ArticlePDF AvailableLiterature Review

Abstract and Figures

R-loops are cellular structures composed of an RNA/DNA hybrid, which is formed when the RNA hybridises to a complementary DNA strand and a displaced single-stranded DNA. R-loops have been detected in various organisms from bacteria to mammals and play crucial roles in regulating gene expression, DNA and histone modifications, immunoglobulin class switch recombination, DNA replication, and genome stability. Recent evidence suggests that R-loops are also involved in molecular mechanisms of neurological diseases and cancer. In addition, mutations in factors implicated in R-loop biology, such as RNase H and SETX (senataxin), lead to devastating human neurodegenerative disorders, highlighting the importance of correctly regulating the level of R-loops in human cells. In this review we summarise current advances in this field, with a particular focus on diseases associated with dysregulation of R-loop structures. We also discuss potential therapeutic approaches for such diseases and highlight future research directions.
The diagram depicts the role of R-loops in human diseases. Loss of wild type protein function is depicted by red crosses. A. Ataxia and motor neuron diseases. Mutations in human RNA/DNA helicase senataxin are associated with AOA2/ALS4 disorders and lead to R-loop accumulation and defects in transcriptional termination by Pol II [16], the maintenance of genome integrity [46], meiotic recombination during spermatogenesis, gene silencing during meiotic sex chromosome inactivation [14], and neuronal differentiation [49]. B. Aicardi-Goutières syndrome (AGS). AGS is associated with mutations in all three subunits of RNase H2, ssDNA 3′–5′ exonuclease TREX1 (DNASEIII), dsRNA-editing enzyme ADAR1, and dNTP triphosphatase SAMHD1; these trigger accumulation of unprocessed nucleic acids, including genomic DNA with incorporated ribonucleotides, R-loops, and retroelement-derived nucleic acids, and result in the immune response characteristic of AGS [65]. C. Trinucleotide expansion diseases. R-loops form over expanded repeats and result in decreased initiation and elongation of RNA Pol II and formation of repressive chromatin marks, which silence the host gene containing expanded repeats [75]. D. Genome instability in cancer. Loss of proteins protecting against abnormal R-loop accumulation, such as FIP1L1, leads to genome instability, one hallmark of cancer [31]. Yellow stars denote double-stranded DNA breaks. E. AID-mediated mutagenesis and translocations in cancer. Single-stranded DNA in R-loops is a substrate for cytidine deamination by activation-induced cytidine deaminase, leading to mutagenesis as indicated by orange stars [21], [88]. These mutations can cause DSB formation, leading to chromosomal translocations. The IgH/c-MYC translocation brings the strong IgH enhancers, shown as yellow box, close to c-MYC, leading to its overexpression in Burkitt's lymphoma [87]. Transcription of IgH/c-MYC starts from a previously inactive promoter downstream of the translocation break point. The IgH locus is depicted in blue, c-MYC gene is in grey. The translocation breakpoint is indicated by a dashed black line. F. Senescence. R-loops formed by the noncoding RNA TERRA accumulate at telomeres in cells deficient of Hpr1 and RNase H. In the absence of telomerase, these R-loops promote Rad52-dependent telomere elongation and delayed senescence. In the absence of telomerase and Rad52, R-loops promote telomere shortening and premature senescence [94].
… 
Content may be subject to copyright.
Review
Out of Balance: R-loops in Human Disease
Matthias Groh, Natalia Gromak*
Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
Abstract: R-loops are cellular structures composed of an
RNA/DNA hybrid, which is formed when the RNA
hybridises to a complementary DNA strand and a
displaced single-stranded DNA. R-loops have been
detected in various organisms from bacteria to mammals
and play crucial roles in regulating gene expression, DNA
and histone modifications, immunoglobulin class switch
recombination, DNA replication, and genome stability.
Recent evidence suggests that R-loops are also involved in
molecular mechanisms of neurological diseases and
cancer. In addition, mutations in factors implicated in R-
loop biology, such as RNase H and SETX (senataxin), lead
to devastating human neurodegenerative disorders,
highlighting the importance of correctly regulating the
level of R-loops in human cells. In this review we
summarise current advances in this field, with a particular
focus on diseases associated with dysregulation of R-loop
structures. We also discuss potential therapeutic ap-
proaches for such diseases and highlight future research
directions.
Introduction
R-loops are three-stranded structures, which form when RNA
hybridises to a complementary DNA strand, forming an RNA/
DNA hybrid, resulting in displacement of the other DNA strand in
this process (Figure 1). The first R-loops were described in 1976,
when their formation in vitro in the presence of 70% formamide
was visualised by electron microscopy (Figure 1) [1]. These
structures were thermodynamically more stable than duplex
DNA, and they remained intact following removal of formamide.
This technique of RNA/DNA hybridisation has been used in over
140 studies to map gene organisation, transcription initiation sites,
and the direction of transcription, as well as measure the quantities
of cellular RNAs [2].
The first evidence for R-loop formation in live bacteria was
obtained in 1994 [3]. This was followed by numerous studies
showing that R-loops exist in different organisms (Figure 1) [4–6].
In living cells, R-loops are thought to form in cis during
transcription, when nascent RNA hybridises to the DNA template
behind the elongating RNA polymerase (Pol) [4]. However, in
contrast to this popular view of cotranscriptional R-loops, recent
studies suggest that RNA transcribed at one locus can hybridise to
homologous DNA at another locus, thus leading to R-loop
formation in trans [7]. In the last five years, the use of an antibody
(S9.6) recognising RNA/DNA hybrids has revolutionised the R-
loop field [8]. Initially, the S9.6 antibody, which detects hybrids as
small as six bp with an affinity of 0.6 nM, was developed as a tool
to enhance the DNA/RNA hybridisation signal in DNA
microarray studies [9,10]. More recently, it has been used to
detect R-loops in vivo and uncover their contribution to
fundamental biological processes in yeast [11,12], plants [13],
mice [14,15], and humans [16–18].
The picture emerging from these studies suggests that R-loops
can be both beneficial and deleterious to cells. Paradoxically, while
they are required for important biological processes, they can also
promote DNA damage and genome instability. In particular, R-
loops have been shown to play an essential positive function in
Escherichia coli plasmid and human mitochondrial DNA replica-
tion [19,20] and during immunoglobulin class switch recombina-
tion, which contributes to the antibody isotype diversity in
activated B cells [21]. R-loops form on many genes in yeast and
human cells [18,22] and have been implicated in regulation of
gene expression. R-loops can repress transcription and promote
transcriptional termination [16,23,24]. Furthermore, R-loops are
clearly associated with epigenetic mechanisms governing tran-
scription, including DNA methylation and posttranslational
histone modifications [18,25–27]. In spite of this growing list of
beneficial R-loop functions, it is also evident that R-loops can be a
dangerous source of DNA damage. They can sensitize DNA to
damaging agents [28], induce transcription-associated recombi-
nation [24], double-strand breaks (DSBs) [29,30], chromosome
breaks, and fragile site instability [31–33], and cause chromosome
loss [34]. Therefore, cells need to tightly regulate the levels of R-
loops to exploit their unique features. Altering the physiological R-
loop balance can impair R-loop-regulated processes, cause
genome instability, and may lead to human diseases. Consequent-
ly, defining the roles of R-loops in the multitude of biological
processes and human disease is likely to develop into one of the
most important and influential areas of R-loop research in the
future.
Proteins in R-loop Biology
The number of proteins associated with R-loop biology has
increased in the last few years, reflecting the diversity of R-loop
processes (Table S1) [4–6]. Many proteins can regulate cellular R-
loop levels either directly or indirectly, mostly by preventing RNA
from hybridising to DNA, thus reducing excessive R-loop
accumulation. Among these are proteins required for efficient
transcriptional elongation, termination, polyadenylation, RNA
splicing, packaging, and export [16,24,28,30,31,34,35]. DNA
topology itself can influence hybridisation of RNA to DNA, and
topoisomerases consequently play important roles in modulating
Citation: Groh M, Gromak N (2014) Out of Balance: R-loops in Human
Disease. PLoS Genet 10(9): e1004630. doi:10.1371/journal.pgen.1004630
Editor: Nancy Maizels, University of Washington School of Medicine, United
States of America
Published September 18, 2014
Copyright: ß2014 Groh, Gromak. This is an open-access article distributed
under the terms of the Creative Commons Attribution License, which permits
unrestricted use, distribution, and reproduction in any medium, provided the
original author and source are credited.
Funding: NG is supported by a Royal Society University Research Fellowship and
MRC NIRG (MR/J007870/1). MG is supported by a PhD studentship from Ataxia
UK/Motor Neuron Disease Association (Gromak/Jun11/6278). The funders had no
role in the preparation of the article.
Competing Interests: The authors have declared that no competing interests
exist.
* Email: natalia.gromak@path.ox.ac.uk
PLOS Genetics | www.plosgenetics.org 1 September 2014 | Volume 10 | Issue 9 | e1004630
R-loop levels [27,33]. Proteins involved in maintenance of genome
integrity can also regulate R-loops, suggesting a dynamic interplay
between DNA repair and R-loop formation [7]. Importantly, cells
possess dedicated enzymes, including the members of the RNase
H family that specifically degrade the RNA in R-loops [36], and
helicases that can unwind RNA/DNA hybrids [12,16].
Recent evidence shows that R-loops can directly affect many
gene expression–associated processes, including DNA methyla-
tion, posttranslational histone modifications, and transcription, by
influencing the function of regulatory proteins [16,18,25,26].
Despite the growing number of proteins involved in R-loop
homeostasis and human disease, many questions still remain
unanswered. For many proteins with documented in vitro RNA/
DNA helicase activity (e.g., Pif1, the MCM complex), in vivo
evidence is generally still lacking (Table S1) [37,38]. Moreover, the
molecular mechanisms underlying interactions between proteins
and R-loops are poorly understood, and in many cases the
connections to disease remain obscure.
R-loops and Neurological Diseases
The biological importance of R-loops in humans is supported
by the fact that mutations in proteins implicated in R-loop
resolution cause devastating human diseases, often related to
neurodegeneration. Mutations in the putative RNA/DNA helicase
SETX cause neurodegenerative diseases, the dominant juvenile
form of amyotrophic lateral sclerosis type 4 (ALS4), and a recessive
form of ataxia oculomotor apraxia type 2 (AOA2) (Figure 2A).
These diseases are characterised by progressive degeneration of
motor neurons in the brain and spinal cord, muscle weakness and
atrophy [39–41].
In addition to its predicted function as an RNA/DNA helicase,
SETX interacts with proteins involved in diverse aspects of RNA
metabolism [42]. Moreover, a single amino acid mutation, which
compromises the function of the yeast homologue Sen1, dramat-
ically changes the Pol II distribution genome-wide, further
supporting the view that SETX/Sen1 functions in the regulation
of transcription [43]. Recently, we demonstrated that SETX is
implicated in transcriptional termination by Pol II in humans [16].
It is required to resolve R-loops at termination elements, releasing
RNA for degradation by the 59–39‘‘torpedo’’ exonuclease Xrn2
prior to termination (Figure 2A) [16,44]. Mutations in the yeast
homologue, Sen1, also lead to a transcriptional termination defect,
associated with accumulation of R-loops and genome instability
[12]. In line with its function in R-loop resolution, SETX/Sen1 is
also involved in maintaining genome integrity by coordinating
transcription, DNA replication, and the DNA damage response
[45–47]. SETX can target the 39–59RNA degradation complex,
the exosome, to sites of transcription-induced DNA damage [48].
Furthermore, SETX protects genome integrity by coordinating
meiotic recombination with transcription during spermatogenesis
and gene silencing during meiotic sex chromosome inactivation [14].
In particular, Setx knock-out mice accumulated DNA double strand
breaks and R-loops and failed to disassemble Rad51 filaments. This
resulted in a failure to cross over, likely due to collision between R-
loops with Holliday junctions [14]. These defects in Setx knock-out
mice lead to male infertility, raising the question as to how this relates
to fertility of male AOA2/ALS4 patients.
Studies in neuronal cells have demonstrated a role for SETX in
neuronal differentiation through fibroblast growth factor 8 (FGF8)
signalling, providing one explanation for the effects of loss-of-
function AOA2 mutations [49]. Surprisingly, overexpression of
dominant mutant forms of SETX did not affect neuritogenesis,
suggesting that a different function of SETX may be affected in
ALS4 patients. However, the interplay between the function of
SETX in R-loop resolution, genome maintenance, and neuronal
differentiation is still unclear. In a recent study, Lavin and
colleagues examined cells from mice with disrupted Atm,Tdp1,
Setx,orAptx genes, which cause ataxia telangiectasia (AT),
spinocerebellar ataxia with axonal neuropathy 1 (SCAN1), AOA2,
and ataxia oculomotor apraxia type 1 (AOA1) disorders,
respectively [15]. These diseases are characterised by a defective
response to DNA damage, suggesting that R-loops may be
implicated in triggering genome instability. Indeed, R-loops were
found to be enriched in proliferating cells (testes), but not in the
brain tissues from Setx,Atm,Tdp1 or Aptx knock-out mice [15].
The enrichment of R-loops in testes correlated with high levels of
DNA damage and apoptosis. The lack of R-loops in brain tissue
questions the association between R-loops and neurodegeneration.
This result is surprising, because inducible R-loops have been
previously detected in neuronal cells at the Snord116 locus, which
is associated with the neurodevelopmental disorder Angelman
syndrome, as discussed below [50]. Furthermore, R-loops were
implicated in inducing DNA damage in nonproliferating cells and
post-mitotic neurons and proposed to contribute to the neurode-
generation seen in AT patients [29]. It is possible that R-loops are
regulated by different mechanisms in proliferating cells and post-
mitotic neurons, thereby leading to different R-loop kinetics and so
preventing their detection in some model systems. In particular, R-
loop accumulation may reflect collisions between transcription and
replication machineries [32,51], events which do not occur in
postmitotic neurons. It should be noted that the mouse models
currently used may not fully recapitulate all aspects of human
neurodegeneration.
Figure 1. History of R-loop research. The diagram depicts major developments in the R-loop field and diseases associated with R-loop
dysregulation.
doi:10.1371/journal.pgen.1004630.g001
PLOS Genetics | www.plosgenetics.org 2 September 2014 | Volume 10 | Issue 9 | e1004630
Figure 2. R-loops and human diseases. The diagram depicts the role of R-loops in human diseases. Loss of wild type protein function is depicted
by red crosses. A. Ataxia and motor neuron diseases. Mutations in human RNA/DNA helicase senataxin are associated with AOA2/ALS4 disorders and
lead to R-loop accumulation and defects in transcriptional termination by Pol II [16], the maintenance of genome integrity [46], meiotic
recombination during spermatogenesis, gene silencing during meiotic sex chromosome inactivation [14], and neuronal differentiation [49]. B.
Aicardi-Goutie
`res syndrome (AGS). AGS is associated with mutations in all three subunits of RNase H2, ssDNA 39–59exonuclease TREX1 (DNASEIII),
dsRNA-editing enzyme ADAR1, and dNTP triphosphatase SAMHD1; these trigger accumulation of unprocessed nucleic acids, including genomic DNA
with incorporated ribonucleotides, R-loops, and retroelement-derived nucleic acids, and result in the immune response characteristic of AGS [65]. C.
Trinucleotide expansion diseases. R-loops form over expanded repeats and result in decreased initiation and elongation of RNA Pol II and formation
of repressive chromatin marks, which silence the host gene containing expanded repeats [75]. D. Genome instability in cancer. Loss of proteins
protecting against abnormal R-loop accumulation, such as FIP1L1, leads to genome instability, one hallmark of cancer [31]. Yellow stars denote
double-stranded DNA breaks. E. AID-mediated mutagenesis and translocations in cancer. Single-stranded DNA in R-loops is a substrate for cytidine
deamination by activation-induced cytidine deaminase, leading to mutagenesis as indicated by orange stars [21,88]. These mutations can cause DSB
formation, leading to chromosomal translocations. The IgH/c-MYC translocation brings the strong IgH enhancers, shown as yellow box, close to
c-MYC, leading to its overexpression in Burkitt’s lymphoma [87]. Transcription of IgH/c-MYC starts from a previously inactive promoter downstream of
the translocation break point. The IgH locus is depicted in blue, c-MYC gene is in grey. The translocation breakpoint is indicated by a dashed black
line. F. Senescence. R-loops formed by the noncoding RNA TERRA accumulate at telomeres in cells deficient of Hpr1 and RNase H. In the absence of
telomerase, these R-loops promote Rad52-dependent telomere elongation and delayed senescence. In the absence of telomerase and Rad52, R-loops
promote telomere shortening and premature senescence [94].
doi:10.1371/journal.pgen.1004630.g002
PLOS Genetics | www.plosgenetics.org 3 September 2014 | Volume 10 | Issue 9 | e1004630
RNase H and Aicardi-Goutie
`res Syndrome (AGS)
In addition to their generation during transcription, RNA/
DNA hybrids can arise due to incorporation of ribonucleotides
into DNA by DNA polymerases during replication. RNase H
enzymes are endonucleases that cleave the RNA of RNA/DNA
hybrids in a sequence-independent manner, thus maintaining
genome stability by resolving R-loops that form during transcrip-
tion and by removing misincorporated ribonucleotides from the
DNA [36]. Eukaryotic cells have two types of these enzymes,
RNase H1 and RNase H2, which have different enzymatic and
site-specific activities [52]. In particular, RNase H1 requires a tract
of at least four ribonucleotides to cleave the RNA/DNA hybrid,
whereas RNase H2 can incise 59to a single ribonucleotide
incorporated within a DNA molecule [36,52]. Therefore, only
RNase H2 can process single ribonucleotides in the DNA, but
both enzymes are capable of eliminating RNA/DNA hybrids.
Unlike in bacteria and unicellular eukaryotic organisms, where
RNase H enzymes are dispensable for viability, both RNase H
enzymes are essential in higher eukaryotes. RNase H1 has been
implicated in mitochondrial DNA (mtDNA) replication during
mouse development, a process likely to be associated with
processing of RNA primers during mtDNA replication [53].
RNase H2 is composed of three different subunits, the catalytic
subunit 2A, and two other subunits, 2B and 2C, all of which are
required for enzyme activity. RNase H2 has been implicated in
recognition and removal of ribonucleotides incorporated into
DNA and hydrolysis of Okazaki fragment RNA primers during
DNA replication [36,54–57]. In addition, recent studies point
towards a role of RNase H2 in R-loop resolution during
transcription in vivo [11,58]. In particular, deletion of Saccharo-
myces cerevisiae RNase H2 imposes transcriptional blocks and R-
loop accumulation over rDNA regions in cells depleted of
Topoisomerase I [11] and transcriptional down-regulation of
genes with higher guanine-cytosine (GC) content at the promoter
regions, which are likely to form stable R-loops [58].
In humans, mutations in any of the three subunits of RNase H2
cause Aicardi-Goutie`res syndrome (AGS), a neurological inflam-
matory disorder, which resembles a congenital viral infection and
is associated with accumulation of ribonucleotides in the DNA
(Figure 2B) [59,60]. Interestingly, AGS can also be triggered by
mutations in single-stranded DNA (ssDNA) 39–59exonuclease
TREX1(DNASEIII) [61], double-stranded RNA (dsRNA)-editing
enzyme ADAR1 [62], and dNTP triphosphatase SAMHD1[63].
These proteins are involved in diverse pathways of nucleic acid
metabolism, although their functions are not yet fully understood.
They have been implicated in degrading ssDNA arising from
endogenous retroelements or replication stress (TREX1), regulat-
ing the intracellular dNTPs pool available for replication and
reverse transcription of these retroelements (SAMHD1), or
altering the immune response to RNA species through RNA
editing of retroelements and microRNAs (ADAR1) [64]. Muta-
tions in these proteins are associated with an accumulation of
unprocessed nucleic acids, which triggers the immune response
characteristic of AGS [64,65].
So far, pathologies linked to AGS mutations in RNase H2 have
been mainly attributed to genome instability caused by accumu-
lation of ribonucleotides in DNA [56,66]. However, a specific
contribution of R-loops and RNA/DNA hybrids to AGS
pathology has not been yet investigated. This research has been
hampered by the difficulty to uncouple the two activities of RNase
H2; its ability to remove ribonucleotides from the DNA and to
resolve R-loops, both of which are affected when RNase H2 is
deleted [52,56]. Nevertheless, several lines of evidence suggest that
R-loops may be involved in AGS pathology. Thus, an AGS-
related mutation in the yeast RNase H2 enzyme resulted in its
reduced RNA/DNA cleavage activity [52]. Since RNase H2
constitutes ,90% of the total cellular RNA/DNA hybrid cleavage
activity, its loss due to AGS mutations may lead to significant
accumulation of R-loops [56]. The importance of RNase H2 is
further highlighted by the fact that mutations in RNase H1 do not
cause AGS, suggesting that RNase H2 may have unique properties
to degrade RNA/DNA hybrids [52]. Indeed, R-loops arising
during DNA replication may be exclusively degraded by RNase
H2, as they may be inaccessible to RNase H1 [52,67]. A recently
generated S. cerevisiae RNase H2 mutant, which possesses R-loop
degrading activity but fails to remove single ribonucleotides from
the DNA [52], will be a useful tool in addressing the contribution
of unresolved transcription-associated R-loops to AGS pathology.
TREX1, ADAR1 and SAMHD1 process retroelement-derived
nucleic acids and help to suppress retroelements expansion in the
host genome and their recognition by the immune system [64].
Interestingly, recent genome-wide studies have demonstrated that
RNA/DNA hybrids are particularly enriched at retrotransposon
elements in yeast cells [22], suggesting that expansion of retro-
elements due to mutations in TREX1, ADAR1 or SAMHD1 may
lead to increased RNA/DNA hybrid levels, contributing to
autoimmunitity in AGS. Indeed, it has recently been demonstrated
that RNA/DNA hybrids can be sensed by toll-like receptor 9
(TLR9) to induce pro-inflammatory cytokine and antiviral
interferon production in dendritic cells [68].
R-loops in Nucleotide Expansion Diseases
Expansions of repetitive sequences have been linked to over
forty human diseases [69], and R-loops have been proposed to
play a role in their pathology [70–73]. Remarkably, R-loops are
formed following transcription of trinucleotide repeats in vitro, in
bacteria and human cells [70,71,73]. Interestingly, the nontem-
plate DNA strand in many repetitive sequences can adopt unusual
DNA structures, including G-quadruplexes and DNA triplexes,
which may further stabilise R-loops [74]. Moreover, R-loops
formed at CTG repeats promote repeat instability characteristic of
these diseases [71].
Recently, we demonstrated that R-loops form over expanded
GAA and CGG repeats in cells from Friedreich’s Ataxia (FRDA)
and Fragile X syndrome (FXS) patients, respectively (Figure 2C)
[75]. The abundance of these stable R-loops correlates with
expansion size, and they colocalise with the repressive chromatin
marks characteristic of these diseases (Figure 2C). R-loops can also
trigger the formation of repressive chromatin and cause transcrip-
tional silencing of the FXN gene, providing a molecular link
between R-loops and the pathology of expansion diseases [75]. In
line with R-loops formed on expanded ‘‘premutation’’ and ‘‘full
mutation’’ CGG-repeat-containing alleles of the FMR1 gene
[75,76], promoter-bound FMR1 mRNA containing trinucleotide
repeats was shown to promote epigenetic silencing in FXS [77].
Importantly, the involvement of R-loops in expansion diseases is
not limited to trinucleotide repeats, since R-loops associated with
expanded hexanucleotide GGGGCC repeats in C9orf72 contrib-
ute to the molecular event leading to amyotrophic lateral sclerosis
(ALS) and frontotemporal dementia (FTD) [78].
R-loops could contribute to the pathology of expansion diseases
in various ways. Similar to R-loops at the 39ends of human genes,
expansion-associated R-loops may form a structural block, directly
interfering with Pol II transcriptional elongation [16,24]. Alterna-
tively, R-loops may nucleate repressive chromatin over the
expansion region, by analogy with heterochromatin formation at
PLOS Genetics | www.plosgenetics.org 4 September 2014 | Volume 10 | Issue 9 | e1004630
centromeres in Schizosaccharomyces pombe [25], or promote
chromatin compaction associated with histone H3S10 phosphor-
ylation, as observed in S. cerevisiae,Caenorhabditis elegans, and
human cells [26]. Furthermore, R-loops could cause the charac-
teristic intergenerational and somatic instability of repeat sequenc-
es [72].
R-loops in Cancer
Genome instability is a hallmark of cancer, and it may actively
drive hereditary tumour development [79,80]. Research in the last
decade has clearly demonstrated that dysregulation of R-loops can
corrupt genome integrity, resulting in increased DNA sensitivity to
damaging agents, formation of DSBs, chromosome breaks, fragile
site instability, chromosome loss, and recombination events [5].
Several mechanisms have therefore evolved to maintain R-loop
levels in balance, and alterations in genome caretaker processes can
affect R-loop levels and genome stability [4]. Moreover, mutations
in proteins controlling R-loop levels have been identified in tumours
(Figure 2D). For example, in eosinophilic leukemia, an oncogenic
translocation renders cleavage and polyadenylation factor FIP1L1
inactive, which has been previously shown to cause increased R-
loop levels, DNA damage and chromosome instability (Figure 2D)
[31]. A similar mechanism was suggested for RNA kinase CLP1,
which is associated with a translocation in mixed lineage leukemia
(MLL) [31]. The histone ubiquitin ligase BRE1 also limits R-loop
levels, and its decreased expression may contribute to the high levels
of genomic instability observed in testicular seminoma [81].
The link between R-loops and cancer has been further
substantiated by the finding that the tumour suppressor BRCA2,
which is mutated in breast and ovarian cancer, is required to prevent
R-loop accumulation and genome instability [82]. These observa-
tions raise the interesting possibility that R-loops may provide
proliferative advantages to tumour cells by promoting genome
instability. This will in turn increase the probability of accumulating
mutations favourable to tumour growth and metastasis. Intriguingly,
recent evidence demonstrates that human oncogenic viruses may
also promote genomic instability through accumulation of R-loops
after infection. Kaposi’s sarcoma-associated herpesvirus (KSHV),
which causes multiple AIDS-related cancers, encodes the ORF57
protein, which can sequester the host hTREX complex, important
for mRNA processing and export [83]. Sequestration of hTREX
leads to KSHV-induced accumulation of R-loops and causes
damage to the host DNA, contributing to tumourigenesis [83].
Whilst some proteins suppress R-loop formation, others may
promote R-loops and so increase genome instability leading to
tumour development. This unexpected function has been shown in
yeast for transcription elongation factor Spt2 and DNA repair
protein Rad51 [7,84]. Overexpression of Spt2 leads to transcription-
dependent chromosomal rearrangements, which are prevented by
RNase H overexpression [84]. Spt2 is structurally related to human
HMG1, which is overexpressed in gastric cancers and malignant
melanomas [84]. However, it is not clear if increased HMG1 levels
promote R-loops and DNA damage in cancer cells. In contrast to its
well-established role in DNA strand exchange during homologous
recombination and DNA repair [85], recent studies have shown that
Rad51 can also mediate R-loop formation and genome instability in
trans, extending the prevailing view that R-loops form cotranscrip-
tionally [7]. Similar to HMG1, RAD51 is overexpressed in human
cancers [7]. However, it remains to be elucidated if RAD51
overexpression in cancers is a consequence of activated DNA repair
pathways, or a cause of genome instability [7].
R-loops have been detected in immunoglobulin (Ig) genes,
where they initiate class switch recombination by exposing single-
stranded DNA, thus providing the substrate for activation-induced
cytidine deaminase (AID), which promotes DSBs and subsequent
translocation between Ig heavy chains [21,86]. Although this
process is essential for generation of antibody isotype diversity,
AID-mediated mutagenesis has also been implicated in patholog-
ical translocations between the Ig loci and other active genes,
leading to production of fusion proteins or oncogenic gene
expression, observed in B cell malignancies (Figure 2E) [87].
Interestingly, R-loops are also found in common translocation
partners of Ig genes, including the oncogene c-MYC [18,27].
Therefore, the simultaneous formation of R-loops in Ig and
transcribed non-Ig genes may induce AID-mediated DSB
formation, leading to pathological translocations (Figure 2E)
[27,88,89]. Interestingly, overexpression of the APOBEC family
of AID-related enzymes in breast cancer have been linked to
genomic mutations, pointing to a potentially broader role of R-
loops and AID/APOBEC-mediated genome instability in cancer
[90].
Changes in gene expression are another central aspect of cancer
[79]. In healthy cells, the expression of tumour suppressor genes
prevents abnormal proliferation and other aspects of tumourigen-
esis [79]. Tumour suppressors are frequently silenced in cancer by
excessive promoter DNA methylation [91]. It has been proposed
that R-loop formation at promoters protects against DNA
methylation by de novo DNA methyltransferase DNMT3B,
thereby keeping genes active [18]. Since R-loops have been
computationally predicted to form at promoters of tumour
suppressor genes BRCA1,RASSF1A, and CDKN2A [92], it is
important to investigate if R-loop levels at these genes are reduced
in cancer and how this relates to the observed DNA hypermethy-
lation.
In contrast to this, efficient transcription of the oncogene c-MYC
requires that R-loop levels are kept low by the activity of DNA
topoisomerase IIIB, which is recruited to arginine-methylated
histones by the tudor domain containing 3 (TDRD3) protein [27].
This R-loop-mediated mechanism of c-MYC gene regulation may
be relevant to tumour progression in breast cancer, which
frequently shows overexpression of both c-MYC and TDRD3
[27,93]. Therefore, it is tempting to speculate that increased
TDRD3 levels suppress R-loops in c-MYC, thereby allowing its
enhanced expression, which correlates with poor cancer prognosis
[93]. However, it still remains to be determined if R-loops play a
specific role in transcription dysregulation in cancer and if this
process differs from R-loop-mediated transcriptional programmes
associated with housekeeping genes.
More recently, R-loops have been implicated in cell senescence, a
mechanism protecting against tumour cell proliferation [79]. In
particular, the telomeric noncoding (nc) RNA TERRA forms R-
loops which are induced when R-loop suppressors such as RNase H
or Thp2 are lost [94,95]. In the absence of telomerase, telomeric R-
loops promote recombination-mediated telomere elongation via
Rad52, and this delays the onset of cellular senescence [94]. In
contrast, in Rad52-deficient cells, R-loop accumulation leads to
telomere shortening and premature senescence [94]. Interestingly,
cells from AOA2 patients with senataxin mutations contain shorter
telomeres, suggesting a possible involvement of SETX in telomere
stability [96]. Telomeric R-loops therefore play a complex and
dynamic role in telomere length maintenance and cellular
proliferative potential (Figure 2F).
In conclusion, multiple lines of evidence point to an involve-
ment of R-loops in cancer biology. Yet it still remains to be
investigated if R-loop levels are indeed regulated differentially in
normal and tumour tissues and if they can directly influence
tumourigenesis.
PLOS Genetics | www.plosgenetics.org 5 September 2014 | Volume 10 | Issue 9 | e1004630
R-loop Therapies
R-loops represent a potential therapeutic target. Despite their
importance in gene regulation, they have yet to be fully exploited
in drug design [97]. Various ligands can target RNA/DNA
hybrids, including ethidium bromide, the aminoglycosides neo-
mycin and paramomycin, and the polyamides distamycin and
netropsin [98]. These compounds recognise RNA/DNA hybrids
through intercalation and binding to the nucleic acid groove.
Although exhibiting high binding affinities to RNA/DNA hybrids,
many of these molecules also bind dsDNA and RNA and are
mutagenic, limiting their potential biological applications [98].
However, recent studies suggest that combining the properties of
these ligands can achieve subnanomolar affinity for RNA/DNA
hybrids. In particular, this has been demonstrated for ligands
linking aminoglycosides to derivatives of ethidium bromide [99],
providing a possible approach for the development of potent and
specific RNA/DNA hybrid ligands in future drug design efforts.
Various compounds that modulate DNA supercoiling and inhibit
DNA topoisomerases, including topotecan and camptothecin, can
also affect R-loop formation in vivo [29,50]. In particular,
topoisomerase inhibitors have recently been used to reactivate the
silenced paternal Ube3a gene, which encodes a ubiquitin E3 ligase,
to compensate for the deleted maternal Ube3a in Angelman
syndrome (AS). AS and Prader-Willi syndrome (PWS) are imprinted
neurodevelopmental disorders that are often caused by large
deletions of human chromosome 15q11–q13 over the Snord116
gene locus, but the deletion differs in its parent-of-origin [100]. In
neurons, only the maternal Ube3a allele is expressed, because the
paternal Ube3a allele is silenced by expression of the ncRNA
Ube3a-ATS (Figure 3A) [101]. AS therapies therefore seek to
reactivate the silenced, but genetically intact, paternal Ube3a allele.
Interestingly, R-loops were recently shown to regulate the neuronal
expression of the paternal Ube3a-ATS transcript, which is essential
for transcriptional silencing of the paternal Ube3a gene [50]. In
particular, treatment with the topoisomerase inhibitor topotecan
increased R-loop levels over the Snord116 locus, resulting in
chromatin decondensation, inhibition of Pol II transcription of
Ube3a-ATS, and concomitant increase in Ube3a expression from
the paternal allele (Figure 3B). This R-loop-mediated reactivation
of paternal Ube3a could therefore compensate for the loss of
maternal Ube3a in AS and so potentially holds promise for targeted
therapies for both AS and PWS (Figure 3B).
It has previously been proposed that R-loops in trinucleotide
expansion diseases could be targeted to suppress repeat expansions
or reactivate silenced genes [72]. A recent study provided direct
evidence that a small molecule is able to suppress R-loop formation
at expanded CGG repeats in the FMR1 gene, thereby preventing
FMR1 epigenetic silencing in FXS [77]. As an alternative
approach, R-loop levels may be indirectly modulated by treatments
that target proteins involved in R-loop biology (Table S1). For
instance, genomic instability caused by a widespread increase of R-
loops due to loss of an R-loop suppressing protein could potentially
be reverted by introduction of an alternative R-loop suppressor.
Recent identification of small-molecule inhibitors for RNase H2
may also provide a powerful new tool for the study of R-loop biology
in health and disease [102]. Furthermore, the S9.6 antibody offers
new opportunities for research and development. In particular, it
has already been used in the development of biosensor systems
[103], detection of miRNA targets[104], and as a key component of
human papillomavirus (HPV) diagnostic kit (Qiagen).
The explosion of studies uncovering the role of R-loops in
health and disease in recent years provides the exciting prospect of
developing new targeted therapeutics for many human disorders.
However, due to the ubiquitous nature of R-loops it will be
important to ensure that efficient treatments are specific.
Conclusions and Future Challenges
R-loops have been implicated in many biological processes in
different organisms. R-loops can play positive and negative roles in
gene expression; they can mediate Ig class switch recombination
and transcriptional termination, affect genome stability, transcrip-
tion, cell cycle progression, and cell viability. Despite the diversity
of these biological processes, the molecular mechanisms associated
with R-loop formation in mammalian cells remain largely
unknown. It is unclear how R-loops can regulate gene expression,
how they are maintained and eliminated in the cells, and which
proteins are involved in the regulation of these processes.
The connections between R-loops and human diseases suggest
that cells have evolved mechanisms to distinguish between
deleterious and beneficial R-loops. However, the evidence discussed
above raises an important question: how can R-loop dysregulation
be mechanistically linked to a variety of human diseases with such
diverse pathologies? One explanation may be that R-loops form in
many genomic locations in healthy cells [16,18,22,27]. Therefore,
unsurprisingly, their dysregulation can affect a large number of
disease-associated genes. This is in contrast to gene-specific R-loop
pathologies, associated with mutations, which result in altered R-
loop levels locally, as observed in the repeat expansion diseases
FRDA and FXS [75,77]. Furthermore, R-loops can have different
intrinsic properties. R-loops at expanded GAA repeats in the FXN
gene are highly stable and trigger transcriptional repression, while R-
loops in the highly-expressed c-Actin gene are easily turned over
[75]. This could, in part, be due to differential activity of R-loop
processing proteins on different classes of genes, as proposed in yeast
[22]. Adding another layer of complexity, the formation of R-loops
can be influenced by cell type [77], cell cycle stage [15], gene length,
and/or GC content and transcriptional level [22,105]. Epigenetic
marks including DNA methylation and post-translational histone
modifications can contribute to further modulation of R-loop levels
[18,27]. Thus, R-loops represent cellular structures that share the
same elementary composition, but may possess different dynamic
properties, which can be affected by any of the aforementioned
processes, thus explaining the wide range of diseases associated with
R-loops.
Despite the lack of mechanistic insights into R-loop-associated
diseases, some common themes, underlying their pathology, are
already becoming obvious. First, there is a strong connection
between R-loop dysregulation and induction of DNA damage and
loss of genome integrity, which contributes to cancer development
[31,81,88], repeat expansion diseases [71], and neurodegeneration
[29,45]. Secondly, R-loops can mediate changes in transcription
locally or globally, contributing to pathologies associated with
repeat expansion diseases [75,77,78], Angelman syndrome [50],
and cancer [27]. However, it is a strong possibility that both of
these pathological themes may overlap in many disorders, as
observed in repeat expansion diseases [71,75,77], and novel
disease themes may be revealed in the future.
One of the major challenges in R-loop field is to investigate the
causes and consequences of R-loop formation in additional models
of human disease. Uncovering further aspects of R-loop biology in
human cells will certainly shed light on many basic biological
questions and have major implications for our understanding of
human disease. Future studies will undoubtedly reveal more
diseases associated with R-loop dysregulation and will provide the
basis for novel therapeutic approaches targeting these so far
overlooked structures in gene expression.
PLOS Genetics | www.plosgenetics.org 6 September 2014 | Volume 10 | Issue 9 | e1004630
Supporting Information
Table S1 Proteins implicated in R-loop biology. For
multiprotein complexes, only subunits directly implicated in R-
loop biology are mentioned in the table. *Asterisk indicates that
protein association with R-loops is based on in vitro evidence.
(DOCX)
Text S1 Supplemental references.
(DOCX)
Acknowledgments
We thank Prof. P. Cook, Prof. N. J. Proudfoot, Prof. C. Norbury and L. M.
Silva for critically reading the manuscript.
References
1. Thomas M, White RL, Davis RW (1976) Hybridization of RNA to double-
stranded DNA: formation of R-loops. Proc Natl Acad Sci U S A 73: 2294–
2298.
2. White RL, Hogness DS (1977) R loop mapping of the 18S and 28S sequences
in the long and short repeating units of Drosophila melanogaster rDNA. Cell
10: 177–192.
3. Drolet M, Bi X, Liu LF (1994) Hypernegative supercoiling of the DNA
template during transcription elongation in vitro. J Biol Chem 269: 2068–2074.
4. Aguilera A, Garcia-Muse T (2012) R loops: from transcription byproducts to
threats to genome stability. Mol Cell 46: 115–124.
5. Hamperl S, Cimprich KA (2014) The contribution of co-transcriptional
RNA:DNA hybrid structures to DNA damage and genome instability. DNA
Repair (Amst) 19: 84–94.
6. Skourti-Stathaki K, Proudfoot NJ (2014) A double-edged sword: R loops as
threats to genome integrity and powerful regulators of gene expression. Genes
Dev 28: 1384–1396.
7. Wahba L, Gore SK, Koshland D (2013) The homologous recombination
machinery modulates the formation of RNA-DNA hybrids and associated
chromosome instability. Elife 2: e00505.
8. Boguslawski SJ, Smith DE, Michalak MA, Mickelson KE, Yehle CO, et al.
(1986) Characterizat ion of monoclonal ant ibody to DNA.RNA and its
application to immunodetection of hybrids. J Immunol Methods 89: 123–
130.
9. Phillips DD, Garboczi DN, Singh K, Hu Z, Leppla SH, et al. (2013) The sub-
nanomolar binding of DNA-RNA hybrids by the single-chain Fv fragment of
antibody S9.6. J Mol Recognit 26: 376–381.
Figure 3. Potential R-loop-based therapeutic approach in Angelman Syndrome (AS). A. Neuronal expression of the paternal ncRNA
Ube3a-ATS represses paternal Ube3a gene in cis [101]. DNA methylation of the Snord116 locus on the maternal allele prevents Ube3a-ATS
transcription, resulting in Ube3a expression from the maternal allele. Transcriptional repression is indicated by red crosses. B. R-loop-mediated re-
activation of silent paternal Ube3a gene provides a targeted therapy for AS. Deletion leading to the loss of maternal Ube3a expression detected in AS
is indicated by the red dashed line. Topotecan treatment increases R-loop levels over the Snord116 locus, resulting in chromatin decondensation,
inhibition of Pol II transcription through Ube3a-ATS, and increased expression of Ube3a from the paternal allele [50].
doi:10.1371/journal.pgen.1004630.g003
PLOS Genetics | www.plosgenetics.org 7 September 2014 | Volume 10 | Issue 9 | e1004630
10. Hu Z, Zhang A, Storz G, Gottesman S, Leppla SH (2006) An antibody-based
microarray assay for small RNA detection. Nucleic Acids Res 34: e52.
11. El Hage A, French SL, Beyer AL, Tollervey D (2010) Loss of Topoisomerase I
leads to R-loop-mediated transcriptional blocks during ribosomal RNA
synthesis. Genes Dev 24: 1546–1558.
12. Mischo HE, Gomez-Gonzalez B, Grzechnik P, Rondon AG, Wei W, et al.
(2011) Yeast Sen1 helicase protects the genome from transcription-associated
instability. Mol Cell 41: 21–32.
13. Sun Q , Csorba T, Skourti-Stathaki K, Proudfoot NJ, Dean C (2013) R-loop
stabilization represses antisense transcription at the Arabidopsis FLC locus.
Science 340: 619–621.
14. Becherel OJ, Yeo AJ, Stellati A, Heng EY, Luff J, et al. (2013) Senataxin plays
an essential role with DNA damage response proteins in meiotic recombination
and gene silencing. PLoS Genet 9: e1003435.
15. Yeo AJ, Becherel OJ, Luff JE, Cullen JK, Wongsurawat T, et al. (2014) R-
Loops in Proliferating Cells but Not in the Brain: Implications for AOA2 and
Other Autosomal Recessive Ataxias. PLoS ONE 9: e90219.
16. Skourti-Stathaki K, Proudfoot NJ, Gromak N (2011) Human senataxin resolves
RNA/DNA hybrids formed at transcriptional pause sites to promote Xrn2-
dependent termination. Mol Cell 42: 794–805.
17. Ginno PA, Lim YW, Lott PL, Korf I, Chedin F (2013) GC skew at the 59and
39ends of human genes links R-loop formation to epigenetic regulation and
transcription termination. Genome Res 23: 1590–1600.
18. Ginno PA, Lott PL, Christensen HC, Korf I, Chedin F (2012) R-loop
formation is a distinctive characteristic of unmethylated human CpG island
promoters. Mol Cell 45: 814–825.
19. Xu B, Clayton DA (1996) RNA-DNA hybrid formation at the human
mitochondrial heavy-strand origin ceases at replication start sites: an
implication for RNA-DNA hybrids serving as primers. EMBO J 15: 3135–
3143.
20. Itoh T, Tomizawa J (1980) Formation of an RNA primer for initiation of
replication of ColE1 DNA by ribonuclease H. Proc Natl Acad Sci U S A 77:
2450–2454.
21. Yu K, Chedin F, Hsieh CL, Wilson TE, Lieber MR (2003) R-loops at
immunoglobulin class switch regions in the chromosomes of stimulated B cells.
Nat Immunol 4: 442–451.
22. Chan YA, Aristizabal MJ, Lu PY, Luo Z, Hamza A, et al. (2014) Genome-
Wide Profiling of Yeast DNA:RNA Hybrid Prone Sites with DRIP-Chip. PLoS
Genet 10: e1004288.
23. Tous C, Aguilera A (2007) Impairment of transcription elongation by R-loops
in vitro. Biochem Biophys Res Commun 360: 428–432.
24. Huertas P, Aguilera A (2003) Cotranscriptionally formed DNA:RNA hybrids
mediate transcription elongation impairment and transcription-associated
recombination. Mol Cell 12: 711–721.
25. Nakama M, Kawakami K, Kajitani T, Urano T, Murakami Y (2012) DNA-
RNA hybrid formation mediates RNAi-directed heterochromatin formation.
Genes Cells 17: 218–233.
26. Castellano-Pozo M, Santos-Pereira JM, Rondon AG, Barroso S, Andujar E,
et al. (2013) R loops are linked to histone H3 S10 phosphorylation and
chromatin condensation. Mol Cell 52: 583–590.
27. Yang Y, McBride KM, Hensley S, Lu Y, Chedin F, et al. (2014) Arginine
methylation facilitates the recruitment of TOP3B to chromatin to prevent R
loop accumulation. Mol Cell 53: 484–497.
28. Santos-Pereira JM, Herrero AB, Garcia-Rubio ML, Marin A, Moreno S, et al.
(2013) The Npl3 hnRNP prevents R-loop-mediated transcription-replication
conflicts and genome instability. Genes Dev 27: 2445–2458.
29. Sordet O, Redon CE, Guirouilh-Barbat J, Smith S, Solier S, et al. (2009)
Ataxia telangiectasia mutated activation by transcription- and topoisomerase I-
induced DNA double-strand breaks. EMBO Rep 10: 887–893.
30. Li X, Manley JL (2005) Inactivation of the SR protein splicing factor ASF/SF2
results in genomic instability. Cell 122: 365–378.
31. Stirling PC, Chan YA, Minaker SW, Aristizabal MJ, Barrett I, et al. (2012) R-
loop-mediated genome instability in mRNA cleavage and polyadenylation
mutants. Genes Dev 26: 163–175.
32. Helmrich A, Ballarino M, Tora L (2011) Collisions between replication and
transcription complexes cause common fragile site instability at the longest
human genes. Mol Cell 44: 966–977.
33. Tuduri S, Crabbe L, Conti C, Tourriere H, Holtgreve-Grez H, et al. (2009)
Topoisomerase I suppresses genomic instability by preventing interference
between replication and transcription. Nat Cell Biol 11: 1315–1324.
34. Wahba L, Amon JD, Koshland D, Vuica-Ross M (2011) RNase H and multiple
RNA biogenesis factors cooperate to prevent RNA:DNA hy brids from
generating genome instability. Mol Cell 44: 978–988.
35. Herrera-Moyano E, Mergui X, Garcia-Rubio ML, Barroso S, Aguilera A
(2014) The yeast and human FACT chromatin-reorganizing complexes solve
R-loop-mediated transcription-replication conflicts. Genes Dev 28: 735–748.
36. Cerritelli SM, Crouch RJ (2009) Ribonuclease H: the enzymes in eukaryotes.
FEBS J 276: 1494–1505.
37. Boule JB, Zakian VA (2007) The yeast Pif1p DNA helicase preferentially
unwinds RNA DNA substrates. Nucleic Acids Res 35: 5809–5818.
38. Shin JH, Kelman Z (2006) The replicative helicases of bacteria, archaea, and
eukarya can unwind RNA-DNA hybrid substrates. J Biol Chem 281: 26914–
26921.
39. Moreira MC, Klur S, Watanabe M, Nemeth AH, Le Ber I, et al. (2004)
Senataxin, the ortholog of a yeast RNA helicase, is mutant in ataxia-ocular
apraxia 2. Nat Genet 36: 225–227.
40. Anheim M, Monga B, Fleury M, Charles P, Barbot C, et al. (2009) Ataxia with
oculomotor apraxia type 2: clinical, biological and genotype/phenotype
correlation study of a cohort of 90 patients. Brain 132: 2688–2698.
41. Chen YZ, Bennett CL, Huynh HM, Blair IP, Puls I, et al. (2004) DNA/RNA
helicase gene mutations in a form of juvenile amyotrophic lateral sclerosis
(ALS4). Am J Hum Genet 74: 1128–1135.
42. Suraweera A, Lim Y, Woods R, Birrell GW, Nasim T, et al. (2009) Functional
role for senataxin, defective in ataxia oculomotor apraxia type 2, in
transcriptional regulation. Hum Mol Genet 18: 3384–3396.
43. Steinmetz EJ, Warren CL, Kuehner JN, Panbehi B, Ansari AZ, et al. (2006)
Genome-wide distribution of yeast RNA polymerase II and its control by Sen1
helicase. Mol Cell 24: 735–746.
44. West S, Gromak N, Proudfoot NJ (2004) Human 59R39exonuclease Xrn2
promotes transcription termination at co-transcriptional cleavage sites. Nature
432: 522–525.
45. Suraweera A, Becherel OJ, Chen P, Rundle N, Woods R, et al. (2007)
Senataxin, defective in ataxia oculomotor apraxia type 2, is involved in the
defense against oxidative DNA damage. J Cell Biol 177: 969–979.
46. Yuce O, West SC (2013) Senataxin, defective in the neurodege nerative
disorder ataxia with oculomotor apraxia 2, lies at the interface of transcription
and the DNA damage response. Mol Cell Biol 33: 406–417.
47. Alzu A, Bermejo R, Begnis M, Lucca C, Piccini D, et al. (2012) Senataxin
associates with replication forks to protect fork int egrity across RN A-
polymerase-II-transcribed genes. Cell 151: 835–846.
48. Richard P, Feng S, Manley JL (2013) A SUMO-dependent interaction between
Senataxin and the exosome, disrupted in the neurodegenerative disease AOA2,
targets the exosome to sites of transcription-induced DNA damage. Genes Dev
27: 2227–2232.
49. Vantaggiato C, Bondioni S, Airoldi G, Bozzato A, Borsani G, et al. (2011)
Senataxin modulates neurite growth through fibroblast growth factor 8
signalling. Brain 134: 1808–1828.
50. Powell WT, Coulson RL, Gonzales ML, Crary FK, Wong SS, et al. (2013) R-
loop formation at Snord116 mediates topotecan inhibition of Ube3a-antisense
and allele-specific chromatin decondensation. Proc Natl Acad Sci U S A 110:
13938–13943.
51. Helmrich A, Ballarino M, Nudler E, Tora L (2013) Transcription-replication
encounters, consequences and genomic instability. Nat Struct Mol Biol 20:
412–418.
52. Chon H, Sparks JL, Rychlik M, Nowotny M, Burgers PM, et al. (2013) RNase
H2 roles in genome integrity revealed by unlinking its activities. Nucleic Acids
Res 41: 3130–3143.
53. Cerritelli SM, Frolova EG, Feng C, Grinberg A, Love PE, et al. (2003) Failure
to produce mitochondrial DNA results in embryonic lethality in Rnaseh1 null
mice. Mol Cell 11: 807–815.
54. Rydberg B, Game J (2002) Excision of misincorporated ribonucleotides in
DNA by RNase H (type 2) and FEN-1 in cell-free extracts. Proc Natl Acad
Sci U S A 99: 16654–16659.
55. Nick McElhinny SA, Kumar D, Clark AB, Watt DL, Watts BE, et al. (2010)
Genome instability due to ribonucleotide incorporation into DNA. Nat Chem
Biol 6: 774–781.
56. Reijns MA, Rabe B, Rigby RE, Mill P, Astell KR, et al. (2012) Enzymatic
removal of ribonucleotides from DNA is essential for mammalian genome
integrity and development. Cell 149: 1008–1022.
57. Lazzaro F, Novarina D, Amara F, Watt DL, Stone JE, et al. (2012) RNase H
and postreplication repair protect cells from ribonucleotides incorporated in
DNA. Mol Cell 45: 99–110.
58. Arana ME, Kerns RT, Wharey L, Gerrish KE, Bushel PR, et al. (2012)
Transcriptional responses to loss of RNase H2 in Saccharomyces cerevisiae.
DNA Repair (Amst) 11: 933–941.
59. Crow YJ, Leitch A, Hayward BE, Garner A, Parmar R, et al. (2006) Mutations
in genes encoding ribonuclease H2 subunits cause Aicardi-Goutieres syndrome
and mimic congenital viral brain infection. Nat Genet 38: 910–916.
60. Crow YJ, Rehwi nkel J (2009) Aicardi-Goutieres syndrome and related
phenotypes: linking nucleic acid metabolism with autoimmunity. Hum Mol
Genet 18: R130–136.
61. Crow YJ, Hayward BE, Parmar R, Robins P, Leitch A, et al. (2006) Mutations
in the gene encoding the 39-59DNA exonuclease TREX1 cause Aicardi-
Goutieres syndrome at the AGS1 locus. Nat Genet 38: 917–920.
62. Rice GI, Kasher PR, Forte GM, Mannion NM, Greenwood SM, et al. (2012)
Mutations in ADAR1 cause Aicardi-Goutieres syndrome associated with a type
I interferon signature. Nat Genet 44: 1243–1248.
63. Rice GI, Bond J, Asipu A, Brunette RL, Manfield IW, et al. (2009) Mutations
involved in Aicardi-Goutieres syndrome implicate SAMHD1 as regulator of
the innate immune response. Nat Genet 41: 829–832.
64. Lee-Kirsch MA, Wolf C, Gunther C (2014) Aicardi-Goutieres syndrome: a
model disease for systemic autoimmunity. Clin Exp Immunol 175: 17–24.
65. Rabe B (2013) Aicardi-Goutieres syndrome: clues from the RNase H2 knock-
out mouse. J Mol Med (Berl) 91: 1235–1240.
66. Hiller B, Achleitner M, Glage S, Naumann R, Behrendt R, et al. (2012)
Mammalian RNase H2 removes ribonucleotides from DNA to maintain
genome integrity. J Exp Med 209: 1419–1426.
PLOS Genetics | www.plosgenetics.org 8 September 2014 | Volume 10 | Issue 9 | e1004630
67. Bubeck D, Reijns MA, Graham SC, Astell KR, Jones EY, et al. (2011) PCNA
directs type 2 RNase H activity on DNA replication and repair substrates.
Nucleic Acids Res 39: 3652–3666.
68. Rigby RE, Webb LM, Mackenzie KJ, Li Y, Leitch A, et al. (2014) RNA:DNA
hybrids are a novel molecular pattern sensed by TLR9. EMBO J 33: 542–558.
69. Lopez Castel A, Cleary JD, Pearson CE (2010) Repeat instability as the basis
for human diseases and as a potential target for therapy. Nat Rev Mol Cell Biol
11: 165–170.
70. Grabczyk E, Mancuso M, Sammarco MC (2007) A persistent RNA.DNA
hybrid formed by transcription of the Friedreich ataxia triplet repeat in live
bacteria, and by T7 RNAP in vitro. Nucleic Acids Res 35: 5351–5359.
71. Lin Y, Dent SY, Wilson JH, Wells RD, Napierala M (2010) R loops stimulate
genetic instability of CTG.CAG repeats. Proc Natl Acad Sci U S A 107: 692–
697.
72. McIvor EI, Polak U, Napierala M (2010) New insights into repeat instability:
role of RNA*DNA hybrids. RNA Biol 7: 551–558.
73. Reddy K, Tam M, Bowater RP, Barber M, Tomlinson M, et al. (2011)
Determinants of R-loop formation at convergent bidirectionally transcribed
trinucleotide repeats. Nucleic Acids Res 39: 1749–1762.
74. Belotserkovskii BP, Mirkin SM, Hanawalt PC (2013) DNA sequences that
interfere with transcription: implications for genome function and stability.
Chem Rev 113: 8620–8637.
75. Groh M, Lufino MM, Wade-Martins R, Gromak N (2014) R-loops associated
with triplet repeat expansions promote gene silencing in Friedreich ataxia and
fragile X syndrome. PLoS Genet 10: e1004318.
76. Loomis EW, Sanz LA, Chedin F, Hagerman PJ (2014) Transcription-
Associated R-Loop Formation across the Human FMR1 CGG-Repeat Region.
PLoS Genet 10: e1004294.
77. Colak D, Zaninovic N, Cohen MS, Rosenwaks Z, Yang WY, et al. (2014)
Promoter-bound trinucleotide repeat mRNA drives epigenetic silencing in
fragile X syndrome. Science 343: 1002–1005.
78. Haeusler AR, Donnelly CJ, Periz G, Simko EA, Shaw PG, et al. (2014)
C9orf72 nucleotide repeat structures initiate molecular cascades of disease.
Nature 507: 195–200.
79. Hanahan D, Weinberg RA (2011) Hallmarks of cancer: the next generation.
Cell 144: 646–674.
80. Negrini S, Gorgoulis VG, Halazonetis TD (2010) Genomic instability–an
evolving hallmark of cancer. Nat Rev Mol Cell Biol 11: 220–228.
81. Chernikova SB, Razorenova OV, Higgins JP, Sishc BJ, Nicolau M, et al. (2012)
Deficiency in mammalian histone H2B ubiquitin ligase Bre1 (Rnf20/Rnf40)
leads to replication stress and chromosomal instability. Cancer Res 72: 2111–
2119.
82. Bhatia V, Barroso SI, Garcia-Rubio ML, Tumini E, Herrera-Moyano E, et al.
(2014) BRCA2 prevents R-loop accumulation and associates with TREX-2
mRNA export factor PCID2. Nature 511: 362–365.
83. Jackson BR, Noerenberg M, Whitehouse A (2014) A novel mechanism
inducing genome instability in Kaposi’s sarcoma-associated herpesvirus
infected cells. PLoS Pathog 10: e1004098.
84. Sikdar N, Banerjee S, Zhang H, Smith S, Myung K (2008) Spt2p defines a new
transcription-dependent gross chromosomal rearrangement pathway. PLoS
Genet 4: e1000290.
85. San Filippo J, Sung P, Klein H (2008) Mechanism of eukaryotic homologous
recombination. Annu Rev Biochem 77: 229–257.
86. Chaudhuri J, Tian M, Khuong C, Chua K, Pinaud E, et al. (2003)
Transcription-targeted DNA deamination by the AID antibody diversification
enzyme. Nature 422: 726–730.
87. Robbiani DF, Nussenzweig MC (2013) Chromosome translocation, B cell
lymphoma, and activation-induced cytidine deaminase. Annu Rev Pathol 8:
79–103.
88. Ruiz JF, Gomez-Gonzalez B, Aguilera A (2011) AID induces double-strand
breaks at immunoglobulin switch regions and c-MYC causing chromosomal
translocations in yeast THO mutants. PLoS Genet 7: e1002009.
89. Duquette ML, Pham P, Goodman MF, Maizels N (2005) AID binds to
transcription-induced structures in c-MYC that map to regions associated with
translocation and hypermutation. Oncogene 24: 5791–5798.
90. Burns MB, Lackey L, Carpenter MA, Rathore A, Land AM, et al. (2013)
APOBEC3B is an enzymatic source of mutation in breast cancer. Nature 494:
366–370.
91. Kulis M, Esteller M (2010) DNA methylation and cancer. Adv Genet 70: 27–56.
92. Wongsurawat T, Jenjaroenpun P, Kwoh CK, Kuznetsov V (2011) Quantitative
model of R-loop forming structures reveals a novel level of RNA-DNA
interactome complexity. Nucleic Acids Res 40: e16.
93. Hynes NE, Stoelzle T (2009) Key signalling nodes in mammary gland
development and cancer: Myc. Breast Cancer Res 11: 210.
94. Balk B, Maicher A, Dees M, Klermund J, Luke-Glaser S, et al. (2013)
Telomeric RNA-DNA hybrids affect telomere-length dynamics and senes-
cence. Nat Struct Mol Biol 20: 1199–1205.
95. Pfeiffer V, Crittin J, Grolimund L, Lingner J (2013) The THO complex
component Thp2 counteracts telomeric R-loops and telomere shortening.
EMBO J 32: 2861–2871.
96. De Amicis A, Piane M, Ferrari F, Fanciulli M, Delia D, et al. (2011) Role of
senataxin in DNA damage and telomeric stability. DNA Repair (Amst) 10:
199–209.
97. Wheelhouse RT, Chaires JB (2010) Drug binding to DNA6RNA hybrid
structures. Methods Mol Biol 613: 55–70.
98. Shaw NN, Arya DP (2008) Recognition of the unique structure of DNA:RNA
hybrids. Biochimie 90: 1026–1039.
99. Shaw NN, Xi H, Arya DP (2008) Molecular recognition of a DNA:RNA
hybrid: sub-nanomolar binding by a neomycin-methidium conjugate. Bioorg
Med Chem Lett 18: 4142–4145.
100. Cassidy SB, Dykens E, Williams CA (2000) Prader-Willi and Angelman
syndromes: sister imprinted disorders. Am J Med Genet 97: 136–146.
101. Meng L, Person RE, Beaudet AL (2012) Ube3a-ATS is an atypical RNA
polymerase II transcript that represses the paternal expression of Ube3a. Hum
Mol Genet 21: 3001–3012.
102. White R, Saxty B, Large J, Kettleborough CA, Jackson AP (2013) Identification
of small-molecule inhibitors of the ribonuclease H2 enzyme. J Biomol Screen
18: 610–620.
103. Sipova H, Zhang S, Dudley AM, Galas D, Wang K, et al. (2010) Surface
plasmon resonance biosensor for rapid label-free detection of microribonucleic
acid at subfemtomole level. Anal Chem 82: 10110–10115.
104. Qavi AJ, Kindt JT, Gleeson MA, Bailey RC (2011) Anti-DNA:RNA antibodies
and silicon photonic microring resonators: increased sensitivity for multiplexed
microRNA detection. Anal Chem 83: 5949–5956.
105. Gomez-Gonzalez B, Garcia-Rubio M, Bermejo R, Gaillard H, Shirahige K,
et al. (2011) Genome-wide function of THO/TREX in active genes prevents
R-loop-dependent replication obstacles. EMBO J 30: 3106–3119.
PLOS Genetics | www.plosgenetics.org 9 September 2014 | Volume 10 | Issue 9 | e1004630
... As aforementioned, the R-loop plays an important role in many physiological processes, its aberrant presence in the genome is associated with several diseases. It is reported that some trinucleotide repeat-associated diseases, neurological diseases, and cancers are related to the R-loop [136] (Table 2). Many proteins with proper function are essential for preventing DNA damage caused by the R-loop and genomic instability. ...
... The non-programmed accumulation of the R-loop is associated with genomic instability. Therefore, there is a potential association between the R-loop and the development of cancers [22,136]. BRCA1 and BRCA2 are two subtypes of breast cancer susceptibility genes. ...
Article
Full-text available
The cell cycle is a crucial biological process that is involved in cell growth, development, and reproduction. It can be divided into G1, S, G2, and M phases, and each period is closely regulated to ensure the production of two similar daughter cells with the same genetic material. However, many obstacles influence the cell cycle, including the R-loop that is formed throughout this process. R-loop is a triple-stranded structure, composed of an RNA: DNA hybrid and a single DNA strand, which is ubiquitous in organisms from bacteria to mammals. The existence of the R-loop has important significance for the regulation of various physiological processes. However, aberrant accumulation of R-loop due to its limited resolving ability will be detrimental for cells. For example, DNA damage and genomic instability, caused by the R-loop, can activate checkpoints in the cell cycle, which in turn induce cell cycle arrest and cell death. At present, a growing number of factors have been proven to prevent or eliminate the accumulation of R-loop thereby avoiding DNA damage and mutations. Therefore, we need to gain detailed insight into the R-loop resolution factors at different stages of the cell cycle. In this review, we review the current knowledge of factors that play a role in resolving the R-loop at different stages of the cell cycle, as well as how mutations of these factors lead to the onset and progression of diseases.
... Although increasing evidence indicates that aberrant R-loops are closely linked to the progression of different cancer types, including LUAD [11,34,35], the causality remains unknown. This study demonstrated that R-loop scores from malignant cells could predict survival outcomes and treatment responses, and it also provided insights into the mechanisms of low-scoring R-loops mediating immune escape and drug resistance. ...
... R-loops are associated with the progression of many cancer types [34,35], including LUAD. Knowledge on the causal relationship between R-loops and disease etiology provides theoretical support for the development of R-loop-based targeted cancer therapeutic strategies [44][45][46]. ...
Article
Full-text available
Dysregulation of R-loop homeostasis is closely related to various human diseases, including cancer. However, the causality of aberrant R-loops in tumor progression remains unclear. In this study, using single-cell RNA-sequencing datasets from lung adenocarcinoma (LUAD), we constructed an R-loop scoring model to characterize the R-loop state according to the identified R-loop regulators related to EGFR mutations, tissue origins, and TNM stage. We then evaluated the relationships of the R-loop score with the tumor microenvironment (TME) and treatment response. Furthermore, the potential roles of FANCI-mediated R-loops in LUAD were explored using a series of in vitro experiments. Results showed that malignant cells with low R-loop scores displayed glycolysis and epithelial–mesenchymal transition pathway activation and immune escape promotion, thereby hampering the antitumor therapeutic effects. Cell communication analysis suggested that low R-loop scores contributed to T cell exhaustion. We subsequently validated the prognostic value of R-loop scores by using bulk transcriptome datasets across 33 tumor types. The R-loop scoring model well predicted patients’ therapeutic response to targeted therapy, chemotherapy, or immunotherapy in 32 independent cohorts. Remarkably, changes in R-loop distribution mediated by FANCI deficiency blocked the activity of Ras signaling pathway, suppressing tumor-cell proliferation and dissemination. In conclusion, this study reveals the underlying molecular mechanism of metabolic reprogramming and T cell exhaustion under R-loop score patterns, and the changes in R-loops mediated by R-loop regulators resulting in tumor progression. Therefore, incorporating anticancer methods based on R-loop or R-loop regulators into the treatment schemes of precision medicine may be beneficial. Supplementary Information The online version contains supplementary material available at 10.1186/s12943-023-01924-6.
... SETX has been identified as a binding partner for BRCA1 and can participate in the termination region of R-loop transcription [19]. When the BRCA1/SETX complex is disrupted, unrepaired ssDNA is broken, resulting in transcriptional-associated genomic instability [20]. SETX mutations are found in approximately 4% of metastatic prostate cancer and 1.2% of prostate adenocarcinoma [21]. ...
Article
Full-text available
Squamous differentiation of prostate cancer, which accounts for less than 1% of all cases, is typically associated with androgen deprivation treatment (ADT) or radiotherapy. This entity is aggressive and exhibits poor prognosis due to limited response to traditional treatment. However, the underlying molecular mechanisms and etiology are not fully understood. Previous findings suggest that squamous cell differentiation may potentially arise from prostate adenocarcinoma (AC), but further validation is required to confirm this hypothesis. This paper presents a case of advanced prostate cancer with a combined histologic pattern, including keratinizing SCC and AC. The study utilized whole-exome sequencing (WES) data to analyze both subtypes and identified a significant overlap in driver gene mutations between them. This suggests that the two components shared a common origin of clones. These findings emphasize the importance of personalized clinical management for prostate SCC, and specific molecular findings can help optimize treatment strategies.
... In addition to external threats that induce damage to DNA, the occurrence of unscheduled R-loops resulting from interference during DNA replication and transcription is a major cause of DNA damage and genome instability (11)(12)(13)(14)(15) . Typically, the R-loop is a nucleic acid structure of DNA / RNA hybrid with a displaced single-stranded DNA transiently formed during RNA transcription ( 11-13 , 16 , 17 ) . ...
Article
Full-text available
Aberrant DNA/RNA hybrids (R-loops) formed during transcription and replication disturbances pose threats to genome stability. DHX9 is an RNA helicase involved in R-loop resolution, but how DHX9 is regulated in response to genotoxic stress remains unclear. Here we report that DHX9 is phosphorylated at S321 and S688, with S321 phosphorylation primarily induced by ATR after DNA damage. Phosphorylation of DHX9 at S321 promotes its interaction with γH2AX, BRCA1 and RPA, and is required for its association with R-loops under genotoxic stress. Inhibition of ATR or expression of the non-phosphorylatable DHX9S321A prevents DHX9 from interacting with RPA and R-loops, leading to the accumulation of stress-induced R-loops. Furthermore, depletion of RPA reduces the association between DHX9 and γH2AX, and in vitro binding analysis confirms a direct interaction between DHX9 and RPA. Notably, cells with the non-phosphorylatable DHX9S321A variant exhibit hypersensitivity to genotoxic stress, while those expressing the phosphomimetic DHX9S321D variant prevent R-loop accumulation and display resistance to DNA damage agents. In summary, we uncover a new mechanism by which ATR directly regulates DHX9 through phosphorylation to eliminate stress-induced R-loops.
... Their removal by enzymes has been extensively studied. It is known that RNase H endonucleases cleave RNA within DNA:RNA hybrids; DNA:RNA helicases such as senataxin (SETX) or aquarius intron-binding spliceosomal factor (AQR) unwind the hybrid double helix; topoisomerases resolve the negative torsional stress behind the transcription bubble to prevent the annealing of nascent RNA with template DNA, and these enzymes can work in concert (Cerritelli and Crouch 2009;Skourti-Stathaki et al. 2011;Groh and Gromak 2014;Amon and Koshland 2016;Marinello et al. 2016;Costantino and Koshland 2018;Manzo et al. 2018;Malig et al. 2020;Niehrs and Luke 2020;Marchena-Cruz et al. 2023). In normally functioning cells, multiple R-loops should be produced and demolished in the right place at the right time, but it is not clear how this is achieved. ...
Article
Full-text available
Non-canonical structures (NCS) refer to the various forms of DNA that differ from the B-conformation described by Watson and Crick. It has been found that these structures are usual components of the genome, actively participating in its essential functions. The present review is focused on the nine kinds of NCS appearing or likely to appear in human ribosomal DNA (rDNA): supercoiling structures, R-loops, G-quadruplexes, i-motifs, DNA triplexes, cruciform structures, DNA bubbles, and A and Z DNA conformations. We discuss the conditions of their generation, including their sequence specificity, distribution within the locus, dynamics, and beneficial and detrimental role in the cell.
Article
Full-text available
Hypoxia is a common feature of solid tumors and is associated with poor patient prognosis, therapy resistance and metastasis. Radiobiological hypoxia (<0.1% O2) is one of the few physiologically relevant stresses that activates both the replication stress/DNA damage response and the unfolded protein response. Recently, we found that hypoxia also leads to the robust accumulation of R-loops, which led us to question here both the mechanism and consequence of hypoxia-induced R-loops. Interestingly, we found that the mechanism of R-loop accumulation in hypoxia is dependent on non-DNA damaging levels of reactive oxygen species. We show that hypoxia-induced R-loops play a critical role in the transcriptional stress response, evidenced by the repression of ribosomal RNA synthesis and the translocation of nucleolin from the nucleolus into the nucleoplasm. Upon depletion of R-loops, we observed a rescue of both rRNA transcription and nucleolin translocation in hypoxia. Mechanistically, R-loops accumulate on the rDNA in hypoxia and promote the deposition of heterochromatic H3K9me2 which leads to the inhibition of Pol I-mediated transcription of rRNA. These data highlight a novel mechanistic insight into the hypoxia-induced transcriptional stress response through the ROS-R-loop-H3K9me2 axis. Overall, this study highlights the contribution of transcriptional stress to hypoxia-mediated tumorigenesis.
Article
Full-text available
Simple Summary R-loops are three-stranded structures consisting of an RNA–DNA hybrid and an unpaired single-stranded DNA (ssDNA), with biological implications in cellular physiology and pathological conditions. R-loops interfere with DNA repair pathways and activate oncogenes, leading to dysregulated cell proliferation, genome instability, and cancer development. R-loops accumulate in several types of cancer cells, including breast, ovarian, prostate, and lung cancer. Studying the biological roles of R-loops in cancer development is potentially beneficial for innovative diagnostic and treatment approaches for cancer. In this review, we focus on recent advances in R-loops’ roles in genome instability, DNA repair, and oncogenic events. Abstract R-loops are unique, three-stranded nucleic acid structures that primarily form when an RNA molecule displaces one DNA strand and anneals to the complementary DNA strand in a double-stranded DNA molecule. R-loop formation can occur during natural processes, such as transcription, in which the nascent RNA molecule remains hybridized with the template DNA strand, while the non-template DNA strand is displaced. However, R-loops can also arise due to many non-natural processes, including DNA damage, dysregulation of RNA degradation pathways, and defects in RNA processing. Despite their prevalence throughout the whole genome, R-loops are predominantly found in actively transcribed gene regions, enabling R-loops to serve seemingly controversial roles. On one hand, the pathological accumulation of R-loops contributes to genome instability, a hallmark of cancer development that plays a role in tumorigenesis, cancer progression, and therapeutic resistance. On the other hand, R-loops play critical roles in regulating essential processes, such as gene expression, chromatin organization, class-switch recombination, mitochondrial DNA replication, and DNA repair. In this review, we summarize discoveries related to the formation, suppression, and removal of R-loops and their influence on genome instability, DNA repair, and oncogenic events. We have also discussed therapeutical opportunities by targeting pathological R-loops.
Article
Full-text available
R loops are three-stranded nucleic acid structures that comprise nascent RNA hybridized with the DNA template, leaving the nontemplate DNA single-stranded. R loops form naturally during transcription even though their persistent formation can be a risky outcome with deleterious effects on genome integrity. On the other hand, over the last few years, an increasingly strong case has been built for R loops as potential regulators of gene expression. Therefore, understanding their function and regulation under these opposite situations is essential to fully characterize the mechanisms that control genome integrity and gene expression. Here we review recent findings about these interesting structures that highlight their opposite roles in cellular fitness.
Article
Full-text available
Kaposi's sarcoma-associated herpesvirus (KSHV) is an oncogenic herpesvirus associated with multiple AIDS-related malignancies. Like other herpesviruses, KSHV has a biphasic life cycle and both the lytic and latent phases are required for tumorigenesis. Evidence suggests that KSHV lytic replication can cause genome instability in KSHV-infected cells, although no mechanism has thus far been described. A surprising link has recently been suggested between mRNA export, genome instability and cancer development. Notably, aberrations in the cellular transcription and export complex (hTREX) proteins have been identified in high-grade tumours and these defects contribute to genome instability. We have previously shown that the lytically expressed KSHV ORF57 protein interacts with the complete hTREX complex; therefore, we investigated the possible intriguing link between ORF57, hTREX and KSHV-induced genome instability. Herein, we show that lytically active KSHV infected cells induce a DNA damage response and, importantly, we demonstrate directly that this is due to DNA strand breaks. Furthermore, we show that sequestration of the hTREX complex by the KSHV ORF57 protein leads to this double strand break response and significant DNA damage. Moreover, we describe a novel mechanism showing that the genetic instability observed is a consequence of R-loop formation. Importantly, the link between hTREX sequestration and DNA damage may be a common feature in herpesvirus infection, as a similar phenotype was observed with the herpes simplex virus 1 (HSV-1) ICP27 protein. Our data provide a model of R-loop induced DNA damage in KSHV infected cells and describes a novel system for studying genome instability caused by aberrant hTREX.
Article
Full-text available
Author Summary Friedreich ataxia and Fragile X syndrome are among 40 human diseases associated with expansion of repeated sequences. In both disorders repeat expansion leads to gene silencing, the molecular mechanism of which is not well understood, impeding the development of specific therapies to treat these disorders. It is proposed that formation of unusual DNA structures (RNA/DNA hybrids, or R-loops) over repeat regions may play a role, but their molecular function has not been investigated in vivo. We show that R-loops form on expanded repeats of FXN and FMR1 genes in cells from FRDA and FXS patients. These R-loops are stable, correlate with repressive chromatin marks and hinder FXN transcription in patient cells. We studied the relationship between repressive chromatin and R-loops. Decrease in the amount of repressive chromatin has no effect on R-loop levels. In contrast, increase in the R-loops leads to transcriptional silencing of FXN gene and formation of repressive chromatin, providing a direct molecular link between R-loops and pathology of expansion diseases. This discovery is important for understanding the basic molecular mechanism underlying the pathology of expansion diseases. The ability of R-loops to trigger transcriptional silencing makes them an attractive target for future therapeutic approaches to treat these diseases.
Article
Full-text available
Expansion of a trinucleotide (CGG) repeat element within the 5' untranslated region (5'UTR) of the human FMR1 gene is responsible for a number of heritable disorders operating through distinct pathogenic mechanisms: gene silencing for fragile X syndrome (>200 CGG) and RNA toxic gain-of-function for FXTAS (∼55-200 CGG). Existing models have focused almost exclusively on post-transcriptional mechanisms, but co-transcriptional processes could also contribute to the molecular dysfunction of FMR1. We have observed that transcription through the GC-rich FMR1 5'UTR region favors R-loop formation, with the nascent (G-rich) RNA forming a stable RNA:DNA hybrid with the template DNA strand, thereby displacing the non-template DNA strand. Using DNA:RNA (hybrid) immunoprecipitation (DRIP) of genomic DNA from cultured human dermal fibroblasts with both normal (∼30 CGG repeats) and premutation (55<CGG<200 repeats) alleles, we provide evidence for FMR1 R-loop formation in human genomic DNA. Using a doxycycline (DOX)-inducible episomal system in which both the CGG-repeat and transcription frequency can be varied, we further show that R-loop formation increases with higher expression levels. Finally, non-denaturing bisulfite mapping of the displaced single-stranded DNA confirmed R-loop formation at the endogenous FMR1 locus and further indicated that R-loops formed over CGG repeats may be prone to structural complexities, including hairpin formation, not commonly associated with other R-loops. These observations introduce a new molecular feature of the FMR1 gene that is directly affected by CGG-repeat expansion and is likely to be involved in the associated cellular dysfunction.
Article
Full-text available
DNA:RNA hybrid formation is emerging as a significant cause of genome instability in biological systems ranging from bacteria to mammals. Here we describe the genome-wide distribution of DNA:RNA hybrid prone loci in Saccharomyces cerevisiae by DNA:RNA immunoprecipitation (DRIP) followed by hybridization on tiling microarray. These profiles show that DNA:RNA hybrids preferentially accumulated at rDNA, Ty1 and Ty2 transposons, telomeric repeat regions and a subset of open reading frames (ORFs). The latter are generally highly transcribed and have high GC content. Interestingly, significant DNA:RNA hybrid enrichment was also detected at genes associated with antisense transcripts. The expression of antisense-associated genes was also significantly altered upon overexpression of RNase H, which degrades the RNA in hybrids. Finally, we uncover mutant-specific differences in the DRIP profiles of a Sen1 helicase mutant, RNase H deletion mutant and Hpr1 THO complex mutant compared to wild type, suggesting different roles for these proteins in DNA:RNA hybrid biology. Our profiles of DNA:RNA hybrid prone loci provide a resource for understanding the properties of hybrid-forming regions in vivo, extend our knowledge of hybrid-mitigating enzymes, and contribute to models of antisense-mediated gene regulation. A summary of this paper was presented at the 26th International Conference on Yeast Genetics and Molecular Biology, August 2013.
Article
Genome instability is central to ageing, cancer and other diseases. It is not only proteins involved in DNA replication or the DNA damage response (DDR) that are important for maintaining genome integrity: from yeast to higher eukaryotes, mutations in genes involved in pre-mRNA splicing and in the biogenesis and export of messenger ribonucleoprotein (mRNP) also induce DNA damage and genome instability. This instability is frequently mediated by R-loops formed by DNA-RNA hybrids and a displaced single-stranded DNA. Here we show that the human TREX-2 complex, which is involved in mRNP biogenesis and export, prevents genome instability as determined by the accumulation of γ-H2AX (Ser-139 phosphorylated histone H2AX) and 53BP1 foci and single-cell electrophoresis in cells depleted of the TREX-2 subunits PCID2, GANP and DSS1. We show that the BRCA2 repair factor, which binds to DSS1, also associates with PCID2 in the cell. The use of an enhanced green fluorescent protein-tagged hybrid-binding domain of RNase H1 and the S9.6 antibody did not detect R-loops in TREX-2-depleted cells, but did detect the accumulation of R-loops in BRCA2-depleted cells. The results indicate that R-loops are frequently formed in cells and that BRCA2 is required for their processing. This link between BRCA2 and RNA-mediated genome instability indicates that R-loops may be a chief source of replication stress and cancer-associated instability.
Article
Accurate DNA replication and DNA repair are crucial for the maintenance of genome stability, and it is generally accepted that failure of these processes is a major source of DNA damage in cells. Intriguingly, recent evidence suggests that DNA damage is more likely to occur at genomic loci with high transcriptional activity. Furthermore, loss of certain RNA processing factors in eukaryotic cells is associated with increased formation of co-transcriptional RNA:DNA hybrid structures known as R-loops, resulting in double-strand breaks (DSBs) and DNA damage. However, the molecular mechanisms by which R-loop structures ultimately lead to DNA breaks and genome instability is not well understood. In this review, we summarize the current knowledge about the formation, recognition and processing of RNA:DNA hybrids, and discuss possible mechanisms by which these structures contribute to DNA damage and genome instability in the cell.