ArticlePDF Available

Long Noncoding RNAs Xist in Three Dimensions

Authors:

Abstract and Figures

The three-dimensional organization of the mammalian genome spatially guides the binding of an RNA to loci for silencing gene expression.
No caption available
… 
No caption available
… 
Content may be subject to copyright.
16 AUGUST 2013 VOL 341 SCIENCE www.sciencemag.org
720
PERSPECTIVES
RNA has traditionally been viewed
as the genomic “messenger” mol-
ecule, carrying the coding informa-
tion from DNA to make proteins. However,
an abundance of short and long noncoding
RNAs also exist, many of which are likely to
have regulatory roles. Little is known about
the mechanisms of action of long noncoding
RNAs, but many appear to act as “guides”
that recruit protein regulatory complexes to
specifi c genomic loci to control gene expres-
sion ( 1). Where these long noncoding RNAs
contact the genome, and how they locate these
regulatory targets, have been open questions.
On page 767 of this issue, Engreitz et al. ( 2)
show that the mouse long noncoding RNA
called X-inactive specifi c transcript (Xist) is
initially targeted to specifi c loci across the X
chromosome using a targeting mechanism
that exploits three-dimensional chromosome
topology.
The Xist RNA orchestrates dosage com-
pensation in female mammalian cells.
Because female mammals have two X chro-
mosomes, inactivating one of them prevents
the expression of twice as many X-chromo-
some genes as in males, which possess only
one X chromosome. In X-inactivation, Xist is
synthesized (17 kilobases in length) from the
X chromosome that will ultimately become
inactivated. It recruits the polycomb repres-
sive complex 2 (PRC2) to switch off gene
expression from one X chromosome in each
female cell ( 3). Eventually, Xist appears to
encapsulate the entire inactive X chromo-
some in a “cloud” (as seen by microscopy),
forming a silencing compartment. The few
genes that escape silencing are seen looping
out of the condensed core ( 4). By develop-
ing and applying a technique to map RNA
interaction sites in the genome, Engreitz et
al. describe these images at a genomic level.
Consistent with microscopic observations,
the authors found that Xist interaction sites
are distributed broadly across the X chromo-
some in mouse fi broblasts, whereas “escape”
genes showed reduced association with Xist.
However, the most interesting questions
relate to how Xist establishes this pattern
of binding during the initiation of X-chro-
mosome inactivation. To investigate these
early events, Engreitz et al. used an induc-
ible system in mouse embryonic stem cells
that allowed them to activate Xist expres-
sion and map Xist interaction sites at defi ned
time points. They observed that Xist does not
spread uniformly outwards along the chro-
mosome sequence, but rather appears to pref-
erentially “jump” from its site of transcription
to certain early binding sites.
One mechanism to explain how Xist could
target specifi c distal loci would be the pres-
ence of high-affi nity binding sites at these
locations. Intriguingly, Engreitz et al. could
not identify any genomic feature to explain
the pattern of Xist early binding. Instead, the
authors considered the three-dimensional
organization of the genome by using pub-
lished data from “Hi-C” ( 5), a high-through-
put “chromosome conformation capture”
technology that allows identifi cation of DNA
sequences in close proximity to each other
within the nucleus. The authors discovered
that the early binding sites for Xist correspond
to loci that are spatially close to the Xist tran-
scription site. This suggested that Xist might
nd its targets by binding sequences nearby
in space, rather than those that are close along
the linear sequence. Importantly, the authors
tested their hypothesis by expressing Xist
from a different position on the X chromo-
some and found a new pattern of Xist early
binding sites that corresponded to sites in
close spatial proximity to the new location.
From these early binding sites, Engreitz
et al. show that Xist requires its A-repeats to
spread across and silence active genes. The
A-repeats are required for PRC2 recruitment
( 6) and for repositioning active genes into the
Xist silencing compartment ( 4). A model is
thus proposed whereby the three-dimensional
chromosome conformation is exploited to
extrude Xist onto its early binding site targets
where it then helps to modify and reorganize
the X-chromosome architecture.
Several key questions remain regarding
the secondary spreading of Xist and con-
trol mechanisms to prevent inappropriate
spreading across genes that escape inactiva-
tion or from transferring to other chromo-
somes. Engreitz et al. found sharp transitions
of Xist binding surrounding escape genes,
which may relate to previous work implicat-
ing CCCTC-binding factor (CTCF) in block-
ing Xist spreading ( 7). In addition to tran-
scription, CTCF controls chromatin archi-
tecture by binding together strands of DNA
and forming chromatin loops. CTCF binding
sites that are adjacent to escape genes may
play a role in segregating these regions from
Xist binding. Preventing spread to other chro-
mosomes requires the nuclear matrix protein
heterogeneous nuclear ribonucleoprotein U
(hnRNP U) ( 8), which may facilitate transfer
of Xist from the transcription site to contact-
ing regions. Although hnRNP U is located
throughout the nucleus, differences in con-
Long Noncoding RNAs Xist
in Three Dimensions
MOLECULAR BIOLOGY
Andrew Dimond and Peter Fraser
The three-dimensional organization of the
mammalian genome spatially guides the
binding of an RNA to loci for silencing gene
expression.
?
?
?
X
i
s
t
X chromosome
Long
noncoding
RNA
Nucleus
Chromosomes
Organizing silence. The Xist long noncoding RNA
binds to sites in close spatial proximity on the X chro-
mosome. A mechanism based on the three-dimen-
sional genome architecture could be a general strat-
egy for targeting long noncoding RNAs.
CREDIT: V. ALTOUNIAN/SCIENCE
Nuclear Dynamics Programme, The Babraham Institute,
Cambridge, UK. E-mail: peter.fraser@babraham.ac.uk
Published by AAAS
on February 6, 2015www.sciencemag.orgDownloaded from on February 6, 2015www.sciencemag.orgDownloaded from
www.sciencemag.org SCIENCE VOL 341 16 AUGUST 2013 721
PERSPECTIVES
The supply, storage, and fl ow of water
under an ice sheet are crucial for its
overall dynamics. The density differ-
ence between ice and water limits subglacial
water pressure to the pressure from overlying
ice. Near this limit, the ice gradually becomes
decoupled from the base (see the figure),
water-filled cavities form, and sediments
become weakened. The velocity of ice mov-
ing over the bedrock increases due to reduced
friction and faster sediment deformation.
Subglacial water pressure is therefore the key
parameter that controls basal motion, ice fl ux,
and the future evolution of an ice sheet. On
page 777 of this issue ( 1), Meierbachtol et al.
present a detailed record of water pressure
variability under one region of the Greenland
ice sheet.
Glacier hydrology has been investigated in
great detail on mountain glaciers, providing
insights into their drainage over the course
of the melt season ( 25). However, similar
measurements have been mostly missing on
the ice sheets. Only very recently, dye-trac-
ing experiments provided information on the
evolution of the hydraulic properties of sub-
glacial drainage ( 6, 7) and their link to surface
velocity variations ( 8). The reason for the
sparse observational data are the diffi culty
of investigating an extended karst-like sys-
tem that can change its characteristics within
days and that is drained by big, violent, and
sediment-laden rivers. Some of the biggest
such rivers are invisible as they emerge from
the ice sheet at great depth in deeply incised
fjords fi lled with icebergs. Directly explor-
ing the under-ice environment is a diffi cult
endeavor because of thick ice, rapidly freez-
ing boreholes, and ice fl ow that can stretch
and rip instrumentation cables.
Meierbachtol et al. explore subglacial con-
ditions on a transect from the Greenland ice
sheet’s margin to its interior, sampling water
pressure at several drill sites. Their main fi nd-
ing is the different characteristics of subgla-
cial water drainage between marginal and
inland areas of the ice sheet. Whereas bore-
holes close to the ice sheet margin showed
large diurnal variations of water pressure, the
inland sites showed persistent high water pres-
sure with small fl uctuations. Their interpreta-
tion is that an effi cient system of high-capac-
ity channels exists under the marginal parts of
Gauging Greenland’s
Subglacial Water
GEOPHYSICS
Martin Lüthi
Subglacial water fl ow regimes differ between
the interior and the margins of the Greenland
Ice Sheet.
CREDIT: MARTIN LÜTHI
Laboratory of Hydraulics, Hydrology and Glaciology (VAW),
ETH Zürich, 8093 Zürich, Switzerland. E-mail: luethi@vaw.
baug.ethz.ch
Freshly exposed, glacially carved bedrock. Sediment-covered areas and solid bedrock form the base of the
Greenland ice sheet. Scratch marks from rocks dragged along this freshly exposed bedrock in West Greenland
illustrate the sliding motion of ice over its base. Meierbachtol et al. provide insights into the mechanisms of
water fl ow, which controls sliding motion.
tact frequency between cis-linked loci and
interchromosomal interactions may inhibit
Xist from establishing a foothold on other
chromosomes. The biggest gaps still to fi ll
are details that explain how Xist eventually
encompasses the entire chromosome. Does
Xist spread from early sites into adjacent
sites by diffusion or transfer across a fairly
static chromosome structure, or do chromo-
some dynamics allow other binding sites to
iteratively contact and collect RNA from the
Xist locus to eventually produce the broad
distribution seen in all cells? Conformational
studies at later time points of X-inactivation
or live-cell studies could potentially provide
a timeline to fi ll these gaps in RNA and chro-
matin dynamics.
Other long noncoding RNAs ( 911) have
been described that exert their regulatory
effects by exploiting the three-dimensional
folding of the genome. The similarities with
Xist suggest that this may be a general tar-
geting mechanism (see the figure). It will
be interesting to see if other, trans-acting
noncoding RNAs (both long and short) also
exploit three-dimensional genome organiza-
tion to target loci on other chromosomes ( 12,
13). With tools to map RNA binding sites and
describe genome conformation now at hand,
answers to these questions are within reach.
References
1. V. A. Moran, R. J. Perera, A. M. Khalil, Nucleic Acids Res.
40, 6391 (2012).
2. J. M. Engreitz et al., Science 341, 1237973 (2013);
10.1126/science.1237973.
3. J. T. Lee, Nat. Rev. Mol. Cell Biol. 12, 815 (2011).
4. J. Chaumeil, P. Le Baccon, A. Wutz, E. Heard, Genes Dev.
20, 2223 (2006).
5. J. R. Dixon et al., Nature 485, 376 (2012).
6. J. Zhao, B. K. Sun, J. A. Erwin, J.-J. Song, J. T. Lee, Science
322, 750 (2008).
7. G. N. Filippova et al., Dev. Cell 8, 31 (2005).
8. Y. Hasegawa et al., Dev. Cell 19, 469 (2010).
9. K. C. Wang et al., Nature 472, 120 (2011).
10. P. G. Maass et al., J. Clin. Invest. 122, 3990 (2012).
11. F. Lai et al., Nature 494, 497 (2013).
12. J. L. Rinn et al., Cell 129, 1311 (2007).
13. M. Guttman et al., Nature 477, 295 (2011).
10.1126/science.1243257
Published by AAAS
DOI: 10.1126/science.1243257
, 720 (2013);341 Science
Andrew Dimond and Peter Fraser
Long Noncoding RNAs Xist in Three Dimensions
This copy is for your personal, non-commercial use only.
clicking here.colleagues, clients, or customers by , you can order high-quality copies for yourIf you wish to distribute this article to others
here.following the guidelines can be obtained byPermission to republish or repurpose articles or portions of articles
): February 6, 2015 www.sciencemag.org (this information is current as of
The following resources related to this article are available online at
http://www.sciencemag.org/content/341/6147/720.full.html
version of this article at: including high-resolution figures, can be found in the onlineUpdated information and services,
http://www.sciencemag.org/content/341/6147/720.full.html#related
found at: can berelated to this article A list of selected additional articles on the Science Web sites
http://www.sciencemag.org/content/341/6147/720.full.html#ref-list-1
, 4 of which can be accessed free:cites 13 articlesThis article
http://www.sciencemag.org/content/341/6147/720.full.html#related-urls
2 articles hosted by HighWire Press; see:cited by This article has been
http://www.sciencemag.org/cgi/collection/molec_biol
Molecular Biology subject collections:This article appears in the following
registered trademark of AAAS. is aScience2013 by the American Association for the Advancement of Science; all rights reserved. The title CopyrightAmerican Association for the Advancement of Science, 1200 New York Avenue NW, Washington, DC 20005.
(print ISSN 0036-8075; online ISSN 1095-9203) is published weekly, except the last week in December, by theScience
on February 6, 2015www.sciencemag.orgDownloaded from
... First, lncRNAs may modulate the action of the protein-coding genes involved in epigenetic patterning and chromatin remodeling or function as scaffolds. A plant lncRNA, COLDAIR, acts in the same way as Xist [72], which serves as scaffolds for the recruitment of PRC2 complexes to specific loci and induces epigenetic silencing [73]. Second, transcription or splicing of lncRNA may modulate gene expression in cis. ...
Article
Full-text available
MdPG1 encoding polygalacturonase in apple (Malus × domestica) is a key gene associated with fruit firmness and texture variations among apple cultivars. However, the causative variants of MdPG1 are still not known. In this study, we identified a SNPA/C variant within an ERF-binding element located in the promoter region of MdPG1. The promoter containing the ERF-binding element with SNPA, rather than the SNPC, could be strongly bound and activated by MdCBF2, a member of the AP2/ERF transcription factor family, as determined by yeast-one-hybrid and dual-luciferase reporter assays. We also demonstrated that the presence of a novel long non-coding RNA, lncRNAPG1, in the promoter of MdPG1 was a causative variant. lncRNAPG1 was specifically expressed in fruit tissues postharvest. lncRNAPG1 could reduce promoter activity when it was fused to the promoter of MdPG1 and a tobacco gene encoding Mg-chelatase H subunit (NtCHLH) in transgenic tobacco cells but could not reduce promoter activity when it was supplied in a separate gene construct, indicating a cis-regulatory effect. Our results provide new insights into genetic regulation of MdPG1 allele expression and are also useful for the development of elite apple cultivars.
... They are predominantly found in the nucleus, are rapidly degraded by exosomes, and were thus originally regarded as transcriptional noise (Struhl 2007). However, recent studies suggest that lncRNAs play important roles in the epigenetic regulation of drug susceptibility in fungi, which will be discussed here (Mikkelsen et al. 2007;Moran et al. 2012;Dimond and Fraser 2013). ...
Article
Global estimates suggest that over 300 million individuals of all ages are affected by serious fungal infections every year, culminating in about 1.7 million deaths. The societal and economic burden on the public health sector due to opportunistic fungal pathogens is quite significant, especially among immunocompromised patients. Despite the high clinical significance of these infectious agents, treatment options are limited with only three major classes of antifungal drugs approved for use. Clinical management of fungal diseases is further compromised by the emergence of antifungal resistant strains. Transcriptional and genetic mechanisms that control drug resistance in human fungal pathogens are well-studied and include drug target alteration, upregulation of drug efflux pumps as well as changes in drug affinity and abundance of target proteins. In this review, we highlight several recently discovered novel post-transcriptional mechanisms that control antifungal resistance, which involve regulation at the translational, post-translational, epigenetic, and mRNA stability levels. The discovery of many of these novel mechanisms has opened new avenues for the development of more effective antifungal treatment strategies and new insights, perspectives, and future directions that will facilitate this process are discussed.
... Here, we found that lncR-125b has 4,424 base pairs, located on chromosome 1 in goat, and has one potential binding site with chi-miR-125b, using the BLAST tool in the Ensembl database (Zerbino et al., 2018) (Figure 1A and Figure S2). Coding potential calculator (CPC) prediction showed that lncR-125b has very low coding potential, similar to that of XIST, a well-known lncRNA (Dimond and Fraser, 2013) (Figure 1B). Next, we examined its expression patterns in different tissues, including heart, liver, spleen, lung, kidney, and LTL muscle. ...
Article
Full-text available
Long noncoding RNAs (lncRNAs) have emerged as essential regulators of skeletal myogenesis, but few myogenesis-associated lncRNAs have been identified and our understanding of their regulatory mechanisms remains limited, particularly in goat. Here, we identified a novel lncRNA, TCONS_00006810 (named lncR-125b), from our previous lncRNA sequencing data on fetal (45, 60, and 105 days of gestation, three biological replicates for each point) and postnatal (3 days after birth, n = 3) goat skeletal muscle, and found that it is highly expressed in skeletal muscle and gradually upregulated during skeletal muscle satellite cell (SMSC) differentiation in goat. Notably, overexpression of lncR-125b accelerated the expression of myogenic differentiation 1 (MyoD 1) and myogenin (MyoG), and the formation of myotubes, and knockdown of lncR-125b showed opposite effects in SMSCs. Results of dual-luciferase assay and quantitative real-time polymerase chain reaction revealed that lncR-125b acts as a molecular sponge for miR-125b and that insulin-like growth factor 2 (IGF2), a critical regulator of skeletal myogenesis, is a direct target gene of miR-125b. Further analyses showed that lncR-125b negatively regulates miR-125b expression and positively regulates IGF2 expression in SMSCs. Mechanistically, lncR-125b promotes SMSC differentiation by functioning as a competing endogenous RNA (ceRNA) for miR-125b to control IGF2 expression. These findings identify lncR-125b as a novel noncoding regulator of muscle cell differentiation and skeletal muscle development in goat.
... Long-chain noncoding RNA (LncRNAs) play key roles in different metabolic pathways in yeast, plant and animals. Recent investigations indicated that they are closely associated with cell differentiation and development, cancer progression, immune response gene mediation, silencing gene expression and neurological diseases occurrence (Carpenter et al., 2013;Dimond and Fraser, 2013;Fatica and Bozzoni, 2014;Guttman et al., 2011;Pauli et al., 2011;Wang et al., 2016b;Yang et al., 2014). In plants, LncRNAs were described to modulate the male sterility of hybrid rice and increased its yield (Ding et al., 2012). ...
... Regulatory ncRNAs are usually classified as small non-coding RNAs and long noncoding RNAs (lncRNAs) according to their lengths ( Shu and Hu, 2010;Pauli et al., 2011). lncRNAs play important roles in cell differentiation and development (Guttman et al., 2011;Pauli et al., 2011;Fatica and Bozzoni, 2014) silencing gene expression in X-chromosome (Dimond and Fraser, 2013), neurological diseases occurrence ( Ponting et al., 2009), cancer progression ( Wapinski and Chang, 2011;Yang et al., 2014) and immune response genes mediation (Carpenter et al., 2013;Wang et al., 2016). In plants, lncRNAs express differentially in various organs and under different treatment conditions, which indicates lncRNAs can modulate gene activity during development and in response to external stimuli ( Kim and Sung, 2012). ...
Article
Full-text available
This article summarizes the current knowledge regarding omics approaches, which include genomics, transcriptomics, proteomics and metabolomics, in the context of bio-hydrogen production in Chlamydomonas reinhardtii. In this paper, critical genes (HydA1, Hyd A2, Sulp, Tla1, Sta7, PFL1) involved in H2 metabolism were identified and analyzed for their function in H2 accumulation. Furthermore, the advantages of gene microarrays and RNA-seq were compared, as well as their applications in transcriptomic analysis of H2 production. Moreover, as a useful tool, proteomic analysis could identify different proteins that participate in H2 metabolism. This review provides fundamental theory and an experimental basis for H2 production, and further research effort is needed in this field.
... Therefore, they have previously been considered transcriptional noise (Struhl, 2007). However, several studies have revealed that lncRNAs play roles in epigenetic gene regulation (Dimond and Fraser, 2013;Mikkelsen et al., 2007;Moran et al., 2012). ...
Article
The emergence of drug-resistant fungi poses a continuously increasing threat to human health. Despite advances in preventive care and diagnostics, resistant fungi continue to cause significant mortality, especially in immunocompromised patients. Therapeutic resources are further limited by current usage of only four major classes of antifungal drugs. Resistance against these drugs has already been observed in pathogenic fungi requiring the development of much needed newer antifungal drugs. Epigenetic changes such as DNA or chromatin modifications alter gene expression levels in response to certain stimuli, including interaction with the host in the case of fungal pathogens. These changes can confer resistance to drugs by altering the expression of target genes or genes encoding drug efflux pumps. Multiple pathogens share many of these epigenetic pathways; thus, targeting epigenetic pathways might also identify drug target candidates for the development of broad-spectrum antifungal drugs. In this review, we discuss the importance of epigenetic pathways in mediating drug resistance in fungi as well as in the development of anti-fungal drugs.
Chapter
Endophytic fungi are ubiquitous microorganisms that reside within healthy plant tissues without causing any apparent harm to the host. The myriad of diverse pharmacological properties makes these fungi a valuable source of bioactive compounds. Research on fungal endophytes has gained significant momentum in recent years, with an increasing focus on their potential for pharmaceuticals. The present scenario of endophytic fungi’s pharmacological potential involves identifying novel compounds, elucidating their mechanisms of action, and evaluating their efficacy and safety. Numerous studies have documented the discovery of endophytic fungi bioactive metabolites with potent immunomodulatory, antimicrobial, anticancer, and antioxidant properties. The demand for such compounds in the pharmaceutical industry is high owing to their therapeutic potential and low toxicity. Improving the yield and quality of bioactive compounds from endophytic fungi is crucial for meeting market demand. Various approaches, including genetic, epigenetic, and physicochemical methods, have been employed to enhance the production of bioactive metabolites. Genetic manipulation of fungal endophytes using techniques such as CRISPR/Cas9 and RNA interference has shown promising results in increasing the yield of biologically active compounds. Epigenetic modifications and physicochemical treatments such as elicitation and fermentation optimization have also been explored to improve bioactive metabolite production. Endophytic fungi have enormous pharmacological potential, which has substantial implications for drug research and development. The market need for such compounds can be met by increasing the production of bioactive substances from endophytic fungi utilizing a variety of methods.
Article
Full-text available
Many researches indicated that long non-coding RNAs (lncRNAs) were involved in the malignant progression of tumors, including Adrenocortical Carcinoma (ACC). However, as for most lncRNAs, their biological behaviors and molecular mechanism remain unclear in ACC. In the present research, weighted gene co-expression network analysis (WGCNA) was used to identify pathologically relevant gene, including lncRNAs. By comparing their expressions in GSE61359 tumors and normal controls, long intergenic non-protein coding RNA 1234 (LINC01234) was selected to investigate the clinical significance, biological function, and mechanism in ACC. Data mining revealed that LINC01234 expression was significantly up-regulated in ACC patients, and a shorter survival time presents in patients with higher LINC01234 expression compared to that in patients with lower LINC01234 expression. Further, LINC01234 silencing resulted in cells growth arrest in vitro and in vivo. Mechanism studies suggested that LINC01234 silencing induced cell cycle arrest, and bromodomain-containing protein 4 (BRD4) overexpression could restore this phenomenon. Further research showed that LINC01234 could mediate BRD4 expression through competitively sequestering microRNA (miR)-140-3p, as evidenced by the positive correlation of LINC01234 with BRD4 and inverse correlation with miR-140-3p expression. Luciferase activity assay also verified the targeting relationship between LINC01234, BRD4 and miR-140-3p. And up-regulated LINC01234 in ACC cells significantly reversed the degradation of BRD4 by miR-140-3p. Collectively, we deduce that LINC01234 functions as a ceRNA to regulate BRD4 expression by sponging miR-140-3p in ACC progress. Our findings have the potential to provide a new target for the diagnosis and treatment of ACC.
Article
Full-text available
Reprogrammed glucose metabolism of enhanced aerobic glycolysis, also known as Warburg effect, which exerts a significant contributor to cancer progression, is regarded as a hallmark of cancer. The roles of long noncoding RNAs (lncRNA) in regulating cancer via metabolic reprogramming are mostly unknown, including esophagal cancer (EC). Here, we showed that how the lncRNA urothelial carcinoma associated 1 (UCA1) exerts pro-oncogene in regulating EC glucose metabolism. Firstly, we found that upregulated UCA1 expression enhances the malignant phenotypes of EC, including poor outcome, larger tumor size, positive lymphatic invasion, and advanced pathological stages. UCA1 silencing could suppress EC cell proliferation and metastasis. Following, bioinformatics analyses revealed that UCA1 regulated the HK2 expression through functioning as a competing endogenous RNA (ceRNA). Mechanistically, UCA1 overexpression could elevate the activation of HK2 oncogenes via inhibition of miR-203 activity, as evidenced by the positive correlation of UCA1 with HK2 and inverse correlation with miR-203 expression. Luciferase activity assay further verified the targeting relationship between UCA1, miR-203, and HK2. Upregulated UCA1 in EC cells significantly suppressed the degradation of HK2 by miR-203. Further research showed that upregulated UCA1 effectively increased the rate of glucose uptake, lactate output, and ECAR value, all of which can be attenuate by HK2 interference and 2-DG, whereas knockdown of UCA1 had the opposite effect. In sum, our findings suggest that the UCA1/miR-203/HK2 axis contributes to EC development by reprogramming tumor glucose metabolism, providing new insight into the management of EC patients.
Article
Full-text available
Many large noncoding RNAs (lncRNAs) regulate chromatin, but the mechanisms by which they localize to genomic targets remain unexplored. We investigated the localization mechanisms of the Xist lncRNA during X-chromosome inactivation (XCI), a paradigm of lncRNA-mediated chromatin regulation. During the maintenance of XCI, Xist binds broadly across the X chromosome. During initiation of XCI, Xist initially transfers to distal regions across the X chromosome that are not defined by specific sequences. Instead, Xist identifies these regions by exploiting the three-dimensional conformation of the X chromosome. Xist requires its silencing domain to spread across actively transcribed regions and thereby access the entire chromosome. These findings suggest a model in which Xist coats the X chromosome by searching in three dimensions, modifying chromosome structure, and spreading to newly accessible locations.
Article
Full-text available
Recent advances in genomic research have revealed the existence of a large number of transcripts devoid of protein-coding potential in multiple organisms. Although the functional role for long non-coding RNAs (lncRNAs) has been best defined in epigenetic phenomena such as X-chromosome inactivation and imprinting, different classes of lncRNAs may have varied biological functions. We and others have identified a class of lncRNAs, termed ncRNA-activating (ncRNA-a), that function to activate their neighbouring genes using a cis-mediated mechanism. To define the precise mode by which such enhancer-like RNAs function, we depleted factors with known roles in transcriptional activation and assessed their role in RNA-dependent activation. Here we report that depletion of the components of the co-activator complex, Mediator, specifically and potently diminished the ncRNA-induced activation of transcription in a heterologous reporter assay using human HEK293 cells. In vivo, Mediator is recruited to ncRNA-a target genes and regulates their expression. We show that ncRNA-a interact with Mediator to regulate its chromatin localization and kinase activity towards histone H3 serine 10. The Mediator complex harbouring disease- displays diminished ability to associate with activating ncRNAs. Chromosome conformation capture confirmed the presence of DNA looping between the ncRNA-a loci and its targets. Importantly, depletion of Mediator subunits or ncRNA-a reduced the chromatin looping between the two loci. Our results identify the human Mediator complex as the transducer of activating ncRNAs and highlight the importance of Mediator and activating ncRNA association in human disease.
Article
Full-text available
Translocations are chromosomal rearrangements that are frequently associated with a variety of disease states and developmental disorders. We identified 2 families with brachydactyly type E (BDE) resulting from different translocations affecting chromosome 12p. Both translocations caused downregulation of the parathyroid hormone-like hormone (PTHLH) gene by disrupting the cis-regulatory landscape. Using chromosome conformation capturing, we identified a regulator on chromosome 12q that interacts in cis with PTHLH over a 24.4-megabase distance and in trans with the sex-determining region Y-box 9 (SOX9) gene on chromosome 17q. The element also harbored a long noncoding RNA (lncRNA). Silencing of the lncRNA, PTHLH, or SOX9 revealed a feedback mechanism involving an expression-dependent network in humans. In the BDE patients, the human lncRNA was upregulated by the disrupted chromosomal association. Moreover, the lncRNA occupancy at the PTHLH locus was reduced. Our results document what we believe to be a novel in cis- and in trans-acting DNA and lncRNA regulatory feedback element that is reciprocally regulated by coding genes. Furthermore, our findings provide a systematic and combinatorial view of how enhancers encoding lncRNAs may affect gene expression in normal development.
Article
Full-text available
The spatial organization of the genome is intimately linked to its biological function, yet our understanding of higher order genomic structure is coarse, fragmented and incomplete. In the nucleus of eukaryotic cells, interphase chromosomes occupy distinct chromosome territories, and numerous models have been proposed for how chromosomes fold within chromosome territories. These models, however, provide only few mechanistic details about the relationship between higher order chromatin structure and genome function. Recent advances in genomic technologies have led to rapid advances in the study of three-dimensional genome organization. In particular, Hi-C has been introduced as a method for identifying higher order chromatin interactions genome wide. Here we investigate the three-dimensional organization of the human and mouse genomes in embryonic stem cells and terminally differentiated cell types at unprecedented resolution. We identify large, megabase-sized local chromatin interaction domains, which we term 'topological domains', as a pervasive structural feature of the genome organization. These domains correlate with regions of the genome that constrain the spread of heterochromatin. The domains are stable across different cell types and highly conserved across species, indicating that topological domains are an inherent property of mammalian genomes. Finally, we find that the boundaries of topological domains are enriched for the insulator binding protein CTCF, housekeeping genes, transfer RNAs and short interspersed element (SINE) retrotransposons, indicating that these factors may have a role in establishing the topological domain structure of the genome.
Article
Full-text available
The recent discovery that the human and other mammalian genomes produce thousands of long non-coding RNAs (lncRNAs) raises many fascinating questions. These mRNA-like molecules, which lack significant protein-coding capacity, have been implicated in a wide range of biological functions through diverse and as yet poorly understood molecular mechanisms. Despite some recent insights into how lncRNAs function in such diverse cellular processes as regulation of gene expression and assembly of cellular structures, by and large, the key questions regarding lncRNA mechanisms remain to be answered. In this review, we discuss recent advances in understanding the biology of lncRNAs and propose avenues of investigation that may lead to fundamental new insights into their functions and mechanisms of action. Finally, as numerous lncRNAs are dysregulated in human diseases and disorders, we also discuss potential roles for these molecules in human health.
Article
Full-text available
Although thousands of large intergenic non-coding RNAs (lincRNAs) have been identified in mammals, few have been functionally characterized, leading to debate about their biological role. To address this, we performed loss-of-function studies on most lincRNAs expressed in mouse embryonic stem (ES) cells and characterized the effects on gene expression. Here we show that knockdown of lincRNAs has major consequences on gene expression patterns, comparable to knockdown of well-known ES cell regulators. Notably, lincRNAs primarily affect gene expression in trans. Knockdown of dozens of lincRNAs causes either exit from the pluripotent state or upregulation of lineage commitment programs. We integrate lincRNAs into the molecular circuitry of ES cells and show that lincRNA genes are regulated by key transcription factors and that lincRNA transcripts bind to multiple chromatin regulatory proteins to affect shared gene expression programs. Together, the results demonstrate that lincRNAs have key roles in the circuitry controlling ES cell state.
Article
Full-text available
The genome is extensively transcribed into long intergenic noncoding RNAs (lincRNAs), many of which are implicated in gene silencing. Potential roles of lincRNAs in gene activation are much less understood. Development and homeostasis require coordinate regulation of neighbouring genes through a process termed locus control. Some locus control elements and enhancers transcribe lincRNAs, hinting at possible roles in long-range control. In vertebrates, 39 Hox genes, encoding homeodomain transcription factors critical for positional identity, are clustered in four chromosomal loci; the Hox genes are expressed in nested anterior-posterior and proximal-distal patterns colinear with their genomic position from 3' to 5'of the cluster. Here we identify HOTTIP, a lincRNA transcribed from the 5' tip of the HOXA locus that coordinates the activation of several 5' HOXA genes in vivo. Chromosomal looping brings HOTTIP into close proximity to its target genes. HOTTIP RNA binds the adaptor protein WDR5 directly and targets WDR5/MLL complexes across HOXA, driving histone H3 lysine 4 trimethylation and gene transcription. Induced proximity is necessary and sufficient for HOTTIP RNA activation of its target genes. Thus, by serving as key intermediates that transmit information from higher order chromosomal looping into chromatin modifications, lincRNAs may organize chromatin domains to coordinate long-range gene activation.
Article
Full-text available
To equalize X-chromosome dosages between the sexes, the female mammal inactivates one of her two X chromosomes. X-chromosome inactivation (XCI) is initiated by expression of Xist, a 17-kb noncoding RNA (ncRNA) that accumulates on the X in cis. Because interacting factors have not been isolated, the mechanism by which Xist induces silencing remains unknown. We discovered a 1.6-kilobase ncRNA (RepA) within Xist and identified the Polycomb complex, PRC2, as its direct target. PRC2 is initially recruited to the X by RepA RNA, with Ezh2 serving as the RNA binding subunit. The antisense Tsix RNA inhibits this interaction. RepA depletion abolishes full-length Xist induction and trimethylation on lysine 27 of histone H3 of the X. Likewise, PRC2 deficiency compromises Xist up-regulation. Therefore, RepA, together with PRC2, is required for the initiation and spread of XCI. We conclude that a ncRNA cofactor recruits Polycomb complexes to their target locus.
Article
The discovery of X-chromosome inactivation (XCI) celebrated its golden anniversary this year. Originally offered as an explanation for the establishment of genetic equality between males and females, 50 years on, XCI presents more than a curious gender-based phenomenon that causes silencing of sex chromosomes. How have the mysteries of XCI unfolded? And what general lessons can be extracted? Several of the cell biological mechanisms that are used to establish the inactive X chromosome, including regulatory networks of non-coding RNAs and unusual nuclear dynamics, are now suspected to hold true for processes occurring on a genome-wide scale.
Article
In XX female mammals, one of the two X chromosomes is epigenetically inactivated to equalize gene expression with XY males. The formation of the inactive X chromosome (Xi) is regulated by an X-linked long noncoding RNA Xist, which accumulates on the entire length of the chromosome in cis and induces heterochromatin formation. However, the mechanism by which Xist RNA "paints" the Xi has long remained elusive. Here, we show that a matrix protein hnRNP U/SP120/SAF-A is required for the accumulation of Xist RNA on the Xi. Xist RNA and hnRNP U interact and upon depletion of hnRNP U, Xist RNA is detached from the Xi and diffusely localized into the nucleoplasm. In addition, ES cells lacking hnRNP U expression fail to form the Xi. We propose that the association with matrix proteins is an essential step in the epigenetic regulation of gene expression by Xist RNA.