserved long noncoding RNA transcribed from a region near the transcription start site of Chd2 and on the same strand, acts in concert with the CHD2 protein to maintain proper Chd2 expression levels. Loss of Chaserr in mice leads to early postnatal lethality in homozygous mice, and severe growth retardation in heterozygotes. Mechanistically, loss of Chaserr leads to substantially increased Chd2 mRNA and protein levels, which in turn lead to transcriptional interference by inhibiting promoters found downstream of highly expressed genes. We further show that Chaserr production represses Chd2 expression solely in cis, and that the phenotypic consequences of Chaserr loss are rescued when Chd2 is perturbed as well. Targeting Chaserr is thus a potential strategy for increasing CHD2 levels in haploinsufficient individuals. The conserved long noncoding RNA Chaserr is transcribed upstream of the chromatin remodeler Chd2. Here, using mouse genetics and high throughput assays, the authors show that Chaserr inhibits expression of Chd2 in cis and is required for postnatal mouse development.
Article
Full-text available
RNA-binding proteins (RBPs) play essential roles in biology and are frequently associated with human disease. Although recent studies have systematically identified individual RNA-binding proteins, their higher-order assembly into ribonucleoprotein (RNP) complexes has not been systematically investigated. Here, we describe a proteomics method for systematic identification of RNP complexes in human cells. We identify 1,428 protein complexes that associate with RNA, indicating that more than 20% of known human protein complexes contain RNA. To explore the role of RNA in the assembly of each complex, we identify complexes that dissociate, change composition, or form stable protein-only complexes in the absence of RNA. We use our method to systematically identify cell-type-specific RNA-associated proteins in mouse embryonic stem cells and finally, distribute our resource, rna.MAP, in an easy-to-use online interface (rna.proteincomplexes.org). Our system thus provides a methodology for explorations across human tissues, disease states, and throughout all domains of life.
Article
Full-text available
Xist RNA, the master regulator of X chromosome inactivation, acts in cis to induce chromosome-wide silencing. Whilst recent studies have defined candidate silencing factors, their relative contribution to repressing different genes, and their relationship with one another is poorly understood. Here we describe a systematic analysis of Xist-mediated allelic silencing in mouse embryonic stem cell-based models. Using a machine learning approach we identify distance to the Xist locus and prior gene expression levels as key determinants of silencing efficiency. We go on to show that Spen, recruited through the Xist A-repeat, plays a central role, being critical for silencing of all except a subset of weakly expressed genes. Polycomb, recruited through the Xist B/C-repeat, also plays a key role, favouring silencing of genes with pre-existing H3K27me3 chromatin. LBR and the Rbm15/m6A-methyltransferase complex make only a minor contribution to gene silencing. Together our results provide a comprehensive model for Xist-mediated chromosome silencing.
Preprint
Full-text available
Mammalian genomes encode tens of thousands of noncoding RNAs. Most noncoding transcripts exhibit nuclear localization and several have been shown to play a role in the regulation of gene expression and chromatin remodelling. To investigate the function of such RNAs, methods to massively map the genomic interacting sites of multiple transcripts have been developed. However, they still present some limitations. Here, we introduce RNA And DNA Interacting Complexes Ligated and sequenced (RADICL-seq), a technology that maps genome-wide RNA-chromatin interactions in intact nuclei. RADICL-seq is a proximity ligation-based methodology that reduces the bias for nascent transcription, while increasing genomic coverage and unique mapping rate efficiency compared to existing methods. RADICL-seq identifies distinct patterns of genome occupancy for different classes of transcripts as well as cell type-specific RNA-chromatin interactions, and emphasizes the role of transcription in the establishment of chromatin structure.
Article
Full-text available
The marsupial inactive X chromosome expresses a long noncoding RNA (lncRNA) called Rsx that has been proposed to be the functional analog of eutherian Xist Despite the possibility that Xist and Rsx encode related functions, the two lncRNAs harbor no linear sequence similarity. However, both lncRNAs harbor domains of tandemly repeated sequence. In Xist, these repeat domains are known to be critical for function. Using k-mer based comparison, we show that the repeat domains of Xist and Rsx unexpectedly partition into two major clusters that each harbor substantial levels of nonlinear sequence similarity. Xist Repeats B, C, and D were most similar to each other and to Rsx Repeat 1, whereas Xist Repeats A and E were most similar to each other and to Rsx Repeats 2, 3, and 4. Similarities at the level of k-mers corresponded to domain-specific enrichment of protein-binding motifs. Within individual domains, protein-binding motifs were often enriched to extreme levels. Our data support the hypothesis that Xist and Rsx encode similar functions through different spatial arrangements of functionally analogous protein-binding domains. We propose that the two clusters of repeat domains in Xist and Rsx function in part to cooperatively recruit PRC1 and PRC2 to chromatin. The physical manner in which these domains engage with protein cofactors may be just as critical to the function of the domains as the protein cofactors themselves. The general approaches we outline in this report should prove useful in the study of any set of RNAs.
Article
Long noncoding RNAs (lncRNAs) cause Polycomb repressive complexes (PRCs) to spread over broad regions of the mammalian genome. We report that in mouse trophoblast stem cells, the Airn and Kcnq1ot1 lncRNAs induce PRC-dependent chromatin modifications over multi-megabase domains. Throughout the Airn-targeted domain, the extent of PRC-dependent modification correlated with intra-nuclear distance to the Airn locus, preexisting genome architecture, and the abundance of Airn itself. Specific CpG islands (CGIs) displayed characteristics indicating that they nucleate the spread of PRCs upon exposure to Airn. Chromatin environments surrounding Xist, Airn, and Kcnq1ot1 suggest common mechanisms of PRC engagement and spreading. Our data indicate that lncRNA potency can be tightly linked to lncRNA abundance and that within lncRNA-targeted domains, PRCs are recruited to CGIs via lncRNA-independent mechanisms. We propose that CGIs that autonomously recruit PRCs interact with lncRNAs and their associated proteins through three-dimensional space to nucleate the spread of PRCs in lncRNA-targeted domains.
Preprint
Many long noncoding RNAs (lncRNAs) regulate gene transcription through binding to histone modification complexes. Therefore, a comprehensive study of nuclear RNAs in a histone modification-specific manner is critical to understand their regulatory mechanisms. Here we develop a method named Profiling Interacting RNAs on Chromatin by deep sequencing (PIRCh-seq), in which we profile chromatin-associated transcriptome in 5 different cell types using antibodies recognizing histone H3 and 6 distinct histone modifications associated with active or repressive chromatin states. PIRCh-seq identified chromatin-associated RNAs with substantially less contamination by nascent transcripts, as compared to existing methods. We classified chromatin-enriched lncRNAs into 6 functional groups based on the patterns of their association with specific histone modifications. LncRNAs were enriched with different chromatin modifications in different cell types, suggesting lncRNAs' regulation may also be cell type-specific. By integrating profiles of RNA secondary structure and RNA m6A modification, we found that RNA bases which bind to chromatin tend to be more single stranded. We discovered hundreds of allele-specific RNA-chromatin interactions, nominating specific single nucleotide variants that alter RNA association with chromatin. These results provide a unique resource to globally study the functions of chromatin-associated lncRNAs and elucidate the basic mechanisms of chromatin-RNA interaction.
Article
The comprehensive but specific identification of RNA-binding proteins as well as the discovery of RNA-associated protein functions remain major challenges in RNA biology. Here we adapt the concept of RNA dependence, defining a protein as RNA dependent when its interactome depends on RNA. We converted this concept into a proteome-wide, unbiased, and enrichment-free screen called R-DeeP (RNA-dependent proteins), based on density gradient ultracentrifugation. Quantitative mass spectrometry identified 1,784 RNA-dependent proteins, including 537 lacking known links to RNA. Exploiting the quantitative nature of R-DeeP, proteins were classified as not, partially, or completely RNA dependent. R-DeeP identified the transcription factor CTCF as completely RNA dependent, and we uncovered that RNA is required for the CTCF-chromatin association. Additionally, R-DeeP allows reconstruction of protein complexes based on co-segregation. The whole dataset is available at http://R-DeeP.dkfz.de, providing proteome-wide, specific, and quantitative identification of proteins with RNA-dependent interactions and aiming at future functional discovery of RNA-protein complexes.