You are on page 1of 7

Drug Resistance Updates 32 (2017) 16–22

Contents lists available at ScienceDirect

Drug Resistance Updates


journal homepage: www.elsevier.com/locate/drup

In cancer, A-to-I RNA editing can be the driver, the passenger, or the MARK
mechanic

Nabeel S. Ganem1, Noa Ben-Asher1, Ayelet T. Lamm
Faculty of Biology, Technion - Israel Institute of Technology, Technion City, Haifa 32000, Israel

A R T I C L E I N F O A B S T R A C T

Keywords: In recent years, A-to-I RNA modifications performed by the Adenosine Deaminase Acting on RNA (ADAR)
ADAR protein family were found to be expressed at altered levels in multiple human malignancies. A-to-I RNA editing
Chemotherapeutic drug sensitivity changes adenosine to inosine on double stranded RNA, thereby changing transcript sequence and structure.
Cancer biomarkers Although A-to-I RNA editing have the potential to change essential mRNA transcripts, affecting their corre-
Gene therapy
sponding protein structures, most of the human editing sites identified to date reside in non-coding repetitive
transcripts such as Alu elements. Therefore, the impact of the hypo- or hyper-editing found in specific cancers
remains unknown. Moreover, it is yet unclear whether or not changes in RNA editing and ADAR expression
levels facilitate or even drive cancer progression or are just a byproduct of other affected pathways. In both
cases, however, the levels of RNA editing and ADAR enzymes can possibly be used as specific biomarkers, as
their levels change differently in specific malignancies. More significantly, recent studies suggest that ADAR
enzymes can be used to reverse the oncogenic process, suggesting a potential for gene therapies. This review
focuses on new findings that suggest that RNA editing by ADARs can affect cancer progression and even for-
mation. We also discuss new possibilities of using ADAR enzymes and RNA editing as cancer biomarkers, in-
dicators of chemotherapeutic drug sensitivity, and even to be themselves potential therapeutic tools.

Introduction to A-to-I RNA editing domain is probably non-functional (Melcher et al., 1996a). ADAR1 also
has Z-DNA binding domains (ZDBD), whose functional activity is cur-
A-to-I RNA editing was discovered 30 years ago and is one of the rently unknown (Herbert et al., 1997).
most studied transcriptome modifications (Bass and Weintraub, 1987; In addition, there are two more ADAR-related genes in mammals,
Rebagliati and Melton, 1987). However, only in the last few years has ADAD1 and ADAD2, which are exclusively expressed in the testis and
A-to-I RNA editing been possibly implicated in cancer formation and are enzymatically inactive with a yet unknown function (Schumacher
progression. Specifically, A-to-I RNA editing is the deamination of et al., 1995). The ADAR1 gene encodes two main isoforms from alter-
adenosine to inosine on double stranded RNAs (dsRNAs) (Fig. 1A), native promoters, ADAR1p110 and ADAR1p150 (Fig. 1B). ADAR1p110
which is performed by ADAR (Adenosine Deaminase Acting on RNA) is the shorter isoform. It is constitutively expressed and mainly loca-
protein family (Bass, 2002). Inosine residues are recognized by the lized in the nucleus. In contrast, ADAR1p150 is mainly cytoplasmic and
cellular machinery as guanosine. Therefore, A-to-I editing in coding its expression is induced by the interferon pathway (Patterson and
regions can recode protein sequences and generate proteins with new Samuel, 1995; Liddicoat et al., 2016). The ADAR2 gene is mostly ex-
functions (reviewed in (Nishikura, 2016)). Currently, there are three pressed in the brain and is localized in the nucleus (Maas and
known ADAR proteins in mammals, ADAR1, ADAR2 and ADAR3. Gommans, 2009). ADAR1 and ADAR2 were shown in vitro to homo-
ADAR1 and ADAR2 are ubiquitously expressed, whereas ADAR3 is dimerize (Cho et al., 2003) and they differ in their editing targets.
exclusively expressed in the brain (Melcher et al., 1996a; Melcher et al., Specific editing sites that alter proteins were mainly found in the brain.
1996b). All ADAR proteins have two main domains, a catalytic dea- One of the most studied edited transcript is that of glutamate ionotropic
minase domain at their C-terminal region and a number of dsRNA receptor AMPA type subunit 2 (GRIA2) (Sommer et al., 1991), which is
binding domains (dsRBD) at their N-terminal region ((Bass, 2002), part of a family of glutamate receptors that are sensitive to α-amino-3-
(Thomas and Beal, 2017), Fig. 1B). The ADAR3 catalytic deaminase hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA), and function as


Corresponding author.
E-mail address: ayeletla@technion.ac.il (A.T. Lamm).
1
These authors contributed equally to this work.

http://dx.doi.org/10.1016/j.drup.2017.09.001

1368-7646/ © 2017 Elsevier Ltd. All rights reserved.


N.S. Ganem et al. Drug Resistance Updates 32 (2017) 16–22

Fig. 1. Schematic representation of the deamination


reaction and human ADAR protein family. (A) An
amine group removal leads to the conversion of
adenosine to inosine by an ADAR enzyme. Guanosine
is shown to illustrate the structural similarity with
inosine. (B) Members of the human ADAR family.
The two main isoforms of ADAR1 are presented,
ADAR1 p110 and ADAR1 p150. The deaminase do-
mains are colored in green, the RNA binding do-
mains (RBD) are colored in red and Z DNA binding
domains (Z −DBD) are colored in blue. Proteins are
scaled. (For interpretation of the references to colour
in this figure legend, the reader is referred to the web
version of this article.)

ligand-activated cation channels. These channels are assembled from 4 childhood (Zhang et al., 2004). ADAR1 mutations are also associated
related subunits, GRIA1-4. RNA editing in GRIA2 is thought to render with Aicardi-Goutières syndrome (AGS; OMIM # 225750), an auto-
the channel impermeable to Ca+2. Codon editing in GRIA2 subunit, somal dominant autoimmune disorder characterized in its most severe
which in some cases is edited to an extent of nearly 100% in transcripts, form by cerebral atrophy, leukodystrophy, intracranial calcifications
is needed to keep neuronal Ca2+ homeostasis. Indeed, mice harboring and chronic cerebrospinal fluid (CSF) lymphocytosis. This syndrome
mutations in ADARs that lack the ability to edit these transcripts exhibit results from the chronic activation of type I interferon (Rice et al.,
an early onset of epileptic seizures and premature death (Brusa et al., 2012). Since editing that affects protein structure is mostly abundant in
1995). RNA editing can also alter splice sites and eliminate stop codons. the nervous system, it is not surprising that changes in transcript editing
Interestingly, ADAR2 edits its own pre-mRNA, resulting in a novel levels were shown to result in neurological disorders including: Alz-
splice site that leads to a truncated protein due to the formation of a heimer’s disease (Gaisler-Salomon et al., 2014), amyotrophic lateral
premature stop codon (Feng et al., 2006). sclerosis (ALS) (Hideyama et al., 2010), and autism spectrum diseases
In recent years, advances in high-throughput sequencing led to the (Eran et al., 2013).
discovery that most editing events occur in clusters in non-coding re- In the current review, we focus on the involvement of ADARs and A-
petitive transcripts, termed ‘hyperediting’, primarily in Alu repeat ele- to-I editing in cancer, including the ability to use editing sites as di-
ments. In fact, editing events occur in more than half of the human agnostic biomarkers, and new developments of using ADAR proteins as
transcriptome (Bazak et al., 2014). The level of editing (i.e., percentage druggable targets for cancer therapeutics.
of transcripts undergoing editing in a particular site) at site-specific
coding sites is higher than that usually found in ‘hyperediting’ sites and The function of ADARs in development and immunity
is mostly associated with ADAR2 function.
Generation of microRNA can also be affected by A-to-I editing. Some Given the substantial amount of editing found in the human tran-
pri-microRNAs undergo RNA editing and this editing prevents their scriptome, it is not surprising that RNA editing by ADARs has essential
processing and affects their ability to produce mature microRNAs roles. Both ADAR1 and ADAR2 are required for normal development
(miRNAs) (Kawahara et al., 2007a; Yang et al., 2006). miRNAs can be and knockout mice for either of these genes die early in development
edited at their seed regions, which can affect their binding efficiency to (Higuchi et al., 2000; Wang et al., 2000). The early embryonic lethality
their targets and even alter their target specificity (Kawahara et al., in ADAR2 knockout mice is partially due to the lack of editing in GRIA2
2007b). Editing in the target miRNA binding site can also affect the RNA, a glutamate ionotropic receptor (see above, (Higuchi et al.,
efficiency of the miRNA binding and regulate the target expression (Cho 2000)). ADAR1 knockout mice phenotypes led to our understanding of
et al., 2017). its possible important role in the innate immune response (Hartner
A-to-I RNA editing is essential in mice and humans. Mutations in et al., 2004; Wang et al., 2004). ADAR1 was found to be a key player in
ADAR genes as well as mutations that affect ADAR’s editing function- the suppression of the antiviral dsRNA sensing type I interferon
ality are associated with several human diseases. Mutations in the pathway (Liddicoat et al., 2015) by preventing self-RNAs from trig-
ADAR1 gene, mainly in its deaminase domain, are associated with the gering the innate immune response (George et al., 2016). ADARs can
pathogenesis of dyschromatosis symmetrica hereditaria (DSH; OMIM # also be pro-viral for specific viruses (reviewed in (Samuel, 2012)).
127400). The latter is a rare autosomal dominant skin disorder char- ADAR1 is also induced by the interferon response (Patterson and
acterized by hyperpigmented and hypopigmented macules on the face Samuel, 1995), which might be linked to the chronic inflammatory
and dorsal aspects of the extremities that appear in infancy or early state observed in several types of cancer (Fumagalli et al., 2015). ADAR

17
N.S. Ganem et al. Drug Resistance Updates 32 (2017) 16–22

and A-to-I RNA editing functions in organismal development and its In metastatic malignant melanomas, ADAR1 is downregulated while
immunity were shown in invertebrates and other organisms as well ADAR2 levels are unchanged (Nemlich et al., 2013). This change in
(reviewed in (Ganem and Lamm, 2017)). expression is accompanied by altered expression of many other genes
and miRNAs, most of which do not undergo RNA editing. In vitro,
ADAR enzymes and A-to-I editing in cancer ADAR1 silencing enhances the malignant phenotype, while conversely,
overexpression of ADAR1 inhibits melanoma cells proliferation
For many years, cancer research focused on mutations in genes that (Nemlich et al., 2013). This inhibition is independent of ADAR1 editing
can lead to cancer initiation and progression, including mutations that activity as catalytically-inactive proteins displayed the same effect as
cause cells to proliferate without constraints, mutations that affect cell the active protein. Downregulation of ADAR1 in melanoma has been
cycle checkpoints, and mutations that induce further mutations by shown to be mediated by overexpression of two miRNAs that target
impairing mechanisms of DNA damage repair. Epigenetics mechanisms ADAR1, miR-17 and miR-432 (Nemlich et al., 2013). High level ex-
were also shown to be important in formation of neoplasia and their pression patterns of ADAR1 with no changes in the expression of
abnormalities, including alterations in methylation patterns or chro- ADAR2 are found in esophageal squamous cell carcinoma (ESCC) (Qin
matin remodeling, which are common in many cancer types (Sharma et al., 2014). Higher expression levels of ADAR1 and higher actual
et al., 2010). Therefore, it was predicted that A-to-I editing would be editing levels have been correlated with poor ESCC patient prognosis
significantly altered in cancer. (Qin et al., 2014).
The expression of ADARs and the corresponding editing capacity While the studies in the abovementioned cancers concentrated on
have been studied in various cancer types, revealing distinct patterns in specific tumor and editing targets, three recent studies comprehensively
different tumors. In glioblastoma multiforme (GBM), the most ag- analyzed the role of ADARs in cancer by using the data of Cancer
gressive type of central nervous system tumor, lower ADAR2-mediated Genome Atlas (TCGA) collection (Fumagalli et al., 2015; Han et al.,
editing was observed, specifically in GRIA2 mRNA, without a corre- 2015; Paz-Yaacov et al., 2015). They found that in most cancer types,
sponding downregulation of ADAR2 expression (Maas et al., 2001). In editing levels are higher than in the normal paired tissues (see examples
vitro and in vivo experiments in which ADAR2 was overexpressed re- in Fig. 2). While all ADAR family members displayed changes in ex-
versed the malignant phenotype of glioblastoma cells (Cenci et al., pression in many tumors, ADAR1 seems to be the most important family
2008; Galeano et al., 2013). In contrast, both ADAR1 and ADAR3 were member in the context of cancer, as alterations in its expression were
found to be upregulated in brain tumors (Cenci et al., 2008; Oakes observed in all the tumors tested. Differential expression is achieved by
et al., 2017), while a different study found that all ADAR gene ex- distinct modalities: gene amplification, alternative splicing and down-
pression levels were reduced (Paz et al., 2007). Interestingly, between regulation through miRNAs. The opposite effects that ADAR1 has on
the two ADAR1 isoforms, only the p110 isoform is expressed in GBM, different tumor types can be attributed to different targets of RNA
since a deletion caused by a splicing variant in the p150 isoform creates editing and different downstream effectors in the different tissues.
a frameshift mutation (Cenci et al., 2008). The lower editing rate in One open question that comes up in many cancer studies, is whether
glioblastoma might result from inhibition of ADAR2 editing by the or not these differences between cancer cells and their paired normal
overexpressed ADAR1, which was shown to inhibit ADAR2 in vitro, in a cells are a byproduct of the many genomic abnormalities that occur in
dose-dependent manner (Cenci et al., 2008). ADAR3 overexpression cancer progression (passenger alterations) or whether the observed
might also be involved as it was shown to inhibit GRIA2 editing, pos- differences are the drivers of cancer progression. Concerning RNA
sibly by competing with ADAR2 on GRIA2 mRNA binding (Oakes et al., editing, the answer is yet unclear. Changes in gene editing levels that
2017). Another possibility is that a splicing variant of ADAR2 is ex- are linked to cancer and in cancer cell microRNA binding sites (Han
pressed, which harbors a diminished catalytic activity (Maas et al., et al., 2015; Paz-Yaacov et al., 2015), might be driver alterations
2001). ADAR2 was also shown to alter the expression and editing levels events. Another indication that ADAR1 overexpression might drive
of microRNAs that are involved in tumorigenesis in glioblastoma cells cancer is that upon knockdown of ADAR1 expression in breast cancer
(Tomaselli et al., 2015). cells, a decrease in cell proliferation and an increase in apoptosis is
Apart from solid tumors, in chronic myeloid leukemia (CML), higher observed (Fumagalli et al., 2015). However, ADAR1 expression is
expression of the inflammatory INF-related pathway and ADAR1p150 known to be induced by the type 1 interferon response (Liddicoat et al.,
isoform were observed (Jiang et al., 2013). Expression of ADAR1p110, 2016; Rice et al., 2012), which is related to the chronic inflammatory
which is not dependent on the inflammation signaling axis, was un- state commonly seen in cancer. Therefore, the changes in RNA editing
changed. In more advanced leukemias, more A-to-I RNA editing was might be a result of the inflammatory environment of the tumors, which
observed, mainly in Alu-repeat sequences (Paz-Yaacov et al., 2015). In constitutes a passenger position (Fumagalli et al., 2015).
vitro studies showed that overexpression of ADAR1p150 drives hema- In addition to showing that cancer cells differ in their ADAR activity
topoietic differentiation towards the myeloid lineage, while ADAR1 from normal cells, these studies have also raised the possibility of using
knockout impairs malignant myeloid progenitor self-renewal (Jiang RNA editing levels as cancer biomarkers and ADAR enzymes as bona
et al., 2013; Steinman et al., 2013). fide drug targets. These possible therapeutic avenues are discussed
In hepatocellular carcinoma (HCC), a disrupted editing balance has below.
been observed in tumor tissues by RNA-seq analysis (Chan et al., 2014).
In addition, ADAR1 upregulation and ADAR2 downregulation corre- A-to-I editing as cancer markers
lated with increased risks of liver cirrhosis and occurrence of hepato-
cellular cancer (Chan et al., 2014). Cancer diagnosis, staging of the disease and the patient’s response to
ADAR1 was shown to edit antizyme inhibitor-1 (AZIN1) mRNA and therapy can be predicted by using unique and reliable biomarkers.
cause amino acid composition change in the protein (Chen et al., 2013). These biomarkers can be specific to a certain type of cancer or can be
Antizyme inhibitors are homologs of ornithine decarboxylase (ODC, the present in several cancer types. Some tumor biomarkers are proteins
key enzyme in polyamine biosynthesis), which are devoid of ornithine that are present on normal and cancer cells or are secreted into the
decarboxylation activity. Hence, antizyme inhibitors function as posi- body’s fluids (Mehta et al., 2010; Petricoin et al., 2006; Sawyers, 2008).
tive regulators of polyamine levels by sequestering antizymes (the latter mRNA levels of specific transcripts, epigenetics changes, and genetic
block the key enzyme in polyamine biosynthesis, ODC) and thereby variations are also common biomarkers. However, in some cases, the
neutralize their effect. AZIN1 editing causes an increase in cell pro- levels of specific tumor markers can be elevated under benign condi-
liferation and high levels of edited AZIN1 were suggested to be a driver tions from unknown reasons. In addition, a specific cancer type can
of HCC (Chen et al., 2013). express distinct biomarkers in different patients due to cancer

18
N.S. Ganem et al. Drug Resistance Updates 32 (2017) 16–22

Fig. 2. Altered ADAR expression and RNA editing levels in human cancers. The scheme presents some findings of changes in ADAR expression (mainly ADAR1) and RNA editing levels in
specific tumors. Alterations in ADAR expression and RNA editing levels are presented by blue and red arrows, respectively. Arrows pointing upwards denote upregulation and arrows
pointing downwards represent downregulation. Data was taken from the following studies: (1) (Han et al., 2015), (2) (Paz-Yaacov et al., 2015), (3) (Fumagalli et al., 2015), (4) (Qin et al.,
2014). Cancer types that appear in black are common to both genders. Prostate adenocarcinoma (PRAD), a male-gendered cancer, appears in blue. Breast cancer, which is predominantly
found in females, appears in pink. (For interpretation of the references to colour in this figure legend, the reader is referred to the web version of this article.)

heterogeneity originating from genomic instability (Mehta et al., 2010; development of novel therapeutic interventions (Han et al., 2015). This
Sawyers, 2008). Clearly however, cancer biomarkers are not available opens the possibility to predict the impact of cancer treatments in in-
yet for various cancer types. This stresses the importance of identifying dividual patients based on specific transcript editing patterns. To ex-
reliable markers for classifying cancer types and for evaluating the plore the RNA editing impact on drug sensitivity, Han et al. (Han et al.,
patient’s response to certain drug treatments (Mehta et al., 2010; 2015) performed cell viability assays on tumor cells after introducing
Sawyers, 2008). In this respect, Paz-Yaacov et al. (Paz-Yaacov et al., mutations in genes that mimic the tumor’s unique RNA editing events
2015) have examined the possibility of using editing levels as a novel such as AZIN1S367G, GRIA2R764G, and COG3I635V. They found an
cancer biomarker. They determined editing levels in Alu sequences in increase in cell survival, which suggests that these editing events have a
several cancer types and found a correlation between low editing levels direct effect on cancer progression. Next, to explore the possibility that
in Alu and higher survival rates in head and neck, liver, and breast these editing events can affect the cancer cells’ response to che-
cancers. Hyper-editing in Alu might reflect loss of genomic stability and motherapeutic drugs, they did a cytotoxicity screen and found that
therefore a more advanced tumor progression (Paz-Yaacov et al., 2015). editing events can affect cancer cell sensitivity to several compounds
However, this finding is currently restricted to certain types of cancer as used for targeted therapeutics. Using the Cancer Cell Line Encyclopedia
some cancer types show hypo-editing (Han et al., 2015). Moreover, as (CCLE) (Barretina et al., 2012), they also found a correlation between
shown above, altered editing levels in specific transcripts can also re- nonsynonymous RNA editing levels and anticancer drug sensitivity.
port on the cancer state. Wang and colleagues did a similar screen using It is also important to note that drugs can also affect the expression
the TCGA dataset to study the correlation of editing levels in cancer levels of ADAR enzymes thereby bringing about hyper- or hypo-editing
types using editing in miRNAs as markers (Wang et al., 2017). They events and possibly editing events in positions that were not edited
found that editing “hot-spots” in miRNAs are associated with estab- before. For example, cocaine can lead to reduced ADAR2 expression
lished tumor subtypes, cancer stage, and patient survival time. Editing and subsequently diminished editing activity (Schmidt et al., 2015).
in miRNAs can affect their target selection, which can be for instance Antidepressant drugs were found to cause alteration in RNA editing
oncogenes and tumor suppressor genes. For example, editing in miR- levels of 5-HT2C (Gurevich et al., 2002) and GluR2 receptors
200b, a key tumor metastasis suppressor, correlates with patient (Burnashev et al., 1995). Furthermore, RNA editing can alter the sen-
prognosis and might serve as a strong biomarker (Wang et al., 2017). sitivity and the pharmacological properties of channels and drug tar-
gets. For example, RNA editing of the Kv1.1 channels alters their sen-
RNA editing and chemotherapeutic drug sensitivity sitivity to the Kv channel blocker 4-aminopyridine (4-AP) and reduce
the ictogenic potential to 4-AP in epileptic rats (Streit et al., 2011). In
While A-to-I RNA editing may be possibly used for cancer diag- addition, due to RNA editing in its α1 subunit, GABA receptor sensi-
nostics and prognosis, it was recently suggested that when editing tivity changes and its deactivation rate increases (Nimmich et al.,
events are dysregulated in cancer, they could target and affect many 2009).
pathways; hence, the latter may serve as possible druggable targets for To conclude, RNA editing can provide a selective effect on cell

19
N.S. Ganem et al. Drug Resistance Updates 32 (2017) 16–22

sensitivity to various drugs by targeting relevant pathways and thus Schneider, 2012) uses the ADAR1 deaminase domain without its RNA
should be considered in the development of novel therapeutics for binding domains (dsRBD) fused to a C-terminus SNAP-tag protein
different indications. conjugated to 5′-O-benzylguanine (BG) modified guide RNA (Fig. 3A).
The guide RNA recognizes its antisense target and forms a secondary
Can A-to-I editing be used for cancer therapeutics? structure that enables editing by the ADAR1 deaminase domain. This
system was shown to successfully diminish a stop codon and correct
Both ADARs and their edited transcripts are involved in cancer missense mutations in high efficiency in vitro and in vivo (Stafforst and
progression. Therefore, ADARs might also be used as targets for novel Schneider, 2012; Vogel et al., 2014), and was even adjusted to be light
cancer therapies. Indeed, knocking out ADAR1 in a CML mouse model activated (Hanswillemenke et al., 2015).
caused rapid leukemic cell death (Steinman et al., 2013) and knocking Another technique uses the ADAR2 deaminase domain fused to a λ
down ADAR1 in breast cancer cell lines caused increased apoptosis phage − N protein, a 22 amino acid peptide, which recognizes and
(Fumagalli et al., 2015). ADAR1 is induced by the interferon response binds to the boxB RNA hairpin (Montiel-Gonzalez et al., 2013). To
pathway, which can be another possible therapeutic target (Patterson target specific RNA, the boxB RNA is fused to an antisense guide RNA
and Samuel, 1995). However, ADARs have many roles in immunity and that can bind to endogenous RNA (Fig. 3B). After recognizing the en-
organismal development, some of which are still unknown (Ganem and dogenous RNA, it can undergo RNA editing by the ADAR2 deaminase
Lamm, 2017; Goldstein et al., 2017). Therefore, targeting ADARs may domain-λ-N fused protein. The system was shown to correct the W496X
cause unforeseen and harmful side effects. mutation in the cystic fibrosis transmembrane conductance regulator
The discovery of the genome editing ability of the CRISPR-Cas9 tool (CFTR) transcript both in vitro and in vivo (Montiel-Gonzalez et al.,
opened new avenues for successful gene therapies (Boettcher and 2013). While the percentage of corrected transcripts was very low,
McManus, 2015; Cox et al., 2015; Fellmann et al., 2017). Although CFTR activity was restored in vivo. The system’s efficiency was im-
other approaches such as delivery of shRNA are already on the market, proved by introducing an E488Q mutation in the deaminase domain, by
CRISPR-Cas9 DNA editing appears to be the most promising (Boettcher increasing the number of the boxB hairpins to two, and by using mul-
and McManus, 2015). However, there are many obstacles that raise tiple λ-N peptides (Montiel-González et al., 2016).
concerns in using CRISPR-Cas9 for human therapies. Some of the con- A very recent approach has been to use a modified target RNA that
cerns are off-target editing in other DNA positions, the inability to ac- recruits endogenous ADARs and fuses them to an antisense fragment
cess the nucleus to reach genomic DNA, and the low efficiency of complementary to the desired target (Fukuda et al., 2017; Heep et al.,
editing (Cox et al., 2015). 2017; Wettengel et al., 2017). The guide RNA is based on a known
In the last few years, several studies suggested some possible ADAR substrate, which was shown to be efficiently edited. The anti-
modalities to exploit the capacity of ADAR proteins to edit RNA for sense fragment is designed as a template for the deamination. It as-
correcting genetic mutations on the mRNA levels or cause mutations in sembles to a dsRNA with the target RNA and undergoes RNA editing by
specific positions (see below and in Fig. 3). The possibility to edit the ADAR deamination motif (Fig. 1C). Efficient target RNAs were
mRNA instead of DNA is very appealing in terms of an enhanced ac- constructed to both ADAR1 isoforms and to ADAR2 (Fukuda et al.,
cessibility to the mRNA, which resides in the cytoplasm, and a more 2017; Heep et al., 2017; Wettengel et al., 2017). This approach for site-
transient approach, which makes off-target editing less of a problem. directed RNA editing might be more suitable for gene therapy and
These new strategies utilize the ability of the ADAR enzymes to dea- mutation recoding since it uses the endogenous ADAR proteins and
minate a certain nucleotide and convert it to another nucleotide. The does not require a newly constructed protein. This methodology was
ability of ADAR enzymes to bind to their target substrates depends on used to correct a recessive stop mutation in PINK1, enabling mi-
the RNA strand’s secondary structure. Altering the secondary structure tochondria autophagy in HeLa cells (Wettengel et al., 2017).
of the desired targets could enable the binding of ADAR enzymes and Although site-directed RNA editing technology is not as advanced as
consequent editing of the target. To date, a few site-directed RNA CRISPR-Cas9 and was not tested yet in variety of disease models or
editing (SDRE) techniques utilizing ADARs exist (for example: (Fukuda cancer cells, it bears a great potential. This is especially true if this
et al., 2017; Montiel-Gonzalez et al., 2013; Stafforst and Schneider, technique will pass the specific A-to-I editing barrier and will enable the
2012)). These techniques use different approaches to convert a single editing of other nucleotides.
nucleotide from adenosine to inosine. One approach (Stafforst and

Fig. 3. Site-directed RNA editing techniques. (A) ADAR1 deaminase domain (DD) fused to SNAP-tag protein which is conjugated to 5′-O-benzylguanine and guide RNA (gRNA). The
gRNA-target RNA complex undergoes RNA editing by the deaminase domain. (B) ADAR2 deaminase domain (DD) fused to a λ phage − N domain (λN-D). λN-D recognizes and binds a
boxB RNA, which is fused to a guide RNA (gRNA). The gRNA-target RNA complex undergoes RNA editing by the deaminase domain. (C) ADAR RNA binding domain (RBD) substrate
transcript fused to a guide RNA (gRNA) recruits the endogenous ADAR enzyme, which in turn binds it and deaminates the gRNA-target RNA complex.

20
N.S. Ganem et al. Drug Resistance Updates 32 (2017) 16–22

Conclusions and future perspectives Feng, Y., Sansam, C.L., Singh, M., Emeson, R.B., 2006. Altered RNA editing in mice
lacking ADAR2 autoregulation. Mol. Cell. Biol. 26, 480–488.
Fukuda, M., Umeno, H., Nose, K., Nishitarumizu, A., Noguchi, R., Nakagawa, H., 2017.
Cancer studies involving ADAR RNA editing are the “new kids on Construction of a guide-RNA for site-directed RNA mutagenesis utilising intracellular
the block”. It is apparent that alterations in ADAR expression and A-to-I RNA editing. Sci. Rep. 7, 41478.
Fumagalli, D., Gacquer, D., Rothé, F., Lefort, A., Libert, F., Brown, D., Kheddoumi, N.,
function in human malignancies affect the expression and activity of Shlien, A., Konopka, T., Salgado, R., Larsimont, D., Polyak, K., Willard-Gallo, K.,
many transcripts and consequently key proteins including oncogenes, Desmedt, C., Piccart, M., Abramowicz, M., Campbell, P.J., Sotiriou, C., Detours, V.,
tumor suppressor genes and their regulators. It is still an open question 2015. Principles governing A-to-I RNA editing in the Breast cancer transcriptome.
Cell Rep. 13, 277–289.
whether or not ADAR editing is a driver of cancer progression or simply Gaisler-Salomon, I., Kravitz, E., Feiler, Y., Safran, M., Biegon, A., Amariglio, N., Rechavi,
a passenger alteration. Nevertheless, RNA editing should be considered G., 2014. Hippocampus-specific deficiency in RNA editing of GluA2 in Alzheimer's
as a biomarker for certain cancer types and stages as well as for ther- disease. Neurobiol. Aging 35, 1785–1791.
Galeano, F., Rossetti, C., Tomaselli, S., Cifaldi, L., Lezzerini, M., Pezzullo, M., Boldrini, R.,
apeutic drug interventions and outcomes. The most exciting aspect is
Massimi, L., Di Rocco, C.M., Locatelli, F., Gallo, A., 2013. ADAR2-editing activity
the possibility of using the ADAR mechanism for treating cancer and inhibits glioblastoma growth through the modulation of the CDC14B/Skp2/p21/p27
other diseases. Transiently targeting specific RNA transcripts is very axis. Oncogene 32, 998–1009.
appealing and with the development of targeted editing, this option Ganem, N.S., Lamm, A.T., 2017. A-to-I RNA editing − thinking beyond the single nu-
cleotide. RNA Biol. http://dx.doi.org/10.1080/15476286.2017.1364830. [Epub
may become a realistic therapeutic approach in the future. ahead of print].
George, C.X., Ramaswami, G., Li, J.B., Samuel, C.E., 2016. Editing of cellular self-RNAs by
Acknowledgements adenosine deaminase ADAR1 suppresses innate immune stress responses. J. Biol.
Chem 291, 6158–6168.
Goldstein, B., Agranat-Tamir, L., Light, D., Ben-Naim Zgayer, O., Fishman, A., Lamm,
We thank Alla Fishman for critical reading of the manuscript and A.T., 2017. A-to-I RNA editing promotes developmental stage-specific gene and
discussions. This work was funded by The Israeli Centers of Research lncRNA expression. Genome Res. 27, 462–470.
Gurevich, I., Tamir, H., Arango, V., Dwork, A.J., Mann, J.J., Schmauss, C., 2002. Altered
Excellence (I-CORE) program, (Center No. 1796/12 to ATL), Israel editing of serotonin 2C receptor pre-mRNA in the prefrontal cortex of depressed
Cancer Research Fund (ICRF), and the Binational Israel-USA Science suicide victims. Neuron 34, 349–356.
Foundation (grant no. 2015091). Han, L., Diao, L., Yu, S., Xu, X., Li, J., Zhang, R., Yang, Y., Werner, H.M.J., Eterovic, A.K.,
Yuan, Y., Li, J., Nair, N., Minelli, R., Tsang, Y.H., Cheung, L.W.T., Jeong, K.J., Roszik,
J., Ju, Z., Woodman, S.E., Lu, Y., Scott, K.L., Li, J.B., Mills, G.B., Liang, H., 2015. The
References genomic landscape and clinical relevance of A-to-I RNA editing in human cancers.
Cancer Cell 28, 515–528.
Hanswillemenke, A., Kuzdere, T., Vogel, P., Jékely, G., Stafforst, T., 2015. Site-Directed
Barretina, J., Caponigro, G., Stransky, N., Venkatesan, K., Margolin, A.A., Kim, S., Wilson,
RNA editing in vivo can Be triggered by the light-driven assembly of an artificial
C.J., Lehár, J., Kryukov, G.V., Sonkin, D., Reddy, A., Liu, M., Murray, L., Berger, M.F.,
riboprotein. J. Am. Chem. Soc. 137, 15875–15881.
Monahan, J.E., Morais, P., Meltzer, J., Korejwa, A., Jané-Valbuena, J., Mapa, F.A.,
Hartner, J.C., Schmittwolf, C., Kispert, A., Muller, A.M., Higuchi, M., Seeburg, P.H., 2004.
Thibault, J., Bric-Furlong, E., Raman, P., Shipway, A., Engels, I.H., Cheng, J., Yu,
Liver disintegration in the mouse embryo caused by deficiency in the RNA-editing
G.K., Yu, J., Aspesi, P., de Silva, M., Jagtap, K., Jones, M.D., Wang, L., Hatton, C.,
enzyme ADAR1. J. Biol. Chem. 279, 4894–4902.
Palescandolo, E., Gupta, S., Mahan, S., Sougnez, C., Onofrio, R.C., Liefeld, T.,
Heep, M., Mach, P., Reautschnig, P., Wettengel, J., Stafforst, T., 2017. Applying human
MacConaill, L., Winckler, W., Reich, M., Li, N., Mesirov, J.P., Gabriel, S.B., Getz, G.,
ADAR1p110 and ADAR1p150 for site-Directed RNA editing-G/C substitution stabi-
Ardlie, K., Chan, V., Myer, V.E., Weber, B.L., Porter, J., Warmuth, M., Finan, P.,
lizes GuideRNAs against editing. Genes (Basel) 8.
Harris, J.L., Meyerson, M., Golub, T.R., Morrissey, M.P., Sellers, W.R., Schlegel, R.,
Herbert, A., Alfken, J., Kim, Y.G., Mian, I.S., Nishikura, K., Rich, A., 1997. A Z-DNA
Garraway, L.A., 2012. The Cancer Cell Line Encyclopedia enables predictive mod-
binding domain present in the human editing enzyme, double-stranded RNA ade-
eling of anticancer drug sensitivity. Nature 483, 603–607.
nosine deaminase. Proc. Natl. Acad. Sci. U. S. A. 94, 8421–8426.
Bass, B.L., Weintraub, H., 1987. A developmentally regulated activity that unwinds RNA
Hideyama, T., Yamashita, T., Suzuki, T., Tsuji, S., Higuchi, M., Seeburg, P.H., Takahashi,
duplexes. Cell 48, 607–613.
R., Misawa, H., Kwak, S., 2010. Induced loss of ADAR2 engenders slow death of
Bass, B.L., 2002. RNA editing by adenosine deaminases that act on RNA. Annu. Rev.
motor neurons from Q/R site-unedited GluR2. J. Neurosci. 30, 11917–11925.
Biochem 71, 817–846.
Higuchi, M., Maas, S., Single, F.N., Hartner, J., Rozov, A., Burnashev, N., Feldmeyer, D.,
Bazak, L., Haviv, A., Barak, M., Jacob-Hirsch, J., Deng, P., Zhang, R., Isaacs, F.J., Rechavi,
Sprengel, R., Seeburg, P.H., 2000. Point mutation in an AMPA receptor gene rescues
G., Li, J.B., Eisenberg, E., Levanon, E.Y., 2014. A-to-I RNA editing occurs at over a
lethality in mice deficient in the RNA-editing enzyme ADAR2. Nature 406, 78–81.
hundred million genomic sites, located in a majority of human genes. Genome Res.
Jiang, Q., Crews, L.A., Barrett, C.L., Chun, H.-J., Court, A.C., Isquith, J.M., Zipeto, M.A.,
24, 365–376.
Goff, D.J., Minden, M., Sadarangani, A., Rusert, J.M., Dao, K.-H.T., Morris, S.R.,
Boettcher, M., McManus, Michael T., 2015. Choosing the Right Tool for the Job: RNAi,
Goldstein, L.S.B., Marra, M.A., Frazer, K.A., Jamieson, C.H.M., 2013. ADAR1 pro-
TALEN, or CRISPR. Mol. Cell 58, 575–585.
motes malignant progenitor reprogramming in chronic myeloid leukemia. Proc. Natl.
Brusa, R., Zimmermann, F., Koh, D.S., Feldmeyer, D., Gass, P., Seeburg, P.H., Sprengel, R.,
Acad. Sci. U. S. A. 110, 1041–1046.
1995. Early-onset epilepsy and postnatal lethality associated with an editing-deficient
Kawahara, Y., Zinshteyn, B., Chendrimada, T.P., Shiekhattar, R., Nishikura, K., 2007a.
GluR-B allele in mice. Science (New York, N.Y.) 270, 1677–1680.
RNA editing of the microRNA-151 precursor blocks cleavage by the Dicer-TRBP
Burnashev, N., Zhou, Z., Neher, E., Sakmann, B., 1995. Fractional calcium currents
complex. EMBO Rep. 8, 763–769.
through recombinant GluR channels of the NMDA, AMPA and kainate receptor
Kawahara, Y., Zinshteyn, B., Sethupathy, P., Iizasa, H., Hatzigeorgiou, A.G., Nishikura, K.,
subtypes. J. Physiol. (Lond.) 485 (Pt 2), 403–418.
2007b. Redirection of silencing targets by adenosine-to-inosine editing of miRNAs.
Cenci, C., Barzotti, R., Galeano, F., Corbelli, S., Rota, R., Massimi, L., Di Rocco, C.,
Science (New York, N.Y.) 315, 1137–1140.
O’Connell, M.A., Gallo, A., 2008. Down-regulation of RNA editing in pediatric as-
Liddicoat, B.J., Piskol, R., Chalk, A.M., Ramaswami, G., Higuchi, M., Hartner, J.C., Li,
trocytomas: ADAR2 editing activity inhibits cell migration and proliferation. J. Biol.
J.B., Seeburg, P.H., Walkley, C.R., 2015. RNA editing by ADAR1 prevents MDA5
Chem. 283, 7251–7260.
sensing of endogenous dsRNA as nonself. Science 349, 1115–1120.
Chan, T.H.M., Lin, C.H., Qi, L., Fei, J., Li, Y., Yong, K.J., Liu, M., Song, Y., Chow, R.K.K.,
Liddicoat, B.J., Chalk, A.M., Walkley, C.R., 2016. ADAR1, inosine and the immune sen-
Ng, V.H.E., Yuan, Y.-F., Tenen, D.G., Guan, X.-Y., Chen, L., 2014. A disrupted RNA
sing system: distinguishing self from non-self. Wiley Interdiscip. Rev. RNA 7,
editing balance mediated by ADARs (Adenosine DeAminases that act on RNA) in
157–172.
human hepatocellular carcinoma. Gut 63, 832–843.
Maas, S., Gommans, W.M., 2009. Novel exon of mammalian ADAR2 extends open reading
Chen, L., Li, Y., Lin, C.H., Chan, T.H.M., Chow, R.K.K., Song, Y., Liu, M., Yuan, Y.-F., Fu,
frame. PLoS One 4, e4225.
L., Kong, K.L., Qi, L., Li, Y., Zhang, N., Tong, A.H.Y., Kwong, D.L.-W., Man, K., Lo,
Maas, S., Patt, S., Schrey, M., Rich, A., 2001. Underediting of glutamate receptor GluR-B
C.M., Lok, S., Tenen, D.G., Guan, X.-Y., 2013. Recoding RNA editing of AZIN1 pre-
mRNA in malignant gliomas. Proc. Natl. Acad. Sci. U. S. A. 98, 14687–14692.
disposes to hepatocellular carcinoma. Nat. Med. 19, 209–216.
Mehta, S., Shelling, A., Muthukaruppan, A., Lasham, A., Blenkiron, C., Laking, G., Print,
Cho, D.-S.C., Yang, W., Lee, J.T., Shiekhattar, R., Murray, J.M., Nishikura, K., 2003.
C., 2010. Predictive and prognostic molecular markers for cancer medicine. Ther.
Requirement of dimerization for RNA editing activity of adenosine deaminases acting
Adv. Med. Oncol. 2, 125–148.
on RNA. J. Biol. Chem. 278, 17093–17102.
Melcher, T., Maas, S., Herb, A., Sprengel, R., Higuchi, M., Seeburg, P.H., 1996a. RED2, a
Cho, C.J., Myung, S.-J., Chang, S., 2017. ADAR1 and MicroRNA; a hidden crosstalk in
brain-specific member of the RNA-specific adenosine deaminase family. J. Biol.
cancer. Int. J. Mol. Sci. 18.
Chem. 271, 31795–31798.
Cox, D.B.T., Platt, R.J., Zhang, F., 2015. Therapeutic genome editing: prospects and
Melcher, T., Maas, S., Herb, A., Sprengel, R., Seeburg, P.H., Higuchi, M., 1996b. A
challenges. Nat. Med. 21, 121–131.
mammalian RNA editing enzyme. Nature 379, 460–464.
Eran, A., Li, J.B., Vatalaro, K., McCarthy, J., Rahimov, F., Collins, C., Markianos, K.,
Montiel-González, M.F., Vallecillo-Viejo, I.C., Rosenthal, J.J.C., 2016. An efficient system
Margulies, D.M., Brown, E.N., Calvo, S.E., Kohane, I.S., Kunkel, L.M., 2013.
for selectively altering genetic information within mRNAs. Nucleic Acids Res. 44,
Comparative RNA editing in autistic and neurotypical cerebella. Mol. Psychiatry 18,
e157.
1041–1048.
Montiel-Gonzalez, M.F., Vallecillo-Viejo, I., Yudowski, G.A., Rosenthal, J.J.C., 2013.
Fellmann, C., Gowen, B.G., Lin, P.-C., Doudna, J.A., Corn, J.E., 2017. Cornerstones of
Correction of mutations within the cystic fibrosis transmembrane conductance
CRISPR-Cas in drug discovery and therapy. Nat. Rev. Drug Discov. 16, 89–100.

21
N.S. Ganem et al. Drug Resistance Updates 32 (2017) 16–22

regulator by site-directed RNA editing. Proc. Natl. Acad. Sci. U. S. A. 110, Pierce, R.C., Sadri-Vakili, G., 2015. ADAR2-dependent GluA2 editing regulates co-
18285–18290. caine seeking. Mol. Psychiatry 20, 1460–1466.
Nemlich, Y., Greenberg, E., Ortenberg, R., Besser, M.J., Barshack, I., Jacob-Hirsch, J., Schumacher, J.M., Lee, K., Edelhoff, S., Braun, R.E., 1995. Distribution of Tenr, an RNA-
Jacoby, E., Eyal, E., Rivkin, L., Prieto, V.G., Chakravarti, N., Duncan, L.M., binding protein, in a lattice-like network within the spermatid nucleus in the mouse.
Kallenberg, D.M., Galun, E., Bennett, D.C., Amariglio, N., Bar-Eli, M., Schachter, J., Biol. Reprod. 52, 1274–1283.
Rechavi, G., Markel, G., 2013. MicroRNA-mediated loss of ADAR1 in metastatic Sharma, S., Kelly, T.K., Jones, P.A., 2010. Epigenetics in cancer. Carcinogenesis 31,
melanoma promotes tumor growth. J. Clin. Invest 123, 2703–2718. 27–36.
Nimmich, M.L., Heidelberg, L.S., Fisher, J.L., 2009. RNA editing of the GABA(A) receptor Sommer, B., Köhler, M., Sprengel, R., Seeburg, P.H., 1991. RNA editing in brain controls a
alpha3 subunit alters the functional properties of recombinant receptors. Neurosci. determinant of ion flow in glutamate-gated channels. Cell 67, 11–19.
Res. 63, 288–293. Stafforst, T., Schneider, M.F., 2012. An RNA-deaminase conjugate selectively repairs
Nishikura, K., 2016. A-to-I editing of coding and non-coding RNAs by ADARs. Nat. Rev. point mutations. Angew. Chem. Int. Ed. Engl. 51, 11166–11169.
Mol. Cell Biol. 17, 83–96. Steinman, R.A., Yang, Q., Gasparetto, M., Robinson, L.J., Liu, X., Lenzner, D.E., Hou, J.,
Oakes, E., Anderson, A., Cohen-Gadol, A., Hundley, H.A., 2017. Adenosine deaminase Smith, C., Wang, Q., 2013. Deletion of the RNA-editing enzyme ADAR1 causes re-
that acts on RNA 3 (ADAR3) binding to glutamate receptor subunit B pre-mRNA gression of established chronic myelogenous leukemia in mice. Int. J. Cancer 132,
inhibits RNA editing in glioblastoma. J. Biol. Chem. 292, 4326–4335. 1741–1750.
Patterson, J.B., Samuel, C.E., 1995. Expression and regulation by interferon of a double- Streit, A.K., Derst, C., Wegner, S., Heinemann, U., Zahn, R.K., Decher, N., 2011. RNA
stranded-RNA-specific adenosine deaminase from human cells: evidence for two editing of Kv1.1 channels may account for reduced ictogenic potential of 4-amino-
forms of the deaminase. Mol. Cell. Biol. 15, 5376–5388. pyridine in chronic epileptic rats. Epilepsia 52, 645–648.
Paz, N., Levanon, E.Y., Amariglio, N., Heimberger, A.B., Ram, Z., Constantini, S., Barbash, Thomas, J.M., Beal, P.A., 2017. How do ADARs bind RNA? New protein-RNA structures
Z.S., Adamsky, K., Safran, M., Hirschberg, A., Krupsky, M., Ben-Dov, I., Cazacu, S., illuminate substrate recognition by the RNA editing ADARs. Bioessays 39 (pp. n/a-
Mikkelsen, T., Brodie, C., Eisenberg, E., Rechavi, G., 2007. Altered adenosine-to-in- n/a).
osine RNA editing in human cancer. Genome Res. 17, 1586–1595. Tomaselli, S., Galeano, F., Alon, S., Raho, S., Galardi, S., Polito, V.A., Presutti, C.,
Paz-Yaacov, N., Bazak, L., Buchumenski, I., Porath, H.T., Danan-Gotthold, M., Vincenti, S., Eisenberg, E., Locatelli, F., Gallo, A., 2015. Modulation of microRNA
Knisbacher, B.A., Eisenberg, E., Levanon, E.Y., 2015. Elevated RNA editing activity is editing, expression and processing by ADAR2 deaminase in glioblastoma. Genome
a major contributor to transcriptomic diversity in tumors. Cell Rep. 13, 267–276. Biol. 16, 5.
Petricoin, E.F., Belluco, C., Araujo, R.P., Liotta, L.A., 2006. The blood peptidome: a higher Vogel, P., Schneider, M.F., Wettengel, J., Stafforst, T., 2014. Improving site-directed RNA
dimension of information content for cancer biomarker discovery. Nat. Rev. Cancer 6, editing in vitro and in cell culture by chemical modification of the guideRNA. Angew.
961–967. Chem. Int. Ed. Engl. 53, 6267–6271.
Qin, Y.-R., Qiao, J.-J., Chan, T.H.M., Zhu, Y.-H., Li, F.-F., Liu, H., Fei, J., Li, Y., Guan, X.- Wang, Q., Khillan, J., Gadue, P., Nishikura, K., 2000. Requirement of the RNA editing
Y., Chen, L., 2014. Adenosine-to-inosine RNA editing mediated by ADARs in eso- deaminase ADAR1 gene for embryonic erythropoiesis. Science 290, 1765–1768.
phageal squamous cell carcinoma. Cancer Res. 74, 840–851. Wang, Q., Miyakoda, M., Yang, W., Khillan, J., Stachura, D.L., Weiss, M.J., Nishikura, K.,
Rebagliati, M.R., Melton, D.A., 1987. Antisense RNA injections in fertilized frog eggs 2004. Stress-induced apoptosis associated with null mutation of ADAR1 RNA editing
reveal an RNA duplex unwinding activity. Cell 48, 599–605. deaminase gene. J. Biol. Chem. 279, 4952–4961.
Rice, G.I., Kasher, P.R., Forte, G.M.A., Mannion, N.M., Greenwood, S.M., Szynkiewicz, M., Wang, Y., Xu, X., Yu, S., Jeong, K.J., Zhou, Z., Han, L., Tsang, Y.H., Li, J., Chen, H.,
Dickerson, J.E., Bhaskar, S.S., Zampini, M., Briggs, T.A., Jenkinson, E.M., Bacino, Mangala, L.S., Yuan, Y., Eterovic, A.K., Lu, Y., Sood, A.K., Scott, K.L., Mills, G.B.,
C.A., Battini, R., Bertini, E., Brogan, P.A., Brueton, L.A., Carpanelli, M., De Laet, C., de Liang, H., 2017. Systematic characterization of A-to-I RNA editing hotspots in
Lonlay, P., del Toro, M., Desguerre, I., Fazzi, E., Garcia-Cazorla, A., Heiberg, A., microRNAs across human cancers. Genome Res. 27, 1112–1125.
Kawaguchi, M., Kumar, R., Lin, J.-P.S.M., Lourenco, C.M., Male, A.M., Marques, W., Wettengel, J., Reautschnig, P., Geisler, S., Kahle, P.J., Stafforst, T., 2017. Harnessing
Mignot, C., Olivieri, I., Orcesi, S., Prabhakar, P., Rasmussen, M., Robinson, R.A., human ADAR2 for RNA repair − recoding a PINK1 mutation rescues mitophagy.
Rozenberg, F., Schmidt, J.L., Steindl, K., Tan, T.Y., van der Merwe, W.G., Vanderver, Nucleic Acids Res. 45, 2797–2808.
A., Vassallo, G., Wakeling, E.L., Wassmer, E., Whittaker, E., Livingston, J.H., Lebon, Yang, W., Chendrimada, T.P., Wang, Q., Higuchi, M., Seeburg, P.H., Shiekhattar, R.,
P., Suzuki, T., McLaughlin, P.J., Keegan, L.P., O'Connell, M.A., Lovell, S.C., Crow, Nishikura, K., 2006. Modulation of microRNA processing and expression through
Y.J., 2012. Mutations in ADAR1 cause Aicardi-Goutières syndrome associated with a RNA editing by ADAR deaminases. Nat. Struct. Mol. Biol. 13, 13–21.
type I interferon signature. Nat. Genet. 44, 1243–1248. Zhang, X.-J., He, P.-P., Li, M., He, C.-D., Yan, K.-L., Cui, Y., Yang, S., Zhang, K.-Y., Gao,
Samuel, C.E., 2012. ADARs: viruses and innate immunity. Curr. Top. Microbiol. Immunol. M., Chen, J.-J., Li, C.-R., Jin, L., Chen, H.-D., Xu, S.-J., Huang, W., 2004. Seven novel
353, 163–195. mutations of the ADAR gene in Chinese families and sporadic patients with dys-
Sawyers, C.L., 2008. The cancer biomarker problem. Nature 452, 548–552. chromatosis symmetrica hereditaria (DSH). Hum. Mutat. 23, 629–630.
Schmidt, H.D., McFarland, K.N., Darnell, S.B., Huizenga, M.N., Sangrey, G.R., Cha, J.H.J.,

22

You might also like